Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Acta Biochim Biophys Sin (Shanghai) ; 50(5): 447-455, 2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29617703

RESUMO

Epidemiologic studies have demonstrated that women account for two-thirds of Alzheimer's disease (AD) cases, for which the decline in circulating gonadal hormone is considered to be one of the major risk factors. In addition, ovarian hormone deficiency may affect ß-amyloid (Aß) deposition, which has a close relationship with autophagic flux. In this study, we investigated the impact of short-term or long-term ovarian hormone deprivation on two mouse models, the non-transgenic (wild-type) and the APP/PS1 double-transgenic AD (2×TgAD) model. Autophagy-related proteins (Beclin1, LC3, and p62) and lysosome-related proteins were detected to evaluate Aß deposition and autophagy. Our results showed that in the group with short-term depletion of ovarian hormones by ovariectomy (ovx), Beclin1, Cathepsin B (Cath-B), and LAMP1 levels were significantly decreased, while the levels of LC3-II and p62 were increased. In the long-term group, however, there was a sharp decline in Beclin1, LC3-II, Cath-B, and LAMP1 expression but not in p62 expression which is increased. It is worthwhile to note that the occurrence of neuritic plaque-induced ovarian hormone loss increased both the Aß level and neuritic plaque deposition in 2×TgAD mice. Therefore, autophagy may play an important role in the pathogenesis of female AD, which is also expected to help post-menopausal patients with AD.


Assuntos
Autofagia , Encéfalo/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Ovariectomia , Placa Amiloide/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Proteínas Relacionadas à Autofagia/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Transgênicos , Ovário/metabolismo , Ovário/cirurgia , Presenilina-1/genética , Presenilina-1/metabolismo
2.
Acta Biochim Biophys Sin (Shanghai) ; 47(4): 258-66, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25736404

RESUMO

The γ-secretase complex catalyzes the final cleavage step of amyloid ß-protein precursor (APP) to generate amyloid ß (Aß) peptide, a pathogenic component of senile plaques in the brain of Alzheimer's disease (AD) patients. Recent studies have shown that presenilin enhancer-2 (Pen-2), presenilin (PS, including PS1 and PS2), nicastrin, and anterior pharynx-defective 1 are essential components of the γ-secretase. The structure and function of Pen-2 in vitro have been well defined. However, little is known about the neuroanatomical distribution and expression of Pen-2 in the central nervous system (CNS) of AD model mice. We report here, using various methods such as immunohistochemical staining and immunoblotting, that Pen-2 is widely expressed at specific neuronal cells of major areas in AD model mice, including the olfactory bulb, basal forebrain, striatum, cortex, hippocampus, amygdala, thalamus, hypothalamus, cerebellum, brainstem, and spinal cord. It is co-expressed with PS1 in specific neuronal cells in mouse brain. Pen-2 is distributed much more extensively than extracellular amyloid deposits, suggesting the importance of other factors in localized amyloid deposition. Pen-2 is localized predominantly in cell membrane and cytoplasma in adult AD mice, but only distributed at cell membrane in controls. At the early stages of postnatal development, the expression level of Pen-2 is relatively high in CNS, but declines, gradually in adult mice. The present study provides an anatomical basis for Pen-2 as a key component of γ-secretase complex in the brain of developing and adult mice, and Pen-2 might be closely related to Aß burden in aging nervous system.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Sistema Nervoso Central/metabolismo , Presenilina-1/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Western Blotting , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Masculino , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Mutação , Presenilina-1/genética , Fatores de Tempo
3.
J Ethnopharmacol ; 335: 118712, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39173724

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Yajieshaba (YJSB), approved by the Yunnan Provincial Food and Drug Administration in 2008, are known for their anti-inflammatory, antiviral, and pro-apoptotic properties, effectively treating Hepatic fibrosis (HF). However, its mechanism of action remains unclear. AIM OF THE STUDY: The objective of this investigation is to explore how YJSB influences the TGF-ß1/Smad signaling pathway as a strategy for reducing HF. METHODS: The establishment of a HF model in mice involved ligation of the common bile duct, followed by administration of YJSB. Body and liver weights were measured, and the liver index calculated. Serum levels of ALT, AST, ALP, TBA, and TBIL were assessed using colorimetric methods. Additionally, liver homogenates were analyzed for PIIINP, Col-IV, LN, HA, and Hyp, as well as TGF-ß1 activity, using ELISA. Histological analyses of liver sections, stained with H&E, Ag, and Masson's trichrome, were performed to examine inflammation and the accumulation of collagen and reticular fibers. These studies aimed to elucidate the pharmacodynamic effects of YJSB on HF in mice with bile duct obstruction. The target pathways of YJSB were preliminarily identified through immunofluorescence detection of TGF-ß1, P-Smad2L, P-Smad2C, P-Smad3L, P-Smad3C, and Smad4 proteins. In vitro experiments included the induction of hepatic stellate cell (HSC-T6) activation by H2O2. A cell injury model was established for HSC-T6, and the CCK-8 assay was used to determine the optimal YJSB concentration and treatment duration. After pirfenidone (PFD) administration, which inhibits the TGF-ß1/Smad pathway, the effects of YJSB on HSC-T6 cell proliferation were observed. ELISA assays quantified Col-III, α-SMA, and Col-I in cell lysates to assess YJSB's impact on collagen synthesis in HSC-T6 cells. Western blot analysis was performed to assess the protein levels within the TGF-ß1/Smad signaling cascade. RESULTS: In the HF mouse model, administration of YJSB notably augmented the body weight and reduced the liver index. Concurrently, there was an elevation in serum concentrations of ALP, AST, ALT, TBA, and TBIL. Similarly, in the liver homogenates of HF mice, increases were observed in the levels of HA, PIIINP, Col-IV, LN, Hyp, and TGF-ß1. Histological assessments using H&E, Ag, and Masson stains indicated a substantial diminution in liver tissue damage. Through immunofluorescence analysis, it was discerned that YJSB modulated the expression of TGF-ß1, P-Smad2L, P-Smad2C, and P-Smad3L downwards, while elevating P-Smad3C and Smad4 protein expressions. Additional investigations revealed a significant reduction in α-SMA, Col-I, and Col-III levels in cell culture fluids, suggesting a decrease in collagen synthesis and a protective role against cellular damage. Western blot analyses demonstrated that the TGF-ß1/Smad pathway inhibitor, PFD, acted in synergy with YJSB, enhancing its regulatory effects on this pathway, decreasing levels of TGF-ß1, P-Smad2L, P-Smad2C, P-Smad3L, and promoting the expression of P-Smad3C. CONCLUSIONS: YJSB demonstrates a pharmacodynamic effect against HF, enhancing liver functionality and effectively mitigating the damage associated with bile duct obstruction. The proposed action mechanism of YJSB involves modulation of the TGF-ß1/Smad signaling pathway. Research indicates that YJSB might play a role in suppressing the movement, programmed cell death, and activation of HSC-T6, potentially decelerating the advancement of hepatic fibrosis.


Assuntos
Colestase , Células Estreladas do Fígado , Peróxido de Hidrogênio , Cirrose Hepática , Transdução de Sinais , Fator de Crescimento Transformador beta1 , Animais , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Fator de Crescimento Transformador beta1/metabolismo , Camundongos , Masculino , Peróxido de Hidrogênio/metabolismo , Colestase/metabolismo , Colestase/patologia , Colestase/tratamento farmacológico , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Transdução de Sinais/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/metabolismo , Proteínas Smad/metabolismo , Camundongos Endogâmicos C57BL , Proteína Smad2/metabolismo
4.
Front Pharmacol ; 15: 1447241, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39170709

RESUMO

Introduction: Gouty nephropathy (GN) arises from factors like excessive purine intake, metabolic disorders or abnormal synthesis, and uric acid hypersaturation in the blood, leading to urate crystal deposition in kidney tissue. DaiTongXiao (DTX) is a remedy used by the Dai people of China. It shows efficacy in lowering uric acid levels and exhibits anti-inflammatory and kidney-protective properties. Methods: A GN rat model was induced using adenine and potassium oxonate. Following DTX administration, various parameters were assessed in urine, serum, and kidney tissue. Western blot analysis evaluated TLR4/MyD88/NF-κB signaling proteins, while immunofluorescence examined NF-κB nuclear expression. Results: DTX treatment improved kidney morphology, increased body weight, and kidney index and enhanced urinary levels of blood urea nitrogen (Bun), 24-h urinary protein, uric acid (UA), and allantoin in GN rats, reducing UA, Bun, creatinine (Cre), cystatin C (CysC), serum amyloid A (SAA), α1-microglobulin (MG), and ß2-MG in serum analysis. Renal tissue assessments showed decreased xanthine oxidase (XOD), hydroxyproline (Hyp), α-smooth muscle actin (α-SMA), and collage type Ⅳ (COL-Ⅳ). Kidney damage severity was notably reduced. DTX lowered serum inflammatory factors like interleukin (IL) -18, tumor necrosis factor-α (TNF-α), C-reactive protein (CRP), transforming growth factor-ß1 (TGF-ß1), and IL-1ß in the rat serum, reducing chemokine monocyte chemoattractant protein-1 (MCP-1) and adhesion factor vascular cell adhesion molecule-1(VCAM-1). Western blotting demonstrated the downregulation of TLR4/MyD88/NF-κB pathway proteins, and immunofluorescence revealed reduced NF-κB expression in renal tissue. Discussion: DTX exhibits significant anti-GN effects by modulating TLR4/MyD88/ NF-κB pathway protein expression, reducing inflammatory factor release, and inhibiting GN progression.

5.
J Ethnopharmacol ; 335: 118611, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39053712

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Allergic rhinitis (AR) stands as a non-infectious inflammatory condition affecting the nasal mucosa, marked by bouts of sneezing, nasal itching, and congestion. This ailment afflicts individuals across all age groups and poses challenges for effective treatment due to its chronic nature. Cangerzisan (CEZS), documented in the Jishengfang compendium, represents a traditional Chinese medicinal formula long utilized for AR management. AIM OF THE STUDY: Investigating mechanism beneath therapeutic effect of CEZS in alleviating AR. MATERIALS AND METHODS: The main active components in CEZS were determined by High Performance Liquid Chromatography (HPLC).The active constituents of CEZS and their corresponding targets were identified through an exhaustive screening process employing TCMSP database. To identify targets relevant to AR, GeneCards, OMIM, and DisGeNET databases were thoroughly applied. Protein-protein interaction (PPI) network was assembled utilizing STRING platform. Potential signaling pathways influenced by CEZS were delineated through GO and KEGG enrichment analyses. Subsequently, an AR model was induced by administering aluminum hydroxide (Al(OH)3) and ovalbumin (OVA) for affecting basal and local sensitization, respectively, facilitating experimental validation of the principal signaling pathways. RESULTS: There were 61 active constituents identified within CEZS, targeting a pool of 129 entities associated with AR treatment. Pathways analysis of KEGG revealed that CEZS potentially inhibits AR advancement via modulating TLR4 signaling pathway. Animal experiments demonstrated that CEZS effectively alleviated symptom scores in guinea pigs with AR. Moreover, it exhibited notable improvements in serum immune and inflammatory factors levels, as well as reduced inflammatory infiltration within nasal mucosa, including goblet and mast cells. CEZS was found to enhance GATA-3 expression while reducing T-bet expression, thereby modulating the TH1/TH2 immune balance. Additionally, CEZS downregulated HMGB1, TLR4, and p-NF-κB/NF-κB protein expressions within nasal mucosa of guinea pigs. CONCLUSIONS: The therapeutic mechanism of CEZS against AR involves rectifying TH1/TH2 immune imbalance and upregulating inflammatory and immune factors through modulating key proteins expression within TLR4 pathway. This targeted regulation effectively impedes AR progression.


Assuntos
Farmacologia em Rede , Rinite Alérgica , Animais , Rinite Alérgica/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Medicamentos de Ervas Chinesas/química , Mapas de Interação de Proteínas , Transdução de Sinais/efeitos dos fármacos , Ovalbumina , Masculino , Feminino , Receptor 4 Toll-Like/metabolismo , Modelos Animais de Doenças , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/imunologia , Cobaias , Antialérgicos/farmacologia , Citocinas/metabolismo
6.
J Ethnopharmacol ; 334: 118553, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38992401

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Functional dyspepsia (FD) is a prevalent gastrointestinal disorder characterised by high incidence and recurrence rates, posing significant health risks. Erpixing Granules (EPX), approved by the National Food and Drug Administration in 2002, are known for their spleen and stomach invigorating properties, effectively treating FD. However, its mechanism of action remains unclear. AIM OF THE STUDY: This study aims to elucidate EPX's mechanism of treating FD through network pharmacology, and experimental validation using FD animal models. METHODS: In this study, the chemical composition of EPX in positive and negative ion modes was analyzed by UHPLC-Q-TOF MS. The mass spectral data were processed and analyzed using MS-DIAL software to automatically match compound fragment information and identify the known components with the compound database to obtain the active components of EPX. SwissTargetPrediction was used to obtain EPX targets, while FD-related targets were sourced from GeneCards, OMIM and DisGeNET databases. A protein-protein interaction (PPI) network was constructed using the STRING platform, and potential signalling pathways of EPX were determined through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, an FD model was established in rates by administering a 0.1% iodoacetamide sucrose solution, followed by tail clamp stimulation to experimentally validate the network pharmacology findings. RESULTS: Our results revealed 139 effective ingredients in EPX, targeting 60 core FD-related genes. PPI network analysis identified EGFR, CTNNB1 and NFκB1 as core target genes. The KEGG pathway analysis indicated that EPX can modulate FD progression through the PI3K/AKT signalling pathway. Animal experiments demonstrated EPX's capacity to increase body mass, food intake and food utilisation efficiency in FD rats, alongside increased gastric juice secretion, pepsin activity, trypsin activity, cholesterol, bile acid and bilirubin activity. HE examination revealed that EPX improved the inflammatory infiltration of gastric mucosal cells in rats. Furthermore, EPX also promoted gastric emptying and intestinal propulsion in mice. These results suggest that EPX improves spleen and stomach function, enhances the protective effect on the spleen and stomach and promotes food digestion and absorption. Immunofluorescence studies revealed upregulated expression of PI3K, AKT and ANO1 proteins in gastric tissue following EPX administration, while Western blotting indicated increased expression of SCF and C-kit proteins. CONCLUSION: Suggesting EPX's anti-FD effect may involve the regulation of the SCF/C-kit signalling pathway and activation of downstream PI3K/AKT signalling pathway, thereby promoting gastrointestinal motility and improving FD symptoms.


Assuntos
Medicamentos de Ervas Chinesas , Dispepsia , Farmacologia em Rede , Mapas de Interação de Proteínas , Animais , Medicamentos de Ervas Chinesas/farmacologia , Dispepsia/tratamento farmacológico , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Modelos Animais de Doenças , Ratos , Animais não Endogâmicos
7.
Int J Mol Med ; 52(6)2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37830152

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid­ß (Aß) in the brain. The gut/brain axis may serve a role in AD pathogenesis. The present study investigated deposition of Aß in the intestinal epithelium and its potential effects on intestinal barrier function in a transgenic mouse model of AD. To investigate alterations in the structure and functionality of the intestinal mucosal barrier in AD model mice, hematoxylin and eosin staining for Paneth cell count, Alcian blue­periodic acid Schiff staining for goblet cells, immunohistochemistry and immunofluorescence for mucin (MUC)2 and wheat germ agglutin expression, transmission electron microscopy for mucosal ultrastructure, FITC­labeled dextran assay for intestinal permeability, quantitative PCR for goblet cell precursor expression and western blot analysis for tight junction proteins, MUC2 and inflammatory cytokine detection were performed. The results showed that AD model mice exhibited excessive Aß deposition in the intestinal epithelium, which was accompanied by increased intestinal permeability, inflammatory changes and decreased expression of tight junction proteins. These alterations in the intestinal barrier led to an increased proliferation of goblet and Paneth cells and increased mucus synthesis. Dysfunction of gut barrier occurs in AD and may contribute to its etiology. Future therapeutic strategies to reverse AD pathology may involve early manipulation of gut physiology and its microbiota.


Assuntos
Doença de Alzheimer , Mucinas , Camundongos , Animais , Mucinas/metabolismo , Doença de Alzheimer/patologia , Junções Íntimas/metabolismo , Mucosa Intestinal/metabolismo , Camundongos Transgênicos , Permeabilidade , Proteínas de Junções Íntimas/metabolismo
8.
Neural Regen Res ; 16(10): 2078-2085, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33642397

RESUMO

Postmenopausal women with Alzheimer's disease (AD) exhibit dramatically reduced sensitivity to estrogen replacement therapy, which is though to be related to an estrogen receptor (ER)α/ERß ratio imbalance arising from a significantly decreased level of ERs of the brain. The aim of our study was to investigate whether valproic acid (VPA) can enhance the beneficial effects of estrogen on cognitive function through restoration of ERα and ERß expression in the brain. We removed the ovaries of female APP/PS1 mice to simulate the low estrogen levels present in postmenopausal women and then administered VPA (30 mg/kg, intraperitoneal injection, once daily), 17ß-estradiol (E2) (2.4 µg, intraperitoneal injection, once daily), liquiritigenin (LG) (50 µg/kg, intragastric infusion, once daily), VPA + E2, or VPA + LG for 4 successive weeks. Compared with treatment with a single drug, treatment with VPA + E2 or VPA + LG significantly increased the level of glycogen synthase kinase 3ß, increased the expression of estrogen receptor α, reduced the expression of small ubiquitin-like modifiers, and increased the level of estrogen receptor ß. This resulted in enhanced sensitivity to estrogen therapy, reduced amyloid ß aggregation, reduced abnormal phosphorylation of the tau protein, reduced neuronal loss, increased dendritic spine and postsynaptic density, and significantly alleviated memory loss and learning impairment in mice. This study was approved by the Chongqing Medical University Animal Protection and Ethics Committee, China on March 6, 2013.

9.
Int J Mol Med ; 44(1): 313-323, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31115485

RESUMO

The abnormal activation of glycogen synthase kinase 3ß (GSK3ß) is one of the mechanisms involved in the pathogenesis of Alzheimer's disease (AD), which results in amyloid ß­peptide (Aß) plaque overproduction, Tau hyperphosphorylation and neuronal loss. A number of studies have reported that the activation of the mammalian target of rapamycin (mTOR) contributes to the generation and deposition of Aß, as well as to the formation of neurofibrillary tangles (NFTs) by inhibiting autophagy. GSK3ß is also involved in the mTOR signalling pathway. However, whether the inhibition of the activation of mTOR via the regulation of the function of GSK3ß affects the pathology of AD remains unclear. In this study, we intraperitoneally injected amyloid precursor protein (APP)/presenilin­1 (PS1) transgenic mice with rapamycin, a known activator of autophagy that inhibits mTOR. Our results revealed that rapamycin treatment decreased senile plaque deposition by reducing APP generation, and downregulating ß­ and γ­secretase activity. Rapamycin also increased Aß clearance by promoting autophagy and reduced Tau hyperphosphorylation by upregulating the levels of insulin­degrading enzyme. Additionally, rapamycin markedly promoted the proliferation of differentiated SH­SY5Y cells stably transfected with the APPswe gene and prevented neuronal loss in the brains of mice in a model of AD. Moreover, rapamycin induced autophagy and promoted autolysosome degradation. In this study, we provide evidence that rapamycin inhibits GSK3ß activation and elevates ß­catenin expression by improving the Wnt3a expression levels, which facilitates the amelioration of AD pathology. On the whole, our findings indicate that rapamycin inhibits the activation of mTOR and alters the Wnt/GSK3ß/ß­catenin signalling pathway; thus, it may serve as a therapeutic target in the treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Sirolimo/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Glicogênio Sintase Quinase 3 beta/genética , Camundongos , Camundongos Transgênicos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Via de Sinalização Wnt/genética , Proteína Wnt3A/genética , beta Catenina/genética
10.
Front Aging Neurosci ; 11: 62, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30971911

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by the deposition of amyloid-ß (Aß) peptides and neurofibrillary tangles (NFTs) and massive loss of neuronal cells in the brain. Adult hippocampus continuously generates new neurons throughout life to shape brain function and impaired neurogenesis may contribute to a series of cognitive deterioration associated with AD. Enhancing endogenous neurogenesis represents a promising strategy that may ameliorate AD-associated cognitive defects. However, neurogenesis-enhancing approaches and underlying mechanisms are still not well studied. Here, using a mouse model of AD amyloid precursor protein (APP/PS1/Nestin-GFP triple transgenic mice, 3xTgAD), we examined the effects of 4 weeks of valproic acid (VPA) treatment on hippocampal neurogenesis in 2- and 6-month-old mice. VPA treatment promoted cell proliferation and increased the density of immature neurons in the dentate gyrus (DG) of the hippocampus of 3xTgAD mice. Consistent with enhanced neurogenesis, behavioral and morphological analysis showed that VPA treatment improved the learning and memory ability of 3xTgAD mice. Mechanistically, VPA treatment increased ß-catenin levels, accumulated the inactive form of glycogen synthase kinase-3ß (GSK-3ß), and induced the expression of NeuroD1, a Wnt target gene involved in neurogenesis, suggesting the activation of the Wnt signaling pathway in the hippocampus of 3xTgAD mice. This study indicates that VPA stimulates neurogenesis in the adult hippocampus of AD mice model through the Wnt pathway, highlighting VPA as a potential therapeutic for treating AD and related diseases.

11.
Exp Neurol ; 283(Pt A): 353-64, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27421879

RESUMO

Alzheimer's disease (AD) is the most common type of dementia frequently responsible for cognitive decline in the elderly. The etiology and molecular mechanism of AD pathogenesis remain inconclusive. Aging and vascular factors are important independent causes and contributors to sporadic AD. Clinical imaging studies showed that cerebral blood flow decreases before cognitive impairment in patients with AD. To investigate the effect of chronic cerebral hypoperfusion (CCH) on cognitive impairment and morphological features, we developed a new manner of CCH mouse model by narrowing bilateral common carotid arteries. Mice started to manifest spatial memory deficits 1month after the surgery and exhibited behavioral changes in a time-dependent manner. Mice also presented memory deficits accompanied with morphological changes at the neuronal and synaptic levels. CCH damaged the normal neuronal morphology and significantly reduced the expression level of PSD95. CCH activated astrocytes, increased the co-expression of GFAP and AQP4, and destroyed the blood-brain barrier (BBB). Furthermore, CCH facilitated intracellular and extracellular Aß deposition by up-regulating γ-secretase and ß-secretase levels. Our results showed good reproducibility of post-CCH pathological processes, which are characterized by neuronal apoptosis, axonal abnormalities, glial activation, BBB damage, amyloid deposition, and cognitive dysfunction; these processes may be used to decipher the complex interplay and pathological process between CCH and AD. This study provides laboratory evidence for the prevention and treatment of cognitive malfunction and AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Transtornos da Memória/etiologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Aquaporina 4/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Ciclina D1/metabolismo , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Ensaio de Imunoadsorção Enzimática , Proteína GAP-43/metabolismo , Guanilato Quinases/metabolismo , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Proteínas do Tecido Nervoso/metabolismo , Sinapses/metabolismo , Sinapses/patologia , Sinapses/ultraestrutura , Fatores de Tempo , Tubulina (Proteína)/metabolismo
12.
CNS Neurosci Ther ; 21(11): 887-97, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26385876

RESUMO

AIM: Tau hyperphosphorylation and amyloid ß-peptide overproduction, caused by altered localization or abnormal activation of glycogen synthase kinase-3ß (GSK-3ß), is a pathogenic mechanism in Alzheimer's disease (AD). Valproic acid (VPA) attenuates senile plaques and neuronal loss. Here, we confirmed that VPA treatment improved spatial memory in amyloid precursor protein (APP)/presenilin 1 (PS 1) double-transgenic mice and investigated the effect of VPA on synaptic structure and neurite outgrowth. METHODS: We used ultrastructural analysis, immunocytochemistry, immunofluorescence staining, and Western blot analysis to assess the effect of VPA treatment in mice. RESULTS: VPA treatment thickened the postsynaptic density, increased the number of presynaptic vesicles, and upregulated the expression of synaptic markers PSD-95 and GAP43. VPA increased neurite length of hippocampal neurons in vivo and in vitro. In VPA-treated AD mouse brain, inactivated GSK-3ß (pSer9-GSK-3ß) was markedly increased, while hyperphosphorylation of tau at Ser396 and Ser262 was decreased; total tau levels remained similar. VPA treatment notably improved pSer133-cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) levels, which are associated with synaptic function and neurite outgrowth. CONCLUSION: VPA improves behavioral deficits in AD, modifies synaptic structure, and accelerates neurite outgrowth, by inhibiting the activity of GSK-3ß, decreasing hyperphosphorylated tau, enhancing CREB and BDNF expression.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Inibidores Enzimáticos/uso terapêutico , Quinase 3 da Glicogênio Sintase/metabolismo , Neuritos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Ácido Valproico/uso terapêutico , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/patologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neuritos/patologia , Neuritos/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/patologia , Presenilina-1/genética , Transdução de Sinais/genética , Sinapses/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Ácido Valproico/farmacologia , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa