Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Stroke ; 43(5): 1383-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22403050

RESUMO

BACKGROUND AND PURPOSE: Systemic administration of Toll-like receptor (TLR) 4 and TLR9 agonists before cerebral ischemia have been shown to reduce ischemic injury by reprogramming the response of the brain to stroke. Our goal was to explore the mechanism of TLR-induced neuroprotection by determining whether a TLR7 agonist also protects against stroke injury. METHODS: C57Bl/6, TNF(-/-), interferon (IFN) regulatory factor 7(-/-), or type I IFN receptor (IFNAR)(-/-) mice were subcutaneously administered the TLR7 agonist Gardiquimod (GDQ) 72 hours before middle cerebral artery occlusion. Infarct volume and functional outcome were determined after reperfusion. Plasma cytokine responses and induction of mRNA for IFN-related genes in the brain were measured. IFNAR(-/-) mice also were treated with the TLR4 agonist (lipopolysaccharide) or the TLR9 agonist before middle cerebral artery occlusion and infarct volumes measured. RESULTS: The results show that GDQ reduces infarct volume as well as functional deficits in mice. GDQ pretreatment provided robust neuroprotection in TNF(-/-) mice, indicating that TNF was not essential. GDQ induced a significant increase in plasma IFNα levels and both IRF7(-/-) and IFNAR(-/-) mice failed to be protected, implicating a role for IFN signaling in TLR7-mediated protection. CONCLUSIONS: Our studies provide the first evidence that TLR7 preconditioning can mediate neuroprotection against ischemic injury. Moreover, we show that the mechanism of protection is unique from other TLR preconditioning ligands in that it is independent of TNF and dependent on IFNAR.


Assuntos
Aminoquinolinas/uso terapêutico , Encéfalo/irrigação sanguínea , Imidazóis/uso terapêutico , Precondicionamento Isquêmico/métodos , Glicoproteínas de Membrana/agonistas , Fármacos Neuroprotetores/uso terapêutico , Receptor de Interferon alfa e beta/fisiologia , Acidente Vascular Cerebral/prevenção & controle , Receptor 7 Toll-Like/agonistas , Animais , Infarto Encefálico/patologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia
2.
J Neuroinflammation ; 8: 140, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21999375

RESUMO

BACKGROUND: Toll-like receptor 4 (TLR4) is activated in response to cerebral ischemia leading to substantial brain damage. In contrast, mild activation of TLR4 by preconditioning with low dose exposure to lipopolysaccharide (LPS) prior to cerebral ischemia dramatically improves outcome by reprogramming the signaling response to injury. This suggests that TLR4 signaling can be altered to induce an endogenously neuroprotective phenotype. However, the TLR4 signaling events involved in this neuroprotective response are poorly understood. Here we define several molecular mediators of the primary signaling cascades induced by LPS preconditioning that give rise to the reprogrammed response to cerebral ischemia and confer the neuroprotective phenotype. METHODS: C57BL6 mice were preconditioned with low dose LPS prior to transient middle cerebral artery occlusion (MCAO). Cortical tissue and blood were collected following MCAO. Microarray and qtPCR were performed to analyze gene expression associated with TLR4 signaling. EMSA and DNA binding ELISA were used to evaluate NFκB and IRF3 activity. Protein expression was determined using Western blot or ELISA. MyD88-/- and TRIF-/- mice were utilized to evaluate signaling in LPS preconditioning-induced neuroprotection. RESULTS: Gene expression analyses revealed that LPS preconditioning resulted in a marked upregulation of anti-inflammatory/type I IFN-associated genes following ischemia while pro-inflammatory genes induced following ischemia were present but not differentially modulated by LPS. Interestingly, although expression of pro-inflammatory genes was observed, there was decreased activity of NFκB p65 and increased presence of NFκB inhibitors, including Ship1, Tollip, and p105, in LPS-preconditioned mice following stroke. In contrast, IRF3 activity was enhanced in LPS-preconditioned mice following stroke. TRIF and MyD88 deficient mice revealed that neuroprotection induced by LPS depends on TLR4 signaling via TRIF, which activates IRF3, but does not depend on MyD88 signaling. CONCLUSION: Our results characterize several critical mediators of the TLR4 signaling events associated with neuroprotection. LPS preconditioning redirects TLR4 signaling in response to stroke through suppression of NFκB activity, enhanced IRF3 activity, and increased anti-inflammatory/type I IFN gene expression. Interestingly, this protective phenotype does not require the suppression of pro-inflammatory mediators. Furthermore, our results highlight a critical role for TRIF-IRF3 signaling as the governing mechanism in the neuroprotective response to stroke.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Isquemia Encefálica , Fator Regulador 3 de Interferon/imunologia , Precondicionamento Isquêmico , Lipopolissacarídeos/farmacologia , Transdução de Sinais , Acidente Vascular Cerebral , Receptor 4 Toll-Like/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Isquemia Encefálica/sangue , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Quimiocinas/sangue , Quimiocinas/imunologia , Citocinas/sangue , Citocinas/imunologia , Perfilação da Expressão Gênica , Humanos , Infarto da Artéria Cerebral Média , Fator Regulador 3 de Interferon/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , NF-kappa B/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
3.
J Cereb Blood Flow Metab ; 28(6): 1235-48, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18364727

RESUMO

Osteopontin (OPN), a large secreted glycoprotein with an arginine, glycine, aspartate (RGD) motif, can bind and signal through cellular integrin receptors. We have shown previously that OPN enhances neuronal survival in the setting of ischemia. Here, we sought to increase the neuroprotective potency of OPN and improve the method of delivery with the goal of identifying a treatment for stroke in humans. We show that thrombin cleavage of OPN improves its ability to ligate integrin receptors and its neuroprotective capacity in models of ischemia. Thrombin-cleaved OPN is a twofold more effective neuroprotectant than the untreated molecule. We also tested whether OPN could be administered intranasally and found that it is efficiently targeted to the brain via intranasal delivery. Furthermore, intranasal administration of thrombin-treated OPN confers protection against ischemic brain injury. Osteopontin mimetics based on the peptide sequences located either N or C terminal to the thrombin cleavage site were generated and tested in models of ischemia. Treatment with successively shorter N-terminal peptides and a phosphorylated C-terminal peptide provided significant neuroprotection against ischemic injury. These findings show that OPN mimetics offer promise for development into new drugs for the treatment of stroke.


Assuntos
Materiais Biomiméticos/administração & dosagem , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Osteopontina/administração & dosagem , Osteopontina/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Administração Intranasal , Sequência de Aminoácidos , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/uso terapêutico , Células Cultivadas , Citoproteção/efeitos dos fármacos , Feminino , Humanos , Integrinas/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Osteopontina/química , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/uso terapêutico , Fosforilação/efeitos dos fármacos , Ligação Proteica , Ratos , Acidente Vascular Cerebral/patologia , Trombina/farmacologia , Fatores de Tempo
4.
J Cereb Blood Flow Metab ; 28(5): 1040-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18183029

RESUMO

Preconditioning with lipopolysaccharide (LPS), a toll-like receptor 4 (TLR4) ligand, provides neuroprotection against subsequent cerebral ischemic brain injury, through a tumor necrosis factor (TNF)alpha-dependent process. Here, we report the first evidence that another TLR, TLR9, can induce neuroprotection. We show that the TLR9 ligand CpG oligodeoxynucleotide (ODN) can serve as a potent preconditioning stimulus and provide protection against ischemic brain injury. Our studies show that systemic administration of CpG ODN 1826 in advance of brain ischemia (middle cerebral artery occlusion (MCAO)) reduces ischemic damage up to 60% in a dose- and time-dependent manner. We also offer evidence that CpG ODN preconditioning can provide direct protection to cells of the central nervous system, as we have found marked neuroprotection in modeled ischemia in vitro. Finally, we show that CpG preconditioning significantly increases serum TNFalpha levels before MCAO and that TNFalpha is required for subsequent reduction in damage, as mice lacking TNFalpha are not protected against ischemic injury by CpG preconditioning. Our studies show that preconditioning with a TLR9 ligand induces neuroprotection against ischemic injury through a mechanism that shares common elements with LPS preconditioning via TLR4.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Precondicionamento Isquêmico/métodos , Lipopolissacarídeos/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptor Toll-Like 9/metabolismo , Animais , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Oligodesoxirribonucleotídeos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Stroke ; 38(9): 2569-76, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17690312

RESUMO

BACKGROUND AND PURPOSE: Mild hypothermia confers profound neuroprotection in ischemia. We recently discovered 2 natural derivatives of thyroxine, 3-iodothyronamine (T(1)AM) and thyronamine (T(0)AM), that when administered to rodents lower body temperature for several hours without induction of a compensatory homeostatic response. We tested whether T(1)AM- and T(0)AM-induced hypothermia protects against brain injury from experimental stroke. METHODS: We tested T(1)AM and T(0)AM 1 hour after and 2 days before stroke in a mouse model of focal ischemia. To determine whether T(1)AM and T(0)AM require hypothermia to protect against stroke injury, the induction of hypothermia was prevented. RESULTS: T(1)AM and T(0)AM administration reduced body temperature from 37 degrees C to 31 degrees C. Mice given T(1)AM or T(0)AM after the ischemic period had significantly smaller infarcts compared with controls. Mice preconditioned with T(1)AM before ischemia displayed significantly smaller infarcts compared with controls. Pre- and postischemia treatments required the induction of hypothermia. T(1)AM and T(0)AM treatment in vitro failed to confer neuroprotection against ischemia. CONCLUSIONS: T(1)AM and T(0)AM, are potent neuroprotectants in acute stroke and T(1)AM can be used as antecedent treatment to induce neuroprotection against subsequent ischemia. Hypothermia induced by T(1)AM and T(0)AM may underlie neuroprotection. T(1)AM and T(0)AM offer promise as treatments for brain injury.


Assuntos
Hipotermia/induzido quimicamente , Fármacos Neuroprotetores , Acidente Vascular Cerebral/patologia , Tironinas , Tiroxina/análogos & derivados , Animais , Comportamento Animal/fisiologia , Temperatura Corporal , Isquemia Encefálica/patologia , Células Cultivadas , Humanos , Precondicionamento Isquêmico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Neurônios/citologia , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Acidente Vascular Cerebral/prevenção & controle , Tironinas/química , Tironinas/metabolismo , Tironinas/farmacologia
6.
J Cereb Blood Flow Metab ; 27(10): 1663-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17327883

RESUMO

Lipopolysaccharide (LPS) preconditioning provides neuroprotection against subsequent cerebral ischemic injury. Tumor necrosis factor-alpha (TNFalpha) is protective in LPS-induced preconditioning yet exacerbates neuronal injury in ischemia. Here, we define dual roles of TNFalpha in LPS-induced ischemic tolerance in a murine model of stroke and in primary neuronal cultures in vitro, and show that the cytotoxic effects of TNFalpha are attenuated by LPS preconditioning. We show that LPS preconditioning significantly increases circulating levels of TNFalpha before middle cerebral artery occlusion in mice and show that TNFalpha is required to establish subsequent neuroprotection against ischemia, as mice lacking TNFalpha are not protected from ischemic injury by LPS preconditioning. After stroke, LPS preconditioned mice have a significant reduction in the levels of TNFalpha (approximately threefold) and the proximal TNFalpha signaling molecules, neuronal TNF-receptor 1 (TNFR1), and TNFR-associated death domain (TRADD). Soluble TNFR1 (s-TNFR1) levels were significantly increased after stroke in LPS-preconditioned mice (approximately 2.5-fold), which may neutralize the effect of TNFalpha and reduce TNFalpha-mediated injury in ischemia. Importantly, LPS-preconditioned mice show marked resistance to brain injury caused by intracerebral administration of exogenous TNFalpha after stroke. We establish an in vitro model of LPS preconditioning in primary cortical neuronal cultures and show that LPS preconditioning causes significant protection against injurious TNFalpha in the setting of ischemia. Our studies suggest that TNFalpha is a twin-edged sword in the setting of stroke: TNFalpha upregulation is needed to establish LPS-induced tolerance before ischemia, whereas suppression of TNFalpha signaling during ischemia confers neuroprotection after LPS preconditioning.


Assuntos
Isquemia Encefálica/metabolismo , Precondicionamento Isquêmico , Lipopolissacarídeos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Animais , Anticorpos/imunologia , Isquemia Encefálica/induzido quimicamente , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células Cultivadas , Transtornos Cerebrovasculares/induzido quimicamente , Transtornos Cerebrovasculares/genética , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Knockout , Ratos , Transdução de Sinais , Solubilidade , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética
7.
J Cereb Blood Flow Metab ; 25(2): 217-25, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15678124

RESUMO

Osteopontin (OPN) is a secreted extracellular phosphoprotein involved in diverse biologic functions, including inflammation, cell migration, and antiapoptotic processes. Here we investigate the neuroprotective potential of OPN to reduce cell death using both in vitro and in vivo models of ischemia. We show that incubation of cortical neuron cultures with OPN protects against cell death from oxygen and glucose deprivation. The effect of OPN depends on the Arg-Gly-Asp (RGD)-containing motif as the protective effect of OPN in vitro was blocked by an RGD-containing hexapeptide, which prevents integrin receptors binding to their ligands. Osteopontin treatment of cortical neuron cultures caused an increase in Akt and p42/p44 MAPK phosphorylation, which is consistent with OPN-inducing neuroprotection via the activation of these protein kinases. Indeed, the protective effect of OPN was reduced by inhibiting the activation of Akt and p42/p44 MAPK using LY294002 and U0126, respectively. The protective effect of OPN was also blocked by the protein synthesis inhibitor cycloheximide, suggesting that the neuroprotective effect of OPN required new protein synthesis. Finally, intracerebral ventricular administration of OPN caused a marked reduction in infarct size after transient middle cerebral artery occlusion in a murine stroke model. These data suggest that OPN is a potent neuroprotectant against ischemic injury.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sialoglicoproteínas/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Western Blotting , Encéfalo/patologia , Isquemia Encefálica/patologia , Células Cultivadas , Cicloeximida/farmacologia , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/química , Oligopeptídeos , Osteopontina , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Sialoglicoproteínas/química , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia
8.
Lancet ; 362(9389): 1028-37, 2003 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-14522533

RESUMO

BACKGROUND: Molecular mechanisms of neuroprotection that lead to ischaemic tolerance are incompletely understood. Identification of genes involved in the process would provide insight into cell survival and therapeutic approaches for stroke. We developed a mouse model of neuroprotection in stroke and did gene expression profiling to identify potential neuroprotective genes and their associated pathways. METHODS: Eight mice per condition were subjected to occlusion of the middle cerebral artery for 15 min (preconditioning), 60 min (injurious ischaemia), or preconditioning followed 72 h later by injurious ischaemia. RNA was extracted from the cortical regions of the ischaemic and non-ischaemic hemispheres. Three pools per condition were generated, and RNA was hybridised to oligonucleotide microarrays for comparison of ischaemic and non-ischaemic hemispheres. Real-time PCR and western blots were used to validate results. Follow-up experiments were done to address the biological relevance of findings. FINDINGS: Microarray analysis revealed changes in gene expression with little overlap among the conditions of injurious ischaemia, ischaemic preconditioning, or both. Injurious ischaemia induced upregulation of gene expression; 49 (86%) of 57 genes regulated showed increased expression in the ischaemic hemisphere. By contrast, preconditioning followed by injurious ischaemia resulted in pronounced downregulation; 47 (77%) of 61 regulated genes showed lower expression. Preconditioning resulted in transcriptional changes involved in suppression of metabolic pathways and immune responses, reduction of ion-channel activity, and decreased blood coagulation. INTERPRETATION: Preconditioning reprogrammes the response to ischaemic injury. Similar changes reported by others support an evolutionarily conserved endogenous response to decreased blood flow and oxygen limitation such as seen during hibernation.


Assuntos
Isquemia Encefálica/genética , Isquemia Encefálica/prevenção & controle , Circulação Cerebrovascular/fisiologia , Expressão Gênica/fisiologia , Hibernação/fisiologia , Precondicionamento Isquêmico/métodos , Acidente Vascular Cerebral/fisiopatologia , Animais , Tempo de Sangramento , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Isquemia Encefálica/fisiopatologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Circulação Cerebrovascular/genética , Modelos Animais de Doenças , Hibernação/genética , Humanos , Técnicas In Vitro , Precondicionamento Isquêmico/normas , Camundongos , Fármacos Neuroprotetores/uso terapêutico , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia
9.
Stroke ; 35(11): 2576-81, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15375302

RESUMO

BACKGROUND AND PURPOSE: Tolerance to ischemic brain injury is induced by several preconditioning stimuli, including lipopolysaccharide (LPS). A small dose of LPS given systemically confers ischemic protection in the brain, a process that appears to involve activation of an inflammatory response before ischemia. We postulated that LPS preconditioning modulates the cellular inflammatory response after cerebral ischemia, resulting in neuroprotection. METHODS: Mice were treated with LPS (0.2 mg/kg) 48 hours before ischemia induced by transient middle cerebral artery occlusion (MCAO). The infarct was measured by 2,3,5-triphenyltetrazolium chloride staining. Microglia/macrophage responses after MCAO were assessed by immunofluorescence and flow cytometry. The effect of MCAO on white blood cells in the brain and peripheral circulation was measured by flow cytometry 48 hours after MCAO. RESULTS: LPS preconditioning induced significant neuroprotection against MCAO. Administration of low-dose LPS before MCAO prevented the cellular inflammatory response in the brain and blood. Specifically, LPS preconditioning suppressed neutrophil infiltration into the brain and microglia/macrophage activation in the ischemic hemisphere, which was paralleled by suppressed monocyte activation in the peripheral blood. CONCLUSIONS: LPS preconditioning induces neuroprotection against ischemic brain injury in a mouse model of stroke. LPS preconditioning suppresses the cellular inflammatory response to ischemia in the brain and circulation. Diminished activation of cellular inflammatory responses that ordinarily exacerbate ischemic injury may contribute to neuroprotection induced by LPS preconditioning.


Assuntos
Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Endotoxinas , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Precondicionamento Isquêmico , Animais , Encéfalo/imunologia , Encéfalo/patologia , Modelos Animais de Doenças , Leucócitos/fisiologia , Lipopolissacarídeos , Ativação de Macrófagos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Monócitos/fisiologia , Infiltração de Neutrófilos
10.
J Cereb Blood Flow Metab ; 23(10): 1151-9, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14526225

RESUMO

Corticotropin releasing hormone (CRH) and its family of related peptides are involved in regulating physiologic responses to multiple stressors, including stroke. Although CRH has been implicated in the exacerbation of injury after stroke, the mechanism remains unclear. After ischemia, both excitotoxic damage and inflammation contribute to the pathology of stroke. CRH is known to potentiate excitotoxic damage in the brain and has been shown to modulate inflammatory responses in the periphery. Here the present authors examine the relative contribution of the two known CRH receptors, CRH-R1 and CRH-R2, to ischemic injury using CRH receptor knockout mice. These results implicate CRH-R1 as the primary mediator of ischemic injury in this mouse model of stroke. In addition, the authors examine a potential role for CRH in inflammatory injury after stroke by identifying functional CRH receptors on astrocytes and microglia, which are cells that are known to be involved in brain inflammation. By single cell PCR, the authors show that microglia and astrocytes express mRNA for both CRH-R1 and CRH-R2. However, CRH-R1 is the primary mediator of cAMP accumulation in response to CRH peptides in these cells. The authors suggest that astrocytes and microglia are cellular targets of CRH, which could serve as a link between CRH and inflammatory responses in ischemic injury via CRH-R1.


Assuntos
Astrócitos/fisiologia , Isquemia Encefálica/fisiopatologia , Microglia/fisiologia , Receptores de Hormônio Liberador da Corticotropina/genética , Animais , Isquemia Encefálica/patologia , Infarto Cerebral/patologia , Infarto Cerebral/fisiopatologia , AMP Cíclico/metabolismo , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , RNA Mensageiro/análise , Receptores de Hormônio Liberador da Corticotropina/metabolismo
11.
Brain Res ; 932(1-2): 110-9, 2002 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11911867

RESUMO

Recent studies indicate that inflammation following cerebral ischemia contributes to neuronal damage. The local activation of resident cells and efficient recruitment of leukocytes into the central nervous system are critical steps in this inflammatory process. Here we describe studies using flow cytometry to examine the temporal pattern of inflammatory cell activation and infiltration following transient middle cerebral artery occlusion (MCAO) in mice. We found an increase in activated microglia/macrophages as early as 18 h post occlusion, which peaked at 48 h and remained abundant at 96 h post occlusion. Neutrophils were significantly increased by 48 h and remained elevated at 96 h post occlusion. T lymphocytes were increased relatively late (72 and 96 h) post occlusion. The flow cytometry data correlate well both quantitatively and qualitatively with immunohistochemistry analysis performed on the same mice. The present study demonstrates the power of flow cytometry in analyzing the inflammatory process following cerebral ischemia and offers temporal information on the cellular changes in mice following transient MCAO.


Assuntos
Isquemia Encefálica/imunologia , Citometria de Fluxo/métodos , Animais , Isquemia Encefálica/patologia , Encefalite/imunologia , Encefalite/patologia , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/imunologia , Neutrófilos/citologia , Neutrófilos/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
12.
J Cereb Blood Flow Metab ; 29(6): 1175-86, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19384334

RESUMO

Primate models are essential tools for translational research in stroke but are reportedly inconsistent in their ability to produce cortical infarcts of reproducible size. Here, we report a new stroke model using a transorbital, reversible, two-vessel occlusion approach in male rhesus macaques that produces consistent and reproducible cortical infarcts. The right middle cerebral artery (distal to the orbitofrontal branch) and both anterior cerebral arteries were occluded with vascular clips. Bilateral occlusion of the anterior cerebral artery was critical for reducing collateral flow to the ipsilateral cortex. Reversible ischemia was induced for 45, 60, or 90 mins (n=2/timepoint) and infarct volume and neurologic outcome were evaluated. The infarcts were located predominantly in the cortex and increased in size with extended duration of ischemia determined by T(2)-weighted magnetic resonance imaging . Infarct volume measured by 2,3,5-triphenyl tetrazolium chloride and cresyl violet staining corroborated magnetic resonance imaging results. Neurologic deficit scores worsened gradually with longer occlusion times. A subset of animals (n=5) underwent 60 mins of ischemia resulting in consistent infarct volumes primarily located to the cortex that correlated well with neurologic deficit scores. This approach offers promise for evaluating therapeutic interventions in stroke.


Assuntos
Isquemia Encefálica/patologia , Modelos Animais de Doenças , Macaca mulatta , Animais , Benzoxazinas , Isquemia Encefálica/sangue , Isquemia Encefálica/cirurgia , Macaca mulatta/cirurgia , Imageamento por Ressonância Magnética , Masculino , Oxazinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA