Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Res Int ; 2017: 8134653, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28529954

RESUMEN

Accounting for high mortality and morbidity rates, intracerebral hemorrhage (ICH) remains one of the most detrimental stroke subtypes lacking a specific therapy. Neuroinflammation contributes to ICH-induced brain injury and is associated with unfavorable outcomes. This study aimed to evaluate whether α7 nicotinic acetylcholine receptor (α7nAChR) stimulation ameliorates neuroinflammation after ICH. Male CD-1 mice and Sprague-Dawley were subjected to intracerebral injection of autologous blood or bacterial collagenase. ICH animals received either α7nAChR agonist PHA-543613 alone or combined with α7nAChR antagonist methyllycaconitine (MLA) or Janus kinase 2 (JAK2) antagonist AG490. Neurobehavioral deficits were evaluated at 24 hours, 72 hours, and 10 weeks after ICH induction. Perihematomal expressions of JAK2, signal transducer and activator of transcription 3 (STAT3), tumor necrosis factor-α (TNF-α), and myeloperoxidase (MPO) were quantified via Western blot. Histologic volumetric analysis of brain tissues was conducted after 10 weeks following ICH induction. PHA-543613 improved short-term neurobehavioral (sensorimotor) deficits and increased activated perihematomal JAK2 and STAT3 expressions while decreasing TNF-α and MPO expressions after ICH. MLA reversed these treatment effects. PHA-543613 also improved long-term neurobehavioral (sensorimotor, learning, and memory) deficits and ameliorated brain atrophy after ICH. These treatment effects were reduced by AG490. α7nAChR stimulation reduced neuroinflammation via activation of the JAK2-STAT3 pathway, thereby ameliorating the short- and long-term sequelae after ICH.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Janus Quinasa 2/genética , Factor de Transcripción STAT3/genética , Receptor Nicotínico de Acetilcolina alfa 7/uso terapéutico , Animales , Transfusión de Sangre Autóloga/métodos , Lesiones Encefálicas/etiología , Lesiones Encefálicas/genética , Lesiones Encefálicas/fisiopatología , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/genética , Hemorragia Cerebral/fisiopatología , Colagenasas/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Inflamación/complicaciones , Inflamación/genética , Inflamación/fisiopatología , Ratones , Neuronas/efectos de los fármacos , Neuronas/patología , Peroxidasa/genética , Quinuclidinas/administración & dosificación , Ratas , Factor de Necrosis Tumoral alfa/genética , Tirfostinos/administración & dosificación , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/genética
2.
J Cereb Blood Flow Metab ; 37(9): 3135-3149, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28155585

RESUMEN

We aim to determine if direct thrombin inhibition by dabigatran will improve long-term brain morphological and neurofunctional outcomes and if potential therapeutic effects are dependent upon reduced PAR-1 stimulation and consequent mTOR activation. Germinal matrix haemorrhage was induced by stereotaxically injecting 0.3 U type VII-S collagenase into the germinal matrix of P7 rat pups. Animals were divided into five groups: sham, vehicle (5% DMSO), dabigatran intraperitoneal, dabigatran intraperitoneal + TFLLR-NH2 (PAR-1 agonist) intranasal, SCH79797 (PAR-1 antagonist) intraperitoneal, and dabigatran intranasal. Neurofunctional outcomes were determined by Morris water maze, rotarod, and foot fault evaluations at three weeks. Brain morphological outcomes were determined by histological Nissl staining at four weeks. Expression levels of p-mTOR/p-p70s6k at three days and vitronectin/fibronectin at 28 days were quantified. Intranasal and intraperitoneal dabigatran promoted long-term neurofunctional recovery, improved brain morphological outcomes, and reduced intracranial pressure at four weeks after GMH. PAR-1 stimulation tended to reverse dabigatran's effects on post-haemorrhagic hydrocephalus development. Dabigatran also reduced expression of short-term p-mTOR and long-term extracellular matrix proteins, which tended to be reversed by PAR-1 agonist co-administration. PAR-1 inhibition alone, however, did not achieve the same therapeutic effects as dabigatran administration.


Asunto(s)
Antitrombinas/uso terapéutico , Dabigatrán/uso terapéutico , Hidrocefalia/prevención & control , Hemorragias Intracraneales/tratamiento farmacológico , Administración Intranasal , Animales , Animales Recién Nacidos , Antitrombinas/administración & dosificación , Dabigatrán/administración & dosificación , Modelos Animales de Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Hidrocefalia/etiología , Hidrocefalia/patología , Hidrocefalia/fisiopatología , Inyecciones Intraperitoneales , Hemorragias Intracraneales/complicaciones , Hemorragias Intracraneales/patología , Hemorragias Intracraneales/fisiopatología , Aprendizaje por Laberinto/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Pirroles/administración & dosificación , Pirroles/farmacología , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inhibidores , Prueba de Desempeño de Rotación con Aceleración Constante
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Acta Neurochir Suppl ; 121: 311-5, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463967

RESUMEN

Neurosurgical procedures can damage viable brain tissue unintentionally by a wide range of mechanisms. This surgically induced brain injury (SBI) can be a result of direct incision, electrocauterization, or tissue retraction. Plasmin, a serine protease that dissolves fibrin blood clots, has been shown to enhance cerebral edema and hemorrhage accumulation in the brain through disruption of the blood brain barrier. Epsilon aminocaproic acid (EAA), a recognized antifibrinolytic lysine analogue, can reduce the levels of active plasmin and, in doing so, potentially can preserve the neurovascular unit of the brain. We investigated the role of EAA as a pretreatment neuroprotective modality in a SBI rat model, hypothesizing that EAA therapy would protect brain tissue integrity, translating into preserved neurobehavioral function. Male Sprague-Dawley rats were randomly assigned to one of four groups: sham (n = 7), SBI (n = 7), SBI with low-dose EAA, 150 mg/kg (n = 7), and SBI with high-dose EAA, 450 mg/kg (n = 7). SBI was induced by partial right frontal lobe resection through a frontal craniotomy. Postoperative assessment at 24 h included neurobehavioral testing and measurement of brain water content. Results at 24 h showed both low- and high-dose EAA reduced brain water content and improved neurobehavioral function compared with the SBI groups. This suggests that EAA may be a useful pretherapeutic modality for SBI. Further studies are needed to clarify optimal therapeutic dosing and to identify mechanisms of neuroprotection in rat SBI models.


Asunto(s)
Ácido Aminocaproico/farmacología , Antifibrinolíticos/farmacología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Procedimientos Neuroquirúrgicos , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Lóbulo Frontal/cirugía , Complicaciones Intraoperatorias , Ratas , Ratas Sprague-Dawley
10.
Acta Neurochir Suppl ; 121: 305-10, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463966

RESUMEN

Surgically induced brain injury (SBI) results in brain edema and neurological decline. Valproic acid (VA) has been shown to be neuroprotective in several experimental brain diseases. In this study, we investigated the pretreatment effect of VA in a rat model of SBI. A total of 57 male Sprague-Dawley rats were use in four groups: sham, SBI + vehicle, SBI + low dose (100 mg/kg) VA, and SBI + high dose (300 mg/kg) VA. SBI was induced by partially resecting right frontal lobes. Shams underwent identical surgical procedures without brain resection. VA or vehicle was administered subcutaneously 30 min prior to SBI. At 24 and 72 h post SBI, neurobehavior and brain water content were assessed as well as matrix metalloproteinases (MMPs) activities. There was significantly higher brain water content within the right frontal lobe in SBI rats than in shams. Without neurobehavioral improvements, the low-dose but not high-dose VA significantly reduced brain edema at 24 h post SBI. The protection tends to persist to 72 h post SBI. At 24 h post SBI, low-dose VA did not significantly reduce the elevated MMP-9 activity associated with SBI. In conclusion, VA pretreatment attenuated brain edema at 24 h after SBI but lacked MMP inhibition. The single dose VA was not associated with neurobehavioral benefits.


Asunto(s)
Conducta Animal/efectos de los fármacos , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Procedimientos Neuroquirúrgicos , Ácido Valproico/farmacología , Animales , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Lóbulo Frontal/cirugía , Complicaciones Intraoperatorias , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley
11.
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
12.
Curr Med Chem ; 22(10): 1214-38, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25620100

RESUMEN

Neonatal brain hemorrhage (NBH) of prematurity is an unfortunate consequence of preterm birth. Complications result in shunt dependence and long-term structural changes such as posthemorrhagic hydrocephalus, periventricular leukomalacia, gliosis, and neurological dysfunction. Several animal models are available to study this condition, and many basic mechanisms, etiological factors, and outcome consequences, are becoming understood. NBH is an important clinical condition, of which treatment may potentially circumvent shunt complication, and improve functional recovery (cerebral palsy, and cognitive impairments). This review highlights key pathophysiological findings of the neonatal vascular-neural network in the context of molecular mechanisms targeting the posthemorrhagic hydrocephalus affecting this vulnerable infant population.


Asunto(s)
Hemorragia Cerebral/congénito , Hemorragia Cerebral/fisiopatología , Recien Nacido Prematuro , Red Nerviosa/irrigación sanguínea , Red Nerviosa/fisiopatología , Hemorragia Cerebral/patología , Líquido Cefalorraquídeo/metabolismo , Humanos , Recién Nacido , Recien Nacido Prematuro/metabolismo
13.
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
14.
J Neurol Sci ; 342(1-2): 101-6, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24819918

RESUMEN

Early brain injury (EBI), following subarachnoid hemorrhage (SAH), comprises blood-brain barrier (BBB) disruption and consequent edema formation. Peripheral leukocytes can infiltrate the injured brain, thereby aggravating BBB leakage and neuroinflammation. Thus, anti-inflammatory pharmacotherapies may ameliorate EBI and provide neuroprotection after SAH. Cannabinoid type 2 receptor (CB2R) agonism has been shown to reduce neuroinflammation; however, the precise protective mechanisms remain to be elucidated. This study aimed to evaluate whether the selective CB2R agonist, JWH133 can ameliorate EBI by reducing brain-infiltrated leukocytes after SAH. Adult male Sprague-Dawley rats were randomly assigned to the following groups: sham-operated, SAH with vehicle, SAH with JWH133 (1.0mg/kg), or SAH with a co-administration of JWH133 and selective CB2R antagonist SR144528 (3.0mg/kg). SAH was induced by endovascular perforation, and JWH133 was administered 1h after surgery. Neurological deficits, brain water content, Evans blue dye extravasation, and Western blot assays were evaluated at 24h after surgery. JWH133 improved neurological scores and reduced brain water content; however, SR144528 reversed these treatment effects. JWH133 reduced Evans blue dye extravasation after SAH. Furthermore, JWH133 treatment significantly increased TGF-ß1 expression and prevented an SAH-induced increase in E-selectin and myeloperoxidase. Lastly, SAH resulted in a decreased expression of the tight junction protein zonula occludens-1 (ZO-1); however, JWH133 treatment increased the ZO-1 expression. We suggest that CB2R stimulation attenuates neurological outcome and brain edema, by suppressing leukocyte infiltration into the brain through TGF-ß1 up-regulation and E-selectin reduction, resulting in protection of the BBB after SAH.


Asunto(s)
Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Quimiotaxis de Leucocito/efectos de los fármacos , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/fisiología , Hemorragia Subaracnoidea/complicaciones , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Edema Encefálico/fisiopatología , Canfanos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Agonistas de Receptores de Cannabinoides/uso terapéutico , Antagonistas de Receptores de Cannabinoides/farmacología , Cannabinoides/farmacología , Cannabinoides/uso terapéutico , Selectina E/biosíntesis , Selectina E/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Leucocitos/citología , Leucocitos/efectos de los fármacos , Masculino , Peroxidasa/biosíntesis , Peroxidasa/efectos de los fármacos , Pirazoles/farmacología , Ratas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Hemorragia Subaracnoidea/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Proteína de la Zonula Occludens-1/biosíntesis , Proteína de la Zonula Occludens-1/efectos de los fármacos
15.
Behav Brain Res ; 264: 151-60, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24518201

RESUMEN

Formation of brain edema after intracerebral hemorrhage (ICH) is highly associated with its poor outcome. However, the relationship between cerebral edema and behavioral deficits has not been thoroughly examined in the preclinical setting. Hence, this study aimed to evaluate the ability of common sensorimotor tests to predict the extent of brain edema in two mouse models of ICH. One hundred male CD-1 mice were subjected to sham surgery or ICH induction via intrastriatal injection of either autologous blood (30 µL) or bacterial collagenase (0.0375U or 0.075U). At 24 and 72 h after surgery, animals underwent a battery of behavioral tests, including the modified Garcia neuroscore (Neuroscore), corner turn test (CTT), forelimb placing test (FPT), wire hang task (WHT) and beam walking (BW). Brain edema was evaluated via the wet weight/dry weight method. Intrastriatal injection of autologous blood or bacterial collagenase resulted in a significant increase in brain water content and associated sensorimotor deficits (p<0.05). A significant correlation between brain edema and sensorimotor deficits was observed for all behavioral tests except for WHT and BW. Based on these findings, we recommend implementing the Neuroscore, CTT and/or FPT in preclinical studies of unilateral ICH in mice.


Asunto(s)
Edema Encefálico/etiología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/etiología , Trastornos Neurológicos de la Marcha/etiología , Animales , Transfusión de Sangre Autóloga/efectos adversos , Colagenasas/toxicidad , Modelos Animales de Enfermedad , Conducta Exploratoria , Hematoma/etiología , Masculino , Ratones , Actividad Motora , Fuerza Muscular , Valor Predictivo de las Pruebas , Propiocepción/fisiología , Desempeño Psicomotor , Factores de Tiempo , Vibrisas/inervación
16.
Transl Stroke Res ; 4(5): 524-32, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24187597

RESUMEN

Although there is evidence that sphingosine-1-phosphate receptor-1 (S1P1) activation occurs following experimental brain injury, there is little information about its metabolic pathway in cerebral ischemia. The purpose of this study was to evaluate the role of the sphingosine metabolic pathway including S1P1, sphingosine kinases 1 (SphK1), and 2 (SphK2) in transient middle cerebral artery occlusion (MCAO). Fifty-eight male Sprague-Dawley rats were used to asses temporal profiles of S1P1, SphK1 and 2 on neurons in infarct and periinfarct cortices at pre-infarct state, 6, and 24 hours after MCAO. The animals were then treated with vehicle and 0.25 mg/kg FTY720, which is an agonist of S1P receptors, and evaluated regarding neurological function, infarct volume, and S1P1 expression on neurons at 24 hours after MCAO. The expressions of S1P1, SphK1, and SphK2 were significantly decreased after MCAO. Labeling of all markers were reduced in the infarct cortex but remained present in the periinfarct cortex, and some were found to be on neurons. Significant improvements of neurological function and brain injury were observed in the FTY720 group compared with the vehicle and untreated groups, although S1P1 expression on neurons was reduced in the FTY720 group compared with the vehicle group. We demonstrated that S1P1, SphK1, and SphK2 were downregulated in the infarct cortex, whereas they were preserved in the periinfarct cortex where FTY720 reduced neuronal injury possibly via S1P1 activation. Our findings suggest that activation of the sphingosine metabolic pathway may be neuroprotective in cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuronas/metabolismo , Esfingosina/metabolismo , Animales , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Inmunosupresores/farmacología , Masculino , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Glicoles de Propileno/farmacología , Ratas , Ratas Sprague-Dawley , Esfingosina/análogos & derivados , Esfingosina/farmacología
17.
Stroke ; 44(12): 3587-90, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24149004

RESUMEN

BACKGROUND AND PURPOSE: This study investigated whether isoflurane ameliorates neurological sequelae after germinal matrix hemorrhage (GMH) through activation of the cytoprotective sphingosine kinase/sphingosine-1-phosphate receptor/Akt pathway. METHODS: GMH was induced in P7 rat pups by intraparenchymal infusion of bacterial collagenase (0.3 U) into the right hemispheric germinal matrix. GMH animals received 2% isoflurane either once 1 hour after surgery or every 12 hours for 3 days. Isoflurane treatment was then combined with sphingosine-1-phosphate receptor-1/2 antagonist VPC23019 or sphingosine kinase 1/2 antagonist N,N-dimethylsphingosine. RESULTS: Brain protein expression of sphingosine kinase-1 and phosphorylated Akt were significantly increased after isoflurane post-treatment, and cleaved caspase-3 was decreased at 24 hours after surgery, which was reversed by the antagonists. Isoflurane significantly reduced posthemorrhagic ventricular dilation and improved motor, but not cognitive, functions in GMH animals 3 weeks after surgery; no improvements were observed after VPC23019 administration. CONCLUSIONS: Isoflurane post-treatment improved the neurological sequelae after GMH possibly by activation of the sphingosine kinase/Akt pathway.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragias Intracraneales/tratamiento farmacológico , Isoflurano/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hemorragias Intracraneales/inducido químicamente , Hemorragias Intracraneales/metabolismo , Isoflurano/farmacología , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores de Lisoesfingolípidos/metabolismo , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Esfingosina/uso terapéutico
18.
PLoS One ; 8(7): e69571, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936048

RESUMEN

Chronic communicating hydrocephalus is a significant health problem affecting up to 20% of survivors of spontaneous subarachnoid hemorrhage (SAH). The development of new treatment strategies is hampered by the lack of well characterized disease models. This study investigated the incidence of chronic hydrocephalus by evaluating the temporal profile of intracranial pressure (ICP) elevation after SAH, induced by endovascular perforation in rats. Twenty-five adult male Sprague-Dawley rats (260-320 g) were subjected to either endovascular perforation or sham surgery. Five animals died after SAH induction. At 7, 14 and 21 days after surgery ICP was measured by stereotaxic puncture of the cisterna magna in SAH (n=10) and SHAM (n=10) animals. On day 21 T-maze test was performed and the number of alterations and latency to decision was recorded. On day 23, samples were processed for histological analyses. The relative ventricle area was evaluated in coronal Nissl stained sections. On day 7 after surgery all animals showed normal ICP. The absolute ICP values were significantly higher in SAH compared to SHAM animals on day 21 (8.26±4.53 mmHg versus 4.38±0.95 mmHg) but not on day 14. Observing an ICP of 10 mmHg as cut-off, 3 animals showed elevated ICP on day 14 and another animal on day 21. The overall incidence of ICP elevation was 40% in SAH animals. On day 21, results of T-maze testing were significantly correlated with ICP values, i.e. animals with elevated ICP showed a lower number of alterations and a delayed decision. Histology yielded a significantly higher (3.59 fold increased) relative ventricle area in SAH animals with ICP elevation compared to SAH animals without ICP elevation. In conclusion, the current study shows that experimental SAH leads to chronic hydrocephalus, which is associated with ICP elevation, behavioral alterations and ventricular dilation in about 40% of SAH animals.


Asunto(s)
Hidrocefalia/etiología , Hemorragia Subaracnoidea/complicaciones , Animales , Conducta Animal , Enfermedad Crónica , Hidrocefalia/patología , Hidrocefalia/fisiopatología , Presión Intracraneal/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Factores de Tiempo
19.
Neurobiol Dis ; 58: 296-307, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23816751

RESUMEN

Neuroinflammation contributes to the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Cytotoxic events following SAH, such as extracellular accumulation of adenosine triphosphate (ATP), may activate the P2X purinoceptor 7 (P2X7R)/cryopyrin inflammasome axis, thus inducing the proinflammatory cytokine IL-1ß/IL-18 secretion. We therefore hypothesized that inhibition of P2X7R/cryopyrin inflammasome axis would ameliorate neuroinflammation after SAH. In the present study, SAH was induced by the endovascular perforation in rats. Small interfering RNAs (siRNAs) of P2X7R or cryopyrin were administered intracerebroventricularly 24h before SAH. Brilliant blue G (BBG), a non-competitive antagonist of P2X7R, was administered intraperitoneally 30min following SAH. Post-assessments including SAH severity score, neurobehavioral test, brain water content, Western blot and immunofluorescence, were performed. Administration of P2X7R and cryopyrin siRNA as well as pharmacologic blockade of P2X7R by BBG ameliorated neurological deficits and brain edema at 24h following SAH. Inhibition of P2X7R/cryopyrin inflammasome axis suppressed caspase-1 activation, which subsequently decreased maturation of IL-1ß/IL-18. To investigate the link between P2X7R and cryopyrin inflammasome in vivo, Benzoylbenzoyl-ATP (BzATP), a P2X7R agonist, was given to lipopolysaccharide (LPS) primed naive rats with scramble or cryopyrin siRNAs. In LPS-primed naive rats, BzATP induced caspase-1 activation and mature IL-1ß release were neutralized by cryopyrin siRNA. Thus, the P2X7R/cryopyrin inflammasome axis may contribute to neuroinflammation via activation of caspase-1 and thereafter mature IL-1ß/IL-18 production following SAH. Therapeutic interventions targeting P2X7R/cryopyrin pathway may be a novel approach to ameliorate EBI following SAH.


Asunto(s)
Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/complicaciones , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Edema Encefálico/etiología , Proteínas Portadoras , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inyecciones Intraventriculares , Lipopolisacáridos , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR , Inhibidores de Agregación Plaquetaria/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Receptores Purinérgicos P2X7/genética , Colorantes de Rosanilina/farmacología , Transducción de Señal/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico
20.
Transl Stroke Res ; 4(4): 432-46, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23894255

RESUMEN

Subarachnoid hemorrhage (SAH), predominantly caused by a ruptured aneurysm, is a devastating neurological disease that has a morbidity and mortality rate higher than 50%. Most of the traditional in vivo research has focused on the pathophysiological or morphological changes of large-arteries after intracisternal blood injection. This was due to a widely held assumption that delayed vasospasm following SAH was the major cause of delayed cerebral ischemia and poor outcome. However, the results of the CONSCIOUS-1 trial implicated some other pathophysiological factors, independent of angiographic vasospasm, in contributing to the poor clinical outcome. The term early brain injury (EBI) has been coined and describes the immediate injury to the brain after SAH, before onset of delayed vasospasm. During the EBI period, a ruptured aneurysm brings on many physiological derangements such as increasing intracranial pressure (ICP), decreased cerebral blood flow (CBF), and global cerebral ischemia. These events initiate secondary injuries such as blood-brain barrier disruption, inflammation, and oxidative cascades that all ultimately lead to cell death. Given the fact that the reversal of vasospasm does not appear to improve patient outcome, it could be argued that the treatment of EBI may successfully attenuate some of the devastating secondary injuries and improve the outcome of patients with SAH. In this review, we provide an overview of the major advances in EBI after SAH research.


Asunto(s)
Isquemia Encefálica/etiología , Hemorragia Subaracnoidea/complicaciones , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Investigación Biomédica Traslacional , Vasoespasmo Intracraneal/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...