Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Vasc Res ; 47(2): 168-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19851078

RESUMO

Vascular smooth muscle cell (SMC) phenotypic modulation plays a key role in atherosclerosis and is classically defined as a switch from a 'contractile' phenotype to a 'synthetic' phenotype, whereby genes that define the contractile SMC phenotype are suppressed and proliferation and/or migratory mechanisms are induced. There is also evidence that SMCs may take on a 'proinflammatory' phenotype, whereby SMCs secrete cytokines and express cell adhesion molecules, e.g. IL-8, IL-6, and VCAM-1, respectively, which may functionally regulate monocyte and macrophage adhesion and other processes during atherosclerosis. Factors that drive the inflammatory phenotype are not limited to cytokines but also include hemodynamic forces imposed on the blood vessel wall and intimate interaction of endothelial cells with SMCs, as well as changes in matrix composition in the vessel wall. However, it is critical to recognize that our understanding of the complex interaction of these multiple signal inputs has only recently begun to shed light on mechanisms that regulate the inflammatory SMC phenotype, primarily through models that attempt to recreate this environment ex vivo. The goal of this review is to summarize our current knowledge in this area and identify some of the key unresolved challenges and questions requiring further study.


Assuntos
Aterosclerose/imunologia , Inflamação/imunologia , Músculo Liso Vascular/imunologia , Miócitos de Músculo Liso/imunologia , Animais , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Hemodinâmica , Humanos , Inflamação/genética , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Fenótipo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Estresse Mecânico , Transcrição Gênica
2.
Arterioscler Thromb Vasc Biol ; 29(5): 725-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19229069

RESUMO

OBJECTIVE: Interleukin-8 (IL-8) is a soluble human-specific chemokine implicated in the development of the chronic inflammatory disease atherosclerosis. Recently, we showed that atheroprone hemodynamics induced IL-8 secretion from endothelial cells (ECs) concurrent with increased EC/smooth muscle cell (SMC) VCAM-1 expression in a human hemodynamic coculture model. Despite an IL-8 association with inflammation, we show here that blocking IL-8 activity during atheroprone flow resulted in increased levels of EC/SMC VCAM-1 expression. We tested the hypothesis that IL-8 limits SMC VCAM-1 expression in response to inflammatory stimuli, either atheroprone flow or cytokine interleukin-1beta (IL-1beta) addition. METHODS AND RESULTS: Atheroprone flow increased monocyte adhesion in both EC/SMCs, concurrent with the induction of VCAM-1 protein. VCAM-1 antisera attenuated this response. IL-1beta upregulated VCAM-1 in SMCs by 3-fold, a response inhibited by the addition of IL-8 at 24 hours. Neither IL-1beta nor IL-8 induced proliferation or migration. Neutralization of the IL-8 receptor, CXCR2, further induced VCAM-1 in the presence of IL-1beta, and phospho-p38 was required for NF-kappaB activation and VCAM-1 expression. Additionally, IL-8 reduced p38 activation and NF-kappaB activity induced by IL-1beta alone. CONCLUSIONS: Together, these findings provide evidence for a novel role whereby IL-8 limits the inflammatory response in ECs/SMCs via VCAM-1 modulation.


Assuntos
Aterosclerose/fisiopatologia , Células Endoteliais/metabolismo , Interleucina-8/fisiologia , Miócitos de Músculo Liso/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Células Cultivadas , Técnicas de Cocultura , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Humanos , Túnica Íntima/metabolismo , Veias Umbilicais/citologia
3.
Am J Pathol ; 173(4): 1220-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18772338

RESUMO

Endothelial cells acquire distinctive molecular signatures in their transformation to an angiogenic phenotype that are indicative of changes in cell behavior and function. Using a rat mesentery model of inflammation-induced angiogenesis and a panel of known endothelial markers (CD31, VE-cadherin, BS-I lectin), we identified a capillary sprout-specific endothelial phenotype that is characterized by the marked down-regulation of CD36, a receptor for the anti-angiogenic molecule thrombospondin-1 (TSP-1). TSP-1/CD36 interactions were shown to regulate angiogenesis in this model as application of TSP-1 inhibited angiogenesis and blockade of both TSP-1 and CD36 accelerated angiogenesis. Vascular endothelial growth factor, which was up-regulated in the in vivo model, elicited a dose- and time-dependent down-regulation of CD36 (ie, to a CD36 low phenotype) in cultured human umbilical vein endothelial cells. Human umbilical vein endothelial cells that had been conditioned to a CD36 low phenotype with VEGF were found to be refractory to anti-proliferative TSP-1 signaling via a CD36-dependent mechanism. The loss of exposure to wall shear stress, which occurs in vivo when previously quiescent cells begin to sprout, also generated a CD36 low phenotype. Ultimately, our results identified the regulation of endothelial cell CD36 expression as a novel mechanism through which VEGF stimulates and sustains capillary sprouting in the presence of TSP-1. Additionally, CD36 was shown to function as a potential molecular linkage through which wall shear stress may regulate both microvessel sprouting and quiescence.


Assuntos
Antígenos CD36/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Transdução de Sinais , Trombospondina 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Humanos , Inflamação , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fenótipo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Estresse Mecânico , Trombospondina 1/farmacologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia
4.
J Muscle Res Cell Motil ; 30(1-2): 41-55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19205907

RESUMO

Lipoma preferred partner (LPP) localizes to focal adhesions/dense bodies, is selectively expressed in smooth muscle cells (SMC) and enhances cell migration. SMCs cultured on denatured collagen or on a rigid substrate, up regulated expression of LPP, its partner palladin, tenascin C (TN-C), phosphorylated focal adhesion kinase (pFAK) and exhibited robust stress fibers. In an endothelial (EC)/SMC hemodynamic flow system, shear stress waveforms mimicking atheroprone flow, applied to the EC layer, significantly decreased expression of SMC LPP and palladin. They were also down regulated with TN-C, in an ApoE murine model of atherosclerosis and with oxidative stress but up regulated in an arterial injury model in response to upstream sequential changes in pFAK, Prx1 and TN-C. In conclusion, expression of LPP and palladin are modulated by a mix of mechanical cues, oxidative stress and substrate composition which translate into their up or down regulation in vessel wall injury and early atherogenesis.


Assuntos
Aterosclerose/metabolismo , Proteínas do Citoesqueleto/biossíntese , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Fosfoproteínas/biossíntese , Resistência ao Cisalhamento , Animais , Aorta/lesões , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Adesão Celular , Movimento Celular , Colágeno/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Proteínas com Domínio LIM , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Fosfoproteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Fibras de Estresse/metabolismo , Tenascina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 28(8): 1534-41, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18556570

RESUMO

OBJECTIVE: The initiation of atherosclerosis is in part dependent on the hemodynamic shear stress environment promoting a proinflammatory phenotype of the endothelium. Previous studies demonstrated increased expression of ER stress protein and unfolded protein response (UPR) regulator, GRP78, within all vascular cells in atherosclerotic lesions and its regulation in the endothelium by several atherosclerotic stressors; however, regulation of GRP78 by shear stress directly has not been established. METHOD AND RESULTS: Using an in vitro model to simulate human arterial shear stress waveforms, atheroprone or atheroprotective flow was applied to human endothelial cells. GRP78 was found to be significantly upregulated (3-fold) in a sustained manner under atheroprone, but not atheroprotective flow up to 24 hours. This response was dependent on both sustained activation of p38, as well integrin alpha2beta1. Increased GRP78 correlated with the activation of the ER stress sensing element (ERSE1) promoter by atheroprone flow as a marker of the UPR. Shear stress regulated GRP78 through increased protein stability when compared to other flow regulated proteins, such as connexin-43 and vascular cell adhesion molecule (VCAM)-1. Increased endothelial expression of GRP78 was also observed in atheroprone versus atheroprotective regions of C57BL6 mice. CONCLUSIONS: This study supports a role of the hemodynamic environment in preferentially inducing GRP78 and the UPR in atheroprone regions, before lesion development, and suggests a potential atheroprotective (ie, prosurvival), compensatory effect in response to ER stress within atherosclerotic lesions.


Assuntos
Células Endoteliais/metabolismo , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Aorta Torácica/citologia , Aorta Torácica/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Chaperona BiP do Retículo Endoplasmático , Hemorreologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos Knockout , Transdução de Sinais , Veias Umbilicais/citologia , Veias Umbilicais/fisiologia , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
6.
Am J Physiol Heart Circ Physiol ; 296(2): H442-52, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19028801

RESUMO

Platelet-derived growth factor (PDGF)-BB is a well-known smooth muscle (SM) cell (SMC) phenotypic modulator that signals by binding to PDGF alphaalpha-, alphabeta-, and betabeta-membrane receptors. PDGF-DD is a recently identified PDGF family member, and its role in SMC phenotypic modulation is unknown. Here we demonstrate that PDGF-DD inhibited expression of multiple SMC genes, including SM alpha-actin and SM myosin heavy chain, and upregulated expression of the potent SMC differentiation repressor gene Kruppel-like factor-4 at the mRNA and protein levels. On the basis of the results of promoter-reporter assays, changes in SMC gene expression were mediated, at least in part, at the level of transcription. Attenuation of the SMC phenotypic modulatory activity of PDGF-DD by pharmacological inhibitors of ERK phosphorylation and by a small interfering RNA to Kruppel-like factor-4 highlight the role of these two pathways in this process. PDGF-DD failed to repress SM alpha-actin and SM myosin heavy chain in mouse SMCs lacking a functional PDGF beta-receptor. Importantly, PDGF-DD expression was increased in neointimal lesions in the aortic arch region of apolipoprotein C-deficient (ApoE(-/-)) mice. Furthermore, human endothelial cells exposed to an atherosclerosis-prone flow pattern, as in vascular regions susceptible to the development of atherosclerosis, exhibited a significant increase in PDGF-DD expression. These findings demonstrate a novel activity for PDGF-DD in SMC biology and highlight the potential contribution of this molecule to SMC phenotypic modulation in the setting of disturbed blood flow.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Linfocinas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Actinas/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/fisiopatologia , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes Reporter , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Linfocinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Fosforilação , Fator de Crescimento Derivado de Plaquetas/genética , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Multimerização Proteica , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/metabolismo , Fluxo Sanguíneo Regional , Estresse Mecânico , Fatores de Tempo , Regulação para Cima , Proteínas Elk-1 do Domínio ets/metabolismo , Calponinas
7.
Am J Physiol Cell Physiol ; 293(6): C1824-33, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913848

RESUMO

Atherosclerosis is an inflammatory disease that preferentially forms at hemodynamically compromised regions of altered shear stress patterns. Endothelial cells (EC) and smooth muscle cells (SMC) undergo phenotypic modulation during atherosclerosis. An in vitro coculture model was developed to determine the role of hemodynamic regulation of EC and SMC phenotypes in coculture. Human ECs and SMCs were plated on a synthetic elastic lamina and human-derived atheroprone, and atheroprotective shear stresses were imposed on ECs. Atheroprone flow decreased genes associated with differentiated ECs (endothelial nitric oxide synthase, Tie2, and Kruppel-like factor 2) and SMCs (smooth muscle alpha-actin and myocardin) and induced a proinflammatory phenotype in ECs and SMCs (VCAM-1, IL-8, and monocyte chemoattractant protein-1). Atheroprone flow-induced changes in SMC differentiation markers were regulated at the chromatin level, as indicated by decreased serum response factor (SRF) binding to the smooth muscle alpha-actin-CC(a/T)(6)GG (CArG) promoter region and decreased histone H(4) acetylation. Conversely, SRF and histone H(4) acetylation were enriched at the c-fos promoter in SMCs. In the presence of atheroprotective shear stresses, ECs aligned with the direction of flow and SMCs aligned more perpendicular to flow, similar to in vivo vessel organization. These results provide a novel mechanism whereby modulation of the EC phenotype by hemodynamic shear stresses, atheroprone or atheroprotective, play a critical role in mechanical-transcriptional coupling and regulation of the SMC phenotype.


Assuntos
Aterosclerose/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , Hemodinâmica/fisiologia , Miócitos de Músculo Liso/patologia , Artérias/fisiopatologia , Aterosclerose/fisiopatologia , Células Cultivadas , Técnicas de Cocultura , Endotélio Vascular/fisiopatologia , Epigênese Genética , Expressão Gênica , Humanos , Inflamação/fisiopatologia , Fenótipo , Proteínas/isolamento & purificação , RNA/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA