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











Base de datos
Intervalo de año de publicación
1.
J Cereb Blood Flow Metab ; 38(10): 1769-1780, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28617154

RESUMEN

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.


Asunto(s)
Encéfalo/patología , Modelos Animales de Enfermedad , Hipertensión Renovascular/patología , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Hipertensión Renovascular/complicaciones , Masculino , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones
2.
Hypertens Res ; 38(11): 723-32, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26084262

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Glioblastoma/patología , Hipertensión/complicaciones , Animales , Presión Arterial , Peso Corporal , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/etiología , Glioblastoma/irrigación sanguínea , Glioblastoma/etiología , Imagen por Resonancia Magnética , Masculino , Distribución Aleatoria , Ratas Endogámicas SHR , Ratas Endogámicas WKY
3.
Acta Biomater ; 15: 77-88, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25556361

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/complicaciones , Portadores de Fármacos/química , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Conducta Animal , Vasos Sanguíneos/efectos de los fármacos , Isquemia Encefálica/fisiopatología , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Laminina/farmacología , Imagen por Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/fisiopatología , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
Oncotarget ; 6(4): 2101-19, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25544764

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioma/terapia , Receptores de Eritropoyetina/metabolismo , Animales , Antineoplásicos Alquilantes/farmacología , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de la radiación , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Quimioradioterapia , Ciclina B1/metabolismo , Dacarbazina/farmacología , Glioma/genética , Glioma/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones Desnudos , Mitosis/efectos de los fármacos , Mitosis/efectos de la radiación , Poliploidía , Interferencia de ARN , Tratamiento con ARN de Interferencia , Receptores de Eritropoyetina/genética , Temozolomida , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Methods Mol Biol ; 982: 79-101, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23456863

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Eritropoyetina/metabolismo , Animales , Isquemia Encefálica/patología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ratones , Arteria Cerebral Media/metabolismo , Arteria Cerebral Media/patología , Ratas
6.
Biol Chem ; 394(4): 529-39, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23399636

RESUMEN

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.


Asunto(s)
Glioma/patología , Hipoxia/diagnóstico , Misonidazol/análogos & derivados , Tomografía de Emisión de Positrones/métodos , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética
7.
J Cereb Blood Flow Metab ; 33(3): 389-95, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23211963

RESUMEN

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.


Asunto(s)
Angiopoyetina 2/biosíntesis , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Animales , Barrera Hematoencefálica/patología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/terapia , Permeabilidad Capilar/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Angiografía por Resonancia Magnética , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Neuro Oncol ; 15(1): 41-56, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115160

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/diagnóstico por imagen , Fluorodesoxiglucosa F18 , Glioblastoma/diagnóstico por imagen , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Radiofármacos , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab , Biomarcadores/análisis , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Ratas , Ratas Desnudas , Tasa de Supervivencia , Temozolomida , Resultado del Tratamiento , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Exp Neurol ; 233(1): 264-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22051176

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Hipertensión/fisiopatología , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/diagnóstico , Precondicionamiento Isquémico , Intercambio Materno-Fetal/fisiología , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Presión Sanguínea , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Femenino , Fuerza de la Mano/fisiología , Hipertensión/diagnóstico , Imagen por Resonancia Magnética , Masculino , Aprendizaje por Laberinto , Actividad Motora , Embarazo , Desempeño Psicomotor/fisiología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Prueba de Desempeño de Rotación con Aceleración Constante , Conducta Espacial
10.
Nucl Med Biol ; 38(6): 781-93, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21843775

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamiento farmacológico , Indoles/farmacología , Imagen por Resonancia Magnética , Misonidazol/análogos & derivados , Tomografía de Emisión de Positrones , Pirroles/farmacología , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Transporte Biológico , Encéfalo/irrigación sanguínea , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Circulación Cerebrovascular/efectos de los fármacos , Modelos Animales de Enfermedad , Glioma/irrigación sanguínea , Glioma/diagnóstico , Glioma/tratamiento farmacológico , Glioma/patología , Indoles/uso terapéutico , Cinética , Misonidazol/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Permeabilidad/efectos de los fármacos , Pirroles/uso terapéutico , Ratas , Sunitinib , Resultado del Tratamiento
11.
Exp Cell Res ; 317(16): 2321-32, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21749867

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioma/tratamiento farmacológico , Receptores de Eritropoyetina/antagonistas & inhibidores , Transducción de Señal/fisiología , Animales , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Núcleo Caudado/patología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Eritropoyetina/antagonistas & inhibidores , Eritropoyetina/genética , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/genética , Glioma/metabolismo , Glioma/patología , Células Hep G2 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Ratas , Ratas Endogámicas F344 , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Eliminación de Secuencia/fisiología , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Stroke ; 42(5): 1412-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21441158

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/patología , Imagen de Difusión por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/métodos , Fibras Nerviosas Mielínicas/patología , Tractos Piramidales/patología , Animales , Conducta Animal/fisiología , Isquemia Encefálica/fisiopatología , Callithrix , Modelos Animales de Enfermedad , Femenino , Masculino , Desempeño Psicomotor/fisiología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología
13.
J Cereb Blood Flow Metab ; 31(2): 504-13, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20648035

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/patología , Hipertensión Renovascular/patología , Hipertensión/genética , Hipertensión/patología , Animales , Presión Sanguínea/fisiología , Enfermedad Crónica , Interpretación Estadística de Datos , Imagen de Difusión por Resonancia Magnética , Progresión de la Enfermedad , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
14.
Eur J Nucl Med Mol Imaging ; 37(12): 2371-80, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20814674

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Microglía/diagnóstico por imagen , Microglía/metabolismo , Receptores de GABA/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
15.
J Cereb Blood Flow Metab ; 30(2): 273-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19794396

RESUMEN

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.


Asunto(s)
Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/fisiopatología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Animales , Conducta Animal/fisiología , Encéfalo/patología , Callithrix , Enfermedad Crónica , Inmunohistoquímica , Imagen por Resonancia Magnética , Actividad Motora , Neurogénesis/fisiología , Recuperación de la Función/fisiología
16.
Stroke ; 40(10): 3349-55, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19628803

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Citoprotección/fisiología , Hipoxia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Hipoxia-Isquemia Encefálica/terapia , Adrenomedulina/antagonistas & inhibidores , Adrenomedulina/metabolismo , Adrenomedulina/farmacología , Animales , Atrofia/fisiopatología , Atrofia/prevención & control , Atrofia/terapia , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Células Cultivadas , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Eritropoyetina/antagonistas & inhibidores , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Hipoxia Encefálica/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/prevención & control , Infarto de la Arteria Cerebral Media/terapia , Masculino , Ratones , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Degeneración Nerviosa/terapia , Estrés Oxidativo/fisiología , Factores de Tiempo
17.
Behav Neurosci ; 123(1): 224-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19170448

RESUMEN

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.


Asunto(s)
Síntomas Conductuales/diagnóstico , Síntomas Conductuales/fisiopatología , Cognición/fisiología , Desempeño Psicomotor/fisiología , Sensación/fisiología , Análisis de Varianza , Animales , Síntomas Conductuales/etiología , Infarto Encefálico/etiología , Infarto Encefálico/patología , Isquemia Encefálica/complicaciones , Modelos Animales de Enfermedad , Imagen por Resonancia Magnética/métodos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Actividad Motora , Pruebas Neuropsicológicas , Equilibrio Postural/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante
18.
Neuro Oncol ; 11(5): 488-502, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19168695

RESUMEN

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.


Asunto(s)
Angiopoyetina 2/genética , Neoplasias Encefálicas/genética , Glioma/genética , Neovascularización Patológica/genética , Angiopoyetina 2/biosíntesis , Animales , Western Blotting , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Glioma/irrigación sanguínea , Glioma/patología , Hemodinámica , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Neovascularización Patológica/patología , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
19.
J Cereb Blood Flow Metab ; 28(9): 1552-63, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18478023

RESUMEN

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.


Asunto(s)
Eritropoyetina/farmacología , Ataque Isquémico Transitorio/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Proliferación Celular , Eritropoyetina/administración & dosificación , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Neuroglía/citología , Neuronas/citología , Ratas , Proteínas Recombinantes
20.
J Neurochem ; 106(3): 1388-403, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18498438

RESUMEN

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.


Asunto(s)
Adrenomedulina/fisiología , Comunicación Autocrina/fisiología , Glucosa/fisiología , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Estrés Oxidativo/fisiología , Comunicación Paracrina/fisiología , Adrenomedulina/biosíntesis , Animales , Astrocitos/metabolismo , Hipoxia de la Célula/fisiología , Células Cultivadas , Glucosa/deficiencia , Ratones , Microglía/metabolismo , Neuronas/fisiología , Oxígeno , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA