Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Rep ; 7(1): 9984, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855586

RESUMO

Tau is a microtubule-associated protein that is highly soluble and natively unfolded. Its dysfunction is involved in the pathogenesis of several neurodegenerative disorders including Alzheimer's disease (AD), where it aggregates within neurons. Deciphering the physiological and pathogenic roles of human Tau (hTau) is crucial to further understand the mechanisms leading to its dysfunction in vivo. We have used a knock-out/knock-in strategy in Drosophila to generate a strain with hTau inserted into the endogenous fly tau locus and expressed under the control of the endogenous fly tau promoter, thus avoiding potential toxicity due to genetic over-expression. hTau knock-in (KI) proteins were expressed at normal, endogenous levels, bound to fly microtubules and were post-translationally modified, hence displaying physiological properties. We used this new model to investigate the effects of acetylation on hTau toxicity in vivo. The simultaneous pseudo-acetylation of hTau at lysines 163, 280, 281 and 369 drastically decreased hTau phosphorylation and significantly reduced its binding to microtubules in vivo. These molecular alterations were associated with ameliorated amyloid beta toxicity. Our results indicate acetylation of hTau on multiple sites regulates its biology and ameliorates amyloid beta toxicity in vivo.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas tau/metabolismo , Acetilação , Animais , Drosophila , Expressão Gênica , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Humanos , Modelos Animais , Fosforilação , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes/metabolismo
2.
Brain ; 140(1): 184-200, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27818384

RESUMO

Alzheimer's disease is characterized by the combined presence of amyloid plaques and tau pathology, the latter being correlated with the progression of clinical symptoms. Neuroinflammatory changes are thought to be major contributors to Alzheimer's disease pathophysiology, even if their precise role still remains largely debated. Notably, to what extent immune responses contribute to cognitive impairments promoted by tau pathology remains poorly understood. To address this question, we took advantage of the THY-Tau22 mouse model that progressively develops hippocampal tau pathology paralleling cognitive deficits and reappraised the interrelationship between tau pathology and brain immune responses. In addition to conventional astroglial and microglial responses, we identified a CD8-positive T cell infiltration in the hippocampus of tau transgenic mice associated with an early chemokine response, notably involving CCL3. Interestingly, CD8-positive lymphocyte infiltration was also observed in the cortex of patients exhibiting frontemporal dementia with P301L tau mutation. To gain insights into the functional involvement of T cell infiltration in the pathophysiological development of tauopathy in THY-Tau22 mice, we chronically depleted T cells using anti-CD3 antibody. Such anti-CD3 treatment prevented hippocampal T cell infiltration in tau transgenic animals and reverted spatial memory deficits, in absence of tau pathology modulation. Altogether, these data support an instrumental role of hippocampal T cell infiltration in tau-driven pathophysiology and cognitive impairments in Alzheimer's disease and other tauopathies.


Assuntos
Anticorpos/uso terapêutico , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Córtex Cerebral/imunologia , Quimiocinas/imunologia , Disfunção Cognitiva/imunologia , Hipocampo/imunologia , Inflamação/imunologia , Tauopatias/imunologia , Idoso , Animais , Disfunção Cognitiva/terapia , Modelos Animais de Doenças , Humanos , Inflamação/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Tauopatias/terapia
3.
Sci Rep ; 6: 22685, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26940749

RESUMO

Dysfunction and accumulation of the microtubule-associated human Tau (hTau) protein into intraneuronal aggregates is observed in many neurodegenerative disorders including Alzheimer's disease (AD). Reversible lysine acetylation has recently emerged as a post-translational modification that may play an important role in the modulation of hTau pathology. Acetylated hTau species have been observed within hTau aggregates in human AD brains and multi-acetylation of hTau in vitro regulates its propensity to aggregate. However, whether lysine acetylation at position 280 (K280) modulates hTau-induced toxicity in vivo is unknown. We generated new Drosophila transgenic models of hTau pathology to evaluate the contribution of K280 acetylation to hTau toxicity, by analysing the respective toxicity of pseudo-acetylated (K280Q) and pseudo-de-acetylated (K280R) mutant forms of hTau. We observed that mis-expression of pseudo-acetylated K280Q-hTau in the adult fly nervous system potently exacerbated fly locomotion defects and photoreceptor neurodegeneration. In addition, modulation of K280 influenced total hTau levels and phosphorylation without changing hTau solubility. Altogether, our results indicate that pseudo-acetylation of the single K280 residue is sufficient to exacerbate hTau neurotoxicity in vivo, suggesting that acetylated K280-hTau species contribute to the pathological events leading to neurodegeneration in AD.


Assuntos
Lisina/metabolismo , Neurotoxinas/toxicidade , Processamento de Proteína Pós-Traducional , Proteínas tau/toxicidade , Acetilação , Doença de Alzheimer/patologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Feminino , Humanos , Neurotoxinas/metabolismo , Agregação Patológica de Proteínas , Proteínas tau/metabolismo
4.
Sci Rep ; 6: 23102, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26976084

RESUMO

Human Tau (hTau) is a highly soluble and natively unfolded protein that binds to microtubules within neurons. Its dysfunction and aggregation into insoluble paired helical filaments is involved in the pathogenesis of Alzheimer's disease (AD), constituting, together with accumulated ß-amyloid (Aß) peptides, a hallmark of the disease. Deciphering both the loss-of-function and toxic gain-of-function of hTau proteins is crucial to further understand the mechanisms leading to neurodegeneration in AD. As the fruit fly Drosophila melanogaster expresses Tau proteins (dTau) that are homologous to hTau, we aimed to better comprehend dTau functions by generating a specific tau knock-out (KO) fly line using homologous recombination. We observed that the specific removal of endogenous dTau proteins did not lead to overt, macroscopic phenotypes in flies. Indeed, survival, climbing ability and neuronal function were unchanged in tau KO flies. In addition, we did not find any overt positive or negative effect of dTau removal on human Aß-induced toxicity. Altogether, our results indicate that the absence of dTau proteins has no major functional impact on flies, and suggests that our tau KO strain is a relevant model to further investigate the role of dTau proteins in vivo, thereby giving additional insights into hTau functions.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas tau/genética , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/fisiologia , Técnicas de Silenciamento de Genes , Expectativa de Vida , Locomoção , Neurônios/fisiologia
5.
Acta Neuropathol ; 130(1): 35-47, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25862636

RESUMO

The involvement of Amyloid-ß (Aß) in the pathogenesis of Alzheimer's disease (AD) is well established. However, it is becoming clear that the amyloid load in AD brains consists of a heterogeneous mixture of Aß peptides, implying that a thorough understanding of their respective role and toxicity is crucial for the development of efficient treatments. Besides the well-studied Aß40 and Aß42 species, recent data have raised the possibility that Aß43 peptides might be instrumental in AD pathogenesis, because they are frequently observed in both dense and diffuse amyloid plaques from human AD brains and are highly amyloidogenic in vitro. However, whether Aß43 is toxic in vivo is currently unclear. Using Drosophila transgenic models of amyloid pathology, we show that Aß43 peptides are mainly insoluble and highly toxic in vivo, leading to the progressive loss of photoreceptor neurons, altered locomotion and decreased lifespan when expressed in the adult fly nervous system. In addition, we demonstrate that Aß43 species are able to trigger the aggregation of the typically soluble and non-toxic Aß40, leading to synergistic toxic effects on fly lifespan and climbing ability, further suggesting that Aß43 peptides could act as a nucleating factor in AD brains. Altogether, our study demonstrates high pathogenicity of Aß43 species in vivo and supports the idea that Aß43 contributes to the pathological events leading to neurodegeneration in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Peptídeos beta-Amiloides/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Feminino , Cabeça/patologia , Cabeça/fisiologia , Atividade Motora/fisiologia , Fragmentos de Peptídeos/genética , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/fisiologia , Agregados Proteicos , RNA Mensageiro/metabolismo , Solubilidade , Análise de Sobrevida
6.
Neurobiol Aging ; 36(2): 730-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25443285

RESUMO

Cognitive decline, the hallmark of Alzheimer's disease, and accompanying neuropsychiatric symptoms share dysfunctions of synaptic processes as a common cellular pathomechanism. Long-term potentiation has proven to be a sensitive tool for the "diagnosis" of such synaptic dysfunctions. Much less, however, is known about how long-term depression (LTD), an alternative mechanism for the storage of memory, is affected by Alzheimer's disease progression. Here, we demonstrate that impaired late LTD (>3 hours) in THY-Tau22 mice can be rescued by either inhibition of glycogen synthase kinase-3 (GSK3ß) activity or by application of the protein-phosphatase 2A agonist selenate. In line with these findings, we observed increased phosphorylation of GSK3ß at Y216 and reduced total phosphatase activity in biochemical assays of hippocampal tissue of THY-Tau22 mice. Interestingly, LTD induction and pharmacologic inhibition of GSK3ß appeared to downregulate GSK3ß activity via a marked upregulation of phosphorylation at the inhibitory Ser9 residue. Our results point to alterations in phosphorylation and/or dephosphorylation homeostasis as key mechanisms underlying the deficits in LTD and hippocampus-dependent learning found in THY-Tau22 mice.


Assuntos
Depressão/genética , Proteínas tau/genética , Doença de Alzheimer/psicologia , Animais , Depressão/diagnóstico , Depressão/tratamento farmacológico , Depressão/psicologia , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hipocampo/metabolismo , Hipocampo/fisiologia , Aprendizagem , Potenciação de Longa Duração , Masculino , Memória , Camundongos Transgênicos , Terapia de Alvo Molecular , Fosforilação , Proteína Fosfatase 2/metabolismo
7.
J Vis Exp ; (86)2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24747743

RESUMO

Two-dimensional gel electrophoresis (2DE) is a powerful tool to uncover proteome modifications potentially related to different physiological or pathological conditions. Basically, this technique is based on the separation of proteins according to their isoelectric point in a first step, and secondly according to their molecular weights by SDS polyacrylamide gel electrophoresis (SDS-PAGE). In this report an optimized sample preparation protocol for little amount of human post-mortem and mouse brain tissue is described. This method enables to perform both two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) and mini 2DE immunoblotting. The combination of these approaches allows one to not only find new proteins and/or protein modifications in their expression thanks to its compatibility with mass spectrometry detection, but also a new insight into markers validation. Thus, mini-2DE coupled to western blotting permits to identify and validate post-translational modifications, proteins catabolism and provides a qualitative comparison among different conditions and/or treatments. Herein, we provide a method to study components of protein aggregates found in AD and Lewy body dementia such as the amyloid-beta peptide and the alpha-synuclein. Our method can thus be adapted for the analysis of the proteome and insoluble proteins extract from human brain tissue and mice models too. In parallel, it may provide useful information for the study of molecular and cellular pathways involved in neurodegenerative diseases as well as potential novel biomarkers and therapeutic targets.


Assuntos
Encéfalo/metabolismo , Immunoblotting/métodos , Proteínas do Tecido Nervoso/análise , Proteoma/análise , Eletroforese em Gel Diferencial Bidimensional/métodos , Animais , Química Encefálica , Carbocianinas/química , Corantes Fluorescentes/química , Humanos , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteoma/metabolismo
8.
Biochem Soc Trans ; 42(2): 587-92, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24646282

RESUMO

AD (Alzheimer's disease) is the most prevalent form of dementia in the aged population. Definitive diagnosis of AD is based on the presence of senile plaques and neurofibrillary tangles that are identified in post-mortem brain specimens. A third pathological component is inflammation. AD results from multiple genetic and environmental risk factors. Among other factors, epidemiological studies report beneficial effects of caffeine, a non-selective antagonist of adenosine receptors. In the present review, we discuss the impact of caffeine and the adenosinergic system in AD pathology as well as consequences in terms of pathology and therapeutics.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Cafeína/uso terapêutico , Antagonistas de Receptores Purinérgicos P1/uso terapêutico , Animais , Humanos , Receptores Purinérgicos P1/metabolismo
9.
Neurobiol Dis ; 58: 179-82, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23732677

RESUMO

Habitual consumption of caffeine, a non-selective adenosine receptor (AR) antagonist, has been suggested to be beneficial in Parkinson's and Alzheimer's diseases. Experimental evidence support that ARs play a role in Huntington's disease (HD) raising the hypothesis that caffeine may be a life-style modifier in HD. To determine a possible relationship between caffeine consumption and age at onset (AAO) in HD, we retrospectively assessed caffeine consumption in 80 HD patients using a dietary survey and determined relationship with AAO. Following adjustment for gender, smoking status and CAG repeat length, caffeine consumption greater than 190mg/day was significantly associated with an earlier AAO. These data support an association between habitual caffeine intake and AAO in HD patients, but further studies are warranted to understand the link between these variables.


Assuntos
Cafeína/efeitos adversos , Doença de Huntington/induzido quimicamente , Doença de Huntington/epidemiologia , Adulto , Idade de Início , Coffea/metabolismo , Feminino , França , Humanos , Doença de Huntington/genética , Modelos Lineares , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Estudos Retrospectivos , Autorrelato , Estatísticas não Paramétricas , Expansão das Repetições de Trinucleotídeos/genética
10.
Diabetes ; 62(5): 1681-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23250356

RESUMO

The τ pathology found in Alzheimer disease (AD) is crucial in cognitive decline. Midlife development of obesity, a major risk factor of insulin resistance and type 2 diabetes, increases the risk of dementia and AD later in life. The impact of obesity on AD risk has been suggested to be related to central insulin resistance, secondary to peripheral insulin resistance. The effects of diet-induced obesity (DIO) on τ pathology remain unknown. In this study, we evaluated effects of a high-fat diet, given at an early pathological stage, in the THY-Tau22 transgenic mouse model of progressive AD-like τ pathology. We found that early and progressive obesity potentiated spatial learning deficits as well as hippocampal τ pathology at a later stage. Surprisingly, THY-Tau22 mice did not exhibit peripheral insulin resistance. Further, pathological worsening occurred while hippocampal insulin signaling was upregulated. Together, our data demonstrate that DIO worsens τ phosphorylation and learning abilities in τ transgenic mice independently from peripheral/central insulin resistance.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Comportamento Animal , Hipocampo/patologia , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Deficiências da Aprendizagem/etiologia , Masculino , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/etiologia , Obesidade/patologia , Obesidade/fisiopatologia , Fosforilação , Processamento de Proteína Pós-Traducional , Distribuição Aleatória , Transdução de Sinais , Comportamento Espacial , Tauopatias/etiologia , Tauopatias/patologia , Tauopatias/fisiopatologia , Regulação para Cima , Proteínas tau/genética
11.
Aging Cell ; 12(1): 11-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23082852

RESUMO

While the spatiotemporal development of Tau pathology has been correlated with occurrence of cognitive deficits in Alzheimer's patients, mechanisms underlying these deficits remain unclear. Both brain-derived neurotrophic factor (BDNF) and its tyrosine kinase receptor TrkB play a critical role in hippocampus-dependent synaptic plasticity and memory. When applied on hippocampal slices, BDNF is able to enhance AMPA receptor-dependent hippocampal basal synaptic transmission through a mechanism involving TrkB and N-methyl-d-Aspartate receptors (NMDAR). Using THY-Tau22 transgenic mice, we demonstrated that hippocampal Tau pathology is associated with loss of synaptic enhancement normally induced by exogenous BDNF. This defective response was concomitant to significant memory impairments. We show here that loss of BDNF response was due to impaired NMDAR function. Indeed, we observed a significant reduction of NMDA-induced field excitatory postsynaptic potential depression in the hippocampus of Tau mice together with a reduced phosphorylation of NR2B at the Y1472, known to be critical for NMDAR function. Interestingly, we found that both NR2B and Src, one of the NR2B main kinases, interact with Tau and are mislocalized to the insoluble protein fraction rich in pathological Tau species. Defective response to BDNF was thus likely related to abnormal interaction of Src and NR2B with Tau in THY-Tau22 animals. These are the first data demonstrating a relationship between Tau pathology and synaptic effects of BDNF and supporting a contribution of defective BDNF response and impaired NMDAR function to the cognitive deficits associated with Tauopathies.


Assuntos
Doença de Alzheimer/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Proteínas tau/genética , Doença de Alzheimer/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transmissão Sináptica/efeitos dos fármacos , Transgenes , Proteínas tau/biossíntese
12.
Transfusion ; 52(8): 1721-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22211513

RESUMO

BACKGROUND: Human blood platelets (PLTs) contain brain-derived neurotrophic factor (BDNF), a neurotrophin that binds to neurotrophic tropomyosin-related kinase B (TrkB) receptor on central nervous system cells. This binding promotes neural synaptic plasticity and memory and prevents neuronal degeneration. Alterations in BDNF homeostasis are associated with aging and are found in several neurodegenerative conditions such as Alzheimer's, Huntington's, and Parkinson's diseases and multiple sclerosis. We have developed PLT viral inactivation and chromatographic fractionation processes and decided here to identify fractions enriched in BDNF. STUDY DESIGN AND METHODS: PLT concentrates (PCs) were treated by solvent/detergent (S/D), extracted by oil, and subjected to fractionation (C18, sulfopropyl [SP]-Sepharose, diethylaminoethyl [DEAE]-Sepharose, or activated charcoal). BDNF and pro-BDNF were evaluated by enzyme-linked immunosorbent assay, and Western blot. TrkB was studied by Western blot. Tri-n-butyl phosphate (TnBP) was quantified by high-performance liquid chromatography, and Triton X-45 by gas chromatography. RESULTS: The mean BDNF content of 2.9 ± 0.7 ng/mL in PC was noted to increase to 56.2 ± 2.4 ng/mL after S/D treatment and remained stable during oil extraction. Approximately 70% of the BDNF content was recovered after C18 chromatography. BDNF did not bind to DEAE-Sepharose and was almost completely adsorbed by charcoal. Chromatography on SP-Sepharose yielded a highly enriched 13-kDa mature BDNF fraction that was more than 170-fold purified, with a mean of 137 ± 29.4 ng/mL and 82% chromatographic recovery, devoid of detectable TnBP and Triton X-45. Pro-BDNF and TrkB proteins were not detected in the PLT extracts. CONCLUSION: We obtained a S/D-treated, highly enriched mature PLT-derived BDNF fraction that could help unveil the pharmacokinetics, pharmacodynamic, and potential therapeutic applications of the BDNF neurotrophin.


Assuntos
Plaquetas/metabolismo , Fator Neurotrófico Derivado do Encéfalo/isolamento & purificação , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fracionamento Celular/métodos , Detergentes/farmacologia , Solventes/farmacologia , Animais , Western Blotting/métodos , Cromatografia Gasosa/métodos , Cromatografia Líquida/métodos , Hipocampo/metabolismo , Humanos , Camundongos , Contagem de Plaquetas , Plaquetoferese/métodos , Receptor trkB/metabolismo
13.
Curr Alzheimer Res ; 9(4): 406-10, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22272617

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular accumulation of amyloid deposits and intracellular neurofibrillary tangles (NFT) composed of hyperphosphorylated Tau proteins. Brain-derived neurotrophic factor (BDNF) is a neurotrophic factor playing a critical role in hippocampal synaptic plasticity and memory and whose levels have been shown reduced in AD brains. While recent data support a pivotal role of ß-amyloid peptides towards BDNF decrease, whether Tau pathology impacts on BDNF expression remains unknown so far. In the present study, we have evaluated this relationship using quantitative PCR, Western blot and ELISA in the THY-Tau22 transgenic strain, known to display a progressive development of both hippocampal AD-like Tau pathology and memory impairments. We observed that Tau pathology was not associated with down-regulation of BDNF at the protein and mRNA levels in this model, suggesting that the alteration of BDNF homeostasis observed in AD patients' brains might rather be ascribed to amyloid pathology.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Tauopatias/patologia , Regulação para Cima/genética , Proteínas tau/genética , Fatores Etários , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , RNA Mensageiro/metabolismo , Tauopatias/genética , Proteínas tau/metabolismo
14.
Curr Alzheimer Res ; 9(4): 397-405, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22272619

RESUMO

Recent data indicate that Tau immunotherapy may be relevant for interfering with neurofibrillary degeneration in Alzheimer disease and related disorders referred to as Tauopathies. The key question for immunotherapy is the choice of the epitope to target. Abnormal phosphorylation is a well-described post-translational modification of Tau proteins and may be a good target. In the present study, we investigated the effects of active immunization against the pathological epitope phospho-Ser422 in the THY-Tau22 transgenic mouse model. Starting from 3-6 months of age, THY-Tau22 mice develop hippocampal neurofibrillary tangle-like inclusions and exhibit phosphorylation of Tau on several AD-relevant Tau epitopes. Three month-old THY-Tau22 mice were immunized with a peptide including the phosphoserine 422 residue while control mice received the adjuvant alone. A specific antibody response against the phospho-Ser422 epitope was observed. We noticed a decrease in insoluble Tau species (AT100- and pS422 immunoreactive) by both biochemical and immunohistochemical means correlated with a significant cognitive improvement using the Y-maze. This Tau immunotherapy may facilitate Tau clearance from the brain toward the periphery since, following immunization, an increase in Tau concentrations was observed in blood. Overall, the present work is, to our knowledge, the first one to demonstrate that active immunotherapy targeting a real pathological epitope such as phospho-Ser422 epitope is efficient. This immunotherapy allows for Tau clearance and improves cognitive deficits promoted by Tau pathology in a well-defined Tau transgenic model.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Imunoterapia Ativa/métodos , Mutação/genética , Serina/metabolismo , Proteínas tau/metabolismo , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Anticorpos/sangue , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/terapia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptídeos/administração & dosagem , Peptídeos/imunologia , Fosforilação/imunologia , Serina/genética , Proteínas tau/genética
15.
Neurobiol Dis ; 43(2): 486-94, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21569847

RESUMO

Tau pathology is encountered in many neurodegenerative disorders known as tauopathies, including Alzheimer's disease. Physical activity is a lifestyle factor affecting processes crucial for memory and synaptic plasticity. Whether long-term voluntary exercise has an impact on Tau pathology and its pathophysiological consequences is currently unknown. To address this question, we investigated the effects of long-term voluntary exercise in the THY-Tau22 transgenic model of Alzheimer's disease-like Tau pathology, characterized by the progressive development of Tau pathology, cholinergic alterations and subsequent memory impairments. Three-month-old THY-Tau22 mice and wild-type littermates were assigned to standard housing or housing supplemented with a running wheel. After 9 months of exercise, mice were evaluated for memory performance and examined for hippocampal Tau pathology, cholinergic defects, inflammation and genes related to cholesterol metabolism. Exercise prevented memory alterations in THY-Tau22 mice. This was accompanied by a decrease in hippocampal Tau pathology and a prevention of the loss of expression of choline acetyltransferase within the medial septum. Whereas the expression of most cholesterol-related genes remained unchanged in the hippocampus of running THY-Tau22 mice, we observed a significant upregulation in mRNA levels of NPC1 and NPC2, genes involved in cholesterol trafficking from the lysosomes. Our data support the view that long-term voluntary physical exercise is an effective strategy capable of mitigating Tau pathology and its pathophysiological consequences.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Terapia por Exercício/métodos , Condicionamento Físico Animal/fisiologia , Proteínas tau/genética , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas tau/efeitos adversos , Proteínas tau/antagonistas & inibidores
16.
Neurobiol Learn Mem ; 95(3): 296-304, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21167950

RESUMO

We evaluated various forms of hippocampus-dependent learning and memory, and hippocampal synaptic plasticity in THY-Tau22 transgenic mice, a murine tauopathy model that expresses double-mutated 4-repeat human tau, and shows neuropathological tau hyperphosphorylation and aggregation throughout the brain. Focussing on hippocampus, immunohistochemical studies in aged THY-Tau22 mice revealed prominent hyper- and abnormal phosphorylation of tau in CA1 region, and an increase in glial fibrillary acidic protein (GFAP) in hippocampus, but without signs of neuronal loss. These mice displayed spatial, social, and contextual learning and memory defects that could not be reduced to subtle neuromotor disability. The behavioral defects coincided with changes in hippocampal synaptic functioning and plasticity as measured in paired-pulse and novel long-term depression protocols. These results indicate that hippocampal tauopathy without neuronal cell loss can impair neural and behavioral plasticity, and further show that transgenic mice, such as the THY-Tau22 strain, might be useful for preclinical research on tauopathy pathogenesis and possible treatment.


Assuntos
Aprendizagem por Associação/fisiologia , Hipocampo/fisiopatologia , Depressão Sináptica de Longo Prazo/fisiologia , Aprendizagem em Labirinto/fisiologia , Tauopatias/fisiopatologia , Análise de Variância , Animais , Condicionamento Clássico/fisiologia , Modelos Animais de Doenças , Medo , Preferências Alimentares , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Masculino , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Comportamento Social , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/genética , Proteínas tau/metabolismo
17.
Biochem Soc Trans ; 38(4): 967-72, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658986

RESUMO

Tau pathology is characterized by intracellular aggregates of abnormally and hyperphosphorylated tau proteins. It is encountered in many neurodegenerative disorders, but also in aging. These neurodegenerative disorders are referred to as tauopathies. Comparative biochemistry of the tau aggregates shows that they differ in both tau isoform phosphorylation and content, which enables a molecular classification of tauopathies. In conditions of dementia, NFD (neurofibrillary degeneration) severity is correlated to cognitive impairment and is often considered as neuronal death. Using tau animal models, analysis of the kinetics of tau phosphorylation, aggregation and neuronal death in parallel to electrophysiological and behavioural parameters indicates a disconnection between cognition deficits and neuronal cell death. Tau phosphorylation and aggregation are early events followed by cognitive impairment. Neuronal death is not observed before the oldest ages. A sequence of events may be the formation of toxic phosphorylated tau species, their aggregation, the formation of neurofibrillary tangles (from pre-tangles to ghost tangles) and finally neuronal cell death. This sequence will last from 15 to 25 years and one can ask whether the aggregation of toxic phosphorylated tau species is a protection against cell death. Apoptosis takes 24 h, but NFD lasts for 24 years to finally kill the neuron or rather to protect it for more than 20 years. Altogether, these data suggest that NFD is a transient state before neuronal death and that therapeutic interventions are possible at that stage.


Assuntos
Neurônios/fisiologia , Proteínas Quinases/metabolismo , Multimerização Proteica/fisiologia , Proteínas tau/metabolismo , Animais , Morte Celular/fisiologia , Precipitação Química , Modelos Animais de Doenças , Humanos , Modelos Biológicos , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Tauopatias/etiologia , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/química , Proteínas tau/fisiologia
18.
Neurobiol Dis ; 35(3): 474-6, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19591938

RESUMO

Based on the pathophysiological role of adenosine A(2A) receptors in HD, we have evaluated the association of the 1976C/T single-nucleotide polymorphism in the ADORA2A gene (rs5751876) with residual age at onset (AAO) in HD. The study population consisted of 791 unrelated patients belonging to the Huntington French Speaking Network. The variability in AAO attributable to the CAG repeats number was calculated by linear regression using the log (AAO) as the dependent variable, and the respective rs5751876 genotypes as independent variables. We show that the rs5751876 variant significantly influences the variability in AAO. The R(2) statistic rose slightly but significantly (p=0.019) when rs5751876 T/T genotype was added to the regression model. Patients harbouring T/T genotype have an earlier AAO of 3.8 years as compared to C/C genotype (p=0.02). Our data thus strengthens the pathophysiological role of A(2A) receptors in Huntington's disease.


Assuntos
Doença de Huntington/epidemiologia , Doença de Huntington/genética , Polimorfismo de Nucleotídeo Único , Receptor A2A de Adenosina/genética , Adolescente , Adulto , Idade de Início , Idoso , Criança , Estudos de Coortes , Genótipo , Humanos , Modelos Lineares , Pessoa de Meia-Idade , Expansão das Repetições de Trinucleotídeos , Adulto Jovem
19.
Curr Alzheimer Res ; 6(2): 152-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19355850

RESUMO

Alzheimer's disease is a neurodegenerative disorder characterized by amyloid deposits and neurofibrillary tangles. Cholinergic dysfunction is also a main pathological feature of the disease. Nevertheless, the links between cholinergic dysfunction and neuropathological hallmarks of Alzheimer's are still unknown. In the present study, we aimed to further investigate Tau aggregation in cholinergic systems, in a Tau transgenic mouse model. THY-Tau22 mice have recently been described as a novel model of Alzheimer-like Tau pathology without motor deficits. This strain presents an age-dependent development of Tau pathology leading to synaptic dysfunctions as well as learning and memory impairments. In the present work, we observed that Tau pathology differentially affects cerebral structures. Interestingly, early Tau pathology was observed in both hippocampus and basal forebrain. Moreover, some morphological as well as functional alterations of the septohippocampal pathway suggest a disconnection between these two key brain regions in Alzheimer's disease. Finally, these data suggest that Tau pathology may participate in cholinergic degeneration.


Assuntos
Doença de Alzheimer/patologia , Hipocampo/patologia , Septo do Cérebro/patologia , Proteínas tau/genética , Proteínas tau/metabolismo , Fatores Etários , Animais , Mapeamento Encefálico , Modelos Animais de Doenças , Glicina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Vias Neurais/patologia , Prolina/genética , Serina/genética , Estilbamidinas/metabolismo , Valina/genética
20.
Curr Pharm Des ; 14(15): 1500-11, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18537673

RESUMO

Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder caused by the expansion of a polymorphic CAG trinucleotide repeat encoding a poly-glutamine tract within the Huntingtin protein. GABAergic enkephalin neurons of the basal ganglia, which show the highest levels of expression of adenosine A(2A) receptors, are the most vulnerable in HD. Such a selective neuronal vulnerability, which occurs despite ubiquitous expression of mutant and normal Huntingtin, has suggested that adenosine A(2A) receptors might play a pathogenetic role in HD. In agreement, changes in A(2A) receptor expression and signaling have been reported in various experimental models of HD. The interpretation of the functional significance of the aberrant A(2A) receptor phenotype in HD mice is however complicated by the conflicting data so far reported on the potential neuroprotective and neurodegenerative effects of these receptors in the brain, with some data suggesting a potential pathogenetic role and some other data suggesting activation of trophic or protective pathways in neurons. The same complex profile has emerged in experimental models of HD, in which both A(2A) receptor agonists and antagonists have shown beneficial effects. The main aim of this review is to critically evaluate whether adenosine A(2A) receptors may represent a suitable target to develop drugs against HD.


Assuntos
Doença de Huntington/metabolismo , Receptor A2A de Adenosina/fisiologia , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Modelos Animais de Doenças , Humanos , Doença de Huntington/tratamento farmacológico , Camundongos , Fármacos Neuroprotetores/uso terapêutico , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA