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.
Development ; 141(15): 2959-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25053429

RESUMO

A complex regulatory network of morphogens and transcription factors is essential for normal cardiac development. Nkx2-5 is among the earliest known markers of cardiac mesoderm that is central to the regulatory pathways mediating second heart field (SHF) development. Here, we have examined the specific requirements for Nkx2-5 in the SHF progenitors. We show that Nkx2-5 potentiates Wnt signaling by regulating the expression of the R-spondin3 (Rspo3) gene during cardiogenesis. R-spondins are secreted factors and potent Wnt agonists that in part regulate stem cell proliferation. Our data show that Rspo3 is markedly downregulated in Nkx2-5 mutants and that Rspo3 expression is regulated by Nkx2-5. Conditional inactivation of Rspo3 in the Isl1 lineage resulted in embryonic lethality secondary to impaired development of SHF. More importantly, we find that Wnt signaling is significantly attenuated in Nkx2-5 mutants and that enhancing Wnt/ß-catenin signaling by pharmacological treatment or by transgenic expression of Rspo3 rescues the SHF defects in the conditional Nkx2-5(+/-) mutants. We have identified a previously unrecognized genetic link between Nkx2-5 and Wnt signaling that supports continued cardiac growth and proliferation during development. Identification of Rspo3 in cardiac development provides a new paradigm in temporal regulation of Wnt signaling by cardiac-specific transcription factors.


Assuntos
Coração/embriologia , Proteínas de Homeodomínio/fisiologia , Trombospondinas/fisiologia , Fatores de Transcrição/fisiologia , Via de Sinalização Wnt , Animais , Sequência de Bases , Linhagem da Célula , Proliferação de Células , Endocárdio/embriologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas , Homologia de Sequência do Ácido Nucleico , Células-Tronco/citologia , Trombospondinas/genética , Fatores de Transcrição/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
2.
Nat Commun ; 4: 1564, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23463007

RESUMO

Haematopoietic cells arise from spatiotemporally restricted domains in the developing embryo. Although studies of non-mammalian animal and in vitro embryonic stem cell models suggest a close relationship among cardiac, endocardial and haematopoietic lineages, it remains unknown whether the mammalian heart tube serves as a haemogenic organ akin to the dorsal aorta. Here we examine the haemogenic activity of the developing endocardium. Mouse heart explants generate myeloid and erythroid colonies in the absence of circulation. Haemogenic activity arises from a subset of endocardial cells in the outflow cushion and atria earlier than in the aorta-gonad-mesonephros region, and is transient and definitive in nature. Interestingly, key cardiac transcription factors, Nkx2-5 and Isl1, are expressed in and required for the haemogenic population of the endocardium. Together, these data suggest that a subset of endocardial/endothelial cells serve as a de novo source for transient definitive haematopoietic progenitors.


Assuntos
Endocárdio/fisiologia , Hematopoese/fisiologia , Animais , Endocárdio/citologia , Endocárdio/embriologia , Endocárdio/ultraestrutura , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Células Eritroides/citologia , Imunofluorescência , Átrios do Coração/citologia , Átrios do Coração/ultraestrutura , Sistema Hematopoético/citologia , Sistema Hematopoético/fisiologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Fígado/metabolismo , Camundongos , Células Mieloides/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Fatores de Transcrição/metabolismo
3.
Development ; 139(24): 4644-55, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23172916

RESUMO

Interneuronal subtype diversity lies at the heart of the distinct molecular properties and synaptic connections that shape the formation of the neuronal circuits that are necessary for the complex spatial and temporal processing of sensory information. Here, we investigate the role of Irx6, a member of the Iroquois homeodomain transcription factor family, in regulating the development of retinal bipolar interneurons. Using a knock-in reporter approach, we show that, in the mouse retina, Irx6 is expressed in type 2 and 3a OFF bipolar interneurons and is required for the expression of cell type-specific markers in these cells, likely through direct transcriptional regulation. In Irx6 mutant mice, presumptive type 3a bipolar cells exhibit an expansion of their axonal projection domain to the entire OFF region of the inner plexiform layer, and adopt molecular features of both type 2 and 3a bipolar cells, highlighted by the ectopic upregulation of neurokinin 3 receptor (Nk3r) and Vsx1. These findings reveal Irx6 as a key regulator of type 3a bipolar cell identity that prevents these cells from adopting characteristic features of type 2 bipolar cells. Analysis of the Irx6;Vsx1 double null retina suggests that the terminal differentiation of type 2 bipolar cells is dependent on the combined expression of the transcription factors Irx6 and Vsx1, but also points to the existence of Irx6;Vsx1-independent mechanisms in regulating OFF bipolar subtype-specific gene expression. This work provides insight into the generation of neuronal subtypes by revealing a mechanism in which opposing, yet interdependent, transcription factors regulate subtype identity.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter/genética , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Neurogênese/genética , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
4.
PLoS One ; 7(3): e32429, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22396765

RESUMO

The spectrum of human congenital malformations known as DiGeorge syndrome (DGS) is replicated in mice by mutation of Tbx1. Vegfa has been proposed as a modifier of DGS, based in part on the occurrence of comparable phenotypes in Tbx1 and Vegfa mutant mice. Many additional genes have been shown to cause DGS-like phenotypes in mice when mutated; these generally intersect in some manner with Tbx1, and therefore impact the same developmental processes in which Tbx1 itself is involved. In this study, using Tie2Cre, we show that endothelial-specific mutation of the gene encoding the VEGFA coreceptor neuropilin-1 (Nrp1) also replicates the most prominent terminal phenotypes that typify DGS. However, the developmental etiologies of these defects are fundamentally different from those caused by absence of TBX1. In Tie2Cre/Nrp1 mutants, initial pharyngeal organization is normal but subsequent pharyngeal organ growth is impaired, second heart field differentiation is normal but cardiac outflow tract cushion organization is distorted, neural crest cell migration is normal, and palatal mesenchyme proliferation is impaired with no change in apoptosis. Our results demonstrate that impairment of VEGF-dependent endothelial pathways leads to a spectrum of DiGeorge syndrome-type malformations, through processes that are distinguishable from those controlled by Tbx1.


Assuntos
Síndrome de DiGeorge/metabolismo , Endotélio/metabolismo , Neuropilinas/genética , Neuropilinas/metabolismo , Animais , Apoptose , Movimento Celular , Proliferação de Células , Síndrome de DiGeorge/genética , Modelos Animais de Doenças , Deleção de Genes , Mesoderma/metabolismo , Camundongos , Camundongos Mutantes , Modelos Biológicos , Morfogênese , Mutação , Crista Neural/citologia , Fenótipo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor TIE-2
5.
Dev Biol ; 361(1): 116-24, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22040871

RESUMO

Disruption of retinoic acid signaling causes a variety of pharyngeal arch artery and great vessel defects, as well as malformations in many other tissues, including those derived from the pharyngeal endoderm. Previous studies implied that arch artery defects in the context of defective RA signaling occur secondary to pharyngeal pouch segmentation defects, although this model has never been experimentally verified. In this study, we examined arch artery morphogenesis during mouse development, and the role of RA in this process. We show in normal embryos that the arch arteries form by vasculogenic differentiation of pharyngeal mesoderm. Using various genetic backgrounds and tissue-specific mutation approaches, we segregate pharyngeal arch artery and pharyngeal pouch defects in RA receptor mutants, and show that RA signal transduction only in pharyngeal mesoderm is required for arch artery formation. RA does not control pharyngeal mesodermal differentiation to endothelium, but instead promotes the aggregation of endothelial cells into nascent vessels. Expression of VE-cadherin was substantially reduced in RAR mutants, and this deficiency may underlie the arch artery defects. The consequences of disrupted mesodermal and endodermal RA signaling were restricted to the 4th and 6th arch arteries and to the 4th pharyngeal pouch, respectively, suggesting that different regulatory mechanisms control the formation of the more anterior arch arteries and pouches.


Assuntos
Artérias/embriologia , Região Branquial/irrigação sanguínea , Células Endoteliais/fisiologia , Mesoderma/metabolismo , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Animais , Antígenos CD/metabolismo , Região Branquial/embriologia , Caderinas/metabolismo , Agregação Celular/fisiologia , Diferenciação Celular/fisiologia , Células Endoteliais/citologia , Imunofluorescência , Técnicas de Inativação de Genes , Imuno-Histoquímica , Hibridização In Situ , Mesoderma/fisiologia , Camundongos , Mutação/genética , Receptores do Ácido Retinoico/genética , Tamoxifeno
6.
Development ; 138(9): 1795-805, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21429986

RESUMO

Secreted factors from the epicardium are believed to be important in directing heart ventricular cardiomyocyte proliferation and morphogenesis, although the specific factors involved have not been identified or characterized adequately. We found that IGF2 is the most prominent mitogen made by primary mouse embryonic epicardial cells and by a newly derived immortalized mouse embryonic epicardial cell line called MEC1. In vivo, Igf2 is expressed in the embryonic mouse epicardium during midgestation heart development. Using a whole embryo culture assay in the presence of inhibitors, we confirmed that IGF signaling is required to activate the ERK proliferation pathway in the developing heart, and that the epicardium is required for this response. Global disruption of the Igf2 gene, or conditional disruption of the two IGF receptor genes Igf1r and Insr together in the myocardium, each resulted in a significant decrease in ventricular wall proliferation and in ventricular wall hypoplasia. Ventricular cardiomyocyte proliferation in mutant embryos was restored to normal at E14.5, concurrent with the establishment of coronary circulation. Our results define IGF2 as a previously unexplored epicardial mitogen that is required for normal ventricular chamber development.


Assuntos
Proliferação de Células , Coração/embriologia , Fator de Crescimento Insulin-Like II/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Coração/efeitos dos fármacos , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , RNA Interferente Pequeno/farmacologia , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Receptor IGF Tipo 2/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
7.
J Cardiovasc Transl Res ; 4(1): 61-5, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21128127

RESUMO

Recent advances in stem cell biology and tissue engineering have put forth new therapeutic paradigms for treatment of myocardial disease. The aim of stem cell therapy for myocardial regeneration has been directed to induce angiogenesis for ischemic heart disease and/or introduction of new cardiomyocytes to improve the mechanical function of the failing heart. Encouraged by positive preliminary results in mouse models of myocardial infarction, clinical trials have utilized autologous skeletal myoblasts and bone-marrow-derived stem cells to treat patients in various clinical settings including acute myocardial injury, chronic angina, and heart failure. These studies have collectively shown, at best, modest improvement in cardiac function. This may be due to the fact that there is little evidence to support actual formation and/or integration of transplanted cells into the recipient myocardium. More recent and emerging data supports the finding that electrical stimulation may be an effective catalyst for sustained functional organization, integration, and maturation of transplanted cell populations into the host myocardium. A therapeutic model that utilizes electrical stimulation and/or achieves cardiac resynchronization in conjunction with stem cell transplantation may be an effective means to achieve successful myocardial regenerative therapy.


Assuntos
Terapia por Estimulação Elétrica , Cardiopatias/terapia , Miocárdio/patologia , Regeneração , Transplante de Células-Tronco , Animais , Terapia Combinada , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Cardiopatias/cirurgia , Humanos , Nicho de Células-Tronco , Resultado do Tratamento
8.
Dev Cell ; 18(3): 480-5, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20230754

RESUMO

In many experimental models and clinical examples, defects in the differentiation of the second heart field (SHF) and heart outflow tract septation defects are combined, although the mechanistic basis for this relationship has been unclear. We found that as the initial SHF population incorporates into the outflow tract, it is replenished from the surrounding progenitor territory. In retinoic acid (RA) receptor mutant mice, this latter process fails, and the outflow tract is shortened and misaligned as a result. As an additional consequence, the outflow tract is misspecified along its proximal-distal axis, which results in ectopic expression of TGFbeta2 and ectopic mesenchymal transformation of the endocardium. Reduction of TGFbeta2 gene dosage in the RA receptor-deficient background restores septation but does not rescue alignment defects, indicating that excess TGFbeta causes septation defects. This may be a common pathogenic pathway when second heart field and septation defects are coupled.


Assuntos
Coração/embriologia , Miocárdio/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Tretinoína/metabolismo , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Gravidez , Receptores do Ácido Retinoico/deficiência , Receptores do Ácido Retinoico/genética , Transdução de Sinais , Fator de Crescimento Transformador beta2/genética
9.
Pediatr Cardiol ; 30(5): 617-25, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19277768

RESUMO

The epicardium is a critical tissue that directs several aspects of heart development, particularly via the secretion of soluble factors. This review summarizes recent approaches that implicate the epicardium as the source of mitogenic factors promoting cardiomyocyte proliferation, as the source of instructive signals that direct compact zone organization (morphogenesis), and as the tissue that directs formation of the coronary vasculature.


Assuntos
Coração/embriologia , Pericárdio/embriologia , Animais , Ciclo Celular/fisiologia , Proliferação de Células , Vasos Coronários/embriologia , Substâncias de Crescimento , Camundongos , Miocárdio , Miócitos Cardíacos/fisiologia , Transdução de Sinais
10.
Nat Med ; 13(4): 455-62, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17351624

RESUMO

Cbl-associated protein (Cap) is a member of a phosphatidylinositol 3-kinase-independent pathway for insulin-stimulated translocation of the glucose transporter GLUT4. Despite this positive role of Cap in glucose uptake, here we show that deletion of the gene encoding Cap (official gene name: Sorbs1) protects against high-fat diet (HFD)-induced insulin resistance in mice while also having an opposite, insulin-sensitizing effect, accompanied by reduced tissue markers of inflammation. Given the emerging role of chronic inflammation in insulin resistance and the macrophage in initiating this inflammatory process, we considered that Sorbs1 deletion from macrophages may have resulted in the observed protection from HFD-induced insulin resistance. Using bone marrow transplantation to generate functional Sorbs1-null macrophages, we show that the insulin-sensitive phenotype can be transferred to wild-type mice by transplantation of Sorbs1-null bone marrow. These studies show that macrophages are an important cell type in the induction of insulin resistance and that Cap has a modulatory role in this function.


Assuntos
Deleção de Genes , Resistência à Insulina/genética , Proteínas dos Microfilamentos/genética , Adipócitos/metabolismo , Animais , Transplante de Medula Óssea , Gorduras na Dieta , Histocitoquímica , Immunoblotting , Resistência à Insulina/fisiologia , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo
11.
Mol Biol Cell ; 18(5): 1723-33, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17332502

RESUMO

In this report, an antisense RNA strategy has allowed us to show that disruption of ALP expression affects the expression of the muscle transcription factors myogenin and MyoD, resulting in the inhibition of muscle differentiation. Introduction of a MyoD expression construct into ALP-antisense cells is sufficient to restore the capacity of the cells to differentiate, illustrating that ALP function occurs upstream of MyoD. It is known that MyoD is under the control of serum response factor (SRF), a transcriptional regulator whose activity is modulated by actin dynamics. A dramatic reduction of actin filament bundles is observed in ALP-antisense cells and treatment of these cells with the actin-stabilizing drug jasplakinolide stimulates SRF activity and restores the capacity of the cells to differentiate. Furthermore, we show that modulation of ALP expression influences SRF activity, the level of its coactivator, MAL, and muscle differentiation. Collectively, these results suggest a critical role of ALP on muscle differentiation, likely via cytoskeletal regulation of SRF.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Fator de Resposta Sérica/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Citoesqueleto/metabolismo , Primers do DNA/genética , Expressão Gênica , Proteínas com Domínio LIM , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/genética , Fator Regulador Miogênico 5/metabolismo , RNA Antissenso/genética , Transfecção
12.
J Am Coll Cardiol ; 48(9 Suppl 1): A15-23, 2006 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-17084280

RESUMO

Heart failure is a growing major cause of human morbidity and mortality worldwide. A wave of new insights from diverse laboratories has begun to uncover new therapeutic strategies that affect the molecular pathways within cardiomyocytes that drive heart failure progression. Using an integrative approach that employs insights from genetic-based studies in mouse and humans and in vivo somatic gene transfer studies, we have uncovered a new link between stress signals mediated by mechanical stretch and defects in sarcoplasmic reticulum (SR) calcium cycling. An intrinsic mechanical stress sensing system is embedded in the Z disc of cardiomyocytes, and defects in stretch responses can lead to heart failure progression and associated increases in wall stress. Reversal of the chronic increases in wall stress by promoting SR calcium cycling can prevent and partially reverse heart failure progression in multiple genetic and acquired model systems of heart failure in both small and large animals. We propose that reversal of advanced heart failure is possible by targeting the defects in SR calcium cycling, which may be a final common pathway for the progression of many forms of heart failure.


Assuntos
Cálcio/metabolismo , Cardiomiopatia Dilatada/genética , Ciclo Celular/genética , Modelos Animais de Doenças , Terapia Genética/métodos , Insuficiência Cardíaca/genética , Proteínas Musculares/fisiologia , Miócitos Cardíacos/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Animais , Proteínas de Ligação ao Cálcio/uso terapêutico , Cardiomiopatia Hipertrófica , Conectina , Cricetinae , Proteínas do Citoesqueleto/genética , Progressão da Doença , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Proteínas com Domínio LIM , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Contração Miocárdica/genética , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/fisiologia , Estresse Mecânico , Função Ventricular Esquerda/genética
13.
Cell ; 117(3): 373-86, 2004 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-15109497

RESUMO

Human mutations in Nkx2-5 lead to progressive cardiomyopathy and conduction defects via unknown mechanisms. To define these pathways, we generated mice with a ventricular-restricted knockout of Nkx2-5, which display no structural defects but have progressive complete heart block, and massive trabecular muscle overgrowth found in some patients with Nkx2-5 mutations. At birth, mutant mice display a hypoplastic atrioventricular (AV) node and then develop selective dropout of these conduction cells. Transcriptional profiling uncovered the aberrant expression of a unique panel of atrial and conduction system-restricted target genes, as well as the ectopic, high level BMP-10 expression in the adult ventricular myocardium. Further, BMP-10 is shown to be necessary and sufficient for a major component of the ventricular muscle defects. Accordingly, loss of ventricular muscle cell lineage specification into trabecular and conduction system myocytes is a new mechanistic pathway for progressive cardiomyopathy and conduction defects in congenital heart disease.


Assuntos
Cardiomiopatias/genética , Linhagem da Célula , Bloqueio Cardíaco/genética , Cardiopatias Congênitas/fisiopatologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Acetilcolinesterase/metabolismo , Envelhecimento , Animais , Animais Recém-Nascidos , Cardiomiopatias/patologia , Condutividade Elétrica , Eletrocardiografia , Deleção de Genes , Expressão Gênica , Perfilação da Expressão Gênica , Marcação de Genes , Genes Reporter , Bloqueio Cardíaco/embriologia , Bloqueio Cardíaco/fisiopatologia , Cardiopatias Congênitas/complicações , Ventrículos do Coração/citologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Knockout , Miócitos Cardíacos/citologia , Reprodutibilidade dos Testes , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA