Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 119: 110208, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37150016

RESUMO

Increasing evidence indicates that an altered immune system is closely linked to the pathophysiology of anxiety disorders, and inhibition of neuroinflammation may represent an effective therapeutic strategy to treat anxiety disorders. Harmine, a beta-carboline alkaloid in various medicinal plants, has been widely reported to display anti-inflammatory and potentially anxiolytic effects. However, the exact underlying mechanisms are not fully understood. Our recent study has demonstrated that dysregulation of neuroplasticity in the basolateral amygdala (BLA) contributes to the pathological processes of inflammation-related anxiety. In this study, using a mouse model of anxiety challenged with Escherichia coli lipopolysaccharide (LPS), we found that harmine alleviated LPS-induced anxiety-like behaviors in mice. Mechanistically, harmine significantly prevented LPS-induced neuroinflammation by suppressing the expression of pro-inflammatory cytokines including IL-1ß and TNF-α. Meanwhile, ex vivo whole-cell slice electrophysiology combined with optogenetics showed that LPS-induced increase of medial prefrontal cortex (mPFC)-driven excitatory but not inhibitory synaptic transmission onto BLA projection neurons, thereby alleviating LPS-induced shift of excitatory/inhibitory balance towards excitation. In addition, harmine attenuated the increased intrinsic neuronal excitability of BLA PNs by reducing the medium after-hyperpolarization. In conclusion, our findings provide new evidence that harmine may exert its anxiolytic effect by downregulating LPS-induced neuroinflammation and restoring the changes in neuronal plasticity in BLA PNs.


Assuntos
Ansiolíticos , Complexo Nuclear Basolateral da Amígdala , Humanos , Complexo Nuclear Basolateral da Amígdala/metabolismo , Ansiolíticos/farmacologia , Ansiolíticos/uso terapêutico , Tonsila do Cerebelo/fisiologia , Harmina/farmacologia , Harmina/uso terapêutico , Doenças Neuroinflamatórias , Lipopolissacarídeos/farmacologia , Plasticidade Neuronal
2.
ASN Neuro ; 13: 17590914211003247, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33853366

RESUMO

LncRNA TUG1 has not yet been reported in cerebral ischemia/reperfusion (I/R) injury. Methylcytosine dioxygenase TET2 is involved in ischemic damage. This study aimed to investigate the effects of TUG1 demethylated by TET2 on I/R-induced inflammatory response and identified its possible mechanisms.We found that TUG1 expression was significantly upregulated in oxygen-glucose deprivation and reoxygenation (OGD/R)-induced SH-SY5Y and SK-N-SH cells. Using the middle cerebral artery occlusion (MCAO) mice, we observed a similar effect. We also found that I/R injury could downregulate miR-200a-3p and upregulate NLRP3 and TET2. The knockdown of TUG1 could alleviate OGD/R-induced inflammatory response through upregulating miR-200a-3p and downregulating NLRP3 and other pro-inflammatory molecules. miR-200a-3p inhibition can partially reverse the effects of TUG1 silencing. Further experiments confirmed that TUG1 sponged miR-200a-3p to diminish miR-200a-3p and promote NLRP3 dependent inflammatory responses. Mechanically, knockdown of TET2 induced low levels of TUG1 and high levels of miR-200a-3p in both SK-N-SH and SH-SY5Y cells. IL-18, IL-1ß, NLRP3, Caspase-1, and GSDMD-N were highly downregulated in OGD/R-induced SK-N-SH and SH-SY5Y cells after TET2 knockdown. TUG1 overexpression could reverse this effect. All the data indicated that TET2 could demethylate TUG1 and contribute to the inflammatory response. In additional experiments using the MCAO mice model, we confirmed knockdown of TET2 attenuated I/R-induced inflammatory response and brain injuries via decreasing TUG1 and increasing miR-200a-3p to inhibit NLRP3 expression. The demethylation of TUG1 by TET2 might aggravate I/R-induced inflammatory injury via modulating NLRP3 by miR-200a-3p. Our data confirmed that TET2 contributed to I/R-induced inflammatory response via the demethylation of TUG1 and regulated TUG1/miR-200a-3p/NLRP3 pathway.


Assuntos
Isquemia Encefálica , Dioxigenases , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Animais , Apoptose , Isquemia Encefálica/genética , Proteínas de Ligação a DNA/genética , Camundongos , MicroRNAs/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , RNA Longo não Codificante/genética , Reperfusão , Traumatismo por Reperfusão/genética
3.
Brain Behav Immun ; 91: 505-518, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33161163

RESUMO

Increasing evidence indicates that excessive inflammatory responses play a crucial role in the pathophysiology of psychiatric diseases, including depression and anxiety disorders. The dysfunctional neural plasticity in amygdala has long been proposed as the vital cause for the progression of psychiatric disorders. However, the effect of neuroinflammation on the functional changes of the amygdala remains largely unknown. Here, by using a mouse model of inflammation induced by lipopolysaccharide (LPS) injection, we investigated the effect of LPS-induced neuroinflammation on the synaptic and non-synaptic plasticity in basolateral amygdala (BLA) projection neurons (PNs) and their contribution to the LPS-induced anxiety- and depressive-like behavior. The results showed that LPS treatment led to the activation of microglia and production of proinflammatory cytokines in the BLA. Furthermore, LPS treatment increased excitatory but not inhibitory synaptic transmission due to the enhanced presynaptic glutamate release, thus leading to the shift of excitatory/inhibitory balance towards excitatory. In addition, the intrinsic neuronal excitability of BLA PNs was also increased by LPS treatment through the loss of expression and function of small-conductance, calcium-activated potassium channel. Chronic fluoxetine pretreatment significantly prevented these neurophysiological changes induced by LPS, and alleviated anxiety and depressive-like behavior, indicating that LPS-induced neuronal dysregulation of BLA PNs may contribute to the development of psychiatry disorders. Collectively, these findings provide evidence that dysregulation of synaptic and non-synaptic transmission in the BLA PNs may account for neuroinflammation-induced anxiety- and depressive-like behavior.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Tonsila do Cerebelo , Ansiedade , Transtornos de Ansiedade , Humanos , Plasticidade Neuronal
4.
Brain Res ; 1736: 146730, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32081533

RESUMO

OBJECTIVES: To investigate the neuroprotective effect of Gingko biloba extract 761 (EGb761) in Alzheimer's disease (AD) models both in vivo and in vitro and the underlying molecular mechanism. METHODS: Cultured BV2 microglial cells were treated with Aß1-42 to establish an in vitro AD model. The in vivo rat AD model was established by injecting Aß1-42. Cells were pre-treated with EGb761, and the proliferation and necroptosis were examined by MTT or flow cytometry assays, respectively. In addition, the membrane potential and oxidative stress were measured. Cognitive function was evaluated by the Morris water maze, and the activation of the JNK signaling pathway was quantified by Western blotting. RESULTS: Cultured BV2 cells exhibited prominent cell death after Aß1-42 induction, and this cell death was alleviated by EGb761 pre-treatment. EGb761 was found to relieve oxidative stress and suppress the membrane potential and calcium overload. EGb761 treatment in AD model rats also improved cognitive function deficits. Both cultured microglial cells and the rat hippocampus exhibited activation of the JNK signaling pathway, and EGb761 relieved this activation in cells. CONCLUSION: Our results showed that EGb761 regulated cell proliferation, suppressed necroptosis and apoptosis, relieved mitochondrial damage, and ameliorated tissue damage to improve cognitive function in AD models. All of these effects may involve the suppression of the JNK signaling pathway.


Assuntos
Doença de Alzheimer/metabolismo , Necroptose/efeitos dos fármacos , Extratos Vegetais/farmacologia , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Transtornos Cognitivos/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Ginkgo biloba , Hipocampo/metabolismo , Humanos , Masculino , Microglia , Mitocôndrias/metabolismo , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Extratos Vegetais/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores
6.
J Stroke Cerebrovasc Dis ; 27(11): 3247-3255, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30093197

RESUMO

BACKGROUND: Cerebral edema, a serious complication of acute cerebral infarction, has a crucial impact on morbidity and mortality in the early stage of cerebral infarction. And aquaporin 4 (AQP4), a bidirectional water transporting protein, plays a pivotal role in edema formation. At experimental model, it has proven that atorvastatin could exert pleiotropic neuroprotection on acute cerebral infarction independent of its cholesterol-lowering action. It was a common protective manifestation that atorvastatin can reduce the infarct volume and cerebral edema. However, little is known about atorvastatin improving ischemic brain edema by regulating AQP4 expression. This study intended to investigate the neuroprotection effects of atorvastatin pretreatment in rats with cerebral ischemia and further explore the potential relationship between atorvastatin and AQP4 expression. METHODS: Fifty-one adult male Sprague Dawley rats were randomly divided into 3 groups: sham, middle cerebral artery occlusion (MCAO), and atorvastatin pretreatment (Ator) group. For Ator group, 20 mg/kg of atorvastatin injectable suspension was administered once for 7days by gavage before operation, whereas the others were administered the same volume of saline matching. Except for sham group, MCAO and Ator groups were subjected to permanent MCAO by modified intraluminal suture method. Infarct volume, neurological deficit, brain water content (BWC), immunohistochemistry, western blot, and polymerase chain reaction (PCR) were measured at 24 hours after MCAO. RESULTS: Compared with sham group, the mNSS, infarct volume, and BWC of ischemic hemisphere were significantly increased (P < 0.001) in MCAO group. Positive cells and protein levels of p-p38MAPK and AQP4 in peri-infarction were significantly increased (P < 0.01). The mRNA levels of p38MAPK and AQP4 were also prominently upregulated (P < 0.01). Interestingly, preadministration of atorvastatin dramatically decreased infarct volume and the BWC of ischemic hemisphere compared with MCAO group (P < 0.05). The overexpressions of p-p38MAPK and AQP4 in peri-infarction were significantly decreased (P < 0.05) and their mRNA levels were downregulated by atorvastatin pretreatment (P < 0.05). Neurological deficits were also dramatically improved (P < 0.001). CONCLUSION: To the best of our knowledge, this is the first study that demonstrates an effect of atorvastatin on expression of AQP4, and we propose that decreased AQP4 expression through a p38MAPK-suppression pathway may be the mechanism of atorvastatin alleviating ischemic cerebral edema.


Assuntos
Aquaporina 4/metabolismo , Atorvastatina/farmacologia , Edema Encefálico/prevenção & controle , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Aquaporina 4/genética , Comportamento Animal/efeitos dos fármacos , Água Corporal/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Edema Encefálico/psicologia , Modelos Animais de Doenças , Regulação para Baixo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/psicologia , Masculino , Atividade Motora/efeitos dos fármacos , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Neurol Sci ; 39(7): 1293-1296, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29480338

RESUMO

Intracranial dural arteriovenous fistulas (DAVFs), constituting approximately 10 to15% of intracranial vascular malformations, are anomalous direct connections between dural arteries and venous sinuses, meningeal veins, or cortical veins; the arterial feeders are various, usually fed by branches of internal carotid, external carotid, or vertebral artery (Santillan et al. CNN 115(3):241-251, 2013; Holoekamp et al. JN 124(6):1752-65, 2016; Terada T et al. JN 80(5):884-9, 1994). Spectrums of clinical presentations are widespread, arranging from pulsatile tinnitus to intracranial hemorrhage. Such DAVFs with rapidly progressive dementia as primary presentation, which has been reported in several literature, are still extremely scarce (Santillan et al. CNN 115(3):241-251, 2013; Holoekamp et al JN 124(6):1752-65, 2016). Up to 2015, similar reports are less than 20 cases (Holoekamp et al. JN 124(6):1752-65, 2016). Herein, we report a patient who was misdiagnosed with encephalitis, presented thalamic dementia, and was ultimately diagnosed of DAVFs.


Assuntos
Malformações Vasculares do Sistema Nervoso Central/complicações , Malformações Vasculares do Sistema Nervoso Central/diagnóstico , Disfunção Cognitiva/diagnóstico , Disfunção Cognitiva/etiologia , Encéfalo/diagnóstico por imagem , Malformações Vasculares do Sistema Nervoso Central/terapia , Disfunção Cognitiva/terapia , Demência/diagnóstico , Demência/etiologia , Demência/terapia , Diagnóstico Diferencial , Erros de Diagnóstico , Progressão da Doença , Encefalite/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade
10.
Cogn Behav Neurol ; 24(3): 128-33, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21945984

RESUMO

BACKGROUND: : In previous studies we and others have demonstrated an association with apolipoprotein (APOE) ε4 genotype and the presence of cognitive deficits in multiple sclerosis (MS). In this follow-up study, we have assessed whether APOE ε4 status exacerbates progression of cognitive deficits in MS. METHODS: : A total of 197 patients with MS were assessed for APOE genotype, and baseline cognitive performance was measured using a standardized battery of tests. One hundred seventy patients (86.3%) were clinically followed up for 1 year and were assessed for progression of cognitive deficits. RESULTS: : The APOE ε4 allele was present in 24.7% of patients. During 1-year follow-up, significant progression of cognitive deficits was found in APOE ε4 carriers (P=0.001) after logistic regression analysis controlling for sex, ethnicity, age, education, disease duration, severity, and subtype. CONCLUSIONS: : APOE ε4 carriers with MS have worsening progression of cognitive deficits than noncarriers. APOE ε4 carrier status predicts cognitive decline in verbal learning and memory.


Assuntos
Apolipoproteína E4/genética , Transtornos Cognitivos/genética , Progressão da Doença , Predisposição Genética para Doença/genética , Esclerose Múltipla/genética , Adulto , Idoso , Transtornos Cognitivos/complicações , Transtornos Cognitivos/psicologia , Feminino , Seguimentos , Genótipo , Heterozigoto , Humanos , Masculino , Memória , Pessoa de Meia-Idade , Esclerose Múltipla/complicações , Esclerose Múltipla/psicologia , Testes Neuropsicológicos , Polimorfismo Genético , Aprendizagem Verbal
11.
PLoS One ; 5(8): e12400, 2010 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-20811645

RESUMO

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) models are important vehicles for studying the effect of infectious elements such as Pertussis toxin (PTx) on disease processes related to acute demyelinating encephalomyelitis (ADEM) or multiple sclerosis (MS). PTx has pleotropic effects on the immune system. This study was designed to investigate the effects of PTx administered intracerebroventricularly (icv) in preventing downstream immune cell infiltration and demyelination of the spinal cord. METHODS AND FINDINGS: EAE was induced in C57BL/6 mice with MOG(35-55). PTx icv at seven days post MOG immunization resulted in mitigation of clinical motor symptoms, minimal T cell infiltration, and the marked absence of axonal loss and demyelination of the spinal cord. Integrity of the blood brain barrier was compromised in the brain whereas spinal cord BBB integrity remained intact. PTx icv markedly increased microglia numbers in the brain preventing their migration to the spinal cord. An in vitro transwell study demonstrated that PTx inhibited migration of microglia. CONCLUSION: Centrally administered PTx abrogated migration of microglia in EAE mice, limiting the inflammatory cytokine milieu to the brain and prevented dissemination of demyelination. The effects of PTx icv warrants further investigation and provides an attractive template for further study regarding the pleotropic effects of infectious elements such as PTx in the pathogenesis of autoimmune disorders.


Assuntos
Movimento Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/patologia , Microglia/efeitos dos fármacos , Microglia/patologia , Toxina Pertussis/administração & dosagem , Toxina Pertussis/farmacologia , Medula Espinal/patologia , Sequência de Aminoácidos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Doenças Desmielinizantes/prevenção & controle , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Leucócitos/imunologia , Camundongos , Dados de Sequência Molecular , Atividade Motora/efeitos dos fármacos , Bainha de Mielina/metabolismo , Permeabilidade/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
12.
Biochem Biophys Res Commun ; 384(4): 466-70, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19422789

RESUMO

Recent clinical studies have identified an association between APOE 4 and cognitive deficits in patients with multiple sclerosis. We induced experimental autoimmune encephalomyelitis (EAE) in APOE knockout (KO) and human APOE 4 knockin (E4) mice to study the interaction of APOE and neuroinflammation on cognition. After EAE induction, KO and E4 showed significant deficits in spatial learning and recall. Regional decreases in choline acetyltransferase localized to the hippocampus. Induction of EAE in a transgenic APOE animal provides a template from which we can decipher the role APOE has on cognition in the context of neuroinflammation.


Assuntos
Apolipoproteína E4/genética , Transtornos Cognitivos/genética , Encefalomielite Autoimune Experimental/fisiopatologia , Memória , Esclerose Múltipla/genética , Comportamento Espacial , Animais , Animais Geneticamente Modificados , Colina O-Acetiltransferase/metabolismo , Transtornos Cognitivos/enzimologia , Encefalomielite Autoimune Experimental/enzimologia , Técnicas de Inativação de Genes , Hipocampo/enzimologia , Humanos , Esclerose Múltipla/enzimologia , Polimorfismo Genético
13.
Autoimmunity ; 42(3): 209-15, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19301202

RESUMO

Myasthenia gravis (MG) is considered as an autoimmune disease mainly mediated by antibodies against acetylcholine receptor. In recent years, other targets related to MG have been the subject of interest. Our previous research found that protein P25 was lower in muscles of MG patients using two-dimensional electrophoresis. In present study, anti-serum to P25 was prepared, immunohistochemistry and ATPase staining revealed that P25 was a muscle specific cytosolic protein and was mainly distributed in type I muscle fibers. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and precise molecular weight derived from mass spectrometer identified P25 as carbonic anhydrase III (CA III). Some members of CA family are related to autoimmune diseases and CA III is recently reported to be involved in rheumatoid arthritis. The results of immunoblot in this report showed that the level of CA III is specifically insufficient in the skeletal muscle of MG patients. The possible roles that CA III play in MG need further elucidation.


Assuntos
Anidrase Carbônica III/deficiência , Músculo Esquelético/enzimologia , Miastenia Gravis/enzimologia , Adolescente , Adulto , Anticorpos Monoclonais/imunologia , Anidrase Carbônica III/imunologia , Citoplasma/enzimologia , Feminino , Humanos , Soros Imunes/imunologia , Masculino , Pessoa de Meia-Idade , Células Musculares/enzimologia , Fibras Musculares de Contração Rápida/enzimologia , Fibras Musculares de Contração Lenta/enzimologia , Músculos Peitorais/enzimologia , Mapeamento de Peptídeos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA