Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 72
1.
Neurobiol Dis ; 193: 106459, 2024 Apr.
Article En | MEDLINE | ID: mdl-38423192

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.


Alzheimer Disease , Cognitive Dysfunction , Humans , Amyloid beta-Peptides/metabolism , Disease Progression , Cognitive Dysfunction/diagnosis , Biomarkers , Amyloidogenic Proteins , Peptide Fragments , tau Proteins
2.
Neurobiol Dis ; 174: 105875, 2022 Nov.
Article En | MEDLINE | ID: mdl-36154878

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.


Alzheimer Disease , Prions , Animals , Male , Mice , Humans , tau Proteins/metabolism , Alzheimer Disease/metabolism , Neurofibrillary Tangles/metabolism , Brain/metabolism , Disease Models, Animal , Prions/metabolism , Retinal Ganglion Cells/metabolism , Injections, Intraocular , Mice, Transgenic
3.
J Intern Med ; 286(4): 398-437, 2019 10.
Article En | MEDLINE | ID: mdl-31286586

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.


Alzheimer Disease/drug therapy , Molecular Targeted Therapy , Amyloid beta-Peptides , Cell- and Tissue-Based Therapy , Combined Modality Therapy , Genetic Therapy , Humans , tau Proteins
4.
Andrology ; 6(6): 854-859, 2018 11.
Article En | MEDLINE | ID: mdl-29984477

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.


A Kinase Anchor Proteins/analysis , Protein Precursors/analysis , Spermatozoa/chemistry , Biomarkers/analysis , Blotting, Western , Cell Shape , Centrifugation, Density Gradient , Fluorescent Antibody Technique , Humans , Male , Microscopy, Immunoelectron , Sperm Count , Sperm Motility , Spermatozoa/ultrastructure
5.
Mol Psychiatry ; 22(6): 874-883, 2017 06.
Article En | MEDLINE | ID: mdl-27113998

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.


Focal Adhesion Kinase 2/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Animals , Biomarkers , Disease Models, Animal , Drosophila/genetics , Focal Adhesion Kinase 2/metabolism , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Risk Factors , tau Proteins/genetics
6.
Mol Psychiatry ; 21(1): 97-107, 2016 Jan.
Article En | MEDLINE | ID: mdl-25450226

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.


Cognition Disorders/physiopathology , Hippocampus/physiopathology , Long-Term Synaptic Depression/physiology , Receptor, Adenosine A2A/metabolism , Tauopathies/physiopathology , Adenosine A2 Receptor Antagonists/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Cognition Disorders/drug therapy , Disease Models, Animal , Glutamic Acid/metabolism , Hippocampus/drug effects , Humans , Long-Term Synaptic Depression/drug effects , Mice, Transgenic , Phosphorylation , RNA, Messenger/metabolism , Receptor, Adenosine A2A/genetics , Tauopathies/drug therapy , Tissue Culture Techniques , Xanthines/pharmacology , gamma-Aminobutyric Acid/metabolism , tau Proteins/genetics , tau Proteins/metabolism
8.
Morphologie ; 99(327): 141-8, 2015 Dec.
Article Fr | MEDLINE | ID: mdl-25908520

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.


Microtubules/metabolism , Spermatogenesis , Spermatozoa/metabolism , Testis/metabolism , tau Proteins/metabolism , Cell Division , Cell Polarity , Cell Shape , Humans , Male , Phosphoproteins/metabolism , Phosphoproteins/physiology , Polymerization , Seminiferous Tubules/metabolism , tau Proteins/genetics
9.
Neuropathol Appl Neurobiol ; 41(1): 59-80, 2015 Feb.
Article En | MEDLINE | ID: mdl-25428793

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.


Disease Models, Animal , Tauopathies/etiology , Tauopathies/metabolism , Translational Research, Biomedical , tau Proteins/physiology , Aging/metabolism , Aging/pathology , Animals , Animals, Genetically Modified , Brain/pathology , Cell Death , Humans , Inclusion Bodies/pathology , Neurons/pathology , tau Proteins/metabolism
10.
Mol Psychiatry ; 19(12): 1326-35, 2014 Dec.
Article En | MEDLINE | ID: mdl-24535457

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.


Aging/blood , Aging/genetics , Amyloid beta-Peptides/blood , Peptide Fragments/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Genome-Wide Association Study , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Polymorphism, Single Nucleotide , White People/genetics
11.
Biochim Biophys Acta ; 1842(4): 654-64, 2014 Apr.
Article En | MEDLINE | ID: mdl-24440524

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.


Exons , Myotonic Dystrophy/genetics , RNA-Binding Proteins/physiology , Response Elements , tau Proteins/genetics , Base Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , RNA Splicing
12.
Mol Psychiatry ; 18(11): 1225-34, 2013 Nov.
Article En | MEDLINE | ID: mdl-23399914

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.


Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Genetic Predisposition to Disease/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , tau Proteins/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Endophenotypes , Gene Expression/genetics , Humans , Mice , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nuclear Proteins/biosynthesis , Plaque, Amyloid/pathology , Polymorphism, Single Nucleotide/genetics , Synaptosomes/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/biosynthesis , tau Proteins/antagonists & inhibitors
13.
Gene Ther ; 20(3): 283-97, 2013 Mar.
Article En | MEDLINE | ID: mdl-22592599

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.


Adenoviridae/genetics , Brain Neoplasms/genetics , Glioblastoma/genetics , Receptors, Somatostatin/genetics , Transduction, Genetic/methods , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , CHO Cells , Capsid Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Cricetinae , Cricetulus , Endocytosis , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Glioblastoma/pathology , Glioblastoma/therapy , HEK293 Cells , Humans , Immunoblotting , Integrins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Receptors, Somatostatin/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Tumor Cells, Cultured
14.
Curr Alzheimer Res ; 8(6): 633-8, 2011 Sep.
Article En | MEDLINE | ID: mdl-21605043

Alzheimer's disease (AD) is a neurodegenerative disorder histologically defined by the cerebral accumulation of amyloid deposits and neurofibrillary tangles composed of hyperphosphorylated tau proteins. Loss of basal forebrain cholinergic neurons is another hallmark of the disease thought to contribute to the cognitive dysfunctions. To this date, the mechanisms underlying cholinergic neurons degeneration remain uncertain. The present study aimed to investigate the relationship between neurofibrillary degeneration and cholinergic defects in AD using THY-Tau22 transgenic mouse model exhibiting a major hippocampal AD-like tau pathology and hyperphosphorylated tau species in the septohippocampal pathway. Here, we report that at a time THY-Tau22 mice display strong reference memory alterations, the retrograde transport of fluorogold through the septohippocampal pathway is altered. This impairment is associated with a significant reduction in the number of choline acetyltransferase (ChAT)-immunopositive cholinergic neurons in the medial septum. Analysis of nerve growth factor (NGF) levels supports an accumulation of the mature neurotrophin in the hippocampus of THY-Tau22 mice, consistent with a decrease of its uptake or retrograde transport by cholinergic terminals. Finally, our data strongly support that tau pathology could be instrumental in the cholinergic neuronal loss observed in AD.


Brain/pathology , Cholinergic Neurons/pathology , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Animals , Brain/metabolism , Cholinergic Neurons/metabolism , Maze Learning/physiology , Memory/physiology , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurons/metabolism , Neurons/pathology , tau Proteins/genetics
15.
Biochim Biophys Acta ; 1812(7): 732-42, 2011 Jul.
Article En | MEDLINE | ID: mdl-21439371

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.


Exons , Gene Silencing , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , tau Proteins/genetics , Base Sequence , Brain/metabolism , CELF Proteins , DNA Primers , Humans , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , tau Proteins/metabolism
16.
Neurology ; 74(1): 42-9, 2010 Jan 05.
Article En | MEDLINE | ID: mdl-19940270

OBJECTIVE: Frontotemporal lobar degeneration (FTLD) encompasses a variety of clinicopathologic entities. The antemortem prediction of the underlying pathologic lesions is reputed to be difficult. This study sought to characterize correlations between 1) the different clinical variants of primary progressive language and speech disorders and 2) the pathologic diagnosis. METHODS: The latter was available for 18 patients having been prospectively monitored in the Lille Memory Clinic (France) between 1993 and 2008. RESULTS: The patients were diagnosed with progressive anarthria (n = 5), agrammatic progressive aphasia (n = 6), logopenic progressive aphasia (n = 1), progressive jargon aphasia (n = 2), typical semantic dementia (n = 2), and atypical semantic dementia (n = 2). All patients with progressive anarthria had a tau pathology at postmortem evaluation: progressive supranuclear palsy (n = 2), Pick disease (n = 2), and corticobasal degeneration (n = 1). All patients with agrammatic primary progressive aphasia had TDP-43-positive FTLD (FTLD-TDP). The patients with logopenic progressive aphasia and progressive jargon aphasia had Alzheimer disease. Both cases of typical semantic dementia had FTLD-TDP. The patients with atypical semantic dementia had tau pathologies: argyrophilic grain disease and corticobasal degeneration. CONCLUSIONS: The different anatomic distribution of the pathologic lesions could explain these results: opercular and subcortical regions in tau pathologies with progressive anarthria, the left frontotemporal cortex in TDP-43-positive frontotemporal lobar degeneration (FTLD-TDP) with agrammatic progressive aphasia, the bilateral lateral and anterior temporal cortex in FTLD-TDP or argyrophilic grain disease with semantic dementia, and the left parietotemporal cortex in Alzheimer disease with logopenic progressive aphasia or jargon aphasia. These correlations have to be confirmed in larger series.


Aphasia, Primary Progressive/pathology , Aphasia, Primary Progressive/physiopathology , Brain/pathology , Speech Disorders/pathology , Aged , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Aphasia, Primary Progressive/classification , Brain Mapping , DNA-Binding Proteins/metabolism , Disease Progression , Female , Frontotemporal Lobar Degeneration/complications , Frontotemporal Lobar Degeneration/pathology , Humans , Language Tests , Male , Middle Aged , Neuropsychological Tests , Postmortem Changes , Predictive Value of Tests , Prions/metabolism , Prospective Studies , Retrospective Studies , Speech Disorders/physiopathology , Statistics as Topic , Tomography Scanners, X-Ray Computed , Tomography, Emission-Computed, Single-Photon/methods , alpha-Synuclein/metabolism , tau Proteins/metabolism
17.
Exp Neurol ; 223(2): 432-8, 2010 Jun.
Article En | MEDLINE | ID: mdl-20005225

Newly proposed diagnostic criteria for Alzheimer's disease include cerebrospinal fluid (CSF) tau levels as one core supportive criterion. The published high sensitivity and specificity figures for CSF tau levels in Alzheimer's disease are offset by the large range of proposed cutoff values (9.6 pg/mL to 1140 pg/mL). This study aimed to provide guidance on how to establish, validate and audit CSF tau cutoff values using an unbiased, two-stage multicentre strategy. Both receiver operator characteristics (ROC) optimised and population-based cutoff values were calculated on a pilot dataset (n=99), validated in a large dataset (n=560) and then compared to the literature. The data suggest using an ROC optimised cutoff level of 323+/-51.7 pg/mL allowing for the published inter-laboratory coefficient of variation of 16%. This cutoff level was confirmed in a prospective audit (n=100). As demand for CSF tau levels will increase globally, the accuracy of local CSF hTau cutoff levels can be compared against this benchmark.


Alzheimer Disease/diagnosis , Chemistry, Clinical/standards , tau Proteins/cerebrospinal fluid , Aged , Alzheimer Disease/cerebrospinal fluid , Benchmarking , Chemistry, Clinical/methods , Female , Humans , Male , Middle Aged , Quality Control , ROC Curve , Reference Values , Reproducibility of Results , Sensitivity and Specificity
18.
Neurology ; 73(11): 847-53, 2009 Sep 15.
Article En | MEDLINE | ID: mdl-19752451

OBJECTIVE: Several lines of evidence indicate that a decrease in the CSF concentration of amyloid beta(42) (Abeta(42)) is a potential biomarker for incident Alzheimer disease. In contrast, studies on plasma Abeta(1-40) and Abeta(1-42) peptide levels have yielded contradictory results. Here, we explored the links between incident dementia and plasma Abeta(1-40) and Abeta(1-42) peptide concentrations in the prospective, population-based Three-City (3C) Study. We also assessed the association between plasma concentrations of truncated Abeta (Abeta(n-40) and Abeta(n-42)) and the risk of dementia. METHODS: During a subsequent 4-year follow-up period, 257 individuals presented incident dementia from 8,414 participants, and a subcohort of 1,185 individuals without dementia was drawn as a control cohort. Plasma levels of Abeta(1-40), Abeta(1-42), Abeta(n-40), and Abeta(n-42) were measured using an xMAP-based assay technology. The association between plasma Abeta peptide levels and the risk of dementia was assessed using Cox proportional hazard models. RESULTS: Of the various Abeta variables analyzed, the Abeta(1-42)/Abeta(1-40) and Abeta(n-42)/Abeta(n-40) ratios presented the strongest association with the risk of dementia: people with a high Abeta(1-42)/Abeta(1-40) or Abeta(n-42)/Abeta(n-40) ratio had a lower risk of developing dementia. These associations were restricted to individuals diagnosed at 2 years of follow-up and the Abeta(n-42)/Abeta(n-40) ratio was mainly associated with the risk of mixed/vascular dementia. CONCLUSION: Plasma Abeta peptide concentrations and Abeta(1-42)/Abeta(1-40) and Abeta(n-42)/Abeta(n-40) ratios may be useful markers to indicate individuals susceptible to short-term risk of dementia.


Amyloid beta-Peptides/blood , Dementia/blood , Peptide Fragments/blood , Aged , Aged, 80 and over , Dementia/diagnosis , Female , France , Humans , Male , Prospective Studies , Reagent Kits, Diagnostic
20.
Ann Pharm Fr ; 67(2): 136-53, 2009 Mar.
Article Fr | MEDLINE | ID: mdl-19298897

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.


Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/biosynthesis , Animals , Antioxidants/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Immunotherapy , Neuroprotective Agents/therapeutic use , tau Proteins/antagonists & inhibitors , tau Proteins/biosynthesis
...