Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 38(10): 1769-1780, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28617154

RESUMO

Although chronic arterial hypertension (CAH) represents the major comorbid factor in stroke, it is rarely integrated in preclinical studies of stroke. The majority of those investigations employ spontaneously hypertensive rats (SHR) which display a susceptibility to ischemic damage independent of hypertension. Here, we used a renovascular model of hypertension (RH) to examine, with magnetic resonance imaging (MRI), brain alterations during the development of hypertension and after brain ischemia. We also examined whether MRI-derived parameters predict the extent of ischemia-induced brain damage. RH was induced according to the two-kidney one-clip model and multiparametric MRI was performed at 3, 6, 9, and 12 weeks after hypertension and also at 10, 50, and 60 min following stroke. Blood pressure values increased progressively and reached a plateau at 6 weeks after RH induction. At 12 weeks, all hypertensive animals displayed spontaneous brain lesions (hemorrhages, deep and cortical lesions, ventricular dilatation), increased apparent diffusion coefficient (ADC) values in the corpus callosum and higher fractional anisotropy in the cortex. Following ischemia, these animals showed larger brain lesions (406 ± 82 vs. 179 ± 36 mm3, p < 0.002) which correlated with ADC values at chronic stage of hypertension. This model of hypertension displays many characteristics of the neuropathology of human CAH. The use of this model in stroke studies is relevant and desirable.


Assuntos
Encéfalo/patologia , Modelos Animais de Doenças , Hipertensão Renovascular/patologia , Acidente Vascular Cerebral/patologia , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Hipertensão Renovascular/complicações , Masculino , Ratos Sprague-Dawley , Acidente Vascular Cerebral/complicações
2.
Hypertens Res ; 38(11): 723-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26084262

RESUMO

Glioblastoma is the most aggressive brain tumor and is almost always fatal. These tumors are highly vascularized and angiogenesis is one of the pre-eminent mechanisms underlying their growth. Chronic arterial hypertension (CAH) is a common and worldwide pathology that markedlly alters the structure and function of the vasculature. Yet, essential hypertension is associated in the brain with potential locally impaired vasoreactivity, disturbed perfusion supply and hypoxia phenomena. Even though CAH is a global burden and has an important impact on brain function, nothing is known about the way this frequent pathology would interact with the evolution of glioma. We sought to determine if arterial hypertension influences gliobastoma growth. In the present study, rat glioma C6 tumor cells were implanted in the caudate-putamen of spontaneously hypertensive rats (SHR) or their normotensive controls, the Wistar-Kyoto (WKY) rats. The evolution of the tumor was sequentially analyzed by multiparametric magnetic resonance imaging and the inflammatory response was examined by histochemistry. We found that CAH significantly attenuates the growth of the tumor as, at 21 days, the volume of the tumor was 85.4±34.7 and 126.1±28.8 mm(3), respectively, in hypertensive and normotensive rats (P<0.02). Moreover, cerebral blood volume and cerebral blood flow were greater in the tumors of hypertensive rats (P<0.05). The lesser growth of the tumor observed in normotensive animals was not due to an enhanced rejection of the tumor cells in WKY rats, the inflammatory response being similar in both groups. For the first time, these results show that CAH impedes the growth of glioblastoma and illustrate the need to further study the impact of hypertension on the evolution of brain tumors.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/patologia , Glioblastoma/patologia , Hipertensão/complicações , Animais , Pressão Arterial , Peso Corporal , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/etiologia , Glioblastoma/irrigação sanguínea , Glioblastoma/etiologia , Imageamento por Ressonância Magnética , Masculino , Distribuição Aleatória , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
3.
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
4.
Oncotarget ; 6(4): 2101-19, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25544764

RESUMO

Hypoxia-inducible genes may contribute to therapy resistance in glioblastoma (GBM), the most aggressive and hypoxic brain tumours. It has been recently reported that erythropoietin (EPO) and its receptor (EPOR) are involved in glioma growth. We now investigated whether EPOR signalling may modulate the efficacy of the GBM current treatment based on chemotherapy (temozolomide, TMZ) and radiotherapy (X-rays). Using RNA interference, we showed on glioma cell lines (U87 and U251) that EPOR silencing induces a G2/M cell cycle arrest, consistent with the slowdown of glioma growth induced by EPOR knock-down. In vivo, we also reported that EPOR silencing combined with TMZ treatment is more efficient to delay tumour recurrence and to prolong animal survival compared to TMZ alone. In vitro, we showed that EPOR silencing not only increases the sensitivity of glioma cells to TMZ as well as X-rays but also counteracts the hypoxia-induced chemo- and radioresistance. Silencing EPOR on glioma cells exposed to conventional treatments enhances senescence and induces a robust genomic instability that leads to caspase-dependent mitotic death by increasing the number of polyploid cells and cyclin B1 expression. Overall these data suggest that EPOR could be an attractive target to overcome therapeutic resistance toward ionising radiation or temozolomide.


Assuntos
Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioma/terapia , Receptores da Eritropoetina/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Senescência Celular/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Quimiorradioterapia , Ciclina B1/metabolismo , Dacarbazina/farmacologia , Glioma/genética , Glioma/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos Nus , Mitose/efeitos dos fármacos , Mitose/efeitos da radiação , Poliploidia , Interferência de RNA , Terapêutica com RNAi , Receptores da Eritropoetina/genética , Temozolomida , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Methods Mol Biol ; 982: 79-101, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23456863

RESUMO

Animal models constitute an indispensable tool to investigate human pathology. Here we describe the procedure to induce permanent and transient cerebral ischemia in the mouse and the rat. The model of transient occlusion of the middle cerebral artery (MCA) is performed by the insertion of an occlusive filament until the origin of the MCA while the permanent occlusion described in the mice is performed by a distal electrocoagulation of the MCA. Those models allow evaluating the efficiency of therapeutic strategy of ischemia from tissular aspect to behavioral and cognitive impairment assessment. They were widely used in the literature to evaluate the efficiency of different drugs including the cytokines and especially erythropoietin (EPO) or its derivatives.


Assuntos
Isquemia Encefálica/metabolismo , Eritropoetina/metabolismo , Animais , Isquemia Encefálica/patologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Camundongos , Artéria Cerebral Média/metabolismo , Artéria Cerebral Média/patologia , Ratos
6.
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
7.
Neuro Oncol ; 15(1): 41-56, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23115160

RESUMO

The individualized care of glioma patients ought to benefit from imaging biomarkers as precocious predictors of therapeutic efficacy. Contrast enhanced MRI and [(18)F]-fluorodeoxyglucose (FDG)-PET are routinely used in clinical settings; their ability to forecast the therapeutic response is controversial. The objectives of our preclinical study were to analyze sensitive µMRI and/or µPET imaging biomarkers to predict the efficacy of anti-angiogenic and/or chemotherapeutic regimens. Human U87 and U251 orthotopic glioma models were implanted in nude rats. Temozolomide and/or bevacizumab were administered. µMRI (anatomical, diffusion, and microrheological parameters) and µPET ([(18)F]-FDG and [(18)F]-fluoro-l-thymidine [FLT]-PET) studies were undertaken soon (t(1)) after treatment initiation compared with late anatomical µMRI evaluation of tumor volume (t(2)) and overall survival. In both models, FDG and FLT uptakes were attenuated at t(1) in response to temozolomide alone or with bevacizumab. The distribution of FLT, reflecting intratumoral heterogeneity, was also modified. FDG was less predictive for treatment efficacy than was FLT (also highly correlated with outcome, P < .001 for both models). Cerebral blood volume was significantly decreased by temozolomide + bevacizumab and was correlated with survival for rats with U87 implants. While FLT was highly predictive of treatment efficacy, a combination of imaging biomarkers was superior to any one alone (P < .0001 in both tumors with outcome). Our results indicate that FLT is a sensitive predictor of treatment efficacy and that predictability is enhanced by a combination of imaging biomarkers. These findings may translate clinically in that individualized glioma treatments could be decided in given patients after PET/MRI examinations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/diagnóstico por imagem , Fluordesoxiglucose F18 , Glioblastoma/diagnóstico por imagem , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab , Biomarcadores/análise , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Ratos , Ratos Nus , Taxa de Sobrevida , Temozolomida , Resultado do Tratamento , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
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
9.
Exp Neurol ; 233(1): 264-72, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22051176

RESUMO

Hypoxic-ischemic (HI) brain injury occurring during the perinatal period is still a major cause of mortality and morbidity. We assessed the impact of maternal hypertension, the most common medical disorder of pregnancy, on the anatomical and functional consequences of HI insult in the immature brain. Rat pups from spontaneously hypertensive (SHR) and normotensive (Wistar Kyoto - WKY) dams were subjected to HI brain damage at post-natal day 7 (P7). Brain lesion and functional deficits were analyzed from 10 min to 35 days after HI, using magnetic resonance imaging (MRI), sensorimotor and cognitive tests. MRI data revealed that SHR pups displayed less brain damage than WKY, attested by an initial smaller lesion followed by a reduced tissue loss at chronic stage (57.1±21.6 and 31.1±27% ipsilateral hemisphere atrophy in WKY and SHR, respectively). Behavioral analyses showed less HI-induced behavioral deficits in motor coordination (rotarod test) and spatial learning (Morris water maze test) in pups from hypertensive dams compared to those from normotensive ones. The data suggest that maternal hypertension causes prenatal stress that may render the immature brain more resistant to subsequent hypoxia-ischemia, related to a preconditioning phenomenon.


Assuntos
Comportamento Animal/fisiologia , Hipertensão/fisiopatologia , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/diagnóstico , Precondicionamento Isquêmico , Troca Materno-Fetal/fisiologia , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Pressão Sanguínea , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Feminino , Força da Mão/fisiologia , Hipertensão/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Aprendizagem em Labirinto , Atividade Motora , Gravidez , Desempenho Psicomotor/fisiologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Teste de Desempenho do Rota-Rod , Comportamento Espacial
10.
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
11.
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
12.
Stroke ; 42(5): 1412-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21441158

RESUMO

BACKGROUND AND PURPOSE: The impact of stroke on white matter is poorly described in preclinical investigations mainly based on rodents with a low white (WM)/gray matter ratio. Using diffusion tensor imaging, we evaluated WM alterations and correlated them with sensorimotor deficits after stroke in the marmoset, a nonhuman primate that displays a WM/gray matter ratio close to that of humans. METHODS: Marmosets underwent a transient brain ischemia (3-hour). Eight serial MRI examinations were made during ischemia and up to 45 days after reperfusion. The sensorimotor deficits were evaluated weekly over 45 days. To assess WM alterations, the SD of the angle of the first eigenvector projection was calculated in the cortex and in the internal and external capsules. The fiber-tracking approach was used to measure the number and the length of bundles. RESULTS: Changes in the apparent diffusion coefficient and the fractional anisotropy values were similar during the temporal evolution of the lesion in the marmoset model of ischemia to that reported in patients with stroke. Despite an absence of visible lesions on T2-MRI and diffusion tensor imaging at the chronic stage, diffusion tensor MRI evidenced alterations in WM by the increase in the standard deviation of the angle of the first eigenvector projection in the cortex, internal and external capsules, and the decrease in the number of bundles of fibers tracked. The disruption of WM was strongly correlated with the chronic sensorimotor deficits. CONCLUSIONS: Despite an absence of a visible ischemic lesion at the chronic stage, diffusion tensor MRI revealed disorganization of WM, which probably underlies the persistence of functional deficits.


Assuntos
Isquemia Encefálica/patologia , Imagem de Difusão por Ressonância Magnética/métodos , Imageamento por Ressonância Magnética/métodos , Fibras Nervosas Mielinizadas/patologia , Tratos Piramidais/patologia , Animais , Comportamento Animal/fisiologia , Isquemia Encefálica/fisiopatologia , Callithrix , Modelos Animais de Doenças , Feminino , Masculino , Desempenho Psicomotor/fisiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
13.
J Cereb Blood Flow Metab ; 31(2): 504-13, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20648035

RESUMO

Although chronic arterial hypertension (CAH) increases the risk of stroke and the severity of the resultant lesion, it is rarely integrated in preclinical studies. Here, we analyzed the impact of CAH on the acute spatiotemporal evolution of the ischemic penumbra as defined by the perfusion-weighted imaging/diffusion-weighted imaging mismatch. Sequential 7T-MRI examinations were performed from 30 minutes up to 4 hours after permanent cerebral ischemia in genetically hypertensive rats (spontaneously hypertensive rats, SHR), renovascular-hypertensive rats (RH-WKY), and their normotensive controls (Wistar-Kyoto rats, WKY). The apparent diffusion coefficient (ADC)-defined lesion was larger in hypertensive rats than in normotensive animals as early as 30 minutes after the ischemia. The ischemic penumbra was smaller in both genetically and renovascular-hypertensive rats (at 30 minutes; SHR=66±25 mm(3), RH-WKY=55±17 mm(3) versus WKY=117±14 mm(3); P<0.008) and there was no significant difference between the perfusion deficit and ADC lesion (mismatch definition of penumbra) as early as 90 minutes after the occlusion. Genetic hypertension and induced renovascular hypertension resulted in larger lesion and smaller penumbra that vanished rapidly. These data support the need to integrate CAH in preclinical studies relative to the treatment of stroke, as failure to do so may lead to preclinical results nonpredictive of clinical trials, which include hypertensive patients.


Assuntos
Isquemia Encefálica/patologia , Hipertensão Renovascular/patologia , Hipertensão/genética , Hipertensão/patologia , Animais , Pressão Sanguínea/fisiologia , Doença Crônica , Interpretação Estatística de Dados , Imagem de Difusão por Ressonância Magnética , Progressão da Doença , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
14.
Eur J Nucl Med Mol Imaging ; 37(12): 2371-80, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20814674

RESUMO

PURPOSE: Neuroinflammation is involved in stroke pathophysiology and might be imaged using radioligands targeting the 18 kDa translocator protein (TSPO). METHODS: We studied microglial reaction in brain areas remote from the primary lesion site in two rodent models of focal cerebral ischaemia (permanent or transient) using [125I]-CLINDE, a promising TSPO single photon emission computed tomography radioligand. RESULTS: In a mouse model of permanent middle cerebral artery occlusion (MCAO), ex vivo autoradiographic studies demonstrated, besides in the ischaemic territory, accumulation of [125I]-CLINDE in the ipsilateral thalamus with a binding that progressed up to 3 weeks after MCAO. [125I]-CLINDE binding markedly decreased in animals pre-injected with either unlabelled CLINDE or PK11195, while no change was observed with flumazenil pre-treatment, demonstrating TSPO specificity. In rats subjected to transient MCAO, [125I]-CLINDE binding in the ipsilateral thalamus and substantia nigra pars reticulata (SNr) was significantly higher than that in contralateral tissue. Moreover, [125I]-CLINDE binding in the thalamus and SNr was quantitatively correlated to the ischaemic volume assessed by MRI in the cortex and striatum, respectively. CONCLUSION: Clinical consequences of secondary neuronal degeneration in stroke might be better treated thanks to the discrimination of neuronal processes using in vivo molecular imaging and potent TSPO radioligands like CLINDE to guide therapeutic interventions.


Assuntos
Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Microglia/diagnóstico por imagem , Microglia/metabolismo , Receptores de GABA/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
15.
J Cereb Blood Flow Metab ; 30(2): 273-85, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19794396

RESUMO

Using multimodal magnetic resonance imaging (MRI), behavioral, and immunohistochemical analyses, we examined pathological changes at the acute, sub-acute, and chronic stages, induced by permanent or temporary ischemia in the common marmoset. Animals underwent either permanent (pMCAO) or 3-h transient (tMCAO) occlusion of the middle cerebral artery (MCAO) by the intraluminal thread approach. MRI scans were performed at 1 h, 8, and 45 days after MCAO. Sensorimotor deficits were assessed weekly up to 45 days after MCAO. Immunohistological studies were performed to examine neuronal loss, astrogliosis, and neurogenesis. Remote lesions were analyzed using retrograde neuronal tracers. At day 8 (D8), the lesion defined on diffusion tensor imaging (DTI)-MRI and T2-MRI was significantly larger in pMCAO as compared with that in the tMCAO group. At D45, the former still displayed abnormal signals in T2-MRI. Post-mortem analyses revealed widespread neuronal loss and associated astrogliosis to a greater extent in the pMCAO group. Neurogenesis was increased in both groups in the vicinity of the lesion. Disconnections between the caudate and the temporal cortex, and between the parietal cortex and the thalamus, were observed. Sensorimotor impairments were more severe and long-lasting in pMCAO relative to tMCAO. The profile of brain damage and functional deficits seen in the marmoset suggests that this model could be suitable to test therapies against stroke.


Assuntos
Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Animais , Comportamento Animal/fisiologia , Encéfalo/patologia , Callithrix , Doença Crônica , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Atividade Motora , Neurogênese/fisiologia , Recuperação de Função Fisiológica/fisiologia
16.
Stroke ; 40(10): 3349-55, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628803

RESUMO

BACKGROUND AND PURPOSE: Inspired from preconditioning studies, ischemic postconditioning, consisting of the application of intermittent interruptions of blood flow shortly after reperfusion, has been described in cardiac ischemia and recently in stroke. It is well known that ischemic tolerance can be achieved in the brain not only by ischemic preconditioning, but also by hypoxic preconditioning. However, the existence of hypoxic postconditioning has never been reported in cerebral ischemia. METHODS: Adult mice subjected to transient middle cerebral artery occlusion underwent chronic intermittent hypoxia starting either 1 or 5 days after ischemia and brain damage was assessed by T2-weighted MRI at 43 days. In addition, we investigated the potential neuroprotective effect of hypoxia applied after oxygen glucose deprivation in primary neuronal cultures. RESULTS: The present study shows for the first time that a late application of hypoxia (5 days) after ischemia reduced delayed thalamic atrophy. Furthermore, hypoxia performed 14 hours after oxygen glucose deprivation induced neuroprotection in primary neuronal cultures. We found that hypoxia-inducible factor-1alpha expression as well as those of its target genes erythropoietin and adrenomedullin is increased by hypoxic postconditioning. Further studies with pharmacological inhibitors or recombinant proteins for erythropoietin and adrenomedullin revealed that these molecules participate in this hypoxia postconditioning-induced neuroprotection. CONCLUSIONS: Altogether, this study demonstrates for the first time the existence of a delayed hypoxic postconditioning in cerebral ischemia and in vitro studies highlight hypoxia-inducible factor-1alpha and its target genes, erythropoietin and adrenomedullin, as potential effectors of postconditioning.


Assuntos
Encéfalo/metabolismo , Citoproteção/fisiologia , Hipóxia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/prevenção & controle , Hipóxia-Isquemia Encefálica/terapia , Adrenomedulina/antagonistas & inibidores , Adrenomedulina/metabolismo , Adrenomedulina/farmacologia , Animais , Atrofia/fisiopatologia , Atrofia/prevenção & controle , Atrofia/terapia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Metabolismo Energético/fisiologia , Eritropoetina/antagonistas & inibidores , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Hipóxia Encefálica/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/prevenção & controle , Infarto da Artéria Cerebral Média/terapia , Masculino , Camundongos , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Degeneração Neural/terapia , Estresse Oxidativo/fisiologia , Fatores de Tempo
17.
Neuro Oncol ; 11(5): 488-502, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19168695

RESUMO

Despite treatment efforts, the median survival in patients with glioblastoma multiforme, the most aggressive form of glioma, does not extend beyond 12-15 months. One of the major pathophysiological characteristics of these tumors is their ability to induce active angiogenesis. Thus, based on the lack of efficient therapies, agents that inhibit angiogenesis are particularly attractive as a therapeutic option. However, it has been recently proposed that although specifically targeting vascular endothelial growth factor, the main angiogenic factor, certainly leads to significant tumor regression, it could also be followed by tumor relapses. In this case, angiogenesis is driven by alternate pathways that include other angiogenic factors. One possible strategy to overcome this therapeutic obstacle is to overexpress antivascular factors such as angiopoietin-2 (Ang2). Here, by using MRI and histological analysis, we studied the vascular events involved in glioma growth impairment induced by Ang2 overexpression. Our results show that an increase in Ang2 expression, during the tumor growth, leads to a significant decrease in tumor growth ( approximately 86%) along with an increase in the survival median ( approximately 70%) but does not modify the tumor vascular area or cerebral blood volume. However, tumor Ang2-derived blood vessels display an abnormal, enlarged morphology. We show that the presence of Ang2 leads to an enhancement of tumor perfusion and a decrease in tumor vessel permeability. Based on our MR image evaluations of hemodynamic tumor vessel changes, we propose that Ang2-derived tumor vessels lead to an inadequate oxygenation of the tumor tissue, leading to impairment in tumor growth.


Assuntos
Angiopoietina-2/genética , Neoplasias Encefálicas/genética , Glioma/genética , Neovascularização Patológica/genética , Angiopoietina-2/biossíntese , Animais , Western Blotting , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Glioma/irrigação sanguínea , Glioma/patologia , Hemodinâmica , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Neovascularização Patológica/patologia , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Regulação para Cima
18.
Behav Neurosci ; 123(1): 224-30, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19170448

RESUMO

Distal occlusion of the middle cerebral artery (dMCAo), which closely mimics human stroke, is one of the most used animal models. However, although assessment of histological and functional outcome is increasingly recommended for preclinical studies, the latter is often excluded because of the high difficulties to estimate, especially in mice, behavioral impairments. The aim of our study was to deeply screen functional consequences of distal permanent MCAo in mice to target relevant behaviors for future studies. A set of sensorimotor and cognitive tests were performed during 3 weeks postsurgery in 2 groups of mice. Afterward, brain infarctions were estimated by histological staining or magnetic resonance imaging. Overall, while no long-term functional impairments could be detected, the adhesive removal was the only test showing a deficit. Interestingly, this sensorimotor impairment was correlated to cortical damage 3 weeks after surgery. In conclusion, despite the fact that dMCAo-induced deficits could not be evidenced by most of our behavioral tests, the authors showed that the adhesive removal test was the only one, sensitive enough, to highlight a long-term deficit. This result suggests therefore that this mouse model of ischemia is relevant to efficiently assess therapeutic strategies with histological but also behavioral analysis, provided that relevant tests are used.


Assuntos
Sintomas Comportamentais/diagnóstico , Sintomas Comportamentais/fisiopatologia , Cognição/fisiologia , Desempenho Psicomotor/fisiologia , Sensação/fisiologia , Análise de Variância , Animais , Sintomas Comportamentais/etiologia , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Isquemia Encefálica/complicações , Modelos Animais de Doenças , Imageamento por Ressonância Magnética/métodos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Atividade Motora , Testes Neuropsicológicos , Equilíbrio Postural/fisiologia , Teste de Desempenho do Rota-Rod
19.
J Cereb Blood Flow Metab ; 28(9): 1552-63, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18478023

RESUMO

Many studies showed beneficial effects of either erythropoietin (EPO) or mesenchymal stem cells (MSCs) treatment in cerebral ischemia. In addition to a neuroprotective role, not only EPO but also MSC favors neurogenesis and functional recovery. In an attempt to further improve postischemic tissue repair, we investigated the effect of a systemic administration of MSC, in the presence or not of EPO, on neurogenesis and functional recovery in a transient focal cerebral ischemia model in the adult rat. Twenty-four hours after ischemia, the rats were divided into four groups, namely vehicle, MSC, EPO, and MSC+EPO, and received a single intravenous injection of MSC (2 x 10(6) cells) and/or a repeated intraperitoneal administration of EPO (1,000 UI/kg) for 3 days. The lesion volume, the MSC outcome, neurogenesis, and functional recovery were assessed 51 days after ischemia. The results showed that cellular proliferation and neurogenesis were increased along the lateral ventricle wall in the MSC+EPO group, whereas no significant effect was observed in groups receiving MSC or EPO alone. This effect was accompanied by an improvement of mnesic performances. Mesenchymal stem cells expressing neuronal or glial markers were detected in the ischemic hemisphere. These results suggest that EPO could act in a synergistic way with MSC to potentiate the postischemic neurogenesis.


Assuntos
Eritropoetina/farmacologia , Ataque Isquêmico Transitório/terapia , Transplante de Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Proliferação de Células , Eritropoetina/administração & dosagem , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Neuroglia/citologia , Neurônios/citologia , Ratos , Proteínas Recombinantes
20.
J Neurochem ; 106(3): 1388-403, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18498438

RESUMO

The understanding of mechanisms involved in ischaemic brain tolerance may provide new therapeutical targets for stroke. In vivo genomic studies revealed an up-regulation of adrenomedullin expression by hypoxic pre-conditioning. Furthermore, adrenomedullin reduced ischaemia-induced brain damage in rodents. However, whether adrenomedullin is involved in hypoxic pre-conditioning-induced tolerance and whether adrenomedullin protects directly neurons against ischaemia remain unknown. Using a neuronal model of hypoxic pre-conditioning and oxygen glucose deprivation (OGD), we showed that 0.1% or 0.5% of O2 pre-conditioning reduced the OGD-induced neuronal death, whereas 1% or 2% of O2 pre-treatment did not induce neuroprotection. Adrenomedullin expression increased following the hypoxic period, and following OGD only in pre-conditioned (0.1% or 0.5% of O2) neurons. Adrenomedullin pre-treatment and post-treatment reduced the OGD-induced neuronal death, partly through PI3kinase-dependent pathway. However, adrenomedullin antagonism during hypoxic pre-conditioning failed to inhibit the neuroprotection whereas adrenomedullin antagonism following OGD abolished the hypoxic pre-conditioning-induced neuroprotection. Finally, we showed that adrenomedullin is involved in neuroprotection induced by endothelial cells and microglia. In contrast, neuroprotection induced by astrocytes occurred through adrenomedullin-independent mechanisms. Altogether, our results suggest that adrenomedullin is an effector of the hypoxic pre-conditioning-induced neuronal tolerance and a potent autocrine and paracrine neuroprotective factor during cerebral ischaemia.


Assuntos
Adrenomedulina/fisiologia , Comunicação Autócrina/fisiologia , Glucose/fisiologia , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Estresse Oxidativo/fisiologia , Comunicação Parácrina/fisiologia , Adrenomedulina/biossíntese , Animais , Astrócitos/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Glucose/deficiência , Camundongos , Microglia/metabolismo , Neurônios/fisiologia , Oxigênio , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...