Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Exp Cell Res ; 317(2): 131-42, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20955695

RESUMO

AIMS: Activin A and transforming growth factor-ß1 (TGF-ß1) belong to the same family of growth and differentiation factors that modulate vascular lesion formation in distinct ways, which we wish to understand mechanistically. METHODS AND RESULTS: We investigated the expression of cell-surface receptors and activation of Smads in human vascular smooth muscle cells (SMCs) and demonstrated that activin receptor-like kinase-1 (ALK-1), ALK-4, ALK-5 and endoglin are expressed in human SMCs. As expected, TGF-ß1 activates Smad1 and Smad2 in these cells. Interestingly, activin A also induces phosphorylation of both Smads, which has not been reported for Smad1 before. Transcriptome analyses of activin A and TGF-ß1 treated SMCs with subsequent Gene-Set Enrichment Analyses revealed that many downstream gene networks are induced by both factors. However, the effect of activin A on expression kinetics of individual genes is less pronounced than for TGF-ß1, which is explained by a more rapid dephosphorylation of Smads and p38-MAPK in response to activin A. Substantial differences in expression of fibronectin, alpha-V integrin and total extracellular collagen synthesis were observed. CONCLUSIONS: Genome-wide mRNA expression analyses clarify the distinct modulation of vascular lesion formation by activin A and TGF-ß1, most significantly because activin A is non-fibrotic.


Assuntos
Receptores de Activinas Tipo II/metabolismo , Ativinas/farmacologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Fator de Crescimento Transformador beta/farmacologia , Receptores de Ativinas Tipo I/metabolismo , Ativinas/genética , Ativinas/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Humanos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Veia Safena/citologia , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Circulation ; 121(18): 2023-32, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20421523

RESUMO

BACKGROUND: Restenosis is the major drawback of percutaneous coronary interventions involving excessive activation and proliferation of vascular smooth muscle cells (SMCs). The nuclear receptor Nurr1 is an early response gene known mainly for its critical role in the development of dopamine neurons. In the present study, we investigated Nurr1 in human and experimental vascular restenosis. METHODS AND RESULTS: In a prospective cohort of 601 patients undergoing percutaneous coronary intervention, including stent placement, we found a strong association between Nurr1 haplotypes and in-stent restenosis risk. Furthermore, Nurr1 is specifically expressed in human in-stent restenosis and induced in cultured human SMCs in response to serum or tumor necrosis factor-alpha. Lentivirus-mediated gain- and loss-of-function experiments in SMCs demonstrated that overexpression of Nurr1 inhibited proliferation, consistent with increased expression of the key cell-cycle inhibitor p27(Kip1), whereas Nurr1 silencing enhanced SMC growth. The tumor necrosis factor-alpha-induced proinflammatory response of SMCs is inhibited by Nurr1, as reflected by reduced interleukin-1beta, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1 expression. Consistent with our in vitro data, endogenous Nurr1 reduced wire injury-induced proliferation and vascular lesion formation in carotid arteries of ApoE(-/-) mice. CONCLUSIONS: Nurr1 haplotypes are associated with human restenosis risk, and Nurr1 is expressed in human in-stent restenosis. In SMCs, Nurr1 inhibits proliferation and inflammatory responses, which explains the inhibition of SMC-rich lesion formation in mice. The recently identified small-molecule drugs that enhance the activity of Nurr1 reveal this nuclear receptor as an attractive novel target for (local) intervention in restenosis.


Assuntos
Doença da Artéria Coronariana/genética , Reestenose Coronária/genética , Músculo Liso Vascular/patologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Vasculite/genética , Angioplastia Coronária com Balão , Animais , Apolipoproteínas E/genética , Divisão Celular/fisiologia , Células Cultivadas , Doença da Artéria Coronariana/epidemiologia , Doença da Artéria Coronariana/terapia , Reestenose Coronária/epidemiologia , Reestenose Coronária/patologia , Vasos Coronários/imunologia , Vasos Coronários/patologia , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença/epidemiologia , Haplótipos , Humanos , Desequilíbrio de Ligação , Camundongos , Camundongos Mutantes , Músculo Liso Vascular/fisiologia , Neovascularização Fisiológica/fisiologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fatores de Risco , Stents , Vasculite/epidemiologia , Vasculite/patologia
3.
Arterioscler Thromb Vasc Biol ; 30(8): 1591-7, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20413732

RESUMO

OBJECTIVE: 6-Mercaptopurine (6-MP), the active metabolite of the immunosuppressive prodrug azathioprine, is commonly used in autoimmune diseases and transplant recipients, who are at high risk for cardiovascular disease. Here, we aimed to gain knowledge on the action of 6-MP in atherosclerosis, with a focus on monocytes and macrophages. METHODS AND RESULTS: We demonstrate that 6-MP induces apoptosis of THP-1 monocytes, involving decreased expression of the intrinsic antiapoptotic factors B-cell CLL/Lymphoma-2 (Bcl-2) and Bcl2-like 1 (Bcl-x(L)). In addition, we show that 6-MP decreases expression of the monocyte adhesion molecules platelet endothelial adhesion molecule-1 (PECAM-1) and very late antigen-4 (VLA-4) and inhibits monocyte adhesion. Screening of a panel of cytokines relevant to atherosclerosis revealed that 6-MP robustly inhibits monocyte chemoattractant chemokine-1 (MCP-1) expression in macrophages stimulated with lipopolysaccharide (LPS). Finally, local delivery of 6-MP to the vessel wall, using a drug-eluting cuff, attenuates atherosclerosis in hypercholesterolemic apolipoprotein E*3-Leiden transgenic mice (P<0.05). In line with our in vitro data, this inhibition of atherosclerosis by 6-MP was accompanied with decreased lesion monocyte chemoattractant chemokine-1 levels, enhanced vascular apoptosis, and reduced macrophage content. CONCLUSIONS: We report novel, previously unrecognized atheroprotective actions of 6-MP in cultured monocytes/macrophages and in a mouse model of atherosclerosis, providing further insight into the effect of the immunosuppressive drug azathioprine in atherosclerosis.


Assuntos
Apolipoproteína E3/metabolismo , Aterosclerose/prevenção & controle , Imunossupressores/farmacologia , Macrófagos/efeitos dos fármacos , Mercaptopurina/farmacologia , Monócitos/efeitos dos fármacos , Animais , Apolipoproteína E3/genética , Apoptose/efeitos dos fármacos , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Imunossupressores/administração & dosagem , Mediadores da Inflamação/metabolismo , Integrina alfa4beta1/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Mercaptopurina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fatores de Tempo , Proteína bcl-X/metabolismo
4.
Circulation ; 120(8): 669-76, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19667240

RESUMO

BACKGROUND: The cyclin-dependent kinase inhibitor p27(kip1) is a key regulator of smooth muscle cell and leukocyte proliferation in vascular disease, including in-stent restenosis. We therefore hypothesized that common genetic variations or single nucleotide polymorphisms in p27(kip1) may serve as a useful tool in risk stratification for in-stent restenosis. METHODS AND RESULTS: Three single nucleotide polymorphisms concerning the p27(kip1) gene (-838C>A, rs36228499; -79C>T, rs34330; +326G>T, rs2066827) were determined in a cohort of 715 patients undergoing coronary angioplasty and stent placement. We discovered that the p27(kip1)-838C>A single nucleotide polymorphism is associated with clinical in-stent restenosis; the -838AA genotype decreases the risk of target vessel revascularization (hazard ratio, 0.28; 95% confidence interval, 0.10 to 0.77). This finding was replicated in another cohort study of 2309 patients (hazard ratio, 0.61; 95% confidence interval, 0.40 to 0.93). No association was detected between this end point and the p27(kip1)-79C>T and +326G>T single nucleotide polymorphisms. We subsequently studied the functional importance of the -838C>A single nucleotide polymorphism and detected a 20-fold increased basal p27(kip1) transcriptional activity of the -838A allele containing promoter. CONCLUSIONS: Patients with the p27(kip1)-838AA genotype have a decreased risk of in-stent restenosis corresponding with enhanced promoter activity of the -838A allele of this cell-cycle inhibitor, which may explain decreased smooth muscle cell proliferation.


Assuntos
Doença da Artéria Coronariana/terapia , Reestenose Coronária/epidemiologia , Reestenose Coronária/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Idoso , Angioplastia Coronária com Balão , Divisão Celular/fisiologia , Doença da Artéria Coronariana/epidemiologia , Doença da Artéria Coronariana/genética , Inibidor de Quinase Dependente de Ciclina p27 , Feminino , Predisposição Genética para Doença/epidemiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Fatores de Risco , Stents
5.
Hum Mutat ; 30(3): 417-23, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19156841

RESUMO

Nuclear receptor subfamily 4, group A, member 2 (NR4A2, also called Nurr1) has lately become of interest with regard to atherogenesis. We examined the association between common variation in the NR4A2 gene and cardiovascular disease in the Rotterdam Study, a prospective population-based study among persons aged > or = 55 years. Three SNPs that tag common haplotypes across a 36-kb region surrounding the NR4A2 gene were determined. Four haplotypes with frequencies >1% covered 96% of the genetic variation. In 5,650 participants without history of coronary heart disease, 729 coronary heart disease events occurred during a median follow-up time of 11.9 years. NR4A2 haplotypes were neither associated with coronary events nor with intima-media thickness (IMT), carotid plaques, or ankle-arm index (AAI). NR4A2 haplotypes showed a tendency toward associations with aortic and coronary calcification (haplo.score global simulation P values 0.076 and 0.075, respectively), which seemed to be based on haplotype 2 (individual P values were both P=0.015). Furthermore, NR4A2 haplotype 3 was associated with higher high-density lipoprotein (HDL) cholesterol and haplotype 4 with lower systolic blood pressure. In conclusion, NR4A2/NURR1 haplotypes were not associated with coronary events, carotid IMT, carotid plaques, or AAI. There was a tendency toward associations with aortic calcification and coronary calcification. Associations for NR4A2 were found with both HDL levels and blood pressure. It remains to be investigated which pathophysiological mechanisms pertain to NR4A2 function in cardiovascular disease.


Assuntos
Doenças Cardiovasculares/genética , Proteínas de Ligação a DNA/genética , Haplótipos , Fatores de Transcrição/genética , Idoso , Aorta/patologia , Pressão Sanguínea , Calcinose/patologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Colesterol/análise , HDL-Colesterol/análise , Vasos Coronários/patologia , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Países Baixos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Razão de Chances , Polimorfismo de Nucleotídeo Único , Estudos Prospectivos , Túnica Íntima/patologia , Túnica Média/patologia
6.
Arterioscler Thromb Vasc Biol ; 28(7): 1339-46, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18467642

RESUMO

OBJECTIVE: Atheroprotective blood flow induces expression of anti-inflammatory Krüppel-like factor 2 (KLF2) and activates antioxidant transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in vascular endothelium. Previously, we obtained KLF2-induced gene expression profiles in ECs, containing several Nrf2 target genes. Our aim was to investigate the role of KLF2 in shear stress-mediated activation of Nrf2 in human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS: Expression of Nrf2 and its targets NAD(P)H dehydrogenase quinone 1 (NQO1) and heme oxygenase (HO-1) was elevated by shear and KLF2. KLF2 knockdown showed that shear-induced expression of NQO1 but not Nrf2 was dependent on KLF2. KLF2 overexpression in absence of flow resulted in more efficient activation of Nrf2 by tert-butyl hydroquinone (tBHQ) through enhanced nuclear localization, and promoted expression of a large panel of Nrf2-dependent genes resulting in superior protection against oxidative stress. Comparison of shear-, KLF2-, and Nrf2-induced transcriptomes showed that the majority of shear-modulated gene sets is influenced by KLF2 or Nrf2. CONCLUSIONS: We report that KLF2 substantially enhances antioxidant activity of Nrf2 by increasing its nuclear localization and activation. The synergistic activity of these two transcription factors forms a major contribution to the shear stress-elicited transcriptome in endothelial cells.


Assuntos
Antioxidantes/metabolismo , Células Endoteliais/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Transporte Ativo do Núcleo Celular , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Indução Enzimática , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Heme Oxigenase-1/biossíntese , Humanos , Fatores de Transcrição Kruppel-Like/genética , NAD(P)H Desidrogenase (Quinona)/biossíntese , Fator 2 Relacionado a NF-E2/genética , Oxidantes/farmacologia , Estresse Oxidativo , Fluxo Pulsátil , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Estresse Mecânico , Transfecção , Regulação para Cima , terc-Butil Hidroperóxido/farmacologia
7.
Circulation ; 115(4): 493-500, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17242285

RESUMO

BACKGROUND: Restenosis is a common complication after percutaneous coronary interventions and is characterized by excessive proliferation of vascular smooth muscle cells (SMCs). We have shown that the nuclear receptor Nur77 protects against SMC-rich lesion formation, and it has been demonstrated that 6-mercaptopurine (6-MP) enhances Nur77 activity. We hypothesized that 6-MP inhibits neointima formation through activation of Nur77. METHODS AND RESULTS: It is demonstrated that 6-MP increases Nur77 activity in cultured SMCs, which results in reduced [3H]thymidine incorporation, whereas Nur77 small interfering RNA knockdown partially restores DNA synthesis. Furthermore, we studied the effect of 6-MP in a murine model of cuff-induced neointima formation. Nur77 mRNA is upregulated in cuffed arteries, with optimal expression after 6 hours and elevated expression up to 7 days after vascular injury. Local perivascular delivery of 6-MP with a drug-eluting cuff significantly inhibits neointima formation in wild-type mice. Locally applied 6-MP does not affect inflammatory responses or apoptosis but inhibits expression of proliferating cell nuclear antigen and enhances protein levels of the cell-cycle inhibitor p27(Kip1) in the vessel wall. An even stronger inhibition of neointima formation in response to local 6-MP delivery was observed in transgenic mice that overexpressed Nur77. In contrast, 6-MP does not alter lesion formation in transgenic mice that overexpress a dominant-negative variant of Nur77 in arterial SMCs, which provides evidence for the involvement of Nur77-like factors. CONCLUSIONS: Enhancement of the activity of Nur77 by 6-MP protects against excessive SMC proliferation and SMC-rich neointima formation. We propose that activation of the nuclear receptor Nur77 is a rational approach to treating in-stent restenosis.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Reestenose Coronária/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Mercaptopurina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Reestenose Coronária/metabolismo , Reestenose Coronária/patologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Implantes de Medicamento , Artéria Femoral/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Fatores de Transcrição/genética , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/patologia , Artérias Umbilicais/citologia
8.
Biochem Biophys Res Commun ; 366(4): 910-6, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18086558

RESUMO

NR4A nuclear receptors are induced in the liver upon fasting and regulate hepatic gluconeogenesis. Here, we studied the role of nuclear receptor Nur77 (NR4A1) in hepatic lipid metabolism. We generated mice expressing hepatic Nur77 using adenoviral vectors, and demonstrate that these mice exhibit a modulation of the plasma lipid profile and a reduction in hepatic triglyceride. Expression analysis of >25 key genes involved in lipid metabolism revealed that Nur77 inhibits SREBP1c expression. This results in decreased SREBP1c activity as is illustrated by reduced expression of its target genes stearoyl-coA desaturase-1, mitochondrial glycerol-3-phosphate acyltransferase, fatty acid synthase and the LDL receptor, and provides a mechanism for the physiological changes observed in response to Nur77. Expression of LXR target genes Abcg5 and Abcg8 is reduced by Nur77, and may suggest involvement of LXR in the inhibitory action of Nur77 on SREBP1c expression. Taken together, our study demonstrates that Nur77 modulates hepatic lipid metabolism through suppression of SREBP1c activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Metabolismo dos Lipídeos , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Adenoviridae , Animais , Regulação da Expressão Gênica , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Triglicerídeos/sangue
9.
Arterioscler Thromb Vasc Biol ; 27(3): 532-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17194892

RESUMO

OBJECTIVE: The flow-responsive Kruppel-like factor 2 (KLF2) is crucial for maintaining endothelial cell quiescence. Here, we describe its detailed effects on transforming growth factor-beta (TGF-beta) signaling, which normally has proatherogenic effects on endothelium. METHODS AND RESULTS: In-depth analysis of genome-wide expression data shows that prolonged lentiviral-mediated overexpression of KLF2 in human umbilical vein endothelial cells (HUVECs) diminishes the expression of a large panel of established TGF-beta-inducible genes. Both baseline and TGF-beta-induced expression levels of plasminogen activator inhibitor 1 (PAI-1) and thrombospondin-1 are greatly diminished by KLF2. Using a combination of ectopic expression, small interfering RNA-mediated knockdown, and promoter activity assays, we show that KLF2 partly inhibits the phosphorylation and subsequent nuclear accumulation of Smad2, thereby suppressing the TGF-beta-induced Smad4-mediated transcriptional activity. This is achieved through TGF-beta-independent induction of inhibitory Smad7. Additionally, a full inhibition of TGF-beta signaling is functionally achieved through a simultaneous suppression of activator protein 1 (AP-1), which is an essential cofactor for TGF-beta-dependent transcription of many genes. CONCLUSIONS: The concerted mechanism by which KLF2 inhibits TGF-beta signaling through induction of inhibitory Smad7 and attenuation of AP-1 activity provides a novel mechanism by which KLF2 contributes to sustaining a quiescent, atheroprotective status of vascular endothelium.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad7/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Western Blotting , Células Cultivadas , Regulação para Baixo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like/farmacologia , Fosforilação , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Proteína Smad7/genética , Fator de Transcrição AP-1/genética , Veias Umbilicais/citologia
10.
Atherosclerosis ; 192(2): 283-90, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16938301

RESUMO

Accumulation of lipid-laden macrophages is a hallmark of atherosclerosis. The relevance of the key transcription factor nuclear factor kappaB (NF-kappaB) for macrophage-derived foam-cell formation has not been unequivocally resolved. Transgenic mice lines were generated in which NF-kappaB activation is specifically inhibited in macrophages by overexpressing a trans-dominant, non-degradable form of IkappaBalpha (IkappaBalpha (32A/36A)) under control of the macrophage-specific SR-A promoter. Alanine substitution of serines 32 and 36 prevents degradation and retains the inactive NF-kappaB/IkappaBalpha (32A/36A) complex in the cytoplasm. Similarly, stable human THP1 monocytic cell lines were generated with integrated copies of IkappaBalpha (32A/36A) cDNA. Upon treatment with oxidized low-density lipoprotein (ox-LDL), murine peritoneal macrophages from transgenic IkappaBalpha (32A/36A) mice, as well as THP1/IkappaBalpha (32A/36A) clones, display decreased lipid loading after differentiation into macrophages. This is accompanied by increased expression of the transcription factors PPARgamma and LXRalpha as well as of the major cholesterol-efflux transporter ABCA1. Paradoxically, mRNA expression of the 'lipid-uptake' receptor CD36 is also increased. Since the net result of these changes is reduction of foam-cell formation, it is proposed that under specific inhibition of NF-kappaB activation, ABCA1-mediated cholesterol efflux prevails over CD36-mediated lipid influx.


Assuntos
Células Espumosas/fisiologia , Macrófagos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Animais , Antígenos CD36/biossíntese , Linhagem Celular , Proteínas de Ligação a DNA/biossíntese , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/fisiologia , Lipoproteínas LDL/metabolismo , Receptores X do Fígado , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Inibidor de NF-kappaB alfa , Receptores Nucleares Órfãos , PPAR gama/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Depuradores Classe A/biossíntese
11.
Arterioscler Thromb Vasc Biol ; 26(10): 2288-94, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16873729

RESUMO

OBJECTIVE: Atherosclerosis is an inflammatory disease in which macrophage activation and lipid loading play a crucial role. In this study, we investigated expression and function of the NR4A nuclear receptor family, comprising Nur77 (NR4A1, TR3), Nurr1 (NR4A2), and NOR-1 (NR4A3) in human macrophages. METHODS AND RESULTS: Nur77, Nurr1, and NOR-1 are expressed in early and advanced human atherosclerotic lesion macrophages primarily in areas of plaque activation/progression as detected by in situ-hybridization and immunohistochemistry. Protein expression localizes to the nucleus. Primary and THP-1 macrophages transiently express NR4A-factors in response to lipopolysaccharide and tumor necrosis factor alpha. Lentiviral overexpression of Nur77, Nurr1, or NOR-1 reduces expression and production of interleukin (IL)-1beta and IL-6 proinflammatory cytokines and IL-8, macrophage inflammatory protein-1alpha and -1beta and monocyte chemoattractant protein-1 chemokines. In addition, NR4A-factors reduce oxidized-low-density lipoprotein uptake, consistent with downregulation of scavenger receptor-A, CD36, and CD11b macrophage marker genes. Knockdown of Nur77 or NOR-1 with gene-specific lentiviral short-hairpin RNAs resulted in enhanced cytokine and chemokine synthesis, increased lipid loading, and augmented CD11b expression, demonstrating endogenous NR4A-factors to inhibit macrophage activation, foam-cell formation, and differentiation. CONCLUSIONS: NR4A-factors are expressed in human atherosclerotic lesion macrophages and reduce human macrophage lipid loading and inflammatory responses, providing further evidence for a protective role of NR4A-factors in atherogenesis.


Assuntos
Aterosclerose/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inflamação/prevenção & controle , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Aterosclerose/patologia , Quimiocinas/antagonistas & inibidores , Citocinas/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Lentivirus/genética , Lipopolissacarídeos/farmacologia , Lipoproteínas LDL/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Esteroides/genética , Fatores de Transcrição/genética
12.
Biochem J ; 400(3): 401-10, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16886909

RESUMO

Lipid rafts and caveolae are biochemically similar, specialized domains of the PM (plasma membrane) that cluster specific proteins. However, they are morphologically distinct, implying different, possibly complementary functions. Two-dimensional gel electrophoresis preceding identification of proteins by MS was used to compare the relative abundance of proteins in DRMs (detergent-resistant membranes) isolated from HUVEC (human umbilical-vein endothelial cells), and caveolae immunopurified from DRM fractions. Various signalling and transport proteins were identified and additional cell-surface biotinylation revealed the majority to be exposed, demonstrating their presence at the PM. In resting endothelial cells, the scaffold of immunoisolated caveolae consists of only few resident proteins, related to structure [CAV1 (caveolin-1), vimentin] and transport (V-ATPase), as well as the GPI (glycosylphosphatidylinositol)-linked, surface-exposed protein CD59. Further quantitative characterization by immunoblotting and confocal microscopy of well-known [eNOS (endothelial nitric oxide synthase) and CAV1], less known [SNAP-23 (23 kDa synaptosome-associated protein) and BASP1 (brain acid soluble protein 1)] and novel [C8ORF2 (chromosome 8 open reading frame 2)] proteins showed different subcellular distributions with none of these proteins being exclusive to either caveolae or DRM. However, the DRM-associated fraction of the novel protein C8ORF2 (approximately 5% of total protein) associated with immunoseparated caveolae, in contrast with the raft protein SNAP-23. The segregation of caveolae from lipid rafts was visually confirmed in proliferating cells, where CAV1 was spatially separated from eNOS, SNAP-23 and BASP1. These results provide direct evidence for the previously suggested segregation of transport and signalling functions between specialized domains of the endothelial plasma membrane.


Assuntos
Cavéolas/metabolismo , Células Endoteliais/metabolismo , Microdomínios da Membrana/metabolismo , Transporte Proteico/fisiologia , Proteoma/metabolismo , Transdução de Sinais/fisiologia , Cavéolas/química , Caveolina 1/metabolismo , Células Cultivadas , Células Endoteliais/ultraestrutura , Humanos , Microdomínios da Membrana/química , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Peptídeos/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas Repressoras/metabolismo
13.
Cardiovasc Res ; 72(2): 231-40, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16945356

RESUMO

OBJECTIVE: The transcription factor KLF2 is considered an important mediator of the anti-inflammatory and anti-thrombotic properties of the endothelium. KLF2 is absent from low-shear, atherosclerosis-prone sites of the vascular tree but is induced by HMG-CoA reductase inhibitors (statins) in vitro. We studied KLF2-dependent induction of important determinants of the atheroprotective status of the endothelium to determine whether pharmacological intervention, e.g. by statins, can potentially replace shear stress. METHODS: Shear stress and statin effects in combination with TNF-alpha were determined in human umbilical vein endothelial cells by quantitative measurements of the steady-state levels and stability of mRNA for KLF2 and its downstream target genes thrombomodulin (TM) and endothelial nitric oxide synthase (eNOS). RESULTS: We demonstrate that prolonged shear stress has a potential that is superior to that of statins to induce the KLF2-dependent expression of eNOS and TM, especially in the presence of the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha). These effects can be attributed to the sustained stabilization of KLF2 mRNA by shear, leading to an increased KLF2 protein expression and concomitant strong induction of KLF2 downstream targets. The stabilization of KLF2 mRNA is demonstrated to be dependent on signaling involving phosphoinositide 3-kinase (PI3K). CONCLUSION: The stabilization of KLF2 steady-state levels, as induced by prolonged shear stress but not by statins, may be essential for sustaining the quiescent, atheroprotective status of the vascular endothelium under inflammatory conditions.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , RNA Mensageiro/metabolismo , Aterosclerose/imunologia , Células Cultivadas , Cromonas/farmacologia , Células Endoteliais/imunologia , Endotélio Vascular/imunologia , Imunofluorescência , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Técnicas In Vitro , Inflamação , Fatores de Transcrição Kruppel-Like/genética , Morfolinas/farmacologia , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína Oncogênica v-akt/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Mecânico , Trombomodulina/genética , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
14.
Eur J Cell Biol ; 85(11): 1131-44, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16959372

RESUMO

Members of the claudin family are involved in formation of barriers that control access to the paracellular space of epithelia. Likewise, endothelium-specific claudin-5 is involved in the function of the blood-brain barrier (BBB). Here, we assessed the role of claudin-5 in non-BBB endothelial barriers using lentiviral-driven overexpression and silencing of claudin-5 in its native environment of primary vascular endothelial cells. Effects were monitored using macromolecular tracers between 342Da and 40kDa. Measurements were made both in absence and presence of transmigrating leukocytes. Freeze-fracture preparations were analyzed for effects at the ultrastructural level. We show that overexpression of claudin-5 leads to formation of elaborate networks of junction strands, which are absent in untransduced endothelial cells. Concomitantly, a modest, non-size-selective enhancement of the barrier function was observed. In contrast, silencing of endogenous claudin-5 does not influence barrier function. The efficient sealing of the endothelium during diapedesis of monocytes or granulocytes is also claudin-5 independent. Collectively, these data provide evidence for a limited contribution of claudin-5 to the barrier function of human umbilical vein endothelial cells (HUVEC), implying that, unlike selective barriers in epithelia, the barrier of non-BBB endothelium seems largely independent of claudin-directed tight junction structures.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteínas de Membrana/metabolismo , Junções Íntimas/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Claudina-5 , Células Endoteliais/ultraestrutura , Imunofluorescência , Técnica de Fratura por Congelamento , Inativação Gênica , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Proteínas de Membrana/genética , Interferência de RNA
15.
Thromb Haemost ; 95(2): 348-53, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16493499

RESUMO

Tissue factor (TF) is a transmembrane protein, which is essential for initiation of the coagulation cascade. TF has been reported to play an important role in the progression of endotoxin (lipopolysaccharide, LPS)-mediated endotoxemia, being induced in numerous tissues, such as kidney, spleen and lung. We developed and validated a rabbit anti-murine TF peptide antiserum to localize TF protein in a murine sepsis model. TF protein distribution was compared to localization of TF mRNA and fibrin deposits, the ultimate resultant of procoagulant TF activity. Evident LPS mediated TF mRNA induction was observed in the tubular area at the cortico-medullar junction in the kidney, and TF activity was increased after 6 hours of endotoxemia. In the spleen, however, TF mRNA was induced in the interfollicular region upon LPS injection, corresponding to increased TF protein in the same area. The clusters of TF-protein positive cells in the spleen are predominantly granulocytes, but no TF mRNA expression was observed within these cells. Based on these observations and the presence of TF-protein positive granulocytes after splenectomy, we hypothesize that granulocytes take-up TF for transport to other locations in order to initiate fibrin formation or to induce pro-inflammatory gene expression upon interaction with factor VIIa.


Assuntos
Regulação da Expressão Gênica , Granulócitos/fisiologia , RNA Mensageiro/análise , Sepse/genética , Tromboplastina/genética , Animais , Endotoxemia , Feminino , Fibrina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Granulócitos/metabolismo , Soros Imunes , Inflamação/genética , Rim , Lipopolissacarídeos/farmacologia , Camundongos , Transporte Proteico , Coelhos , Baço , Tromboplastina/análise , Tromboplastina/metabolismo , Distribuição Tecidual
16.
Circ Res ; 90(10): 1128-34, 2002 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-12039804

RESUMO

Activin A alters the characteristics of human arterial smooth muscle cells (SMCs) toward a contractile, quiescent phenotype. We hypothesize that activin A may prevent SMC-rich neointimal hyperplasia. Here, we study the effect of adenovirus-mediated expression of activin A on neointima formation in vitro and in vivo. Human saphenous vein organ cultures, in which a neointima is formed spontaneously, were infected either with activin A- or lacZ-adenovirus. Activin A-overexpression reduces neointima formation by 78%, whereas no significant reduction was observed after control infection. In addition, the effect of activin A on neointima formation was assessed in vivo in mice with cuffed femoral arteries. In activin A adenovirus-infected mice (IV injection), neointimal hyperplasia is reduced by 77% compared with the SMC-rich neointima in mock-infected or in noninfected mice. Cultured human saphenous vein SMCs and murine aorta SMCs were incubated with activin A and an increased expression of SM22alpha and SM alpha-actin mRNA, and SM alpha-actin protein was demonstrated. Laser-capture microdissection on sections of cuffed murine arteries and subsequent real-time RT-PCR established in vivo induction of SM alpha-actin mRNA in the media of activin A-treated mice. In summary, activin A inhibits neointima formation in vitro and in vivo by preventing SMC dedifferentiation.


Assuntos
Ativinas/genética , Adenoviridae/genética , Arteriopatias Oclusivas/prevenção & controle , Subunidades beta de Inibinas/genética , Músculo Liso Vascular/metabolismo , Ativinas/metabolismo , Ativinas/farmacologia , Animais , Arteriopatias Oclusivas/metabolismo , Arteriopatias Oclusivas/patologia , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Artéria Femoral/citologia , Artéria Femoral/patologia , Vetores Genéticos , Humanos , Hiperplasia , Subunidades beta de Inibinas/metabolismo , Subunidades beta de Inibinas/farmacologia , Cinética , Camundongos , Contração Muscular , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Técnicas de Cultura de Órgãos , Fenótipo , RNA Mensageiro/biossíntese , Veia Safena/anatomia & histologia , Veia Safena/citologia , Túnica Íntima/patologia
17.
Circulation ; 106(12): 1530-5, 2002 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-12234960

RESUMO

BACKGROUND: Smooth muscle cells (SMCs) play a key role in intimal thickening in atherosclerosis and restenosis. The precise signaling pathways by which the proliferation of SMCs is regulated are largely unknown. The TR3 orphan receptor, the mitogen-induced nuclear orphan receptor (MINOR), and the nuclear receptor of T cells (NOT) are a subfamily of transcription factors belonging to the nuclear receptor superfamily and are induced in activated SMCs. In this study, we investigated the role of these transcription factors in SMC proliferation in atherogenesis. METHODS AND RESULTS: Multiple human vascular specimens at distinct stages of atherosclerosis (lesion types II to V by American Heart Association classification) derived from 14 different individuals were studied for expression of these transcription factors. We observed expression of TR3, MINOR, and NOT in neointimal SMCs, whereas no expression was detected in medial SMCs. Adenovirus-mediated expression of a dominant-negative variant of TR3, which suppresses the transcriptional activity of each subfamily member, increases DNA synthesis and decreases p27(Kip1) protein expression in cultured SMCs. We generated transgenic mice that express this dominant-negative variant or full-length TR3 under control of a vascular SMC-specific promoter. Carotid artery ligation of transgenic mice that express the dominant-negative variant of TR3 in arterial SMCs, compared with lesions formed in wild-type mice, results in a 3-fold increase in neointimal formation, whereas neointimal formation is inhibited 5-fold in transgenic mice expressing full-length TR3. CONCLUSIONS: Our results reveal that TR3 and possibly other members of this transcription factor subfamily inhibit vascular lesion formation. These transcription factors could serve as novel targets in the treatment of vascular disease.


Assuntos
Arteriosclerose/etiologia , Proteínas de Ligação a DNA/fisiologia , Músculo Liso Vascular/metabolismo , Receptores de Esteroides , Receptores dos Hormônios Tireóideos , Fatores de Transcrição/fisiologia , Adenoviridae/genética , Animais , Arteriosclerose/genética , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Cardiotônicos/metabolismo , Artérias Carótidas/cirurgia , DNA/biossíntese , Proteínas de Ligação a DNA/genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , RNA Mensageiro/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
18.
Arterioscler Thromb Vasc Biol ; 22(12): 1978-83, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12482822

RESUMO

OBJECTIVE: We previously reported that plasminogen activator inhibitor 1 (PAI-1), in the presence of vitronectin (VN), inhibits thrombin activity in vitro. Furthermore, we demonstrated in human atherosclerotic plaques the colocalization of thrombin, PAI-1, and VN, as well as activity of thrombin and PAI-1. Here, we show that PAI-1 is a local thrombin inhibitor in vivo. METHODS AND RESULTS: We used the murine carotid artery ligation model to assess the role of PAI-1 and VN in stenosis by using PAI-1-deficient (PAI-1(-/-)) and VN(-/-) mice. Ligation resulted in a smooth muscle cell (SMC)-rich intima without infiltrating cells. We show that PAI-1(-/-) and VN(-/-) mice generate a larger intima than wild-type mice as the result of more extensive SMC proliferation, as evidenced by cell counting and staining for proliferating cell-nuclear antigen. CONCLUSIONS: In PAI-1(-/-) mice, excessive intima formation is prevented by the thrombin-specific inhibitor hirudin. Finally, immunohistochemical analysis revealed PAI-1, VN, and (pro)thrombin antigen in intimal lesions. Our observations are compatible with inhibition of thrombin-mediated SMC proliferation by PAI-1/VN complexes.


Assuntos
Artérias Carótidas , Estenose das Carótidas/metabolismo , Estenose das Carótidas/prevenção & controle , Modelos Animais de Doenças , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Vitronectina/fisiologia , Animais , Artérias Carótidas/cirurgia , Divisão Celular/fisiologia , Tecido Conjuntivo/química , Hirudinas/farmacologia , Imuno-Histoquímica , Ligadura/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/citologia , Inibidor 1 de Ativador de Plasminogênio/deficiência , Inibidor 1 de Ativador de Plasminogênio/imunologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Protrombina/imunologia , Protrombina/metabolismo , Túnica Íntima/química , Túnica Íntima/citologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/fisiologia , Túnica Média/química , Vitronectina/deficiência , Vitronectina/imunologia , Vitronectina/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 23(9): 1535-40, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842839

RESUMO

OBJECTIVE: Endothelial cells play a pivotal role in vascular homeostasis. In this study, we investigated the function of the nerve growth factor-induced protein-B (NGFI-B) subfamily of nuclear receptors comprising the TR3 orphan receptor (TR3), mitogen-induced nuclear orphan receptor (MINOR), and nuclear orphan receptor of T cells (NOT) in endothelial cells. METHODS AND RESULTS: The mRNA expression of TR3, MINOR, and NOT in atherosclerotic lesions was assessed in human vascular specimens. Each of these factors is expressed in smooth muscle cells, as described before, and in subsets of endothelial cells, implicating that they might affect endothelial cell function. Adenoviral overexpression of TR3 in cultured endothelial cells resulted in decreased [3H]thymidine incorporation, whereas a dominant-negative TR3 variant that inhibits the activity of endogenous TR3-like factors enhanced DNA synthesis. TR3 interfered with progression of the cell cycle by upregulating p27Kip1 and downregulating cyclin A, whereas expression levels of a number of other cell cycle-associated proteins remained unchanged. CONCLUSIONS: These findings demonstrate that TR3 is a modulator of endothelial cell proliferation and arrests endothelial cells in the G1 phase of the cell cycle by influencing cell cycle protein levels. We hypothesize involvement of TR3 in the maintenance of integrity of the vascular endothelium.


Assuntos
Ciclo Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Receptores de Esteroides/biossíntese , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/biossíntese , Receptores dos Hormônios Tireóideos/fisiologia , Adenoviridae/genética , Divisão Celular/genética , Divisão Celular/fisiologia , Células Cultivadas , DNA Complementar/genética , Endotélio Vascular/virologia , Regulação da Expressão Gênica/genética , Técnicas de Transferência de Genes , Variação Genética/genética , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Peptídeos/genética , Peptídeos/fisiologia , Estrutura Terciária de Proteína/genética , Receptores de Esteroides/deficiência , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/deficiência , Receptores dos Hormônios Tireóideos/genética , Deleção de Sequência/genética , Células-Tronco/química , Células-Tronco/metabolismo , Células-Tronco/virologia , Ativação Transcricional/genética , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Replicação Viral/genética
20.
Arterioscler Thromb Vasc Biol ; 23(1): 64-8, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12524226

RESUMO

OBJECTIVE: Cardiac ankyrin repeat protein (CARP) is a transcription factor-related protein that has been studied most extensively in the heart. In the present study, we investigated the expression and the potential function of CARP in human and murine atherosclerosis. METHODS AND RESULTS: CARP expression was observed by in situ hybridization in endothelial cells lining human atherosclerotic plaques, whereas lesion macrophages were devoid of CARP. Furthermore, we established that CARP mRNA and smooth muscle (SM) alpha-actin antigen both colocalized in a subset of intimal smooth muscle cells (SMCs), whereas no CARP mRNA was encountered in quiescent SMCs in the media. The CARP mRNA-expressing intimal SMCs were distinct from intimal SMCs that synthesized the activation marker osteopontin or proliferating cell nuclear antigen. In addition, we showed that activin A, a member of the TGFbeta superfamily that prevents SMC-rich lesion formation, induced CARP mRNA expression in cultured SMCs. CONCLUSIONS: Based on our data and the knowledge that CARP reduces the proliferation of cultured SMCs, we propose that CARP is involved in inhibition of vascular lesion formation.


Assuntos
Ativinas/fisiologia , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Subunidades beta de Inibinas/fisiologia , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/fisiologia , Proteínas Repressoras/biossíntese , Proteínas Repressoras/fisiologia , Adulto , Idoso , Animais , Repetição de Anquirina/fisiologia , Arteriosclerose/prevenção & controle , Divisão Celular/fisiologia , Células Cultivadas , Reestenose Coronária/metabolismo , Reestenose Coronária/patologia , Endotélio Vascular/química , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Humanos , Artéria Ilíaca/química , Artéria Ilíaca/metabolismo , Artéria Ilíaca/patologia , Macrófagos/química , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas Musculares/biossíntese , Proteínas Musculares/fisiologia , Músculo Liso Vascular/patologia , RNA Mensageiro/biossíntese , Veia Safena/química , Veia Safena/metabolismo , Veia Safena/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA