Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Glia ; 63(12): 2208-19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26200696

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Glutatión/metabolismo , Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/metabolismo , Colesterol/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Oxidación-Reducción , Placa Amiloide/metabolismo
2.
Acta Pharmacol Sin ; 33(4): 438-44, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22407229

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Ginsenósidos/farmacología , Hipocampo/citología , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Neuronas/metabolismo , Panax/química , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
3.
Zhonghua Yi Xue Za Zhi ; 92(5): 330-5, 2012 Feb 07.
Artículo en Zh | MEDLINE | ID: mdl-22490838

RESUMEN

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.


Asunto(s)
Ginsenósidos/farmacología , FN-kappa B/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular Tumoral , Ratones , Neuroblastoma , Regiones Promotoras Genéticas
4.
Yao Xue Xue Bao ; 46(9): 1065-71, 2011 Sep.
Artículo en Zh | MEDLINE | ID: mdl-22121776

RESUMEN

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.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteína GAP-43/metabolismo , Neurogénesis/efectos de los fármacos , Daño por Reperfusión/metabolismo , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/etiología , Bromodesoxiuridina/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Compuestos Orgánicos/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/etiología
5.
Yao Xue Xue Bao ; 45(7): 853-9, 2010 Jul.
Artículo en Zh | MEDLINE | ID: mdl-20931782

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Diterpenos/farmacología , Fragmentos de Péptidos/toxicidad , Fenantrenos/farmacología , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Diterpenos/aislamiento & purificación , Femenino , Feto , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fenantrenos/aislamiento & purificación , Fosforilación , Plantas Medicinales/química , Embarazo , Ratas , Ratas Sprague-Dawley , Tripterygium/química
6.
Glia ; 57(11): 1227-38, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19170180

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Diterpenos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Subunidad p50 de NF-kappa B/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Fenantrenos/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Línea Celular , Células Cultivadas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Microglía/fisiología , Neuroinmunomodulación/efectos de los fármacos , Neuronas/fisiología
8.
Mol Neurodegener ; 11(1): 51, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27406263

RESUMEN

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.


Asunto(s)
Apolipoproteína E4/genética , Demencia/metabolismo , Memoria/fisiología , Neuronas/metabolismo , Aprendizaje Espacial/fisiología , Estrés Psicológico/fisiopatología , Envejecimiento , Animales , Apolipoproteína E4/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteína Reelina
9.
Mol Neurobiol ; 53(9): 6397-6406, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26582466

RESUMEN

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.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Diterpenos/farmacología , PPAR gamma/metabolismo , Fenantrenos/farmacología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diterpenos/química , Humanos , Ratones , Ratones Transgénicos , Fenantrenos/química , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos
10.
Chin Med J (Engl) ; 129(15): 1835-44, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-27453234

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Trastornos del Conocimiento/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Células Cultivadas , Senescencia Celular/genética , Senescencia Celular/fisiología , Cognición/fisiología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Chin Med J (Engl) ; 129(23): 2845-2852, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27901000

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Neuronas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Caspasa 12/metabolismo , Chaperón BiP del Retículo Endoplásmico , Lóbulo Frontal/metabolismo , Proteínas de Choque Térmico/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Factor de Transcripción CHOP/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Respuesta de Proteína Desplegada/fisiología
12.
Sheng Li Xue Bao ; 57(2): 154-60, 2005 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-15830099

RESUMEN

The present study was aimed to investigate the effects of ginsenoside Rb1 on okadaic acid (OA)-induced Tau hyperphosphorylation in hippocampal neurons of Sparague-Dawley rat and to explore its possible mechanism. Animals were randomly divided into four groups. Group 1 received dimethysulphoxide (DMSO) injection (vehicle group), group 2 only received OA injection (OA group), group 3 was pretreated with Rb1 and then received OA injection (Rb1 pretreatment group), and the group 4 was an intact control group. The animals in group 3 were injected intraperitoneally with various doses of Rb1 at 5, 10, and 20 mg/kg (once a day for 14 d). On the thirteen day of pretreatment, animals in Rb1 pretreatment group as well as animals in OA group received a bolus injection of 0.483 microg of OA (1.5 microl of solution in DMSO) at right dorsal aspect of hippocampus to induce Tau hyperphosphrylation. The brains were harvested one day after the last treatment. In all groups, the morphology of neurofibrils, phosphorylation of Tau protein, and the activity of phosphatase 2A (PP2A) were investigated. In OA group, the Bielschowski's assay revealed darkened and uneven neurofibrils staining in the hippocampus. The immunohistochemistry results showed a significant increase in Thr(231) phosphorylation of Tau protein in OA group relative to the control group (P<0.01). OA injection also markedly decreased PP2A activity (P<0.01). Western blot confirmed Thr(231) phosphorylation of Tau protein and it also detected phosphorylation of Ser(396) of Tau protein. The animals with Rb1 pretreatment displayed even staining of neurofibrils and normal pattern of fiber organization. Rb1 pretreatment also attenuated Thr(231) and Ser(396) hyperphosphorylations of Tau protein, and restored PP2A activity compared to the OA group (P<0.01). These results indicate that OA-induced hyperphosphorylation of Tau protein in rat hippocampal neurons can be attenuated by the pretreatment of ginsenoside Rb1. These data also implicate that Rb1 has potential neuroprotective effects on Tau-related neuropathology.


Asunto(s)
Ginsenósidos/farmacología , Hipocampo/citología , Neuronas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Masculino , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Ácido Ocadaico , Fosforilación/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
13.
Yao Xue Xue Bao ; 40(3): 225-30, 2005 Mar.
Artículo en Zh | MEDLINE | ID: mdl-15952593

RESUMEN

AIM: To explore the effect and the possible mechanism of ginsenoside Rb1 on beta-amyloid peptide (beta-AP)(25-35) -induced tau protein hyperphosphorylation in cortical neurons. METHODS: Western blotting and immunocytochemical staining were used to detect tau phosphorylation level, total tau and glycogen synthase kinase-3beta (GSK-3beta) in cortical neurons. RESULTS: After exposure to beta-AP(25-35) (20 micromol x L(-1)) for 12 h, the levels of tau protein phosphorylation in the sites of Ser 396, Ser 199/202, Thr 231 and total tau were raised. Meanwhile, the expression of GSK-3beta also increased. Pretreatment with ginsenoside Rbl or lithium chloride, a specific inhibitor of GSK-3beta, markedly reduced beta-AP(25-35)-induced tau hyperphosphorylation and the expression of GSK-3beta. CONCLUSION: Ginsenoside Rb1 can attenuate beta AP(25-35)-induced tau protein hyperphosphorylation in cortical neurons by inhibiting the expression of GSK-3beta.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Corteza Cerebral/metabolismo , Ginsenósidos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Proteínas tau/metabolismo , Animales , Corteza Cerebral/citología , Femenino , Feto , Ginsenósidos/aislamiento & purificación , Glucógeno Sintasa Quinasa 3 beta , Neuronas/metabolismo , Panax/química , Fosforilación , Plantas Medicinales/química , Ratas , Ratas Sprague-Dawley
14.
Chin Med J (Engl) ; 128(16): 2220-7, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26265617

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Biomarcadores/análisis , Glucosa/metabolismo , Bulbo Olfatorio/metabolismo , Amiloide/análisis , Animales , Animales Modificados Genéticamente , Fluorodesoxiglucosa F18/metabolismo , Ratones , Tomografía de Emisión de Positrones
15.
Mol Neurodegener ; 10: 7, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25871877

RESUMEN

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.


Asunto(s)
Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Cognición/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/genética , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Regulación hacia Abajo , Memoria/fisiología , Ratones Noqueados , Mutación/genética , Transducción de Señal/fisiología
16.
Eur J Pharmacol ; 473(1): 1-7, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12877931

RESUMEN

In Parkinson's disease, neuroprotective therapy to rescue dopamine neurons has been proposed. Ginsenoside Rg1, one of the biologically active ingredients of ginseng, may be a candidate neuroprotective drug. In the present study, the mechanism underlying the neuroprotection provided by ginsenosde Rg1 was studied against apoptosis induced by exogenous dopamine in PC12 cells. Pretreatment with ginsenoside Rg1 markedly reduced the generation of dopamine-induced reactive oxygen species and the release of mitochondrial cytochrome c into the cytosol, and subsequently inhibited the activation of caspase-3. In addition, Rg1 pretreatment also reduced inducible nitric oxide (NO) synthase protein level and NO production. These results suggested that ginsenoside Rg1 may attenuate dopamine-induced apoptotic cell death through suppression of intracellular oxidative stress, and that it may rescue or protect dopamine neurons in Parkinson's disease.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Dopamina/metabolismo , Ginsenósidos/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Western Blotting , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Grupo Citocromo c/efectos de los fármacos , Grupo Citocromo c/metabolismo , Fragmentación del ADN , Dopamina/farmacología , Citometría de Flujo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Células PC12 , Panax/química , Ratas
17.
Yao Xue Xue Bao ; 38(3): 176-80, 2003 Mar.
Artículo en Zh | MEDLINE | ID: mdl-12830711

RESUMEN

AIM: To explore possible signal transmission way through which ginsenoside Rg1 protect cells from MPP(+)-induced apoptosis. METHODS: The apoptosis of SHSY5Y induced by 1-methyl-4-phenylpyridinium (MPP+) was observed by AO-EB staining. Flow cytometry was used to quantitate the reactive oxygen species (ROS). Western Blotting was used to detect the c-jun NH2-terminal kinase (JNK) activity in SHSY5Y cells. Immunocytochemistry staining was used to detect cleaved Caspase-3 positive cells. RESULTS: MPP+ was shown to induce apoptosis in SHSY5Y cells. The percentage of apoptotic SHSY5Y cells induced by MPP+ was obviously lower in those groups pretreated with 10 mumol.L-1 Rg1 or 2.5 mmol.L-1 N-acetylcysyteine (NAC). It showed more ROS in MPP+ groups than in control. JNK activity increased with time within 72 hours in 1 mmol.L-1 MPP+ group. Simultaneously, it showed decrease of ROS, less activity of JNK and lower expression of cleaved Caspase-3 in 10 mumol.L-1 Rg1 and 2.5 mmol.L-1 NAC pretreated groups compared with groups treated with MPP+ only. CONCLUSION: Rg1 protects against MPP(+)-induced apoptosis in SHSY5Y cells and the effect might be attributed to its removal of ROS, inhibition of the activity of JNK and expression of cleaved Caspase-3.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Apoptosis/efectos de los fármacos , Ginsenósidos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fármacos Neuroprotectores/farmacología , 1-Metil-4-fenilpiridinio/antagonistas & inhibidores , Caspasas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Ginsenósidos/aislamiento & purificación , Humanos , MAP Quinasa Quinasa 4 , Neuroblastoma/patología , Panax/química , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
18.
Yao Xue Xue Bao ; 38(6): 401-4, 2003 Jun.
Artículo en Zh | MEDLINE | ID: mdl-14513795

RESUMEN

AIM: To explore whether the oligonucleotide uptake in hematological tumor cells is related to cellular species and proliferation. METHODS: Intracellular mean fluorescence intensity was measured by flow cytometry. RESULTS: After treatment with FITC-labeled G3139 at the concentration of 0.60 mumol.L-1 for 4 h, the G3139 uptake into peripheral blood mononuclear cell and bone marrow mononuclear cell in hematological tumor patients was significantly higher than that in normal control. There was different uptake of G3139 among the malignant hematological tumor cell strains, and the uptake in cells derived from monocyte, B lymphocyte and myeloid cell was much higher than that in cells derived from T lymphocyte. After treatment with all-trans retinoic acid (ATRA), HL60 cell proliferation was markedly inhibited and the uptake of G3139 decreased significantly. CONCLUSION: Hematological tumor cells were capable of taking up oligonucleotide, and the oligonucleotide uptake in hematological tumor cells is related to its cellular species and its activation.


Asunto(s)
Genes bcl-2/genética , Leucemia/metabolismo , Oligonucleótidos Antisentido/metabolismo , Tionucleótidos/metabolismo , Transporte Biológico , División Celular/fisiología , Células HL-60 , Humanos , Leucemia/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Leucocitos Mononucleares/metabolismo , Linfoma no Hodgkin/metabolismo , Linfoma no Hodgkin/patología , Tretinoina/farmacología , Células Tumorales Cultivadas
19.
Yao Xue Xue Bao ; 39(9): 673-6, 2004 Sep.
Artículo en Zh | MEDLINE | ID: mdl-15606011

RESUMEN

AIM: To explore the possible role of p21, cyclin E and cyclin-dependent kinase 2 (CDK2) in the protection of ginsenoside Rg1 against tert-butylhydroperoxide (t-BHP)-induced senescence in WI-38 cells. METHODS: The cellular ultrastructure, cytometric assay and beta-galactosidase (beta-gal) cytochemistry staining were used to evaluate cell senescence. The levels of of p21, cyclin E and CDK2 protein were detected by Western blot. RESULTS: Pretreatment with Rg1 significantly attenuated t-BHP-induced senescence in WI-38 cells. Simultaneously, compared with cells treated with t-BHP alone, Rg1 pretreatment markedly decreased the level of p21 protein and increased the levels of CDK2 and cyclin E. CONCLUSION: p21, cyclin E and CDK2 may be involved in the process of ginsenoside Rg1 protection against t-BHP-induced senescence in WI-38 cells.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Senescencia Celular/efectos de los fármacos , Ciclina E/metabolismo , Ginsenósidos/farmacología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Línea Celular , Quinasa 2 Dependiente de la Ciclina , Fibroblastos/citología , Fibroblastos/metabolismo , Ginsenósidos/aislamiento & purificación , Humanos , Panax/química , Plantas Medicinales/química , terc-Butilhidroperóxido/antagonistas & inhibidores
20.
Behav Brain Res ; 258: 8-18, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24140565

RESUMEN

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.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Cognición/efectos de los fármacos , Diterpenos/farmacología , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Fenantrenos/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/fisiopatología , Diterpenos/uso terapéutico , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Plasticidad Neuronal/fisiología , Fenantrenos/uso terapéutico , Sinapsis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA