Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2193: 49-65, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32808258

RESUMEN

Traumatic brain injury (TBI) is a heterogeneous brain injury which represents one of the leading causes of mortality and disability worldwide. Rodent TBI models are helpful to examine the cellular and molecular mechanisms after injury. Controlled cortical impact (CCI) is one of the most commonly used TBI models in rats and mice, based on its consistency of injury and ease of implementation. Here, we describe a CCI protocol to induce a moderate contusion to the somatosensory motor cortex. We provide additional protocols for monitoring animals after CCI induction.


Asunto(s)
Bioensayo/métodos , Lesiones Traumáticas del Encéfalo/fisiopatología , Lesiones Encefálicas/fisiopatología , Contusiones/fisiopatología , Animales , Modelos Animales de Enfermedad , Ratones , Ratas
2.
Glia ; 68(3): 528-542, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31670865

RESUMEN

Mild-traumatic brain injury (mTBI) represents ~80% of all emergency room visits and increases the probability of developing long-term cognitive disorders in children. To date, molecular and cellular mechanisms underlying post-mTBI cognitive dysfunction are unknown. Astrogliosis has been shown to significantly alter astrocytes' properties following brain injury, potentially leading to significant brain dysfunction. However, such alterations have never been investigated in the context of juvenile mTBI (jmTBI). A closed-head injury model was used to study jmTBI on postnatal-day 17 mice. Astrogliosis was evaluated using glial fibrillary acidic protein (GFAP), vimentin, and nestin immunolabeling in somatosensory cortex (SSC), dentate gyrus (DG), amygdala (AMY), and infralimbic area (ILA) of prefrontal cortex in both hemispheres from 1 to 30 days postinjury (dpi). In vivo T2-weighted-imaging (T2WI) and diffusion tensor imaging (DTI) were performed at 7 and 30 dpi to examine tissue level structural alterations. Increased GFAP-labeling was observed up to 30 dpi in the ipsilateral SSC, the initial site of the impact. However, vimentin and nestin expression was not perturbed by jmTBI. The morphology of GFAP positive cells was significantly altered in the SSC, DG, AMY, and ILA up to 7 dpi that some correlated with magnetic resonance imaging changes. T2WI and DTI values were significantly altered at 30 dpi within these brain regions most prominently in regions distant from the impact site. Our data show that jmTBI triggers changes in astrocytic phenotype with a distinct spatiotemporal pattern. We speculate that the presence and time course of astrogliosis may contribute to pathophysiological processes and long-term structural alterations following jmTBI.


Asunto(s)
Astrocitos/metabolismo , Conmoción Encefálica/patología , Lesiones Encefálicas/patología , Traumatismos Cerrados de la Cabeza/patología , Animales , Encéfalo/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Imagen por Resonancia Magnética/métodos , Ratones
3.
J Cereb Blood Flow Metab ; 40(1): 163-176, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30354902

RESUMEN

Complex cellular and molecular events occur in the neurovascular unit after stroke, such as blood-brain barrier (BBB) dysfunction and inflammation that contribute to neuronal death, neurological deterioration and mortality. Caveolin-1 (Cav-1) has distinct physiological functions such as caveolae formation associated with endocytosis and transcytosis as well as in signaling pathways. Cav-1 has been proposed to be involved in BBB dysfunction after brain injury; however, its precise role is poorly understood. The goal of this study was to characterize the expression and effect of Cav-1 deletion on outcome in the first week in a transient Middle Cerebral Artery Occlusion stroke model. We found increased Cav-1 expression in new blood vessels in the lesion and in reactive astrocytes in the peri-lesion areas. In Cav-1 KO mice, the lesion volume was larger and the behavioral outcome worse than in WT mice. Cav-1 KO mice exhibited reduced neovascularization and modified astrogliosis, without formation of a proper glial scar around the lesion at three days post injury, coinciding with aggravated outcomes. Altogether, these results point towards a potential protective role of endogenous Cav-1 in the first days after ischemia by promoting neovascularization, astrogliosis and scar formation.


Asunto(s)
Caveolina 1/fisiología , Neovascularización Patológica/patología , Plasticidad Neuronal/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Astrocitos/patología , Barrera Hematoencefálica/patología , Caveolina 1/metabolismo , Caveolina 1/farmacología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Ratones , Ratones Noqueados , Neovascularización Patológica/etiología
4.
J Neurosci Res ; 98(1): 9-18, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30430614

RESUMEN

Brain edema is a common feature of brain injuries, which leads to increased intracranial pressure (ICP) and ischemia that worsen outcome. Current management of edema focuses on reduction of ICP, but there are no treatments targeting the molecular players directly involved in edema process. The perivascular astrocyte endfeet are critical in maintaining brain homeostasis with ionic and water exchange; in this context, aquaporins (AQPs), astrocyte water channels, have emerged as privileged targets for edema modulation. However, AQPs can facilitate either accumulation or drainage of water, depending on the osmotic gradients between extra-intracellular space; and thus inhibition of AQPs leads to different outcomes depending on specific tissue characteristics and time post-injury. Most of this knowledge has been gathered from the study of AQP4, the best characterized AQP and the one that has the biggest impact on water movement. In addition to the level of expression, the ratio of AQP4 isoforms (m1, m23 or mz), the spatial distribution of AQP4 into orthogonal arrays of particles, and the interaction of AQP4 with neighboring ionic channels and gap junctions could directly impact edema formation. Although there are no specific AQP4 pharmacological blockers, the development of AQP4 siRNA offers a promising therapeutic tool. Given the complex dynamics of AQP4, therapies targeting AQP4 should carefully take into account the particular features of the injury (e.g., hemorrhagic vs. non-hemorrhagic) and different times after injury (e.g., phase of edema formation vs. resolution).


Asunto(s)
Acuaporinas/metabolismo , Barrera Hematoencefálica/metabolismo , Edema Encefálico/metabolismo , Encéfalo/metabolismo , Animales , Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA