Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cardiovasc Transl Res ; 5(5): 654-65, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22566269

RESUMO

Recent reports indicate that the adult mammalian heart is capable of limited, but measurable, cardiomyocyte turnover. While the lineage origin of the newly formed cardiomyocytes is not entirely understood, mounting evidence suggest that the epicardium and subepicardium may represent an important source of cardiac stem or progenitor cells. Stem cell niches are characterized by low oxygen tension, where stem cells preferentially utilize cytoplasmic glycolysis to meet their energy demands. However, it is unclear if the heart harbors similar hypoxic regions, or whether these regions house metabolically distinct cardiac progenitor populations. Here we identify the epicardium and subepicardium as the cardiac hypoxic niche based on [corrected] capillary density quantification, and localization of Hif-1α in the uninjured heart. We further demonstrate that this hypoxic microenvironment houses a metabolically distinct population of glycolytic progenitor cells. Finally, we show that Hif-1α regulates the glycolytic phenotype and progenitor properties of these cells. These findings highlight important anatomical and functional properties of the epicardial and subepicardial microenvironment, and the potential role of hypoxia signaling in regulation of cardiac progenitors.


Assuntos
Microambiente Celular , Metabolismo Energético , Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Pericárdio/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Hipóxia Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica , Glicólise , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Estresse Oxidativo , Perfusão , Fenótipo , Interferência de RNA , Transdução de Sinais , Transfecção
2.
Am J Hum Genet ; 87(2): 289-96, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20691403

RESUMO

The focal facial dermal dysplasias (FFDDs) are a group of inherited developmental disorders in which the characteristic diagnostic feature is bitemporal scar-like lesions that resemble forceps marks. To date, the genetic defects underlying these ectodermal dysplasias have not been determined. To identify the gene defect causing autosomal-recessive Setleis syndrome (type III FFDD), homozygosity mapping was performed with genomic DNAs from five affected individuals and 26 members of the consanguineous Puerto Rican (PR) family originally described by Setleis and colleagues. Microsatellites D2S1397 and D2S2968 were homozygous in all affected individuals, mapping the disease locus to 2q37.3. Haplotype analyses of additional markers in the PR family and a consanguineous Arab family further limited the disease locus to approximately 3 Mb between D2S2949 and D2S2253. Of the 29 candidate genes in this region, the bHLH transcription factor, TWIST2, was initially sequenced on the basis of its known involvement in murine facial development. Homozygous TWIST2 nonsense mutations, c.324C>T and c.486C>T, were identified in the affected members of the Arab and PR families, respectively. Characterization of the expressed mutant proteins, p.Q65X and p.Q119X, by electrophoretic mobility shift assays and immunoblot analyses indicated that they were truncated and unstable. Notably, Setleis syndrome patients and Twist2 knockout mice have similar facial features, indicating the gene's conserved role in mammalian development. Although human TWIST2 and TWIST1 encode highly homologous bHLH transcription factors, the finding that TWIST2 recessive mutations cause an FFDD and dominant TWIST1 mutations cause Saethre-Chotzen craniocynostosis suggests that they function independently in skin and bone development.


Assuntos
Anormalidades Múltiplas/genética , Códon sem Sentido/genética , Homozigoto , Proteínas Repressoras/genética , Proteína 1 Relacionada a Twist/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Pré-Escolar , Mapeamento Cromossômico , Cromossomos Humanos Par 3/genética , Fácies , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas Nucleares/química , Linhagem , Fenótipo , Porto Rico , Proteínas Repressoras/química , Alinhamento de Sequência , Síndrome , Proteína 1 Relacionada a Twist/química , Emirados Árabes Unidos
3.
Invest Ophthalmol Vis Sci ; 51(11): 5561-70, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20574024

RESUMO

PURPOSE: Twist2 is a member of a family of bHLH transcription factors critical for normal mesenchymal proliferation and differentiation. In this study, the authors analyzed the role of Twist2 in the eye and cornea through examination of a Twist2 loss-of-function mouse mutant. METHODS: Twist2 expression during eye development in the mouse was investigated using RT-PCR and mRNA slide in situ hybridization. Lineage tracing was performed using Cre reporter mice. Morphometric analyses were performed, and cell proliferation and cell death were investigated by immunohistochemistry using Ki67 and cleaved caspase 3 antibodies, respectively. RESULTS: In the mouse, Twist2 is expressed first in the periocular mesenchyme and subsequently in the corneal stroma and endothelium of the developing eye. Loss of Twist2 function leads to corneal thinning and a reduced population of stromal keratocytes. The reduction in the stromal cell population can be traced back to embryonic stages during which the proliferation of stromal progenitor cells is impaired and to the reduced number of proliferating cells in the corneal limbus postnatally. Adult Twist2-null mice display enophthalmia and blepharophimosis. Corneal thinning in mutant mice is not accompanied by glaucoma, an association reported in human patients. CONCLUSIONS: Twist2 is required for normal corneal keratocyte proliferation and eyelid morphogenesis in the mouse. Loss of Twist2 function leads to corneal thinning because of the reduction in stromal keratocyte proliferation.


Assuntos
Proliferação de Células , Córnea/embriologia , Córnea/patologia , Substância Própria/embriologia , Proteínas Repressoras/fisiologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Animais Recém-Nascidos , Apoptose , Blefarofimose/genética , Blefarofimose/patologia , Caspase 3/metabolismo , Diferenciação Celular/fisiologia , Substância Própria/metabolismo , Enoftalmia/genética , Enoftalmia/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Genótipo , Sequências Hélice-Alça-Hélice/fisiologia , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
PLoS Biol ; 6(12): e316, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19090621

RESUMO

Basic helix-loop-helix (bHLH) transcription factors play critical roles in lymphoid and erythroid development; however, little is known about their role in myeloid lineage development. In this study, we identify the bHLH transcription factor Twist-2 as a key negative regulator of myeloid lineage development, as manifested by marked increases in mature myeloid populations of macrophages, neutrophils, and basophils in Twist-2-deficient mice. Mechanistic studies demonstrate that Twist-2 inhibits the proliferation as well as differentiation of granulocyte macrophage progenitors (GMP) by interacting with and inhibiting the transcription factors Runx1 and C/EBPalpha. Moreover, Twist-2 was found to have a contrasting effect on cytokine production: inhibiting the production of proinflammatory cytokines such as interleukin-12 (IL-12) and interferon-gamma (IFNgamma) while promoting the regulatory cytokine IL-10 by myeloid cells. The data from further analyses suggest that Twist-2 activates the transcription factor c-Maf, leading to IL-10 expression. In addition, Twist-2 was found to be essential for endotoxin tolerance. Thus, this study reveals the critical role of Twist-2 in regulating the development of myeloid lineages, as well as the function and inflammatory responses of mature myeloid cells.


Assuntos
Células Progenitoras Mieloides/fisiologia , Proteínas Repressoras/fisiologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Endotoxinas/farmacologia , Interferon gama/antagonistas & inibidores , Interleucina-10/biossíntese , Interleucina-12/antagonistas & inibidores , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-maf/biossíntese , Proteínas Repressoras/biossíntese , Proteína 1 Relacionada a Twist/biossíntese
5.
J Exp Med ; 203(8): 1891-901, 2006 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-16831897

RESUMO

Type I interferons (IFNs) are pleiotropic cytokines with antiviral and immunomodulatory properties. The immunosuppressive actions of type I IFNs are poorly understood, but IFN-mediated suppression of TNFalpha production has been implicated in the regulation of inflammation and contributes to the effectiveness of type I IFNs in the treatment of certain autoimmune and inflammatory diseases. In this study, we investigated mechanisms by which type I IFNs suppress induction of TNFalpha production by immune complexes, Fc receptors, and Toll-like receptors. Suppression of TNFalpha production was mediated by induction and activation of the Axl receptor tyrosine kinase and downstream induction of Twist transcriptional repressors that bind to E box elements in the TNF promoter and suppress NF-kappaB-dependent transcription. Twist expression was activated by the Axl ligand Gas6 and by protein S and apoptotic cells. These results implicate Twist proteins in regulation of TNFalpha production by antiinflammatory factors and pathways, and provide a mechanism by which type I IFNs and Axl receptors suppress inflammatory cytokine production.


Assuntos
Inflamação/imunologia , Interferon-alfa/farmacologia , Proteínas Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Células Cultivadas , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Fc/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Receptor Tirosina Quinase Axl
6.
Dev Cell ; 6(3): 423-35, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15030764

RESUMO

Runx2 is necessary and sufficient for osteoblast differentiation, yet its expression precedes the appearance of osteoblasts by 4 days. Here we show that Twist proteins transiently inhibit Runx2 function during skeletogenesis. Twist-1 and -2 are expressed in Runx2-expressing cells throughout the skeleton early during development, and osteoblast-specific gene expression occurs only after their expression decreases. Double heterozygotes for Twist-1 and Runx2 deletion have none of the skull abnormalities observed in Runx2(+/-) mice, a Twist-2 null background rescues the clavicle phenotype of Runx2(+/-) mice, and Twist-1 or -2 deficiency leads to premature osteoblast differentiation. Furthermore, Twist-1 overexpression inhibits osteoblast differentiation without affecting Runx2 expression. Twist proteins' antiosteogenic function is mediated by a novel domain, the Twist box, which interacts with the Runx2 DNA binding domain to inhibit its function. In vivo mutagenesis confirms the antiosteogenic function of the Twist box. Thus, relief of inhibition by Twist proteins is a mandatory event precluding osteoblast differentiation.


Assuntos
Diferenciação Celular/fisiologia , Fatores de Regulação Miogênica/fisiologia , Proteínas Nucleares/fisiologia , Osteoblastos/fisiologia , Osteogênese/fisiologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Northern Blotting/métodos , Western Blotting/métodos , Células Cultivadas , Chlorocebus aethiops , Subunidade alfa 1 de Fator de Ligação ao Core , Análise Mutacional de DNA/métodos , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica/fisiologia , Heterozigoto , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Regulação Miogênica/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Testes de Precipitina/métodos , Prolina/genética , Estrutura Terciária de Proteína/fisiologia , RNA/análise , Ratos , Proteínas Repressoras/genética , Serina/genética , Esqueleto , Coloração e Rotulagem , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia , Transfecção/métodos , Proteína 1 Relacionada a Twist
7.
Development ; 130(13): 3063-74, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12756187

RESUMO

Human craniosynostosis syndromes, resulting from activating or neomorphic mutations in fibroblast growth factor receptor 2 (FGFR2), underscore an essential role for FGFR2 signaling in skeletal development. Embryos harboring homozygous null mutations in FGFR2 die prior to skeletogenesis. To address the role of FGFR2 in normal bone development, a conditional gene deletion approach was adopted. Homologous introduction of cre recombinase into the Dermo1 (Twist2) gene locus resulted in robust expression of CRE in mesenchymal condensations giving rise to both osteoblast and chondrocyte lineages. Inactivation of a floxed Fgfr2 allele with Dermo1-cre resulted in mice with skeletal dwarfism and decreased bone density. Although differentiation of the osteoblast lineage was not disturbed, the proliferation of osteoprogenitors and the anabolic function of mature osteoblasts were severely affected.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Osteoblastos/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Repressoras , Transdução de Sinais/fisiologia , Animais , Densidade Óssea , Osso e Ossos/diagnóstico por imagem , Diferenciação Celular/fisiologia , Linhagem da Célula , Condrócitos/citologia , Condrócitos/fisiologia , Craniossinostoses/genética , Craniossinostoses/metabolismo , Deleção de Genes , Humanos , Hibridização In Situ , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Fenótipo , Radiografia , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
Cell ; 112(2): 169-80, 2003 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-12553906

RESUMO

During Drosophila embryogenesis, the dorsal transcription factor activates the expression of twist, a transcription factor required for mesoderm formation. We show here that the mammalian twist proteins, twist-1 and -2, are induced by a cytokine signaling pathway that requires the dorsal-related protein RelA, a member of the NF-kappaB family of transcription factors. Twist-1 and -2 repress cytokine gene expression through interaction with RelA. Mice homozygous for a twist-2 null allele or doubly heterozygous for twist-1 and -2 alleles show elevated expression of proinflammatory cytokines, resulting in perinatal death from cachexia. These findings reveal an evolutionarily conserved signaling circuit in which twist proteins regulate cytokine signaling by establishing a negative feedback loop that represses the NF-kappaB-dependent cytokine pathway.


Assuntos
Citocinas/genética , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , NF-kappa B/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fatores de Transcrição , Animais , Apoptose/efeitos dos fármacos , Caquexia/genética , Citocinas/farmacologia , Genes Letais , Inflamação/genética , Camundongos , Camundongos Knockout , Mutação/genética , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Pele/patologia , Fator de Transcrição RelA , Transcrição Gênica , Ativação Transcricional , Fator de Necrose Tumoral alfa/farmacologia , Proteína 1 Relacionada a Twist
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...