Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 115(10): 2401-2406, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29467285

RESUMO

Endothelial cell (EC)-enriched protein coding genes, such as endothelial nitric oxide synthase (eNOS), define quintessential EC-specific physiologic functions. It is not clear whether long noncoding RNAs (lncRNAs) also define cardiovascular cell type-specific phenotypes, especially in the vascular endothelium. Here, we report the existence of a set of EC-enriched lncRNAs and define a role for spliced-transcript endothelial-enriched lncRNA (STEEL) in angiogenic potential, macrovascular/microvascular identity, and shear stress responsiveness. STEEL is expressed from the terminus of the HOXD locus and is transcribed antisense to HOXD transcription factors. STEEL RNA increases the number and integrity of de novo perfused microvessels in an in vivo model and augments angiogenesis in vitro. The STEEL RNA is polyadenylated, nuclear enriched, and has microvascular predominance. Functionally, STEEL regulates a number of genes in diverse ECs. Of interest, STEEL up-regulates both eNOS and the transcription factor Kruppel-like factor 2 (KLF2), and is subject to feedback inhibition by both eNOS and shear-augmented KLF2. Mechanistically, STEEL up-regulation of eNOS and KLF2 is transcriptionally mediated, in part, via interaction of chromatin-associated STEEL with the poly-ADP ribosylase, PARP1. For instance, STEEL recruits PARP1 to the KLF2 promoter. This work identifies a role for EC-enriched lncRNAs in the phenotypic adaptation of ECs to both body position and hemodynamic forces and establishes a newer role for lncRNAs in the transcriptional regulation of EC identity.


Assuntos
Cromatina/metabolismo , Células Endoteliais , Neovascularização Fisiológica , RNA Longo não Codificante , Animais , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Hemodinâmica , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos SCID , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
2.
Circulation ; 130(14): 1168-78, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25186941

RESUMO

BACKGROUND: Cell-based therapies to augment endothelial cells (ECs) hold great therapeutic promise. Here, we report a novel approach to generate functional ECs directly from adult fibroblasts. METHODS AND RESULTS: Eleven candidate genes that are key regulators of endothelial development were selected. Green fluorescent protein (GFP)-negative skin fibroblasts were prepared from Tie2-GFP mice and infected with lentiviruses allowing simultaneous overexpression of all 11 factors. Tie2-GFP(+) cells (0.9%), representing Tie2 gene activation, were detected by flow cytometry. Serial stepwise screening revealed 5 key factors (Foxo1, Er71, Klf2, Tal1, and Lmo2) that were required for efficient reprogramming of skin fibroblasts into Tie2-GFP(+) cells (4%). This reprogramming strategy did not involve pluripotency induction because neither Oct4 nor Nanog was expressed after 5 key factor transduction. Tie2-GFP(+) cells were isolated using fluorescence-activated cell sorting and designated as induced ECs (iECs). iECs exhibited endothelium-like cobblestone morphology and expressed EC molecular markers. iECs possessed endothelial functions such as Bandeiraea simplicifolia-1 lectin binding, acetylated low-density lipoprotein uptake, capillary formation on Matrigel, and nitric oxide production. The epigenetic profile of iECs was similar to that of authentic ECs because the promoters of VE-cadherin and Tie2 genes were demethylated. mRNA profiling showed clustering of iECs with authentic ECs and highly enriched endothelial genes in iECs. In a murine model of hind-limb ischemia, iEC implantation increased capillary density and enhanced limb perfusion, demonstrating the in vivo viability and functionality of iECs. CONCLUSIONS: We demonstrated the first direct conversion of adult fibroblasts to functional ECs. These results suggest a novel therapeutic modality for cell therapy in ischemic vascular disease.


Assuntos
Células Endoteliais/citologia , Fibroblastos/citologia , Terapia Genética/métodos , Isquemia/terapia , Doenças Vasculares/terapia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores Etários , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Fibroblastos/fisiologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Fluorescência Verde/genética , Membro Posterior/irrigação sanguínea , Isquemia/patologia , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Nus , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Pele/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Doenças Vasculares/patologia
3.
Development ; 139(21): 3973-85, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22932696

RESUMO

During murine embryogenesis, the Ets factor Erg is highly expressed in endothelial cells of the developing vasculature and in articular chondrocytes of developing bone. We identified seven isoforms for the mouse Erg gene. Four share a common translational start site encoded by exon 3 (Ex3) and are enriched in chondrocytes. The other three have a separate translational start site encoded by Ex4 and are enriched in endothelial cells. Homozygous Erg(ΔEx3/ΔEx3) knockout mice are viable, fertile and do not display any overt phenotype. By contrast, homozygous Erg(ΔEx4/ΔEx4) knockout mice are embryonic lethal, which is associated with a marked reduction in endocardial-mesenchymal transformation (EnMT) during cardiac valve morphogenesis. We show that Erg is required for the maintenance of the core EnMT regulatory factors that include Snail1 and Snail2 by binding to their promoter and intronic regions.


Assuntos
Endocárdio/metabolismo , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Mesoderma/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Endocárdio/embriologia , Genótipo , Mesoderma/embriologia , Camundongos , Camundongos Knockout , Morfogênese , Proteínas Oncogênicas/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulador Transcricional ERG
4.
Blood ; 121(21): 4404-12, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23529929

RESUMO

We previously demonstrated that the first intron of the human von Willebrand factor (vWF) is required for gene expression in the endothelium of transgenic mice. Based on this finding, we hypothesized that RNA splicing plays a role in mediating vWF expression in the vasculature. To address this question, we used transient transfection assays in human endothelial cells and megakaryocytes with intron-containing and intronless human vWF promoter-luciferase constructs. Next, we generated knockin mice in which LacZ was targeted to the endogenous mouse vWF locus in the absence or presence of the native first intron or heterologous introns from the human ß-globin, mouse Down syndrome critical region 1, or hagfish coagulation factor X genes. In both the in vitro assays and the knockin mice, the loss of the first intron of vWF resulted in a significant reduction of reporter gene expression in endothelial cells but not megakaryocytes. This effect was rescued to varying degrees by the introduction of a heterologous intron. Intron-mediated enhancement of expression was mediated at a posttranscriptional level. Together, these findings implicate a role for intronic splicing in mediating lineage-specific expression of vWF in the endothelium.


Assuntos
Linhagem da Célula/genética , Endotélio Vascular/fisiologia , Splicing de RNA/genética , Fator de von Willebrand/genética , Animais , Bovinos , Éxons/genética , Técnicas de Introdução de Genes , Hemostasia/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Íntrons/genética , Óperon Lac , Camundongos , Regiões Promotoras Genéticas/genética , Especificidade da Espécie
5.
Blood ; 121(17): 3531-40, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23449636

RESUMO

Proximal promoter DNA methylation has been shown to be important for regulating gene expression. However, its relative contribution to the cell-specific expression of endothelial cell (EC)-enriched genes has not been defined. We used methyl-DNA immunoprecipitation and bisulfite conversion to analyze the DNA methylation profile of EC-enriched genes in ECs vs nonexpressing cell types, both in vitro and in vivo. We show that prototypic EC-enriched genes exhibit functional differential patterns of DNA methylation in proximal promoter regions of most (eg, CD31, von Willebrand factor [vWF], VE-cadherin, and intercellular adhesion molecule-2), but not all (eg, VEGFR-1 and VEGFR-2), EC-enriched genes. Comparable findings were evident in cultured ECs, human blood origin ECs, and murine aortic ECs. Promoter-reporter episomal transfection assays for endothelial nitric oxide synthase, VE-cadherin, and vWF indicated functional promoter activity in cell types where the native gene was not active. Inhibition of DNA methyltransferase activity indicated important functional relevance. Importantly, profiling DNA replication timing patterns indicated that EC-enriched gene promoters with differentially methylated regions replicate early in S-phase in both expressing and nonexpressing cell types. Collectively, these studies highlight the functional importance of promoter DNA methylation in controlling vascular EC gene expression.


Assuntos
Metilação de DNA , Período de Replicação do DNA , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Regiões Promotoras Genéticas/genética , Fase S/fisiologia , Animais , Antígenos CD/genética , Aorta/citologia , Aorta/metabolismo , Caderinas/genética , Bovinos , Moléculas de Adesão Celular/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Derme/citologia , Derme/metabolismo , Endotélio Vascular/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Fator de von Willebrand/genética
6.
J Biol Chem ; 287(9): 6582-91, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22235125

RESUMO

ETS-related gene (ERG) is a member of the ETS transcription factor family. Our previous studies have shown that ERG expression is highly enriched in endothelial cells (EC) both in vitro and in vivo. ERG expression is markedly repressed in response to inflammatory stimuli. It has been shown that ERG is a positive regulator of several EC-restricted genes including VE-cadherin, endoglin, and von Willebrand factor, and a negative regulator of other genes such as interleukin (IL)-8 and intercellular adhesion molecule (ICAM)-1. In this study we have identified a novel role for ERG in the regulation of EC barrier function. ERG knockdown results in marked increases in EC permeability. This is associated with a significant increase of stress fiber and gap formation in EC. Furthermore, we identify CLDN5 as a downstream target of ERG in EC. Thus, our results suggest that ERG plays a pivotal role in regulating EC barrier function and that this effect is mediated in part through its regulation of CLDN5 gene expression.


Assuntos
Permeabilidade Capilar/fisiologia , Claudinas/genética , Células Endoteliais/metabolismo , Transativadores/metabolismo , Ativação Transcricional/fisiologia , Adenoviridae/genética , Permeabilidade Capilar/efeitos dos fármacos , Claudina-5 , Vasos Coronários/citologia , Citoesqueleto/fisiologia , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/fisiologia , Microvasos/citologia , Mutagênese Sítio-Dirigida , Artéria Pulmonar/citologia , Transativadores/genética , Regulador Transcricional ERG , Fator de Necrose Tumoral alfa/farmacologia
7.
J Biol Chem ; 287(5): 3559-72, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22158614

RESUMO

Matrix metalloproteinase (MMP)-13 has a pivotal, rate-limiting function in cartilage remodeling and degradation due to its specificity for cleaving type II collagen. The proximal MMP13 promoter contains evolutionarily conserved E26 transformation-specific sequence binding sites that are closely flanked by AP-1 and Runx2 binding motifs, and interplay among these and other factors has been implicated in regulation by stress and inflammatory signals. Here we report that ELF3 directly controls MMP13 promoter activity by targeting an E26 transformation-specific sequence binding site at position -78 bp and by cooperating with AP-1. In addition, ELF3 binding to the proximal MMP13 promoter is enhanced by IL-1ß stimulation in chondrocytes, and the IL-1ß-induced MMP13 expression is inhibited in primary human chondrocytes by siRNA-ELF3 knockdown and in chondrocytes from Elf3(-/-) mice. Further, we found that MEK/ERK signaling enhances ELF3-driven MMP13 transactivation and is required for IL-1ß-induced ELF3 binding to the MMP13 promoter, as assessed by chromatin immunoprecipitation. Finally, we show that enhanced levels of ELF3 co-localize with MMP13 protein and activity in human osteoarthritic cartilage. These studies define a novel role for ELF3 as a procatabolic factor that may contribute to cartilage remodeling and degradation by regulating MMP13 gene transcription.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Metaloproteinase 13 da Matriz/biossíntese , Osteoartrite/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Cartilagem Articular/patologia , Condrócitos/patologia , Proteínas de Ligação a DNA/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/farmacologia , Metaloproteinase 13 da Matriz/genética , Camundongos , Camundongos Knockout , Osteoartrite/genética , Osteoartrite/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-ets , Elementos de Resposta/genética , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/genética
8.
Blood ; 117(1): 342-51, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20980682

RESUMO

A region of the human von Willebrand factor (VWF) gene between -2812 and the end of the first intron (termed vWF2) was previously shown to direct expression in the endothelium of capillaries and a subset of larger blood vessels in the heart and skeletal muscle. Here, our goal was to delineate the DNA sequences responsible for this effect. A series of constructs containing deletions or mutations of vWF2 coupled to LacZ were targeted to the Hprt locus of mice, and the resulting animals were analyzed for reporter gene expression. The findings demonstrate that DNA sequences between -843 and -620 are necessary for expression in capillary but not large vessel endothelium in heart and skeletal muscle. Further, expression of VWF in capillaries and larger vessels of both tissues required the presence of a native or heterologous intron. In vitro assays implicated a role for ERG-binding ETS motif at -56 in mediating basal expression of VWF. In Hprt-targeted mice, mutation of the ETS consensus motif resulted in loss of LacZ expression in the endothelium of the heart and skeletal muscle. Together, these data indicate that distinct DNA modules regulate vascular bed-specific expression of VWF.


Assuntos
Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Coração/fisiologia , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas/genética , Fator de von Willebrand/genética , Animais , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Endotélio Vascular/citologia , Feminino , Humanos , Hipoxantina Fosforribosiltransferase/genética , Íntrons/genética , Óperon Lac , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Regulador Transcricional ERG , Fator de von Willebrand/metabolismo
9.
Blood ; 118(4): 1145-53, 2011 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-21628409

RESUMO

ERG is a member of the ETS transcription factor family that is highly enriched in endothelial cells (ECs). To further define the role of ERG in regulating EC function, we evaluated the effect of ERG knock-down on EC lumen formation in 3D collagen matrices. Blockade of ERG using siRNA completely interferes with EC lumen formation. Quantitative PCR (QPCR) was used to identify potential downstream gene targets of ERG. In particular, we identified RhoJ as the Rho GTPase family member that is closely related to Cdc42 as a target of ERG. Knockdown of ERG expression in ECs led to a 75% reduction in the expression of RhoJ. Chromatin immunoprecipitation and transactivation studies demonstrated that ERG could bind to functional sites in the proximal promoter of the RhoJ gene. Knock-down of RhoJ similarly resulted in a marked reduction in the ability of ECs to form lumens. Suppression of either ERG or RhoJ during EC lumen formation was associated with a marked increase in RhoA activation and a decrease in Rac1 and Cdc42 activation and their downstream effectors. Finally, in contrast to other Rho GTPases, RhoJ exhibits a highly EC-restricted expression pattern in several different tissues, including the brain, heart, lung, and liver.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Transativadores/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Western Blotting , Técnicas de Silenciamento de Genes , Humanos , Imunoprecipitação , Lasers , Camundongos , Camundongos Nus , Microdissecção , Morfogênese , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Regulador Transcricional ERG
10.
Blood ; 114(27): 5557-66, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-19822898

RESUMO

Vascular endothelial growth factor receptor 1 (VEGFR1) is a marker for endothelial-specific gene expression. We previously reported that the human VEGFR1 promoter (between -748 and +284) contains information for expression in the intact endothelium of transgenic mice. The objective of this study was to dissect the cis-regulatory elements underlying VEGFR1 promoter activity in vitro and in vivo. In primary endothelial cells, binding sites for E74-like factor 1 (ELF-1; between -49 and -52), cyclic adenosine monophosphate response element binding (CREB; between -74 and -81), and early growth response factor 1/3 (EGR-1/3; between -16 to -25) were shown to play a positive role in gene transcription, whereas a putative E26 transformation-specificsequence (ETS) motif between -36 and -39 had a net negative effect on promoter activity. When targeted to the Hprt locus of mice, mutations of the ELF-1 binding site and the CRE element reduced promoter activity in the embryonic vasculature and resulted in a virtual loss of expression in adult endothelium. Postnatally, the EGR binding site mutant displayed significantly reduced promoter activity in a subset of vascular beds. In contrast, mutation of the -39 ETS site resulted in increased LacZ staining in multiple vascular beds. Together, these results provide new insights into the transcriptional regulatory mechanisms of VEGFR1.


Assuntos
Proteína de Ligação a CREB/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Sequência de Bases , Sítios de Ligação/genética , Sítios de Ligação/fisiologia , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Homologia de Sequência do Ácido Nucleico
11.
J Neural Transm (Vienna) ; 118(1): 23-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20972807

RESUMO

CD3ζ is a subunit of the CD3 molecule that, until recently, appeared restricted to T cells and natural killer cells. However, experimental studies have demonstrated a role of CD3ζ in dendritic outgrowth in the visual system as well as in synaptic plasticity. Given the increasing evidence for uncharacteristic recapitulation of neurodevelopmental processes in neurodegenerative diseases, in this study, we evaluated brains from subjects with Parkinson's disease and Lewy body dementia for evidence of aberrant CD3 expression. Our data shows marked CD3ζ in association with the α-synuclein containing pathological lesions, i.e., Lewy bodies and Lewy neurites, in the brains of subjects with Parkinson's disease and Lewy body dementia. This finding raises the novel concept of CD3 dysregulation in these disorders as a pathogenic factor and also furthers the increasing evidence that the recall of aberrant neurodevelopmental processes underlies the pathogenesis of neurodegenerative diseases.


Assuntos
Complexo CD3/metabolismo , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Idoso , Idoso de 80 Anos ou mais , Encéfalo/patologia , Feminino , Humanos , Imuno-Histoquímica , Corpos de Lewy/patologia , Corpos de Lewy/ultraestrutura , Masculino , Pessoa de Meia-Idade , alfa-Sinucleína/metabolismo
12.
Circ Res ; 104(9): 1049-57, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19359602

RESUMO

ERG (Ets-related gene) is an ETS transcription factor that has recently been shown to regulate a number of endothelial cell (EC)-restricted genes including VE-cadherin, von Willebrand factor, endoglin, and intercellular adhesion molecule-2. Our preliminary data demonstrate that unlike other ETS factors, ERG exhibits a highly EC-restricted pattern of expression in cultured primary cells and several adult mouse tissues including the heart, lung, and brain. In response to inflammatory stimuli, such as tumor necrosis factor-alpha, we observed a marked reduction of ERG expression in ECs. To further define the role of ERG in the regulation of normal EC function, we used RNA interference to knock down ERG. Microarray analysis of RNA derived from ERG small interfering RNA- or tumor necrosis factor-alpha-treated human umbilical vein (HUV)ECs revealed significant overlap (P<0.01) in the genes that are up- or downregulated. Of particular interest to us was a significant change in expression of interleukin (IL)-8 at both protein and RNA levels. Exposure of ECs to tumor necrosis factor-alpha is known to be associated with increased neutrophil attachment. We observed that knockdown of ERG in HUVECs is similarly associated with increased neutrophil attachment compared to control small interfering RNA-treated cells. This enhanced adhesion could be blocked with IL-8 neutralizing or IL-8 receptor blocking antibodies. ERG can inhibit the activity of the IL-8 promoter in a dose dependent manner. Direct binding of ERG to the IL-8 promoter in ECs was confirmed by chromatin immunoprecipitation. In summary, our findings support a role for ERG in promoting antiinflammatory effects in ECs through repression of inflammatory genes such as IL-8.


Assuntos
Células Endoteliais/metabolismo , Inflamação/prevenção & controle , Interleucina-8/metabolismo , Regiões Promotoras Genéticas , Transativadores/metabolismo , Transcrição Gênica , Animais , Adesão Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Endotoxemia/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-8/genética , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Proteínas Oncogênicas/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Transativadores/genética , Fatores de Transcrição , Regulador Transcricional ERG , Fator de Necrose Tumoral alfa/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 30(10): 1990-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20671229

RESUMO

OBJECTIVE: MicroRNA plays important roles in vascular biology, but the regulation of endothelial-specific microRNA is not well characterized. MicroRNA-126 (miR-126) is highly expressed in endothelial cells, and it regulates angiogenesis and vascular inflammation. Here we show that the transcription factors Ets-1 and Ets-2 regulate miR-126 expression. METHODS AND RESULTS: A genomic region between -71 and -100 bp upstream of the miR-126 transcriptional start site is critical for transactivation of the gene containing miR-126. This genomic region contains a potential Ets binding site. Mutations within the Ets binding site block transactivation, and Ets-1 and Ets-2 interact with this critical genomic region. Knockdown of endogenous Ets-1 and Ets-2 decreases miR-126 expression. Finally, knockdown of miR-126 alters regulation of an Ets-1 target gene. CONCLUSIONS: Taken together, these data show that the transcription factors Ets-1 and Ets-2 play a key role in controlling the expression of miR-126 and suggest that miR-126 may mediate some of their vascular effects.


Assuntos
Células Endoteliais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteína Proto-Oncogênica c-ets-2/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Proteínas de Ligação ao Cálcio , Células Cultivadas , Primers do DNA/genética , Família de Proteínas EGF , Fatores de Crescimento Endotelial/genética , Perfilação da Expressão Gênica , Humanos , Luciferases/genética , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Proteínas/genética , Deleção de Sequência , Transdução de Sinais , Ativação Transcricional
14.
J Biol Chem ; 284(42): 29109-24, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19674970

RESUMO

Endothelial phenotypes are highly regulated in space and time by both transcriptional and post-transcriptional mechanisms. There is increasing evidence that the GATA family of transcription factors function as signal transducers, coupling changes in the extracellular environment to changes in downstream target gene expression. Here we show that human primary endothelial cells derived from large blood vessels express GATA2, -3, and -6. Of these factors, GATA3 was expressed at the highest levels. In DNA microarrays of human umbilical vein endothelial cells (HUVEC), small interfering RNA-mediated knockdown of GATA3 resulted in reduced expression of genes associated with angiogenesis, including Tie2. At a functional level, GATA3 knockdown inhibited angiopoietin (Ang)-1-mediated but not vascular endothelial cell growth factor (VEGF)-mediated AKT signaling, cell migration, survival, and tube formation. In electrophoretic gel mobility shift assays and chromatin immunoprecipitation, GATA3 was shown to bind to regulatory regions within the 5'-untranslated region of the Tie2 gene. In co-immunoprecipitation and co-transfection assays, GATA3 and the Ets transcription factor, ELF1, physically interacted and synergized to transactivate the Tie2 promoter. GATA3 knockdown blocked the ability of Ang-1 to attenuate vascular endothelial cell growth factor stimulation of vascular cell adhesion molecule-1 expression and monocytic cell adhesion. Moreover, exposure of human umbilical vein endothelial cells to tumor necrosis factor-alpha resulted in marked down-regulation of GATA3 expression and reduction in Tie2 expression. Together, these findings suggest that GATA3 is indispensable for Ang-1-Tie2-mediated signaling in large vessel endothelial cells.


Assuntos
Fator de Transcrição GATA3/fisiologia , Regulação da Expressão Gênica , Receptor TIE-2/biossíntese , Angiopoietina-1/metabolismo , Células Endoteliais/citologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fator de Transcrição GATA3/metabolismo , Humanos , Imunoprecipitação , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Receptor TIE-2/química , Fator de Necrose Tumoral alfa/metabolismo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
BMC Genomics ; 11: 342, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20509943

RESUMO

BACKGROUND: In this study, we used a systematic bioinformatics analysis approach to elucidate genes that exhibit an endothelial cell (EC) restricted expression pattern, and began to define their regulation, tissue distribution, and potential biological role. RESULTS: Using a high throughput microarray platform, a primary set of 1,191 transcripts that are enriched in different primary ECs compared to non-ECs was identified (LCB >3, FDR <2%). Further refinement of this initial subset of transcripts, using published data, yielded 152 transcripts (representing 109 genes) with different degrees of EC-specificity. Several interesting patterns emerged among these genes: some were expressed in all ECs and several were restricted to microvascular ECs. Pathway analysis and gene ontology demonstrated that several of the identified genes are known to be involved in vasculature development, angiogenesis, and endothelial function (P < 0.01). These genes are enriched in cardiovascular diseases, hemorrhage and ischemia gene sets (P < 0.001). Most of the identified genes are ubiquitously expressed in many different tissues. Analysis of the proximal promoter revealed the enrichment of conserved binding sites for 26 different transcription factors and analysis of the untranslated regions suggests that a subset of the EC-restricted genes are targets of 15 microRNAs. While many of the identified genes are known for their regulatory role in ECs, we have also identified several novel EC-restricted genes, the function of which have yet to be fully defined. CONCLUSION: The study provides an initial catalogue of EC-restricted genes most of which are ubiquitously expressed in different endothelial cells.


Assuntos
Biologia Computacional , Células Endoteliais/metabolismo , Genes , Sítios de Ligação , Diferenciação Celular/genética , Linhagem Celular , Doença/genética , Células Endoteliais/citologia , Células Endoteliais/patologia , Perfilação da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Neovascularização Fisiológica/genética , Especificidade de Órgãos , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/metabolismo
16.
Blood ; 112(6): 2336-9, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18519813

RESUMO

We recently demonstrated that the 3-kb 5'-flanking region of the human ROBO4 gene directs endothelial cell-specific expression in vitro and in vivo. Moreover, a GA-binding protein (GABP)-binding motif at -119 was necessary for mediating promoter activity in vitro. The goal of the present study was to confirm the functional relevance of the -119 GABP-binding site in vivo. To that end, the Hprt locus of mice was targeted with a Robo4-LacZ transgenic cassette in which the GABP site was mutated. In other studies, the GABP mutation was introduced into the endogenous mouse Robo4 locus in which LacZ was knocked-in. Compared with their respective controls, the mutant promoters displayed a significant reduction in activity in embryoid bodies, embryos, and adult animals. Together, these data provide strong support for the role of the GABP-binding motif in mediating Robo4 expression in the intact endothelium.


Assuntos
Endotélio/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Animais , Sítios de Ligação/genética , Embrião de Mamíferos , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Experimentais , Distribuição Tecidual , Transplante Heterólogo
17.
Circ Res ; 103(2): 194-202, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18556575

RESUMO

The success of therapeutic vascularization and tissue engineering will rely on our ability to create vascular networks using human cells that can be obtained readily, can be expanded safely ex vivo, and can produce robust vasculogenic activity in vivo. Here we describe the formation of functional microvascular beds in immunodeficient mice by coimplantation of human endothelial and mesenchymal progenitor cells isolated from blood and bone marrow. Evaluation of implants after 1 week revealed an extensive network of human blood vessels containing erythrocytes, indicating the rapid formation of functional anastomoses within the host vasculature. The implanted endothelial progenitor cells were restricted to the luminal aspect of the vessels; mesenchymal progenitor cells were adjacent to lumens, confirming their role as perivascular cells. Importantly, the engineered vascular networks remained patent at 4 weeks in vivo. This rapid formation of long-lasting microvascular networks by postnatal progenitor cells obtained from noninvasive sources constitutes an important step forward in the development of clinical strategies for tissue vascularization.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica/fisiologia , Engenharia Tecidual/métodos , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Animais , Células da Medula Óssea/fisiologia , Comunicação Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Sangue Fetal/fisiologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Pericitos/citologia , Pericitos/fisiologia , Medicina Regenerativa , Transplante Heterólogo
18.
Vasc Med ; 15(4): 321-31, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20621955

RESUMO

The differentiation of embryonic stem cells along the endothelial cell lineage requires a tightly coordinated sequence of events that are regulated in both space and time. Although significant gaps remain in this process, major strides have been made over the past 10 years in identifying the growth factors, signal transduction pathways, and transcription factors that function together as critical mediators of this process. Examples of some of the signal transduction pathways include the hedgehog (HH), WNT, BMP, and Notch pathways. A complex interplay between growth factors, and activation of a variety of signal transduction pathways leads to the induction of transcriptional programs that promote the differentiation of embryonic stem cells along the endothelial lineage and ultimately into arterial, venous, and lymphatic endothelial cells. The purpose of this review is to summarize the recent advances in our understanding of the molecular mechanisms underlying endothelial differentiation.


Assuntos
Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos
19.
BMC Dev Biol ; 9: 72, 2009 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20030844

RESUMO

BACKGROUND: The molecular mechanisms that govern stem cell differentiation along the endothelial lineage remain largely unknown. Ets related gene (ERG) has recently been shown to participate in the transcriptional regulation of a number of endothelial specific genes including VE-cadherin (CD144), endoglin, and von Willebrand's Factor (vWF). The specific role of the ETS factor ERG during endothelial differentiation has not been evaluated. RESULTS: ERG expression and function were evaluated during the differentiation of embryonic stem cells into embryoid bodies (EB). The results of our study demonstrate that ERG is first expressed in a subpopulation of vascular endothelial growth factor receptor 2 (VEGF-R2) expressing cells that also express VE-cadherin. During ES cell differentiation, ERG expression remains restricted to cells of the endothelial lineage that eventually coalesce into primitive vascular structures within embryoid bodies. ERG also exhibits an endothelial cell (EC)-restricted pattern during embryogenesis. To further define the role of ERG during ES cell differentiation, we used a knockdown strategy to inhibit ERG expression. Delivery of three independent shRNA led to 70-85% reductions in ERG expression during ES cell differentiation compared to no change with control shRNA. ERG knockdown was associated with a marked reduction in the number of ECs, the expression of EC-restricted genes, and the formation of vascular structures. CONCLUSION: The ETS factor ERG appears to be a critical regulator of EC differentiation.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células Endoteliais/citologia , Proteínas Oncogênicas/metabolismo , Animais , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Proteínas Oncogênicas/genética , Fatores de Transcrição , Regulador Transcricional ERG
20.
Cancer Sci ; 100(2): 269-77, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19200258

RESUMO

Shikonin and beta-hydroxyisovalerylshikonin (beta-HIVS) from Lithospermum erythrorhizon inhibit angiogenesis via inhibition of vascular endothelial growth factor receptors (VEGFR) in an adenosine triphosphate-non-competitive manner, although the underlying molecular mechanism has not been fully understood. In the present study, we found that beta-HIVS inhibited angiogenesis within chicken chorioallantoic membrane approximately threefold more efficiently than shikonin. beta-HIVS also significantly inhibited angiogenesis in two other assays, induced either by Lewis lung carcinoma cells implanted in mouse dorsal skin or by VEGF in s.c. implanted Matrigel plugs and metastasis of Lewis lung carcinoma cells to lung. Therefore, using beta-HIVS as a bioprobe, we investigated the molecular mechanism of shikonin's anti-angiogenic actions. beta-HIVS inhibited the phosphorylation and expression of VEGFR2 and Tie2 without affecting VEGFR1 and fibroblast growth factor receptor 1 levels. beta-HIVS suppressed the phosphorylation but not the expression of extracellular signal-regulated kinase, and an Sp1-dependent transactivation of the VEGFR2 and Tie2 promoters, thereby suppressing the proliferation of vascular endothelial and progenitor cells. This was mimicked by an Sp1 inhibitor mithramycin A and partially rescued by Sp1 overexpression. These results implicate potential use of shikonin and beta-HIVS as leading compounds for clinical application in the future by virtue of their unique properties including: (i) inhibition of VEGFR2 and Tie2 phosphorylation in an adenosine triphosphate-non-competitive manner; (ii) simultaneous inhibition of the phosphorylation and expression of VEGFR2 and Tie2; and (iii) bifunctional inhibition of the growth in endothelial cells and vascular remodeling.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Medicamentos de Ervas Chinesas/farmacologia , Naftoquinonas/farmacologia , Neovascularização Patológica/prevenção & controle , Animais , Apoptose , Western Blotting , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Galinhas , Membrana Corioalantoide/metabolismo , Membrana Corioalantoide/patologia , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Técnicas Imunoenzimáticas , Imunoprecipitação , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA