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










Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 13(1): 103, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27165310

RESUMO

BACKGROUND: Neuroinflammation in utero may contribute to brain injury resulting in life-long neurological disabilities. The pivotal role of the efferent cholinergic anti-inflammatory pathway (CAP) in controlling inflammation, e.g., by inhibiting the HMGB1 release, via the macrophages' α7 nicotinic acetylcholine receptor (α7nAChR) has been described in adults, but its importance in the fetus is unknown. Moreover, it is unknown whether CAP may also exert anti-inflammatory effects on the brain via the anatomically predominant afferent component of the vagus nerve. METHODS: We measured microglial activation in the ovine fetal brain near term 24 h after the umbilical cord occlusions mimicking human labor versus controls (no occlusions) by quantifying HMGB1 nucleus-to-cytosol translocation in the Iba1+ and α7nAChR+ microglia. Based on multiple clinical studies in adults and our own work in fetal autonomic nervous system, we gauged the degree of CAP activity in vivo using heart rate variability measure RMSSD that reflects fluctuations in vagus nerve activity. RESULTS: RMSSD correlated to corresponding plasma IL-1ß levels at R = 0.57 (p = 0.02, n = 17) and to white matter microglia cell counts at R = -0.89 (p = 0.03). The insult increased the HMGB1 translocation in α7nAChR+ microglia in a brain region-dependent manner (p < 0.001). In parallel, RMSSD at 1 h post insult correlated with cytosolic HMGB1 of thalamic microglia (R = -0.94, p = 0.005), and RMSSD at pH nadir correlated with microglial α7nAChR in the white matter (R = 0.83, p = 0.04). Overall, higher RMSSD values correlated with lower HMGB1 translocation and higher α7nAChR intensity per area in a brain region-specific manner. CONCLUSIONS: Afferent fetal CAP may translate increased vagal cholinergic signaling into suppression of cerebral inflammation in response to near-term hypoxic acidemia as might occur during labor. Our findings suggest a new control mechanism of fetal neuroinflammation via the vagus nerve, providing novel possibilities for its non-invasive monitoring in utero and for targeted treatment.


Assuntos
Encefalite/etiologia , Encefalite/terapia , Hipóxia Fetal/complicações , Nervo Vago/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Encéfalo/patologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Proteínas de Ligação ao Cálcio , Proteínas de Ligação a DNA/metabolismo , Diagnóstico por Computador , Modelos Animais de Doenças , Encefalite/sangue , Feminino , Hipóxia Fetal/sangue , Hipóxia Fetal/terapia , Feto , Regulação da Expressão Gênica/fisiologia , Proteína HMGB1/metabolismo , Frequência Cardíaca/fisiologia , Interleucina-1beta/sangue , Interleucina-6/sangue , Masculino , Proteínas dos Microfilamentos , Microglia/metabolismo , Microglia/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ovinos , Nervo Vago/embriologia , Estimulação do Nervo Vago
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...