Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Biol Chem ; 394(4): 529-39, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23399636

RESUMO

Despite multiple advances in cancer therapies, patients with glioblastoma (GBM) still have a poor prognosis. Numerous glioma models are used not only for the development of innovative therapies but also to optimize conventional ones. Given the significance of hypoxia in drug and radiation resistance and that hypoxia is widely observed among GBM, the establishment of a reliable method to map hypoxia in preclinical human models may contribute to the discovery and translation of future and more targeted therapies. The aim of this study was to compare the hypoxic status of two commonly used human orthotopic glioma models (U87 and U251) developed in rats and studied by noninvasive hypoxia imaging with 3-[18F]fluoro-1-(2-nitro-1-imidazolyl)-2-propanol-micro-positron emission tomography ([18F]-FMISO-µPET). In parallel, because of the relationships between angiogenesis and hypoxia, we used magnetic resonance imaging (MRI), histology, and immunohistochemistry to characterize the tumoral vasculature. Although all tumors were detectable in T2-weighted MRI and 2-deoxy-2-[18F]fluoro-d-glucose-µPET, only the U251 model exhibited [18F]-FMISO uptake. Additionally, the U251 tumors were less densely vascularized than U87 tumors. Our study demonstrates the benefits of noninvasive imaging of hypoxia in preclinical models to define the most reliable one for translation of future therapies to clinic based on the importance of intratumoral oxygen tension for the efficacy of chemotherapy and radiotherapy.


Assuntos
Glioma/patologia , Hipóxia/diagnóstico , Misonidazol/análogos & derivados , Tomografia por Emissão de Pósitrons/métodos , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética
2.
Exp Cell Res ; 317(16): 2321-32, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21749867

RESUMO

Hypoxia has been shown to be one of the major events involved in EPO expression. Accordingly, EPO might be expressed by cerebral neoplastic cells, especially in glioblastoma, known to be highly hypoxic tumours. The expression of EPOR has been described in glioma cells. However, data from the literature remain descriptive and controversial. On the basis of an endogenous source of EPO in the brain, we have focused on a potential role of EPOR in brain tumour growth. In the present study, with complementary approaches to target EPO/EPOR signalling, we demonstrate the presence of a functional EPO/EPOR system on glioma cells leading to the activation of the ERK pathway. This EPO/EPOR system is involved in glioma cell proliferation in vitro. In vivo, we show that the down-regulation of EPOR expression on glioma cells reduces tumour growth and enhances animal survival. Our results support the hypothesis that EPOR signalling in tumour cells is involved in the control of glioma growth.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioma/tratamento farmacológico , Receptores da Eritropoetina/antagonistas & inibidores , Transdução de Sinais/fisiologia , Animais , Astrócitos/metabolismo , Neoplasias Encefálicas/metabolismo , Núcleo Caudado/patologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Eritropoetina/antagonistas & inibidores , Eritropoetina/genética , Eritropoetina/metabolismo , Eritropoetina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/genética , Glioma/metabolismo , Glioma/patologia , Células Hep G2 , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Endogâmicos F344 , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Deleção de Sequência/fisiologia , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cereb Cortex ; 21(7): 1695-702, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21127017

RESUMO

Vascular-specific growth factor angiopoietin-2 (Ang2) is mainly involved during vascular network setup. Recently, Ang2 was suggested to play a role in adult neurogenesis, affecting migration and differentiation of adult neuroblasts in vitro. However, to date, no data have reported an effect of Ang2 on neurogenesis during embryonic development. As we detected Ang2 expression in the developing cerebral cortex at embryonic day E14.5 and E16.5, we used in utero electroporation to knock down Ang2 expression in neuronal progenitors located in the cortical ventricular zone (VZ) to examine the role of Ang2 in cortical embryonic neurogenesis. Using this strategy, we showed that radial migration from the VZ toward the cortical plate of Ang2-knocked down neurons is altered as well as their morphology. In parallel, we observed a perturbation of intermediate progenitor population and the surrounding vasculature. Taken together, our results show for the first time that, in addition to its role during brain vasculature setup, Ang2 is also involved in embryonic cortical neurogenesis and especially in the radial migration of projection neurons.


Assuntos
Angiopoietina-2/fisiologia , Neurogênese/fisiologia , Telencéfalo/embriologia , Telencéfalo/crescimento & desenvolvimento , Angiopoietina-2/genética , Animais , Animais Recém-Nascidos , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Feminino , Técnicas de Silenciamento de Genes/métodos , Camundongos , Neurogênese/genética , Neurônios/citologia , Neurônios/fisiologia , Gravidez , Telencéfalo/irrigação sanguínea
4.
Clin Cancer Res ; 15(11): 3697-704, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19435839

RESUMO

PURPOSE: Epidermal growth factor receptor (EGFR) signal transduction pathways are implicated in malignant glioma aggressiveness and promote tumor cell invasion, proliferation, and angiogenesis. Nevertheless, response to EGFR tyrosine kinase inhibitor gefitinib (Iressa, ZD1839) has been disappointing in clinical trials. One potential explanation may come from the diversity of molecular alterations seen in gliomas. To validate that hypothesis, we have investigated responses to gefitinib on various tumor parameters in human malignant gliomas that exhibited different molecular alterations. EXPERIMENTAL DESIGN: We used a panel of six human malignant gliomas from established xenografts characterized for their genetic (EGFR, PTEN, TP53, and CDKN2A) and molecular (EGFR, PTEN, ERK, and Akt) alterations. Tumors were treated with gefitinib (1 or 10 micromol/L) for prolonged periods (8 or 16 days) in an organotypic brain slice model that allowed quantification of invasion, proliferation, and angiogenesis. RESULTS: In nontreated tumors, EGFR amplification was associated with profuse tumor cell invasion. After treatment, invasion was inhibited in tumors with EGFR amplification in a dose-dependent manner. Treatment had only antiproliferative effect in two of three tumors with EGFR amplification. Tumors with PTEN loss were resistant to treatment. We did not observe shrinkage of the tumors after treatment. None of the tumors had mutations of the EGFR kinase domain. Gefitinib had similar antiangiogenic effect in all of the tumors. CONCLUSIONS: Gefitinib reduces cell invasion in EGFR amplified tumors. PTEN loss of expression seems to be a determinant of resistance. Interestingly, inhibition of angiogenesis by gefitinib seems independent on the EGFR genetic status of the tumors.


Assuntos
Proliferação de Células , Receptores ErbB/metabolismo , Glioma/patologia , Neovascularização Patológica/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gefitinibe , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Nus , Mutação , Invasividade Neoplásica , Neovascularização Patológica/genética , Técnicas de Cultura de Órgãos , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Interferência de RNA , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Acta Biomater ; 15: 77-88, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25556361

RESUMO

Few effective therapeutic interventions are available to limit brain damage and functional deficits after ischaemic stroke. Within this context, mesenchymal stem cell (MSC) therapy carries minimal risks while remaining efficacious through the secretion of trophic, protective, neurogenic and angiogenic factors. The limited survival rate of MSCs restricts their beneficial effects. The usefulness of a three-dimensional support, such as a pharmacologically active microcarrier (PAM), on the survival of MSCs during hypoxia has been shown in vitro, especially when the PAMs were loaded with vascular endothelial growth factor (VEGF). In the present study, the effect of MSCs attached to laminin-PAMs (LM-PAMs), releasing VEGF or not, was evaluated in vivo in a model of transient stroke. The parameters assessed were infarct volume, functional recovery and endogenous cellular reactions. LM-PAMs induced the expression of neuronal markers by MSCs both in vitro and in vivo. Moreover, the prolonged release of VEGF increased angiogenesis around the site of implantation of the LM-PAMs and facilitated the migration of immature neurons towards the ischaemic tissue. Nonetheless, MSCs/LM-PAMs-VEGF failed to improve sensorimotor functions. The use of LM-PAMs to convey MSCs and to deliver growth factors could be an effective strategy to repair the brain damage caused by a stroke.


Assuntos
Isquemia Encefálica/complicações , Portadores de Fármacos/química , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Comportamento Animal , Vasos Sanguíneos/efeitos dos fármacos , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Laminina/farmacologia , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/fisiopatologia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
J Cereb Blood Flow Metab ; 33(3): 389-95, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23211963

RESUMO

Most forms of cerebral ischemia are characterized by damage to the entire neurovascular unit, which leads to an increase in the permeability of the blood-brain barrier (BBB). In response to permanent focal cerebral ischemia in mice, we detected an early concomitant increase in the expression of the vascular endothelial growth factor (VEGF), a key inducer of vascular leakage and pathological blood vessel growth, and of angiopoietin-2 (Ang2), which is closely associated with VEGF in vascular remodeling. Thus, the aim of this study was to evaluate the role of Ang2 alone, or in combination with VEGF, in the acute phase of cerebral ischemia. The effect of these angiogenic factors on the ischemic lesion volume was evaluated by magnetic resonance imaging. We observed that timely administration of VEGF exacerbates ischemic damage. In contrast, Ang2 decreases the ischemic volume and this beneficial effect is maintained in the presence of VEGF. This investigation reports, for the first time, a protective role of Ang2 following cerebral ischemia, an action associated with a reduced BBB permeability. We propose that Ang2 represents a pertinent molecular target for the treatment of cerebral ischemia since acute brain damage may be limited by a pharmacological protection of the vascular compartment.


Assuntos
Angiopoietina-2/biossíntese , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Animais , Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/terapia , Permeabilidade Capilar/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Angiografia por Ressonância Magnética , Camundongos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
7.
Nucl Med Biol ; 38(6): 781-93, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21843775

RESUMO

INTRODUCTION: No direct proof has been brought to light in a link between hypoxic changes in glioma models and the effects of antiangiogenic treatments. Here, we assessed the sensitivity of the detection of hypoxia through the use of (18)F-fluoromisonidazole positron emission tomography ([(18)F]-FMISO PET) in response to the evolution of the tumor and its vasculature. METHODS: Orthotopic glioma tumors were induced in rats after implantation of C6 or 9L cells. Sunitinib was administered from day (D) 17 to D24. At D17 and D24, multiparametric magnetic resonance imaging was performed to characterize tumor growth and vasculature. Hypoxia was assessed by [(18)F]-FMISO PET. RESULTS: We showed that brain hypoxic volumes are related to glioma volume and its vasculature and that an antiangiogenic treatment, leading to an increase in cerebral blood volume and a decrease in vessel permeability, is accompanied by a decrease in the degree of hypoxia. CONCLUSIONS: We propose that [(18)F]-FMISO PET and multiparametric magnetic resonance imaging are pertinent complementary tools in the evaluation of the effects of an antiangiogenic treatment in glioma.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamento farmacológico , Indóis/farmacologia , Imageamento por Ressonância Magnética , Misonidazol/análogos & derivados , Tomografia por Emissão de Pósitrons , Pirróis/farmacologia , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Transporte Biológico , Encéfalo/irrigação sanguínea , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Glioma/irrigação sanguínea , Glioma/diagnóstico , Glioma/tratamento farmacológico , Glioma/patologia , Indóis/uso terapêutico , Cinética , Misonidazol/metabolismo , Neovascularização Patológica/tratamento farmacológico , Permeabilidade/efeitos dos fármacos , Pirróis/uso terapêutico , Ratos , Sunitinibe , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA