Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 91(13-14): 716-22, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22365955

RESUMO

AIMS: Endothelin (ET) antagonists show promise in animal models of cirrhosis and portal hypertension. The aim was to pharmacologically characterise the expression of endothelin receptors in human liver, hepatic artery and portal vein. MAIN METHODS: Immunofluorescence staining, receptor autoradiography and competition binding assays were used to localise and quantify ET receptors on hepatic parenchyma, hepatic artery and portal vein in human cirrhotic or normal liver. Additional experiments were performed to determine the affinity and selectivity of ET antagonists for liver ET endothelin receptors. An endothelial cell ET(B) knockout murine model was used to examine the function of sinusoid endothelial ET(B) receptors. KEY FINDINGS: ET(B) receptors predominated in normal human liver and displayed the highest ratio (ET(B):ET(A) 63:47) compared with other peripheral tissues. In two patients examined, liver ET(B) expression was up-regulated in cirrhosis (ET(B):ET(A) 83:17). Both sub-types localised to the media of normal portal vein but ET(B) receptors were downregulated fivefold in the media of cirrhotic portal vein. Sinusoid diameter was fourfold smaller in endothelial cell ET(B) knockout mice. The liver morphology of ET(B) knockout mice was markedly different to normal murine liver, with loss of the wide spread sinusoidal pattern. In the knockout mice, sinusoids were reduced in both number and absolute diameter, while large intrahepatic veins were congested with red blood cells. SIGNIFICANCE: These data support a role for the ET system in cirrhosis of the liver and suggest that endothelial ET(B) blockade may cause sinusoidal constriction which may contribute to hepatotoxicity associated with some endothelin antagonists.


Assuntos
Cirrose Hepática/patologia , Fígado/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina B/genética , Animais , Autorradiografia , Ligação Competitiva , Artéria Hepática/metabolismo , Humanos , Isoxazóis/farmacologia , Fígado/patologia , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Fenilpropionatos/farmacologia , Veia Porta/metabolismo , Veia Porta/patologia , Piridazinas/farmacologia , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Tiofenos/farmacologia , Regulação para Cima
2.
Am J Pathol ; 168(6): 1793-807, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16723696

RESUMO

The precise cellular and molecular mechanisms regulating adventitial vasa vasorum neovascularization, which occurs in the pulmonary arterial circulation in response to hypoxia, remain unknown. Here, using a technique to isolate and culture adventitial fibroblasts (AdvFBs) and vasa vasorum endothelial cells (VVECs) from the adventitia of pulmonary arteries, we report that hypoxia-activated pulmonary artery AdvFBs exhibited pro-angiogenic properties and influenced the angiogenic phenotype of VVEC, in a process of cell-cell communication involving endothelin-1 (ET-1). We demonstrated that AdvFBs, either via co-culture or conditioned media, stimulated VVEC proliferation and augmented the self-assembly and integrity of cord-like networks that formed when VVECs where cultured on Matrigel. In addition, hypoxia-activated AdvFBs produced ET-1, suggesting a paracrine role for this pro-angiogenic molecule in these processes. When co-cultured on Matrigel, AdvFBs and VVECs self-assembled into heterotypic cord-like networks, a process augmented by hypoxia but attenuated by either selective endothelin receptor antagonists or oligonucleotides targeting prepro-ET-1 mRNA. From these observations, we propose that hypoxia-activated AdvFBs exhibit pro-angiogenic properties and, as such, communicate with VVECs, in a process involving ET-1, to regulate vasa vasorum neovascularization occurring in the adventitia of pulmonary arteries in response to chronic hypoxia.


Assuntos
Endotelina-1/metabolismo , Endotélio Vascular/metabolismo , Fibroblastos/patologia , Hipóxia , Artéria Pulmonar/metabolismo , Animais , Bovinos , Técnicas de Cultura de Células/métodos , Proliferação de Células , Células Cultivadas/metabolismo , Modelos Animais de Doenças , Hipertensão/patologia , Microscopia de Fluorescência
3.
Am J Pathol ; 168(2): 659-69, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16436679

RESUMO

Vascular remodeling in chronic hypoxic pulmonary hypertension includes marked fibroproliferative changes in the pulmonary artery (PA) adventitia. Although resident PA fibroblasts have long been considered the primary contributors to these processes, we tested the hypothesis that hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage, termed fibrocytes. Using two neonatal animal models (rats and calves) of chronic hypoxic pulmonary hypertension, we demonstrated a dramatic perivascular accumulation of mononuclear cells of a monocyte/macrophage lineage (expressing CD45, CD11b, CD14, CD68, ED1, ED2). Many of these cells produced type I collagen, expressed alpha-smooth muscle actin, and proliferated, thus exhibiting mesenchymal cell characteristics attributed to fibrocytes. The blood-borne origin of these cells was confirmed in experiments wherein circulating monocytes/macrophages of chronically hypoxic rats were in vivo-labeled with DiI fluorochrome via liposome delivery and subsequently identified in the remodeled pulmonary, but not systemic, arterial adventitia. The DiI-labeled cells that appeared in the vessel wall expressed monocyte/macrophage markers and procollagen. Selective depletion of this monocytic cell population, using either clodronate-liposomes or gadolinium chloride, prevented pulmonary adventitial remodeling (ie, production of collagen, fibronectin, and tenascin-C and accumulation of myofibroblasts). We conclude that circulating mesenchymal precursors of a monocyte/macrophage lineage, including fibrocytes, are essential contributors to hypoxia-induced pulmonary vascular remodeling.


Assuntos
Hipertensão Pulmonar/metabolismo , Hipóxia/metabolismo , Macrófagos/fisiologia , Monócitos/fisiologia , Circulação Pulmonar/fisiologia , Células-Tronco/fisiologia , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Bovinos , Linhagem da Célula , Proliferação de Células , Colágeno Tipo I/metabolismo , Fibroblastos/patologia , Fibroblastos/fisiologia , Fibronectinas/metabolismo , Hipertensão Pulmonar/patologia , Lipossomos , Masculino , Músculo Liso Vascular/metabolismo , Artéria Pulmonar/patologia , Ratos , Ratos Endogâmicos WKY , Tenascina/metabolismo
6.
J Appl Physiol (1985) ; 98(2): 715-21, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15649883

RESUMO

Data are rapidly accumulating in support of the idea that circulating monocytes and/or mononuclear fibrocytes are recruited to the pulmonary circulation of chronically hypoxic animals and that these cells play an important role in the pulmonary hypertensive process. Hypoxic induction of monocyte chemoattractant protein-1, stromal cell-derived factor-1, vascular endothelial growth factor-A, endothelin-1, and tumor growth factor-beta(1) in pulmonary vessel wall cells, either directly or indirectly via signals from hypoxic lung epithelial cells, may be a critical first step in the recruitment of circulating leukocytes to the pulmonary circulation. In addition, hypoxic stress appears to induce release of increased numbers of monocytic progenitor cells from the bone marrow, and these cells may have upregulated expression of receptors for the chemokines produced by the lung circulation, which thus facilitates their specific recruitment to the pulmonary site. Once present, macrophages/fibrocytes may exert paracrine effects on resident pulmonary vessel wall cells stimulating proliferation, phenotypic modulation, and migration of resident fibroblasts and smooth muscle cells. They may also contribute directly to the remodeling process through increased production of collagen and/or differentiation into myofibroblasts. In addition, they could play a critical role in initiating and/or supporting neovascularization of the pulmonary artery vasa vasorum. The expanded vasa network may then act as a conduit for further delivery of circulating mononuclear cells to the pulmonary arterial wall, creating a feedforward loop of pathological remodeling. Future studies will need to determine the mechanisms that selectively induce leukocyte/fibrocyte recruitment to the lung circulation under hypoxic conditions, their direct role in the remodeling process via production of extracellular matrix and/or differentiation into myofibroblasts, their impact on the phenotype of resident smooth muscle cells and adventitial fibroblasts, and their role in the neovascularization observed in hypoxic pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/imunologia , Hipóxia/imunologia , Leucócitos/imunologia , Pulmão/imunologia , Ativação Linfocitária/imunologia , Circulação Pulmonar/imunologia , Animais , Humanos , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Modelos Imunológicos
7.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L668-78, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12754186

RESUMO

Information is rapidly emerging regarding the important role of the arterial vasa vasorum in a variety of systemic vascular diseases. In addition, increasing evidence suggests that progenitor cells of bone marrow (BM) origin may contribute to postnatal neovascularization and/or vascular wall thickening that is characteristic in some forms of systemic vascular disease. Little is known regarding postnatal vasa formation and the role of BM-derived progenitor cells in the setting of pulmonary hypertension (PH). We sought to determine the effects of chronic hypoxia on the density of vasa vasorum in the pulmonary artery and to evaluate if BM-derived progenitor cells contribute to the increased vessel wall mass in a bovine model of hypoxia-induced PH. Quantitative morphometric analyses of lung tissue from normoxic and hypoxic calves revealed that hypoxia results in a dramatic expansion of the pulmonary artery adventitial vasa vasorum. Flow cytometric analysis demonstrated that cells expressing the transmembrane tyrosine kinase receptor for stem cell factor, c-kit, are mobilized from the BM in the circulation in response to hypoxia. Immunohistochemistry revealed an increase in the expression of c-kit+ cells together with vascular endothelial growth factor, fibronectin, and thrombin in the hypoxia-induced remodeled pulmonary artery vessel wall. Circulating mononuclear cells isolated from neonatal calves exposed to hypoxia were found to differentiate into endothelial and smooth muscle cell phenotypes depending on culture conditions. From these observations, we suggest that the vasa vasorum and circulating progenitor cells could be involved in vessel wall thickening in the setting of hypoxia-induced PH.


Assuntos
Hipóxia/patologia , Neovascularização Patológica/patologia , Artéria Pulmonar/patologia , Células-Tronco/patologia , Animais , Animais Recém-Nascidos , Bovinos , Diferenciação Celular , Células Cultivadas , Eritrócitos , Hipertensão Pulmonar/patologia , Masculino , Proteínas Proto-Oncogênicas c-kit/análise , Artéria Pulmonar/química , Vasa Vasorum/química , Vasa Vasorum/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...