Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Blood ; 122(24): 3982-92, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24108462

RESUMO

Endothelial cells (ECs) lining arteries and veins have distinct molecular/functional signatures. The underlying regulatory mechanisms are incompletely understood. Here, we established a specific fingerprint of freshly isolated arterial and venous ECs from human umbilical cord comprising 64 arterial and 12 venous genes, representing distinct functions/pathways. Among the arterial genes were 8 transcription factors (TFs), including Notch target HEY2, the current "gold standard" determinant for arterial EC (aEC) specification. Culture abrogated differential gene expression in part due to gradual loss of canonical Notch activity and HEY2 expression. Notably, restoring HEY2 expression or Delta-like4-induced Notch signaling in cultured ECs only partially reinstated the aEC gene signature, whereas combined overexpression of the 8 TFs restored this fingerprint more robustly. Whereas some TFs stimulated few genes, others boosted a large proportion of arterial genes. Although there was some overlap and cross-regulation, the TFs largely complemented each other in regulating the aEC gene profile. Finally, overexpression of the 8 TFs in human umbilical vein ECs conveyed an arterial-like behavior upon their implantation in a Matrigel plug in vivo. Thus, our study shows that Notch signaling determines only part of the aEC signature and identifies additional novel and complementary transcriptional players in the complex regulation of human arteriovenous EC identity.


Assuntos
Artérias/citologia , Células Endoteliais/metabolismo , Fatores de Transcrição/genética , Transcriptoma , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Análise por Conglomerados , Redes Reguladoras de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
2.
J Clin Invest ; 118(2): 505-14, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18172550

RESUMO

Despite progress in cardiovascular research, a cure for peripheral vascular disease has not been found. We compared the vascularization and tissue regeneration potential of murine and human undifferentiated multipotent adult progenitor cells (mMAPC-U and hMAPC-U), murine MAPC-derived vascular progenitors (mMAPC-VP), and unselected murine BM cells (mBMCs) in mice with moderate limb ischemia, reminiscent of intermittent claudication in human patients. mMAPC-U durably restored blood flow and muscle function and stimulated muscle regeneration, by direct and trophic contribution to vascular and skeletal muscle growth. This was in contrast to mBMCs and mMAPC-VP, which did not affect muscle regeneration and provided only limited and transient improvement. Moreover, mBMCs participated in a sustained inflammatory response in the lower limb, associated with progressive deterioration in muscle function. Importantly, mMAPC-U and hMAPC-U also remedied vascular and muscular deficiency in severe limb ischemia, representative of critical limb ischemia in humans. Thus, unlike BMCs or vascular-committed progenitors, undifferentiated multipotent adult progenitor cells offer the potential to durably repair ischemic damage in peripheral vascular disease patients.


Assuntos
Extremidades/irrigação sanguínea , Isquemia/terapia , Células-Tronco Multipotentes/transplante , Animais , Vasos Sanguíneos/citologia , Transplante de Medula Óssea , Diferenciação Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Células Musculares/citologia
3.
J Nucl Med ; 48(7): 1216-23, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17574988

RESUMO

UNLABELLED: Peripheral arterial occlusive disease (PAOD) is a leading cause of mortality and morbidity in the western world. The development of noninvasive methods for assessment and comparison of the efficacy of novel therapies in animal models is of great importance. METHODS: Hindlimb ischemia was induced in nude mice by ligation and excision of the left femoral artery (n = 5) or the left iliac artery (n = 10). Assessment of limb perfusion was performed by small-animal PET analysis after intravenous injection of (13)N-ammonia between 24 h and 30 d after surgery using the ratio of perfusion between the left limb (ischemic) and the right limb (control). Activity concentration per area unit was calculated in regions of interest placed on 1-mm-thick images for numeric calculations, and the iliac and the femoral models were compared. In addition, histopathologic studies were performed to assess the degree of necrosis (hematoxylin-eosin) and fibrosis (sirius red). Immunohistochemistry analyses for identification of arterioles (alpha-smooth muscle actin) and endothelium-capillaries-(Bandeiraea simplicifolia I [BS-I] lectin) were also performed. RESULTS: Perfusion in both hindlimbs of control animals was similar (median of the left-to-right ratio = 0.99). Twenty-four hours after ischemia, perfusion of the ischemic limb (% mean +/- SD) was 33.3 +/- 10.6 and 22.1 +/- 9.9 in the femoral and iliac models, respectively. Spontaneous recovery of perfusion in the hindlimb that underwent surgery was significantly lower in the iliac model at day +15 (73.2 +/- 15.5 vs. 51.9 +/- 11.3; P < 0.01). Fibrosis increased progressively until day +30, whereas muscle necrosis was maximal at day +7 with a moderate reduction by day +30. In accordance with this positive effect, there was a statistically significant increase in the area covered with smooth muscle-coated vessels (arterioles) at day +30 in comparison with day 7 (P < 0.05). In addition, a correlation between (13)N-ammonia uptake and the amount of necrosis (r = -0.73; P = 0.06) and fibrosis (r = -0.67; P = 0.05) at day +30 was found. CONCLUSION: (13)N-Ammonia imaging allows semiquantitative evaluation of hindlimb perfusion in surgical mouse models of acute hindlimb ischemia. Although spontaneous perfusion recovery is observed in both models, the iliac model shows a substantially lower recovery and is hence better suited for assessment of new therapeutic strategies for acute hindlimb ischemic disease.


Assuntos
Amônia , Arteriopatias Oclusivas/diagnóstico por imagem , Membro Posterior/irrigação sanguínea , Radioisótopos de Nitrogênio , Doenças Vasculares Periféricas/diagnóstico por imagem , Animais , Arteriopatias Oclusivas/patologia , Fibrose , Isquemia/diagnóstico por imagem , Isquemia/patologia , Masculino , Camundongos , Camundongos Nus , Necrose , Doenças Vasculares Periféricas/patologia , Tomografia por Emissão de Pósitrons/métodos , Fluxo Sanguíneo Regional
4.
Cell Transplant ; 20(2): 259-69, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20719064

RESUMO

There is a need for comparative studies to determine which cell types are better candidates to remedy ischemia. Here, we compared human AC133(+) cells and multipotent adult progenitor cells (hMAPC) in a mouse model reminiscent of critical limb ischemia. hMAPC or hAC133(+) cell transplantation induced a significant improvement in tissue perfusion (measured by microPET) 15 days posttransplantation compared to controls. This improvement persisted for 30 days in hMAPC-treated but not in hAC133(+)-injected animals. While transplantation of hAC133(+) cells promoted capillary growth, hMAPC transplantation also induced collateral expansion, decreased muscle necrosis/fibrosis, and improved muscle regeneration. Incorporation of differentiated hAC133(+) or hMAPC progeny into new vessels was limited; however, a paracrine angio/arteriogenic effect was demonstrated in animals treated with hMAPC. Accordingly, hMAPC-conditioned, but not hAC133(+)-conditioned, media stimulated vascular cell proliferation and prevented myoblast, endothelial, and smooth muscle cell apoptosis in vitro. Our study suggests that although hAC133(+) cell and hMAPC transplantation both contribute to vascular regeneration in ischemic limbs, hMAPC exert a more robust effect through trophic mechanisms, which translated into collateral and muscle fiber regeneration. This, in turn, conferred tissue protection and regeneration with longer term functional improvement.


Assuntos
Células-Tronco Adultas/citologia , Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Células-Tronco Multipotentes/citologia , Peptídeos/metabolismo , Transplante de Células-Tronco , Antígeno AC133 , Animais , Apoptose , Vasos Sanguíneos/crescimento & desenvolvimento , Proliferação de Células , Citoproteção , Membro Posterior/patologia , Humanos , Isquemia/patologia , Masculino , Camundongos , Camundongos Nus , Músculos/fisiopatologia , Neovascularização Fisiológica , Regeneração , Reperfusão , Sobrevivência de Tecidos
5.
Blood ; 109(6): 2634-42, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17090652

RESUMO

Many stem cell types have been shown to differentiate into endothelial cells (ECs); however, their specification to arterial or venous endothelium remains unexplored. We tested whether a specific arterial or venous EC fate could be induced in human multipotent adult progenitor cells (hMAPCs) and AC133(+) cells (hAC133(+)). In vitro, in the presence of VEGF(165), hAC133(+) cells only adopted a venous and microvascular EC phenotype, while hMAPCs differentiated into both arterial and venous ECs, possibly because hMAPCs expressed significantly more sonic hedgehog (Shh) and its receptors as well as Notch 1 and 3 receptors and some of their ligands. Accordingly, blocking either of those pathways attenuated in vitro arterial EC differentiation from hMAPCs. Complementarily, stimulating these pathways by addition of Delta-like 4 (Dll-4), a Notch ligand, and Shh to VEGF(165) further boosted arterial differentiation in hMAPCs both in vitro and in an in vivo Matrigel model. These results represent the first demonstration of adult stem cells with the potential to be differentiated into different types of ECs in vitro and in vivo and provide a useful human model to study arteriovenous specification.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular , Neovascularização Fisiológica , Antígeno AC133 , Células-Tronco Adultas/efeitos dos fármacos , Animais , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Separação Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Glicoproteínas/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Camundongos Nus , Microscopia Eletrônica , Fragmentos de Peptídeos/farmacologia , Peptídeos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA