Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Chem Biol ; 17(5): 1073-1081, 2022 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-35471821

RESUMO

We sought to develop a small-molecule activator of interferon regulatory factor 3 (IRF3), an essential innate immune transcription factor, which could potentially be used therapeutically in multiple disease settings. Using a high-throughput screen, we identified small-molecule entities that activate a type I interferon response, with minimal off-target NFκB activation. We identified 399 compounds at a hit rate of 0.24% from singlicate primary screening. Secondary screening included the primary hits and additional compounds with similar chemical structures obtained from other library sources and resulted in 142 candidate compounds. The hit compounds were sorted and ranked to identify compound groups with activity in both human and mouse backgrounds to facilitate animal model engagement for translational development. Chemical modifications within two groups of small molecules produced leads with improved activity over original hits. Furthermore, these leads demonstrated activity in ex vivo cytokine release assays from human blood- and mouse bone marrow-derived macrophages. Dependence on IRF3 was demonstrated using bone marrow-derived macrophages from IRF3-deficient mice, which were not responsive to the molecules. To identify the upstream pathway leading to IRF3 activation, we used a library of CRISPR knockout cell lines to test the key innate immune adaptor and receptor molecules. These studies indicated a surprising toll-interleukin-1 receptor-domain-containing-adapter-inducing interferon-ß-dependent but TLR3/4-independent mechanism of IRF3 activation.


Assuntos
Fator Regulador 3 de Interferon , Transdução de Sinais , Animais , Antivirais/farmacologia , Desenvolvimento de Medicamentos , Fator Regulador 3 de Interferon/metabolismo , Macrófagos/metabolismo , Camundongos
2.
Stroke ; 47(1): 262-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26564103

RESUMO

BACKGROUND AND PURPOSE: Preconditioning with poly-l-lysine and carboxymethylcellulose (ICLC) provides robust neuroprotection from cerebral ischemia in a mouse stroke model. However, the receptor that mediates neuroprotection is unknown. As a synthetic double-stranded RNA, poly-ICLC may bind endosomal Toll-like receptor 3 or one of the cytosolic retinoic acid-inducible gene-I-like receptor family members, retinoic acid-inducible gene-I, or melanoma differentiation-associated protein 5. Activation of these receptors culminates in type I interferons (IFN-α/ß) induction-a response required for poly-ICLC-induced neuroprotection. In this study, we investigate the receptor required for poly-ICLC-induced neuroprotection. METHODS: Toll-like receptor 3, melanoma differentiation-associated protein 5-, and IFN-promoter stimulator 1-deficient mice were treated with poly-ICLC 24 hours before middle cerebral artery occlusion. Infarct volume was measured 24 hours after stroke to identify the receptor signaling pathways involved in protection. IFN-α/ß induction was measured in plasma samples collected 6 hours after poly-ICLC treatment. IFN-ß-deficient mice were used to test the requirement of IFN-ß for poly-ICLC-induced neuroprotection. Mice were treated with recombinant IFN-α-A to test the role of IFN-α as a potential mediator of neuroprotection. RESULTS: Poly-ICLC induction of both neuroprotection and systemic IFN-α/ß requires the cytosolic receptor melanoma differentiation-associated protein 5 and the adapter molecule IFN-promoter stimulator 1, whereas it is independent of Toll-like receptor 3. IFN-ß is not required for poly-ICLC-induced neuroprotection. IFN-α treatment protects against stroke. CONCLUSIONS: Poly-ICLC preconditioning is mediated by melanoma differentiation-associated protein 5 and its adaptor molecule IFN-promoter stimulator 1. This is the first evidence that a cytosolic receptor can mediate neuroprotection, providing a new target for the development of therapeutic agents to protect the brain from ischemic injury.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , RNA Helicases DEAD-box/metabolismo , Precondicionamento Isquêmico/métodos , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/prevenção & controle , Animais , Carboximetilcelulose Sódica/análogos & derivados , Carboximetilcelulose Sódica/metabolismo , Carboximetilcelulose Sódica/uso terapêutico , Helicase IFIH1 Induzida por Interferon , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Poli I-C/metabolismo , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Polilisina/metabolismo , Polilisina/uso terapêutico
3.
Acta Neurochir Suppl ; 121: 39-44, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463920

RESUMO

Stroke activates an inflammatory response that results in the infiltration of peripheral immune cells into the ischemic area, contributing to exacerbation of tissue damage. However, evidence indicates that inflammatory cell infiltration can also promote neuroprotection through regulatory immune cells that mitigate injury. These immune regulatory cells may also be important mediators of neuroprotection associated with preconditioning, a phenomenon whereby small exposure to a potential harmful stimulus is able to induce protection against a subsequent ischemic event. The elucidation of mechanisms that allow these immune cells to confer neuroprotection is critical to developing new therapeutic strategies against acute stroke. In the present review, we discuss the dual role of peripheral immune cells in stroke-related brain injury and neuroprotection. Furthermore, we report new data from our laboratory that supports the important role of peripheral cells and their interaction with the brain endothelium for the establishment of the protective phenotype in preconditioning.


Assuntos
Precondicionamento Isquêmico , Linfócitos/imunologia , Macrófagos/imunologia , Neuroproteção/imunologia , Neutrófilos/imunologia , Acidente Vascular Cerebral/imunologia , Animais , Humanos
4.
J Cereb Blood Flow Metab ; 32(12): 2193-200, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23010947

RESUMO

Systemic preconditioning with the TLR9 ligand CpG induces neuroprotection against brain ischemic injury through a tumor necrosis factor (TNF)-dependent mechanism. It is unclear how systemic administration of CpG engages the brain to induce the protective phenotype. To address this, we created TLR9-deficient reciprocal bone marrow chimeric mice lacking TLR9 on either hematopoietic cells or radiation-resistant cells of nonhematopoietic origin. We report that wild-type mice reconstituted with TLR9-deficient hematopoietic cells failed to show neuroprotection after systemic CpG preconditioning. Further, while hematopoietic expression of TLR9 is required for CpG-induced neuroprotection it is not sufficient to restore protection to TLR9-deficient mice that are reconstituted with hematopoietic cells bearing TLR9. To determine whether the absence of protection was associated with TNF, we examined TNF levels in the systemic circulation and the brain. We found that although TNF is required for CpG preconditioning, systemic TNF levels did not correlate with the protective phenotype. However, induction of cerebral TNF mRNA required expression of TLR9 on both hematopoietic and nonhematopoietic cells and correlated with neuroprotection. In accordance with these results, we show the therapeutic potential of intranasal CpG preconditioning, which induces brain TNF mRNA and robust neuroprotection with no concomitant increase in systemic levels of TNF.


Assuntos
Adjuvantes Imunológicos/farmacologia , Transplante de Medula Óssea , Isquemia Encefálica/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Receptor Toll-Like 9/biossíntese , Quimeras de Transplante/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Knockout , Receptor Toll-Like 9/genética , Quimeras de Transplante/genética , Transplante Homólogo , Fator de Necrose Tumoral alfa/genética
5.
PLoS One ; 7(6): e36465, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22745654

RESUMO

The innate immune system plays important roles in a number of disparate processes. Foremost, innate immunity is a first responder to invasion by pathogens and triggers early defensive responses and recruits the adaptive immune system. The innate immune system also responds to endogenous damage signals that arise from tissue injury. Recently it has been found that innate immunity plays an important role in neuroprotection against ischemic stroke through the activation of the primary innate immune receptors, Toll-like receptors (TLRs). Using several large-scale transcriptomic data sets from mouse and mouse macrophage studies we identified targets predicted to be important in controlling innate immune processes initiated by TLR activation. Targets were identified as genes with high betweenness centrality, so-called bottlenecks, in networks inferred from statistical associations between gene expression patterns. A small set of putative bottlenecks were identified in each of the data sets investigated including interferon-stimulated genes (Ifit1, Ifi47, Tgtp and Oasl2) as well as genes uncharacterized in immune responses (Axud1 and Ppp1r15a). We further validated one of these targets, Ifit1, in mouse macrophages by showing that silencing it suppresses induction of predicted downstream genes by lipopolysaccharide (LPS)-mediated TLR4 activation through an unknown direct or indirect mechanism. Our study demonstrates the utility of network analysis for identification of interesting targets related to innate immune function, and highlights that Ifit1 can exert a positive regulatory effect on downstream genes.


Assuntos
Proteínas de Transporte/metabolismo , Imunidade Inata/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Linhagem Celular , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
6.
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
7.
Stroke ; 40(3 Suppl): S34-7, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19064776

RESUMO

BACKGROUND AND PURPOSE: Systemic administration of cytosine-guanine (CpG) oligodeoxynucleotides provides neuroprotection against subsequent cerebral ischemic injury. We examined the genomic response of leukocytes and brain cells after ischemia in the context of CpG preconditioning. METHODS: RNA was isolated from circulating leukocytes and ischemic cortex 3 and 24 hours after middle cerebral artery occlusion after CpG or saline pretreatment and subjected to microarray analysis. Genes uniquely upregulated in CpG-pretreated mice were examined for overrepresented transcriptional regulatory elements. RESULTS: CpG preconditioning induced a novel response to middle cerebral artery occlusion within circulating leukocytes that was dominated by natural killer cell-associated genes and the GATA-3 transcriptional regulatory element. Preconditioning also caused a novel brain response to stroke that was dominated by Type I interferon, interferon-associated genes, and transcriptional regulatory elements. CONCLUSIONS: CpG preconditioning invokes novel leukocyte and brain responses to stroke. In this, CpG may be a unique preconditioning agent, coordinating peripheral and brain responses to protect against ischemic injury.


Assuntos
Isquemia Encefálica/prevenção & controle , Isquemia Encefálica/fisiopatologia , Encefalite/fisiopatologia , Precondicionamento Isquêmico , Receptores Toll-Like/fisiologia , Doença Aguda , Animais , Arteriopatias Oclusivas/patologia , Arteriopatias Oclusivas/fisiopatologia , Arteriopatias Oclusivas/prevenção & controle , Isquemia Encefálica/patologia , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Artérias Cerebrais/fisiopatologia , Fosfatos de Dinucleosídeos/farmacologia , Modelos Animais de Doenças , Encefalite/metabolismo , Encefalite/patologia , Fator de Transcrição GATA3/metabolismo , Interferon Tipo I/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/prevenção & controle
8.
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
9.
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
10.
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
11.
Genes Dev ; 20(19): 2673-86, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16983145

RESUMO

Carcinoma cells initiate the metastatic cascade by inserting invasive pseudopodia through breaches in the basement membrane (BM), a specialized barrier of cross-linked, extracellular matrix macromolecules that underlies epithelial cells and ensheaths blood vessels. While BM invasion is the sine qua non of the malignant phenotype, the molecular programs that underlie this process remain undefined. To identify genes that direct BM remodeling and transmigration, we coupled high-resolution electron microscopy with an ex vivo model of invasion that phenocopies the major steps observed during the transition of carcinoma in situ to frank malignancy. Herein, a triad of membrane-anchored proteases, termed membrane type-1, type-2, and type-3 metalloproteinases, are identified as the triggering agents that independently confer cancer cells with the ability to proteolytically efface the BM scaffolding, initiate the assembly of invasive pseudopodia, and propagate transmigration. These studies characterize the first series of gene products capable of orchestrating the entire BM remodeling program that distinguishes the carcinomatous phenotype.


Assuntos
Membrana Basal/fisiologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Animais , Membrana Basal/enzimologia , Membrana Basal/ultraestrutura , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Chlorocebus aethiops , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 15 da Matriz/genética , Metaloproteinase 15 da Matriz/metabolismo , Metaloproteinase 16 da Matriz/genética , Metaloproteinase 16 da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana/genética , Camundongos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Mutação/genética , Invasividade Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/fisiopatologia , RNA Interferente Pequeno/genética , Ratos , Inibidores Teciduais de Metaloproteinases/metabolismo
12.
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
13.
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
14.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA