Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Mol Cell Neurosci ; 74: 34-41, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27013422

RESUMO

Processing of the amyloid precursor protein (APP) and amyloid beta (Aß) has been for decades in the center of Alzheimer's disease (AD) research. Beside many other variables, lipids, especially cholesterol and its derivatives, are discussed to contribute to AD pathogenesis. Several studies show that cholesterol affects APP metabolism. Also the converse mechanism, the direct influence of Aß on cholesterol metabolism, has been described. To further investigate this crosstalk between cholesterol- and APP metabolism, a high-fat feeding study was conducted with animals overexpressing human APPSL and/or human ApoB-100. The impact of diet and genotype on cerebral cholesterol metabolism and content as well as spatial learning and memory was examined. While behavioral performance was not influenced by this high fat diet (HFD), reduction of cortical free cholesterol levels and mRNA expression patterns under normal diet and HFD conditions in human APPSL overexpressing mice argue for an important role of APP in cerebral lipid metabolism. From our results we conclude that increased APP metabolism in ApoBxAPP and APPSL mice induces mechanisms to reduce free cholesterol levels.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Córtex Cerebral/metabolismo , Colesterol/metabolismo , Doença de Alzheimer/sangue , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apolipoproteína B-100/genética , Apolipoproteína B-100/metabolismo , Colesterol/sangue , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/metabolismo , Feminino , Genótipo , Homeostase , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL
2.
J Neuroinflammation ; 11: 84, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24886182

RESUMO

BACKGROUND: Beyond cognitive decline, Alzheimer's disease (AD) is characterized by numerous neuropathological changes in the brain. Although animal models generally do not fully reflect the broad spectrum of disease-specific alterations, the APPSL mouse model is well known to display early plaque formation and to exhibit spatial learning and memory deficits. However, important neuropathological features, such as neuroinflammation and lipid peroxidation, and their progression over age, have not yet been described in this AD mouse model. METHODS: Hippocampal and neocortical tissues of APPSL mice at different ages were evaluated. One hemisphere from each mouse was examined for micro- and astrogliosis as well as concomitant plaque load. The other hemisphere was evaluated for lipid peroxidation (quantified by a thiobarbituric acid reactive substances (TBARS) assay), changes in Aß abundance (Aß38, Aß40 and Aß42 analyses), as well as determination of aggregated Aß content (Amorfix A4 assay). Finally, correlation analyses were performed to illustrate the time-dependent correlation between neuroinflammation and Aß load (soluble, insoluble, fibrils), or lipid peroxidation, respectively. RESULTS: As is consistent with previous findings, neuroinflammation starts early and shows strong progression over age in the APPSL mouse model. An analyses of concomitant Aß load and plaque deposition revealed a similar progression, and high correlations between neuroinflammation markers and soluble or insoluble Aß or fibrillar amyloid plaque loads were observed. Lipid peroxidation, as measured by TBARS levels, correlates well with neuroinflammation in the neocortex but not the hippocampus. The hippocampal lipid peroxidation correlated strongly with the increase of LOC positive fiber load, whereas neocortical TBARS levels were unrelated to amyloidosis. CONCLUSIONS: These data illustrate for the first time the progression of major AD related neuropathological features other than plaque load in the APPSL mouse model. Specifically, we demonstrate that microgliosis and astrocytosis are prominent aspects of this AD mouse model. The strong correlation of neuroinflammation with amyloid burden and lipid peroxidation underlines the importance of these pathological factors for the development of AD. The new finding of a different relation of lipid peroxidation in the hippocampus and neocortical regions show that the model might contribute to the understanding of complex pathological mechanisms and their interplay in AD.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide/genética , Encefalite/etiologia , Hipocampo/patologia , Neocórtex/patologia , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/patologia , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
3.
J Alzheimers Dis ; 80(2): 813-829, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33579845

RESUMO

BACKGROUND: Alzheimer's disease (AD) and other tauopathies are neurodegenerative disorders characterized by cellular accumulation of aggregated tau protein. Tau pathology within these disorders is accompanied by chronic neuroinflammation, such as activation of the classical complement pathway by complement initiation factor C1q. Additionally, about half of the AD cases present with inclusions composed of aggregated alpha-synuclein called Lewy bodies. Lewy bodies in disorders such as Parkinson's disease and Lewy body dementia also frequently occur together with tau pathology. OBJECTIVE: Immunotherapy is currently the most promising treatment strategy for tauopathies. However, the presence of multiple pathological processes within tauopathies makes it desirable to simultaneously target more than one disease pathway. METHODS: Herein, we have developed three bispecific antibodies based on published antibody binding region sequences. One bispecific antibody binds to tau plus alpha-synuclein and two bispecific antibodies bind to tau plus C1q. RESULTS: Affinity of the bispecific antibodies to their targets compared to their monospecific counterparts ranged from nearly identical to one order of magnitude lower. All bispecific antibodies retained binding to aggregated protein in patient-derived brain sections. The bispecific antibodies also retained their ability to inhibit aggregation of recombinant tau, regardless of whether the tau binding sites were in IgG or scFv format. Mono- and bispecific antibodies inhibited cellular seeding induced by AD-derived pathological tau with similar efficacy. Finally, both Tau-C1q bispecific antibodies completely inhibited the classical complement pathway. CONCLUSION: Bispecific antibodies that bind to multiple pathological targets may therefore present a promising approach to treat tauopathies and other neurodegenerative disorders.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anticorpos Biespecíficos/farmacologia , Complemento C1q/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Anticorpos Biespecíficos/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Complemento C1q/imunologia , Humanos , Tauopatias/metabolismo , alfa-Sinucleína/imunologia , Proteínas tau/imunologia
4.
Front Neurosci ; 10: 232, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27313503

RESUMO

Besides the continued focus on Aß and Tau in Alzheimer's disease (AD), it is increasingly evident that other pathologic characteristics, such as vascular alterations or inflammation, are associated with AD. Whether these changes are an initial cause for the onset of AD or occur as a result of the disease in late stages is still under debate. In the present study, the impact of the high-fat diet (HFD) induced vascular risk factor hyperlipidemia on Aß levels and clearance as well as cerebral vasculature and blood-brain barrier (BBB) integrity was examined in mice. For this purpose, human APP transgenic (APPSL) and wildtype (WT) mice were fed a HFD for 12 weeks. Plasma and tissues were subsequently investigated for Aß distribution and concentrations of several vascular markers. Decreased plasma Aß together with increased levels of insoluble Aß and amyloid plaques in the brains of HFD fed APPSL mice point toward impaired Aß clearance due to HFD. Additionally, HFD induced manifold alterations in the cerebral vasculature and BBB integrity exclusively in human APP overexpressing mice but not in wildtype mice. Therefore, HFD appears to enhance Aß dependent vascular/BBB dysfunction in combination with an increased proportion of cerebral to plasma Aß in APPSL mice.

5.
Neurobiol Aging ; 34(10): 2379-88, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23643485

RESUMO

During their lifetime, people are commonly exposed to several vascular risk factors that may affect brain ageing and cognitive function. In the last few years, increasing evidence suggests that pathological plasma lipid profiles contribute to the pathogenesis of late-onset Alzheimer's disease. Importantly, hypercholesterolemia, especially elevated low-density lipoprotein cholesterol values, that is, increased apolipoprotein B-100 (ApoB-100) levels, represents an independent risk factor. In this study, the effects of ApoB-100 overexpression, either alone or in combination with cerebral expression of human amyloid precursor protein (hAPP), on cognitive functions and brain pathology were assessed. Our results show that ApoB-100 overexpression induces memory decline and increases cerebral lipid peroxidation and amyloid beta levels compared to those in wild-type animals. Although double-transgenic ApoBxAPP animals did not develop more distinct behavioral deficits than single-transgenic hAPP littermates, hApoB-100 expression caused additional pathophysiological features, such as high LDL and low HDL-cholesterol levels, increased lipid peroxidation, and pronounced ApoB-100 accumulation in cerebral vessels. Thus, our results indicate that ApoBxAPP mice might better reflect the situation of elderly humans than hAPPsl overexpression alone.


Assuntos
Apolipoproteína B-100/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Cognição , Expressão Gênica , Camundongos Transgênicos , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Selvagens , Apolipoproteína B-100/genética , Encéfalo/fisiopatologia , HDL-Colesterol/deficiência , LDL-Colesterol/metabolismo , Feminino , Humanos , Hipercolesterolemia , Peroxidação de Lipídeos , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Risco
6.
J Mol Neurosci ; 47(1): 192-203, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22351109

RESUMO

The role of hyperphosphorylation of the microtubule-associated protein tau in the pathological processes of several neurodegenerative diseases is becoming better understood. Consequently, development of new compounds capable of preventing tau hyperphosphorylation is an increasingly hot topic. For this reason, dependable in vitro and in vivo models that reflect tau hyperphosphorylation in human diseases are needed. In this study, we generated and validated an in vitro model appropriate to test potential curative and preventive compound effects on tau phosphorylation. For this purpose, a stably transfected SH-SY5Y cell line was constructed over-expressing mutant human tau441 (SH-SY5Y-TMHT441). Analyses of expression levels and tau phosphorylation status in untreated cells confirmed relevance to human diseases. Subsequently, the effect of different established kinase inhibitors on tau phosphorylation (e.g., residues Thr231, Thr181, and Ser396) was examined. It was shown with several methods including immunosorbent assays and mass spectrometry that the phosphorylation pattern of tau in SH-SY5Y-TMHT441 cells can be reliably modulated by these compounds, specifically targeting JNK, GSK-3, CDK1/5, and CK1. These four protein kinases are known to be involved in in vivo tau phosphorylation and are therefore authentic indicators for the suitability of this new cell culture model for tauopathies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Testes Genéticos/métodos , Farmacogenética/métodos , Proteínas tau/genética , Doença de Alzheimer/patologia , Animais , Linhagem Celular Tumoral , Desenho de Fármacos , Humanos , Camundongos , Camundongos Transgênicos , Mutagênese/fisiologia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Neuroblastoma/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/metabolismo , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa