Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Fluids Barriers CNS ; 21(1): 23, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38433215

RESUMEN

BACKGROUND: The active transport of molecules into the brain from blood is regulated by receptors, transporters, and other cell surface proteins that are present on the luminal surface of endothelial cells at the blood-brain barrier (BBB). However, proteomic profiling of proteins present on the luminal endothelial cell surface of the BBB has proven challenging due to difficulty in labelling these proteins in a way that allows efficient purification of these relatively low abundance cell surface proteins. METHODS: Here we describe a novel perfusion-based labelling workflow: in vivo glycocapture. This workflow relies on the oxidation of glycans present on the luminal vessel surface via perfusion of a mild oxidizing agent, followed by subsequent isolation of glycoproteins by covalent linkage of their oxidized glycans to hydrazide beads. Mass spectrometry-based identification of the isolated proteins enables high-confidence identification of endothelial cell surface proteins in rats and mice. RESULTS: Using the developed workflow, 347 proteins were identified from the BBB in rat and 224 proteins in mouse, for a total of 395 proteins in both species combined. These proteins included many proteins with transporter activity (73 proteins), cell adhesion proteins (47 proteins), and transmembrane signal receptors (31 proteins). To identify proteins that are enriched in vessels relative to the entire brain, we established a vessel-enrichment score and showed that proteins with a high vessel-enrichment score are involved in vascular development functions, binding to integrins, and cell adhesion. Using publicly-available single-cell RNAseq data, we show that the proteins identified by in vivo glycocapture were more likely to be detected by scRNAseq in endothelial cells than in any other cell type. Furthermore, nearly 50% of the genes encoding cell-surface proteins that were detected by scRNAseq in endothelial cells were also identified by in vivo glycocapture. CONCLUSIONS: The proteins identified by in vivo glycocapture in this work represent the most complete and specific profiling of proteins on the luminal BBB surface to date. The identified proteins reflect possible targets for the development of antibodies to improve the crossing of therapeutic proteins into the brain and will contribute to our further understanding of BBB transport mechanisms.


Asunto(s)
Barrera Hematoencefálica , Proteoma , Ratas , Ratones , Animales , Células Endoteliales , Proteómica , Encéfalo , Microvasos , Proteínas de la Membrana , Polisacáridos
2.
J Biol Chem ; 291(53): 27007-27022, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27864367

RESUMEN

Phaseic acid (PA) is a phytohormone regulating important physiological functions in higher plants. Here, we show the presence of naturally occurring (-)-PA in mouse and rat brains. (-)-PA is exclusively present in the choroid plexus and the cerebral vascular endothelial cells. Purified (-)-PA has no toxicity and protects cultured cortical neurons against glutamate toxicity through reversible inhibition of glutamate receptors. Focal occlusion of the middle cerebral artery elicited a significant induction in (-)-PA expression in the cerebrospinal fluid but not in the peripheral blood. Importantly, (-)-PA induction only occurred in the penumbra area, indicting a protective role of PA in the brain. Indeed, elevating the (-)-PA level in the brain reduced ischemic brain injury, whereas reducing the (-)-PA level using a monoclonal antibody against (-)-PA increased ischemic injury. Collectively, these studies showed for the first time that (-)-PA is an endogenous neuroprotective molecule capable of reversibly inhibiting glutamate receptors during ischemic brain injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Ácido Glutámico/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Receptores de Glutamato/química , Sesquiterpenos/uso terapéutico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Células Cultivadas , Infarto de la Arteria Cerebral Media/etiología , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/metabolismo
3.
Sci Rep ; 5: 7890, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25601765

RESUMEN

Semaphorin 3A (Sema3A) increased significantly in mouse brain following cerebral ischemia. However, the role of Sema3A in stroke brain remains unknown. Our aim was to determine wether Sema3A functions as a vascular permeability factor and contributes to ischemic brain damage. Recombinant Sema3A injected intradermally to mouse skin, or stereotactically into the cerebral cortex, caused dose- and time-dependent increases in vascular permeability, with a degree comparable to that caused by injection of a known vascular permeability factor vascular endothelial growth factor receptors (VEGF). Application of Sema3A to cultured endothelial cells caused disorganization of F-actin stress fibre bundles and increased endothelial monolayer permeability, confirming Sema3A as a permeability factor. Sema3A-mediated F-actin changes in endothelial cells were through binding to the neuropilin2/VEGFR1 receptor complex, which in turn directly activates Mical2, a F-actin modulator. Down-regulation of Mical2, using specific siRNA, alleviated Sema3A-induced F-actin disorganization, cellular morphology changes and endothelial permeability. Importantly, ablation of Sema3A expression, cerebrovascular permeability and brain damage were significantly reduced in response to transient middle cerebral artery occlusion (tMCAO) and in a mouse model of cerebral ischemia/haemorrhagic transformation. Together, these studies demonstrated that Sema3A is a key mediator of cerebrovascular permeability and contributes to brain damage caused by cerebral ischemia.


Asunto(s)
Lesiones Encefálicas/genética , Permeabilidad Capilar/genética , Semaforina-3A/genética , Accidente Cerebrovascular/genética , Actinas/genética , Actinas/metabolismo , Animales , Apoptosis/genética , Lesiones Encefálicas/fisiopatología , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Permeabilidad Capilar/fisiología , Corteza Cerebral/lesiones , Corteza Cerebral/patología , Proteínas del Citoesqueleto/biosíntesis , Modelos Animales de Enfermedad , Humanos , Ratones , Proteínas de Microfilamentos , Neuropilina-2/metabolismo , Semaforina-3A/administración & dosificación , Accidente Cerebrovascular/fisiopatología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
4.
Methods Mol Biol ; 1135: 157-75, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24510863

RESUMEN

Angiogenesis, the sprouting of new capillaries from preexisting vessels, is an integral part of both normal development and numerous pathological conditions such as tumor growth, inflammation, and stroke. The development of angiogenesis assays has been critical in understanding this process in both the context of disease and normal physiology. With the growing availability of antibodies against angiogenic markers as well as advances in microscopy and imaging analysis software, a more comprehensive assessment of the angiogenesis process is beginning to take form (Milner et al., Stroke 39:191-197, 2008; Freitas-Andrade et al., J Cereb Blood Flow Metab 32:663-675, 2012; Li et al., Glia 58:1157-1167, 2010; Dore-Duffy and LaManna, Antioxid Redox Signal 9:1363-1371, 2007). This chapter describes an in vivo method of inducing brain angiogenesis in mice by chronic exposure to mild hypoxia. In addition, a detailed procedure of quantifying angiogenesis using multiple immunofluorescent labeling of mouse brain tissue sections is also presented.


Asunto(s)
Encéfalo/irrigación sanguínea , Neovascularización Fisiológica , Animales , Hipoxia de la Célula , Circulación Cerebrovascular , Crioultramicrotomía , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Microscopía Fluorescente
5.
Neoplasia ; 15(5): 554-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633927

RESUMEN

Insulin-like growth factor-binding protein 4 (IGFBP-4/IBP-4) has potent IGF-independent anti-angiogenic and antitumorigenic effects. In this study, we demonstrated that these activities are located in the IGFBP-4 C-terminal protein fragment (CIBP-4), a region containing a thyroglobulin type 1 (Tg1) domain. Proteins bearing Tg1 domains have been shown to inhibit cathepsins, lysosomal enzymes involved in basement membrane degradation and implicated in tumor invasion and angiogenesis. In our studies, CIBP-4 was shown to internalize and co-localize with lysosomal-like structures in both endothelial cells (ECs) and glioblastoma U87MG cells. CIBP-4 also inhibited both growth factor-induced EC tubulogenesis in Matrigel and the concomitant increases in intracellular cathepsin B (CatB) activity. In vitro assays confirmed CIBP-4 capacity to block recombinant CatB activity. Biodistribution analysis of intravenously injected CIBP-4-Cy5.5 in a glioblastoma tumor xenograft model indicated targeted accumulation of CIBP-4 in tumors. Most importantly, CIBP-4 reduced tumor growth in this animal model by 60%. Pleiotropic anti-angiogenic and anti-tumorigenic activities of CIBP-4 most likely underlie its observed therapeutic potential against glioblastoma.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Catepsina B/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Fragmentos de Péptidos/farmacología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacocinética , Animales , Catepsina B/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Glioblastoma/enzimología , Glioblastoma/patología , Células HEK293 , Humanos , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/farmacocinética , Masculino , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacocinética , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Neurobiol Dis ; 54: 24-31, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23473743

RESUMEN

Increased brain infiltration of polymorphonuclear neutrophils (PMNs) occurs early after stroke and is important in eliciting brain inflammatory response during stroke recovery. In order to understand the molecular mechanism of PMN entry, we investigated the expression and requirement for Slit1, a chemorepulsive guidance cue, and its cognate receptor, Robo1, in a long-term recovery mouse model of cerebral ischemia. The expression levels of Robo1 were significantly decreased bilaterally at 24h following reperfusion. Robo1 expression levels remained suppressed in the ipsilateral cortex until 28d post MCAO-reperfusion, while the levels of Robo1 in the contralateral cortex recovered to the level of sham-operated mouse by 7d reperfusion. Circulating PMNs express high levels of Slit1, but not Robo1. Influx of PMNs into the ischemic core area occurred early (24h) after cerebral ischemia, when endothelial Robo1 expression was significantly reduced in the ischemic brain, indicating that Robo1 may form a repulsive barrier to PMN entry into the brain parenchyma. Indeed, blocking Slit1 on PMNs in a transwell migration assay in combination with an antibody blocking of Robo1 on human umbilical vein endothelial cells (HUVEC) significantly increased PMN transmigration during oxygen glucose deprivation, an in vitro model of ischemia. Collectively, in the normal brain, the presence of Slit1 on PMNs, and Robo1 on cerebral endothelial cells, generated a repulsive force to prevent the infiltration of PMNs into the brain. During stroke recovery, a transient reduction in Robo1 expression on the cerebral endothelial cells allowed the uncontrolled infiltration of Slit1-expressing PMNs into the brain causing inflammatory reactions.


Asunto(s)
Encéfalo/metabolismo , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neutrófilos/metabolismo , Receptores Inmunológicos/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Western Blotting , Encéfalo/inmunología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Accidente Cerebrovascular/inmunología , Transfección , Proteínas Roundabout
7.
J Neurochem ; 122(4): 764-74, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22681613

RESUMEN

Microglia are the 'immune cells' of the brain and their activation plays a vital role in the pathogenesis of many neurodegenerative diseases. Activated microglia produce high levels of pro-inflammatory factors, such as TNFα, causing neurotoxicity. Here we show that vimentin played a key role in controlling microglia activation and neurotoxicity during cerebral ischemia. Deletion of vimentin expression significantly impaired microglia activation in response to LPS in vitro and transient focal cerebral ischemia in vivo. Reintroduction of the functional vimentin gene back into vimentin knockout microglia restored their response to LPS. More importantly, impairment of microglia activation significantly protected brain from cerebral ischemia-induced neurotoxicity. Collectively, we demonstrate a previously unknown function of vimentin in controlling microglia activation.


Asunto(s)
Isquemia Encefálica/patología , Activación de Macrófagos/fisiología , Microglía/fisiología , Vimentina/fisiología , Animales , Western Blotting , Encéfalo/patología , Separación Celular , Técnica del Anticuerpo Fluorescente Indirecta , Procesamiento de Imagen Asistido por Computador , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/patología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microscopía Confocal , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/fisiopatología , Plásmidos/efectos de los fármacos , Plásmidos/genética , Daño por Reperfusión/patología , Tetraciclina/farmacología , Vimentina/genética
8.
Methods Mol Biol ; 763: 423-39, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21874469

RESUMEN

The blood-brain barrier (BBB) disruption following cerebral ischemia (stroke) contributes to the development of life-threatening brain edema. Recent studies suggested that the ischemic BBB disruption is not uniform throughout the affected brain region. The aim of this study was to establish in vivo optical imaging methods to assess the size selectivity and spatial distribution of the BBB disruption after a focal cerebral ischemia. The BBB permeability was assessed in mice subjected to a 60-min middle cerebral artery occlusion and 24 h of reperfusion using in vivo time domain near-infrared optical imaging after contrast enhancement with two tracers of different molecular size, Cy5.5 (1 kDa) and Cy5.5 conjugated with bovine serum albumin (BSA) (67 kDa). Volumetric reconstruction of contrast-enhanced brain areas in vivo and ex vivo indicated that the BSA-Cy5.5-enhancement is identical to the volume of infarct determined by TTC staining, whereas the volume of enhancement with Cy5.5 was 40% greater. The volume differential between areas of BBB disruption for small and large-size molecules could be useful for determining the size of peri-infarct tissues (penumbra) that can respond to neuroprotective therapies.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Tomografía Computarizada de Haz Cónico/métodos , Medios de Contraste/metabolismo , Ataque Isquémico Transitorio/metabolismo , Imagen Molecular/métodos , Daño por Reperfusión/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/patología , Carbocianinas/análisis , Carbocianinas/metabolismo , Bovinos , Medios de Contraste/análisis , Modelos Animales de Enfermedad , Fluorescencia , Histocitoquímica , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Ataque Isquémico Transitorio/etiología , Ataque Isquémico Transitorio/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Microtomía , Daño por Reperfusión/fisiopatología , Albúmina Sérica/análisis , Albúmina Sérica/metabolismo , Sales de Tetrazolio/análisis
9.
Cancer Biol Ther ; 12(7): 634-46, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21795858

RESUMEN

Insulin-like growth factor binding protein 7 (IGFBP7) is downregulated in several solid cancers. IGFBP7 has been proposed to act as a tumor suppressor gene through mechanisms involving senescence and apoptotic pathways. The tumor suppressor effect of IGFBP7 in glioblastoma multiforme (GBM) was examined in this study using two human GBM cell lines, U87MG and T98G. Exogenously applied IGFBP7 (20 and 100 nM) significantly reduced U87MG (~70 and ~75%, respectively) and T98G (~37 and ~50%, respectively) cell growth in soft agar. IGFBP7 stimulated senescence-associated ß-galactosidase in both U87MG and T98G cells without stimulating apoptosis (annexin V and propidium iodide staining, expression of SMARCB1 or BNIP3L and caspase cleavage) or affecting phosphorylation of p44/42 MAPK. The inhibitory effect of IGFBP7 on U87MG cell growth was further assessed in vivo using U87MG cells grafted on the chick chorioallantoic membrane. In this model, U87MG cells formed solid and highly vascularized tumors that were reduced in size (~40%) when treated with 500 nM IGFBP7 compared with control tumors. Vessels in IGFBP7-treated tumors were clustered, unevenly distributed and associated with higher number of α-SMA positive cells compared with those in untreated tumors. IGFBP7 induced both aortic smooth muscle cell (AoSMC) chemoattraction and endothelial cell (EC) transdifferentiation into a SM-like cell phenotype. U87MG conditioned media-induced IGFBP7 expression in ECs was significantly inhibited by the cross-talk/interaction with SMCs. This study indicates that IGFBP7 suppresses U87MG tumor cell growth, induces cell senescence and participates in tumor vessel stabilization by promoting SMC/pericyte recruitment and differentiation.


Asunto(s)
Neoplasias del Sistema Nervioso Central/irrigación sanguínea , Neoplasias del Sistema Nervioso Central/patología , Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Animales , Anexina A5/metabolismo , Aorta/citología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/metabolismo , Embrión de Pollo , Membrana Corioalantoides , Endotelio Vascular/citología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Músculo Liso Vascular/efectos de los fármacos , Fosforilación , beta-Galactosidasa/metabolismo
10.
PLoS One ; 6(6): e20808, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21687673

RESUMEN

Gangliosides, a member of the glycosphingolipid family, are heterogeneously expressed in biological membranes and are particularly enriched within the central nervous system. Gangliosides consist of mono- or poly-sialylated oligosaccharide chains of variable lengths attached to a ceramide unit and are found to be intimately involved in brain disease development. The purpose of this study is to examine the spatial profile of ganglioside species using matrix-assisted laser desorption/ionization (MALDI) imaging (IMS) following middle cerebral artery occlusion (MCAO) reperfusion injury in the mouse. IMS is a powerful method to not only discriminate gangliosides by their oligosaccharide components, but also by their carbon length within their sphingosine base. Mice were subjected to a 30 min unilateral MCAO followed by long-term survival (up to 28 days of reperfusion). Brain sections were sprayed with the matrix 5-Chloro-2-mercaptobenzothiazole, scanned and analyzed for a series of ganglioside molecules using an Applied Biosystems 4800 MALDI TOF/TOF. Traditional histological and immunofluorescence techniques were performed to assess brain tissue damage and verification of the expression of gangliosides of interest. Results revealed a unique anatomical profile of GM1, GD1 and GT1b (d18:1, d20:1 as well as other members of the glycosphingolipid family). There was marked variability in the ratio of expression between ipsilateral and contralateral cortices for the various detected ganglioside species following MCAO-reperfusion injury. Most interestingly, MCAO resulted in the transient induction of both GM2 and GM3 signals within the ipsilateral hemisphere; at the border of the infarcted tissue. Taken together, the data suggest that brain region specific expression of gangliosides, particularly with respect to hydrocarbon length, may play a role in neuronal responses to injury.


Asunto(s)
Encéfalo/metabolismo , Gangliósidos/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Ataque Isquémico Transitorio/metabolismo , Imagen Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Cerebro/metabolismo , Cerebro/patología , Cerebro/fisiopatología , Gangliósidos/química , Regulación de la Expresión Génica , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Especificidad de Órganos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Esfingosina/química , Factores de Tiempo
11.
Proteomics ; 10(18): 3272-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20718007

RESUMEN

Cerebral ischemia (CI) induces dramatic changes in synaptic structure and function that precedes delayed post-ischemic neuronal death. Here, a proteomic analysis was used to identify the effects of focal CI on synaptosomal protein levels. Contralateral and ipsilateral synaptosomes, prepared from adult mice subjected to 60 min middle cerebral artery occlusion, were isolated following 3, 6 and 20 h of reperfusion. Synaptosomal protein samples (n=3) were labeled using the cleavable ICAT system prior to analysis with nanoLC-MS/MS. Each sample was analyzed by LC-MS to identify differential expressions using InDEPT software and differentially expressed peptides were identified by targeted LC-MS/MS. A total of 62 differentially expressed proteins were identified and Gene Ontology classification (cellular component) indicated that the majority of the proteins were located in the mitochondria and other components consistent with synaptic localization. The observed alterations in synaptic protein levels poorly correlated with gene expression, indicating the involvement of post-transcriptional regulatory mechanisms in determining post-ischemic synaptic protein content. Additionally, immunohistochemistry analysis of prosaposin (Psap) and saposin C (SapC) indicates that CI disrupts Psap processing and glycosphingolipid metabolism. These results demonstrate that the synapse is adversely affected by CI and may play a role in mediating post-ischemic neuronal viability.


Asunto(s)
Isquemia Encefálica/metabolismo , Lisosomas/química , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Sinaptosomas/química , Animales , Isquemia Encefálica/genética , Regulación de la Expresión Génica , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteómica , Saporinas , Sinaptosomas/metabolismo
12.
Brain Res ; 1344: 209-16, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20493826

RESUMEN

Membrane rafts, rich in sphingolipids and cholesterol, are membrane microdomains important in neuronal domain-specific signaling events such as during axonal outgrowth and neuronal death. The present study seeks to determine the spatiotemporal association of several axonal guidance signaling molecules with membrane rafts. These molecules are Neuropilin-1 (NRP-1), Fer Kinase, and collapsin response mediator proteins (CRMPs), which are known to have important functions in axonal outgrowth and neuronal death caused by cerebral ischemia. Mice were subjected to sham or a 1h unilateral middle cerebral artery occlusion (MCAO) followed by a time course of reperfusion up to 24h. Brain cortices were separated and membrane rafts were extracted based on their insolubility in Triton X-100 and separation by sucrose gradient fractionation. We demonstrate the early and transient induction of NRP-1 and CRMP-2 in membrane rafts in both ipsilateral and contralateral hemispheres, in contrast to an early, but sustained elevation of Fer kinase and other CRMPs (1, 3, 4, 5) in response to unilateral MCAO. The fact that NRP1/Fer kinase/CRMP-2 co-localize in membrane rafts early during ischemic injury suggests that the membrane rafts may form a scaffold to support and initiate NRP1/Fer/CRMP-2-mediated signal transduction in neuronal damage response during ischemia-reperfusion. Further understanding of the time-specific and membrane domain-specific protein-protein interaction may lead to the identification of therapeutic targets for stroke.


Asunto(s)
Membrana Celular/metabolismo , Lateralidad Funcional/fisiología , Regulación de la Expresión Génica/fisiología , Infarto de la Arteria Cerebral Media/patología , Neuropilina-1/metabolismo , Fosfotransferasas/metabolismo , Semaforina-3A/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Fosfotransferasas/clasificación , Reperfusión/métodos , Factores de Tiempo
13.
J Biol Chem ; 285(13): 9908-9918, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20133938

RESUMEN

Neuropilins (NRPs) are receptors for the major chemorepulsive axonal guidance cue semaphorins (Sema). The interaction of Sema3A/NRP1 during development leads to the collapse of growth cones. Here we show that Sema3A also induces death of cultured cortical neurons through NRP1. A specific NRP1 inhibitory peptide ameliorated Sema3A-evoked cortical axonal retraction and neuronal death. Moreover, Sema3A was also involved in cerebral ischemia-induced neuronal death. Expression levels of Sema3A and NRP1, but not NRP2, were significantly increased early during brain reperfusion following transient focal cerebral ischemia. NRP1 inhibitory peptide delivered to the ischemic brain was potently neuroprotective and prevented the loss of motor functions in mice. The integrity of the injected NRP1 inhibitory peptide into the brain remained unchanged, and the intact peptide permeated the ischemic hemisphere of the brain as determined using MALDI-MS-based imaging. Mechanistically, NRP1-mediated axonal collapse and neuronal death is through direct and selective interaction with the cytoplasmic tyrosine kinase Fer. Fer RNA interference effectively attenuated Sema3A-induced neurite retraction and neuronal death in cortical neurons. More importantly, down-regulation of Fer expression using Fer-specific RNA interference attenuated cerebral ischemia-induced brain damage. Together, these studies revealed a previously unknown function of NRP1 in signaling Sema3A-evoked neuronal death through Fer in cortical neurons.


Asunto(s)
Neuropilina-1/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Semaforina-3A/química , Animales , Encéfalo/metabolismo , Isquemia Encefálica/patología , Muerte Celular , Regulación Enzimológica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuropilina-1/química , Péptidos/química , Unión Proteica , Interferencia de ARN , Transducción de Señal
14.
Neurosci Res ; 66(4): 396-401, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20036291

RESUMEN

Neuropilin 2 (NRP2) is a type I transmembrane protein that binds to distinct members of the class III secreted Semaphorin subfamily. NRP2 plays important roles in repulsive axon guidance, angiogenesis and vasculogenesis through partnering with co-receptors such as vascular endothelial growth factor receptors (VEGFRs) during development. Emerging evidence also suggests that NRP2 contributes to injury response and environment changes in adult brains. In this study, we examined the contribution of NRP2 gene to cerebral ischemia-induced brain injury using NRP2 deficient mouse. To our surprise, the lack of NRP2 expression does not affect the outcome of brain injury induced by transient occlusion of the middle cerebral artery (MCAO) in mouse. The cerebral vasculature in terms of the middle cerebral artery anatomy and microvessel density in the cerebral cortex of NRP2 deficient homozygous (NRP2(-/-)) mice are normal and almost identical to those of the heterozygous (NRP2(+/-)) and wild type (NRP2(+/+)) littermates. MCAO (1h) and 24h reperfusion caused a brain infarction of 23% (compared to the contralateral side) in NRP2(-/-) mice, which is not different from those in NRP2(+/- and +/+) mice at 22 and 21%, respectively (n=19, p>0.05). Correspondingly, NRP2(-/-) mouse also showed a similar level of deterioration of neurological functions after stroke compared with their NRP2(+/- and +/+) littermates. Oxygen-glucose-deprivation (OGD) caused a significant neuronal death in NRP2(-/-) cortical neurons, at the level similar to that in NRP(+/+) cortical neurons (72% death in NRP(-/-) neurons vs. 75% death in NRP2(+/+) neurons; n=4; p>0.05). Together, these loss-of-function studies demonstrated that despite of its critical role in neuronal guidance and vascular formation during development, NRP2 expression dose not affect adult brain response to cerebral ischemia.


Asunto(s)
Corteza Cerebral/patología , Glucosa/deficiencia , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/patología , Neuronas/patología , Neuropilina-2/genética , Oxígeno/fisiología , Animales , Infarto Encefálico/etiología , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Supervivencia Celular , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Ataque Isquémico Transitorio/etiología , Ataque Isquémico Transitorio/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropilina-2/biosíntesis
15.
J Neurochem ; 111(3): 870-81, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19735446

RESUMEN

Intracellular calcium influx through NMDA receptors triggers a cascade of deleterious signaling events which lead to neuronal death in neurological conditions such as stroke. However, it is not clear as to the molecular mechanism underlying early damage response from axons and dendrites which are important in maintaining a network essential for the survival of neurons. Here, we examined changes of axons treated with glutamate and showed the appearance of betaIII-tubulin positive varicosities on axons before the appearance of neuronal death. Dizocilpine blocked the occurrence of varicosities on axons suggesting that these microstructures were mediated by NMDA receptor activities. Despite early increased expression of pCaMKII and pMAPK after just 10 min of glutamate treatment, only inhibitors to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and calpain prevented the occurrence of axonal varicosities. In contrast, inhibitors to Rho kinase, mitogen-activated protein kinase and phosphoinositide 3-kinase were not effective, nor were they able to rescue neurons from death, suggesting CaMKII and calpain are important in axon survival. Activated CaMKII directly phosphorylates collapsin response mediator protein (CRMP) 2 which is independent of calpain-mediated cleavage of CRMP2. Over-expression of CRMP2, but not the phosphorylation-resistant mutant CRMP2-T555A, increased axonal resistance to glutamate toxicity with reduced numbers of varicosities. The levels of both pCRMP2 and pCaMKII were also increased robustly within early time points in ischemic brains and which correlated with the appearance of axonal varicosities in the ischemic neurons. Collectively, these studies demonstrated an important role for CaMKII in modulating the integrity of axons through CRMP2 during excitotoxicity-induced neuronal death.


Asunto(s)
Axones/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ácido Glutámico/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas Fluorescentes Verdes/genética , Infarto de la Arteria Cerebral Media/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Semaforina-3A/farmacología , Transducción de Señal/efectos de los fármacos , Transfección/métodos , Tubulina (Proteína)/metabolismo
16.
Brain Res ; 1269: 185-97, 2009 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-19285050

RESUMEN

Vascular basement membrane (BM) stabilizes brain vessels and inhibits endothelial cell cycle. Cerebral ischemia causes BM breakdown with the loss of structural BM components including collagens and laminins. In this study, the expression changes of the BM proteoglycan agrin, and the non-structural BM constituent SPARC (BM-40, osteonectin), were studied in brain vessels after global cerebral ischemia. A transient 20-min forebrain ischemia followed by 1, 6 or 24 h of reperfusion was induced in adult Sprague-Dawley rats by combined bilateral common carotid artery occlusion and hypotension (42-45 mm Hg). In a separate group of animals, a mild (32 degrees C) post-ischemic hypothermia was induced for 6 h, starting immediately after ischemia. RNA from approximately 500 brain vessels (20-100 microm) extracted by laser-capture microdissection (LCM) microscopy was used to determine the expression of proteoglycans agrin and SPARC mRNAs by quantitative PCR (Q-PCR). Protein expression was determined by immunohistochemistry in adjacent tissue sections. The BBB permeability was assessed using (3)H-sucrose as an in vivo tracer and by examining fibrinogen immunoreactivity in tissue sections. A transient global brain ischemia resulted in a significant (ANOVA, p<0.05; 6 animals/group) reduction in agrin and SPARC mRNAs in LCM-captured brain vessels 24 h after reperfusion. A time-dependent loss of agrin and SPARC from the BM during reperfusion was also observed by immunochemistry. A 6-h post-ischemic hypothermia reduced SPARC and agrin mRNA and protein losses, BBB transfer constant for (3)H-sucrose as well as fibrinogen extravasation 24 h after reperfusion. It is conluded that a transient post-ischemic hypothermia stabilizes brain vessels and reduces BBB disruption in part by preventing proteolytic degradation of regulatory BM constituents, SPARC and agrin.


Asunto(s)
Agrina/genética , Barrera Hematoencefálica/fisiología , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Hipotermia Inducida , Osteonectina/genética , Agrina/metabolismo , Animales , Membrana Basal/patología , Membrana Basal/fisiología , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Encéfalo/fisiología , Femenino , Fibrinógeno/metabolismo , Laminina/metabolismo , Masculino , Osteonectina/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sacarosa/farmacocinética , Tritio
17.
Biochem Biophys Res Commun ; 367(1): 109-15, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18162177

RESUMEN

Strategies to provide neuroprotection and to promote regenerative axonal outgrowth in the injured brain are thwarted by the plethora of axon growth inhibitors and the ligand promiscuity of some of their receptors. Especially, new neurons derived from ischemia-stimulated neurogenesis must integrate this multitude of inhibitory molecular cues, generated as a result of cortical damage, into a functional response. More often than not the response is one of growth cone collapse, axonal retraction and neuronal death. Therefore, characterization of the expression of inhibitory molecules in long-term surviving ischemic brains following stroke is important for designing selective therapeutics. Here, we describe a long-term recovery mouse model for cerebral ischemia in which a brief transient occlusion of the middle cerebral artery (30min) was followed by up to 30 days of long-term reperfusion. Significantly decreased grip strength motor function and increased expression of one of the major repulsive guidance cues, Semaphorin 3A (Sema3A) and its receptor Neuropilin1 (NRP1) occurred in brains of these mice. Interestingly, increased Doublecortin (DCX) expression occurred only in the lateral ventricular wall zone, but not in the dentate gyrus granule cell layer on the ischemic side of the brain. Importantly, no DCX positive cells were detected in the infarct core region after 30d ischemic recovery. Collectively, these studies demonstrated the sustained elevation of Sema3A/NRP1 expression in the ischemic territory, which may contribute to the inhibitory microenvironment responsible for preventing new neurons from entering the infarct area. This model will be of use as a platform for testing anti-inhibitory therapies to stroke.


Asunto(s)
Isquemia Encefálica , Regulación de la Expresión Génica/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Regulación de la Expresión Génica/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Modelos Biológicos , Neuropéptidos/genética , Neuropilina-1/genética , Semaforina-3A/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
18.
J Neurosci Methods ; 168(2): 443-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18155299

RESUMEN

The fatty acid salt, sodium caprate (C10) is a well recognized drug absorption enhancer in intestine because of its ability to widen tight junctions in the epithelial cell lining. Caprate's potential usefulness to similarly alter the blood-brain barrier (BBB) tight junctions of brain vasculature and enhance CNS drug delivery has undergone little investigation. Adult SD rats were anesthetized and C10 was infused into the left internal carotid artery (dosing parameters: 10-30 mM, 1 or 2 ml min(-1), for 0.5-1.5 min). Beginning 5 or 60 min after infusion an i.v. bolus of [3H]mannitol was allowed to circulate for 30 min and degree of BBB leakiness measured as magnitude of the transfer constant (Ki, nl g(-1)s(-1)) for blood to brain mannitol permeation determined from brain and plasma samples. In initial experiments identical C10 infusions caused dramatic BBB opening in some rats, e.g., 10-fold increase in Ki, but not in others. Higher dosing produced consistent opening measured 5-35 or 60-90 min post-infusion but was also toxic as shown by severe brain edema and cardio-respiratory failure. The variable effect of moderate doses was attributed to the fact that arterial blood pressure markedly increased during C10 infusion and may have altered the flow dynamics of cerebrovascular caprate distribution from rat to rat. We modified the procedure by temporarily withdrawing blood to produce hypovolemia and systemic arterial hypotension during C10 infusion. Caprate infusions of 15-25 mM, 2 ml min(-1) for 1 min, produced reliable dose-related openings that lasted as much as an hour, were reversible, and accompanied by little or moderate edema, depending on dose. These findings confirm an earlier report showing that intracarotid caprate infusion opens the BBB but also show that control of the temporary hypertensive response produced by intracarotid caprate infusion is key to tailoring the dosage to consistently achieve graded, reversible BBB opening.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Arterias Carótidas/fisiología , Ácidos Decanoicos/administración & dosificación , Ácidos Decanoicos/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Ácidos Decanoicos/toxicidad , Diuréticos , Relación Dosis-Respuesta a Droga , Hipovolemia/fisiopatología , Infusiones Intraarteriales , Inulina , Masculino , Manitol , Ratas , Ratas Sprague-Dawley , Uniones Estrechas/efectos de los fármacos
19.
Mol Imaging ; 7(6): 248-62, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19123995

RESUMEN

The blood-brain barrier (BBB) disruption following cerebral ischemia can be exploited to deliver imaging agents and therapeutics into the brain. The aim of this study was (a) to establish novel in vivo optical imaging methods for longitudinal assessment of the BBB disruption and (b) to assess size selectivity and temporal patterns of the BBB disruption after a transient focal ischemia. The BBB permeability was assessed using in vivo time domain near-infrared optical imaging after contrast enhancement with either free Cy5.5 (1 kDa) or Cy5.5 conjugated with bovine serum albumin (BSA) (67 kDa) in mice subjected to either 60- or 20-minute transient middle cerebral artery occlusion (MCAO) and various times of reperfusion (up to 14 days). In vivo imaging observations were corroborated by ex vivo brain imaging and microscopic analyses of fluorescent tracer extravasation. The in vivo optical contrast enhancement with Cy5.5 was spatially larger than that observed with BSA-Cy5.5. Longitudinal studies after a transient 20-minute MCAO suggested a bilateral BBB disruption, more pronounced in the ipsilateral hemisphere, peaking at day 7 and resolving at day 14 after ischemia. The area differential between the BBB disruption for small and large molecules could potentially be useful as a surrogate imaging marker for assessing perinfarct tissues to which neuroprotective therapies of appropriate sizes could be delivered.


Asunto(s)
Barrera Hematoencefálica/patología , Arteria Cerebral Media/patología , Accidente Cerebrovascular/patología , Animales , Permeabilidad de la Membrana Celular , Circulación Coronaria , Modelos Animales de Enfermedad , Cinética , Masculino , Ratones , Microscopía Fluorescente , Reperfusión
20.
J Cereb Blood Flow Metab ; 28(1): 99-110, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17519975

RESUMEN

Synaptic pathology is observed during hypoxic events in the central nervous system in the form of altered dendrite structure and conductance changes. These alterations are rapidly reversible, on the return of normoxia, but are thought to initiate subsequent neuronal cell death. To characterize the effects of hypoxia on regulators of synaptic stability, we examined the temporal expression of cell adhesion molecules (CAMs) in synaptosomes after transient middle cerebral artery occlusion (MCAO) in mice. We focused on events preceding the onset of ischemic neuronal cell death (<48 h). Synaptosome preparations were enriched in synaptically localized proteins and were free of endoplasmic reticulum and nuclear contamination. Electron microscopy showed that the synaptosome preparation was enriched in spheres (approximately 650 nm in diameter) containing secretory vesicles and postsynaptic densities. Forebrain mRNA levels of synaptically located CAMs was unaffected at 3 h after MCAO. This is contrasted by the observation of consistent downregulation of synaptic CAMs at 20 h after MCAO. Examination of synaptosomal CAM protein content indicated that certain adhesion molecules were decreased as early as 3 h after MCAO. For comparison, synaptosomal Agrn protein levels were unaffected by cerebral ischemia. Furthermore, a marked increase in the levels of p-Ctnnb1 in ischemic synaptosomes was observed. p-Ctnnb1 was detected in hippocampal fiber tracts and in cornu ammonis 1 neuronal nuclei. These results indicate that ischemia induces a dysregulation of a subset of synaptic proteins that are important regulators of synaptic plasticity before the onset of ischemic neuronal cell death.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Vesículas Secretoras/metabolismo , Sinapsis/metabolismo , Sinaptosomas/metabolismo , Agrina/metabolismo , Animales , Muerte Celular , Hipocampo/metabolismo , Hipocampo/ultraestructura , Infarto de la Arteria Cerebral Media/patología , Ratones , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/ultraestructura , Prosencéfalo/metabolismo , Prosencéfalo/ultraestructura , ARN Mensajero/metabolismo , Vesículas Secretoras/ultraestructura , Sinapsis/ultraestructura , Sinaptosomas/ultraestructura , Factores de Tiempo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...