Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
J Neurochem ; 140(5): 776-786, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28054340

RESUMO

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.


Assuntos
Cloridrato de Fingolimode/farmacologia , Hemorragias Intracranianas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Água Corporal/metabolismo , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Cognição/efeitos dos fármacos , Feminino , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/psicologia , Contagem de Leucócitos , Masculino , Oxidiazóis/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Gravidez , Pironas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tiofenos/farmacologia , Proteínas de Junções Íntimas/metabolismo , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
2.
Neurobiol Dis ; 107: 66-72, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28286182

RESUMO

INTRODUCTION: Postoperative cerebral edema is a devastating complication in neurosurgical patients. Loss of blood-brain barrier integrity has been shown to lead to the development of brain edema following neurosurgical procedures. The aim of this study was to evaluate preconditioning with Crotalus helleri venom (Cv-PC) as a potential preventive therapy for reducing postoperative brain edema in the rodent SBI model. C. helleri venom is known to contain phospholipase A2 (PLA2), an enzyme upstream to cyclooxygenase-2 (COX-2) in the inflammatory cascade, acts to increase the production of inflammatory mediators, such as prostaglandins. We hypothesize that Cv-PC will downregulate the response of the COX-2 pathway to injury, thereby reducing the inflammatory response and the development of brain edema after SBI. MATERIALS AND METHODS: 75 male Sprague Dawley rats (280-330g) were divided to the following groups-naïve+vehicle, naïve+Cv-PC, sham, vehicle, Cv-PC, Cv-PC+NS398 (COX-2 inhibitor). Vehicle preconditioned and Cv-PC animals received either three daily subcutaneous doses of saline or C. helleri venom at 72h, 48h, and 24h prior to surgery. In Cv-PC+NS398 animals, NS398 was administered intraperitoneally 1h prior to each Cv-PC injection. Sham-operated animals received craniotomy only, whereas SBI animals received a partial right frontal lobectomy. Neurological testing and brain water content were assessed at 24h and 72h after SBI; COX-2 and PGE2 expression was assessed at 24h postoperatively by Western blot and immunohistochemistry, respectively. RESULTS: At 24h after SBI, the vehicle-treated animals were observed to have increased brain water content (83.1±0.2%) compared to that of sham animals (80.2±0.1%). The brain water content of vehicle-treated animals at 72h post-SBI was elevated at 83.3±0.2%. Cv-PC-treated animals with doses of 10% LD50 had significantly reduced brain water content of 81.92±0.7% and 81.82±0.3% at 24h and 72h, respectively, after SBI compared to that of vehicle-treated animals, while Cv-PC with 5% LD50 doses showed brain water content that trended lower but did not reach statistical significance. At 24h and 72h post-SBI, Cv-PC-treated animals had significantly higher neurological score than vehicle-treated animals. The COX-2 over-expression characterized in SBI was attenuated in Cv-PC-treated animals; NS398 reversed the protective effect of Cv-PC on COX-2 expression. Cv-PC tempered the over-expression of the inflammatory marker PGE2. CONCLUSION: Our findings indicate that Cv-PC may provide a promising therapy for reducing postoperative edema and improving neurological function after neurosurgical procedures.


Assuntos
Edema Encefálico/prevenção & controle , Encéfalo/cirurgia , Lobo Frontal/lesões , Complicações Intraoperatórias/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Complicações Pós-Operatórias/tratamento farmacológico , Venenos de Serpentes/administração & dosagem , Animais , Água Corporal/efeitos dos fármacos , Edema Encefálico/etiologia , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Crotalus , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Epiderme/efeitos dos fármacos , Epiderme/imunologia , Epiderme/patologia , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/patologia , Lobo Frontal/cirurgia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Complicações Intraoperatórias/metabolismo , Complicações Intraoperatórias/patologia , Masculino , Procedimentos Neurocirúrgicos , Nitrobenzenos/farmacologia , Complicações Pós-Operatórias/metabolismo , Complicações Pós-Operatórias/patologia , Ratos Sprague-Dawley , Sulfonamidas/farmacologia
3.
Neurobiol Dis ; 87: 124-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26739391

RESUMO

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.


Assuntos
Antígenos CD36/metabolismo , Hematoma/fisiopatologia , Hemorragias Intracranianas/fisiopatologia , PPAR gama/metabolismo , Anilidas/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Antígenos CD36/genética , Fármacos do Sistema Nervoso Central/farmacologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Hematoma/tratamento farmacológico , Hematoma/patologia , Hemorragias Intracranianas/tratamento farmacológico , Hemorragias Intracranianas/patologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/fisiologia , Microglia/efeitos dos fármacos , Microglia/fisiologia , Fármacos Neuroprotetores/farmacologia , PPAR gama/antagonistas & inibidores , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , RNA Interferente Pequeno/administração & dosagem , Distribuição Aleatória , Ratos Sprague-Dawley , Regulação para Cima
4.
Acta Neurochir Suppl ; 121: 49-53, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463922

RESUMO

Cerebral iron overload contributes to free-radical damage and secondary brain injury following intracerebral hemorrhage (ICH). Phlebotomy most effectively removes iron from the human body, compared with any pharmacological agent (e.g., chelator), and does not impact mean arterial blood pressure. For centuries, this ancient method was a treatment for stroke. This is the first controlled scientific evaluation of this approach after ICH. Femoral catheterization occurred at 30 min following collagenase infusion. Three different exsanguination volumes were tested: 1, 2, 3 ml (approximately 5-15 % (normotensive) loss of total blood volume; or 3.33-10 ml/kg) compared with ICH and sham controls. Brain water content, hemorrhage size, and neuroscore were measured 24 h later. Preliminary analysis of the data demonstrated that therapeutic phlebotomy occurring shortly after ICH in adult rats significantly decreased brain edema and hemorrhagic size at 1 day after the brain injury. However, the neuroscore was unchanged compared with untreated animals. Therefore, exsanguination therapy after ICH using the traditional phlebotomy approach may eventually ameliorate early brain injury (hemorrhage and edema) in further human studies, despite equivocal changes in the short-term neurological functional ability. In meantime, translational studies must further delineate the involvement of specific neuroprotective molecules, sympathetic responses, hemodynamic-vasoactive mediators, or neuroendocrine factors involved in this apparent postconditioning approach following ICH in rodents.


Assuntos
Edema Encefálico/prevenção & controle , Hemorragia Cerebral/terapia , Flebotomia/métodos , Animais , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Exsanguinação , Masculino , Colagenase Microbiana/toxicidade , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença
5.
Acta Neurochir Suppl ; 121: 99-102, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463930

RESUMO

Brain edema is routinely measured using the wet-dry method. Volume, however, is the sum total of all cerebral tissues, including water. Therefore, volumetric change following injury may not be adequately quantified using percentage of edema. We thus tested the hypothesis that dried brains can be reconstituted with water and then re-measured to determine the actual volume. Subarachnoid hemorrhage (SAH) was induced by endovascular perforation in adult male Sprague-Dawley rats (n = 30). Animals were euthanized at 24 and 72 h after evaluation of neurobehavior for determination of brain water content. Dried brains were thereafter reconstituted with equal parts of water (lost from brain edema) and centrifuged to remove air bubbles. The total volume was quantified using hydrostatic (underwater) physics principles that 1 ml water (mass) = 1 cm(3) (volume). The amount of additional water needed to reach a preset level marked on 2-ml test tubes was added to that lost from brain edema, and from the brain itself, to determine the final volume. SAH significantly increased both brain water and volume while worsening neurological function in affected rats. Volumetric measurements demonstrated significant brain swelling after SAH, in addition to the brain edema approach. This modification of the "wet-dry" method permits brain volume determination using valuable post hoc dried brain tissue.


Assuntos
Edema Encefálico/patologia , Encéfalo/patologia , Hemorragia Subaracnóidea/patologia , Animais , Comportamento Animal , Edema Encefálico/fisiopatologia , Modelos Animais de Doenças , Procedimentos Endovasculares , Masculino , Tamanho do Órgão , Punções , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/fisiopatologia
6.
Acta Neurochir Suppl ; 121: 63-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463924

RESUMO

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.


Assuntos
Hemorragia Cerebral/terapia , Pós-Condicionamento Isquêmico/métodos , Animais , Animais Recém-Nascidos , Comportamento Animal , Encéfalo/fisiopatologia , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/fisiopatologia , Modelos Animais de Doenças , Extremidades , Aprendizagem em Labirinto , Colagenase Microbiana/toxicidade , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Teste de Desempenho do Rota-Rod
7.
Acta Neurochir Suppl ; 121: 209-12, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463950

RESUMO

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.


Assuntos
Comportamento Animal/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Hemorragia Cerebral/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Hemorragias Intracranianas/metabolismo , Administração Intranasal , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/metabolismo , Edema Encefálico , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Modelos Animais de Doenças , Hemoglobinas/efeitos dos fármacos , Hemoglobinas/metabolismo , Hidrocefalia , Hemorragias Intracranianas/patologia , Hemorragias Intracranianas/fisiopatologia , Permeabilidade , Ratos , Ratos Sprague-Dawley
8.
Acta Neurochir Suppl ; 121: 203-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463949

RESUMO

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.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Hemorragias Intracranianas/metabolismo , Oligopeptídeos/farmacologia , Pirróis/farmacologia , Quinazolinas/farmacologia , Animais , Animais Recém-Nascidos , Western Blotting , Encéfalo/patologia , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Hemorragias Intracranianas/patologia , Hemorragias Intracranianas/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptor PAR-1/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores
9.
Acta Neurochir Suppl ; 121: 217-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463952

RESUMO

Germinal matrix hemorrhage (GMH) is the most common and devastating neurological problem of premature infants. Current treatment is largely ineffective and GMH has been nonpreventable. Osteopontin (OPN) is an endogenous protein that has been shown to be neuroprotective, however, it has not been tested in GMH. P7 neonatal rats were subjected to stereotactic ganglionic eminence collagenase infusion. Groups were as follows: (1) sham, (2) GMH + vehicle, (3) GMH + intranasal OPN. Seventy-two hours later, the animals were evaluated using righting reflex, blood-brain barrier (BBB) permeability by Evans blue dye leakage, brain water content, and hemoglobin assay. Intranasal OPN improved outcomes after GMH by attenuation of brain swelling, BBB function, re-bleeding, and neurological outcomes. OPN may play an important role in enhancing neuroprotective brain signaling following GMH. These observed effects may offer novel possibilities for therapy in this patient population.


Assuntos
Comportamento Animal/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Hemorragias Intracranianas/metabolismo , Osteopontina/farmacologia , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Edema Encefálico/etiologia , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Hemoglobinas/efeitos dos fármacos , Hemoglobinas/metabolismo , Hemorragias Intracranianas/complicações , Hemorragias Intracranianas/patologia , Hemorragias Intracranianas/fisiopatologia , Ratos , Ratos Sprague-Dawley
10.
Acta Neurochir Suppl ; 121: 213-6, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463951

RESUMO

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.


Assuntos
Encéfalo/efeitos dos fármacos , Hemorragias Intracranianas/metabolismo , Oligopeptídeos/farmacologia , Pirróis/farmacologia , Quinazolinas/farmacologia , Receptor PAR-1/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores , Serina-Treonina Quinases TOR/efeitos dos fármacos , Trombina/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Imunossupressores/farmacologia , Hemorragias Intracranianas/patologia , Hemorragias Intracranianas/fisiopatologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo
11.
Acta Neurochir Suppl ; 121: 263-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463959

RESUMO

Osteopontin (OPN) is a neuroprotective molecule that is upregulated following rodent neonatal hypoxic-ischemic (nHI) brain injury. Because Rac1 is a regulator of blood-brain barrier (BBB) stability, we hypothesized a role for this in OPN signaling. nHI was induced by unilateral ligation of the right carotid artery followed by hypoxia (8 % oxygen for 2 h) in P10 Sprague-Dawley rat pups. Intranasal (iN) OPN was administered at 1 h post-nHI. Groups consisted of: (1) Sham, (2) Vehicle, (3) OPN, and (4) OPN + Rac1 inhibitor (NSC23766). Evans blue dye extravasation (BBB permeability) was quantified 24 h post-nHI, and brain edema at 48 h. Increased BBB permeability and brain edema following nHI was ameliorated in the OPN treatment group. However, those rat pups receiving OPN co-treatment with the Rac1 inhibitor experienced no improvement compared with vehicle. OPN protects the BBB following nHI, and this was reversed by Rac1 inhibitor (NSC23766).


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Edema Encefálico/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Osteopontina/farmacologia , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Aminoquinolinas/farmacologia , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/metabolismo , Artérias Carótidas/cirurgia , Ligadura , Permeabilidade , Pirimidinas/farmacologia , Ratos Sprague-Dawley , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Acta Neurochir Suppl ; 121: 111-4, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463932

RESUMO

The leading cause of morbidity and mortality in infants is hypoxia-ischemia (HI). The current therapies for HI have limited success, in part due to a lack of understanding of HI pathophysiology and underlying mechanisms. Herein, a neonatal rat model of HI was used to examine the changes in brain swelling and infarct volume over 4 days after HI. Forty-four P10 rat pups were sacrificed at 2, 3, or 4 days post-HI. After sacrifice, the brains were removed, sliced, and stained with TTC (2,3,5-triphenyl-2H-tetrazolium chloride). Images of TTC-stained brains were used for measurement of the ipsilateral hemisphere brain volumes and infarct volumes, calculated using standard equations. The hemispheric brain volumes of HI animals in all groups was lower than that of sham animals and decreased as the post-HI sacrifice time increased. The infarct volume of HI animals was larger than that of sham animals. Infarct volumes tended to decrease over the days post-HI. The change in infarct volume is likely the result of a combination of brain growth and repair mechanisms. However, changes in the hemispheric brain volume may include tissue growth and repair mechanism, so also may be a limitation of the current algorithm used for calculating ipsilateral hemisphere brain volume.


Assuntos
Edema Encefálico/patologia , Infarto Encefálico/patologia , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Progressão da Doença , Tamanho do Órgão , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
13.
Stroke ; 46(6): 1710-3, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25931468

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Lesões Encefálicas/tratamento farmacológico , Hemorragia Cerebral/tratamento farmacológico , Imunossupressores/farmacologia , Nitrobenzenos/farmacologia , Receptor PAR-1/antagonistas & inibidores , Receptores de Trombina/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Sulfonamidas/farmacologia , Animais , Animais Recém-Nascidos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patologia , Ciclo-Oxigenase 2/metabolismo , Ratos
14.
J Neurosci Res ; 93(1): 94-103, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25196554

RESUMO

Blood-brain barrier (BBB) disruption and consequent edema formation contribute to the development of early brain injury following subarachnoid hemorrhage (SAH). Various cerebrovascular insults result in increased platelet-derived growth factor receptor (PDGFR)-α stimulation, which has been linked to BBB breakdown and edema formation. This study examines whether imatinib, a PDGFR inhibitor, can preserve BBB integrity in a rat endovascular perforation SAH model. Imatinib (40 or 120 mg/kg) or a vehicle was administered intraperitoneally at 1 hr after SAH induction. BBB leakage, brain edema, and neurological deficits were evaluated. Total and phosphorylated protein expressions of PDGFR-α, c-Src, c-Jun N-terminal kinase (JNK), and c-Jun were measured, and enzymatic activities of matrix metalloproteinase (MMP)-2 and MMP-9 were determined in the injured brain. Imatinib treatment significantly ameliorated BBB leakage and edema formation 24 hr after SAH, which was paralleled by improved neurological functions. Decreased brain expressions of phosphorylated PDGFR-α, c-Src, JNK, and c-Jun as well as reduced MMP-9 activities were found in treated animals. PDGFR-α inhibition preserved BBB integrity following experimental SAH; however, the protective mechanisms remain to be elucidated. Targeting PDGFR-α signaling might be advantageous to ameliorate early brain injury following SAH.


Assuntos
Benzamidas/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Hemorragia Subaracnóidea/patologia , Animais , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Permeabilidade Capilar/fisiologia , Modelos Animais de Doenças , Mesilato de Imatinib , Imunoprecipitação , MAP Quinase Quinase 4/metabolismo , Masculino , Exame Neurológico , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo
15.
Stroke ; 45(3): 828-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24425124

RESUMO

BACKGROUND AND PURPOSE: Transforming growth factor-ß (TGF-ß) overproduction and activation of the TGF-ß pathway are associated with the development of brain injury following germinal matrix hemorrhage (GMH) in premature infants. We examined the effects of GMH on the level of TGF-ß1 in a novel rat collagenase-induced GMH model and determined the effect of inhibition of the TGF receptor I. METHODS: In total, 92 seven-day old (P7) rats were used. Time-dependent effects of GMH on the level of TGF-ß1 and TGF receptor I were evaluated by Western blot. A TGF receptor I inhibitor (SD208) was administered daily for 3 days, starting either 1 hour or 3 days after GMH induction. The effects of GMH and SD208 on the TGF-ß pathway were evaluated by Western blot at day 3. The effects of GMH and SD208 on cognitive and motor function were also assessed. The effects of TGF receptor I inhibition by SD208 on GMH-induced brain injury and underlying molecular pathways were investigated by Western blot, immunofluorescence, and morphology studies 24 days after GMH. RESULTS: GMH induced significant delay in development, caused impairment in both cognitive and motor functions, and resulted in brain atrophy in rat subjects. GMH also caused deposition of both vitronectin (an extracellular matrix protein) and glial fibrillary acidic protein in perilesion areas, associated with development of hydrocephalus. SD208 ameliorated GMH-induced developmental delay, improved cognitive and motor functions, and attenuated body weight loss. SD208 also decreased vitronectin and glial fibrillary acidic protein deposition and decreased GMH-induced brain injury. CONCLUSIONS: Increased level of TGF-ß1 and activation of the TGF-ß pathway associate with the development of brain injury after GMH. SD208 inhibits GMH-induced activation of the TGF-ß pathway and leads to an improved developmental profile, partial recovery of cognitive and motor functions, and attenuation of GMH-induced brain atrophy and hydrocephalus.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/fisiopatologia , Hemorragias Intracranianas/tratamento farmacológico , Hemorragias Intracranianas/fisiopatologia , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/fisiopatologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adulto , Animais , Atrofia , Western Blotting , Ventrículos Cerebrais/patologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Hidrocefalia/etiologia , Hidrocefalia/patologia , Imuno-Histoquímica , Doenças do Sistema Nervoso/etiologia , Exame Neurológico , Gravidez , Pteridinas/farmacologia , Pteridinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Sobrevida , Vitronectina/metabolismo , Redução de Peso/efeitos dos fármacos
16.
Stroke ; 45(8): 2475-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24947291

RESUMO

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.


Assuntos
Encéfalo/efeitos dos fármacos , Desferroxamina/uso terapêutico , Hemorragias Intracranianas/tratamento farmacológico , Aprendizagem em Labirinto/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Desferroxamina/administração & dosagem , Modelos Animais de Doenças , Hemorragias Intracranianas/fisiopatologia , Aprendizagem em Labirinto/fisiologia , Atividade Motora/fisiologia , Ratos , Fatores de Tempo
17.
Stroke ; 44(12): 3587-90, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24149004

RESUMO

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.


Assuntos
Encéfalo/efeitos dos fármacos , Hemorragias Intracranianas/tratamento farmacológico , Isoflurano/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Modelos Animais de Doenças , Hemorragias Intracranianas/induzido quimicamente , Hemorragias Intracranianas/metabolismo , Isoflurano/farmacologia , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptores de Lisoesfingolipídeo/metabolismo , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Esfingosina/uso terapêutico
18.
Crit Care Med ; 41(5): 1266-75, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23388512

RESUMO

OBJECTIVE: Hydrogen inhalation was neuroprotective in several brain injury models. Its mechanisms are believed to be related to antioxidative stress. We investigated the potential neurovascular protective effect of hydrogen inhalation especially effect on mast cell activation in a mouse model of intracerebral hemorrhage. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: One hundred seventy-one 8-week-old male CD-1 mice were used. INTERVENTIONS: Collagenase-induced intracerebral hemorrhage model in 8-week-old male CD-1 mice was used. Hydrogen was administrated via spontaneous inhalation. The blood-brain barrier permeability and neurologic deficits were investigated at 24 and 72 hours after intracerebral hemorrhage. Mast cell activation was evaluated by Western blot and immuno-staining. The effects of hydrogen inhalation on mast cell activation were confirmed in an autologous blood injection model intracerebral hemorrhage. MEASUREMENT AND MAIN RESULTS: At 24 and 72 hours post intracerebral hemorrhage, animals showed blood-brain barrier disruption, brain edema, and neurologic deficits, accompanied with phosphorylation of Lyn kinase and release of tryptase, indicating mast cell activation. Hydrogen treatment diminished phosphorylation of Lyn kinase and release of tryptase, decreased accumulation and degranulation of mast cells, attenuated blood-brain barrier disruption, and improved neurobehavioral function. CONCLUSION: Activation of mast cells following intracerebral hemorrhage contributed to increase of blood-brain barrier permeability and brain edema. Hydrogen inhalation preserved blood-brain barrier disruption by prevention of mast cell activation after intracerebral hemorrhage.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas/prevenção & controle , Hemorragia Cerebral/tratamento farmacológico , Hidrogênio/administração & dosagem , Mastócitos/efeitos dos fármacos , Administração por Inalação , Análise de Variância , Animais , Western Blotting , Edema Encefálico/etiologia , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Movimento Celular/efeitos dos fármacos , Hemorragia Cerebral/complicações , Colagenases/farmacologia , Modelos Animais de Doenças , Hidrogênio/farmacocinética , Peróxido de Hidrogênio/metabolismo , Imuno-Histoquímica , Masculino , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos , Distribuição Aleatória , Valores de Referência , Taxa de Sobrevida
19.
Stroke ; 43(3): 844-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22207510

RESUMO

BACKGROUND AND PURPOSE: Perihematomal edema formation and consequent cell death contribute to the delayed brain injury evoked by intracerebral hemorrhage (ICH). We aimed to evaluate the effect of α7 nicotinic acetylcholine receptor (α7nAChR) stimulation on behavior, brain edema, and neuronal apoptosis. Furthermore, we aimed to determine the role of the proapoptotic glycogen synthase kinase-3ß (GSK-3ß) after experimental ICH. METHODS: Male CD-1 mice (n=109) were subjected to intracerebral infusion of autologous blood (n=88) or sham surgery (n=21). ICH animals received vehicle administration, 4 or 12 mg/kg of α7nAChR agonist PHA-543613, 12 mg/kg of α7nAChR agonist PNU-282987, 6 mg/kg of α7nAChR antagonist methyllycaconitine (MLA), 15 µg/kg of phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin, or PHA-543613 combined with MLA or wortmannin. Behavioral deficits and brain water content were evaluated at 24 and 72 hours after surgery. Western blotting and immunofluorescence staining were used for the quantification and localization of activated Akt (p-Akt), GSK-3ß (p-GSK-3ß), and cleaved caspase-3 (CC3). Neuronal cell death was quantified through terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). RESULTS: α7nAChR stimulation improved neurological outcome and reduced brain edema at 24 and 72 hours after surgery (P<0.05 compared with vehicle). Furthermore, PHA-543613 treatment increased p-Akt and decreased p-GSK-3ß and CC3 expressions in the ipsilateral hemisphere (P<0.05, respectively), which was reversed by MLA and wortmannin. P-Akt, p-GSK-3ß, and CC3 were generally localized in neurons. PHA-543613 reduced neuronal cell death in the perihematomal area (P<0.05). CONCLUSIONS: α7nAChR stimulation improved functional and morphological outcomes after experimental ICH in mice. PHA-543613 reduced the expression of proapoptotic GSK-3ß through the PI3K-Akt signaling pathway.


Assuntos
Benzamidas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hemorragias Intracranianas/tratamento farmacológico , Fármacos Neuroprotetores , Agonistas Nicotínicos/farmacologia , Quinuclidinas/farmacologia , Receptores Nicotínicos/fisiologia , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Western Blotting , Água Corporal/metabolismo , Química Encefálica/efeitos dos fármacos , Edema Encefálico/etiologia , Edema Encefálico/patologia , Caspase 3/fisiologia , Inibidores de Caspase , Imunofluorescência , Glicogênio Sintase Quinase 3 beta , Hemorragias Intracranianas/psicologia , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Receptores Nicotínicos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/psicologia , Resultado do Tratamento , Receptor Nicotínico de Acetilcolina alfa7
20.
Neurobiol Dis ; 46(1): 204-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22300708

RESUMO

Fibroblast growth factors (FGFs) maintain and promote vascular integrity; however whether FGFs protect the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unexplored. In this present study, we hypothesized that exogenous FGF administration attenuates brain injury after ICH, specifically by preserving endothelial adherens junctions, therefore reducing vasogenic brain edema and attenuating neurofunctional deficits in mice subjected to experimental ICH. Acid fibroblast growth factor (FGF1) or basic fibroblast growth factor (FGF2) was administered intracerebroventricularly (ICV) at 0.5 h after intrastriatal injection of bacterial collagenase (cICH) or autologous whole blood (bICH). Fibroblast growth factor receptor (FGFR) inhibitor PD173074 and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 were additionally administered with FGF2. The selective Rho-associated coiled-coil forming protein serine/threonine kinase (ROCK) inhibitor Y27632 was independently administered at 0.5 h after cICH. Brain water content and neurofunctional deficits were evaluated at 24 and 72h after ICH induction. Evans blue extravasation as well as Western blot analysis for the quantification of activated FGFR, Akt, Ras-related C3 botulinum toxin substrate 1 (Rac1), Ras homolog gene family member A (RhoA) and adherens junction proteins (p120-catenin, ß-catenin and VE-cadherin) were conducted at 72 h post-cICH. FGF treatment reduced perihematomal brain edema and improved neurofunctional deficits at 72 h after experimental ICH (p<0.05, compared to vehicle); however, FGFR and PI3K inhibition reversed these neuroprotective effects. Exogenous FGF2 increased activated FGFR, Akt, and Rac1 but reduced activated RhoA protein expression at 72 h after cICH (p<0.05, compared to vehicle), which was reversed by FGFR and PI3K inhibition. Y27632 treatment reduced brain injury at 72 h after cICH (p<0.05, compared to vehicle) and increased the expression of catenins (p120-catenin, ß-catenin). In conclusion, our findings suggest that exogenous FGF treatment reduced RhoA activity via FGFR-induced activation of the PI3K-Akt-Rac1 signaling pathway, thus preserving BBB integrity, and therefore attenuating secondary brain injury after experimental ICH in mice.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Hemorragia Cerebral Traumática/tratamento farmacológico , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Animais , Barreira Hematoencefálica/fisiologia , Hemorragia Cerebral Traumática/metabolismo , Hemorragia Cerebral Traumática/fisiopatologia , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas Recombinantes/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa