Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 135(5): 943-57, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26375300

RESUMO

Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) have an extraordinary capacity to withstand prolonged and profound reductions in blood flow and oxygen delivery to the brain without incurring any cellular damage. As such, the hibernation torpor of I. tridecemlineatus provides a valuable model of tolerance to ischemic stress. Herein, we report that during hibernation torpor, a marked reduction in the phosphorylation of the ribosomal protein S6 (rpS6) occurs within the brains of I. tridecemlineatus. Of note, rpS6 phosphorylation was shown to increase in the brains of rats that underwent an occlusion of the middle cerebral artery. However, such an increase was attenuated after the implementation of an ischemic preconditioning paradigm. In addition, cultured cortical neurons treated with the rpS6 kinase (S6K) inhibitors, D-glucosamine or PF4708671, displayed a decrease in rpS6 phosphorylation and a subsequent increase in tolerance to oxygen/glucose deprivation, an in vitro model of ischemic stroke. Collectively, such evidence suggests that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning. Further identification and characterization of the mechanisms used by hibernating species to increase ischemic tolerance may eventually clarify how the loss of homeostatic control that occurs during and after cerebral ischemia in the clinic can ultimately be minimized and/or prevented. Mammalian hibernation provides a valuable model of tolerance to ischemic stress. Herein, we demonstrate that marked reductions in the phosphorylation of ribosomal protein S6 (rpS6), extracellular signal-regulated kinase family of mitogen-activated protein (MAP) kinase p44/42 (p44/42MAPK) and ribosomal protein S6 kinase (S6K) occur within the brains of both hibernating squirrels and rats, which have undergone an ischemic preconditioning paradigm. We therefore propose that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning, via a suppression of protein synthesis and/or energy consumption.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hibernação , Infarto da Artéria Cerebral Média/prevenção & controle , Precondicionamento Isquêmico , Proteína S6 Ribossômica/metabolismo , Animais , Células Cultivadas , Córtex Cerebral , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Glucose/deficiência , Masculino , Dados de Sequência Molecular , Neurônios/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteína S6 Ribossômica/genética , Sciuridae , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Tempo
2.
Brain Res ; 1388: 148-56, 2011 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-21376021

RESUMO

Toll-like receptor (TLR) signaling plays an important role in cerebral ischemia, but downstream signaling events, which can be organ-specific, are incompletely understood. We thereby investigated involvement of the MyD88-dependent (MyD88) and MyD88-independent (TRIF) TLR signaling pathways in 2 in vitro and 2 in vivo models of cerebral ischemia. For in vitro studies, we used a model of oxygen-glucose deprivation (OGD) followed by flow cytometric analysis to determine:1) viability of PC12 cells following knock-down with MyD88 siRNA compared to negative control siRNA and 2) viability, apoptosis and necrosis of cortical neurons from MyD88 null (-/-) , TRIF mutant, and wild type (WT) mice. In addition, in vivo, 1) We examined CA1 neuronal survival 7 days after global forebrain ischemia and 2) infarct volumes 24h after Middle Cerebral Artery Occlusion (MCAO) in all 3 types of mice. OGD: 1) There were no differences in either percent viability of PC12 cells transfected with MyD88 compared to negative control siRNA or 2) in percent viability, apoptosis and necrosis of cortical neurons from MyD88-/-,TRIF mutant and WT mice. Global ischemia: neuronal survival was similar in all 3 groups of mice. Finally, MCAO: infarct volumes were not statistically different among all 3 groups of mice: MyD88-/-, 23.9±9.9 mm(3), TRIF mutant, 26.7±5.8 mm(3) and WT, 17.9±8.4mm(3). These findings show that disruption of MyD88 or TRIF signaling does not confer protection in brain ischemia and suggests the possibility of additional or alternate downstream adaptors during TLR signaling in cerebral ischemia.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Isquemia Encefálica/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Western Blotting , Separação Celular , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necrose/metabolismo , Células PC12 , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Ratos
3.
J Cereb Blood Flow Metab ; 30(6): 1188-99, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20197780

RESUMO

In this study, we examined the relationship between tissue and blood levels of matrix metalloproteinase (MMP)-2 and MMP-9 through gelatin zymography at multiple time points after experimental stroke. We additionally investigated the association between these levels and the evidence of blood-cerebrospinal fluid (CSF) barrier (BCSFB) and blood-brain barrier (BBB) disruption on post-contrast fluid-attenuated inversion-recovery (FLAIR) imaging. Increased plasma MMP-9 was associated with BCSFB disruption at 1h post-reperfusion. Ventricular enhancement ipsilateral to the stroke was 500+/-100%, significantly higher than sham, 24, and 48 h groups. Increased tissue MMP-2 and MMP-9 were associated with BBB disruption at 48 h post-reperfusion. Parenchymal enhancement was 60+/-20% for a volume equivalent to 260+/-80 mm(3). Although the percent enhancement was comparable across groups, the volume of enhancing lesion was significantly higher at 48 h (260+/-80 mm(3), 100%) in comparison to 1 h (8+/-3 mm(3), 3%) and 24 h (51 mm(3), 18%). These findings support the use of imaging markers of BCSFB and BBB status as indirect measures of MMP regulation in the blood and brain tissue. The methods presented herein should be useful in understanding the link between MMPs, barrier integrity, and subsequent hemorrhagic transformation.


Assuntos
Barreira Hematoencefálica/metabolismo , Metaloproteinase 2 da Matriz/sangue , Metaloproteinase 9 da Matriz/sangue , Acidente Vascular Cerebral/sangue , Animais , Biomarcadores/sangue , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Endogâmicos SHR , Fatores de Tempo
4.
J Cereb Blood Flow Metab ; 29(7): 1229-39, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19417758

RESUMO

At this time, the pathophysiology of macrophage involvement and their role in stroke progression are poorly understood. Recently, T2- and T2*-weighted magnetic resonance imaging (MRI), after intravenous administration of iron-oxide particles, have been used to understand the inflammatory cascade. Earlier studies report that image enhancement after stroke is from iron-laden macrophages; however, they do not account for potential blood-brain barrier disruption and nonspecific contrast enhancement. In this study, spontaneously hypertensive rats were preloaded with Feridex 7 days before stroke, permitting the labeling of bone-marrow-derived macrophages. Three-dimensional gradient-echo imaging showed average signal decreases of 13% to 23% in preloaded animals, concentrated on the lesion periphery and reaching a maximum on days 2 to 4 after stroke. Immunohistochemistry showed ED-2+, PB+, MHC-II+ and TNF-alpha+ perivascular macrophages (PVM), meningeal macrophages (MM), and choroid plexus macrophages (CPM). ED-1+ and IBA+ tissue macrophages and/or activated microglia were located throughout the lesion, but were PB-. These findings indicate the following: (1) Feridex preloading permits tracking of the central nervous system (CNS)-resident macrophages (PVM, MM, and CPM) and (2) CNS-resident macrophages likely play an integral role in the inflammatory cascade through antigen presentation and expression of proinflammatory cytokines. Further refinement of this method should permit noninvasive monitoring of inflammation and potential evaluation of antiinflammatory therapies in preclinical models of stroke.


Assuntos
Sistema Nervoso Central/imunologia , Ferro , Macrófagos/fisiologia , Imageamento por Ressonância Magnética/métodos , Óxidos , Acidente Vascular Cerebral/imunologia , Animais , Meios de Contraste , Dextranos , Óxido Ferroso-Férrico , Infarto da Artéria Cerebral Média/imunologia , Inflamação/imunologia , Macrófagos/patologia , Nanopartículas de Magnetita , Masculino , Métodos , Técnicas de Sonda Molecular , Ratos , Ratos Endogâmicos SHR
5.
J Cereb Blood Flow Metab ; 29(3): 606-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19107136

RESUMO

Neuroblasts in the subventricular zone (SVZ) proliferate markedly after brain ischemia, and migrate to the site of injury along with blood vessels. However, a large fraction of stroke-generated neuroblasts die shortly after being born, in part, because of local inflammation. In spontaneously hypertensive rats (SHRs) subjected to permanent middle cerebral artery occlusion, we primed E-selectin-specific regulatory T cells (Tregs) by repetitive intranasal administration of recombinant E-selectin to target local secretion of immunomodulating, antiinflammatory cytokines to activating blood vessel segments. E-selectin-tolerized SHRs had decreased infarction volumes, and increased numbers of Tregs in the cervical lymph nodes and ischemic brain. The brain Tregs were distributed primarily in periinfarct regions. E-selectin tolerization did not alter cellular proliferation in the ipsilateral SVZ after stroke, but the expression of tumor necrosis factor on vascular niche blood vessels was suppressed and both doublecortin protein levels and the number of newly generated neuroblasts or neurons were increased in the brain. This enhanced survival of neural progenitor cells and neurons was paralleled by improved functional performance. These studies suggest that E-selectin-specific Tregs can modulate the efficacy of neurogenesis after ischemia and promote repair after brain injury.


Assuntos
Selectina E/imunologia , Mucosa Nasal/imunologia , Neurônios/patologia , Células-Tronco/patologia , Acidente Vascular Cerebral/prevenção & controle , Linfócitos T/imunologia , Administração Intranasal , Animais , Morte Celular , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Proteína Duplacortina , Selectina E/administração & dosagem , Tolerância Imunológica , Imunidade nas Mucosas/imunologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/imunologia , Neurônios/imunologia , Ratos , Ratos Endogâmicos SHR , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Células-Tronco/imunologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
6.
J Cereb Blood Flow Metab ; 28(2): 341-53, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17637705

RESUMO

Vascular cognitive impairment (VCI) is the second most prevalent type of dementia in the world. The white matter damage that characterizes the common subcortical ischemic form of VCI can be modeled by ligating both common carotid arteries in the Wistar rat to induce protracted cerebral hypoperfusion. In this model, we find that repetitive intranasal administration of recombinant E-selectin to induce mucosal tolerance and to target immunomodulation to activating blood vessels potently suppresses both white matter (and possibly gray matter) damage and markers of vessel activation (tumor necrosis factor and E-selectin); it also preserves behavioral function in T-maze spontaneous alternation, T-maze spatial discrimination memory retention, and object recognition tests. Immunomodulation may be an effective novel strategy to prevent progression of VCI.


Assuntos
Encéfalo/patologia , Demência Vascular/prevenção & controle , Selectina E/farmacologia , Tolerância Imunológica/fisiologia , Imunidade nas Mucosas/fisiologia , Transtornos da Memória/prevenção & controle , Animais , Demência Vascular/patologia , Demência Vascular/fisiopatologia , Discriminação Psicológica/efeitos dos fármacos , Feminino , Hipersensibilidade Tardia/fisiopatologia , Tolerância Imunológica/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Imunoensaio , Fatores Imunológicos/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Ratos , Ratos Wistar , Reconhecimento Psicológico/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
7.
Brain Res ; 1132(1): 177-84, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17188657

RESUMO

Evidence that inflammatory and immune mechanisms may have a critical role in the development of vasospasm after subarachnoid hemorrhage is accumulating. We examined, therefore, whether induction of immunological tolerance to the adhesion molecule that is uniquely expressed on activated endothelium, E-selectin, could inhibit the vasospasm provoked by subarachnoid blood in a rat subarachnoid hemorrhage model. We found that intranasal instillation of E-selectin every other day for 10 days on a mucosal tolerization schedule suppressed delayed type hypersensitivity to E-selectin confirming tolerance to that molecule and markedly suppressed basilar artery spasm after subarachnoid hemorrhage. The results of this proof-of-concept study suggest that agents that can mimic the local effects of the mediators of mucosal tolerance could have therapeutic potential for the management of post-subarachnoid hemorrhage vasospasm.


Assuntos
Selectina E/farmacologia , Encefalite/complicações , Tolerância Imunológica/efeitos dos fármacos , Terapia de Imunossupressão/métodos , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/tratamento farmacológico , Administração Intranasal , Animais , Artéria Basilar/efeitos dos fármacos , Artéria Basilar/imunologia , Artéria Basilar/fisiopatologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Selectina E/imunologia , Encefalite/imunologia , Encefalite/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Hipersensibilidade Tardia/imunologia , Hipersensibilidade Tardia/fisiopatologia , Tolerância Imunológica/imunologia , Masculino , Ratos , Ratos Endogâmicos SHR , Hemorragia Subaracnóidea/imunologia , Hemorragia Subaracnóidea/fisiopatologia , Resultado do Tratamento , Vasoespasmo Intracraniano/imunologia , Vasoespasmo Intracraniano/fisiopatologia
8.
Neurosci Lett ; 411(3): 200-5, 2007 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-17127001

RESUMO

Peripheral benzodiazepine receptors (PBRs) are upregulated on activated microglia. We recently developed a promising positron emission tomography (PET) ligand, [11C]PBR28, with high affinity and excellent ratio of specific to nonspecific binding. We assessed the ability of [11C]PBR28 PET to localize PBRs in a rat permanent middle cerebral artery occlusion (MCAO) model of neuroinflammation. [11C]PBR28 was intravenously administered to rats at 4 and 7 days after permanent MCAO. In all experiments, arterial blood was sampled for compartmental modeling of regional distribution volumes, and rat brains were sampled after imaging for in vitro [3H]PK 11195 autoradiography and histological evaluation. [11C]PBR28 PET and [3H]PK 11195 autoradiography showed similar areas of increased PBRs, especially in the peri-ischemic core. Results from these in vivo and in vitro methods were strongly correlated. In this first study to demonstrate neuroinflammation in vivo with small animal PET, [11C]PBR28 had adequate sensitivity to localize and quantify the associated increase in PBRs.


Assuntos
Acetamidas/farmacocinética , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/metabolismo , Éteres Fenílicos/farmacocinética , Tomografia por Emissão de Pósitrons , Receptores de GABA-A/metabolismo , Animais , Autorradiografia/métodos , Isquemia Encefálica/patologia , Isótopos de Carbono/farmacocinética , Processamento de Imagem Assistida por Computador/métodos , Isoquinolinas/farmacocinética , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Distribuição Tecidual
9.
J Cereb Blood Flow Metab ; 25(2): 154-162, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15647748

RESUMO

Although ischemic tolerance has been described in a variety of primary cell culture systems, no similar in vitro models have been reported with any cell line. A model of ischemic preconditioning in the rat pheochromocytoma PC12 cell line is described here. When compared to nonpreconditioned cells, preexposure of PC12 cells to 6 hours of oxygen and glucose deprivation (OGD) significantly increased cell viability after 15 hours of OGD 24 hours later. Flow cytometry analysis of cells labeled with specific markers for apoptosis, Annexin V, and Hoechst 33342, and of DNA content, revealed that apoptosis is involved in OGD-induced PC12 cell death and that preconditioning of the cells mainly counteracts the effect of apoptosis. Immunocytochemistry of caspase-3, a central executioner in the apoptotic process, further confirmed the activation of apoptotic pathways in OGD-induced PC12 cell death. This model may be useful to investigate the cellular mechanisms involved in neuronal transient tolerance following ischemia.


Assuntos
Precondicionamento Isquêmico/métodos , Modelos Biológicos , Células PC12/patologia , Animais , Anexina A5/metabolismo , Apoptose/fisiologia , Benzimidazóis/metabolismo , Caspase 3 , Caspases/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Ratos
10.
Proc Natl Acad Sci U S A ; 100(25): 15107-12, 2003 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-14645708

RESUMO

We have demonstrated that induction of mucosal tolerance to E-selectin, a cytokine-inducible adhesion molecule restricted to activating blood vessels, prevents ischemic and hemorrhagic stroke in spontaneously hypertensive, genetically stroke-prone (SHR-SP) rats. We now examine whether mucosal tolerance to E-selectin has protective effects in ischemic brain damage after permanent middle cerebral artery occlusion (MCAO) in SHR-SP rats and whether these effects are related to generation of regulatory T cells. Rats were exposed to intranasal administration of E-selectin every other day for 10 days (single tolerization group) or on two tolerization schedules separated by 11 days (booster tolerization group). Control groups received PBS on corresponding schedules. MCAO was performed 48 h after the last dose of E-selectin or PBS. There were 45.8% and 37.9% (P < 0.05) decreases of infarction volume in the E-selectin booster group compared with the PBS group at 6 and 48 h, respectively. Single tolerization with E-selectin had only a slight trend toward a decrease in infarction volume (6.3%). CD8-positive cells were decreased in brains of E-selectin booster animals (46.6%, P < 0.01) compared with controls; splenocyte-culture supernatant levels of IL-10 were increased (59.3%, P < 0.05) in E-selectin booster animals. A decrease of infarction volume (34%, P < 0.05) was also observed in SHR-SP rats subjected to MCAO after adoptive transfer of splenocytes from E-selectin-tolerized compared with PBS-tolerized donors. The results indicate that, in addition to preventing stroke, mucosal tolerance to E-selectin is cytoprotective. Thus, immunomodulation targeted to activated blood vessel segments can both reduce stroke occurrence and attenuate brain damage if a stroke supervenes.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/patologia , Selectina E/química , Selectina E/imunologia , Infarto da Artéria Cerebral Média/prevenção & controle , Animais , Peso Corporal , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Selectina E/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Interleucina-10/metabolismo , Fluxometria por Laser-Doppler , Antígenos Comuns de Leucócito/biossíntese , Masculino , Infarto do Miocárdio , Ratos , Ratos Endogâmicos SHR , Linfócitos T/metabolismo , Fatores de Tempo
11.
J Cereb Blood Flow Metab ; 23(6): 748-55, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12796723

RESUMO

Brain cells produce cytokines and chemokines during the inflammatory process after stroke both in animal models and in patients. Monocyte chemoattractant protein 1 (MCP-1), one of the proinflammatory chemokines, can attract monocytes to the tissue where MCP-1 is overexpressed. However, the role of MCP-1 elevation in stroke has not been explored in detail. The authors hypothesized that elevated MCP-1 levels would lead to increased influx of monocytes and increased brain infarction size in stroke induced by middle cerebral artery occlusion with partial reperfusion. There were no differences in blood pressure, blood flow, or vascular architecture between wild-type mice and transgenic MBP-JE mice. Twenty-four to 48 hours after middle cerebral artery occlusion, brain infarction volumes after ischemia were significantly larger in MBP-JE mice than in wild-type controls and were accompanied by increased local transmigration and perivascular accumulation of macrophages and neutrophils. These results indicate that MCP-1 can contribute to inflammatory injury in stroke.


Assuntos
Quimiocina CCL2/genética , Infarto da Artéria Cerebral Média/patologia , Ataque Isquêmico Transitório/patologia , Macrófagos/patologia , Neutrófilos/patologia , Animais , Pressão Sanguínea , Encéfalo/imunologia , Encéfalo/patologia , Circulação Cerebrovascular , Círculo Arterial do Cérebro/anatomia & histologia , Modelos Animais de Doenças , Feminino , Expressão Gênica/imunologia , Infarto da Artéria Cerebral Média/imunologia , Ataque Isquêmico Transitório/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Artéria Cerebral Média/anatomia & histologia , Monócitos/imunologia , Monócitos/patologia , Neutrófilos/imunologia , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia
12.
Stroke ; 33(9): 2156-63, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12215580

RESUMO

BACKGROUND AND PURPOSE: Inflammatory and immune mechanisms can precipitate cerebrovascular thrombosis and hemorrhage. Immunologic tolerance can be induced to a specific antigen by intranasal instillation of that antigen. Lymphocytes tolerized in this way provide local immunosuppression on restimulation with the same antigen. This study tests whether tolerization of lymphocytes to E-selectin can suppress local vessel activation and prevent stroke. METHODS: Spontaneously hypertensive genetically stroke-prone rats (n=113) were distributed among the following studies: comparison of ischemic infarcts/intraparenchymal hemorrhages after single or repetitive tolerization schedules with ovalbumin, E-selectin, or PBS; comparison of E-selectin tolerization- and PBS tolerization-induced suppression of delayed-type hypersensitivity in animals subsequently sensitized to E-selectin; and comparison of PBS-, ovalbumin-, and E-selectin-tolerized groups (after intravenous lipopolysaccharide to activate vessels) regarding transforming growth factor-beta1-positive splenocyte counts, plasma interferon-gamma levels, anti-human E-selectin antibodies, endothelial intercellular adhesion molecule-1, and anti-endothelial cell antibodies. RESULTS: Nasal instillation of E-selectin, which is specifically expressed on activated endothelium, potently inhibited the development of ischemic and hemorrhagic strokes in spontaneously hypertensive stroke-prone rats with untreated hypertension. Repeated schedules of tolerization were required to maintain the resistance to stroke. Suppression of delayed-type hypersensitivity to E-selectin and increased numbers of transforming growth factor-beta1-positive splenocytes showed that intranasal exposure to E-selectin induced immunologic tolerance. E-selectin tolerization also reduced endothelial activation and immune responses after intravenous lipopolysaccharide, as shown by marked suppression of intercellular adhesion molecule-1 expression, anti-endothelial cell antibodies on luminal endothelium, and plasma interferon-gamma levels compared with the control condition. CONCLUSIONS: The novel findings in this study support further investigation of immunologic tolerance as applied to the prevention of stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Córtex Cerebral/efeitos dos fármacos , Hemorragia Cerebral/prevenção & controle , Selectina E/imunologia , Imunidade nas Mucosas/imunologia , Acidente Vascular Cerebral/prevenção & controle , Animais , Isquemia Encefálica/complicações , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/patologia , Hemorragia Cerebral/complicações , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/patologia , Avaliação Pré-Clínica de Medicamentos , Selectina E/administração & dosagem , Predisposição Genética para Doença , Hipersensibilidade Tardia/imunologia , Tolerância Imunológica , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Instilação de Medicamentos , Lipopolissacarídeos/administração & dosagem , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Mucosa Nasal/imunologia , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ratos , Ratos Endogâmicos SHR , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia , Taxa de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA