Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Development ; 129(8): 2031-42, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11934868

RESUMO

Connexin43 knockout mice die neonatally from conotruncal heart malformation and outflow obstruction. Previous studies have indicated the involvement of neural crest perturbations in these cardiac anomalies. We provide evidence for the involvement of another extracardiac cell population, the proepicardial cells. These cells give rise to the vascular smooth muscle cells of the coronary arteries and cardiac fibroblasts in the heart. We have observed the abnormal presence of fibroblast and vascular smooth muscle cells in the infundibular pouches of the connexin43 knockout mouse heart. In addition, the connexin43 knockout mice exhibit a variety of coronary artery patterning defects previously described for neural crest-ablated chick embryos, such as anomalous origin of the coronary arteries, absent left or right coronary artery, and accessory coronary arteries. However, we show that proepicardial cells also express connexin43 gap junctions abundantly. The proepicardial cells are functionally well coupled, and this coupling is significantly reduced with the loss of connexin43 function. Further analysis revealed an elevation in the speed of cell locomotion and cell proliferation rate in the connexin43-deficient proepicardial cells. A parallel analysis of proepicardial cells in transgenic mice with dominant negative inhibition of connexin43 targeted only to neural crest cells showed none of these coupling, proliferation or migration changes. These mice exhibit outflow obstruction, but no infundibular pouches. Together these findings indicate an important role for connexin43 in coronary artery patterning, a role that probably involves the proepicardial and cardiac neural crest cells. We discuss the potential involvement of connexin43 in human cardiovascular anomalies involving the coronary arteries.


Assuntos
Conexina 43/fisiologia , Vasos Coronários/embriologia , Junções Comunicantes/metabolismo , Neovascularização Fisiológica/fisiologia , Animais , Biomarcadores , Diferenciação Celular , Divisão Celular , Movimento Celular , Conexina 43/genética , Circulação Coronária , Coração , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/biossíntese , Proteínas Musculares/biossíntese , Músculo Liso Vascular/metabolismo , Miosinas/biossíntese , Pericárdio/embriologia
3.
J Biol Chem ; 276(19): 16418-24, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-11279108

RESUMO

Serum response factor (SRF) plays an important role in regulating smooth muscle cell (SMC) development and differentiation. To understand the molecular mechanisms underlying the activity of SRF in SMCs, the two CArG box-containing elements in the arterial SMC-specific SM22alpha promoter, SME-1 and SME-4, were functionally and biochemically characterized. Mutations that abolish binding of SRF to the SM22alpha promoter totally abolish promoter activity in transgenic mice. Moreover, a multimerized copy of either SME-1 or SME-4 subcloned 5' of the minimal SM22alpha promoter (base pairs -90 to +41) is necessary and sufficient to restrict transgene expression to arterial SMCs in transgenic mice. In contrast, a multimerized copy of the c-fos SRE is totally inactive in arterial SMCs and substitution of the c-fos SRE for the CArG motifs within the SM22alpha promoter inactivates the 441-base pair SM22alpha promoter in transgenic mice. Deletion analysis revealed that the SME-4 CArG box alone is insufficient to activate transcription in SMCs and additional 5'-flanking nucleotides are required. Nuclear protein binding assays revealed that SME-4 binds SRF, YY1, and four additional SMC nuclear proteins. Taken together, these data demonstrate that binding of SRF to specific CArG boxes is necessary, but not sufficient, to restrict transgene expression to SMCs in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Genes fos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/fisiologia , Proteínas Nucleares/metabolismo , Regiões 3' não Traduzidas/genética , Células 3T3 , Regiões 5' não Traduzidas/genética , Animais , Aorta , Artérias/embriologia , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Células HeLa , Coração/embriologia , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Ratos , Deleção de Sequência , Fator de Resposta Sérica , Fatores de Transcrição/metabolismo , beta-Galactosidase/genética
4.
Mol Cell Biol ; 21(4): 1336-44, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11158319

RESUMO

SM22alpha is a 22-kDa smooth muscle cell (SMC) lineage-restricted protein that physically associates with cytoskeletal actin filament bundles in contractile SMCs. To examine the function of SM22alpha, gene targeting was used to generate SM22alpha-deficient (SM22(-/-LacZ)) mice. The gene targeting strategy employed resulted in insertion of the bacterial lacZ reporter gene at the SM22alpha initiation codon, permitting precise analysis of the temporal and spatial pattern of SM22alpha transcriptional activation in the developing mouse. Northern and Western blot analyses confirmed that the gene targeting strategy resulted in a null mutation. Histological analysis of SM22(+/-LacZ) embryos revealed detectable beta-galactosidase activity in the unturned embryonic day 8.0 embryo in the layer of cells surrounding the paired dorsal aortae concomitant with its expression in the primitive heart tube, cephalic mesenchyme, and yolk sac vasculature. Subsequently, during postnatal development, beta-galactosidase activity was observed exclusively in arterial, venous, and visceral SMCs. SM22alpha-deficient mice are viable and fertile. Their blood pressure and heart rate do not differ significantly from their control SM22alpha(+/-) and SM22alpha(+/+) littermates. The vasculature and SMC-containing tissues of SM22alpha-deficient mice develop normally and appear to be histologically and ultrastructurally similar to those of their control littermates. Taken together, these data demonstrate that SM22alpha is not required for basal homeostatic functions mediated by vascular and visceral SMCs in the developing mouse. These data also suggest that signaling pathways that regulate SMC specification and differentiation from local mesenchyme are activated earlier in the angiogenic program than previously recognized.


Assuntos
Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Músculo Liso/citologia , Músculo Liso/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/fisiologia , Desenvolvimento Muscular , Proteínas Musculares/fisiologia , Músculo Liso/crescimento & desenvolvimento , Transdução de Sinais , Ativação Transcricional
5.
Mol Ther ; 1(6): 555-65, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10933980

RESUMO

Gene transfer of Fas ligand (CD95L) using adenoviral vectors has been shown to generate apoptotic responses and potent inflammatory reactions that can be used to induce the regression of malignancies in vivo, but these vectors also cause significant hepatotoxicity that may limit their clinical utility. Here we describe an adenoviral vector encoding CD95L with restricted gene expression that reduces its toxicity in vivo. Preclinical efficacy and gene expression studies of lineage-restricted CD95L adenoviral vectors were performed. To enhance its cytotoxicity and reduce potential systemic effects, a noncleavable CD95L was made by deleting a segment containing the cleavage site (CD95L deltaQP). Higher CD95L expression of this mutant was observed on the tumor cell surface, together with a reduction in the release of soluble CD95L. This CD95L cleavage mutant was then expressed under control of a smooth muscle-specific promoter, SM22apha, and analyzed for its ability to suppress the growth of tumors of smooth muscle origin in vivo. Growth of human leiomyosarcomas but not gliomas was inhibited after ADV gene transfer into tumor-bearing immunodeficient mice. In contrast to viral promoters, in which mortality was uniformly seen after injection of 10(12) particles, no significant hepatic injury or systemic toxicity was observed in mice, and the maximum tolerated dose was increased > or = 10- to 100-fold. These findings suggest that restricted specificity of adenoviral CD95L gene expression enhances the safety of this approach for cancer gene therapy.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos , Glicoproteínas de Membrana/genética , Neoplasias/terapia , Animais , Apoptose , Proteína Ligante Fas , Expressão Gênica , Terapia Genética/efeitos adversos , Glioma/imunologia , Glioma/patologia , Glioma/terapia , Humanos , Leiomiossarcoma/imunologia , Leiomiossarcoma/patologia , Leiomiossarcoma/terapia , Fígado/lesões , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso/imunologia , Mutação , Neoplasias/imunologia , Neoplasias/patologia , Regiões Promotoras Genéticas , Segurança , Células Tumorais Cultivadas
6.
J Biol Chem ; 275(39): 30387-93, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-10866994

RESUMO

Prolonged serum deprivation induces a structurally and functionally contractile phenotype in about 1/6 of cultured airway myocytes, which exhibit morphological elongation and accumulate abundant contractile apparatus-associated proteins. We tested the hypothesis that transcriptional activation of genes encoding these proteins accounts for their accumulation during this phenotypic transition by measuring the transcriptional activities of the murine SM22 and human smooth muscle myosin heavy chain promoters during transient transfection in subconfluent, serum fed or 7 day serum-deprived cultured canine tracheal smooth muscle cells. Contrary to our expectation, SM22 and smooth muscle myosin heavy chain promoter activities (but not viral murine sarcoma virus-long terminal repeat promoter activity) were decreased in long term serum-deprived myocytes by at least 8-fold. Because serum response factor (SRF) is a required transcriptional activator of these and other smooth muscle-specific promoters, we evaluated the expression and function of SRF in subconfluent and long term serum-deprived cells. Whole cell SRF mRNA and protein were maintained at high levels in serum-deprived myocytes, but SRF transcription-promoting activity, nuclear SRF binding to consensus CArG sequences, and nuclear SRF protein were reduced. Furthermore, immunocytochemistry revealed extranuclear redistribution of SRF in serum-deprived myocytes; nuclear localization of SRF was restored after serum refeeding. These results uncover a novel mechanism for physiological control of smooth muscle-specific gene expression through extranuclear redistribution of SRF and consequent down-regulation of its transcription-promoting activity.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso/fisiologia , Cadeias Pesadas de Miosina/genética , Proteínas Nucleares/metabolismo , Animais , Transporte Biológico , Compartimento Celular , Meios de Cultura Livres de Soro , Citoplasma/metabolismo , Proteínas de Ligação a DNA/isolamento & purificação , Cães , Regulação para Baixo , Regulação da Expressão Gênica , Músculo Liso/citologia , Regiões Promotoras Genéticas , Fator de Resposta Sérica , Traqueia/citologia , Fator de Transcrição AP-2 , Fatores de Transcrição/isolamento & purificação
7.
J Biol Chem ; 275(26): 19949-54, 2000 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-10779506

RESUMO

Gene targeting studies have demonstrated that the zinc finger transcription factor GATA-6 lies upstream in a transcriptional cascade that controls differentiation of the visceral endoderm. To understand the function of GATA-6 in the visceral endoderm and to identify genes regulated by GATA-6 in this tissue, subtractive hybridization was performed using template cDNAs derived from differentiated wild-type embryonic stem (ES) cells and GATA-6(-/-) ES cells, respectively. These analyses revealed that the gene encoding Dab2, a mitogen-responsive phosphoprotein, is differentially expressed in wild-type and GATA-6-deficient ES cells. Consistent with these findings, Dab2 is expressed in the visceral endoderm of wild-type embryos but not in the visceral endoderm of GATA-6-deficient embryos. Cotransfection experiments demonstrate that the human Dab2 promoter can be transactivated by forced expression of GATA-6 in NIH-3T3 cells. In contrast, forced expression of GATA-4 does not transactivate the human Dab2 promoter and Dab2 is expressed in the visceral endoderm of GATA-4 null embryos. Surprisingly, the specificity of GATA-6-induced transactivation of the Dab2 promoter is not mediated through its zinc finger DNA-binding domain. Taken together, these data demonstrate that the mitogen-responsive phosphoprotein Dab2 is a downstream target of GATA-6 in the visceral endoderm. Moreover, these data demonstrate that molecular mechanisms have evolved that direct, and distinguish, the functional specificity of GATA family members when they are developmentally coexpressed.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular , Proteínas de Ligação a DNA/metabolismo , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fosfoproteínas/metabolismo , Proteínas , Fatores de Transcrição/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Sequência de Bases , Diferenciação Celular , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição GATA4 , Fator de Transcrição GATA6 , Genes Supressores de Tumor , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/genética , Plasmídeos , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Ativação Transcricional , Proteínas Supressoras de Tumor
8.
Mol Med ; 6(11): 983-91, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11147575

RESUMO

BACKGROUND: Gene transfer into vascular smooth muscle cells (vsmcs) holds promise for studying the pathogenesis of arterial disorders. However, a potential limitation of vectors with heterologous promoters is organ toxicity resulting from unrestricted transgene expression. Vascular smooth muscle cell-specific gene expression could increase the safety of vectors for vascular diseases. MATERIALS AND METHODS: To develop vectors that target gene expression to vsmcs, we constructed vectors encoding human placental alkaline phosphatase (hpAP) and chloramphenicol transferase (CAT) driven by a 441-bp region of the murine SM22alpha promoter (AdSM22alpha-hpAP). RESULTS: Transfection of AdSM22alpha-hpAP into vascular and nonvascular cells resulted in the expression of alkaline phosphatase (AP) in primary arterial and venous smcs, but not in primary endothelial cells or National Institutes of Health (NIH) 3T3 cells. Expression of AP was observed on 32.5 +/- 1.4% of primary pig vsmcs-infected AdSM22alpha-hpAP at a multiplicity of infection (MOI) of 500; whereas, infection with AdCMV-hpAP resulted in 100 +/- 0.0% expression at a MOI of 250. In vitro, expression from the heterologous cytomegalovirus (CMV) promoter was approximately 10(3)-fold higher in vsmcs, compared with the SM22alpha promoter. Following introduction of AdSM22alpha-hpAP vectors into balloon-injured pig arteries, AP recombinant protein was detected in neointimal (2.23 +/- 1.14%) and medial (0.56 +/- 0.21%) smcs, but not in endothelial or adventitial cells. In contrast, AdCMV-hpAP vectors led to AP expression in intimal endothelial and smcs cells (39.14 +/- 10.09%) and medial smcs (2.84 +/- 1.05%). AP expression was not observed in endothelial or vsmcs following transfection with the control vector, adenoviral vector lacking E1 (AddeltaE1). CONCLUSIONS: The SM22alpha promoter programs recombinant gene expression exclusively to vascular smcs in vitro and in vivo. Although expression levels are lower than with heterologous promoters, these vectors may provide a safe and effective tool for gene therapy of vascular diseases.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Regiões Promotoras Genéticas , Animais , Camundongos
9.
Genes Dev ; 12(22): 3579-90, 1998 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9832509

RESUMO

GATA6 belongs to a family of zinc finger transcription factors that play important roles in transducing nuclear events that regulate cellular differentiation and embryonic morphogenesis in vertebrate species. To examine the function of GATA6 during embryonic development, gene targeting was used to generate GATA6-deficient (GATA6(-/-)) ES cells and mice harboring a null mutation in GATA6. Differentiated embryoid bodies derived from GATA6(-/-) ES cells lack a covering layer of visceral endoderm and severely attenuate, or fail to express, genes encoding early and late endodermal markers, including HNF4, GATA4, alpha-fetoprotein (AFP), and HNF3beta. Homozygous GATA6(-/-) mice died between embryonic day (E) 6.5 and E7. 5 and exhibited a specific defect in endoderm differentiation including severely down-regulated expression of GATA4 and absence of HNF4 gene expression. Moreover, widespread programmed cell death was observed within the embryonic ectoderm of GATA6-deficient embryos, a finding also observed in HNF4-deficient embryos. Consistent with these data, forced expression of GATA6 activated the HNF4 promoter in nonendodermal cells. Finally, to examine the function of GATA6 during later embryonic development, GATA6(-/-)-C57BL/6 chimeric mice were generated. lacZ-tagged GATA6(-/-) ES cells contributed to all embryonic tissues with the exception of the endodermally derived bronchial epithelium. Taken together, these data suggest a model in which GATA6 lies upstream of HNF4 in a transcriptional cascade that regulates differentiation of the visceral endoderm. In addition, these data demonstrate that GATA6 is required for establishment of the endodermally derived bronchial epithelium.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Endoderma/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Fosfoproteínas/genética , Fatores de Transcrição/genética , Vísceras/crescimento & desenvolvimento , Células 3T3 , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Embrião de Mamíferos/patologia , Fator de Transcrição GATA6 , Marcação de Genes , Genótipo , Fator 4 Nuclear de Hepatócito , Histocitoquímica , Hibridização In Situ , Pulmão/embriologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica , RNA Mensageiro/genética , Ativação Transcricional/genética
10.
J Clin Invest ; 100(5): 1006-14, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9276717

RESUMO

Gene transfer using replication-defective adenoviruses (RDAd) holds promise for the treatment of vascular proliferative disorders, but is potentially limited by the capacity of these viruses to infect multiple cell lineages. We have generated an RDAd vector, designated AdSM22-lacZ, which encodes the bacterial lacZ reporter gene under the transcriptional control of the smooth muscle cell (SMC)-specific SM22alpha promoter. Here, we show that in vitro AdSM22-lacZ programs expression of the lacZ reporter gene in primary rat aortic SMCs and immortalized A7r5 SMCs, but not in primary human umbilical vein endothelial cells (HUVECs) or NIH 3T3 cells. Consistent with these results, after intraarterial administration of AdSM22-lacZ to control and balloon-injured rat carotid arteries, beta-galactosidase activity was detected within SMCs of the tunica media and neointima, but not within endothelial or adventitial cells. Moreover, intravenous administration of AdSM22-lacZ did not result in lacZ gene expression in the liver or lungs. Finally, we have shown that direct injection of AdSM22-lacZ into SMC-containing tissues such as the ureter and bladder results in high-level transgene expression in visceral SMCs. Taken together, these results demonstrate that transgene expression after infection with an RDAd vector can be regulated in an SMC lineage-restricted fashion by using a transcriptional cassette containing the SMC-specific SM22alpha promoter. The demonstration of an efficient gene delivery system targeted specifically to SMCs provides a novel means to restrict expression of recombinant gene products to vascular or visceral SMCs in vivo.


Assuntos
Adenoviridae/genética , Terapia Genética , Músculo Liso Vascular , Músculo Liso Vascular/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Vírus Defeituosos/genética , Vetores Genéticos , Humanos , Óperon Lac , Músculo Esquelético/metabolismo , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Transgenes , Replicação Viral
11.
Mol Cell Biol ; 17(4): 2266-78, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9121477

RESUMO

The SM22alpha promoter has been used as a model system to define the molecular mechanisms that regulate smooth muscle cell (SMC) specific gene expression during mammalian development. The SM22alpha gene is expressed exclusively in vascular and visceral SMCs during postnatal development and is transiently expressed in the heart and somites during embryogenesis. Analysis of the SM22alpha promoter in transgenic mice revealed that 280 bp of 5' flanking sequence is sufficient to restrict expression of the lacZ reporter gene to arterial SMCs and the myotomal component of the somites. DNase I footprint and electrophoretic mobility shift analyses revealed that the SM22alpha promoter contains six nuclear protein binding sites (designated smooth muscle elements [SMEs] -1 to -6, respectively), two of which bind serum response factor (SRF) (SME-1 and SME-4). Mutational analyses demonstrated that a two-nucleotide substitution that selectively eliminates SRF binding to SME-4 decreases SM22alpha promoter activity in arterial SMCs by approximately 90%. Moreover, mutations that abolish binding of SRF to SME-1 and SME-4 or mutations that eliminate each SME-3 binding activity totally abolished SM22alpha promoter activity in the arterial SMCs and somites of transgenic mice. Finally, we have shown that a multimerized copy of SME-4 (bp -190 to -110) when linked to the minimal SM22alpha promoter (bp -90 to +41) is necessary and sufficient to direct high-level transcription in an SMC lineage-restricted fashion. Taken together, these data demonstrate that distinct transcriptional regulatory programs control SM22alpha gene expression in arterial versus visceral SMCs. Moreover, these data are consistent with a model in which combinatorial interactions between SRF and other transcription factors that bind to SME-4 (and that bind directly to SRF) activate transcription of the SM22alpha gene in arterial SMCs.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas dos Microfilamentos , Proteínas Musculares/genética , Músculo Liso/citologia , Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Células Cultivadas , DNA/genética , DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Óperon Lac , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Desenvolvimento Muscular , Músculo Liso/crescimento & desenvolvimento , Músculo Liso Vascular/citologia , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/metabolismo , Mutação , Regiões Promotoras Genéticas , Ratos , Fator de Resposta Sérica
12.
Genes Dev ; 11(8): 1048-60, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9136932

RESUMO

Previous studies have suggested that the GATA4 transcription factor plays an important role in regulating mammalian cardiac development. In the studies described in this report we have used gene targeting to produce GATA4-deficient mice. Homozygous GATA4-deficient (GATA4-/-) mice died between 8.5 and 10.5 days post coitum (dpc). GATA4-/- embryos displayed severe defects in both rostral-to-caudal and lateral-to-ventral folding, which were reflected in a generalized disruption of the ventral body pattern. This resulted in the defective formation of an organized foregut and anterior intestinal pore, the failure to close both the amniotic cavity and yolk sac, and the uniform lack of a ventral pericardial cavity and heart tube. Analysis of cardiac development in the GATA4-/- mice demonstrated that these embryos developed splanchnic mesoderm, which differentiated into primitive cardiac myocytes that expressed contractile proteins. However, consistent with the observed defect in ventral morphogenesis, these GATA4-/- procardiomyocytes failed to migrate to the ventral midline to form a linear heart tube and instead formed aberrant cardiac structures in the anterior and dorsolateral regions of the embryo. The defect in ventral migration of the GATA4-/- procardiomyocytes was not cell intrinsic because GATA4-/- cardiac myocytes and endocardial cells populated the hearts of GATA4-/- -C57BL/6 chimeric mice. Taken together, these results demonstrated that GATA4 is not essential for the specification of the cardiac cell lineages. However, they define a critical role for GATA4 in regulating the rostral-to-caudal and lateral-to-ventral folding of the embryo that is needed for normal cardiac morphogenesis.


Assuntos
Padronização Corporal , Proteínas de Ligação a DNA/fisiologia , Coração/embriologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Quimera , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Sistema Digestório/embriologia , Desenvolvimento Embrionário e Fetal , Feminino , Fator de Transcrição GATA4 , Fator de Transcrição GATA6 , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Mesoderma , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Morfogênese , Miocárdio/química , Miocárdio/citologia , RNA Mensageiro/análise , Fatores de Transcrição/genética , Vísceras/química , Vísceras/embriologia , Saco Vitelino/química , Saco Vitelino/embriologia
13.
Dev Biol ; 183(1): 21-36, 1997 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-9119112

RESUMO

Members of the GATA family of zinc finger transcription factors regulate critical steps of cellular differentiation during vertebrate development. In the studies described in this report, we have isolated and functionally characterized the murine GATA-5 cDNA and protein and defined the temporal and spatial pattern of GATA-5 gene expression during mammalian development. The amino terminus of the mouse GATA-5 protein shares high level amino acid sequence identity with the murine GATA-4 and -6 proteins, but not with other members of the GATA family. GATA-5 binds to the functionally important CEF-1 nuclear protein binding site in the cardiac-specific slow/cardiac troponin C (cTnC) transcriptional enhancer and overexpression of GATA-5 transactivates the cTnC enhancer in noncardiac muscle cell lines. During embryonic and postnatal development, the pattern of GATA-5 gene expression differs significantly from that of other GATA family members. In the primitive streak embryo, GATA-5 mRNA is detectable in the precardiac mesoderm. Within the embryonic heart, the GATA-5 gene is expressed within the atrial and ventricular chambers (ED 9.5), becomes restricted to the atrial endocardium (ED 12.5), and is subsequently not expressed in the heart during late fetal and postnatal development. Moreover, coincident with the earliest steps in lung development, only the GATA-5 gene is expressed within the pulmonary mesenchyme. Finally, the GATA-5 gene is expressed in tissue-restricted subsets of smooth muscle cells (SMCs), including bronchial SMCs and SMCs in the bladder wall. These data are consistent with a model in which GATA-5 performs a unique temporally and spatially restricted function in the embryonic heart and lung. Moreover, these data suggest that GATA-5 may play an important role in the transcriptional program(s) that underlies smooth muscle cell diversity.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transativadores/genética , Fatores de Transcrição/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Clonagem Molecular , DNA Complementar , Desenvolvimento Embrionário e Fetal , Fator de Transcrição GATA5 , Gástrula/química , Coração/embriologia , Pulmão/embriologia , Mesoderma/química , Camundongos , Dados de Sequência Molecular , Músculo Liso/química , Músculo Liso/citologia , Especificidade de Órgãos , RNA Mensageiro/análise , Homologia de Sequência de Aminoácidos
14.
J Biol Chem ; 272(13): 8515-24, 1997 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-9079680

RESUMO

GATA-4 is one of the earliest developmental markers of the precardiac mesoderm, heart, and gut and has been shown to activate regulatory elements controlling transcription of genes encoding cardiac-specific proteins. To elucidate the molecular mechanisms underlying the transcriptional activity of the GATA-4 protein, structure-function analyses were performed. These analyses revealed that the C-terminal zinc finger and adjacent basic domain of GATA-4 is bifunctional, modulating both DNA-binding and nuclear localization activities. The N terminus of the protein encodes two independent transcriptional Activation Domains (amino acids 1-74 and amino acids 130-177). Amino acid residues were identified within each domain that are required for transcriptional activation. Finally, we have shown that regions of Xenopus GATA-5 and -6 corresponding to Activation Domains I and II, respectively, function as potent transcriptional activators. The identification and functional characterization of two evolutionarily conserved transcriptional Activation Domains within the GATA-4/5/6 subfamily suggests that each of these domains modulates critical functions in the transcriptional regulatory program(s) encoded by GATA-4, -5, and -6 during vertebrate development. As such these data provide novel insights into the molecular mechanisms that control development of the heart.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transcrição Gênica , Dedos de Zinco , Células 3T3 , Sequência de Aminoácidos , Animais , Células COS , Sequência Conservada , DNA/metabolismo , Fator de Transcrição GATA4 , Fator de Transcrição GATA5 , Fator de Transcrição GATA6 , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Alinhamento de Sequência , Deleção de Sequência , Relação Estrutura-Atividade , Troponina C/genética
15.
Dev Biol ; 177(1): 309-22, 1996 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-8660897

RESUMO

Members of the GATA family of zinc finger transcription factors play important roles in the development of several mesodermally derived cell lineages. In the studies described in this report, we have isolated and functionally characterized the murine GATA-6 cDNA and protein and defined the temporal and spatial patterns of GATA-6 gene expression during mammalian development. The GATA-6 and -4 proteins share high-level amino acid sequence identity over a proline-rich region at the amino terminus of the protein that is not conserved in other GATA family members. GATA-6 binds to a functionally important nuclear protein binding site within the cardiac-specific cardiac troponin C (cTnC) transcriptional enhancer. Moreover, the cTnC promoter enhancer can be transactivated by overexpression of GATA-6 in noncardiac muscle cells. During early murine embryonic development, the patterns of GATA-6 and -4 gene expression are similar, with expression of GATA-6 restricted to the precardiac mesoderm, the embryonic heart tube, and the primitive gut. However, coincident with the onset of vasculogenesis and development of the respiratory and urogenital tracts, only the GATA-6 gene is expressed in arterial smooth muscle cells, the developing bronchi, and the urogenital ridge and bladder. These data are consistent with a model in which GATA-6 functions in concert with GATA-4 to direct tissue-specific gene expression during formation of the mammalian heart and gastrointestinal tract, but performs a unique function in programming lineage-restricted gene expression in the arterial system, the bladder, and the embryonic lung.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/fisiologia , Fatores de Transcrição/genética , Dedos de Zinco/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Sistema Cardiovascular/embriologia , Linhagem da Célula/genética , Células Clonais , DNA Complementar/isolamento & purificação , Sistema Digestório/embriologia , Fator de Transcrição GATA6 , Pulmão/embriologia , Camundongos , Dados de Sequência Molecular , Músculo Esquelético/embriologia , Músculo Liso Vascular/embriologia , RNA/análise , Ratos , Troponina/genética , Troponina C , Sistema Urogenital/embriologia
16.
Mol Cell Biol ; 16(4): 1676-86, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8657143

RESUMO

The protein product of the c-fps/fes (c-fes) proto-oncogene has been implicated in the normal development of myeloid cells (macrophages and neutrophils). mRNA for c-fes has been detected exclusively in myeloid cells and vascular endothelial cells in adult mammals. Although a 13-kilobase-pair (kb) human c-fes transgene exhibits high levels of expression in mice, the sequences that confer myeloid-cell-specific expression of the human c-fes gene have not been defined. Transient-transfection experiments demonstrated that plasmids containing 446 bp of c-fes 5'-flanking sequences linked to a luciferase reporter gene were active exclusively in myeloid cells. No other DNA element within the 13-kb human c-fes locus contained positive cis-acting elements, with the exception of a weakly active region within the 3'-flanking sequences. DNase I footprinting assays revealed four distinct sites that bind myeloid nuclear proteins (-408 to -386, -293 to -254, -76 to -65, and -34 to +3). However, the first two footprints resided in sequences that were largely dispensable for transient activity. Plasmids containing 151 bp of 5'-flanking sequences confer myeloid-cell-specific gene expression. Electrophoretic mobility shift analyses demonstrated that the 151-bp region contains nuclear protein binding sites for Sp1, PU.1, and/or Elf-1, and a novel factor. This unidentified factor binds immediately 3' of the PU.1/Elf-1 sites and appears to be myeloid cell specific. Mutation of the PU.1/Elf-1 site or the 3' site (FP4-3') within the context of the c-fes promoter resulted in substantially reduced activity in transient transfections. Furthermore, transient-cotransfection assay demonstrated that PU.1 (and not Elf-1) can transactivate the c-fes promoter in nonmyeloid cell lines. We conclude that the human c-fes gene contains a strong myeloid-cell-specific promoter that is regulated by Sp1, PU.1, and a novel transcription factor.


Assuntos
Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Fator de Transcrição Sp1/genética , Transativadores , Animais , Sequência de Bases , Linhagem Celular , Humanos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Neutrófilos/metabolismo , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-fes
17.
J Biol Chem ; 271(1): 395-403, 1996 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-8550594

RESUMO

Calponin-h1 is a 34-kDa myofibrillar thin filament, actin-binding protein that is expressed exclusively in smooth muscle cells (SMCs) in adult animals. To examine the molecular mechanisms that regulate SMC-specific gene expression, we have examined the temporal, spatial, and cell cycle-regulated patterns of expression of calponin-h1 gene expression and isolated and structurally characterized the murine calponin-h1 gene. Calponin-h1 mRNA is expressed exclusively in SMC-containing tissues in adult animals. During murine embryonic development, calponin-h1 gene expression is (i) detectable in E9.5 embryos in the dorsal aorta, cardiac outflow tract, and tubular heart, (ii) sequentially up-regulated in SMC-containing tissues, and (iii) down-regulated to non-detectable levels in the heart during late fetal development. In addition, the gene is expressed in resting rat aortic SMCs, but its expression is rapidly down-regulated when growth-arrested cells reenter phase G1 of the cell cycle and proliferate. Calponin-h1 is encoded by a 10.7-kilobase single copy gene composed of seven exons, which is part of a multigene family. Transient transfection analyses demonstrated that 1.5 kilobases of calponin-h1 5'-flanking sequence is sufficient to program high level transcription of a luciferase reporter gene in cultured primary rat aortic SMCs and the smooth muscle cell line, A7r5. Taken together, these data suggest that the calponin-h1 gene will serve as an excellent model system with which to examine the molecular mechanisms that regulate SMC lineage specification, differentiation, and phenotypic modulation.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso Vascular/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Clonagem Molecular , DNA Complementar , Humanos , Camundongos , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Família Multigênica , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fenótipo , Regiões Promotoras Genéticas , Ratos , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Calponinas
18.
J Biol Chem ; 270(22): 13460-9, 1995 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-7768949

RESUMO

SM22 alpha is expressed exclusively in smooth muscle-containing tissues of adult animals and is one of the earliest markers of differentiated smooth muscle cells (SMCs). To examine the molecular mechanisms that regulate SMC-specific gene expression, we have isolated and structurally characterized the murine SM22 alpha gene. SM22 alpha is a 6.2-kilobase single copy gene composed of five exons. SM22 alpha mRNA is expressed at high levels in the aorta, uterus, lung, and intestine, and in primary cultures of rat aortic SMCs, and the SMC line, A7r5. In contrast to genes encoding SMC contractile proteins, SM22 alpha gene expression is not decreased in proliferating SMCs. Transient transfection experiments demonstrated that 441 base pairs of SM22 alpha 5'-flanking sequence was necessary and sufficient to program high level transcription of a luciferase reporter gene in both primary rat aortic SMCs and A7r5 cells. DNA sequence analyses revealed that the 441-base pair promoter contains two CArG/SRF boxes, a CACC box, and one potential MEF-2 binding site, cis-acting elements which are each important regulators of striated muscle transcription. Taken together, these studies have identified the murine SM22 alpha promoter as an excellent model system for studies of developmentally regulated, lineage-specific gene expression in SMCs.


Assuntos
Proteínas dos Microfilamentos , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Ciclo Celular/genética , Células Cultivadas , Clonagem Molecular , DNA Complementar , Camundongos , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Ratos
19.
J Physiol ; 483 ( Pt 1): 131-9, 1995 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-7776227

RESUMO

1. Beat-to-beat performance of myocardium is highly dependent on sarcomere length. The physiological basis for this effect is not well understood but presumably includes alterations in the extent of overlap between thick and thin filaments. Sarcomere length dependence of activation also appears to be involved since length-tension relationships in cardiac muscle are usually steeper than those in skeletal muscle. 2. An explanation recently proposed to account for the difference between length-tension relationships is that the cardiac isoform of troponin C (cTnC) has intrinsic properties that confer greater length-dependent changes in the Ca2+ sensitivity of tension than does skeletal troponin C (sTnC), presumably due to greater length-dependent changes in the Ca(2+)-binding affinity of cTnC. To test this hypothesis, transgenic mice were developed in which fast sTnC was expressed ectopically in the heart. This allowed a comparison of the length dependence of the Ca2+ sensitivity of tension between myocytes having thin filaments that contained either endogenous cTnC or primarily sTnC. 3. In myocytes from both transgenic and normal mice, the Ca2+ sensitivity of tension increased similarly when mean sarcomere length was increased from approximately 1.83 to 2.23 microns. In both cases, the mid-point (pCa50) of the tension-pCa (i.e. -log[Ca2+]) relationship shifted 0.12 +/- 0.01 pCa units (mean +/- S.E.M.) in the direction of lower Ca2+. 4. We conclude that the Ca2+ sensitivity of tension in myocytes changes as a function of sarcomere length but is independent of the isoform of troponin C present in the thin filaments.


Assuntos
Cálcio/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Troponina/fisiologia , Citoesqueleto de Actina/química , Animais , Separação Celular , Expressão Gênica , Camundongos , Camundongos Transgênicos , Sarcômeros/fisiologia , Troponina/análise , Troponina/genética , Troponina C
20.
Science ; 267(5197): 518-22, 1995 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-7824950

RESUMO

Vascular smooth muscle cell (SMC) proliferation in response to injury is an important etiologic factor in vascular proliferative disorders such as atherosclerosis and restenosis after balloon angioplasty. The retinoblastoma gene product (Rb) is present in the unphosphorylated and active form in quiescent primary arterial SMCs, but is rapidly inactivated by phosphorylation in response to growth factor stimulation in vitro. A replication-defective adenovirus encoding a nonphosphorylatable, constitutively active form of Rb was constructed. Infection of cultured primary rat aortic SMCs with this virus inhibited growth factor-stimulated cell proliferation in vitro. Localized arterial infection with the virus at the time of balloon angioplasty significantly reduced SMC proliferation and neointima formation in both the rat carotid and porcine femoral artery models of restenosis. These results demonstrate the role of Rb in regulating vascular SMC proliferation and suggest a gene therapy approach for vascular proliferative disorders associated with arterial injury.


Assuntos
Genes do Retinoblastoma , Terapia Genética , Músculo Liso Vascular/citologia , Proteína do Retinoblastoma/fisiologia , Doenças Vasculares/terapia , Adenoviridae/genética , Adenoviridae/fisiologia , Angioplastia com Balão , Animais , Sequência de Bases , Sangue , Artérias Carótidas/virologia , Divisão Celular , Modelos Animais de Doenças , Artéria Femoral/virologia , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Músculo Liso Vascular/patologia , Músculo Liso Vascular/virologia , Ratos , Ratos Sprague-Dawley , Suínos , Doenças Vasculares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...