Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Chin Med J (Engl) ; 129(23): 2845-2852, 2016 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-27901000

RESUMO

BACKGROUND: Amyloid ß (Aß) deposits and the endoplasmic reticulum stress (ERS) are both well established in the development and progression of Alzheimer's disease (AD). However, the mechanism and role of Aß-induced ERS in AD-associated pathological progression remain to be elucidated. METHODS: The five familial AD (5×FAD) mice and wild-type (WT) mice aged 2, 7, and 12 months were used in the present study. Morris water maze test was used to evaluate their cognitive performance. Immunofluorescence and Western blot analyses were used to examine the dynamic changes of pro-apoptotic (CCAAT/enhancer-binding protein homologous protein [CHOP] and cleaved caspase-12) and anti-apoptotic factors (chaperone glucose-regulated protein [GRP] 78 and endoplasmic reticulum-associated protein degradation-associated ubiquitin ligase synovial apoptosis inhibitor 1 [SYVN1]) in the ERS-associated unfolded protein response (UPR) pathway. RESULTS: Compared with age-matched WT mice, 5×FAD mice showed higher cleaved caspase-3, lower neuron-positive staining at the age of 12 months, but earlier cognitive deficit at the age of 7 months (all P < 0.05). Interestingly, for 2-month-old 5×FAD mice, the related proteins involved in the ERS-associated UPR pathway, including CHOP, cleaved caspase-12, GRP 78, and SYVN1, were significantly increased when compared with those in age-matched WT mice (all P < 0.05). Moreover, ERS occurred mainly in neurons, not in astrocytes. CONCLUSIONS: These findings suggest that compared with those of age-matched WT mice, ERS-associated pro-apoptotic and anti-apoptotic proteins are upregulated in 2-month-old 5×FAD mice, consistent with intracellular Aß aggregation in neurons.


Assuntos
Doença de Alzheimer/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Neurônios/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/fisiologia , Western Blotting , Caspase 12/metabolismo , Chaperona BiP do Retículo Endoplasmático , Lobo Frontal/metabolismo , Proteínas de Choque Térmico/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fator de Transcrição CHOP/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Resposta a Proteínas não Dobradas/fisiologia
3.
Mol Neurodegener ; 11(1): 51, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27406263

RESUMO

BACKGROUND: Apolipoprotein E (ApoE) is a major lipid carrier that supports lipid transport and injury repair in the brain. The APOE ε4 allele is associated with depression, mild cognitive impairment (MCI) and dementia; however, the precise molecular mechanism through which ApoE4 influences the risk of disease development remains unknown. To address this gap in knowledge, we investigated the potential effects of chronic unpredictable mild stress (CUMS) on ApoE3 and ApoE4 target replacement (ApoE3-TR and ApoE4-TR) mice. RESULTS: All ApoE-TR mice exposed to CUMS at 3 months old recovered from a depression-like state by the age of 12 months. Of note, ApoE4-TR mice, unlike age-matched ApoE3-TR mice, displayed impaired spatial cognitive abilities, loss of GABAergic neurons, decreased expression of Reelin, PSD95, SYN and Fyn, and reduced phosphorylation of NMDAR2B and CREB. CONCLUSION: These results suggest that early-life stress may mediate cognitive impairment in middle-age ApoE4-TR mice through sustained reduction of GABAergic neurons and Reelin expression, which might further diminish the activation of the Fyn/NMDAR2B signaling pathway.


Assuntos
Apolipoproteína E4/genética , Demência/metabolismo , Memória/fisiologia , Neurônios/metabolismo , Aprendizagem Espacial/fisiologia , Estresse Psicológico/fisiopatologia , Envelhecimento , Animais , Apolipoproteína E4/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteína Reelina
4.
Chin Med J (Engl) ; 129(15): 1835-44, 2016 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-27453234

RESUMO

BACKGROUND: Amyloid ß (Aß) has been established as a key factor for the pathological changes in the brains of patients with Alzheimer's disease (AD), and cellular senescence is closely associated with aging and cognitive impairment. However, it remains blurred whether, in the AD brains, Aß accelerates the neuronal senescence and whether this senescence, in turn, impairs the cognitive function. This study aimed to explore the expression of senescence-associated genes in the hippocampal tissue from young to aged 5XFAD mice and their age-matched wild type (WT) mice to determine whether senescent neurons are present in the transgenic AD mouse model. METHODS: The 5XFAD mice and age-matched wild type mice, both raised from 1 to 18 months, were enrolled in the study. The senescence-associated genes in the hippocampus were analyzed and differentially expressed genes (DEGs) were screened by quantitative real-time polymerase chain reaction. Cognitive performance of the mice was evaluated by Y-maze and Morris water maze tests. Oligomeric Aß (oAß) (1-42) was applied to culture primary neurons to simulate the in vivo manifestation. Aging-related proteins were detected by Western blotting analysis and immunofluorescence. RESULTS: In 5XFAD mice, of all the DEGs, the senescence-associated marker p16 was most significantly increased, even at the early age. It was mainly localized in neurons, with a marginal expression in astrocytes (labeled as glutamine synthetase), nil expression in activated microglia (labeled as Iba1), and negatively correlated with the spatial cognitive impairments of 5XFAD mice. oAß (1-42) induced the production of senescence-related protein p16, but not p53 in vitro, which was in line with the in vivo manifestation. CONCLUSIONS: oAß-accelerated neuronal senescence may be associated with the cognitive impairment in 5XFAD mice. Senescence-associated marker p16 can serve as an indicator to estimate the cognitive prognosis for AD population.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Células Cultivadas , Senescência Celular/genética , Senescência Celular/fisiologia , Cognição/fisiologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Reação em Cadeia da Polimerase em Tempo Real
5.
Mol Neurobiol ; 53(9): 6397-6406, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26582466

RESUMO

Due to its apparent rate-limiting function, BACE1 (ß-secretase) appears to be a prime target for prevention of amyloid-ß (Aß) generation in brains with Alzheimer's disease (AD). The activity of BACE1 is regulated by peroxisome proliferator-activated receptor-γ (PPARγ), a transcription factor binding site of the BACE1 promoter, indicating that PPARγ may be a potential target for AD treatment. Several studies have demonstrated that PPARγ activation is involved in the immunostimulation of amyloid-ß precursor protein processing by nonsteroidal anti-inflammatory drugs (NSAIDs). The present study found that tripchlorolide (T4), with a similar chemical structure to that of NSAIDs, decreased the levels of Aß secreted in N2a-APP695 cells. T4 treatment reduced the mRNA and protein levels of BACE1 and the protein level of sAPPß, a cleaved N-terminal fragment of APP by BACE1. The treatment also translocated PPARγ from cytoplasm to nuclear. Intriguingly, T4, like pioglitazone (a PPARγ agonist), suppressed the BACE1 activity in N2a-APP695 cells, which was attenuated by GW9662 (a PPARγ antagonist). These results indicate that T4 may be a PPARγ agonist to enhance the binding of nuclear PPARγ to the BACE1 promoter, which may in turn inhibit the transcription and translation of BACE1, suppress the activity of BACE1, and ultimately attenuate the generation of Aß. Due to its capability to alter Aß generation and to protect central neural system against the neurotoxicity of Aß, T4 may serve as a promising agent in modulating Aß-related pathology in Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Diterpenos/farmacologia , PPAR gama/metabolismo , Fenantrenos/farmacologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Diterpenos/química , Humanos , Camundongos , Camundongos Transgênicos , Fenantrenos/química , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
6.
Chin Med J (Engl) ; 128(16): 2220-7, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-26265617

RESUMO

BACKGROUND: Early diagnosis assumes a vital role in an effective treatment of Alzheimer's disease (AD). Most of the current studies can only make an AD diagnosis after the manifestation of typical clinical symptoms. The present study aimed to investigate typical and other biomarkers of AD to find a possible early biomarker. METHODS: A total of 14 5XFAD mice (at 3 and 6 months old), with 14 age-matched wild-type (WT) mice as control, were enrolled in this case-control study. Morris water maze test was performed to evaluate the cognitive function; buried food pellet test and olfactory maze test were employed to investigate the olfactory function; immunofluorescence to detect amyloid deposition and positron emission tomography to examine 2-deoxy-2-(18F) fluoro-D-glucose ([18F]-FDG) uptake in the hippocampus and cerebral cortex. RESULTS: With the increasing age, cognitive performance (P = 0.0262) and olfactory function were significantly deteriorated (day 1 P = 0.0012, day 2 P = 0.0031, day 3 P = 0.0160, respectively) and the (18F)-FDG uptake was markedly decreased in multi-cerebral regions including the olfactory bulb (P < 0.0001), hippocampus (P = 0.0121), and cerebral cortex (P < 0.0001). Of note, in 3-month-old 5XFAD mice, a significant decline of (18F)-FDG uptake in the olfactory bulb was found when compared with that of age-matched WT mice (P = 0.023) while no significant difference was present when the uptakes in other cerebral regions were compared. CONCLUSIONS: The decline of (18F)-FDG uptake in the olfactory bulb occurs earlier than other incidents, serving as an earlier in vivo biological marker of AD in 5XFAD mice and making early diagnosis of AD possibly.


Assuntos
Doença de Alzheimer/diagnóstico , Biomarcadores/análise , Glucose/metabolismo , Bulbo Olfatório/metabolismo , Amiloide/análise , Animais , Animais Geneticamente Modificados , Fluordesoxiglucose F18/metabolismo , Camundongos , Tomografia por Emissão de Pósitrons
7.
Glia ; 63(12): 2208-19, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26200696

RESUMO

Oxidative stress plays an important role in the progression of Alzheimer's disease (AD) and other neurodegenerative conditions. Glutathione (GSH), the major antioxidant in the central nervous system, is primarily synthesized and released by astrocytes. We determined if ß-amyloid (Aß42), crucially involved in Alzheimer's disease, affected GSH release. Monomeric Aß (mAß) stimulated GSH release from cultured cortical astrocytes more effectively than oligomeric Aß (oAß) or fibrillary Aß (fAß). Monomeric Aß increased the expression of the transporter ABCC1 (also referred to as MRP1) that is the main pathway for GSH release. GSH release from astrocytes, with or without mAß stimulation, was reduced by pharmacological inhibition of ABCC1. Astrocytes robustly express connexin proteins, especially connexin43 (Cx43), and mAß also stimulated Cx43 hemichannel-mediated glutamate and GSH release. Aß-stimulation facilitated hemichannel opening in the presence of normal extracellular calcium by reducing astrocyte cholesterol level. Aß treatment did not alter the intracellular concentration of reduced or oxidized glutathione. Using a mouse model of AD with early onset Aß deposition (5xFAD), we found that cortical ABCC1 was significantly increased in temporal register with the surge of Aß levels in these mice. ABCC1 levels remained elevated from 1.5 to 3.5 months of age in 5xFAD mice, before plunging to subcontrol levels when amyloid plaques appeared. Similarly, in cultured astrocytes, prolonged incubation with aggregated Aß, but not mAß, reduced induction of ABCC1 expression. These results support the hypothesis that in the early stage of AD pathogenesis, less aggregated Aß increases GSH release from astrocytes (via ABCC1 transporters and Cx43 hemichannels) providing temporary protection from oxidative stress which promotes AD development.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Glutationa/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Córtex Cerebral/metabolismo , Colesterol/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/antagonistas & inibidores , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Oxirredução , Placa Amiloide/metabolismo
8.
Mol Neurodegener ; 10: 7, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25871877

RESUMO

BACKGROUND: Alzheimer's disease (AD) causes progressive loss of memory and cognition, exacerbated by APOE4, the greatest genetic risk factor for AD. One proposed mechanism for apolipoprotein E (apoE) effects on cognition is via NMDAR-dependent signaling. APOE genotype-specific effects on this pathway were dissected using EFAD-transgenic (Tg) mice (5xFAD mice, that over-express human amyloid-beta (Aß) via 5 familial-AD (FAD) mutations, and express human apoE), and 5xFAD/APOE-knockout (KO) mice. Previous data from EFAD-Tg mice demonstrate age-dependent (2-6 months), apoE-specific effects on the development of Aß pathology. This study tests the hypothesis that apoE4 impairs cognition via modulation of NMDAR-dependent signaling, specifically via a loss of function by comparison of E4FAD mice with 5xFAD/APOE-KO mice, E3FAD and E2FAD mice. RESULTS: Using female E2FAD, E3FAD, E4FAD and 5xFAD/APOE-KO mice aged 2-, 4-, and 6-months, the Y-maze and Morris water maze behavioral tests were combined with synaptic protein levels as markers of synaptic viability. The results demonstrate a greater age-induced deficit in cognition and reduction in PSD95, drebrin and NMDAR subunits in the E4FAD and 5xFAD/APOE-KO mice compared with E2FAD and E3FAD mice, consistent with an apoE4 loss of function. Interestingly, for NMDAR-mediated signaling, the levels of p-CaMK-II followed this same apoE-specific pattern as cognition, while the levels of p-CREB and BDNF demonstrate an apoE4 toxic gain of function: E2FAD > E3FAD > 5xFAD/APOE-KO > E4FAD. CONCLUSION: These findings suggest that compared with E2FAD and E3FAD, E4FAD and 5xFAD/APOE-KO mice exhibit enhanced age-induced reductions in cognition and key synaptic proteins via down-regulation of an NMDAR signaling pathway, consistent with an apoE4 loss of function. However, levels of p-CREB and BDNF, signaling factors common to multiple pathways, suggest a gain of toxic function. Publications in this field present contradictory results as to whether APOE4 imparts a loss or gain of function. As with the results reported herein, the overall effect of APOE4 on a given CNS-specific measure will be the product of multiple overlapping mechanisms. Thus, caution remains critical in determining whether APOE gene inactivation or therapies that correct the loss of positive function related to apoE4, are the appropriate therapeutic response.


Assuntos
Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Cognição/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/genética , Envelhecimento , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Regulação para Baixo , Memória/fisiologia , Camundongos Knockout , Mutação/genética , Transdução de Sinais/fisiologia
9.
Behav Brain Res ; 258: 8-18, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24140565

RESUMO

Deficits in cognition and performance accompanying age-related neurodegenerative diseases such as Alzheimer's disease (AD) are closely associated with the impairment of synaptic plasticity. Here, using a mouse model of senescence-accelerated P8 (SAMP8), we reported the role of tripchlorolide (T4), an extract of the natural herb Tripterygium wilfordii Hook F, in improving cognitive deficits and promoting the long-term potentiation (LTP) of hippocampal slices via the N-methyl-D-aspartate receptor (NMDAR)-dependent signaling pathway. Our results demonstrated that chronic administration of T4 at low doses (0.25, 1.0, or 4.0 µg/kg per day, injected intraperitoneally for 75 days) significantly improved learning and memory function in aged SAMP8 mice, as indicated by a chain of behavioral tests including the Y-maze and Morris water maze. Additionally, T4 reversed the impaired LTP in hippocampal CA1 regions of SAMP8 mice in a dose-dependent manner. Moreover, it upregulated the levels of phospho-NMDAR1, postsynaptic density-95 (PSD-95), phospho-calcium-calmodulin dependent kinase II (CaMKII), phospho-CREB and brain derived neurotrophic factor (BDNF) in the hippocampus. This indicates that T4 prevents the impairment of NMDAR-mediated synaptic plasticity-related signal molecules. At optimal doses, T4 did not show significant side-effects on blood counts, blood biochemical measures, or survival of the mice. This novel mechanism in reversing age-related synaptic dysfunction and NMDAR functional deficits suggests that T4 can halt the manifestation of a key early-stage event in AD. With the consideration of SAMP8 mice as a model to develop therapeutic interventions for AD, our findings provide new insight into the clinical application of tripchlorolide in AD treatment.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Cognição/efeitos dos fármacos , Diterpenos/farmacologia , Hipocampo/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Fenantrenos/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Envelhecimento/metabolismo , Animais , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/fisiopatologia , Diterpenos/uso terapêutico , Hipocampo/fisiopatologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Plasticidade Neuronal/fisiologia , Fenantrenos/uso terapêutico , Sinapses/fisiologia
10.
Artigo em Chinês | MEDLINE | ID: mdl-23662411

RESUMO

OBJECTIVE: To observe time points of the expressions of basic fibroblast growth factor (bFGF), growth associated protein-43 (GAP-43) and neurogenesis after cerebral ischemia/reperfusion in rats and explore its possible mechanism of neurogenesis. METHODS: Models of middle cerebral artery occlusion (MCAO) were established in SD rats which were divided into 3 d, 7 d, 14 d and 28 d groups (n = 6). The neurological severity was evaluated by neurological severity scores (NSS) and scores of motor test (SMT). Neuronal injury in the boundary zone of the infarction area was evaluated by TUNEL and Nissl staining; The expressions of bFGF and GAP-43 and neurogenesis were evaluated by Western blot and 5-bromodeoxyuridine (Brdu) fluorescence staining, respectively. RESULTS: It showed up neurologic impairment and motor dysfunction after cerebral ischemia/reperfusion in rats at 3 d, the numbers of neuron apoptosis also peaked at 3d, the protein levels of bFGF and GAP-43 were significantly increased in time-dependent manner, peaked at 7 d and then decreased gradually, meanwhile, Brdu and NeuN double fluorescence staining displayed scattered Brdu-and NeuN-positive cells in the boundary zone of the infarction area. CONCLUSION: These results suggest that the upregulation of bFGF and GAP-43 may contribute to the neurogenesis after cerebral ischemia/reperfusion.


Assuntos
Isquemia Encefálica , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteína GAP-43/metabolismo , Neurogênese , Traumatismo por Reperfusão , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia
11.
Neurosci Bull ; 29(1): 121-4, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23361522

RESUMO

During recent years, major advances have been made in neuroscience, i.e., asynchronous release, three-dimensional structural data sets, saliency maps, magnesium in brain research, and new functional roles of long non-coding RNAs. Especially, the development of optogenetic technology provides access to important information about relevant neural circuits by allowing the activation of specific neurons in awake mammals and directly observing the resulting behavior. The Grand Research Plan for Neural Circuits of Emotion and Memory was launched by the National Natural Science Foundation of China. It takes emotion and memory as its main objects, making the best use of cutting-edge technologies from medical science, life science and information science. In this paper, we outline the current status of neural circuit studies in China and the technologies and methodologies being applied, as well as studies related to the impairments of emotion and memory. In this phase, we are making efforts to repair the current deficiencies by making adjustments, mainly involving four aspects of core scientific issues to investigate these circuits at multiple levels. Five research directions have been taken to solve important scientific problems while the Grand Research Plan is implemented. Future research into this area will be multimodal, incorporating a range of methods and sciences into each project. Addressing these issues will ensure a bright future, major discoveries, and a higher level of treatment for all affected by debilitating brain illnesses.


Assuntos
Encéfalo/fisiologia , Emoções/fisiologia , Memória/fisiologia , Vias Neurais/fisiologia , Neurociências/métodos , China , Humanos
12.
Zhonghua Yi Xue Za Zhi ; 92(5): 330-5, 2012 Feb 07.
Artigo em Chinês | MEDLINE | ID: mdl-22490838

RESUMO

OBJECTIVE: To explore whether or not ginsenoside Rg1 can modify the metabolism of amyloid precursor protein (APP) and the generation of amyloid beta (Aß) by nuclear factor-kappa B (NF-κB). METHODS: N2a/APP695 cells, a mutated APP-overexpressing neuronal cell line, was used to mimic the APP metabolism and Aß generation in vitro. The BACE1 mRNA and protein levels were detected by RT-PCR (reverse transcription-polymerase chain reaction) and Western blot respectively. Then the expression levels and subcellular localization of NF-κB were detected by Western blot and confocal laser scanning microscope respectively. RESULTS: The treatment of ginsenoside Rg1 at a dose of 2.5 µmol/L decreased the levels of Aß1-40 and Aß1-42 (13.3 ± 4.3) ng/ml vs (12.0 ± 5.4) ng/ml in N2a/APP695 cells, decreased the protein level of BACE1 (BACE1/ß-actin 0.26 ± 0.05), increased the protein level of NF-κB p65 (p-p65/p65 0.93 ± 0.02) and resulted in the translocation of NF-κB from cytoplasm to nucleus. Quinazoline inhibited the activation of NF-κB with a reduction of p-p65 and p-p65/p65 in N2a/APP695 cells and increased the BACE1 protein level. And the treatment of ginsenoside Rg1 showed similar changes in N2a/APP695 cells when compared with the treatment of quinazoline alone. CONCLUSION: Ginsenoside Rg1 may modify the metabolism of APP by enhancing the nuclear binding of NF-κB to BACE1 promoter and inhibiting the transcription and translation of BACE1.


Assuntos
Ginsenosídeos/farmacologia , NF-kappa B/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular Tumoral , Camundongos , Neuroblastoma , Regiões Promotoras Genéticas
13.
Acta Pharmacol Sin ; 33(4): 438-44, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22407229

RESUMO

AIM: To investigate the effect of ginsenoside Rb1 on voltage-gated calcium currents in cultured rat hippocampal neurons and the modulatory mechanism. METHODS: Cultured hippocampal neurons were prepared from Sprague Dawley rat embryos. Whole-cell configuration of the patch-clamp technique was used to record the voltage-gated calcium currents (VGCCs) from the hippocampal neurons,and the effect of Rb1 was examined. RESULTS: Rb1 (2-100 µmol/L) inhibited VGCCs in a concentration-dependent manner, and the current was mostly recovered upon wash-out. The specific L-type Ca(2+) channel inhibitor nifedipine (10 µmol/L) occluded Rb1-induced inhibition on VGCCs. Neither the selective N-type Ca(2+) channel blocker ω-conotoxin-GVIA (1 µmol/L), nor the selective P/Q-type Ca(2+) channel blocker ω-agatoxin IVA (30 nmol/L) diminished Rb1-sensitive VGCCs. Rb1 induced a leftward shift of the steady-state inactivation curve of I(Ca) to a negative potential without affecting its activation kinetics or reversal potential in the I-V curve. The inhibitory effect of Rb1 was neither abolished by the adenylyl cyclase activator forskolin (10 µmol/L), nor by the PKA inhibitor H-89 (10 µmol/L). CONCLUSION: Ginsenoside Rb1 selectively inhibits the activity of L-type voltage-gated calcium channels, without affecting the N-type or P/Q-type Ca(2+) channels in hippocampal neurons. cAMP-PKA signaling pathway is not involved in this effect.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Ginsenosídeos/farmacologia , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Animais , Células Cultivadas , Neurônios/metabolismo , Panax/química , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
14.
Eur J Pharmacol ; 675(1-3): 15-21, 2012 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-22166376

RESUMO

The level of ß-site APP-cleaving enzyme 1 (BACE1) has been documented to increase in the brains of patients with Alzheimer's disease, which has resulted in elevation of ß-amyloid (Aß) peptides. As a transcription factor binding site of the BACE1 promoter, peroxisome proliferator-activated receptor-γ (PPARγ) response element regulates the activity of the BACE1 promoter activity, indicating that PPARγ may become a potential target for Alzheimer's disease treatment. Recent studies have demonstrated that ginsenoside Rg1 which is an effective component of extracts of ginseng can prevent memory loss and improve cognitive function in a variety of animal models. However, the underlying mechanism remains unclear. In the present study, we found that Rg1 decreased the levels of Aß1₋40 and Aß1₋42 secreted in N2a-APP695 cells. The expression levels of both BACE1 mRNA and protein as well as ß-CTFs, a cleavaged C-terminal fragment of APP by BACE1, were reduced in cells treated with Rg1. Moreover, Rg1 treatment led to a translocation of PPARγ from cytoplasm to nuclear. Intriguingly, Rg1, like pioglitazone (a PPARγ agonist), suppressed BACE1 activity in N2a-APP695 cells, while its effect on BACE1 activity was attenuated by GW9662 (a PPARγ antagonist). These results indicate that Rg1 may be a PPARγ agonist to enhance the binding of nuclear PPARγ to the BACE1 promoter, which may in turn inhibit the transcription and translation of BACE1, suppress the activity of BACE1, and ultimately attenuate Aß generation. Therefore, ginsenoside Rg1 may serve as a promising agent in modulating Aß-related pathology in Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Ginsenosídeos/farmacologia , Neurônios/efeitos dos fármacos , PPAR gama/metabolismo , Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/genética , Linhagem Celular , Fármacos do Sistema Nervoso Central/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo
15.
Yao Xue Xue Bao ; 46(9): 1065-71, 2011 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-22121776

RESUMO

This study is to observe the effect of ilexonin A (IA) on the expression of basic fibroblast growth factor (bFGF) and growth associated protein-43 (GAP-43), and neurogenesis after cerebral ischemia-reperfusion in rats and explore its possible mechanism of protecting neuronal injury. Models of middle cerebral artery occlusion (MCAO) were established in SD rats. Before and after two hours ischemia-reperfusion, IA (20 and 40 mg x kg(-1)) was injected immediately and on 3, 7, 14, and 28 d once a day. The neurological severity was evaluated by neurological severity scores (NSS); neuronal injury in the boundary zone of the infarction area was evaluated by TUNEL and Niss1 staining. The expressions of bFGF and GAP-43 and neurogenesis were evaluated by Western blotting and 5-bromodeoxyuridine (Brdu) fluorescence staining, respectively. After treatment with IA, the NSS of treatment groups were lower than that of the models (3 and 7 d). The number of TUNEL positive neurons decreased and Nissl positive neurons increased at the same time (3 d). The expressions of bFGF and GAP-43 increased significantly in the boundary zone of the infarction area when compared to model group. Moreover, IA markedly enhanced the neurogenesis in the brain after ischemia-reperfusion, which revealed an increase of Brdu/NeuN positive cells in the boundary zone of the infarction area. The possible mechanism of protecting neuronal injury of IA may be related to inhibition on neuronal apoptosis, upregulation of bFGF and GAP-43, and neurogenesis in boundary zone of infarction after cerebral ischemia-reperfusion.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteína GAP-43/metabolismo , Neurogênese/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Isquemia Encefálica/etiologia , Bromodesoxiuridina/metabolismo , Infarto da Artéria Cerebral Média/complicações , Masculino , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Compostos Orgânicos/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia
16.
Mol Neurodegener ; 6: 45, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21718498

RESUMO

BACKGROUND: Reactive microglia are associated with ß-amyloid (Aß) deposit and clearance in Alzhiemer's Disease (AD). Paradoxically, entocranial resident microglia fail to trigger an effective phagocytic response to clear Aß deposits although they mainly exist in an "activated" state. Oligomeric Aß (oAß), a recent target in the pathogenesis of AD, can induce more potent neurotoxicity when compared with fibrillar Aß (fAß). However, the role of the different Aß forms in microglial phagocytosis, induction of inflammation and oxidation, and subsequent regulation of phagocytic receptor system, remain unclear. RESULTS: We demonstrated that Aß(1-42) fibrils, not Aß(1-42) oligomers, increased the microglial phagocytosis. Intriguingly, the pretreatment of microglia with oAß(1-42) not only attenuated fAß(1-42)-triggered classical phagocytic response to fluorescent microspheres but also significantly inhibited phagocytosis of fluorescent labeled fAß(1-42). Compared with the fAß(1-42) treatment, the oAß(1-42) treatment resulted in a rapid and transient increase in interleukin 1ß (IL-1ß) level and produced higher levels of tumor necrosis factor-α (TNF-α), nitric oxide (NO), prostaglandin E2 (PGE2) and intracellular superoxide anion (SOA). The further results demonstrated that microglial phagocytosis was negatively correlated with inflammatory mediators in this process and that the capacity of phagocytosis in fAß(1-42)-induced microglia was decreased by IL-1ß, lippolysaccharide (LPS) and tert-butyl hydroperoxide (t-BHP). The decreased phagocytosis could be relieved by pyrrolidone dithiocarbamate (PDTC), a nuclear factor-κB (NF-κB) inhibitor, and N-acetyl-L-cysteine (NAC), a free radical scavenger. These results suggest that the oAß-impaired phagocytosis is mediated through inflammation and oxidative stress-mediated mechanism in microglial cells. Furthermore, oAß(1-42) stimulation reduced the mRNA expression of CD36, integrin ß1 (Itgb1), and Ig receptor FcγRIII, and significantly increased that of formyl peptide receptor 2 (FPR2) and scavenger receptor class B1 (SRB1), compared with the basal level. Interestingly, the pre-stimulation with oAß(1-42) or the inflammatory and oxidative milieu (IL-1ß, LPS or t-BHP) significantly downregulated the fAß(1-42)-induced mRNA over-expression of CD36, CD47 and Itgb1 receptors in microglial cells. CONCLUSION: These results imply that Aß oligomers induce a potent inflammatory response and subsequently disturb microglial phagocytosis and clearance of Aß fibrils, thereby contributing to an initial neurodegenerative characteristic of AD. Antiinflammatory and antioxidative therapies may indeed prove beneficial to delay the progression of AD.

17.
Yao Xue Xue Bao ; 45(7): 853-9, 2010 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-20931782

RESUMO

This study is to explore whether the Wnt/beta-catenin signaling pathway is involved in the process of tripchlorolide (T4) protecting against oligomeric Abeta(1-42)-induced neuronal apoptosis. Primary cultured cortical neurons were used for the experiments on day 6 or 7. The oligomeric Abeta(1-42) (5 micromol x L(-1) for 24 h) was applied to induce neuronal apoptosis. Prior to treatment with Abeta(1-42) for 24 h, the cultured neurons were pre-incubated with T4 (2.5, 10, and 40 nmol x L(-1)), Wnt3a (Wnt signaling agonists) and Dkk1 (inhibitors) for indicated time. Then the cell viability, neuronal apoptosis, and protein levels of Wnt, glycogen synthase kinase 3beta (GSK3beta), beta-catenin and phospho-beta-catenin were measured by MTT assay, TUNEL staining and Western blotting, respectively. The result demonstrated that oligomeric Abeta(1-42) induced apoptotic neuronal cell death in a time- and dose-dependent manner. Pretreatment with T4 significantly increased the neuronal cell survival and attenuated neuronal apoptosis. Moreover, oligomeric Abeta(1-42)-induced phosphorylation of beta-catenin and GSK3beta was markedly inhibited by T4. Additionally, T4 stabilized cytoplasmic beta-catenin. These results indicate that tripchlorolide protects against the neurotoxicity of Abeta by regulating Wnt/beta-catenin signaling pathway. This may provide insight into the clinical application of tripchlorolide to Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Apoptose/efeitos dos fármacos , Diterpenos/farmacologia , Fragmentos de Peptídeos/toxicidade , Fenantrenos/farmacologia , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Diterpenos/isolamento & purificação , Feminino , Feto , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/isolamento & purificação , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fenantrenos/isolamento & purificação , Fosforilação , Plantas Medicinais/química , Gravidez , Ratos , Ratos Sprague-Dawley , Tripterygium/química
18.
J Alzheimers Dis ; 19(3): 977-89, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20157253

RESUMO

It is well established that the presence of soluble amyloid-beta protein (Abeta) correlates with the severity of dementia in Alzheimer's disease (AD). Several lines of evidence indicate that cyclic AMP responsive element binding protein (CREB) and protein kinase A (PKA) are involved in soluble Abeta-trigged disruption of synaptic plasticity in early AD. Previously we demonstrated the beneficial effects of ginsenoside Rg1 on Abeta-induced neuronal insult. Therefore, in the present study, we examined the effects of long-term consumption of Rg1 on the cerebral Abeta content and PKA/CREB signaling molecules, as well as cognitive performance in senescence-accelerated mouse prone 8 (SAMP8). Notably, a significant dose-dependent reduction of soluble Abeta(1-40) was shown in the hippocampus of SAMP8 mice after administration with ginsenoside Rg1 for 3 months. Furthermore, Rg1 treatment resulted in a significant decrease of hippocampal PKA RIIalpha level (isoform IIalpha of the regulatory subunit of PKA). In contrast, phospho-CREB and brain derived neurotrophic factor (BDNF) levels were dramatically increased in the hippocampus of SAMP8 treated with Rg1. Additionally, administration of ginsenoside Rg1 consequently improved learning and memory outcomes in SAMP8 mice. These data suggest that long-term consumption of ginsenoside Rg1 may delay cognitive decline, associated with significant effects on Abeta generation, PKA/CREB activity, as well as BDNF content in the brain. These data provide further support for the therapeutic or intervention potency of ginsenoside Rg1 in the early stage of AD.


Assuntos
Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Cognição/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ginsenosídeos/farmacologia , Envelhecimento/efeitos dos fármacos , Animais , Fármacos do Sistema Nervoso Central/administração & dosagem , Ginsenosídeos/administração & dosagem , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos
19.
Glia ; 57(11): 1227-38, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19170180

RESUMO

Recent research has focused on soluble oligomeric assemblies of beta-amyloid peptides (Abeta) as the proximate cause of neuroinflammation, synaptic loss, and the eventual dementia associated with Alzheimer's disease (AD). In this study, tripchlorolide (T4), an extract of Tripterygium wilfordii Hook. F (TWHF), was studied as a novel agent to suppress neuroinflammatory process in microglial cells and to protect neuronal cells against microglia-mediated oligomeric Abeta toxicity. T4 significantly attenuated oligomeric Abeta(1-42)-induced release of inflammatory productions such as tumor necrosis factor-alpha, interleukin-1beta, nitric oxide (NO), and prostaglandin E2. It also downregulated the protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in microglial cells. Further molecular mechanism study demonstrated that T4 inhibited the nuclear translocation of nuclear factor-kappaB (NF-kappaB) without affecting I-kappaBalpha phosphorylation. It repressed Abeta-induced JNK phosphorylation but not ERK or p38 MAPK. The inhibition of NF-kappaB and JNK by T4 is correlated with the suppression of inflammatory mediators in Abeta-stimulated microglial cells. These results suggest that T4 protects neuronal cells by blocking inflammatory responses of microglial cells to oligomeric Abeta(1-42) and that T4 acts on the signaling of NF-kappaB and JNK, which are involved in the modulation of inflammatory response. Therefore, T4 may be an effective agent in modulating neuroinflammatory process in AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Diterpenos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microglia/efeitos dos fármacos , Subunidade p50 de NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/toxicidade , Fenantrenos/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Linhagem Celular , Células Cultivadas , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/fisiologia , Neuroimunomodulação/efeitos dos fármacos , Neurônios/fisiologia
20.
Biochem Pharmacol ; 76(3): 362-72, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18602088

RESUMO

A large body of evidence has suggested a strong association between neuroinflammation and the pathogenesis of many neurodegenerative diseases. Therefore, it is a good target for therapeutic treatment. So far, studies have proven anti-inflammatory herbal medicine and its constituents to be effective in slowing down the neurodegenerative process. The present study tested tripchlorolide, an extract of Tripterygium wilfordii Hook F (TWHF), as a novel agent to suppress inflammatory process in microglia. It showed this novel agent to be cytotoxic at a dose of 20-40 nM to primary microglia and BV-2 microglial cells but not to primary cortical neurons and Neuro-2A cells in vitro. Moreover, tripchlorolide protected primary cortical neurons and Neuro-2A cells from neuroinflammatory toxicity induced by the conditioned media from lipopolysaccharide (LPS)-stimulated microglia, which resulted in a significant decrease in their cell survival. The changes of the inflammatory mediators in this process were further investigated. In the LPS-stimulated microglia, the production of tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), nitric oxide (NO), prostaglandin E(2) (PGE(2)), and intracellular superoxide anion (SOA) was markedly attenuated by tripchlorolide at a dose of 1.25-10 nM in a dose-dependent manner. Furthermore, the production of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) was also significantly inhibited by tripchlorolide in both mRNA and protein levels. These results suggest that tripchlorolide can protect neuronal cells via a mechanism involving inhibition of inflammatory responses of microglia to pathological stimulations. Therefore, it is potentially a highly effective therapeutic agent in treating neuroninflammatory diseases.


Assuntos
Diterpenos/farmacologia , Lipopolissacarídeos/toxicidade , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas , Fenantrenos/farmacologia , Animais , Western Blotting , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados , Ciclo-Oxigenase 2/biossíntese , Dinoprostona/imunologia , Diterpenos/isolamento & purificação , Interleucina-1beta/imunologia , Camundongos , Microglia/imunologia , Neurônios/imunologia , Fármacos Neuroprotetores/isolamento & purificação , Síndromes Neurotóxicas/imunologia , Síndromes Neurotóxicas/prevenção & controle , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Fenantrenos/isolamento & purificação , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...