Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 193: 106459, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38423192

RESUMO

BACKGROUND AND PURPOSE: Blood-based biomarkers are a non-invasive solution to predict the risk of conversion of mild cognitive impairment (MCI) to dementia. The utility of free plasma amyloid peptides (not bound to plasma proteins and/or cells) as an early indicator of conversion to dementia is still debated, as the results of studies have been contradictory. In this context, we investigated whether plasma levels of the free amyloid peptides Aß1-42 and Aß1-40 and the free plasma Aß1-42/Aß1-40 ratio are associated with the conversion of MCI to dementia, in particular AD, over three years of follow-up in a subgroup of the BALTAZAR cohort. We also compared their predictive value to that of total plasma Aß1-42 and Aß1-40 levels and the total plasma Aß1-42/Aß1-40 ratio. METHODS: The plasma Aß1-42 and Aß1-40 peptide assay was performed using the INNO-BIA kit (Fujirebio Europe). Free amyloid levels (defined by the amyloid fraction directly accessible to antibodies of the assay) were obtained with the undiluted plasma, whereas total amyloid levels were obtained after the dilution of plasma (1/3) with a denaturing buffer. Free and total Aß1-42 and Aß1-40 levels were measured at inclusion for a subgroup of participants (N = 106) with mild cognitive impairment (MCI) from the BALTAZAR study (a large-scale longitudinal multicenter cohort with a three-year follow-up). Associations between conversion and the free/total plasma Aß1-42 and Aß1-40 levels and Aß1-42/Aß1-40 ratio were analyzed using logistic and Cox Proportional Hazards models. Demographic, clinical, cognitive (MMSE, ADL and IADL), APOE, and MRI characteristics (relative hippocampal volume) were compared using non-parametric (Mann-Whitney) or parametric (Student) tests for quantitative variables and Chi-square or Fisher exact tests for qualitative variables. RESULTS: The risk of conversion to dementia was lower for patients in the highest quartile of free plasma Aß1-42/Aß1-40 (≥ 25.8%) than those in the three lower quartiles: hazard ratio = 0.36 (95% confidence interval [0.15-0.87]), after adjustment for age, sex, education, and APOE ε4 (p-value = 0.022). This was comparable to the risk of conversion in the highest quartile of total plasma Aß1-42/Aß1-40: hazard ratio = 0.37 (95% confidence interval [0.16-0.89], p-value = 0.027). However, while patients in the highest quartile of total plasma Aß1-42/Aß1-40 showed higher MMSE scores and a higher hippocampal volume than patients in the three lowest quartiles of total plasma Aß1-42/Aß1-40, as well as normal CSF biomarker levels, the patients in the highest quartile of free plasma Aß1-42/Aß1-40 did not show any significant differences in MMSE scores, hippocampal volume, or CSF biomarker levels relative to the three lowest quartiles of free plasma Aß1-42/Aß1-40. CONCLUSION: The free plasma Aß1-42/Aß1-40 ratio is associated with a risk of conversion from MCI to dementia within three years, with performance comparable to that of the total plasma Aß1-42/Aß1-40 ratio. Threshold levels of the free and total plasma Aß1-42/Aß1-40 ratio could be determined, with a 60% lower risk of conversion for patients above the threshold than those below.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Humanos , Peptídeos beta-Amiloides/metabolismo , Progressão da Doença , Disfunção Cognitiva/diagnóstico , Biomarcadores , Proteínas Amiloidogênicas , Fragmentos de Peptídeos , Proteínas tau
2.
Neurobiol Dis ; 174: 105875, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36154878

RESUMO

Neurofibrillary tangles (NFT), a neuronal lesion found in Alzheimer's disease (AD), are composed of fibrillary aggregates of modified forms of tau proteins. The propagation of NFT follows neuroanatomical pathways suggesting that synaptically connected neurons could transmit tau pathology by the recruitment of normal tau in a prion-like manner. Moreover, the intracerebral injection of pathological tau from AD brains induces the seeding of normal tau in mouse brain. Creutzfeldt-Jacob disease has been transmitted after ocular transplants of cornea or sclera and the scrapie agent can spread across the retino-tectal pathway after intraocular injection of scrapie mouse brain homogenates. In AD, a tau pathology has been detected in the retina. To investigate the potential risk of tau pathology transmission during eye surgery using AD tissue material, we have analysed the development of tau pathology in the visual pathway of mice models expressing murine tau, wild-type or mutant human tau after intraocular injection of pathological tau proteins from AD brains. Although these pathological tau proteins were internalized in retinal ganglion cells, they did not induce aggregation of endogenous tau nor propagation of a tau pathology in the retino-tectal pathway after a 6-month incubation period. These results suggest that retinal ganglion cells exhibit a resistance to develop a tau pathology, and that eye surgery is not a major iatrogenic risk of transmission of tau pathology, contrary to what has been observed for transmission of infectious prions in prion diseases.


Assuntos
Doença de Alzheimer , Príons , Animais , Masculino , Camundongos , Humanos , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Emaranhados Neurofibrilares/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Príons/metabolismo , Células Ganglionares da Retina/metabolismo , Injeções Intraoculares , Camundongos Transgênicos
3.
J Intern Med ; 286(4): 398-437, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31286586

RESUMO

Alzheimer's disease (AD), the most frequent cause of dementia, is escalating as a global epidemic, and so far, there is neither cure nor treatment to alter its progression. The most important feature of the disease is neuronal death and loss of cognitive functions, caused probably from several pathological processes in the brain. The main neuropathological features of AD are widely described as amyloid beta (Aß) plaques and neurofibrillary tangles of the aggregated protein tau, which contribute to the disease. Nevertheless, AD brains suffer from a variety of alterations in function, such as energy metabolism, inflammation and synaptic activity. The latest decades have seen an explosion of genes and molecules that can be employed as targets aiming to improve brain physiology, which can result in preventive strategies for AD. Moreover, therapeutics using these targets can help AD brains to sustain function during the development of AD pathology. Here, we review broadly recent information for potential targets that can modify AD through diverse pharmacological and nonpharmacological approaches including gene therapy. We propose that AD could be tackled not only using combination therapies including Aß and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood-brain barrier targets that have been studied recently. We also make a case for the role of gut microbiota in AD. Our hope is to promote the continuing research of diverse targets affecting AD and promote diverse targeting as a near-future strategy.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Terapia de Alvo Molecular , Peptídeos beta-Amiloides , Terapia Baseada em Transplante de Células e Tecidos , Terapia Combinada , Terapia Genética , Humanos , Proteínas tau
4.
Mol Psychiatry ; 22(6): 874-883, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27113998

RESUMO

A recent genome-wide association meta-analysis for Alzheimer's disease (AD) identified 19 risk loci (in addition to APOE) in which the functional genes are unknown. Using Drosophila, we screened 296 constructs targeting orthologs of 54 candidate risk genes within these loci for their ability to modify Tau neurotoxicity by quantifying the size of >6000 eyes. Besides Drosophila Amph (ortholog of BIN1), which we previously implicated in Tau pathology, we identified p130CAS (CASS4), Eph (EPHA1), Fak (PTK2B) and Rab3-GEF (MADD) as Tau toxicity modulators. Of these, the focal adhesion kinase Fak behaved as a strong Tau toxicity suppressor in both the eye and an independent focal adhesion-related wing blister assay. Accordingly, the human Tau and PTK2B proteins biochemically interacted in vitro and PTK2B co-localized with hyperphosphorylated and oligomeric Tau in progressive pathological stages in the brains of AD patients and transgenic Tau mice. These data indicate that PTK2B acts as an early marker and in vivo modulator of Tau toxicity.


Assuntos
Quinase 2 de Adesão Focal/genética , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Animais , Biomarcadores , Modelos Animais de Doenças , Drosophila/genética , Quinase 2 de Adesão Focal/metabolismo , Loci Gênicos/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Fatores de Risco , Proteínas tau/genética
5.
Mol Psychiatry ; 21(1): 97-107, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450226

RESUMO

Consumption of caffeine, a non-selective adenosine A2A receptor (A2AR) antagonist, reduces the risk of developing Alzheimer's disease (AD) in humans and mitigates both amyloid and Tau burden in transgenic mouse models. However, the impact of selective A2AR blockade on the progressive development of AD-related lesions and associated memory impairments has not been investigated. In the present study, we removed the gene encoding A2AR from THY-Tau22 mice and analysed the subsequent effects on both pathological (Tau phosphorylation and aggregation, neuro-inflammation) and functional impairments (spatial learning and memory, hippocampal plasticity, neurotransmitter profile). We found that deleting A2ARs protect from Tau pathology-induced deficits in terms of spatial memory and hippocampal long-term depression. These effects were concomitant with a normalization of the hippocampal glutamate/gamma-amino butyric acid ratio, together with a global reduction in neuro-inflammatory markers and a decrease in Tau hyperphosphorylation. Additionally, oral therapy using a specific A2AR antagonist (MSX-3) significantly improved memory and reduced Tau hyperphosphorylation in THY-Tau22 mice. By showing that A2AR genetic or pharmacological blockade improves the pathological phenotype in a Tau transgenic mouse model, the present data highlight A2A receptors as important molecular targets to consider against AD and Tauopathies.


Assuntos
Transtornos Cognitivos/fisiopatologia , Hipocampo/fisiopatologia , Depressão Sináptica de Longo Prazo/fisiologia , Receptor A2A de Adenosina/metabolismo , Tauopatias/fisiopatologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Animais , Transtornos Cognitivos/tratamento farmacológico , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Humanos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Camundongos Transgênicos , Fosforilação , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/genética , Tauopatias/tratamento farmacológico , Técnicas de Cultura de Tecidos , Xantinas/farmacologia , Ácido gama-Aminobutírico/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Biochim Biophys Acta ; 1842(4): 654-64, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24440524

RESUMO

The splicing of the microtubule-associated protein Tau is regulated during development and is found to be deregulated in a growing number of pathological conditions such as myotonic dystrophy type I (DM1), in which a reduced number of isoforms is expressed in the adult brain. DM1 is caused by a dynamic and unstable CTG repeat expansion in the DMPK gene, resulting in an RNA bearing long CUG repeats (n>50) that accumulates in nuclear foci and sequesters CUG-binding splicing factors of the muscle blind-like (MBNL) family, involved in the splicing of Tau pre-mRNA among others. However, the precise mechanism leading to Tau mis-splicing and the role of MBNL splicing factors in this process are poorly understood. We therefore used new Tau minigenes that we developed for this purpose to determine how MBNL1 and MBNL2 interact to regulate Tau exon 2 splicing. We demonstrate that an intronic region 250 nucleotides downstream of Tau exon 2 contains cis-regulatory splicing enhancers that are sensitive to MBNL and that bind directly to MBNL1. Both MBNL1 and MBNL2 act as enhancers of Tau exon 2 inclusion. Intriguingly, the interaction of MBNL1 and MBNL2 is required to fully reverse the mis-splicing of Tau exon 2 induced by the trans-dominant effect of long CUG repeats, similar to the DM1 condition. In conclusion, both MBNL1 and MBNL2 are involved in the regulation of Tau exon 2 splicing and the mis-splicing of Tau in DM1 is due to the combined inactivation of both.


Assuntos
Éxons , Distrofia Miotônica/genética , Proteínas de Ligação a RNA/fisiologia , Elementos de Resposta , Proteínas tau/genética , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Dados de Sequência Molecular , Splicing de RNA
7.
Neuropathol Appl Neurobiol ; 41(1): 59-80, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25428793

RESUMO

Our aims are to review animal models of tauopathies, which include a number of brain disorders with various aetiologies, including ageing, genetics, infectious diseases, toxins, trauma and other unknown factors. Tauopathies are characterized by the accumulation of filaments of the microtubule-associated tau protein. The different aetiopathogeneses and distinct molecular events involved in tau aggregation have led to the development of various animal models for these diseases. In this review, rather than listing all current models, we focus on specific animal models addressing, among others, the question of tau hyperphosphorylation, tau aggregation and tau spreading. Physiological conditions, including normal ageing and hibernation, may exhibit tau phosphorylation and some aspects of tauopathies. However, most of the models of tauopathies involve genetically modified animals (mostly rodents, but also fruit fly, zebrafish and worm). Some of these models have been crucial for the development of therapeutic approaches in humans. The present review shows the difficulty in pinpointing a specific mechanism that may be targeted in tauopathies but also opens up new avenues for innovative therapeutic strategies.


Assuntos
Modelos Animais de Doenças , Tauopatias/etiologia , Tauopatias/metabolismo , Pesquisa Translacional Biomédica , Proteínas tau/fisiologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Animais Geneticamente Modificados , Encéfalo/patologia , Morte Celular , Humanos , Corpos de Inclusão/patologia , Neurônios/patologia , Proteínas tau/metabolismo
8.
Mol Psychiatry ; 19(12): 1326-35, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24535457

RESUMO

Amyloid beta (Aß) peptides are the major components of senile plaques, one of the main pathological hallmarks of Alzheimer disease (AD). However, Aß peptides' functions are not fully understood and seem to be highly pleiotropic. We hypothesized that plasma Aß peptides concentrations could be a suitable endophenotype for a genome-wide association study (GWAS) designed to (i) identify novel genetic factors involved in amyloid precursor protein metabolism and (ii) highlight relevant Aß-related physiological and pathophysiological processes. Hence, we performed a genome-wide association meta-analysis of four studies totaling 3 528 healthy individuals of European descent and for whom plasma Aß1-40 and Aß1-42 peptides levels had been quantified. Although we did not observe any genome-wide significant locus, we identified 18 suggestive loci (P<1 × 10(-)(5)). Enrichment-pathway analyses revealed canonical pathways mainly involved in neuronal functions, for example, axonal guidance signaling. We also assessed the biological impact of the gene most strongly associated with plasma Aß1-42 levels (cortexin 3, CTXN3) on APP metabolism in vitro and found that the gene protein was able to modulate Aß1-42 secretion. In conclusion, our study results suggest that plasma Aß peptides levels are valid endophenotypes in GWASs and can be used to characterize the metabolism and functions of APP and its metabolites.


Assuntos
Envelhecimento/sangue , Envelhecimento/genética , Peptídeos beta-Amiloides/sangue , Fragmentos de Peptídeos/sangue , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Polimorfismo de Nucleotídeo Único , População Branca/genética
9.
Morphologie ; 99(327): 141-8, 2015 Dec.
Artigo em Francês | MEDLINE | ID: mdl-25908520

RESUMO

The Tau protein (Tubulin Associated Unit) is a phosphoprotein of the microtubule-associated protein family (MAPs). Its role is the regulation of the microtubule polymerization. The Tau protein is naturally present in brain, heart, muscle, lung, kidney, pancreas and liver. An expression of Tau protein and RNA messengers was also highlighted in the testis that is an organ rich in microtubules. The role of microtubules is essential in the stabilization of the cellular shape and in cell divisions. In the testis, Tau protein could be involved in the division process of the spermatogenesis by acting on the microtubular dynamics in the arrangement of the spermatozoon polarity. This review synthesizes the current knowledge, the localization and the main functions of the Tau protein focused on the testis. The localization and the potential roles of the Tau protein during the spermatogenesis are discussed by emphasizing the link with the microtubular structures of seminiferous tubules.


Assuntos
Microtúbulos/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Testículo/metabolismo , Proteínas tau/metabolismo , Divisão Celular , Polaridade Celular , Forma Celular , Humanos , Masculino , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Polimerização , Túbulos Seminíferos/metabolismo , Proteínas tau/genética
10.
Mol Psychiatry ; 18(11): 1225-34, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23399914

RESUMO

Genome-wide association studies (GWAS) have identified a region upstream the BIN1 gene as the most important genetic susceptibility locus in Alzheimer's disease (AD) after APOE. We report that BIN1 transcript levels were increased in AD brains and identified a novel 3 bp insertion allele ∼28 kb upstream of BIN1, which increased (i) transcriptional activity in vitro, (ii) BIN1 expression levels in human brain and (iii) AD risk in three independent case-control cohorts (Meta-analysed Odds ratio of 1.20 (1.14-1.26) (P=3.8 × 10(-11))). Interestingly, decreased expression of the Drosophila BIN1 ortholog Amph suppressed Tau-mediated neurotoxicity in three different assays. Accordingly, Tau and BIN1 colocalized and interacted in human neuroblastoma cells and in mouse brain. Finally, the 3 bp insertion was associated with Tau but not Amyloid loads in AD brains. We propose that BIN1 mediates AD risk by modulating Tau pathology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Predisposição Genética para Doença/genética , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética , Proteínas tau/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Endofenótipos , Expressão Gênica/genética , Humanos , Camundongos , Degeneração Neural/genética , Degeneração Neural/patologia , Proteínas Nucleares/biossíntese , Placa Amiloide/patologia , Polimorfismo de Nucleotídeo Único/genética , Sinaptossomos/patologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Proteínas tau/antagonistas & inibidores
11.
Gene Ther ; 20(3): 283-97, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22592599

RESUMO

Glioblastomas are the most aggressive of the brain tumors occurring in adults and children. Currently available chemotherapy prolongs the median survival time of patients by only 4 months. The low efficiency of current treatments is partly owing to the blood-brain barrier, which restricts the penetration of most drugs into the central nervous system. Locoregional treatment strategies thus become mandatory. In this context, viral tools are of great interest for the selective delivery of genes into tumoral cells. Gliomas express high levels of type 2 somatostatin receptors (sstr2A), pinpointing them as suitable targets for the improvement of transduction efficiency in these tumors. We designed a new adenoviral vector based on the introduction of the full-length somatostatin (SRIF (somatotropin release-inhibiting factor)) sequence into the HI loop of the HAdV fiber protein. We demonstrate that (i) HAdV-5-SRIF uptake into cells is mediated by sstr2A, (ii) our vector drives high levels of gene expression in cells expressing endogenous sstr2A, with up to 65-fold enhancement and (iii) low doses of HAdV-5-SRIF are sufficient to infect high-grade human primary glioblastoma cells. Adenoviral vectors targeting SRIF receptors might thus represent a promising therapeutic approach to brain tumors.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Receptores de Somatostatina/genética , Transdução Genética/métodos , Adenoviridae/metabolismo , Sequência de Aminoácidos , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Células CHO , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Cricetinae , Cricetulus , Endocitose , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/farmacocinética , Glioblastoma/patologia , Glioblastoma/terapia , Células HEK293 , Humanos , Immunoblotting , Integrinas/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Células Tumorais Cultivadas
12.
Biochim Biophys Acta ; 1812(7): 732-42, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21439371

RESUMO

Tau is the proteinaceous component of intraneuronal aggregates common to neurodegenerative diseases called Tauopathies, including myotonic dystrophy type 1. In myotonic dystrophy type 1, the presence of microtubule-associated protein Tau aggregates is associated with a mis-splicing of Tau. A toxic gain-of-function at the ribonucleic acid level is a major etiological factor responsible for the mis-splicing of several transcripts in myotonic dystrophy type 1. These are probably the consequence of a loss of muscleblind-like 1 (MBNL1) function or gain of CUGBP1 and ETR3-like factor 1 (CELF1) splicing function. Whether these two dysfunctions occur together or separately and whether all mis-splicing events in myotonic dystrophy type 1 brain result from one or both of these dysfunctions remains unknown. Here, we analyzed the splicing of Tau exons 2 and 10 in the brain of myotonic dystrophy type 1 patients. Two myotonic dystrophy type 1 patients showed a mis-splicing of exon 10 whereas exon 2-inclusion was reduced in all myotonic dystrophy type 1 patients. In order to determine the potential factors responsible for exon 10 mis-splicing, we studied the effect of the splicing factors muscleblind-like 1 (MBNL1), CUGBP1 and ETR3-like factor 1 (CELF1), CUGBP1 and ETR3-like factor 2 (CELF2), and CUGBP1 and ETR3-like factor 4 (CELF4) or a dominant-negative CUGBP1 and ETR-3 like factor (CELF) factor on Tau exon 10 splicing by ectopic expression or siRNA. Interestingly, the inclusion of Tau exon 10 is reduced by CUGBP1 and ETR3-like factor 2 (CELF2) whereas it is insensitive to the loss-of-function of muscleblind-like 1 (MBNL1), CUGBP1 and ETR3-like factor 1 (CELF1) gain-of-function, or a dominant-negative of CUGBP1 and ETR-3 like factor (CELF) factor. Moreover, we observed an increased expression of CUGBP1 and ETR3-like factor 2 (CELF2) only in the brain of myotonic dystrophy type 1 patients with a mis-splicing of exon 10. Taken together, our results indicate the occurrence of a mis-splicing event in myotonic dystrophy type 1 that is induced neither by a loss of muscleblind-like 1 (MBNL1) function nor by a gain of CUGBP1 and ETR3-like factor 1 (CELF1) function but is rather associated to CUGBP1 and ETR3-like factor 2 (CELF2) gain-of-function.


Assuntos
Éxons , Inativação Gênica , Proteínas do Tecido Nervoso/genética , Proteínas de Ligação a RNA/genética , Proteínas tau/genética , Sequência de Bases , Encéfalo/metabolismo , Proteínas CELF , Primers do DNA , Humanos , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas tau/metabolismo
14.
Mol Psychiatry ; 14(1): 106-16, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17893704

RESUMO

To more rapidly identify candidate genes located within chromosomal regions of interest defined by genome scan studies in Alzheimer's disease (AD), we have developed a customized microarray containing all the ORFs (n=2741) located within nine of these regions. Levels of gene expression were assessed in total RNA from brain tissue of 12 controls and 12 AD patients. Of all genes showing differential expression, we focused on the ornithine transcarbamylase (OTC) gene on Xp21.1., a key enzyme of the urea cycle which we found to be expressed in AD brains but not in controls, as confirmed by RT-PCR. We also detected mRNA expression of all the other urea cycle enzymes in AD brains. Immunochemistry experiments revealed that the OTC expression was strictly restricted to vascular endothelial cells in brain. Furthermore, OTC activity was 880% increased in the CSF of probable AD cases compared with controls. We analysed the association of the OTC -389 G/A and -241 A/G promoter polymorphisms with the risk of developing AD. We observed that rare haplotypes may be associated with the risk of AD through a possible modulation of the methylation of the OTC promoter. In conclusion, our results suggest the involvement of a new pathway in AD brains involving the urea cycle.


Assuntos
Doença de Alzheimer/enzimologia , Expressão Gênica/fisiologia , Ornitina Carbamoiltransferase/metabolismo , Polimorfismo de Nucleotídeo Único , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Encéfalo/metabolismo , Análise Mutacional de DNA/métodos , Feminino , Genótipo , Humanos , Masculino , Análise em Microsséries/métodos , Ornitina Carbamoiltransferase/genética , Fatores Sexuais
15.
Mol Psychiatry ; 14(11): 1004-16, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19204726

RESUMO

The only recognized genetic determinant of the common forms of Alzheimer's disease (AD) is the epsilon 4 allele of the apolipoprotein E gene (APOE). To identify new candidate genes, we recently performed transcriptomic analysis of 2741 genes in chromosomal regions of interest using brain tissue of AD cases and controls. From 82 differentially expressed genes, 1156 polymorphisms were genotyped in two independent discovery subsamples (n=945). Seventeen genes exhibited at least one polymorphism associated with AD risk, and following correction for multiple testing, we retained the interleukin (IL)-33 gene. We first confirmed that the IL-33 expression was decreased in the brain of AD cases compared with that of controls. Further genetic analysis led us to select three polymorphisms within this gene, which we analyzed in three independent case-control studies. These polymorphisms and a resulting protective haplotype were systematically associated with AD risk in non-APOE epsilon 4 carriers. Using a large prospective study, these associations were also detected when analyzing the prevalent and incident AD cases together or the incident AD cases alone. These polymorphisms were also associated with less cerebral amyloid angiopathy (CAA) in the brain of non-APOE epsilon 4 AD cases. Immunohistochemistry experiments finally indicated that the IL-33 expression was consistently restricted to vascular capillaries in the brain. Moreover, IL-33 overexpression in cellular models led to a specific decrease in secretion of the A beta(40) peptides, the main CAA component. In conclusion, our data suggest that genetic variants in IL-33 gene may be associated with a decrease in AD risk potentially in modulating CAA formation.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Interleucinas/genética , Interleucinas/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Apolipoproteína E4/genética , Encéfalo/metabolismo , Células COS , Estudos de Casos e Controles , Linhagem Celular Transformada , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Chlorocebus aethiops , Feminino , Seguimentos , Carga Genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Interleucina-33 , Cooperação Internacional , Masculino , Neuroblastoma , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fragmentos de Peptídeos/metabolismo , Polimorfismo de Nucleotídeo Único , Modelos de Riscos Proporcionais , RNA Mensageiro/metabolismo , Estudos Retrospectivos , Transfecção/métodos
16.
Ann Pharm Fr ; 67(2): 136-53, 2009 Mar.
Artigo em Francês | MEDLINE | ID: mdl-19298897

RESUMO

What is an innovative therapeutics for the Alzheimer's disease? An already used therapeutics which appeals to a recent and innovative concept or a therapeutic still putative based on tracks turned out experimentally but which still ask to be supported by man? Some therapeutic used at present are based on often former observations (anti-acetylcholinesterasic strategy) or more recent (antiglutamatergic strategy) but cannot be really considered as therapeutic innovative. They will be reviewed thus quickly because treated somewhere else. Potentially innovative therapeutics arise from recent headways and are there often only because of their stammerings. If the biology of Tau proteins is well-known, its therapeutic approach is little developed. On the contrary, therapeutics approaches turns essentially around the peptide amyloid, whether its training or the cellular consequences of its overproduction. This article is centred on the various therapeutic approaches which we can prospectively propose and which are very promising for some and for the others, collide with abstract or theoretical problems which will be approached here.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/biossíntese , Animais , Antioxidantes/uso terapêutico , Inibidores da Colinesterase/uso terapêutico , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Humanos , Imunoterapia , Fármacos Neuroprotetores/uso terapêutico , Proteínas tau/antagonistas & inibidores , Proteínas tau/biossíntese
17.
Biochim Biophys Acta ; 1773(9): 1428-37, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17624454

RESUMO

The peptidyl prolyl cis-trans isomerase Pin1 and the Inhibitor of Apoptosis Protein (IAP) Survivin are two major proteins involved in cancer. They both modulate apoptosis, mitosis, centrosome duplication and neuronal development but until now no functional relationship has been reported between these two proteins. We tested Pin1-induced regulation of Survivin in neuroblastoma cells. Pin1 overexpression in SY5Y neuroblastoma cells decreased Survivin levels. Immunocytochemical studies indicated that they partially co-localized in interphase and mitotic cells. Co-immunoprecipitation further demonstrates the existence of a Pin1/Survivin complex. Pin1-induced effect on Survivin was confirmed in COS cells. RT-PCR and mutagenesis experiments suggested that this Pin1-induced decrease of Survivin occurred at the protein level. Survivin downregulation depended on the binding ability of Pin1 but was not related to the single Thr-Pro site, suggesting an indirect relationship into a protein complex. Finally, this functional regulation of Survivin by Pin1 is reciprocal since Pin1 silencing led to an increase in Survivin levels. The characterization of this functional relationship between Pin1 and Survivin might help to better understand mitosis control and cancer mechanisms.


Assuntos
Regulação para Baixo , Proteínas Inibidoras de Apoptose/metabolismo , Peptidilprolil Isomerase/metabolismo , Animais , Sítios de Ligação , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , DNA Complementar , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Mutação , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Peptidilprolil Isomerase/genética , Testes de Precipitina , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
18.
Andrology ; 6(6): 854-859, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29984477

RESUMO

BACKGROUND: A-kinase anchor protein 4 (AKAP4) and its precursor pro-AKAP4 are two major proteins in spermatozoa of rodents and mammals. Although researchers have characterized the AKAP4 expression in various species, the protein's expression in humans has not been described in detail. OBJECTIVES: The objective of this study was to characterize human pro-AKAP4 more precisely (notably the definition of its localization and expression levels in human spermatozoa and testes). MATERIALS AND METHODS: pro-AKAP4 protein expression levels were assessed by Western blotting. The pro-AKAP4's localization in spermatozoa and testes was determined using immunofluorescence staining and immunogold electron microscopy. Furthermore, pro-AKAP4 protein expression levels were assessed in a series of 77 human semen samples, and associations with semen parameters were evaluated. RESULTS: Western blotting revealed a 100-kDa band in human sperm protein extracts. The pro-AKAP4 was immunolocalized in the fibrous sheath of the flagellum of ejaculated spermatozoa and in elongated spermatids in human testes. A Western blot analysis of 77 normozoospermic semen samples evidenced striking differences in pro-AKAP4 levels from one to another sample (median [interquartile range] integrated optical density = 305 [49-1038]). No correlations were found for pro-AKAP4 levels on one hand and semen volume, sperm concentration, sperm count, sperm motility, or sperm morphology on the other (p > 0.05 for all). However, pro-AKAP4 levels were positively correlated with motility after density gradient centrifugation of the semen (r = 0.224, p = 0.049). DISCUSSION: AKAP4 protein might be activated as an alternative pathway to rescue sperm motility. In human spermatozoa, pro-AKAP4 might therefore be a 'reservoir' of mature AKAP4. CONCLUSION: This study generated new knowledge about pro-AKAP4 in human semen, which may be of interest in the management of male infertility.


Assuntos
Proteínas de Ancoragem à Quinase A/análise , Precursores de Proteínas/análise , Espermatozoides/química , Biomarcadores/análise , Western Blotting , Forma Celular , Centrifugação com Gradiente de Concentração , Imunofluorescência , Humanos , Masculino , Microscopia Imunoeletrônica , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Espermatozoides/ultraestrutura
19.
Curr Protein Pept Sci ; 7(3): 179-94, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16787258

RESUMO

PIN1 participates in the regulation of a number of signalling pathways in the cell involving protein phosphorylation/dephosphorylation. Its role seems to be an essential control level in addition to the protein phosphorylation by proline-directed kinases. Its cellular function includes regulation of the cell cycle by interaction with phosphorylated mitotic proteins such as Cdc25 and transcription factors such as p53. PIN1 was shown to be involved in the malignant transformation of cells in breast cancer, by up regulation of cyclinD1 and is thought to be involved in the development of the AD by regulating the function of phosphorylated Tau. We propose here to discuss the molecular function of PIN1 at the atomic level based on data from the recent literature and our own results obtained by the technique of Nuclear Magnetic Resonance. PIN1 specifically interacts with pThr/pSer-Pro motifs and is constituted by two domains: a WW N-terminal domain that binds pThr/pSer-Pro epitopes and a prolyl cis/trans isomerase C-terminal catalytic domain. An exception to this organisation is found in the plant PIN1 homologous enzymes, like PIN1At from Arabidopsis thaliana, that are constituted of the sole catalytic domain. The molecular function of PIN1, binding to and isomerization of pThr/pSer-Pro bonds, are thought to lead to several functional consequences. In a first mode of action, exemplified by its competition with the CKS protein, the interaction with PIN1 prevents interaction with other regulatory proteins, like ubiquitin-ligases that lead to degradation pathways. In a second mode of action, the idea is largely accepted that the local isomerization modifies the global conformation of the protein substrate and hence its intrinsic activity, although this has never been directly demonstrated. Finally, isomerization catalysis is thought to regulate the (de)phosphorylation of specific pThr/pSer-Pro motifs, exemplified by the stimulation of the dephosphorylation of pThr231 of Tau by the PP2A phosphatase.


Assuntos
Peptidilprolil Isomerase/química , Peptidilprolil Isomerase/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Arabidopsis , Sítios de Ligação , Domínio Catalítico , Humanos , Dados de Sequência Molecular , Peptidilprolil Isomerase de Interação com NIMA , Ressonância Magnética Nuclear Biomolecular , Peptidilprolil Isomerase/genética , Fosforilação , Conformação Proteica , Alinhamento de Sequência , Transdução de Sinais , Relação Estrutura-Atividade , Especificidade por Substrato
20.
FEBS Lett ; 579(1): 1-5, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15620682

RESUMO

Increasing evidence suggests that an inhibition of the proteasome, as demonstrated in Parkinson's disease, might be involved in Alzheimer's disease. In this disease and other Tauopathies, Tau proteins are hyperphosphorylated and aggregated within degenerating neurons. In this state, Tau is also ubiquitinated, suggesting that the proteasome might be involved in Tau proteolysis. Thus, to investigate if proteasome inhibition leads to accumulation, hyperphosphorylation and aggregation of Tau, we used neuroblastoma cells overexpressing Tau proteins. Surprisingly, we showed that the inhibition of the proteasome led to a bidirectional degradation of Tau. Following this result, the cellular mechanisms that may degrade Tau were investigated.


Assuntos
Doença de Alzheimer/metabolismo , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas tau/metabolismo , Anticorpos Fosfo-Específicos/imunologia , Caspases/análise , Caspases/metabolismo , Extratos Celulares/química , Linhagem Celular Tumoral , Humanos , Leupeptinas/farmacologia , Fosforilação , Poli(ADP-Ribose) Polimerases/análise , Poli(ADP-Ribose) Polimerases/metabolismo , Complexo de Endopeptidases do Proteassoma/análise , Inibidores de Proteassoma , Proteínas tau/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA