Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Brain ; 147(8): 2691-2705, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38964748

RESUMO

Early pathological upregulation of adenosine A2A receptors (A2ARs), one of the caffeine targets, by neurons is thought to be involved in the development of synaptic and memory deficits in Alzheimer's disease (AD) but mechanisms remain ill-defined. To tackle this question, we promoted a neuronal upregulation of A2AR in the hippocampus of APP/PS1 mice developing AD-like amyloidogenesis. Our findings revealed that the early upregulation of A2AR in the presence of an ongoing amyloid pathology exacerbates memory impairments of APP/PS1 mice. These behavioural changes were not linked to major change in the development of amyloid pathology but rather associated with increased phosphorylated tau at neuritic plaques. Moreover, proteomic and transcriptomic analyses coupled with quantitative immunofluorescence studies indicated that neuronal upregulation of the receptor promoted both neuronal and non-neuronal autonomous alterations, i.e. enhanced neuroinflammatory response but also loss of excitatory synapses and impaired neuronal mitochondrial function, presumably accounting for the detrimental effect on memory. Overall, our results provide compelling evidence that neuronal A2AR dysfunction, as seen in the brain of patients, contributes to amyloid-related pathogenesis and underscores the potential of A2AR as a relevant therapeutic target for mitigating cognitive impairments in this neurodegenerative disorder.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Transtornos da Memória , Camundongos Transgênicos , Neurônios , Receptor A2A de Adenosina , Sinapses , Animais , Transtornos da Memória/metabolismo , Transtornos da Memória/genética , Transtornos da Memória/patologia , Camundongos , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Sinapses/metabolismo , Sinapses/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Hipocampo/metabolismo , Hipocampo/patologia , Presenilina-1/genética , Modelos Animais de Doenças , Placa Amiloide/patologia , Placa Amiloide/metabolismo , Masculino , Camundongos Endogâmicos C57BL
2.
Biomedicines ; 10(2)2022 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-35203451

RESUMO

Tau proteins are known to be mainly involved in regulation of microtubule dynamics. Besides this function, which is critical for axonal transport and signal transduction, tau proteins also have other roles in neurons. Moreover, tau proteins are turned into aggregates and consequently trigger many neurodegenerative diseases termed tauopathies, of which Alzheimer's disease (AD) is the figurehead. Such pathological aggregation processes are critical for the onset of these diseases. Among the various causes of tau protein pathogenicity, abnormal tau mRNA metabolism, expression and dysregulation of tau post-translational modifications are critical steps. Moreover, the relevance of tau function to general mRNA metabolism has been highlighted recently in tauopathies. In this review, we mainly focus on how mRNA metabolism impacts the onset and development of tauopathies. Thus, we intend to portray how mRNA metabolism of, or mediated by, tau is associated with neurodegenerative diseases.

3.
Brain ; 142(11): 3636-3654, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31599329

RESUMO

Accumulating data support the role of tau pathology in cognitive decline in ageing and Alzheimer's disease, but underlying mechanisms remain ill-defined. Interestingly, ageing and Alzheimer's disease have been associated with an abnormal upregulation of adenosine A2A receptor (A2AR), a fine tuner of synaptic plasticity. However, the link between A2AR signalling and tau pathology has remained largely unexplored. In the present study, we report for the first time a significant upregulation of A2AR in patients suffering from frontotemporal lobar degeneration with the MAPT P301L mutation. To model these alterations, we induced neuronal A2AR upregulation in a tauopathy mouse model (THY-Tau22) using a new conditional strain allowing forebrain overexpression of the receptor. We found that neuronal A2AR upregulation increases tau hyperphosphorylation, potentiating the onset of tau-induced memory deficits. This detrimental effect was linked to a singular microglial signature as revealed by RNA sequencing analysis. In particular, we found that A2AR overexpression in THY-Tau22 mice led to the hippocampal upregulation of C1q complement protein-also observed in patients with frontotemporal lobar degeneration-and correlated with the loss of glutamatergic synapses, likely underlying the observed memory deficits. These data reveal a key impact of overactive neuronal A2AR in the onset of synaptic loss in tauopathies, paving the way for new therapeutic approaches.


Assuntos
Complemento C1q/metabolismo , Neurônios/metabolismo , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Sinapses/patologia , Tauopatias/genética , Tauopatias/patologia , Animais , Autopsia , Degeneração Lobar Frontotemporal/genética , Degeneração Lobar Frontotemporal/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Transtornos da Memória/etiologia , Transtornos da Memória/psicologia , Camundongos , Camundongos Transgênicos , Mutação , Aprendizagem Espacial , Tauopatias/psicologia , Proteínas tau/genética
4.
Neurobiol Dis ; 129: 217-233, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30928644

RESUMO

Alzheimer's Disease is a devastating dementing disease involving amyloid deposits, neurofibrillary tangles, progressive and irreversible cognitive impairment. Today, only symptomatic drugs are available and therapeutic treatments, possibly acting at a multiscale level, are thus urgently needed. To that purpose, we designed multi-effects compounds by synthesizing drug candidates derived by substituting a novel N,N'-disubstituted piperazine anti-amyloid scaffold and adding acetylcholinesterase inhibition property. Two compounds were synthesized and evaluated. The most promising hybrid molecule reduces both the amyloid pathology and the Tau pathology as well as the memory impairments in a preclinical model of Alzheimer's disease. In vitro also, the compound reduces the phosphorylation of Tau and inhibits the release of Aß peptides while preserving the processing of other metabolites of the amyloid precursor protein. We synthetized and tested the first drug capable of ameliorating both the amyloid and Tau pathology in animal models of AD as well as preventing the major brain lesions and associated memory impairments. This work paves the way for future compound medicines against both Alzheimer's-related brain lesions development and the associated cognitive impairments.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/efeitos dos fármacos , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Piperazinas/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Memória/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Placa Amiloide/patologia
5.
Neurobiol Dis ; 125: 14-22, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30665005

RESUMO

Accumulation of hyper-phosphorylated and aggregated Tau proteins is a neuropathological hallmark of Alzheimer's Disease (AD) and Tauopathies. AD patient brains also exhibit insulin resistance. Whereas, under normal physiological conditions insulin signaling in the brain mediates plasticity and memory formation, it can also regulate peripheral energy homeostasis. Thus, in AD, brain insulin resistance affects both cognitive and metabolic changes described in these patients. While a role of Aß oligomers and APOE4 towards the development of brain insulin resistance emerged, contribution of Tau pathology has been largely overlooked. Our recent data demonstrated that one of the physiological function of Tau is to sustain brain insulin signaling. We postulated that under pathological conditions, hyper-phosphorylated/aggregated Tau is likely to lose this function and to favor the development of brain insulin resistance. This hypothesis was substantiated by observations from patient brains with pure Tauopathies. To address the potential link between Tau pathology and brain insulin resistance, we have evaluated the brain response to insulin in a transgenic mouse model of AD-like Tau pathology (THY-Tau22). Using electrophysiological and biochemical evaluations, we surprisingly observed that, at a time when Tau pathology and cognitive deficits are overt and obvious, the hippocampus of THY-Tau22 mice exhibits enhanced response to insulin. In addition, we demonstrated that the ability of i.c.v. insulin to promote body weight loss is enhanced in THY-Tau22 mice. In line with this, THY-Tau22 mice exhibited a lower body weight gain, hypoleptinemia and hypoinsulinemia and finally a metabolic resistance to high-fat diet. The present data highlight that the brain of transgenic Tau mice exhibit enhanced brain response to insulin. Whether these observations are ascribed to the development of Tau pathology, and therefore relevant to human Tauopathies, or unexpectedly results from the Tau transgene overexpression is debatable and discussed.


Assuntos
Encéfalo/metabolismo , Insulina/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas tau/genética
6.
J Exp Med ; 214(8): 2257-2269, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28652303

RESUMO

The molecular pathways underlying tau pathology-induced synaptic/cognitive deficits and neurodegeneration are poorly understood. One prevalent hypothesis is that hyperphosphorylation, misfolding, and fibrillization of tau impair synaptic plasticity and cause degeneration. However, tau pathology may also result in the loss of specific physiological tau functions, which are largely unknown but could contribute to neuronal dysfunction. In the present study, we uncovered a novel function of tau in its ability to regulate brain insulin signaling. We found that tau deletion leads to an impaired hippocampal response to insulin, caused by altered IRS-1 and PTEN (phosphatase and tensin homologue on chromosome 10) activities. Our data also demonstrate that tau knockout mice exhibit an impaired hypothalamic anorexigenic effect of insulin that is associated with energy metabolism alterations. Consistently, we found that tau haplotypes are associated with glycemic traits in humans. The present data have far-reaching clinical implications and raise the hypothesis that pathophysiological tau loss-of-function favors brain insulin resistance, which is instrumental for cognitive and metabolic impairments in Alzheimer's disease patients.


Assuntos
Encéfalo/metabolismo , Resistência à Insulina , Proteínas tau/fisiologia , Animais , Encéfalo/fisiologia , Disfunção Cognitiva/etiologia , Haplótipos , Hipocampo/fisiologia , Humanos , Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Sci Rep ; 6: 31493, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27510168

RESUMO

Caffeine is associated with procognitive effects in humans by counteracting overactivation of the adenosine A2A receptor (A2AR), which is upregulated in the human forebrain of aged and Alzheimer's disease (AD) patients. We have previously shown that an anti-A2AR therapy reverts age-like memory deficits, by reestablishment of the hypothalamic-pituitary-adrenal (HPA) axis feedback and corticosterone circadian levels. These observations suggest that A2AR over-activation and glucocorticoid dysfunction are key events in age-related hippocampal deficits; but their direct connection has never been explored. We now show that inducing A2AR overexpression in an aging-like profile is sufficient to trigger HPA-axis dysfunction, namely loss of plasmatic corticosterone circadian oscillation, and promotes reduction of GR hippocampal levels. The synaptic plasticity and memory deficits triggered by GR in the hippocampus are amplified by A2AR over-activation and were rescued by anti-A2AR therapy; finally, we demonstrate that A2AR act on GR nuclear translocation and GR-dependent transcriptional regulation. We provide the first demonstration that A2AR is a major regulator of GR function and that this functional interconnection may be a trigger to age-related memory deficits. This supports the idea that the procognitive effects of A2AR antagonists, namely caffeine, on Alzheimer's and age-related cognitive impairments may rely on its ability to modulate GR actions.


Assuntos
Envelhecimento/fisiologia , Disfunção Cognitiva/genética , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Receptores de Glucocorticoides/metabolismo , Envelhecimento/metabolismo , Animais , Cafeína/metabolismo , Núcleo Celular/metabolismo , Disfunção Cognitiva/metabolismo , Corticosterona/metabolismo , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Camundongos Transgênicos , Modelos Animais , Plasticidade Neuronal , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Ratos
8.
Sci Rep ; 5: 9659, 2015 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-25974414

RESUMO

Tau is a central player in Alzheimer's disease (AD) and related Tauopathies, where it is found as aggregates in degenerating neurons. Abnormal post-translational modifications, such as truncation, are likely involved in the pathological process. A major step forward in understanding the role of Tau truncation would be to identify the precise cleavage sites of the several truncated Tau fragments that are observed until now in AD brains, especially those truncated at the N-terminus, which are less characterized than those truncated at the C-terminus. Here, we optimized a proteomics approach and succeeded in identifying a number of new N-terminally truncated Tau species from the human brain. We initiated cell-based functional studies by analyzing the biochemical characteristics of two N-terminally truncated Tau species starting at residues Met11 and Gln124 respectively. Our results show, interestingly, that the Gln124-Tau fragment displays a stronger ability to bind and stabilize microtubules, suggesting that the Tau N-terminal domain could play a direct role in the regulation of microtubule stabilization. Future studies based on our new N-terminally truncated-Tau species should improve our knowledge of the role of truncation in Tau biology as well as in the AD pathological process.


Assuntos
Doença de Alzheimer/patologia , Microtúbulos/fisiologia , Tubulina (Proteína)/metabolismo , Proteínas tau/genética , Acetilação , Doença de Alzheimer/genética , Encéfalo/patologia , Linhagem Celular , Humanos , Degeneração Neural/metabolismo , Fosforilação , Ligação Proteica/genética , Processamento de Proteína Pós-Traducional , Proteômica , Proteínas tau/metabolismo
9.
FASEB J ; 29(8): 3171-81, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25888602

RESUMO

Tauopathies, including Alzheimer's disease (AD), are neurodegenerative diseases associated with the pathologic aggregation of human brain Tau protein. Neuronal Tau is involved in microtubule (MT) formation and stabilization. We showed previously that the immunophilin FK506-binding protein of MW ∼52 kDa (FKBP52) interferes with this function of full-length Tau and provokes aggregation of a disease-related mutant of Tau. To dissect the molecular interaction between recombinant human FKBP52 and Tau, here, we study the effect of FKBP52 on a functional Tau fragment (Tau-F4, Ser(208)-Ser(324)) containing part of the proline- rich region and MT-binding repeats. Therefore, we perform MT assembly and light-scattering assays, blue native PAGE analysis, electron microscopy, and Tau seeding experiments in SH-SY5Y human neuroblastoma cells. We show that FKBP52 (6 µM) prevents MT formation generated by Tau-F4 (5 µM) and induces Tau-F4 oligomerization and aggregation. Electron microscopy analyses show granular oligomers and filaments of Tau-F4 after short-time FKBP52 incubation. We demonstrate that the terminal parts of Tau interfere with the effects of FKBP52. Finally, we find that FKBP52-induced Tau-F4 oligomers cannot only generate in vitro, direct conformational changes in full-length Tau and that their uptake into neuronal cells can equally lead to aggregation of wild-type endogenous Tau. This suggests a potential prion-like property of these particular Tau-F4 aggregates. Collectively, our results strengthen the hypothesis of FKBP52 involvement in the Tau pathogenicity process.


Assuntos
Príons/metabolismo , Ligação Proteica/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Microtúbulos/metabolismo , Ratos , Ratos Sprague-Dawley , Tauopatias/metabolismo
10.
ACS Chem Neurosci ; 6(4): 559-69, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25611616

RESUMO

The amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD). Preventing deregulated APP processing by inhibiting amyloidogenic processing of carboxy-terminal fragments (APP-CTFs), and reducing the toxic effect of amyloid beta (Aß) peptides remain an effective therapeutic strategy. We report the design of piperazine-containing compounds derived from chloroquine structure and evaluation of their effects on APP metabolism and ability to modulate the processing of APP-CTF and the production of Aß peptide. Compounds which retained alkaline properties and high affinity for acidic cell compartments were the most effective. The present study demonstrates that (1) the amino side chain of chloroquine can be efficiently substituted by a bis(alkylamino)piperazine chain, (2) the quinoline nucleus can be replaced by a benzyl or a benzimidazole moiety, and (3) pharmacomodulation of the chemical structure allows the redirection of APP metabolism toward a decrease of Aß peptide release, and increased stability of APP-CTFs and amyloid intracellular fragment. Moreover, the benzimidazole compound 29 increases APP-CTFs in vivo and shows promising activity by the oral route. Together, this family of compounds retains a lysosomotropic activity which inhibits lysosome-related Aß production, and is likely to be beneficial for therapeutic applications in AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Cloroquina/análogos & derivados , Fármacos Neuroprotetores/química , Quinolinas/química , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cloroquina/química , Cloroquina/farmacologia , Desenho de Fármacos , Feminino , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Fármacos Neuroprotetores/farmacologia , Fragmentos de Peptídeos/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Quinolinas/farmacologia , Água/química
11.
Neurobiol Aging ; 35(9): 2079-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24780254

RESUMO

Tau pathology found in Alzheimer's disease (AD) is crucial in cognitive decline. Epidemiologic evidences support that habitual caffeine intake prevents memory decline during aging and reduces the risk to develop Alzheimer's disease. So far, experimental studies addressed the impact of caffeine in models mimicking the amyloid pathology of AD. However, in vivo effects of caffeine in a model of AD-like tauopathy remain unknown. Here, we evaluated effects of chronic caffeine intake (0.3 g/L through drinking water), given at an early pathologic stage, in the THY-Tau22 transgenic mouse model of progressive AD-like tau pathology. We found that chronic caffeine intake prevents from the development of spatial memory deficits in tau mice. Improved memory was associated with reduced hippocampal tau phosphorylation and proteolytic fragments. Moreover, caffeine treatment mitigated several proinflammatory and oxidative stress markers found upregulated in the hippocampus of THY-Tau22 animals. Together, our data support that moderate caffeine intake is beneficial in a model of AD-like tau pathology, paving the way for future clinical evaluation in AD patients.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Cafeína/administração & dosagem , Hipocampo/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Cafeína/metabolismo , Cafeína/farmacologia , Modelos Animais de Doenças , Hipocampo/patologia , Masculino , Transtornos da Memória/prevenção & controle , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteólise/efeitos dos fármacos
12.
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
13.
Eur J Med Chem ; 79: 184-93, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24735644

RESUMO

Insulin degrading enzyme (IDE) is a highly conserved zinc metalloprotease that is involved in the clearance of various physiologically peptides like amyloid-beta and insulin. This enzyme has been involved in the physiopathology of diabetes and Alzheimer's disease. We describe here a series of small molecules discovered by screening. Co-crystallization of the compounds with IDE revealed a binding both at the permanent exosite and at the discontinuous, conformational catalytic site. Preliminary structure-activity relationships are described. Selective inhibition of amyloid-beta degradation over insulin hydrolysis was possible. Neuroblastoma cells treated with the optimized compound display a dose-dependent increase in amyloid-beta levels.


Assuntos
Acetatos/farmacologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Imidazóis/química , Insulisina/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Acetatos/síntese química , Acetatos/química , Peptídeos beta-Amiloides/metabolismo , Relação Dose-Resposta a Droga , Humanos , Hidrólise , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
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
15.
Oncotarget ; 5(3): 754-63, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24504508

RESUMO

MUC1 is a membrane-anchored mucin and its cytoplasmic tail (CT) can interact with many signaling pathways and act as a co-transcription factor to activate genes involved in tumor progression and metastasis. MUC1 is overexpressed in renal cell carcinoma with correlation to prognosis and has been implicated in the hypoxic pathway, the main renal carcinogenetic pathway. In this context, we assessed the effects of MUC1 overexpression on renal cancer cells properties. Using shRNA strategy and/or different MUC1 constructs, we found that MUC1-extracellular domain and MUC1-CT are involved in increase of migration, cell viability, resistance to anoikis and in decrease of cell aggregation in cancer cells. Invasiveness depends only on MUC1-CT. Then, by using siRNA strategy and/or pharmacological inhibitors or peptides, we showed that sheddases ADAM10, ADAM17 and gamma-secretase are necessary for MUC1 C-terminal subunit (MUC1-C) nuclear location and in increase of invasion property. Finally, MUC1 overexpression increases ADAM10/17 protein expression suggesting a positive regulatory loop. In conclusion, we report that MUC1 acts in renal cancer progression and MUC1-C nuclear localization drives invasiveness of cancer cells through a sheddase/gamma secretase dependent pathway. MUC1 appears as a therapeutic target by blocking MUC1 cleavage or nuclear translocation by using pharmacological approach and peptide strategies.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Neoplasias Renais/metabolismo , Mucina-1/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Progressão da Doença , Xenoenxertos , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Camundongos SCID , Mucina-1/genética , Subunidades Proteicas , Transdução de Sinais , Transfecção
16.
J Alzheimers Dis ; 37(4): 777-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23948912

RESUMO

Age-related cognitive decline and neurodegenerative diseases are a growing challenge for society. Accumulation of tau pathology has been proposed to partially contribute to these impairments. This study provides a behavioral characterization during aging of transgenic mice bearing tau mutations. THY-Tau22 mice were evaluated at ages wherein tau neuropathology in this transgenic mouse model is low (3-4 months), moderate (6-7 months), or extensive (>9 months). Spatial memory was found to be impaired only after 9 months of age in THY-Tau22 mice, whereas non-spatial memory was affected as early as 6 months, appearing to offer an opportunity for assessing potential therapeutic agents in attenuating or preventing tauopathies through modulation of tau kinetics.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Envelhecimento/patologia , Animais , Transtornos Cognitivos/patologia , Progressão da Doença , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Fosforilação/genética
17.
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
18.
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
19.
Neurobiol Aging ; 34(3): 757-69, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22926167

RESUMO

A prerequisite to dephosphorylation at Ser-Pro or Thr-Pro motifs is the isomerization of the imidic peptide bond preceding the proline. The peptidyl-prolyl cis/trans isomerase named Pin1 catalyzes this mechanism. Through isomerization, Pin1 regulates the function of a growing number of targets including the microtubule-associated tau protein and is supposed to be deregulated Alzheimer's disease (AD). Using proteomics, we showed that Pin1 is posttranslationally modified on more than 5 residues, comprising phosphorylation, N-acetylation, and oxidation. Although Pin1 expression remained constant, Pin1 posttranslational two-dimensional pattern was modified by tau overexpression in a tau-inducible neuroblastoma cell line, in our THY-Tau22 mouse model of tauopathy as well as in AD. Interestingly, in all of these systems, Pin1 modifications were very similar. In AD brain tissue when compared with control, Pin1 is hyperphosphorylated at serine 16 and found in the most insoluble hyperphosphorylated tau fraction of AD brain tissue. Furthermore, in all tau pathology conditions, acetylation of Pin1 may also contribute to the differences observed. In conclusion, Pin1 displays several posttranslational modifications, which are specific in tauopathies and may be useful as biomarker.


Assuntos
Encéfalo/metabolismo , Peptidilprolil Isomerase/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Tauopatias/metabolismo , Proteínas tau/metabolismo , Acetilação , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Peptidilprolil Isomerase de Interação com NIMA , Oxirredução , Fosforilação/fisiologia , Prolina/metabolismo , Proteoma , Serina/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...