Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33653955

RESUMO

Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood-brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret ), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti-ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret ;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.


Assuntos
Barreira Hematoencefálica/imunologia , Encefalomielite Autoimune Experimental/imunologia , Homeostase/imunologia , Leucócitos/imunologia , Pericitos/imunologia , Animais , Barreira Hematoencefálica/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Homeostase/genética , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos/patologia , Camundongos , Camundongos Transgênicos , Pericitos/patologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
2.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32586981

RESUMO

Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor ß-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.


Assuntos
Movimento Celular , Endotélio Vascular/metabolismo , Pericitos/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Animais , Vasos Sanguíneos , Neoplasias do Colo/irrigação sanguínea , Transição Epitelial-Mesenquimal , Matriz Extracelular , Técnicas de Inativação de Genes , Hibridização Genética , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Melanoma/irrigação sanguínea , Melanoma/secundário , Camundongos , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Pericitos/metabolismo , Ativação Plaquetária/fisiologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Trombocitopenia , Hipóxia Tumoral , Microambiente Tumoral
3.
Biotechnol Bioeng ; 112(9): 1916-26, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25786390

RESUMO

With a view toward reduction of graft loss, we explored pancreatic islet transplantation within fibrin matrices rendered pro-angiogenic by incorporation of minimal doses of vascular endothelial growth factor-A165 and platelet-derived growth factor-BB presented complexed to a fibrin-bound integrin-binding fibronectin domain. Engineered matrices allowed for extended release of pro-angiogenic factors and for their synergistic signaling with extracellular matrix-binding domains in the post-transplant period. Aprotinin addition delayed matrix degradation and prolonged pro-angiogenic factor availability within the graft. Both subcutaneous (SC) and epididymal fat pad (EFP) sites were evaluated. We show that in the SC site, diabetes reversal in mice transplanted with 1,000 IEQ of syngeneic islets was not observed for islets transplanted alone, while engineered matrices resulted in a diabetes median reversal time (MDRT) of 38 days. In the EFP site, the MDRT with 250 IEQ of syngeneic islets within the engineered matrices was 24 days versus 86 days for islets transplanted alone. Improved function of engineered grafts was associated with enhanced and earlier (by day 7) angiogenesis. Our findings show that by engineering the transplant site to promote prompt re-vascularization, engraftment and long-term function of islet grafts can be improved in relevant extrahepatic sites.


Assuntos
Fibrina/química , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Becaplermina , Proliferação de Células/efeitos dos fármacos , Humanos , Hidrogéis/química , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-sis/química , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/farmacologia , Fator A de Crescimento do Endotélio Vascular/química
4.
Cardiovasc Res ; 71(3): 557-65, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16831408

RESUMO

OBJECTIVE: Extracellular retention of PDGF-B has been proposed to play an important role in PDGF-B signalling. We used the PDGF-B retention motif knockout mouse (RetKO) to study the effects of retention motif deletion on development of micro- and macrovascular structure and function. METHODS: Passive and active properties of conduit vessels were studied using myograph techniques and histological examination. Capillary structure and function was studied using measurements of capillary density in skeletal muscle and by assessing aerobic physical performance in a treadmill setup. Cardiac function was assessed using echocardiography. RESULTS: Myograph experiments revealed an increased diameter and stiffness of the aorta in RetKO. Histological examination showed increased media collagen content and a decreased number of aortic wall layers, however with a similar number of vascular smooth muscle cells. This outward eutrophic remodelling of the aorta was accompanied by endothelial dysfunction. RetKO showed decreased capillary density in skeletal muscle and signs of a defective delivery of capillary oxygen to skeletal muscle, as shown by a decreased physical performance. In RetKO mice, echocardiography revealed an adaptive eccentric cardiac hypertrophy. CONCLUSION: We conclude that retention of PDGF-B during development is essential for a normal conduit vessel function in the adult mouse. Furthermore, PDGF-B retention is also necessary for the development of an adequate capillary density, and thereby for a normal oxygen delivery to skeletal muscle. The lack of primary effects on cardiac function supports the redundant role of PDGF-B in cardiac development.


Assuntos
Aorta Torácica/fisiopatologia , Músculo Esquelético/irrigação sanguínea , Proteínas Proto-Oncogênicas c-sis/fisiologia , Animais , Aorta Torácica/patologia , Pressão Sanguínea , Capilares/patologia , Capilares/fisiopatologia , Cardiomegalia/diagnóstico , Cardiomegalia/genética , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Miografia , Consumo de Oxigênio , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Renina/sangue , Túnica Média/metabolismo
5.
FASEB J ; 20(10): 1703-5, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16807374

RESUMO

Normal blood microvessels are lined by pericytes, which contribute to microvessel development and stability through mechanisms that are poorly understood. Pericyte deficiency has been implicated in the pathogenesis of microvascular abnormalities associated with diabetes and tumors. However, the unambiguous identification of pericytes is still a problem because of cellular heterogeneity and few available molecular markers. Here we describe an approach to identify pericyte markers based on transcription profiling of pericyte-deficient brain microvessels isolated from platelet-derived growth factor (PDGF-B)-/- and PDGF beta receptor (PDGFRbeta)-/- mouse mutants. The approach was validated by the identification of known pericyte markers among the most down-regulated genes in PDGF-B-/- and PDGFRbeta-/- microvessels. Of candidates for novel pericyte markers, we selected ATP-sensitive potassium-channel Kir6.1 (also known as Kcnj8) and sulfonylurea receptor 2, (SUR2, also known as Abcc9), both part of the same channel complex, as well as delta homologue 1 (DLK1) for in situ hybridization, which demonstrated their specific expression in brain pericytes of mouse embryos. We also show that Kir6.1 is highly expressed in pericytes in brain but undetectable in pericytes in skin and heart. The three new brain pericyte markers are signaling molecules implicated in ion transport and intercellular signaling, potentially opening new windows on pericyte function in brain microvessels.


Assuntos
Encéfalo/citologia , Capilares/citologia , Análise em Microsséries/métodos , Pericitos/química , Proteínas Proto-Oncogênicas c-sis/deficiência , Receptor beta de Fator de Crescimento Derivado de Plaquetas/deficiência , Transportadores de Cassetes de Ligação de ATP/análise , Animais , Biomarcadores/análise , Encéfalo/irrigação sanguínea , Proteínas de Ligação ao Cálcio , Embrião de Mamíferos , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , Canais KATP , Proteínas de Membrana/análise , Camundongos , Camundongos Mutantes , Canais de Potássio Corretores do Fluxo de Internalização/análise , Receptores de Droga , Proteínas Repressoras/análise , Receptores de Sulfonilureias , Distribuição Tecidual
6.
Am J Pathol ; 167(3): 901-12, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127167

RESUMO

Both innate and adaptive immunity contribute to the progression of inflammatory-fibrotic lesions of atherosclerosis. Although platelet-derived growth factor (PDGF)-B has been investigated as a stimulant of smooth muscle cells in vascular diseases, its effects on the immune response during disease have not been evaluated in vivo. We used hematopoietic chimeras generated after lethal irradiation of ApoE-/- recipients to test the role of PDGF in atherosclerosis. Monocyte accumulation in early atherosclerotic lesions increased 1.9-fold in ApoE-/-/PDGF-B-/- chimeras. Lymphocytes from null chimeras showed a 1.6- to 2.0-fold increase in the number of activated CD4(+) T cells and a 2.5-fold elevation of interferon-gamma-secreting CD4(+) T cells on ex vivo challenge with modified low-density lipoprotein. Splenocyte transcript levels were also altered with a twofold decrease in interleukin-10 and 1.7- and 3.0-fold increases in interleukin-18 and CCR 5, respectively. These cellular and molecular changes were consistent with a shift to a proinflammatory phenotype in null chimeras. Our data also demonstrated for the first time the presence of a recently discovered family of negative regulators of innate and adaptive immunity, the suppressors of cytokine signaling (SOCS), in developing atherosclerotic lesions. Thus, our studies identify two independent negative immune regulatory pathways-PDGF-B and SOCS-that may help limit lesion expansion.


Assuntos
Apolipoproteínas E/deficiência , Arteriosclerose/complicações , Arteriosclerose/imunologia , Células Sanguíneas/metabolismo , Inflamação/etiologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Animais , Apolipoproteínas E/genética , Arteriosclerose/genética , Arteriosclerose/patologia , Células da Medula Óssea/metabolismo , Proteínas de Transporte/metabolismo , Quimera , Perfilação da Expressão Gênica , Predisposição Genética para Doença , Ligantes , Ativação Linfocitária , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Proteínas Repressoras/metabolismo , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Linfócitos T/imunologia , Fatores de Transcrição/metabolismo
7.
J Cell Biochem ; 95(4): 859-67, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15838875

RESUMO

Platelet-derived growth factor-B (PDGF-B) and its receptors play essential roles in the complex process of blood vessel maturation and they therefore constitute promising targets for therapeutic strategies against blood vessel-related diseases. Additionally, they are involved in the autocrine stimulation of tumor cells and have been suggested to regulate tumor stroma fibroblasts. Our study aimed to identify genes that are regulated directly by PDGF-B, or indirectly via the recruitment of perivascular cells, in the context of an intact tissue. We used a subtractive cloning technique to compare gene transcription in the brains of wild-type (WT) mice and syngenic mice deficient of PDGF-B leading to a defect in the recruitment of perivascular cells. The resulting 147 differentially expressed sequences contained early and late PDGF-B target genes, and genes implicated in blood vessel maturation-related pathways. Additionally, gene clusters for specific biological processes such as cell migration and intracellular transport were identified. Of eight randomly selected sequences, six were found expressed in cultured cells of mesenchymal origin, two of them inducible by exogenous PDGF-BB. The collection of cDNA presented here provides insights into the changes provoked by the removal of one growth factor of a complete tissue and might be the basis for the identification of novel players in the complex process of blood vessel maturation.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-sis/metabolismo , Transcrição Gênica , Animais , Encéfalo/irrigação sanguínea , Clonagem Molecular , DNA Complementar/genética , Perfilação da Expressão Gênica , Camundongos , Camundongos Knockout , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-sis/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Ativação Transcricional
8.
Biochem Biophys Res Commun ; 321(2): 413-23, 2004 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-15358192

RESUMO

Hepatic stellate cells (HSCs) and transdifferentiated myofibroblasts are the principal producers of excessive extracellular matrix in liver fibrosis and cirrhosis. Activation of HSC is regulated by several cytokines and growth factors, including platelet-derived growth factor B-chain (PDGF-B), a potent mitogen for HSC, and overexpressed during hepatic fibrogenesis. Previous studies showed that MAPK and phosphatidylinositol 3' kinase are key signaling pathways involved in PDGF-induced stimulation of HSC. Based on the involvement of PDGF-B in fibrogenesis, reducing ligand stimulation of proliferative cytokine- or growth factor receptors interfering with receptor signaling therefore presents an interesting strategy for hepatic fibrosis prevention or interruption. We therefore generated an adenoviral vector serotype 5 (Ad5) expressing an antisense mRNA of the PDGF B-chain (Ad5-CMV-asPDGF) for application in an experimentally induced liver fibrogenesis model. The transgene clearly showed the ability to down-regulate endogenous PDGF B-chain and PDGFRbeta mRNA in culture-activated HSC and rat livers. The asPDGF mRNA also attenuates experimental liver fibrogenesis indicated by reduced levels of alpha-SMA and collagen type I expression.


Assuntos
DNA Antissenso/genética , Cirrose Hepática/genética , Cirrose Hepática/prevenção & controle , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Actinas/genética , Adenoviridae/genética , Animais , Células Cultivadas , Colágeno/genética , Modelos Animais de Doenças , Regulação para Baixo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Masculino , Proteínas Proto-Oncogênicas c-sis/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
9.
EMBO J ; 21(16): 4307-16, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12169633

RESUMO

Loss of pericytes from the capillary wall is a hallmark of diabetic retinopathy, however, the pathogenic significance of this phenomenon is unclear. In previous mouse gene knockout models leading to pericyte deficiency, prenatal lethality has so far precluded analysis of postnatal consequences in the retina. We now report that endothelium-restricted ablation of platelet-derived growth factor-B generates viable mice with extensive inter- and intra-individual variation in the density of pericytes throughout the CNS. We found a strong inverse correlation between pericyte density and the formation of a range of retinal microvascular abnormalities strongly reminiscent of those seen in diabetic humans. Proliferative retinopathy invariably developed when pericyte density was <50% of normal. Our data suggest that a reduction of the pericyte density is sufficient to cause retinopathy in mice, implying that pericyte loss may also be a causal pathogenic event in human diabetic retinopathy.


Assuntos
Retinopatia Diabética/patologia , Pericitos/patologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Capilares/patologia , Retinopatia Diabética/etiologia , Retinopatia Diabética/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Marcação de Genes , Camundongos , Camundongos Transgênicos , Pericitos/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , Retina/patologia , Vasos Retinianos/patologia
10.
Am J Pathol ; 159(5): 1869-76, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11696447

RESUMO

The hypothesis that wound repair is augmented by delivery of platelet-derived growth factor (PDGF) from platelets and macrophages is an attractive extrapolation from the known activities of PDGF in cell culture and in vivo. To test this hypothesis in mice, we prepared hematopoietic chimeras, in which the hematopoietic system of a normal adult mouse was replaced by the hematopoietic system of a PDGF B-chain -/- or +/+ donor. We initiated local granulation tissue formation either by implanting small surgical sponges to elicit a foreign body granulation tissue response, or by ligating the left common carotid to form an organized thrombus. We found that the absence of hematopoietic PDGF B-chain did not decrease the extent of granulation tissue or vascular lesion formation, and that the vascularization of both lesions increased by approximately 100%. We conclude that PDGF B-chain from cells of hematopoietic origin, including platelets and macrophages, is not important for granulation tissue formation, and that it reduces vascularization of granulation issue, probably through disabling of the short-range chemotactic gradients of PDGF that are important for recruiting pericytes/smooth muscle cells to the endothelium of new vessels.


Assuntos
Tecido de Granulação/fisiopatologia , Hematopoese/fisiologia , Neovascularização Fisiológica/fisiologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/fisiologia , Animais , Artérias , Quimera , Reação a Corpo Estranho/fisiopatologia , Tecido de Granulação/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/genética , Trombose/etiologia
11.
Blood ; 97(7): 1990-8, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11264163

RESUMO

Platelet-derived growth factor (PDGF)-B and PDGF beta-receptor (PDGFR beta) deficiency in mice is embryonic lethal and results in cardiovascular, renal, placental, and hematologic disorders. The hematologic disorders are described, and a correlation with hepatic hypocellularity is demonstrated. To explore possible causes, the colony-forming activity of fetal liver cells in vitro was assessed, and hematopoietic chimeras were demonstrated by the transplantation of mutant fetal liver cells into lethally irradiated recipients. It was found that mutant colony formation is equivalent to that of wild-type controls. Hematopoietic chimeras reconstituted with PDGF-B(-/-), PDGFR beta(-/-), or wild-type fetal liver cells show complete engraftment (greater than 98%) with donor granulocytes, monocytes, B cells, and T cells and display none of the cardiovascular or hematologic abnormalities seen in mutants. In mouse embryos, PDGF-B is expressed by vascular endothelial cells and megakaryocytes. After birth, expression is seen in macrophages and neurons. This study demonstrates that hematopoietic PDGF-B or PDGFR beta expression is not required for hematopoiesis or integrity of the cardiovascular system. It is argued that metabolic stress arising from mutant defects in the placenta, heart, or blood vessels may lead to impaired liver growth and decreased production of blood cells. The chimera models in this study will serve as valuable tools to test the role of PDGF in inflammatory and immune responses. (Blood. 2001;97:1990-1998)


Assuntos
Vasos Sanguíneos/anormalidades , Doenças Fetais/genética , Hematopoese/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Anemia/embriologia , Anemia/genética , Anemia/metabolismo , Animais , Vasos Sanguíneos/embriologia , Transplante de Medula Óssea , Desenvolvimento Embrionário e Fetal/genética , Eritroblastose Fetal/genética , Eritroblastose Fetal/metabolismo , Feminino , Doenças Fetais/sangue , Doenças Fetais/patologia , Coração Fetal/anormalidades , Transplante de Tecido Fetal , Genes Letais , Teste de Complementação Genética , Genótipo , Transplante de Células-Tronco Hematopoéticas , Inflamação , Rim/anormalidades , Rim/embriologia , Fígado/citologia , Fígado/embriologia , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Placenta/fisiopatologia , Gravidez , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Quimera por Radiação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/deficiência , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Organismos Livres de Patógenos Específicos , Estresse Fisiológico/embriologia , Estresse Fisiológico/genética , Estresse Fisiológico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...