Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 5(10): e13566, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21042414

RESUMO

5-Androstene-3ß,7ß,17ß-triol (ß-AET), an active metabolite of dehydroepiandrosterone (DHEA), reversed glucocorticoid (GC)-induced suppression of IL-6, IL-8 and osteoprotegerin production by human osteoblast-like MG-63 cells and promoted osteoblast differentiation of human mesenchymal stem cells (MSCs). In a murine thermal injury model that includes glucocorticoid-induced osteopenia, ß-AET significantly (p<0.05) preserved bone mineral content, restored whole body bone mineral content and endochondral growth, suggesting reversal of GC-mediated decreases in chondrocyte proliferation, maturation and osteogenesis in the growth plate. In men and women, levels of ß-AET decline with age, consistent with a role for ß-AET relevant to diseases associated with aging. ß-AET, related compounds or synthetic derivatives may be part of effective therapeutic strategies to accelerate tissue regeneration and prevent or treat diseases associated with aging such as osteoporosis.


Assuntos
Envelhecimento , Androstenóis/farmacologia , Doenças Ósseas Metabólicas/fisiopatologia , Queimaduras/fisiopatologia , Osteoporose/fisiopatologia , Absorciometria de Fóton , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Doenças Ósseas Metabólicas/etiologia , Queimaduras/complicações , Diferenciação Celular , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade
2.
Nat Biotechnol ; 25(8): 911-20, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17664940

RESUMO

Vascular endothelial growth factor (VEGF) is an essential regulator of normal and abnormal blood vessel growth. A monoclonal antibody (mAb) that targets VEGF suppresses tumor growth in murine cancer models and human patients. We investigated cellular and molecular events that mediate refractoriness of tumors to anti-angiogenic therapy. Inherent anti-VEGF refractoriness is associated with infiltration of the tumor tissue by CD11b+Gr1+ myeloid cells. Recruitment of these myeloid cells is also sufficient to confer refractoriness. Combining anti-VEGF treatment with a mAb that targets myeloid cells inhibits growth of refractory tumors more effectively than anti-VEGF alone. Gene expression analysis in CD11b+Gr1+ cells isolated from the bone marrow of mice bearing refractory tumors reveals higher expression of a distinct set of genes known to be implicated in active mobilization and recruitment of myeloid cells. These findings indicate that, in our models, refractoriness to anti-VEGF treatment is determined by the ability of tumors to prime and recruit CD11b+Gr1+ cells.


Assuntos
Antineoplásicos/administração & dosagem , Antígeno CD11b/metabolismo , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Receptores de Quimiocinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Camundongos , Camundongos Endogâmicos C57BL , Resultado do Tratamento
3.
Blood ; 107(2): 550-7, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16189273

RESUMO

Vascular endothelial growth factor-A (VEGF-A) and its 2 transmembrane tyrosine-kinase receptors, VEGFR-1 and VEGFR-2, constitute a ligand-receptor signaling system that is crucial for developmental angiogenesis. VEGF-B and placental growth factor (PlGF) activate VEGFR-1 selectively, however, mice lacking either ligand display only minor developmental defects. We hypothesized that the relative contributions of VEGF-B and PlGF to VEGFR-1 signaling may be masked in the presence of VEGF-A, which is abundantly expressed during postnatal development. To test this hypothesis, neonatal or adult mice were treated with a monoclonal antibody (G6-23-IgG) blocking murine VEGF-A or a soluble VEGFR-1 receptor IgG chimeric construct [mFlt(1-3)-IgG], which neutralizes VEGF-A, VEGF-B, and PlGF. Both compounds attenuated growth and survival of neonatal mice to similar extents and the pathophysiologic alterations, including a reduction in organ size and vascularization, changes in gene expression, and hematologic end points, were essentially indistinguishable. In adult mice, we observed only minor changes in response to treatment, which were similar between both anti-VEGF compounds. In conclusion, our findings suggest that PlGF and VEGF-B do not compensate during conditions of VEGF-A blockade, suggesting a minor role for compensatory VEGFR-1 signaling during postnatal development and vascular homeostasis in adults. The absence of compensatory VEGFR-1 signaling by VEGF-B and PlGF may have important implications for the development of anticancer strategies targeting the VEGF ligand/receptor system.


Assuntos
Neoplasias Experimentais/terapia , Neovascularização Patológica , Proteínas da Gravidez/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator B de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Mutantes , Camundongos Nus , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Fator de Crescimento Placentário , Proteínas da Gravidez/antagonistas & inibidores , Proteínas da Gravidez/imunologia , Taxa de Sobrevida , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator B de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator B de Crescimento do Endotélio Vascular/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
J Biol Chem ; 281(2): 951-61, 2006 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-16278208

RESUMO

To fully assess the role of VEGF-A in tumor angiogenesis, antibodies that can block all sources of vascular endothelial growth factor (VEGF) are desired. Selectively targeting tumor-derived VEGF overlooks the contribution of host stromal VEGF. Other strategies, such as targeting VEGF receptors directly or using receptor decoys, result in inhibiting not only VEGF-A but also VEGF homologues (e.g. placental growth factor, VEGF-B, and VEGF-C), which may play a role in angiogenesis. Here we report the identification of novel anti-VEGF antibodies, B20 and G6, from synthetic antibody phage libraries, which block both human and murine VEGF action in vitro. Their affinity-improved variants completely inhibit three human tumor xenografts in mice of skeletal muscle, colorectal, and pancreatic origins (A673, HM-7, and HPAC). Avastin, which only inhibits the tumor-derived human VEGF, is approximately 90% effective at inhibiting HM-7 and A673 growth but is <50% effective at inhibiting HPAC growth. Indeed, HPAC tumors contain more host stroma invasion and stroma-derived VEGF than other tumors. Thus, the functional contribution of stromal VEGF varies greatly among tumors, and systemic blockade of both tumor and stroma-derived VEGF is sufficient for inhibiting the growth of tumor xenografts.


Assuntos
Fator A de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/química , Cinética , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Transplante de Neoplasias , Neovascularização Patológica , Biblioteca de Peptídeos , Ligação Proteica , Especificidade da Espécie , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/imunologia
5.
J Gene Med ; 5(1): 11-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12516047

RESUMO

BACKGROUND: Hemophilia B is an X-linked bleeding disorder that results from a deficiency in functional coagulation factor IX (hFIX). In patients lacking FIX, the intrinsic coagulation pathway is disrupted leading to a lifelong, debilitating and sometimes fatal disease. METHODS: We have developed an ex vivo gene therapy system using genetically modified bone marrow stromal cells (BMSCs) as a platform for sustained delivery of therapeutic proteins into the general circulation. This model exploits the ability of BMSCs to form localized ectopic ossicles when transplanted in vivo. BMSCs were transduced with MFG-hFIX, a retroviral construct directing the expression of hFIX. The biological activity of hFIX expressed by these cells was assessed in vitro and in vivo. RESULTS: Transduced cells produced biologically active hFIX in vitro with a specific activity of 90% and expressed hFIX at levels of approximately 497 ng/10(6) cells/24 h and 322 ng/10(6) cells/24 h for human and porcine cells, respectively. The secretion of hFIX was confirmed by Western blot analysis of the conditioned medium using a hFIX-specific antibody. Transduced BMSCs (8 x 10(6) cells per animal) were transplanted within scaffolds into subcutaneous sites in immunocompromised mice. At 1 week post-implantation, serum samples contained hFIX at levels greater than 25 ng/ml. Circulating levels of hFIX gradually decreased to 11.5 ng/ml at 1 month post-implantation and declined to a stable level at 6.1 ng/ml at 4 months. CONCLUSIONS: These findings demonstrate that genetically modified BMSCs can continuously secrete biologically active hFIX from self-contained ectopic ossicles in vivo, and thus represent a novel delivery system for releasing therapeutic proteins into the circulation.


Assuntos
Células da Medula Óssea/metabolismo , Fator IX/genética , Terapia Genética , Células Estromais/metabolismo , Animais , Vetores Genéticos , Humanos , Retroviridae , Suínos , Transdução Genética
6.
Nature ; 417(6892): 954-8, 2002 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12087404

RESUMO

Vascular endothelial growth factor (VEGF) is a principal regulator of blood vessel formation and haematopoiesis, but the mechanisms by which VEGF differentially regulates these processes have been elusive. Here we describe a regulatory loop by which VEGF controls survival of haematopoietic stem cells (HSCs). We observed a reduction in survival, colony formation and in vivo repopulation rates of HSCs after ablation of the VEGF gene in mice. Intracellularly acting small-molecule inhibitors of VEGF receptor (VEGFR) tyrosine kinase dramatically reduced colony formation of HSCs, thus mimicking deletion of the VEGF gene. However, blocking VEGF by administering a soluble VEGFR-1, which acts extracellularly, induced only minor effects. These findings support the involvement in HSC survival of a VEGF-dependent internal autocrine loop mechanism (that is, the mechanism is resistant to inhibitors that fail to penetrate the intracellular compartment). Not only ligands selective for VEGF and VEGFR-2 but also VEGFR-1 agonists rescued survival and repopulation of VEGF-deficient HSCs, revealing a function for VEGFR-1 signalling during haematopoiesis.


Assuntos
Comunicação Autócrina , Fatores de Crescimento Endotelial/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Linfocinas/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Clonais/citologia , Células Clonais/efeitos dos fármacos , Células Clonais/metabolismo , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Citometria de Fluxo , Deleção de Genes , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Linfocinas/antagonistas & inibidores , Linfocinas/genética , Camundongos , Camundongos Knockout , Comunicação Parácrina , Proteínas Proto-Oncogênicas/agonistas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/farmacologia , Receptores de Fatores de Crescimento/agonistas , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Solubilidade , Transdução Genética , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA