Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hippocampus ; 31(9): 935-956, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33960056

RESUMO

Neuron-restrictive silencing factor (NRSF) is a zinc-finger transcription factor that regulates expression of a diverse set of genes. However, NRSF function in brain development still remains elusive. In the present study, we generated NRSF-conditional knockout (NRSF-cKO) mice by hGFAP-Cre/loxp system to study the effect of NRSF deficiency on brain development. Results showed that NRSF conditional knockout caused a smaller hippocampus and a thinner granule cell layer (GCL) in mice. Moreover, the reduction and disarrangement of GFAP+ cells in subgranular zone (SGZ) of NRSF-cKO mice was accompanied with the decreased number of premature neurons, neural stem cells (NSCs) and neural progenitor cells (NPCs), as well as compromising the majority of mitotically active cells. The analysis of postnatal development of hippocampus indicated the existence of an abnormality at postnatal day (P) 8, rather than at P1, in NRSF-cKO mice, although the densities of Ki67+ cells with mitotic ability in dentate gyrus were relatively unaffected at P1 and P8. Meanwhile, NRSF deficiency led to abnormal organization of SGZ at P8 during postnatal development. RNA-Seq analysis revealed 79 deregulated genes in hippocampus of NRSF-cKO mice at P8, which were involved in p53 signal transduction, neuron migration and negative regulation of cell proliferation, etc. The deregulation of p53 pathway in NRSF-cKO mice at P1 and P8 was evidenced, of which p21/Cdkn1a was accumulated in a portion of NSCs and NPCs in hippocampus during postnatal development. Together, these results, for the first time, revealed that NRSF could significantly influence the postnatal development of hippocampus, especially the formation of SGZ.


Assuntos
Células-Tronco Neurais , Neurônios , Animais , Giro Denteado , Hipocampo , Camundongos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia
2.
Acta Pharmacol Sin ; 39(10): 1582-1589, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29795362

RESUMO

Both in vivo and in vitro studies have shown the beneficial effects of the delta-opioid receptor (DOR) on neurodegeneration in hypoxia/ischemia. We previously reported that DOR stimulation with [(D-Ala2, D-Leu5) enkephalin] (DADLE), a potent DOR agonist, for both a short (minutes) and long (days) time has notable protective effects against sodium azide (NaN3)-induced cell injury in primary cultured rat cortical neurons. We further demonstrated that short-term DADLE stimulation increased neuronal survival through the PKC-mitochondrial ERK pathway. However, the mechanisms underlying long-term neuroprotection by DADLE remain unclear. Here, we showed that DOR stimulation with DADLE (0.1 µmol/L) for 2 d selectively activates the PI3K/Akt/NF-κB pathway in NaN3-treated neurons; this activation increased Bcl-2 expression, attenuated Cyto c release and promoted neuronal survival. Further investigation revealed that sustained DADLE stimulation increased Bcl-2 expression by enhancing NF-κB binding to the Bcl-2 promoter and upregulating the histone acetylation levels of the Bcl-2 promoter. Our results demonstrate that prolonged DADLE exposure epigenetically promotes Bcl-2 expression and elicits neuroprotective effects in the NaN3 model via the PI3K/Akt/NF-κB pathway.


Assuntos
Leucina Encefalina-2-Alanina/farmacologia , Epigênese Genética/efeitos dos fármacos , Neuroproteção/fisiologia , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , Citocromos c/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Regulação para Cima
3.
J Alzheimers Dis ; 49(3): 829-44, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26577520

RESUMO

Stress is an important risk factor of Alzheimer's disease (AD). It has been evidenced that stress could induce tau phosphorylation and increase tau insolubility in brain; however, little is known about the interactional effect of stress with aging on tauopathy. Therefore, we explored the effects of aging on stress-induced tauopathy and the potential mechanism in mouse model of chronic restraint stress (CRS). Here we found that in general, the level of phosphorylated tau (P-tau) was higher in brain of middle-aged mice than that in adult mice under physiological conditions. CRS-induced tau phosphorylation and its insolubility were more prominent in middle-aged mice. The increase of AT8-labeled insoluble P-tau was dramatic in middle-aged mice, which was highly ubiquitinated but did not form PHF structures. The levels of chaperones were relatively lower in middle-aged mice brain; CRS further reduced the expression, especially for HDJ2/HSP40. CRS also suppressed the expression of Pin1, the peptidylprolyl cis/trans isomerase, in middle-aged mice but not in adult mice. Downregulation of HSP40 or Pin1 caused an increase of transfected extraneous tau in 293 cells. Rosmarinic acid (RA) could effectively suppress the elevation of P-tau and insoluble P-tau formation induced by CRS, and reversed the abnormal changes of chaperones and Pin1 particularly in middle-aged mice. Taken together, our findings provided evidence that aging could be a promoting factor in stress-induced tauopathy, which was relevant with malregulation of chaperones and Pin1, and RA might be a promising beneficial agent for stress-induced tauopathy.


Assuntos
Envelhecimento , Antioxidantes/uso terapêutico , Encéfalo/efeitos dos fármacos , Cinamatos/uso terapêutico , Depsídeos/uso terapêutico , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Hormônio Liberador da Corticotropina/metabolismo , Modelos Animais de Doenças , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Imunoeletrônica , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/metabolismo , Fosforilação , Fatores Desencadeantes , Receptores da Corticotropina/metabolismo , Restrição Física/efeitos adversos , Estresse Psicológico/etiologia , Estresse Psicológico/patologia , Transfecção , Ácido Rosmarínico
4.
Sheng Li Xue Bao ; 66(2): 107-17, 2014 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-24777400

RESUMO

To investigate the murine double minute 2 (MDM2) localization and expression pattern in brain, immunohistochemistry, immunofluorescent staining and immunoblotting methods were used to analyze it in brains of Kunming mice during postnatal development, in brains of adult SD rats and in primarily cultured neurons. The distribution of MDM2 and markers of axon initial segment (AIS) was analyzed by double immunolabeling. In addition, Nutlin-3, a MDM2 antagonist, was injected into hippocampus to analyze the effect on the distribution of MDM2 and AIS protein Nav1.6 in AIS. The results showed that the dynamic expression patterns of MDM2 protein in cerebral cortex and hippocampus of Kunming mice after birth were different. However, it was similar that MDM2 was gradually enriched to AIS during postnatal development, especially after postnatal day 7. The MDM2 in AIS was also observed in different brain regions of adult SD rat brain and in primarily cultured neurons, where MDM2 was colocalized with AIS markers such as AnkG and Nav1.6. In addition, hippocampal injection of Nutlin-3 could induce the loss of the characteristic distribution of MDM2 in AIS. Moreover, Nutlin-3 not only caused a decrease of Nav1.6 distributing in AIS, but also disrupted the polarized distribution of MAP2 in neurons. These results indicate that MDM2 can be enriched at the AIS of adult rodent brain, which might play a role in regulation of the maintenance of AIS function and neuronal polarity.


Assuntos
Axônios/metabolismo , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Animais , Imidazóis/farmacologia , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.6/metabolismo , Piperazinas/farmacologia , Ratos , Ratos Sprague-Dawley
5.
Sheng Li Xue Bao ; 65(3): 253-62, 2013 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-23788181

RESUMO

Small ubiquitin-related modifiers (SUMOs) belong to an important class of ubiquitin like proteins. SUMOylation is a post-translational modification process that regulates the functional properties of many proteins, among which are several proteins implicated in neurodegenerative diseases. This study was aimed to investigate the changes of SUMO-1 expression and modification, and the relationship between SUMO-1 and Alzheimer's disease (AD) pathology in APP/PS1 transgenic AD mice. Using Western blot, co-immunoprecipitation and immunofluorescent staining methods, the SUMO-1 expression and modification and its relation to tau, amyloid precursor protein (APP) and ß-amyloid protein (Aß) in the 12-month-old APP/PS1 transgenic AD mice were analyzed. The results showed that: (1) Compared with the normal wild-type mice, the expression and modification of SUMO-1 increased in brain of AD mice, which was accompanied by an increase of ubiquitination; (2) In RIPA soluble protein fraction of cerebral cortex, co-immunoprecipitation analysis showed tau SUMOylated by SUMO-1 increased in AD mice, however, AT8 antibody labeled phosphorylated tau was less SUMOylated whereas PS422 antibody labeled phosphorylated tau was similar to control mice; (3) Double immunofluorescent staining showed that SUMO-1 could distributed in amyloid plaques, appearing that some of SUMO-1 diffused in centre of some plaques and some of SUMO-1 co-localized with AT8 labeled phosphorylated tau forming punctate aggregates around amyloid plaques which was concerned as dystrophic neurites, however, less Aß, APP and PS422 labeled phosphorylated tau were found co-localized with SUMO-1. These results suggest that SUMO-1 expression and modification increase abnormally in transgenic AD mice, which may participate in modulation of the formation of senile plaques and dystrophic neurites.


Assuntos
Doença de Alzheimer/fisiopatologia , Placa Amiloide/fisiopatologia , Proteína SUMO-1/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/patologia , Camundongos , Camundongos Transgênicos , Neuritos/patologia , Fosforilação , Sumoilação , Proteínas tau/metabolismo
6.
Neurotox Res ; 21(4): 368-78, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22147269

RESUMO

Our previous study has shown that acteoside, an antioxidative phenylethanoid glycoside, protect against beta-amyloid (Aß)-induced cytotoxicity in vitro. However, the precise protective mechanisms remains unclear. Heme oxygenase-1 (HO-1) is a crucial factor in the response to oxidative injury, protecting neurons against Aß-induced injury. In the present study we examined to determine whether acteoside upregulates HO-1 expression, and thereby protects PC12 cells against Aß-induced cell death. It was revealed that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We showed that acteoside increased HO-1 expression in vitro and in vivo. Acteoside treatment resulted in nuclear translocation of the transcription factor NF-E2-related factor 2 (Nrf2). Acteoside activated both ERK and PI3 K/Akt, and treatments with the specific ERK inhibitor PD98059, the PI3 K inhibitor LY294002, and the specific Nrf2 siRNA suppressed the acteoside-induced HO-1 expression. The HO-1 inhibitor ZnPP, PD98059, and LY294002 markedly abolished the neuroprotective effect of acteoside against Aß-induced neurotoxicity. Taken together, these results demonstrate that acteoside is an activator of Nrf2 and inducer of HO-1 expression. We also showed that acteoside increased HO-1 expression through activation of ERK and PI3 K/Akt signal pathways in vitro. Upregulation of HO-1 by acteoside may involve in the neuroprotection against Aß-induced neurotoxicity.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Glucosídeos/farmacologia , Heme Oxigenase-1/biossíntese , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Fenóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Peptídeos beta-Amiloides/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glucosídeos/antagonistas & inibidores , Heme Oxigenase-1/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/metabolismo , Degeneração Neural/induzido quimicamente , Células PC12 , Fenóis/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
7.
Int J Neurosci ; 122(6): 290-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22185089

RESUMO

Accumulation of the amyloid ß protein (Aß) in the brain is an important step in the pathogenesis of Alzheimer's disease. Many molecules could bind with Aß, among which some molecules mediate Aß neuronal toxicity. Thus, it is of interest to study the binding proteins of Aß, and the functions that might be affected by Aß. In the present study, we observed that accumulation of α-subunit of ATP synthase is associated with aggregates of Aß proteins in amyloid plaques of amyloid precursor protein/presennillin-1 transgenic mice, and identified the α-subunit of ATP synthase as one of the Aß binding proteins on the plasma membrane of neural cells by Western blot and mass spectrometry. In order to evaluate the consequences of the interaction between Aß and surface α-subunit of ATP synthase, the extracellular ATP generation was analyzed, which showed that aggregated Aß partially inhibited the extracellular generation of ATP, but was unable to significantly induce a decrease in cell surface ATP synthase α on neurons. These results suggest that the cell surface ATP synthase α is a binding protein for Aß on neural cells, the functional inhibition of surface ATP synthase might be involved in machinery of brain malfunction in Aß-mediated pathogenesis of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Trifosfato de Adenosina/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1/genética , Ratos , Ratos Sprague-Dawley
8.
Neurosci Bull ; 27(3): 163-72, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21614099

RESUMO

OBJECTIVE: Munc18-1 has an important role in neurotransmitter release, and controls every step in the exocytotic pathway in the central nervous system. In the present study, whether epileptic seizure causes a change of Munc18 localization in neuronal nuclei was analyzed. METHODS: Epilepsy models were established by injection of kainic acid (KA) solution into hippocampus of Sprague-Dawley (SD) rats or intraperitoneal injection of KA in Kunming mice. The hippocampal neurons were prepared from embryonic day 18 SD rats, and cultured in neurobasal medium, followed by treatment with glutamate for 3 h. Neuronal and glial nuclei of hippocampus were separated by sucrose density gradient centrifugation. The nucleus-enriched fractions were stained with 0.1% Cresyl Violet for morphological assay. Immunochemistry and immunoelectron microscopy with anti-Munc18-1 antibody were used to determine the nuclear localization of Munc18-1. Immunoblotting was used to detect the protein level of Munc18-1. RESULTS: The localization of Munc18-1 in nucleus of rat hippocampal neuron was confirmed by immunochemistry, immunoelectron microscopy, and immunoblotting detection of neuronal nucleus fraction. In animals receiving intrahippocampal or intraperitoneal injection of KA, immunostaining revealed that the expression of Munc18-1 decreased in pyramidal cell layer of CA regions, as well as in hilus and granular cell layer of dentate gyrus in hippocampus. Moreover, immunoblotting analysis showed that the expression level of Munc18-1 in nucleus fraction of hippocampus significantly decreased in KA-treated animals. The relationship between the change of Munc18-1 expression in neuronal nuclei and neuronal over-activation was also tested in primary cultured neurons. After treatment with 50 µmol/L glutamate acid for 3 h, Munc18-1 level was decreased in nucleus fraction and increased in cytoplasmic fraction of primary cultured neurons. CONCLUSION: These results suggest that excitatory stimulation can induce the distribution change of Munc18-1 in neuron, which may subsequently modulate neuronal functions in brain.


Assuntos
Núcleo Celular/metabolismo , Epilepsia/metabolismo , Proteínas Munc18/metabolismo , Neurônios/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Agonistas de Aminoácidos Excitatórios , Hipocampo/citologia , Hipocampo/metabolismo , Imuno-Histoquímica , Ácido Caínico , Masculino , Camundongos , Camundongos Endogâmicos , Neuroglia/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Cell Biol Int ; 35(1): 81-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20626349

RESUMO

The ATP synthase is known to play important roles in ATP generation and proton translocation within mitochondria. Here, we now provide evidence showing the presence of functional ecto-ATP synthase on the neuronal surface. Immunoblotting revealed that the α, ß subunits of ATP synthase F1 portion are present in isolated fractions of plasma membrane and biotin-labelled surface protein from primary cultured neurons; the surface distribution of α, ß subunits was also confirmed by immunofluorescence staining. Moreover, α and ß subunits were also found in fractions of plasma membrane and lipid rafts isolated from rat brain, and flow cytometry analysis showed α subunits on the surface of acutely isolated brain cells. Activity assays showed that the extracellular ATP generation of cultured neurons could be compromised by α, ß subunit antibodies and ATP synthase inhibitors. pH(i) (intracellular pH) analysis demonstrated that at low extracellular pH, α or ß subunit antibodies decreased pHi of primary cultured neurons. Therefore, ATP synthase on the surface of neurons may be involved in the machineries of extracellular ATP generation and pH(i) homoeostasis.


Assuntos
Complexos de ATP Sintetase/fisiologia , Equilíbrio Ácido-Base , Trifosfato de Adenosina/biossíntese , Líquido Extracelular/metabolismo , Neurônios/metabolismo , Animais , Membrana Celular/enzimologia , Células Cultivadas , Embrião de Mamíferos/citologia , Feminino , Concentração de Íons de Hidrogênio , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
10.
Brain Res ; 1360: 159-67, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-20828541

RESUMO

Astaxanthin (ATX), the most abundant flavonoids in propolis, has been proven to exert neuroprotective property against glutamate-induced neurotoxicity and ischemia-reperfusion-induced apoptosis. Previous study have revealed that ATX can rescue PC12 cells from Aß(25-35)-induced apoptotic death. However, the mechanisms by which ATX mediates its therapeutic effects in vitro are unclear. In the present study, we explored the underlying mechanisms involved in the protective effects of ATX on the Aß(25-35)-induced cytotoxicity in SH-SY5Y cells. Pre-treatment with ATX for 4h significantly reduced the Aß(25-35)-induced viability loss, apoptotic rate and attenuated Aß-mediated ROS production. In addition, ATX inhibited Aß(25-35)-induced lowered membrane potential, decreased Bcl-2/Bax ratio. We also demonstrated that ATX could prevent the activation of p38MAPK kinase pathways induced by Aß. Moreover, we for the first time have revealed the ATX increased antioxidant enzyme heme oxygenase-1 (HO-1) expression in concentration-dependent and time-dependent manners, which were correlated with its protective effect against Aß(25-35)-induced injury. Because the inhibitor of HO-1 activity, ZnPP reversed the protective effect of ATX against Aß(25-35)-induced cell death. We also demonstrated that the specific ERK inhibitor, PD98059, concentration-dependently blocked on ATX-induced HO-1 expression, and meanwhile PD98059 reversed the protective effect of ATX against Aß25-35-induced cell death. Taken together, these findings suggest that astaxanthin can induce HO-1 expression through activation of ERK signal pathways, thereby protecting the SH-SY5Y cells from Aß(25-35)-induced oxidative cell death.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/toxicidade , Heme Oxigenase-1/biossíntese , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Fármacos Neuroprotetores , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Proteínas Reguladoras de Apoptose/biossíntese , Western Blotting , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Flavonoides/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Humanos , Metaloproteinases da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos , Xantofilas/antagonistas & inibidores , Xantofilas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Neurosci Bull ; 26(4): 273-81, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20651809

RESUMO

OBJECTIVE: Munc18 is considered as an intracellular protein that plays an important role in exocytosis of neurotransmitters. Previous studies have demonstrated the presence of autoantibodies against Munc18 in a subgroup of Rasmussen's encephalitis patients. However, the machinery of Munc18 autoimmunity is still elusive. The present study was aimed to investigate Munc18 release from primary cultured neurons, Munc18 distribution on the outer plasma membrane of neurons, and the neurotoxicity of Munc18 antibody. METHODS: The cerebral cortical neurons from embryonic day 17 Sprague-Dawley rats were prepared and cultured in neurobasal medium. The proteins in culture medium were precipitated with 10 % trichloroacetic acid, and analyzed by immunoblotting. The proteins on neuronal surface were biotinylated with EZ-Link-sulfo-NHS-LC-Biotin, and collected with avidin-conjugated agarose beads followed by immunoblotting analysis. For cell surface immunofluorescent staining, the living neurons were labeled with anti-Munc18 antibody at 4 degrees C. Neuronal injury was assessed by lactate dehydrogenase(LDH) release. RESULTS: Munc18 was detected in culture medium by immunoblotting analysis. After treatment with 50 micromol/L glutamate for 1 h, Munc18 content in medium was increased. Meanwhile, beta-actin and syntaxin1 were not detected in culture medium, and LDH release was not significantly increased. Moreover, glutamate enhanced Munc18 distribution on outer plasma membrane. Living neuron staining also demonstrated the localization of Munc18 on neuronal surface after glutamate treatment, especially at contacting regions between neurons. Glutamate-induced increase of surface Munc18 distribution was suppressed by NMDA receptor antagonist MK801, but not by AMPA receptor antagonist NBQX. Moreover, compared with c-Fos antibody, Munc18 antibody could induce neuronal injury, when culture medium contained the components of serum. CONCLUSION: A portion of Munc18 can be released from neurons or distributed on neuronal surface, which can be enhanced by glutamate treatment via activation of NMDA receptors. Besides, Munc18 antibody-induced neuronal injury depends on the serum components.


Assuntos
Córtex Cerebral/citologia , Ácido Glutâmico/farmacologia , Proteínas Munc18/metabolismo , Neurônios/efeitos dos fármacos , Animais , Biotinilação/métodos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Transporte Proteico/efeitos dos fármacos , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
12.
Brain Res ; 1347: 132-41, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20513368

RESUMO

Microtubule-associated proteins (MAPs) play a critical role in maintaining normal cytoskeletal architecture and functions. In the present study, we aim to explore the effects of the emotional stressor, chronic restraint stress, on the expression levels and localization of tau and MAP2. We found that after chronic restraint stress, soluble hyperphosphorylated tau was greatly increased, whereas MAP2 was decreased. Moreover, immunohistochemistry analysis demonstrated that phosphorylated tau and MAP2 displayed the similar subcellular distribution pattern after chronic restraint stress. Robust hyperphosphorylated tau immunolabeling was observed both in cortex and hippocampus of stressed animals and mainly located to perikaryal/dendritic elements. After stress, the MAP2 was mainly distributed in the perikaryal compartments, discontinuous dendrites and neuropil. Moreover, the distribution pattern of MAP2 in hippocampus significantly changed. Immunofluorescence double labeling indicated that hyperphosphorylated tau increased in the regions where there displayed an decrease of MAP2. These results suggest that the involvement of repeated restraint stress may not only induce phosphorylation state of tau and distribution of phosphorylated tau, but also alter the content and neuronal distribution of MAP2. Tau and MAP2 are most important MAPs for neuronal cells, the subcellular distribution change of them might be link to functional change of neurons after emotional stress.


Assuntos
Córtex Cerebral/metabolismo , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Estresse Psicológico/patologia , Proteínas tau/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Restrição Física/métodos , Estresse Psicológico/etiologia
13.
Biochem Biophys Res Commun ; 390(4): 1294-8, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19878659

RESUMO

Huntington's disease (HD) and other polyglutamine (polyQ) neurodegenerative diseases are characterized by neuronal accumulation of the disease protein, suggesting that the cellular ability to handle abnormal proteins is compromised. As a multi-subunit protein localized in the mitochondria of eukaryotic cells, the F(0)F(1)-ATP synthase alpha belongs to the family of stress proteins HSP60. Currently, mounting evidences indicate F(0)F(1)-ATP synthase alpha may play a role in neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Recently, ATP synthase alpha was reported to have protective and therapeutic roles in primary cardiacmyocytes of iron-overloaded rats by lowering ROS production. However, little is understood about the role of ATP synthase alpha in cell death and neurodegeneration. Here, we demonstrate that overexpression of ATP synthase alpha suppresses huntingtin (htt) polyQ aggregation and toxicity in transfected SH-SY5Y cell lines. Overexpression of ATP synthase alpha is able to protect cell death caused by polyglutamine-expanded htt. Transient overexpression of ATP synthase alpha suppresses the aggregate formation by estimation of polyQ aggregation, Western blot analysis, and filter trap assay (FTA) in transfected SH-SY5Y cells. These results indicated that ATP synthase alpha has a strong inhibitory effect on polyglutamine aggregate formation and toxicity in vitro, and suggest a novel neuroprotective role of ATP synthase alpha.


Assuntos
Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , ATPases Translocadoras de Prótons/biossíntese , Animais , Apoptose , Linhagem Celular Tumoral , Humanos , Proteína Huntingtina , Camundongos , Peptídeos/farmacologia , Ratos
14.
Neurosci Lett ; 465(3): 276-81, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19766167

RESUMO

The biological function of full-length amyloid-beta protein precursor (APP), the precursor of Abeta, is not fully understood. Mounting studies reported that antibody binding to cell surface APP causes neuronal injury. However, the mechanism of cell surface APP mediating neuronal injury remains to be determined. Colocalization of APP with integrin on cell surface leads us to suppose that focal adhesion (FA) related mechanism is involved in surface APP-mediated neuronal injury. In the present study, results demonstrated that primary cultured neurons treated with antibody against APP-N-terminal not only caused neuronal injury and aberrant morphologic changes of neurite, but also induced reaction of FA proteins appearing an acute increase then decrease pattern. Moreover, the elevation of tyrosine phosphorylation of FA proteins including paxillin and focal adhesion kinase (FAK), and down-regulated expression of protein tyrosine phosphatase (PTP1B) induced by APP antibody were prevented by inhibitor of Src protein kinases 4-amino-5-(4-chlorophenyl)-7(t-butyl) pyrazol (3,4-D) pyramide (PP2) and G protein inhibitor pertussis toxin (PTX), implying that Src family kinase and G protein play roles in APP-induced FA signals. In addition, pretreatment with PTX and PP2 was able to suppress APP-antibody induced neuronal injury. Taken together, the results suggest a novel mechanism for APP mediating neuronal injury through deregulating FA signals.


Assuntos
Precursor de Proteína beta-Amiloide/imunologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Proteína-Tirosina Quinases de Adesão Focal/imunologia , Neurônios/imunologia , Neurônios/patologia , Transdução de Sinais/imunologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/imunologia , Ratos , Transdução de Sinais/efeitos dos fármacos
15.
Brain Res ; 1252: 183-91, 2009 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-19056363

RESUMO

It is recognized in recent years that activation of delta-opioid receptor (DOR) elicits neuroprotection against hypoxia and ischemia. However, the underlying mechanisms are not well understood yet. Mitochondrial dysfunction plays a key role in hypoxic neuronal injury, but the effect of DOR activation on neurons with a mitochondrial respiratory chain deficiency is poorly elucidated. In this study we tested the effects of DOR activation and inhibition on cultured cortical neurons after inhibiting mitochondrial respiratory chain with sodium azide (NaN(3)) in days 8 cultures. Neuronal injury was assessed by lactate dehydrogenase release. Changes in DOR proteins were investigated using an antibody against the N-terminus of the DOR, which recognizes the 60, 48, and 32 kDa proteins. Our main findings are that 1) delta- but not mu-opioid receptor activation reduces NaN(3)-induced neuronal damage, and this neuroprotective effect is abolished by DOR antagonist (naltrindole, NTI); 2) prolonged DOR inhibition with NTI further increases NaN(3)-induced neuronal damage; 3) NaN(3) treatment down-regulates DOR protein levels in neurons, and the 60 and 32 kDa proteins are particularly sensitive; 4) DADLE, besides activating DOR directly, also reverses the decrease of neuronal DOR protein levels induced by NaN(3), which may contribute greatly to its neuroprotective effect; 5) NTI reverses NaN(3)-induced down-regulation of DOR proteins as well, the effect of NTI amplifying NaN(3)-induced neuronal damage therefore is probably due to its inhibition on DOR activity only. In conclusion, these data suggest that DOR activation plays an important role in neuroprotection against mitochondrial respiratory chain injury.


Assuntos
Mitocôndrias/metabolismo , Neurônios/metabolismo , Receptores Opioides delta/metabolismo , Animais , Western Blotting , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Transporte de Elétrons/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Leucina Encefalina-2-Alanina/farmacologia , Imunoprecipitação , L-Lactato Desidrogenase/metabolismo , Mitocôndrias/efeitos dos fármacos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/metabolismo , Azida Sódica/farmacologia
16.
Zhong Xi Yi Jie He Xue Bao ; 6(1): 68-72, 2008 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-18184549

RESUMO

OBJECTIVE: To explore whether there are beta-amyloid protein (Abeta) binding elements in heart-beneficial recipe (HBR, a compound traditional Chinese herbal medicine), which can ameliorate the cytotoxicity of Abeta. METHODS: The extract of HBR and Abeta(1-40) were co-precipitated, and the Abeta(1-40) in pellets was detected by immunoblotting. Affi-gel-Abeta(1-40) was constructed, and Affi-gel-Abeta(1-40) affinity elements from the extract of HBR were analyzed by high-performance liquid chromatography (HPLC). The assay of lactic dehydrogenase (LDH) release from the primary cultured rat cortex neurons was used to evaluate the cytotoxicity of Abeta(1-42), and the protection effects of the HBR serum and the Affi-gel-Abeta(1-40) treated HBR serum. RESULTS: Immunoblotting examination showed Abeta(1-40) could be co-precipitated with components of HBR following co-incubation, and the amount of Abeta(1-40) within pellets decreased when the HBR extract was diluted. Abeta(1-40) affinity elements from the extract of HBR, eluted from Affi-gel-Abeta(1-40) by glycine solution (pH=2.5), could be detected by HPLC-fluorescent detector system. The analysis of LDH release showed that exposure of neurons to 5 micromol/L Abeta(1-42) for 48 h caused a significant increase of LDH release in either a serum free or 10% serum contained culture condition (P<0.01). The rat HBR serum was able to suppress Abeta(1-42) induced LDH release (P<0.05), whereas Affi-gel-Abeta(1-40) treated HBR serum still maintained the ability to attenuate Abeta(1-42) induced LDH release although the effect was somewhat decreased compared with Affi-gel treated HBR serum. CONCLUSION: There are Abeta affinity components in HBR, which could not increase the Abeta cytotoxicity, but might be able to inhibit the cytotoxicity of Abeta. The results implied that the exploration of Abeta affinity elements from Chinese medicinal recipe which is effective for Alzheimer disease, might be an important direction in Alzheimer disease therapeutic research area.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Medicamentos de Ervas Chinesas/farmacologia , Neurônios/citologia , Fármacos Neuroprotetores/farmacologia , Animais , Células Cultivadas , Interações Medicamentosas , Medicamentos de Ervas Chinesas/química , Masculino , Fármacos Neuroprotetores/química , Ratos
17.
Neurosci Lett ; 406(3): 178-82, 2006 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-16919876

RESUMO

Accumulating evidence indicates that the aberrant re-entry of post-mitotic neurons into the G2/M phase of cell cycle and the resulting mitotic catastrophe may contribute to the pathogenesis of Alzheimer's disease. However, the cellular event that drives the differentiated neurons to abnormally enter G2/M phase remains elusive. Similarly, whether mitotic catastrophe is indeed one of the death pathways for differentiated neurons is not clear. Previous studies revealed that okadaic acid (OA), a phosphatase inhibitor that induces AD like pathological changes, evokes mitotic changes in neuroblastoma cells. In this study, we examined the in vivo effects of OA on cyclin B1 expression, the induction of mitosis, and subsequent mitotic catastrophe. We found that cyclin B1 expression in adult neurons was significantly increased after injecting OA into rat frontal cortex, which also increased tau protein phosphorylation. Interestingly, cyclin B1 and phosphorylated tau were well co-localized around the OA injection site, but were only partially co-localized in other brain regions. Staining with toluidine blue, Giemsa dye or propidium iodide revealed typical mitotic and mitotic catastrophe-like morphological changes with irregular arrangement of condensed chromatin and chromosome fibers in a few cells. Furthermore, the strong cyclin B1 staining in these cells suggests that cyclin B1 promoted G2 to M phase transition is required for the mitotic catastrophe. The detection of neuron-specific enolase in a portion of these cells demonstrated that at least part them are neuron. All together, our results suggest that the disturbance of the protein kinase-phosphatase system caused by OA is sufficient to induce neuronal cyclin B1 expression, force neurons into the mitotic phase of cell cycle, and cause mitotic catastrophe.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Ciclina B/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Mitose/efeitos dos fármacos , Ácido Okadáico/farmacologia , Animais , Western Blotting/métodos , Ciclina B1 , Imunofluorescência/métodos , Masculino , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Proteínas tau/metabolismo
18.
Neurosci Lett ; 388(1): 13-6, 2005 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-16005567

RESUMO

The abnormal hyperphosphorylation of tau protein in brain is attributed to a number of neurodegenerative diseases such as Alzheimer disease. It has been reported that cold water stress (CWS) could cause rapid reversible tau phosphorylation in brain. To explore the possible long-tem effects of CWS on tau phosphorylation, we employed the immunoblot and immunohistochemical methods to analyze the phosphorylation of tau in the hippocampus of mice subjected to CWS. Results showed that CWS stimulation caused not only an early phase reversible tau phosphorylation, but also a later phase tau phosphorylation after 6h. The distribution pattern of phosphorylated tau (P-tau) in the later phase was different to that of early phase. At 1h after CWS, defined as early phase, P-tau was strikingly located in the mossy fibers and nerve terminals at the molecular layer of dentate gray (DG), whereas at 12h, defined as later phase, P-tau was dominantly located in the somatodendritic compartments of neurons in DG and CA3/CA1 regions, but obviously decreased in the mossy fibers and nerve terminals of molecular layer. These findings demonstrate that CWS leads to prominent changes of tau phosphorylation and P-tau localization in the hippocampus in a time dependent manner.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Estresse Psicológico/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/psicologia , Animais , Causalidade , Temperatura Baixa/efeitos adversos , Giro Denteado/metabolismo , Giro Denteado/fisiopatologia , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Masculino , Camundongos , Fibras Musgosas Hipocampais/metabolismo , Fibras Musgosas Hipocampais/fisiopatologia , Fosforilação , Terminações Pré-Sinápticas/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia , Fatores de Tempo , Regulação para Cima/fisiologia
19.
Cell Res ; 13(2): 111-20, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12737519

RESUMO

RMP was reported to regulate transcription via competing with HBx to bind the general transcription factor IIB (TFIIB) and interacting with RPB5 subunit of RNA polymerase II as a corepressor of transcription regulator. However, our present research uncovered that RMP also regulates the transcription through interaction with the general transcription factors IIF (TFIIF), which assemble in the preinitiation complex and function in both transcription initiation and elongation. With in vitro pull-down assay and Far-Western analysis, we demonstrated that RMP could bind with bacterially expressed recombinant RAP30 and RAP74 of TFIIF subunits. In the immunoprecipitation assay in COS1 cells cotransfected with FLAG-tagged RMP or its mutants, GST-fused RAP30 and RAP74 were co-immunoprecipitated with RMP in approximately equal molar ratio, which suggests that RAP30 and RAP74 interact with RMP as a TFIIF complex. Interestingly both RAP30 and RAP74 interact with the same domain (D5) of the C-terminal RMP of 118-amino-acid residuals which overlaps with its TFIIB-binding domain. Internal deletion of D5 region of RMP abolished its binding ability with both subunits of TFIIF, while D5 domain alone was sufficient to interact with TFIIF subunits. The result of luciferase assay showed that overexpression of RMP, but not the mutant RMP lacking D5 region, suppressed the transcription activated by Gal-VP16, suggesting that interaction with TFIIF is required for RMP to suppress the activated transcription. The interaction between RMP and TFIIF may be an additional passway for RMP to regulate the transcription, or alternatively TFIIF may cooperate with RPB5 and TFIIB for the corepressor function of RMP.


Assuntos
Proteínas de Transporte/genética , Células Eucarióticas/metabolismo , Genes Reguladores/genética , Genes Supressores/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Fatores de Transcrição TFII/genética , Transcrição Gênica/genética , Ativação Transcricional/genética , Animais , Sítios de Ligação/genética , Células COS , Estrutura Terciária de Proteína/genética , RNA Polimerase II/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Repressoras
20.
Sheng Li Xue Bao ; 55(2): 142-6, 2003 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-12715101

RESUMO

Recent evidence indicates that the aberrant neuronal expression of mitotic proteins in Alzheimer's disease (AD) brain may be related to AD pathological changes. To investigate whether the toxicity of beta-amyloid protein (Abeta) induces mitotic proteins expression in adult rat brain, we used immunohistochemical and integral optical density analytic method to analyze the adult rat brains, which had been injected with Abeta(25-35) into unilateral amygdala. Results showed that the levels of neurofibrillary tangle (NFT) related phosphorylated tau protein and apoptosis related protein Bax were increased in Abeta(25-35) injected rat brains, meanwhile the aberrantly expression of mitotic protein cyclin A and cyclin B1 was also detected at 7 d after operation, but the level of cyclin A decreased and cyclin B1 disappeared at 21 d. Immunofluorescence double labeling presented that cyclin B1 was partially co-localized with Bax or phosphorylated tau protein, whereas Bax and phosphorylated tau protein seldom co-localized. These results suggest that Abeta causes mitotic protein expression in adult brain neurons, which may die through apoptosis or may be affected by AD NFT-related tau phosphorylation.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Peptídeos beta-Amiloides/toxicidade , Ciclina A/metabolismo , Ciclina B1/metabolismo , Fragmentos de Peptídeos/toxicidade , Proteína X Associada a bcl-2/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Masculino , Neurônios/metabolismo , Fosforilação , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...