Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Nat Neurosci ; 27(4): 643-655, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38424324

RÉSUMÉ

Dipeptide repeat proteins are a major pathogenic feature of C9orf72 amyotrophic lateral sclerosis (C9ALS)/frontotemporal dementia (FTD) pathology, but their physiological impact has yet to be fully determined. Here we generated C9orf72 dipeptide repeat knock-in mouse models characterized by expression of 400 codon-optimized polyGR or polyPR repeats, and heterozygous C9orf72 reduction. (GR)400 and (PR)400 knock-in mice recapitulate key features of C9ALS/FTD, including cortical neuronal hyperexcitability, age-dependent spinal motor neuron loss and progressive motor dysfunction. Quantitative proteomics revealed an increase in extracellular matrix (ECM) proteins in (GR)400 and (PR)400 spinal cord, with the collagen COL6A1 the most increased protein. TGF-ß1 was one of the top predicted regulators of this ECM signature and polyGR expression in human induced pluripotent stem cell neurons was sufficient to induce TGF-ß1 followed by COL6A1. Knockdown of TGF-ß1 or COL6A1 orthologues in polyGR model Drosophila exacerbated neurodegeneration, while expression of TGF-ß1 or COL6A1 in induced pluripotent stem cell-derived motor neurons of patients with C9ALS/FTD protected against glutamate-induced cell death. Altogether, our findings reveal a neuroprotective and conserved ECM signature in C9ALS/FTD.


Sujet(s)
Sclérose latérale amyotrophique , Démence frontotemporale , Cellules souches pluripotentes induites , Animaux , Humains , Souris , Démence frontotemporale/anatomopathologie , Sclérose latérale amyotrophique/métabolisme , Facteur de croissance transformant bêta-1 , Protéine C9orf72/génétique , Protéine C9orf72/métabolisme , Cellules souches pluripotentes induites/métabolisme , Motoneurones/métabolisme , Drosophila , Matrice extracellulaire/métabolisme , Dipeptides/métabolisme , Expansion de séquence répétée de l'ADN/génétique
3.
Nucleic Acids Res ; 49(13): 7713-7731, 2021 07 21.
Article de Anglais | MEDLINE | ID: mdl-34233002

RÉSUMÉ

Liquid-liquid phase separation (LLPS) of proteins and RNAs has emerged as the driving force underlying the formation of membrane-less organelles. Such biomolecular condensates have various biological functions and have been linked to disease. The protein Fused in Sarcoma (FUS) undergoes LLPS and mutations in FUS have been causally linked to the motor neuron disease Amyotrophic Lateral Sclerosis (ALS-FUS). LLPS followed by aggregation of cytoplasmic FUS has been proposed to be a crucial disease mechanism. However, it is currently unclear how LLPS impacts the behaviour of FUS in cells, e.g. its interactome. Hence, we developed a method allowing for the purification of LLPS FUS-containing droplets from cell lysates. We observe substantial alterations in the interactome, depending on its biophysical state. While non-LLPS FUS interacts mainly with factors involved in pre-mRNA processing, LLPS FUS predominantly binds to proteins involved in chromatin remodelling and DNA damage repair. Interestingly, also mitochondrial factors are strongly enriched with LLPS FUS, providing a potential explanation for the observed changes in mitochondrial gene expression in mouse models of ALS-FUS. In summary, we present a methodology to investigate the interactomes of phase separating proteins and provide evidence that LLPS shapes the FUS interactome with implications for function and disease.


Sujet(s)
Protéine FUS de liaison à l'ARN/métabolisme , Noyau de la cellule/métabolisme , Chromatine/métabolisme , Cytoplasme/métabolisme , Granulations cytoplasmiques/métabolisme , Cellules HEK293 , Cellules HeLa , Humains , Cartographie d'interactions entre protéines , ARN messager/métabolisme , Petit ARN nucléaire/métabolisme , Protéine FUS de liaison à l'ARN/composition chimique , Protéine FUS de liaison à l'ARN/isolement et purification
4.
Sci Adv ; 7(30)2021 07.
Article de Anglais | MEDLINE | ID: mdl-34290090

RÉSUMÉ

FUsed in Sarcoma (FUS) is a multifunctional RNA binding protein (RBP). FUS mutations lead to its cytoplasmic mislocalization and cause the neurodegenerative disease amyotrophic lateral sclerosis (ALS). Here, we use mouse and human models with endogenous ALS-associated mutations to study the early consequences of increased cytoplasmic FUS. We show that in axons, mutant FUS condensates sequester and promote the phase separation of fragile X mental retardation protein (FMRP), another RBP associated with neurodegeneration. This leads to repression of translation in mouse and human FUS-ALS motor neurons and is corroborated in vitro, where FUS and FMRP copartition and repress translation. Last, we show that translation of FMRP-bound RNAs is reduced in vivo in FUS-ALS motor neurons. Our results unravel new pathomechanisms of FUS-ALS and identify a novel paradigm by which mutations in one RBP favor the formation of condensates sequestering other RBPs, affecting crucial biological functions, such as protein translation.


Sujet(s)
Sclérose latérale amyotrophique , Maladies neurodégénératives , Sclérose latérale amyotrophique/génétique , Animaux , Protéine du syndrome X fragile/génétique , Souris , Mutation , Biosynthèse des protéines , Protéine FUS de liaison à l'ARN/génétique
5.
iScience ; 24(12): 103463, 2021 Dec 17.
Article de Anglais | MEDLINE | ID: mdl-34988393

RÉSUMÉ

Amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) is a fatal neurodegenerative disorder, and continued innovation is needed for improved understanding and for developing therapeutics. We have created next-generation genomically humanized knockin mouse models, by replacing the mouse genomic region of Sod1, Tardbp (TDP-43), and Fus, with their human orthologs, preserving human protein biochemistry and splicing with exons and introns intact. We establish a new standard of large knockin allele quality control, demonstrating the utility of indirect capture for enrichment of a genomic region of interest followed by Oxford Nanopore sequencing. Extensive analysis shows that homozygous humanized animals only express human protein at endogenous levels. Characterization of humanized FUS animals showed that they are phenotypically normal throughout their lifespan. These humanized strains are vital for preclinical assessment of interventions and serve as templates for the addition of coding or non-coding human ALS/FTD mutations to dissect disease pathomechanisms, in a physiological context.

6.
Nat Commun ; 11(1): 6341, 2020 12 11.
Article de Anglais | MEDLINE | ID: mdl-33311468

RÉSUMÉ

Mutations in the RNA-binding protein Fused in Sarcoma (FUS) cause early-onset amyotrophic lateral sclerosis (ALS). However, a detailed understanding of central RNA targets of FUS and their implications for disease remain elusive. Here, we use a unique blend of crosslinking and immunoprecipitation (CLIP) and NMR spectroscopy to identify and characterise physiological and pathological RNA targets of FUS. We find that U1 snRNA is the primary RNA target of FUS via its interaction with stem-loop 3 and provide atomic details of this RNA-mediated mode of interaction with the U1 snRNP. Furthermore, we show that ALS-associated FUS aberrantly contacts U1 snRNA at the Sm site with its zinc finger and traps snRNP biogenesis intermediates in human and murine motor neurons. Altogether, we present molecular insights into a FUS toxic gain-of-function involving direct and aberrant RNA-binding and strengthen the link between two motor neuron diseases, ALS and spinal muscular atrophy (SMA).


Sujet(s)
Sclérose latérale amyotrophique/métabolisme , Petit ARN nucléaire/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Protéine FUS de liaison à l'ARN/métabolisme , Petites ribonucléoprotéines nucléaires U1/métabolisme , Sclérose latérale amyotrophique/génétique , Animaux , Lignée cellulaire , Prédisposition génétique à une maladie/génétique , Humains , Souris , Souris knockout , Modèles moléculaires , Motoneurones/métabolisme , Mutation , Motifs et domaines d'intéraction protéique , Petit ARN nucléaire/composition chimique , Protéine FUS de liaison à l'ARN/composition chimique , Petites ribonucléoprotéines nucléaires U1/composition chimique
7.
Nucleic Acids Res ; 48(12): 6889-6905, 2020 07 09.
Article de Anglais | MEDLINE | ID: mdl-32479602

RÉSUMÉ

Mutations in the RNA-binding protein FUS cause amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease. FUS plays a role in numerous aspects of RNA metabolism, including mRNA splicing. However, the impact of ALS-causative mutations on splicing has not been fully characterized, as most disease models have been based on overexpressing mutant FUS, which will alter RNA processing due to FUS autoregulation. We and others have recently created knockin models that overcome the overexpression problem, and have generated high depth RNA-sequencing on FUS mutants in parallel to FUS knockout, allowing us to compare mutation-induced changes to genuine loss of function. We find that FUS-ALS mutations induce a widespread loss of function on expression and splicing. Specifically, we find that mutant FUS directly alters intron retention levels in RNA-binding proteins. Moreover, we identify an intron retention event in FUS itself that is associated with its autoregulation. Altered FUS levels have been linked to disease, and we show here that this novel autoregulation mechanism is altered by FUS mutations. Crucially, we also observe this phenomenon in other genetic forms of ALS, including those caused by TDP-43, VCP and SOD1 mutations, supporting the concept that multiple ALS genes interact in a regulatory network.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Homéostasie/génétique , Protéine FUS de liaison à l'ARN/génétique , Animaux , Cytoplasme/génétique , Protéines de liaison à l'ADN/génétique , Modèles animaux de maladie humaine , Régulation de l'expression des gènes/génétique , Humains , Introns/génétique , Mutation perte de fonction , Souris , Souris knockout , Mutation/génétique , Épissage des ARN/génétique , Superoxide dismutase-1/génétique , Protéine contenant la valosine/génétique
8.
Cell Rep ; 30(11): 3655-3662.e2, 2020 03 17.
Article de Anglais | MEDLINE | ID: mdl-32187538

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurodegenerative disease resulting from a complex interplay between genetics and environment. Impairments in axonal transport have been identified in several ALS models, but in vivo evidence remains limited, thus their pathogenetic importance remains to be fully resolved. We therefore analyzed the in vivo dynamics of retrogradely transported, neurotrophin-containing signaling endosomes in nerve axons of two ALS mouse models with mutations in the RNA processing genes TARDBP and FUS. TDP-43M337V mice, which show neuromuscular pathology without motor neuron loss, display axonal transport perturbations manifesting between 1.5 and 3 months and preceding symptom onset. Contrastingly, despite 20% motor neuron loss, transport remained largely unaffected in FusΔ14/+ mice. Deficiencies in retrograde axonal transport of signaling endosomes are therefore not shared by all ALS-linked genes, indicating that there are mechanistic distinctions in the pathogenesis of ALS caused by mutations in different RNA processing genes.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Transport axonal , Protéines de liaison à l'ADN/génétique , Endosomes/métabolisme , Mutation/génétique , Protéine FUS de liaison à l'ARN/génétique , Transduction du signal , Animaux , Femelle , Humains , Mâle , Souris de lignée C57BL , Motoneurones/métabolisme , Cellules réceptrices sensorielles/métabolisme
9.
Brain ; 140(11): 2797-2805, 2017 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-29053787

RÉSUMÉ

Mutations in FUS are causative for amyotrophic lateral sclerosis with a dominant mode of inheritance. In trying to model FUS-amyotrophic lateral sclerosis (ALS) in mouse it is clear that FUS is dosage-sensitive and effects arise from overexpression per se in transgenic strains. Novel models are required that maintain physiological levels of FUS expression and that recapitulate the human disease-with progressive loss of motor neurons in heterozygous animals. Here, we describe a new humanized FUS-ALS mouse with a frameshift mutation, which fulfils both criteria: the FUS Delta14 mouse. Heterozygous animals express mutant humanized FUS protein at physiological levels and have adult onset progressive motor neuron loss and denervation of neuromuscular junctions. Additionally, we generated a novel antibody to the unique human frameshift peptide epitope, allowing specific identification of mutant FUS only. Using our new FUSDelta14 ALS mouse-antibody system we show that neurodegeneration occurs in the absence of FUS protein aggregation. FUS mislocalization increases as disease progresses, and mutant FUS accumulates at the rough endoplasmic reticulum. Further, transcriptomic analyses show progressive changes in ribosomal protein levels and mitochondrial function as early disease stages are initiated. Thus, our new physiological mouse model has provided novel insight into the early pathogenesis of FUS-ALS.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Modèles animaux de maladie humaine , Mutation avec décalage du cadre de lecture , Souris , Agrégation pathologique de protéines/génétique , Protéine FUS de liaison à l'ARN/génétique , Sclérose latérale amyotrophique/métabolisme , Animaux , Réticulum endoplasmique rugueux/métabolisme , Dosage génique , Analyse de profil d'expression de gènes , Techniques de knock-in de gènes , Hétérozygote , Humains , Mitochondries/métabolisme , Motoneurones/métabolisme , Jonction neuromusculaire/métabolisme , Agrégation pathologique de protéines/métabolisme , Protéine FUS de liaison à l'ARN/métabolisme , Protéines ribosomiques/génétique
10.
Acta Neuropathol ; 131(3): 411-25, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26646779

RÉSUMÉ

Prion diseases are a group of fatal neurodegenerative disorders characterised by the accumulation of misfolded prion protein (PrP(Sc)) in the brain. The critical relationship between aberrant protein misfolding and neurotoxicity currently remains unclear. The accumulation of aggregation-prone proteins has been linked to impairment of the ubiquitin-proteasome system (UPS) in a variety of neurodegenerative disorders, including Alzheimer's, Parkinson's and Huntington's diseases. As the principal route for protein degradation in mammalian cells, this could have profound detrimental effects on neuronal function and survival. Here, we determine the temporal onset of UPS dysfunction in prion-infected Ub(G76V)-GFP reporter mice, which express a ubiquitin fusion proteasome substrate to measure in vivo UPS activity. We show that the onset of UPS dysfunction correlates closely with PrP(Sc) deposition, preceding earliest behavioural deficits and neuronal loss. UPS impairment was accompanied by accumulation of polyubiquitinated substrates and found to affect both neuronal and astrocytic cell populations. In prion-infected CAD5 cells, we demonstrate that activation of the UPS by the small molecule inhibitor IU1 is sufficient to induce clearance of polyubiquitinated substrates and reduce misfolded PrP(Sc) load. Taken together, these results identify the UPS as a possible early mediator of prion pathogenesis and promising target for development of future therapeutics.


Sujet(s)
Protéines PrPSc/métabolisme , Maladies à prions/métabolisme , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Animaux , Technique de Western , Modèles animaux de maladie humaine , Immunotransfert , Immunohistochimie , Hybridation in situ , Souris , Souris de lignée C57BL , Souris transgéniques , Microscopie confocale , Maladies à prions/anatomopathologie
11.
J Neurosci ; 35(7): 3155-73, 2015 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-25698751

RÉSUMÉ

The charged multivesicular body proteins (Chmp1-7) are an evolutionarily conserved family of cytosolic proteins that transiently assembles into helical polymers that change the curvature of cellular membrane domains. Mutations in human CHMP2B cause frontotemporal dementia, suggesting that this protein may normally control some neuron-specific process. Here, we examined the function, localization, and interactions of neuronal Chmp2b. The protein was highly expressed in mouse brain and could be readily detected in neuronal dendrites and spines. Depletion of endogenous Chmp2b reduced dendritic branching of cultured hippocampal neurons, decreased excitatory synapse density in vitro and in vivo, and abolished activity-induced spine enlargement and synaptic potentiation. To understand the synaptic effects of Chmp2b, we determined its ultrastructural distribution by quantitative immuno-electron microscopy and its biochemical interactions by coimmunoprecipitation and mass spectrometry. In the hippocampus in situ, a subset of neuronal Chmp2b was shown to concentrate beneath the perisynaptic membrane of dendritic spines. In synaptoneurosome lysates, Chmp2b was stably bound to a large complex containing other members of the Chmp family, as well as postsynaptic scaffolds. The supramolecular Chmp assembly detected here corresponds to a stable form of the endosomal sorting complex required for transport-III (ESCRT-III), a ubiquitous cytoplasmic protein complex known to play a central role in remodeling of lipid membranes. We conclude that Chmp2b-containing ESCRT-III complexes are also present at dendritic spines, where they regulate synaptic plasticity. We propose that synaptic ESCRT-III filaments may function as a novel element of the submembrane cytoskeleton of spines.


Sujet(s)
Complexes de tri endosomique requis pour le transport/déficit , Protéines de tissu nerveux/déficit , Synapses/physiologie , Animaux , Cellules cultivées , Simulation numérique , Dendrites/métabolisme , Dendrites/ultrastructure , Complexes de tri endosomique requis pour le transport/génétique , Agonistes des acides aminés excitateurs/pharmacologie , Potentiels post-synaptiques excitateurs/effets des médicaments et des substances chimiques , Potentiels post-synaptiques excitateurs/génétique , Femelle , Hippocampe/cytologie , Humains , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Mâle , Souris , Souris de lignée C57BL , Microscopie immunoélectronique , Mutation/génétique , N-Méthyl-aspartate/pharmacologie , Protéines de tissu nerveux/génétique , Neurones/cytologie , Neurones/ultrastructure , Densité post-synaptique/métabolisme , Densité post-synaptique/ultrastructure , Rats , Rat Sprague-Dawley , Synapses/ultrastructure ,
12.
Neurobiol Aging ; 36(1): 546.e1-7, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25179228

RÉSUMÉ

An expanded hexanucleotide repeat in the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Although 0-30 hexanucleotide repeats are present in the general population, expansions >500 repeats are associated with C9ALS/FTD. Large C9ALS/FTD expansions share a common haplotype and whether these expansions derive from a single founder or occur more frequently on a predisposing haplotype is yet to be determined and is relevant to disease pathomechanisms. Furthermore, although cases carrying 50-200 repeats have been described, their role and the pathogenic threshold of the expansions remain to be identified and carry importance for diagnostics and genetic counseling. We present clinical and genetic data from a UK ALS cohort and report the detailed molecular study of an atypical somatically unstable expansion of 90 repeats. Our results across different tissues provide evidence for the pathogenicity of this repeat number by showing they can somatically expand in the central nervous system to the well characterized pathogenic range. Our results support the occurrence of multiple expansion events for C9ALS/FTD.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Études de cohortes , Expansion de séquence répétée de l'ADN/génétique , Protéines/génétique , Protéine C9orf72 , Démence frontotemporale/génétique , Humains , Royaume-Uni
13.
Science ; 345(6201): 1192-1194, 2014 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-25103406

RÉSUMÉ

An expanded GGGGCC repeat in C9orf72 is the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis. A fundamental question is whether toxicity is driven by the repeat RNA itself and/or by dipeptide repeat proteins generated by repeat-associated, non-ATG translation. To address this question, we developed in vitro and in vivo models to dissect repeat RNA and dipeptide repeat protein toxicity. Expression of pure repeats, but not stop codon-interrupted "RNA-only" repeats in Drosophila caused adult-onset neurodegeneration. Thus, expanded repeats promoted neurodegeneration through dipeptide repeat proteins. Expression of individual dipeptide repeat proteins with a non-GGGGCC RNA sequence revealed that both poly-(glycine-arginine) and poly-(proline-arginine) proteins caused neurodegeneration. These findings are consistent with a dual toxicity mechanism, whereby both arginine-rich proteins and repeat RNA contribute to C9orf72-mediated neurodegeneration.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Expansion de séquence répétée de l'ADN/génétique , Drosophila melanogaster/génétique , Démence frontotemporale/génétique , Protéines/génétique , Sclérose latérale amyotrophique/anatomopathologie , Animaux , Protéine C9orf72 , Lignée cellulaire tumorale , Dipeptides/métabolisme , Modèles animaux de maladie humaine , Escherichia coli , Démence frontotemporale/anatomopathologie , Humains , Neurones/métabolisme , Neurones/anatomopathologie
14.
Neurobiol Aging ; 35(2): 443.e1-3, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24041967

RÉSUMÉ

Trinucleotide repeat disorders are a heterogeneous group of diseases caused by the expansion, beyond a pathogenic threshold, of unstable DNA tracts in different genes. Sequence interruptions in the repeats have been described in the majority of these disorders and may influence disease phenotype and heritability. Spinal bulbar muscular atrophy (SBMA) is a motor neuron disease caused by a CAG trinucleotide expansion in the androgen receptor (AR) gene. Diagnostic testing and previous research have relied on fragment analysis polymerase chain reaction to determine the AR CAG repeat size, and have therefore not been able to assess the presence of interruptions. We here report a sequencing study of the AR CAG repeat in a cohort of SBMA patients and control subjects in the United Kingdom. We found no repeat interruptions to be present, and we describe differences between sequencing and traditional sizing methods.


Sujet(s)
Amyotrophies/génétique , Récepteurs aux androgènes/génétique , Expansion de trinucléotide répété/génétique , Études de cohortes , Humains , Amyotrophies/diagnostic , Réaction de polymérisation en chaîne , Analyse de séquence d'ADN/méthodes , Royaume-Uni
15.
J Neurol Neurosurg Psychiatry ; 85(5): 506-8, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24309268

RÉSUMÉ

BACKGROUND: Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are progressive neurodegenerative disorders that share significant clinical, pathological and genetic overlap and are considered to represent different ends of a common disease spectrum. Mutations in Profilin1 have recently been described as a rare cause of familial ALS. The PFN1 E117G missense variant has been described in familial and sporadic cases, and also found in controls, casting doubt on its pathogenicity. Interpretation of such variants represents a significant clinical-genetics challenge. OBJECTIVE AND RESULTS: Here, we combine a screen of a new cohort of 383 ALS patients with multiple-sequence datasets to refine estimates of the ALS and FTD risk associated with PFN1 E117G. Together, our cohorts add up to 5118 ALS and FTD cases and 13 089 controls. We estimate a frequency of E117G of 0.11% in controls and 0.25% in cases. Estimated odds after population stratification is 2.44 (95% CI 1.048 to ∞, Mantel-Haenszel test p=0.036). CONCLUSIONS: Our results show an association between E117G and ALS, with a moderate effect size.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Mutation/génétique , Profilines/génétique , Sujet âgé , Études de cohortes , Démence frontotemporale/génétique , Humains , Mâle , Adulte d'âge moyen , Facteurs de risque , Royaume-Uni
16.
Nat Rev Genet ; 13(1): 14-20, 2011 Dec 16.
Article de Anglais | MEDLINE | ID: mdl-22179716

RÉSUMÉ

Mouse models have become an invaluable tool for understanding human health and disease owing to our ability to manipulate the mouse genome exquisitely. Recent progress in genomic analysis has led to an increase in the number and type of disease-causing mutations detected and has also highlighted the importance of non-coding regions. As a result, there is increasing interest in creating 'genomically' humanized mouse models, in which entire human genomic loci are transferred into the mouse genome. The technical challenges towards achieving this aim are large but are starting to be tackled with success.


Sujet(s)
Chromosomes artificiels de mammifère/génétique , Ciblage de gène/méthodes , Techniques de transfert de gènes , Souris transgéniques/génétique , Transgènes/génétique , Animaux , Modèles animaux de maladie humaine , Maladies génétiques congénitales/génétique , Étude d'association pangénomique/méthodes , Humains , Souris , Régions promotrices (génétique) , Recombinaison génétique , Éléments de régulation transcriptionnelle
17.
J Neurosci ; 28(33): 8189-98, 2008 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-18701681

RÉSUMÉ

Ubiquitin-positive intraneuronal inclusions are a consistent feature of the major human neurodegenerative diseases, suggesting that dysfunction of the ubiquitin proteasome system is central to disease etiology. Research using inhibitors of the 20S proteasome to model Parkinson's disease is controversial. We report for the first time that specifically 26S proteasomal dysfunction is sufficient to trigger neurodegenerative disease. Here, we describe novel conditional genetic mouse models using the Cre/loxP system to spatially restrict inactivation of Psmc1 (Rpt2/S4) to neurons of either the substantia nigra or forebrain (e.g., cortex, hippocampus, and striatum). PSMC1 is an essential subunit of the 26S proteasome and Psmc1 conditional knock-out mice display 26S proteasome depletion in targeted neurons, in which the 20S proteasome is not affected. Impairment of specifically ubiquitin-mediated protein degradation caused intraneuronal Lewy-like inclusions and extensive neurodegeneration in the nigrostriatal pathway and forebrain regions. Ubiquitin and alpha-synuclein neuropathology was evident, similar to human Lewy bodies, but interestingly, inclusion bodies contained mitochondria. We support this observation by demonstrating mitochondria in an early form of Lewy body (pale body) from Parkinson's disease patients. The results directly confirm that 26S dysfunction in neurons is involved in the pathology of neurodegenerative disease. The model demonstrates that 26S proteasomes are necessary for normal neuronal homeostasis and that 20S proteasome activity is insufficient for neuronal survival. Finally, we are providing the first reproducible genetic platform for identifying new therapeutic targets to slow or prevent neurodegeneration.


Sujet(s)
Encéphale/enzymologie , Corps d'inclusion/enzymologie , Corps de Lewy/enzymologie , Dégénérescence nerveuse/enzymologie , Dégénérescence nerveuse/génétique , Neurones/enzymologie , Proteasome endopeptidase complex/déficit , Animaux , Encéphale/anatomopathologie , Femelle , Humains , Corps d'inclusion/génétique , Corps d'inclusion/anatomopathologie , Corps de Lewy/génétique , Corps de Lewy/anatomopathologie , Mâle , Souris , Souris knockout , Souris transgéniques , Dégénérescence nerveuse/anatomopathologie , Neurones/anatomopathologie , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/physiologie
19.
Essays Biochem ; 41: 187-203, 2005.
Article de Anglais | MEDLINE | ID: mdl-16250906

RÉSUMÉ

The ubiquitin proteasome system (UPS) has emerged from obscurity to be seen as a major player in all regulatory processes in the cell. The concentrations of key proteins in diverse regulatory pathways are controlled by post-translational ubiquitination and degradation by the 26 S proteasome. These regulatory cascades include growth-factor-controlled signal-transduction pathways and multiple points in the cell cycle. The cell cycle is orchestrated by a combination of cyclin-dependent kinases, kinase inhibitors and protein phosphorylation, together with the timely and specific degradation of cyclins and kinase inhibitors at critical points in the cell cycle by the UPS. These processes provide the irreversibility needed for movement of the cycle through gap 1 (G1), DNA synthesis (S), gap 2 (G2) and mitosis (M). The molecular events include cell-size control, DNA replication, DNA repair, chromosomal rearrangements and cell division. It is doubtful whether these events could be achieved without the temporally and spatially regulated combination of protein phosphorylation and ubiquitin-dependent degradation of key cell-cycle regulatory proteins. The oncogenic transformation of cells is a multistep process that can be triggered by mutation of genes for proteins involved in regulatory processes from the cell surface to the nucleus. Since the UPS has critical functions at all these levels of control, it is to be expected that UPS activities will be central to cell transformation and cancer progression.


Sujet(s)
Tumeurs/métabolisme , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Animaux , Cycle cellulaire , Réparation de l'ADN , Substances de croissance/métabolisme , Humains , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Protéine du rétinoblastome/métabolisme , Transduction du signal , Protéine p53 suppresseur de tumeur/métabolisme , Ubiquitin-protein ligases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...