Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Rep ; 10(1): 21252, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277513

RESUMEN

Neurovascular injury has been proposed as a universal pathological hallmark of traumatic brain injury (TBI) with molecular markers of angiogenesis and endothelial function associated with injury severity and morbidity. Sex differences in the neurovasculature response post-TBI may contribute to the differences seen in how males and females respond to injury. Steady-state contrast enhanced magnetic resonance imaging (SSCE-MRI) can be used to non-invasively assess the neurovasculature and may be a useful tool in understanding and predicting outcomes post-TBI. Here we used SSCE-MRI to investigate the neurovasculature of male and female rats at 48 h after an experimental TBI, and how these changes related to neuromotor function at 1-week post-TBI. In addition to TBI induced changes, we found that female rats had greater vessel density, greater cerebral blood volumes and performed better on a neuromotor task than their male counterparts. These results suggest that acute post-TBI cerebrovascular function is worse in males, and that this may contribute to the greater functional deficits observed post-injury. Furthermore, these results highlight the potential of SSCE-MRI to provide insights into the cerebral microvasculature post-TBI. Future studies, incorporating both males and females, are warranted to investigate the evolution of these changes and the underlying mechanisms.


Asunto(s)
Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Animales , Encéfalo/diagnóstico por imagen , Modelos Animales de Enfermedad , Femenino , Espectroscopía de Resonancia Magnética , Masculino , Ratas , Ratas Long-Evans
2.
Microcirculation ; 27(7): e12646, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32608116

RESUMEN

OBJECTIVE: It is not known how activation of the hypoxia-inducible factor (HIF) pathway in pericytes, cells of the microvascular wall, influences new capillary growth. We tested the hypothesis that HIF-activated pericytes promote angiogenesis in a neonatal model of spinal cord injury (SCI). METHODS: Human placental pericytes stimulated with cobalt chloride and naïve pericytes were injected into the site of a thoracic hemi-section of the spinal cord in rat pups on postnatal day three (P3). Hindlimb motor recovery and Doppler blood flow perfusion at the site of transection were measured on P10. Immunohistochemistry was used to visualize vessel and neurofilament density for quantification. RESULTS: Injection of HIF-activated pericytes resulted in greater vascular density in males but did not result in improved motor function for males or females. Injection of non-HIF-activated pericytes resulted improved motor function recovery in both sexes (males, 2.722 ± 0.31-fold score improvement; females, 3.824 ± 0.58-fold score improvement, P < .05) but produced no significant changes in vessel density. CONCLUSIONS: HIF-activated pericytes promote vascular density in males post-SCI. Acute delivery of non-HIF-activated pericytes at the site of injury can improve motor recovery post-SCI.


Asunto(s)
Pericitos/fisiología , Pericitos/trasplante , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Animales , Animales Recién Nacidos , Velocidad del Flujo Sanguíneo , Proliferación Celular , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Miembro Posterior , Humanos , Locomoción/fisiología , Masculino , Neovascularización Fisiológica , Ratas , Recuperación de la Función/fisiología , Factores Sexuales , Médula Espinal/irrigación sanguínea , Médula Espinal/patología , Traumatismos de la Médula Espinal/rehabilitación
3.
Brain ; 139(Pt 7): 1919-38, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27289302

RESUMEN

There are no treatments in clinical practice known to mitigate the neurobiological processes that convert a healthy brain into an epileptic one, a phenomenon known as epileptogenesis. Downregulation of protein phosphatase 2A, a protein that causes the hyperphosphorylation of tau, is implicated in neurodegenerative diseases commonly associated with epilepsy, such as Alzheimer's disease and traumatic brain injury. Here we used the protein phosphatase 2A activator sodium selenate to investigate the role of protein phosphatase 2A in three different rat models of epileptogenesis: amygdala kindling, post-kainic acid status epilepticus, and post-traumatic epilepsy. Protein phosphatase 2A activity was decreased, and tau phosphorylation increased, in epileptogenic brain regions in all three models. Continuous sodium selenate treatment mitigated epileptogenesis and prevented the biochemical abnormalities, effects which persisted after drug withdrawal. Our studies indicate that limbic epileptogenesis is associated with downregulation of protein phosphatase 2A and the hyperphosphorylation of tau, and that targeting this mechanism with sodium selenate is a potential anti-epileptogenic therapy.


Asunto(s)
Anticonvulsivantes/farmacología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Proteína Fosfatasa 2/metabolismo , Ácido Selénico/farmacología , Proteínas tau/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/complicaciones , Electroencefalografía , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Epilepsia/etiología , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/farmacología , Excitación Neurológica , Imagen por Resonancia Magnética , Masculino , Fosforilación , Proteína Fosfatasa 2/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas tau/efectos de los fármacos
4.
Am J Physiol Cell Physiol ; 309(9): C600-7, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26289751

RESUMEN

Endothelial cell migration is a fundamental process during angiogenesis and, therefore, a point of intervention for therapeutic strategies aimed at controlling pathologies involving blood vessel growth. We sought to determine the role of the gap junction protein connexin 43 (Cx43) in key features of angiogenesis in the central nervous system. We used an in vitro model to test the hypothesis that a complex of interacting proteins, including Cx43 and zonula occludens-1 (ZO-1), regulates the migratory behavior of cerebral endothelium. With knockdown and overexpression experiments, we demonstrate that the rate of healing following scrape-wounding of endothelium is regulated by the level of Cx43 protein expression. The effects on cell motility and proliferation were independent of gap junction communication as cells were sensitive to altered Cx43 expression in single plated cells. Coupling of Cx43/ZO-1 critically regulates this process as demonstrated with the use of a Cx43 α-carboxy terminus 1 peptide mimetic (αCT1) and overexpression of a mutant ZO-1 with the Cx43-binding PDZ2 domain deleted. Disrupting the Cx43/ZO-1 complex with these treatments resulted in collapse of the organized F-actin cytoskeleton and the appearance of actin nodes. Preincubation with the myosin 2 inhibitors blebbistatin or Y-27632 disrupted the Cx43/ZO-1 complex and inhibited cell spreading at the leading edge of migration. Cells studied individually in time-lapse open field locomotion assays wandered less when Cx43/ZO-1 interaction was disrupted without significant change in speed, suggesting that faster wound healing is a product of linearized migration. In contrast to the breakdown of F-actin architecture, microtubule architecture was not obviously affected by treatments. This study provides new insight into the fundamental regulatory mechanisms of cerebral endothelial cell locomotion. Cx43 tethers the F-actin cytoskeleton through a ZO-1 linker and supports cell spreading and exploration during locomotion. Here, we demonstrate that releasing this actin-coupled tether shifts the balance of directional migration control to a more linear movement that enhances the rate of wound healing.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Encéfalo/irrigación sanguínea , Movimiento Celular , Forma de la Célula , Conexina 43/metabolismo , Células Endoteliales/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Animales , Sitios de Unión , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Conexina 43/genética , Células Endoteliales/efectos de los fármacos , Ratones , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , Transducción de Señal , Transfección , Cicatrización de Heridas
5.
Microcirculation ; 22(8): 711-23, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26243428

RESUMEN

OBJECTIVES: The mechanisms involved in activating pericytes, cells that ensheath capillaries, to engage in the formation of new capillaries, angiogenesis, remain unknown. In this study, the hypothesis was tested that pericytes could be stimulated to promote angiogenesis by driving the HIF pathway. METHODS: Pericytes were stimulated with CoCl2 to activate the HIF pathway. Stimulated pericytes were cocultured with endothelial cells in a wound healing assay and in a 3D collagen matrix assay of angiogenesis. A culture system of spinal cord tissue was used to assess microvascular outcomes after treatment with stimulated pericytes. Pharmaceutical inhibition of exosome production was also performed. RESULTS: Treatment with stimulated pericytes resulted in faster wound healing (1.92 ± 0.18 fold increase, p < 0.05), greater endothelial cord formation (2.9 ± 0.14 fold increase, p < 0.05) in cell culture assays, and greater vascular density (1.78 ± 0.23 fold increase, p < 0.05) in spinal cord tissue. Exosome secretion and the physical presence of stimulated pericytes were necessary in the promotion of angiogenic outcomes. CONCLUSIONS: These results elucidate a mechanism that may be exploited to enhance features of angiogenesis in the CNS.


Asunto(s)
Capilares/metabolismo , Exosomas/metabolismo , Neovascularización Fisiológica , Pericitos/metabolismo , Capilares/citología , Hipoxia de la Célula , Humanos , Pericitos/citología
6.
Am J Pathol ; 184(9): 2573-85, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25016930

RESUMEN

Mild to moderate hyperhomocysteinemia is prevalent in humans and is implicated in neurovascular diseases, including recently in certain retinal diseases. Herein, we used hyperhomocysteinemic mice deficient in the Cbs gene encoding cystathionine-ß-synthase (Cbs(+/-)) to evaluate retinal vascular integrity. The Cbs(+/+) (wild type) and Cbs(+/-) (heterozygous) mice (aged 16 to 52 weeks) were subjected to fluorescein angiography and optical coherence tomography to assess vasculature in vivo. Retinas harvested for cryosectioning or flat mount preparations were subjected to immunofluorescence microscopy to detect blood vessels (isolectin-B4), angiogenesis [anti-vascular endothelial growth factor (VEGF) and anti-CD105], gliosis [anti-glial fibrillary acidic protein (GFAP)], pericytes (anti-neural/glial antigen 2), blood-retinal barrier [anti-zonula occludens protein 1 (ZO-1) and anti-occludin], and hypoxia [anti-pimonidazole hydrochloride (Hypoxyprobe-1)]. Levels of VEGF, GFAP, ZO-1, and occludin were determined by immunoblotting. Results of these analyses showed a mild vascular phenotype in young mice, which progressed with age. Fluorescein angiography revealed progressive neovascularization and vascular leakage in Cbs(+/-) mice; optical coherence tomography confirmed new vessels in the vitreous by 1 year. Immunofluorescence microscopy demonstrated vascular patterns consistent with ischemia, including a capillary-free zone centrally and new vessels with capillary tufts midperipherally in older mice. This was associated with increased VEGF, CD105, and GFAP and decreased ZO-1/occludin levels in the Cbs(+/-) retinas. Retinal vein occlusion was observed in some Cbs(+/-) mouse retinas. We conclude that mild to moderate elevation of homocysteine in Cbs(+/-) mice is accompanied by progressive alterations in retinal vasculature characterized by ischemia, neovascularization, incompetent blood-retinal barrier, and vascular occlusion.


Asunto(s)
Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Hiperhomocisteinemia/patología , Vasos Retinianos/patología , Animales , Heterocigoto , Hiperhomocisteinemia/genética , Ratones , Ratones Mutantes , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa , Neovascularización Retiniana/genética , Neovascularización Retiniana/patología
7.
Microcirculation ; 21(6): 541-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24655004

RESUMEN

OBJECTIVE: To test the hypothesis that Hcy impairs angiogenic outgrowth through an iNOS-dependent mechanism. METHODS: Adult C57Bl/6 mouse choroid explants were used in angiogenic outgrowth assays. Mouse microvascular endothelial cells were studied in culture during scrape-induced migration and dispersed cell locomotion experiments. Activity of iNOS was manipulated with pharmacology (1400 W), siRNA, and by use of choroid explants from iNOS knockout mice (iNOS(-/-)). RESULTS: Hcy (20 µM) significantly decreased the area of endothelial outgrowth without altering the number of cells in the choroid explant angiogenic assay, resulting in more densely packed outgrowth. Hcy prevented the outward orientation of actin filaments and decreased the number of actin projections along the leading edge of outgrowth. Hcy also slowed outgrowth from the edge of a scraped endothelial monolayer and in cultures of dispersed cells, Hcy impaired cell locomotion without affecting proliferation. Inhibition of iNOS activity rescued the effect of Hcy on area of explant outgrowth, cell density, number of projections, cell locomotion, and rate of outgrowth following scraping. CONCLUSIONS: Hcy impairs microvascular endothelial outgrowth, but not proliferation, by disrupting cell locomotion through an iNOS-dependent mechanism.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Endotelio Vascular/enzimología , Homocisteína/farmacología , Microvasos/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Masculino , Ratones , Ratones Noqueados , Microvasos/citología , Óxido Nítrico Sintasa de Tipo II/genética
8.
PLoS One ; 8(5): e63951, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696861

RESUMEN

Hyperhomocysteinemia (HHcy) is a risk factor for cognitive impairment. The purpose of this study was to determine the temporal pattern of cerebral pathology in a mouse model of mild HHcy, because understanding this time course provides the basis for understanding the mechanisms involved. C57Bl/6 mice with heterozygous deletion cystathionine ß-synthase (cbs (+/-); Het) were used as a model of mild HHcy along with their wild-type littermates (cbs (+/+); WT). Mice were 'young' (5.3±0.2 months of age) and 'old' (16.6±0.9 months of age). Blood-brain barrier (BBB) permeability was quantified from Evans blue and sodium fluorescein extravasation. Microvascular architecture was assessed by z-stack confocal microscopy. Leukoaraiosis was measured from Luxol fast blue stained slides of paraffin brain sections. Inflammation was quantified using standard antibody-based immunohistochemical techniques. Cognitive function was assessed using the Morris water maze. BBB permeability was significantly greater in Het vs. WT mice at all ages (p<0.05). There were no differences in microvascular architecture among the groups. Compared with all other groups, old Het mice had significantly greater leukoaraiosis, inflammation in the fornix, and cognitive impairment (p<0.05). In mild HHcy, increased permeability of the BBB precedes the onset of cerebral pathology. This new paradigm may play a role in the progression of disease in HHcy.


Asunto(s)
Barrera Hematoencefálica/patología , Encéfalo/patología , Hiperhomocisteinemia/fisiopatología , Animales , Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Azul de Evans/química , Hiperhomocisteinemia/patología , Leucoaraiosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal
9.
J Cereb Blood Flow Metab ; 32(5): 825-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22186670

RESUMEN

Hyperhomocysteinemia (HHcy) disrupts nitric oxide (NO) signaling and increases nitrative stress in cerebral microvascular endothelial cells (CMVECs). This is mediated, in part, by protein nitrotyrosinylation (3-nitrotyrosine; 3-NT) though the mechanisms by which extracellular homocysteine (Hcy) generates intracellular 3-NT are unknown. Using a murine model of mild HHcy (cbs(+/-) mouse), we show that 3-NT is significantly elevated in cerebral microvessels with concomitant reductions in serum NO bioavailability as compared with wild-type littermate controls (cbs(+/+)). Directed pharmacology identified a receptor-dependent mechanism for 3-NT formation in CMVECs. Homocysteine increased expression of inducible NO synthase (iNOS) and formation of 3-NT, both of which were blocked by inhibition of metabotropic glutamate receptor-5 (mGluR5) with the specific antagonist 2-methyl-6-(phenylethynyl) pyridine hydrochloride. Activation of mGluR5 is both sufficient and necessary to drive the nitrative stress because direct activation using the mGluR5-specific agonist (RS)-2-chloro-5-hydroxyphenylglycine also increased iNOS expression and 3-NT formation while knockdown of mGluR5 receptor expression by short hairpin RNA (shRNA) blocked their increase in response to Hcy. Nitric oxide derived from iNOS was required for Hcy-mediated formation of 3-NT because the effect was blocked by 1400W. These results provide the first evidence for a receptor-dependent process that explains how plasma Hcy levels control intracellular nitrative stress in cerebral microvascular endothelium.


Asunto(s)
Encéfalo/metabolismo , Endotelio/metabolismo , Hiperhomocisteinemia/metabolismo , Óxido Nítrico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Estrés Fisiológico , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Encéfalo/irrigación sanguínea , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Glicina/análogos & derivados , Glicina/farmacología , Homocisteína/genética , Homocisteína/metabolismo , Hiperhomocisteinemia/genética , Ratones , Ratones Noqueados , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Fenilacetatos/farmacología , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/genética , Tirosina/análogos & derivados , Tirosina/genética , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA