Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci Res ; 98(1): 121-128, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30667078

RESUMEN

Currently, there is no effective treatment for germinal matrix hemorrhage and intraventricular hemorrhage (GMH-IVH), a common and often fatal stroke subtype in premature infants. Secondary brain injury after GMH-IVH is known to involve blood clots that contribute to inflammation and neurological deficits. Furthermore, the subsequent blood clots disrupt normal cerebrospinal fluid circulation and absorption after GMH-IVH, contributing to posthemorrhagic hydrocephalus (PHH). Clinically, GMH-IVH severity is graded on a I to IV scale: Grade I is confined to the germinal matrix, grade II includes intraventricular hemorrhage, grade III includes intraventricular hemorrhage with extension into dilated ventricles, and grade IV includes intraventricular hemorrhage with extension into dilated ventricles as well as parenchymal hemorrhaging. GMH-IVH hematoma volume is the best prognostic indicator, where patients with higher grades have worsened outcomes. Various preclinical studies have shown that rapid hematoma resolution quickly ameliorates inflammation and improves neurological outcomes. Current experimental evidence identifies alternatively activated microglia as playing a pivotal role in hematoma clearance. In this review, we discuss the pathophysiology of GMH-IVH in the development of PHH, microglia/macrophage's role in the neonatal CNS, and established/potential therapeutic targets that enhance M2 microglia/macrophage phagocytosis of blood clots after GMH-IVH.


Asunto(s)
Encéfalo/metabolismo , Hemorragias Intracraneales/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Encéfalo/patología , Humanos , Hidrocefalia/etiología , Hidrocefalia/metabolismo , Hidrocefalia/patología , Recién Nacido , Recien Nacido Prematuro , Hemorragias Intracraneales/complicaciones , Hemorragias Intracraneales/patología , Macrófagos/patología , Microglía/patología
2.
J Neurosci Res ; 98(1): 105-120, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30793349

RESUMEN

In addition to being the leading cause of morbidity and mortality in premature infants, germinal matrix hemorrhage (GMH) is also the leading cause of acquired infantile hydrocephalus. The pathophysiology of posthemorrhagic hydrocephalus (PHH) development after GMH is complex and vaguely understood, although evidence suggests fibrosis and gliosis in the periventricular and subarachnoid spaces disrupts normal cerebrospinal fluid (CSF) dynamics. Theories explaining general hydrocephalus etiology have substantially evolved from the original bulk flow theory developed by Dr. Dandy over a century ago. Current clinical and experimental evidence supports a new hydrodynamic theory for hydrocephalus development involving redistribution of vascular pulsations and disruption of Starling forces in the brain microcirculation. In this review, we discuss CSF flow dynamics, history and development of theoretical hydrocephalus pathophysiology, and GMH epidemiology and etiology as it relates to PHH development. We highlight known mechanisms and propose new avenues that will further elucidate GMH pathophysiology, specifically related to hydrocephalus.


Asunto(s)
Plexo Coroideo/metabolismo , Hidrocefalia/metabolismo , Hemorragias Intracraneales/metabolismo , Transducción de Señal/fisiología , Plexo Coroideo/patología , Humanos , Hidrocefalia/etiología , Hidrocefalia/patología , Lactante , Recién Nacido , Recien Nacido Prematuro , Hemorragias Intracraneales/complicaciones , Hemorragias Intracraneales/patología
3.
J Neurosci Res ; 98(1): 168-178, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31157469

RESUMEN

Spontaneous intracerebral hemorrhage (ICH) is the deadliest stroke subtype and neuroinflammation is a critical component of the pathogenesis following ICH. Annexin A1-FPR2 signaling has been shown to play a protective role in animal stroke models. This study aimed to assess whether Annexin A1 attenuated neuroinflammation and brain edema after ICH and investigate the underlying mechanisms. Male CD-1 mice were subjected to collagenase-induced ICH. Annexin A1 was administered at 0.5 hr after ICH. Brain water content measurement, short-term and long-term neurobehavioral tests, Western blot and immnunofluorescence were performed. Results showed that Annexin A1 effectively attenuated brain edema, improved short-term neurological function and ameliorated microglia activation after ICH. Annexin A1 also improved memory function at 28 days after ICH. However, these beneficial effects were abolished with the administration of FPR2 antagonist Boc-2. Furthermore, AnxA1/FPR2 signaling may confer protective effects via inhibiting p38-associated inflammatory cascade. Our study demonstrated that Annexin A1/FPR2/p38 signaling pathway played an important role in attenuating neuroinflammation after ICH and that Annexin A1 could be a potential therapeutic strategy for ICH patients.


Asunto(s)
Anexina A1/farmacología , Edema Encefálico/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Receptores de Formil Péptido/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anexina A1/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Colagenasas , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo
4.
Cereb Cortex ; 29(8): 3482-3495, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-30192926

RESUMEN

Intraventricular hemorrhage (IVH) is a common complication of prematurity in infants born at 23-28 weeks of gestation. Survivors exhibit impaired growth of the cerebral cortex and neurodevelopmental sequeale, but the underlying mechanism(s) are obscure. Previously, we have shown that neocortical neurogenesis continues until at least 28 gestational weeks. This renders the prematurely born infants vulnerable to impaired neurogenesis. Here, we hypothesized that neurogenesis is impaired by IVH, and that signaling through GSK3ß, a critical intracellular kinase regulated by Wnt and other pathways, mediates this effect. These hypotheses were tested observationally in autopsy specimens from premature infants, and experimentally in a premature rabbit IVH model. Significantly, in premature infants with IVH, the number of neurogenic cortical progenitor cells was reduced compared with infants without IVH, indicating acutely decreased neurogenesis. This finding was corroborated in the rabbit IVH model, which further demonstrated reduction of upper layer cortical neurons after longer survival. Both the acute reduction of neurogenic progenitors, and the subsequent decrease of upper layer neurons, were rescued by treatment with AR-A014418, a specific inhibitor of GSK3ß. Together, these results indicate that IVH impairs late stages of cortical neurogenesis, and suggest that treatment with GSK3ß inhibitors may enhance neurodevelopment in premature infants with IVH.


Asunto(s)
Apoptosis/efectos de los fármacos , Hemorragia Cerebral Intraventricular/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Tiazoles/farmacología , Urea/análogos & derivados , Animales , Western Blotting , Estudios de Casos y Controles , Recuento de Células , Proliferación Celular , Corteza Cerebral , Hemorragia Cerebral Intraventricular/patología , Modelos Animales de Enfermedad , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Humanos , Inmunohistoquímica , Recien Nacido Extremadamente Prematuro , Recién Nacido , Antígeno Ki-67/metabolismo , Ventrículos Laterales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Factor de Transcripción PAX6/metabolismo , Fosforilación , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción SOXB1/metabolismo , Proteínas de Dominio T Box/metabolismo , Urea/farmacología , Sustancia Blanca
5.
Cereb Cortex ; 29(12): 4932-4947, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30877788

RESUMEN

Preterm-born children suffer from neurological and behavioral disorders. Herein, we hypothesized that premature birth and non-maternal care of preterm newborns might disrupt neurobehavioral function, hippocampal dendritic arborization, and dendritic spine density. Additionally, we assessed whether 17ß-estradiol (E2) replacement or the TrkB receptor agonist, 7,8-dihydroxyflavone (DHF), would reverse compromised dendritic development and cognitive function in preterm newborns. These hypotheses were tested by comparing preterm (E28.5) rabbit kits cared and gavage-fed by laboratory personnel and term-kits reared and breast-fed by their mother doe at an equivalent postconceptional age. Neurobehavioral tests showed that both premature-birth and formula-feeding with non-maternal care led to increased anxiety behavior, poor social interaction, and lack of novelty preference compared with term-kits. Dendritic branching and number of total or mushroom dendritic spines were reduced in the CA1 field of preterm-kits compared with term controls. While CDC42 and Rac1/2/3 expression levels were lower, RhoA-activity was higher in preterm-kits compared with term controls. Both E2 and DHF treatment reversed prematurity-induced reduction in spine density, reduced total RhoA-GTPase levels, and enhanced cognitive function. Hence, prematurity and non-maternal care result in cognitive deficits, and reduced dendritic arbors and spines in CA1. E2 replacement or DHF treatment might reverse changes in dendritic spines and improve neurodevelopment in premature infants.


Asunto(s)
Cognición/fisiología , Espinas Dendríticas/patología , Estradiol/farmacología , Hipocampo/patología , Nacimiento Prematuro/fisiopatología , Receptor trkB/agonistas , Animales , Cognición/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Estrógenos/farmacología , Femenino , Flavonas/farmacología , Hipocampo/efectos de los fármacos , Privación Materna , Embarazo , Nacimiento Prematuro/patología , Conejos , Receptor trkB/efectos de los fármacos
6.
J Neurosci ; 38(34): 7378-7391, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30037831

RESUMEN

Development of cortical interneurons continues until the end of human pregnancy. Premature birth deprives the newborns from the supply of maternal estrogen and a secure intrauterine environment. Indeed, preterm infants suffer from neurobehavioral disorders. This can result from both preterm birth and associated postnatal complications, which might disrupt recruitment and maturation of cortical interneurons. We hypothesized that interneuron subtypes, including parvalbumin-positive (PV+), somatostatin-positive (SST+), calretinin-positive (CalR+), and neuropeptide Y-positive (NPY+) interneurons, were recruited in the upper and lower cortical layers in a distinct manner with advancing gestational age. In addition, preterm birth would disrupt the heterogeneity of cortical interneurons, which might be reversed by estrogen treatment. These hypotheses were tested by analyzing autopsy samples from premature infants and evaluating the effect of estrogen supplementation in prematurely delivered rabbits. The PV+ and CalR+ neurons were abundant, whereas SST+ and NPY+ neurons were few in cortical layers of preterm human infants. Premature birth of infants reduced the density of PV+ or GAD67+ neurons and increased SST+ interneurons in the upper cortical layers. Importantly, 17 ß-estradiol treatment in preterm rabbits increased the number of PV+ neurons in the upper cortical layers relative to controls at postnatal day 14 (P14) and P21 and transiently reduced SST population at P14. Moreover, protein and mRNA levels of Arx, a key regulator of cortical interneuron maturation and migration, were higher in estrogen-treated rabbits relative to controls. Therefore, deficits in PV+ and excess of SST+ neurons in premature newborns are ameliorated by estrogen replacement, which can be attributed to elevated Arx levels. Estrogen replacement might enhance neurodevelopmental outcomes in extremely preterm infants.SIGNIFICANCE STATEMENT Premature birth often leads to neurodevelopmental delays and behavioral disorders, which may be ascribed to disturbances in the development and maturation of cortical interneurons. Here, we show that preterm birth in humans is associated with reduced population of parvalbumin-positive (PV+) neurons and an excess of somatostatin-expressing interneurons in the cerebral cortex. More importantly, 17 ß-estradiol treatment increased the number of PV+ neurons in preterm-born rabbits, which appears to be mediated by an elevation in the expression of Arx transcription factor. Hence the present study highlights prematurity-induced reduction in PV+ neurons in human infants and reversal in their population by estrogen replacement in preterm rabbits. Because preterm birth drops plasma estrogen level 100-fold, estrogen replacement in extremely preterm infants might improve their developmental outcome and minimize neurobehavioral disorders.


Asunto(s)
Corteza Cerebral/patología , Estradiol/farmacología , Enfermedades del Prematuro/patología , Interneuronas/efectos de los fármacos , Animales , Animales Recién Nacidos , Calbindina 2/análisis , Recuento de Células , Femenino , Edad Gestacional , Glutamato Descarboxilasa/análisis , Humanos , Recién Nacido , Recien Nacido Prematuro , Interneuronas/química , Interneuronas/clasificación , Interneuronas/fisiología , Masculino , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuropéptido Y/análisis , Parvalbúminas/análisis , Conejos , Somatostatina/análisis , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
7.
J Neurochem ; 140(5): 776-786, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28054340

RESUMEN

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) agonist, is clinically available to treat multiple sclerosis and is showing promise in treating stroke. We investigated if fingolimod provides long-term protection from experimental neonatal germinal matrix hemorrhage (GMH), aiming to support a potential mechanism of acute fingolimod-induced protection. GMH was induced in P7 rats by infusion of collagenase (0.3 U) into the right ganglionic eminence. Animals killed at 4 weeks post-GMH received low- or high-dose fingolimod (0.25 or 1.0 mg/kg) or vehicle, and underwent neurocognitive testing before histopathological evaluation. Subsequently, a cohort of animals killed at 72 h post-GMH received 1.0 mg/kg fingolimod; the specific S1PR1 agonist, SEW2871; or fingolimod co-administered with the S1PR1/3/4 inhibitor, VPC23019, or the Rac1 inhibitor, EHT1864. All drugs were injected intraperitoneally 1, 24, and 48 h post-surgery. At 72 h post-GMH, brain water content, extravasated Evans blue dye, and hemoglobin were measured as well as the expression levels of phospho-Akt, Akt, GTP-Rac1, Total-Rac1, ZO1, occludin, and claudin-3 determined. Fingolimod significantly improved long-term neurocognitive performance and ameliorated brain tissue loss. At 72 h post-GMH, fingolimod reduced brain water content and Evans blue dye extravasation as well as reversed GMH-induced loss of tight junctional proteins. S1PR1 agonism showed similar protection, whereas S1PR or Rac1 inhibition abolished the protective effect of fingolimod. Fingolimod treatment improved functional and morphological outcomes after GMH, in part, by tempering acute post-hemorrhagic blood-brain barrier disruption via the activation of the S1PR1/Akt/Rac1 pathway.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Hemorragias Intracraneales/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Proteína de Unión al GTP rac1/metabolismo , Animales , Agua Corporal/metabolismo , Encéfalo/patología , Química Encefálica/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Cognición/efectos de los fármacos , Femenino , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/psicología , Recuento de Leucocitos , Masculino , Oxadiazoles/farmacología , Fosfoserina/análogos & derivados , Fosfoserina/farmacología , Embarazo , Pironas/farmacología , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Tiofenos/farmacología , Proteínas de Uniones Estrechas/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores
9.
Neurobiol Dis ; 87: 124-33, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26739391

RESUMEN

Germinal matrix hemorrhage remains the leading cause of morbidity and mortality in preterm infants in the United States with little progress made in its clinical management. Survivors are often afflicted with long-term neurological sequelae, including cerebral palsy, mental retardation, hydrocephalus, and psychiatric disorders. Blood clots disrupting normal cerebrospinal fluid circulation and absorption after germinal matrix hemorrhage are thought to be important contributors towards post-hemorrhagic hydrocephalus development. We evaluated if upregulating CD36 scavenger receptor expression in microglia and macrophages through PPARγ stimulation, which was effective in experimental adult cerebral hemorrhage models and is being evaluated clinically, will enhance hematoma resolution and ameliorate long-term brain sequelae using a neonatal rat germinal matrix hemorrhage model. PPARγ stimulation (15d-PGJ2) increased short-term PPARγ and CD36 expression levels as well as enhanced hematoma resolution, which was reversed by a PPARγ antagonist (GW9662) and CD36 siRNA. PPARγ stimulation (15d-PGJ2) also reduced long-term white matter loss and post-hemorrhagic ventricular dilation as well as improved neurofunctional outcomes, which were reversed by a PPARγ antagonist (GW9662). PPARγ-induced upregulation of CD36 in macrophages and microglia is, therefore, critical for enhancing hematoma resolution and ameliorating long-term brain sequelae.


Asunto(s)
Antígenos CD36/metabolismo , Hematoma/fisiopatología , Hemorragias Intracraneales/fisiopatología , PPAR gamma/metabolismo , Anilidas/farmacología , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Antígenos CD36/genética , Fármacos del Sistema Nervioso Central/farmacología , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Hematoma/tratamiento farmacológico , Hematoma/patología , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/patología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/fisiología , Microglía/efectos de los fármacos , Microglía/fisiología , Fármacos Neuroprotectores/farmacología , PPAR gamma/antagonistas & inhibidores , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , ARN Interferente Pequeño/administración & dosificación , Distribución Aleatoria , Ratas Sprague-Dawley , Regulación hacia Arriba
10.
Acta Neurochir Suppl ; 121: 63-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463924

RESUMEN

Germinal matrix hemorrhage (GMH) is the most common and devastating neurological injury of premature infants, and current treatment approaches are ineffective. Remote ischemic postconditioning (RIPC) is a method by which brief limb ischemic stimuli protect the injured brain. We hypothesized that RIPC can improve outcomes following GMH in rats. Neonatal rats (P7) were subjected to either stereotactic ganglionic eminence collagenase infusion or sham surgery. Groups were as follows: sham (n = 0), GMH non-RIPC (n = 10), GMH + 1 week RIPC (n = 10), GMH + 2 weeks RIPC (n = 10). Neurobehavior analysis at the fourth week consisted of Morris water maze (MWM) and rotarod (RR). This was followed by euthanasia for histopathology on day 28. Both 1- and 2-week RIPC showed significant improvement in FF and RR motor testing compared with untreated animals (i.e., GMH without RIPC). RIPC treatment also improved cognition (MWM) and attenuated neuropathological ventricular enlargement (hydrocephalus) in juvenile animals following GMH. RIPC is a safe and noninvasive approach that improved sensorimotor and neuropathological outcomes following GMH in rats. Further studies are needed to evaluate for mechanisms of neuroprotection.


Asunto(s)
Hemorragia Cerebral/terapia , Poscondicionamiento Isquémico/métodos , Animales , Animales Recién Nacidos , Conducta Animal , Encéfalo/fisiopatología , Hemorragia Cerebral/inducido químicamente , Hemorragia Cerebral/fisiopatología , Modelos Animales de Enfermedad , Extremidades , Aprendizaje por Laberinto , Colagenasa Microbiana/toxicidad , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante
11.
Acta Neurochir Suppl ; 121: 209-12, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463950

RESUMEN

Germinal matrix hemorrhage (GMH) is the most devastating neurological problem of premature infants. Current treatment strategies are ineffective and brain injury is unpreventable. Insulin-like growth factor 1 (IGF-1) is an endogenous protein shown to have multiple neuroprotective properties. We therefore hypothesized that IGF-1 would reduce brain injury after GMH. Neonatal rats (P7 age) received stereotactic collagenase into the right ganglionic eminence. The following groups were studied: (1) sham, (2) GMH + vehicle, (3) GMH + intranasal IGF-1. Three days later, the animals were evaluated using the righting-reflex (early neurobehavior), Evans blue dye leakage (blood-brain barrier (BBB) permeability), brain water content (edema), and hemoglobin assay (extent of bleeding). Three weeks later, juvenile rats were tested using a water maze (delayed neurobehavior), and then were sacrificed on day 28 for assessment of hydrocephalus (ventricular size). Intranasal IGF-1 treated animals had improved neurological function, and amelioration of BBB permeability, edema, and re-bleeding. IGF-1 may play a part in protective brain signaling following GMH, and our observed protective effect may offer new promise for treatment targeting this vulnerable patient population.


Asunto(s)
Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Hemorragia Cerebral/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Hemorragias Intracraneales/metabolismo , Administración Intranasal , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/metabolismo , Edema Encefálico , Hemorragia Cerebral/patología , Hemorragia Cerebral/fisiopatología , Modelos Animales de Enfermedad , Hemoglobinas/efectos de los fármacos , Hemoglobinas/metabolismo , Hidrocefalia , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Permeabilidad , Ratas , Ratas Sprague-Dawley
12.
Acta Neurochir Suppl ; 121: 203-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463949

RESUMEN

Germinal matrix hemorrhage (GMH) is a major cause of brain damage in prematurity and has long-lasting neurological implications. The development of brain inflammation contributes to brain injury, leading to a lifetime of neurologic deficits. PAR-1 and 4 receptors are involved with inflammatory pathways after brain hemorrhage in adult models of stroke, of which cyclooxygenase-2 (COX-2) is a potential mediator. We therefore hypothesized a role for PAR-1, 4/ COX-2 signaling following GMH. Postnatal day 7 Sprague-Dawley rats were subjected to GMH induction, which entailed stereotactic collagenase infusion into the ganglionic eminence. Animals were euthanized at two time points: 72 h (short-term) or 4 weeks (long-term). Short-term COX-2 expression was evaluated in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective COX-2 inhibitor (NS-398); and the neurobehavioral and pathological examinations were performed 4 weeks later. Pharmacological PAR-1, 4 antagonism normalized COX-2 expression following GMH and reduced hydrocephalus. Early inhibition of COX-2 by NS-398 improved long-term neurobehavioral outcomes. COX-2 signaling plays an important role in brain injury following neonatal GMH, possibly through upstream PAR-1, 4 receptor mechanisms.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Hemorragias Intracraneales/metabolismo , Oligopéptidos/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/patología , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores
13.
Acta Neurochir Suppl ; 121: 213-6, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463951

RESUMEN

Germinal matrix hemorrhage (GMH) is the most common cause of neurological complications of prematurity and has lasting implications. PAR-1 and PAR-4 receptors are involved with upstream signaling pathways following brain hemorrhage in adult models of stroke, of which the mammalian target of rapamycin (mTOR) is a potential downstream mediator. Therefore, we hypothesized a role for PAR-1, -4/ mTOR signaling following GMH brain injury. Postnatal day 7 Sprague-Dawley rats were subjected to GMH through stereotactic infusion of collagenase into the right ganglionic eminence. Rodents were euthanized at 72 h (short term), or 4 weeks (long term). Short-term mTOR expression was evaluated by Western blot in the context of PAR-1 (SCH-79797) and PAR-4 (P4pal10) inhibition. Pups in the long-term group were administered the selective mTOR inhibitor (rapamycin) with neurobehavioral and brain pathological examinations performed at 4 weeks. Pharmacological PAR-1, -4 antagonism normalized the increased mTOR expression following GMH. Early inhibition of mTOR by rapamycin improved long-term outcomes in rats. Mammalian-TOR signaling plays an important role in brain injury following neonatal GMH, possibly involving upstream PAR-1, -4 mechanisms.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragias Intracraneales/metabolismo , Oligopéptidos/farmacología , Pirroles/farmacología , Quinazolinas/farmacología , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/efectos de los fármacos , Trombina/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Inmunosupresores/farmacología , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
14.
Acta Neurochir Suppl ; 121: 237-41, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463955

RESUMEN

Hemorrhagic transformation occurs in as many as 48 % of stroke patients and is a major contributor to post-insult morbidity and mortality. Experimental models of hemorrhagic transformation are utilized for understanding the mechanisms behind its development, as well as for investigating potential therapeutics for prevention and reduction of bleeding. Thoroughly studying animal models of hemorrhagic transformation is critically important for testing novel treatments. Thus far, no study has examined the progression of brain swelling and hemorrhagic transformation after transient middle cerebral artery occlusion (MCAO). Herein, we investigate the development of infarction, brain swelling, and hemorrhagic transformation following MCAO in hyperglycemic rats. Twenty-five Sprague-Dawley rats were subjected to either 1.5 h of MCAO or sham surgery 15 min after induction of hyperglycemia. Animals were sacrificed at 0.25, 1, 3, or 24 h after reperfusion for measurement of infarct volume, brain swelling, and hemoglobin volume. Within 15 min of reperfusion, the infarct volume was significantly larger than in sham animals and did not increase in size over the 24 h. However, both brain swelling and hemorrhagic transformation, which began immediately after reperfusion, increase over 24 h after reperfusion.


Asunto(s)
Glucemia/metabolismo , Edema Encefálico/metabolismo , Hemorragia Cerebral/metabolismo , Hiperglucemia/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Animales , Edema Encefálico/etiología , Hemorragia Cerebral/etiología , Modelos Animales de Enfermedad , Glucosa/farmacología , Hiperglucemia/inducido químicamente , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Riesgo , Edulcorantes/farmacología , Factores de Tiempo
15.
Stroke ; 46(6): 1710-3, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25931468

RESUMEN

BACKGROUND AND PURPOSE: This study examines the role of thrombin's protease-activated receptor (PAR)-1, PAR-4 in mediating cyclooxygenase-2 and mammalian target of rapamycin after germinal matrix hemorrhage. METHODS: Germinal matrix hemorrhage was induced by intraparenchymal infusion of bacterial collagenase into the right ganglionic eminence of P7 rat pups. Animals were treated with PAR-1, PAR-4, cyclooxygenase-2, or mammalian target of rapamycin inhibitors by 1 hour, and ≤5 days. RESULTS: We found increased thrombin activity 6 to 24 hours after germinal matrix hemorrhage, and PAR-1, PAR-4, inhibition normalized cyclooxygenase-2, and mammalian target of rapamycin by 72 hours. Early treatment with NS398 or rapamycin substantially improved long-term outcomes in juvenile animals. CONCLUSIONS: Suppressing early PAR signal transduction, and postnatal NS398 or rapamycin treatment, may help reduce germinal matrix hemorrhage severity in susceptible preterm infants.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Inmunosupresores/farmacología , Nitrobencenos/farmacología , Receptor PAR-1/antagonistas & inhibidores , Receptores de Trombina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Sulfonamidas/farmacología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Ciclooxigenasa 2/metabolismo , Ratas
16.
Neurobiol Dis ; 82: 349-358, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176793

RESUMEN

BACKGROUND AND PURPOSE: Edema formation, inflammation and increased blood-brain barrier permeability contribute to poor outcomes after intracerebral hemorrhage (ICH). This study examined the therapeutic effect of dimethyl fumarate (DMF), a fumaric acid ester that activates nuclear factor erythroid-2 related factor 2 (Nrf2) and Nrf2 heterodimerization effector protein musculo-aponeurotic fibrosarcoma-G (MAFG) in a murine ICH model. METHODS: Male CD-1 mice (n=176) were subjected to intrastriatal infusion of bacterial collagenase (n=126), autologous blood (n=18) or sham surgery (n=32). Four (4) animals not subjected to ICH (naive) were also included in the study. After ICH, animals either received vehicle, dimethyl fumarate (10 mg or 100 mg/kg) or casein kinase 2 inhibitor (E)-3-(2,3,4,5-tetrabromophenyl)acrylic acid (TBCA). Thirty-two mice also received scrambled siRNA or MAFG siRNA 24h before ICH. Brain water content and neurological function were evaluated. RESULTS: Dimethyl fumarate reduced Evans blue dye extravasation, decreased brain water content, and improved neurological deficits at 24 and 72 h after ICH. Casein kinase 2 inhibitor TBCA and MAFG siRNA prevented the effect of dimethyl fumarate on brain edema and neurological function. After ICH, ICAM-1 levels increased and casein kinase 2 levels decreased. Dimethyl fumarate reduced ICAM-1 but enhanced casein kinase 2 levels. Again, casein kinase 2 inhibitor TBCA and MAFG siRNA abolished the effect of dimethyl fumarate on ICAM-1 and casein kinase 2. Dimethyl fumarate preserved pNrf2 and MAFG expression in the nuclear lysate after ICH and the effect of dimethyl fumarate was abolished by casein kinase 2 inhibitor TBCA and MAFG siRNA. Dimethyl fumarate reduced microglia activation in peri-hematoma areas after ICH. The protective effect of dimethyl fumarate on brain edema and neurological function was also observed in a blood injection mouse model. CONCLUSION: Dimethyl fumarate ameliorated inflammation, reduced blood-brain barrier permeability, and improved neurological outcomes by casein kinase 2 and Nrf2 signaling pathways after experimental ICH in mice.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Hemorragia Cerebral/tratamiento farmacológico , Dimetilfumarato/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Acrilatos/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/enzimología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/enzimología , Quinasa de la Caseína II/antagonistas & inhibidores , Hemorragia Cerebral/enzimología , Colagenasas , Modelos Animales de Enfermedad , Molécula 1 de Adhesión Intercelular/metabolismo , Factor de Transcripción MafG/genética , Factor de Transcripción MafG/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/enzimología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
17.
Stroke ; 45(8): 2475-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24947291

RESUMEN

BACKGROUND AND PURPOSE: This study investigated if acute and delayed deferoxamine treatment attenuates long-term sequelae after germinal matrix hemorrhage (GMH). METHODS: Bacterial collagenase (0.3 U) was infused intraparenchymally into the right hemispheric ganglionic eminence in P7 rat pups to induce GMH. GMH animals received either deferoxamine or vehicle twice a day for 7 consecutive days. Deferoxamine administration was initiated at either 1 hour or 72 hours post-GMH. Long-term neurocognitive deficits and motor coordination were assessed using Morris water maze, rotarod, and foot fault tests between day 21 to 28 post-GMH. At 28 days post-GMH, brain morphology was assessed and extracellular matrix protein (fibronectin and vitronectin) expression was determined. RESULTS: Acute and delayed deferoxamine treatment improved long-term motor and cognitive function at 21 to 28 days post-GMH. Attenuated neurofunction was paralleled with improved overall brain morphology at 28 days post-GMH, reducing white matter loss, basal ganglia loss, posthemorrhagic ventricular dilation, and cortical loss. GMH resulted in significantly increased expression of fibronectin and vitronectin, which was reversed by acute and delayed deferoxamine treatment. CONCLUSIONS: Acute and delayed deferoxamine administration ameliorated long-term sequelae after GMH.


Asunto(s)
Encéfalo/efectos de los fármacos , Deferoxamina/uso terapéutico , Hemorragias Intracraneales/tratamiento farmacológico , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Animales , Animales Recién Nacidos , Deferoxamina/administración & dosificación , Modelos Animales de Enfermedad , Hemorragias Intracraneales/fisiopatología , Aprendizaje por Laberinto/fisiología , Actividad Motora/fisiología , Ratas , Factores de Tiempo
18.
Neurochem Res ; 39(9): 1621-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24482021

RESUMEN

Acute central nervous system (CNS) injuries, including stroke, traumatic brain injury (TBI), and spinal cord injury (SCI), are common causes of human disabilities and deaths, but the pathophysiology of these diseases is not fully elucidated and, thus, effective pharmacotherapies are still lacking. Valproic acid (VPA), an inhibitor of histone deacetylation, is mainly used to treat epilepsy and bipolar disorder with few complications. Recently, the neuroprotective effects of VPA have been demonstrated in several models of acute CNS injuries, such as stroke, TBI, and SCI. VPA protects the brain from injury progression via anti-inflammatory, anti-apoptotic, and neurotrophic effects. In this review, we focus on the emerging neuroprotective properties of VPA and explore the underlying mechanisms. In particular, we discuss several potential related factors in VPA research and present the opportunity to administer VPA as a novel neuropective agent.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Ácido Valproico/uso terapéutico , Animales , Femenino , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
19.
Stroke ; 44(6): 1743-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23613493

RESUMEN

BACKGROUND AND PURPOSE: Blood-brain barrier disruption and consequent vasogenic edema formation codetermine the clinical course of intracerebral hemorrhage (ICH). This study examined the effect of PHA-543613, a novel α7 nicotinic acetylcholine receptor agonist, on blood-brain barrier preservation after ICH. METHODS: Male CD-1 mice, subjected to intrastriatal blood infusion, received PHA-543613 alone or in combination with α7 nicotinic acetylcholine receptor antagonist methyllycaconitine or phosphatidylinositol 3-kinase inhibitor wortmannin. RESULTS: PHA-543613 alone, but not in combination with methyllycaconitine or wortmannin, inhibited glycogen synthase kinase-3ß, thus, stabilizing ß-catenin and tight junction proteins, which was paralleled by improved blood-brain barrier stability and ameliorated neurofunctional deficits in ICH animals. CONCLUSIONS: PHA-543613 preserved blood-brain barrier integrity after ICH, possibly through phosphatidylinositol 3-kinase-Akt-induced inhibition of glycogen synthase kinase-3ß and ß-catenin stabilization.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Hemorragia Cerebral/fisiopatología , Quinuclidinas/farmacología , Receptores Nicotínicos/fisiología , Transducción de Señal/efectos de los fármacos , Aconitina/análogos & derivados , Aconitina/farmacología , Androstadienos/farmacología , Animales , Barrera Hematoencefálica/fisiología , Hemorragia Cerebral/metabolismo , Claudina-3/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Wortmanina , beta Catenina/metabolismo
20.
Stroke ; 44(5): 1410-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23539525

RESUMEN

BACKGROUND AND PURPOSE: Plasma thrombin concentration is increased after subarachnoid hemorrhage (SAH). However, the role of thrombin receptor (protease-activated receptor-1 [PAR-1]) in endothelial barrier disruption has not been studied. The aims of this study were to investigate the role of PAR-1 in orchestrating vascular permeability and to assess the potential therapeutics of a PAR-1 antagonist, SCH79797, through maintaining vascular integrity. METHODS: SCH79797 was injected intraperitoneally into male Sprauge-Dawley rats undergoing SAH by endovascular perforation. Assessment was conducted at 24 hours after SAH for brain water content, Evans blue content, and neurobehavioral testing. To explore the role of PAR-1 activation and the specific mechanism of SCH79797's effect after SAH, Western blot, immunoprecipitation, and immunofluorescence of hippocampus tissue were performed. A p21-activated kinase-1 (PAK1) inhibitor, IPA-3, was used to explore the underlying protective mechanism of SCH79797. RESULTS: At 24 hours after SAH, animals treated with SCH79797 demonstrated a reduction in brain water content, Evans blue content, and neurobehavioral deficits. SCH79797 also attenuated PAR-1 expression and maintained the level of vascular endothelial-cadherin, an important component of adherens junctions. Downstream to PAR-1, c-Src-dependent activation of p21-activated kinase-1 led to an increased serine/threonine phosphorylation of vascular endothelial-cadherin; immunoprecipitation results revealed an enhanced binding of phosphorylated vascular endothelial-cadherin with endocytosis orchestrator ß-arrestin-2. These pathological states were suppressed after SCH79797 treatment. CONCLUSIONS: PAR-1 activation after SAH increases microvascular permeability, at least, partly through a PAR-1-c-Src-p21-activated kinase-1-vascular endothelial-cadherin phosphorylation pathway. Through suppressing PAR-1 activity, SCH79797 plays a protective role in maintaining microvascular integrity after SAH.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Pirroles/uso terapéutico , Quinazolinas/uso terapéutico , Receptor PAR-1/antagonistas & inhibidores , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Disulfuros/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Naftoles/farmacología , Permeabilidad/efectos de los fármacos , Pirroles/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/patología , Quinasas p21 Activadas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA