Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 34(49): 16467-81, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25471584

RESUMO

Intrauterine infection (chorioamnionitis) aggravates neonatal hypoxic-ischemic (HI) brain injury, but the mechanisms linking systemic inflammation to the CNS damage remain uncertain. Here we report evidence for brain influx of T-helper 17 (TH17)-like lymphocytes to coordinate neuroinflammatory responses in lipopolysaccharide (LPS)-sensitized HI injury in neonates. We found that both infants with histological chorioamnionitis and rat pups challenged by LPS/HI have elevated expression of the interleukin-23 (IL-23) receptor, a marker of early TH17 lymphocytes, in the peripheral blood mononuclear cells. Post-LPS/HI administration of FTY720 (fingolimod), a sphingosine-1-phosphate receptor agonist that blocks lymphocyte trafficking, mitigated the influx of leukocytes through the choroid plexus and acute induction of nuclear factor-κB signaling in the brain. Subsequently, the FTY720 treatment led to attenuated blood-brain barrier damage, fewer cluster of differentiation 4-positive, IL-17A-positive T-cells in the brain, less proinflammatory cytokine, and better preservation of growth and white matter functions. The FTY720 treatment also provided dose-dependent reduction of brain atrophy, rescuing >90% of LPS/HI-induced brain tissue loss. Interestingly, FTY720 neither opposed pure-HI brain injury nor directly inhibited microglia in both in vivo and in vitro models, highlighting its unique mechanism against inflammation-sensitized HI injury. Together, these results suggest that the dual hit of systemic inflammation and neonatal HI injury triggers early onset of the TH17/IL-17-mediated immunity, which causes severe brain destruction but responds remarkably to the therapeutic blockade of lymphocyte trafficking.


Assuntos
Movimento Celular/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Inflamação/prevenção & controle , Ativação Linfocitária/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Animais , Animais Recém-Nascidos , Atrofia/tratamento farmacológico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Corioamnionite/tratamento farmacológico , Corioamnionite/metabolismo , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Cloridrato de Fingolimode , Humanos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Recém-Nascido , Lipopolissacarídeos , Linfócitos/citologia , NF-kappa B/metabolismo , Gravidez , Propilenoglicóis/uso terapêutico , Ratos , Receptores de Interleucina/metabolismo , Esfingosina/farmacologia , Esfingosina/uso terapêutico , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Substância Branca/efeitos dos fármacos
2.
Stroke ; 44(9): 2623-2627, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23881953

RESUMO

BACKGROUND AND PURPOSE: Plasminogen activator inhibitor-I (PAI-1), a ≈50-kDa serine protease inhibitor, markedly reduces the extravascular toxicity of tissue-type plasminogen activator in experimental hypoxic-ischemic (HI) brain injury of newborns. However, the current treatment with PAI-1 requires intracerebroventricle injection to cross the blood-brain barrier, which is an invasive procedure of limited clinical potential. Thus, we tested whether intranasal administration of PAI-1 can bypass blood-brain barrier and mitigate neonatal HI brain injury. METHODS: Rat pups were subjected to HI, with or without lipopolysaccharide pre-exposure, followed by intranasal delivery of a stable-mutant form of PAI-1 (CPAI). RESULTS: Immunoblotting showed that CPAI sequentially entered the olfactory bulbs and cerebral cortex after intranasal delivery and reduced ≈75% of brain atrophy in HI or lipopolysaccharide-sensitized HI injury. Mechanistically, CPAI attenuated HI-induced plasminogen activators and lipopolysaccharide/HI-induced nuclear factor-κB signaling, neuroinflammation, and blood-brain barrier permeability. CONCLUSIONS: Intranasal delivery of CPAI is an effective treatment of experimental HI brain injury of newborns. Clinical application of this experimental therapy merits further investigation.


Assuntos
Hipóxia-Isquemia Encefálica/tratamento farmacológico , Inibidor 1 de Ativador de Plasminogênio/administração & dosagem , Inativadores de Plasminogênio/administração & dosagem , Inativadores de Plasminogênio/uso terapêutico , Administração Intranasal , Animais , Animais Recém-Nascidos , Atrofia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Masculino , Inibidor 1 de Ativador de Plasminogênio/uso terapêutico , Ratos , Ratos Wistar , Inibidores de Serina Proteinase/administração & dosagem , Inibidores de Serina Proteinase/uso terapêutico
3.
Sci Transl Med ; 5(193): 193ra90, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843451

RESUMO

Intracranial hemorrhage in preterm neonates may result in neonatal mortality and functional disabilities, but its pathogenic mechanisms are poorly defined and better therapies are needed. We used a tetracycline-regulated transgenic system to test whether the induction of vascular endothelial growth factor (VEGF) in the germinal matrix leads to intracranial hemorrhage. This genetic strategy initially induced a dense network of loosely adjoined endothelial cells and pericytes near lateral ventricles, similar to the immature vascular rete in human fetal brains. Yet, this rich vascular network transformed into low-vasculature patches correlated with hemorrhage and caspase-3 activation near birth. Gene expression and biochemical analyses suggested that downstream mediators of VEGF in this network include transcriptional factors ETS1 and HIF2α (hypoxia-inducible factor 2α), components of the PDGFß (platelet-derived growth factor ß) and TGFß (transforming growth factor-ß) receptor signaling pathways, matrix metalloproteinase-9 (MMP-9), and cathepsins. Prenatal administration of glucocorticoids markedly reduced mortality and cerebral hemorrhage in mutant animals, as in human neonates. This protective effect was not due to blocking vasculogenesis, but was instead associated with inhibition of neurovascular proteases, notably MMP-9, cathepsin B, and caspase-3. Collectively, these results support a causative role of VEGF in perinatal cerebral hemorrhage and implicate its downstream proteases as potential therapeutic targets.


Assuntos
Hemorragia Cerebral/enzimologia , Hemorragia Cerebral/patologia , Peptídeo Hidrolases/biossíntese , Prosencéfalo/enzimologia , Prosencéfalo/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Betametasona/farmacologia , Betametasona/uso terapêutico , Caspase 3/metabolismo , Catepsina B/metabolismo , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/genética , Modelos Animais de Doenças , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/patologia , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Perfilação da Expressão Gênica , Glucocorticoides/farmacologia , Glucocorticoides/uso terapêutico , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Fenótipo , Prosencéfalo/irrigação sanguínea , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Tetraciclina/farmacologia
4.
Exp Neurol ; 247: 447-455, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23353638

RESUMO

Perinatal infection aggravates neonatal hypoxic-ischemic (HI) brain injury and may interfere with therapeutic hypothermia. While the NF-κB signaling pathway has been implicated in microglia activation in infection-sensitized HI, the current therapeutic strategies rely on systemic intervention, which could impair neonatal immunity and increase the risk of severe infection. To devise a brain-targeted anti-NF-κB strategy, we examined the effects of intranasal delivery of tat-NBD peptides in two animal models of neonatal infection-sensitized HI. Kinetic experiments showed that tat-NBD peptides entered the olfactory bulbs rapidly (10-30 min) and peaked in the cerebral cortex around 60 min after intranasal application in P7 rats. Further, intranasal delivery of 1.4 mg/kg tat-NBD, which is only 7% of the intravenous dose in past studies, markedly attenuated NF-κB signaling, microglia activation, and brain damage triggered by HI with 4 or 72 h pre-exposure to the bacterial endotoxin lipopolysaccharide (LPS). In contrast, intranasal delivery of mutant tat-NBD peptides or systemic application of minocycline failed to block LPS-sensitized HI injury. Yet, intranasal delivery of up to 5.6 mg/kg tat-NBD peptides immediately after pure-HI insult showed little protection, likely due to its rapid clearance from the brain and inability to inhibit parenchymal plasminogen activators. Together, these results suggest a novel therapy of infection-sensitized HI brain injury in newborns.


Assuntos
Hipóxia-Isquemia Encefálica/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intracelular/administração & dosagem , NF-kappa B/antagonistas & inibidores , Peptídeos/administração & dosagem , Administração Intranasal , Análise de Variância , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Hipóxia-Isquemia Encefálica/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Espectroscopia de Ressonância Magnética , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas dos Microfilamentos/metabolismo , Mutação/fisiologia , Ratos , Sais de Tetrazólio
5.
Cereb Cortex ; 23(5): 1218-29, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22556277

RESUMO

Intrauterine infection exacerbates neonatal hypoxic-ischemic (HI) brain injury and impairs the development of cerebral cortex. Here we used low-dose lipopolysaccharide (LPS) pre-exposure followed by unilateral cerebral HI insult in 7-day-old rats to study the pathogenic mechanisms. We found that LPS pre-exposure blocked the HI-induced proteolytic activity of tissue-type plasminogen activator (tPA), but significantly enhanced NF-κB signaling, microglia activation, and the production of pro-inflammatory cytokines in newborn brains. Remarkably, these pathogenic responses were all blocked by intracerebroventricular injection of a stable-mutant form of plasminogen activator protein-1 called CPAI. Similarly, LPS pre-exposure amplified, while CPAI therapy mitigated HI-induced blood-brain-barrier damage and the brain tissue loss with a therapeutic window at 4 h after the LPS/HI insult. The CPAI also blocks microglia activation following a brain injection of LPS, which requires the contribution by tPA, but not the urinary-type plasminogen activator (uPA), as shown by experiments in tPA-null and uPA-null mice. These results implicate the nonproteolytic tPA activity in LPS/HI-induced brain damage and microglia activation. Finally, the CPAI treatment protects near-normal motor and white matter development despite neonatal LPS/HI insult. Together, because CPAI blocks both proteolytic and nonproteolytic tPA neurotoxicity, it is a promising therapeutics of neonatal HI injury either with or without infection.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/prevenção & controle , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/prevenção & controle , Lipopolissacarídeos , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Animais Recém-Nascidos , Encefalite/induzido quimicamente , Encefalite/metabolismo , Encefalite/prevenção & controle , Hipóxia-Isquemia Encefálica/induzido quimicamente , Camundongos , Ratos
6.
Proc Natl Acad Sci U S A ; 108(18): 7607-12, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21502507

RESUMO

The organization of neural progenitors in the developing mammalian neuroepithelium is marked by cadherin-based adherens junctions. Whereas RhoA, a founding member of the small Rho GTPase family, has been shown to play important roles in epithelial adherens junctions, its physiological roles in neural development remain uncertain due to the lack of specific loss-of-function studies. Here, we show that RhoA protein accumulates at adherens junctions in the developing mouse brain and colocalizes to the cadherin-catenin complex. Conditional deletion of RhoA in midbrain and forebrain neural progenitors using Wnt1-Cre and Foxg1-Cre mice, respectively, disrupts apical adherens junctions and causes massive dysplasia of the brain. Furthermore, RhoA-deficient neural progenitor cells exhibit accelerated proliferation, reduction of cell- cycle exit, and increased expression of downstream target genes of the hedgehog pathway. Consequently, both lines of conditional RhoA-deficient embryos exhibit expansion of neural progenitor cells and exencephaly-like protrusions. These results demonstrate a critical role of RhoA in the maintenance of apical adherens junctions and the regulation of neural progenitor proliferation in the developing mammalian brain.


Assuntos
Junções Aderentes/metabolismo , Encéfalo/embriologia , Proliferação de Células , Células-Tronco Neurais/metabolismo , Proteína rhoA de Ligação ao GTP/deficiência , Animais , Bromodesoxiuridina , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Indóis , Camundongos , Camundongos Mutantes , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA