Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 42
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Commun Med (Lond) ; 3(1): 170, 2023 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-38017287

RÉSUMÉ

BACKGROUND: Charcot-Marie-Tooth disease type 1A (CMT1A) is one of the most common hereditary peripheral neuropathies caused by duplication of 1.5 Mb genome region including PMP22 gene. We aimed to correct the duplication in human CMT1A patient-derived iPS cells (CMT1A-iPSCs) by genome editing and intended to analyze the effect on Schwann cells differentiated from CMT1A-iPSCs. METHODS: We designed multiple gRNAs targeting a unique sequence present at two sites that sandwich only a single copy of duplicated peripheral myelin protein 22 (PMP22) genes, and selected one of them (gRNA3) from screening their efficiencies by T7E1 mismatch detection assay. AAV2-hSaCas9-gRNAedit was generated by subcloning gRNA3 into pX601-AAV-CMV plasmid, and the genome editing AAV vector was infected to CMT1A-iPSCs or CMT1A-iPSC-derived Schwann cell precursors. The effect of the genome editing AAV vector on myelination was evaluated by co-immunostaining of myelin basic protein (MBP), a marker of mature myelin, and microtubule-associated protein  2(MAP2), a marker of neurites or by electron microscopy. RESULTS: Here we show that infection of CMT1A-iPS cells (iPSCs) with AAV2-hSaCas9-gRNAedit expressing both hSaCas9 and gRNA targeting the tandem repeat sequence decreased PMP22 gene duplication by 20-40%. Infection of CMT1A-iPSC-derived Schwann cell precursors with AAV2-hSaCas9-gRNAedit normalized PMP22 mRNA and PMP22 protein expression levels, and also ameliorated increased apoptosis and impaired myelination in CMT1A-iPSC-derived Schwann cells. CONCLUSIONS: In vivo transfer of AAV2-hSaCas9-gRNAedit to peripheral nerves could be a potential therapeutic modality for CMT1A patient after careful examinations of toxicity including off-target mutations.


Charcot-Marie-Tooth disease type 1A (CMT1A) is a common heritable form of the condition that develops when nerves in the body's extremities, such as the hands, feet and arms, are damaged due to an extra copy of PMP22 gene being incorrectly produced. Currently, no known therapies exist. Here, we developed a method to delete the additional copy of PMP22 gene by 20­40% to prevent overproduction. Our results show that this method can reduce PMP22 protein production, leading to near normal production in patient's nerve cells. Further safety assessments should now be undertaken. If the treatment is safe for patients it could become a therapeutic option for CMT1A patients.

2.
Commun Biol ; 4(1): 1175, 2021 10 11.
Article de Anglais | MEDLINE | ID: mdl-34635772

RÉSUMÉ

DNA damage is increased in Alzheimer's disease (AD), while the underlying mechanisms are unknown. Here, we employ comprehensive phosphoproteome analysis, and identify abnormal phosphorylation of 70 kDa subunit of Ku antigen (Ku70) at Ser77/78, which prevents Ku70-DNA interaction, in human AD postmortem brains. The abnormal phosphorylation inhibits accumulation of Ku70 to the foci of DNA double strand break (DSB), impairs DNA damage repair and eventually causes transcriptional repression-induced atypical cell death (TRIAD). Cells under TRIAD necrosis reveal senescence phenotypes. Extracellular high mobility group box 1 (HMGB1) protein, which is released from necrotic or hyper-activated neurons in AD, binds to toll-like receptor 4 (TLR4) of neighboring neurons, and activates protein kinase C alpha (PKCα) that executes Ku70 phosphorylation at Ser77/78. Administration of human monoclonal anti-HMGB1 antibody to post-symptomatic AD model mice decreases neuronal DSBs, suppresses secondary TRIAD necrosis of neurons, prevents escalation of neurodegeneration, and ameliorates cognitive symptoms. TRIAD shares multiple features with senescence. These results discover the HMGB1-Ku70 axis that accounts for the increase of neuronal DNA damage and secondary enhancement of TRIAD, the cell death phenotype of senescence, in AD.


Sujet(s)
Maladie d'Alzheimer/anatomopathologie , Altération de l'ADN , Réparation de l'ADN , Protéine HMGB1/physiologie , Autoantigène Ku/métabolisme , Transduction du signal/génétique , Animaux , Protéine HMGB1/génétique , Souris , Souris transgéniques , Phosphorylation
3.
Commun Biol ; 4(1): 961, 2021 08 12.
Article de Anglais | MEDLINE | ID: mdl-34385591

RÉSUMÉ

Multiple gene mutations cause familial frontotemporal lobar degeneration (FTLD) while no single gene mutations exists in sporadic FTLD. Various proteins aggregate in variable regions of the brain, leading to multiple pathological and clinical prototypes. The heterogeneity of FTLD could be one of the reasons preventing development of disease-modifying therapy. We newly develop a mathematical method to analyze chronological changes of PPI networks with sequential big data from comprehensive phosphoproteome of four FTLD knock-in (KI) mouse models (PGRNR504X-KI, TDP43N267S-KI, VCPT262A-KI and CHMP2BQ165X-KI mice) together with four transgenic mouse models of Alzheimer's disease (AD) and with APPKM670/671NL-KI mice at multiple time points. The new method reveals the common core pathological network across FTLD and AD, which is shared by mouse models and human postmortem brains. Based on the prediction, we performed therapeutic intervention of the FTLD models, and confirmed amelioration of pathologies and symptoms of four FTLD mouse models by interruption of the core molecule HMGB1, verifying the new mathematical method to predict dynamic molecular networks.


Sujet(s)
Maladie d'Alzheimer/étiologie , Modèles animaux de maladie humaine , Dégénérescence lobaire frontotemporale/étiologie , Maladie d'Alzheimer/anatomopathologie , Animaux , Dégénérescence lobaire frontotemporale/anatomopathologie , Humains , Souris , Souris transgéniques , Modèles théoriques
4.
ACS Chem Neurosci ; 12(16): 3015-3027, 2021 08 18.
Article de Anglais | MEDLINE | ID: mdl-34319089

RÉSUMÉ

Tau aggregation is a central hallmark of tauopathies such as frontotemporal lobar degeneration and progressive supranuclear palsy as well as of Alzheimer's disease, and it has been a target for therapeutic development. Herein, we unexpectedly found that hepta-histidine (7H), an inhibitor of the interaction between Ku70 and Huntingtin proteins, suppresses aggregation of Tau-R3 peptides in vitro. Addition of the trans-activator of transcription (TAT) sequence (YGRKKRRQRRR) derived from the TAT protein to 7H increased its permeability into cells, and TAT-7H treatment of iPS cell-derived neurons carrying Tau or APP mutations suppressed Tau phosphorylation. These results indicate that 7H is a promising lead compound for developing anti-aggregation drugs against Tau-related neurodegenerative diseases including Alzheimer's disease (AD).


Sujet(s)
Maladie d'Alzheimer , Dégénérescence lobaire frontotemporale , Tauopathies , Maladie d'Alzheimer/traitement médicamenteux , Histidine , Humains , Protéines tau
5.
Life Sci Alliance ; 4(7)2021 07.
Article de Anglais | MEDLINE | ID: mdl-34130995

RÉSUMÉ

The early-stage pathologies of frontotemporal lobal degeneration (FTLD) remain largely unknown. In VCPT262A-KI mice carrying VCP gene mutation linked to FTLD, insufficient DNA damage repair in neural stem/progenitor cells (NSCs) activated DNA-PK and CDK1 that disabled MCM3 essential for the G1/S cell cycle transition. Abnormal neural exit produced neurons carrying over unrepaired DNA damage and induced early-stage transcriptional repression-induced atypical cell death (TRIAD) necrosis accompanied by the specific markers pSer46-MARCKS and YAP. In utero gene therapy expressing normal VCP or non-phosphorylated mutant MCM3 rescued DNA damage, neuronal necrosis, cognitive function, and TDP43 aggregation in adult neurons of VCPT262A-KI mice, whereas similar therapy in adulthood was less effective. The similar early-stage neuronal necrosis was detected in PGRNR504X-KI, CHMP2BQ165X-KI, and TDPN267S-KI mice, and blocked by embryonic treatment with AAV-non-phospho-MCM3. Moreover, YAP-dependent necrosis occurred in neurons of human FTLD patients, and consistently pSer46-MARCKS was increased in cerebrospinal fluid (CSF) and serum of these patients. Collectively, developmental stress followed by early-stage neuronal necrosis is a potential target for therapeutics and one of the earliest general biomarkers for FTLD.


Sujet(s)
Dégénérescence lobaire frontotemporale/anatomopathologie , Cellules souches neurales/métabolisme , Protéine contenant la valosine/métabolisme , Animaux , Cycle cellulaire , Lignage cellulaire/génétique , Cellules cultivées , Altération de l'ADN/génétique , Altération de l'ADN/physiologie , Protéines de liaison à l'ADN/métabolisme , Dégénérescence lobaire frontotemporale/liquide cérébrospinal , Dégénérescence lobaire frontotemporale/génétique , Expression des gènes/génétique , Régulation de l'expression des gènes/génétique , Souris , Souris de lignée C57BL , Mutation , Nécrose/métabolisme , Nécrose/anatomopathologie , Cellules souches neurales/anatomopathologie , Neurones/métabolisme , Protéine contenant la valosine/génétique
6.
Nat Commun ; 11(1): 507, 2020 01 24.
Article de Anglais | MEDLINE | ID: mdl-31980612

RÉSUMÉ

The timing and characteristics of neuronal death in Alzheimer's disease (AD) remain largely unknown. Here we examine AD mouse models with an original marker, myristoylated alanine-rich C-kinase substrate phosphorylated at serine 46 (pSer46-MARCKS), and reveal an increase of neuronal necrosis during pre-symptomatic phase and a subsequent decrease during symptomatic phase. Postmortem brains of mild cognitive impairment (MCI) rather than symptomatic AD patients reveal a remarkable increase of necrosis. In vivo imaging reveals instability of endoplasmic reticulum (ER) in mouse AD models and genome-edited human AD iPS cell-derived neurons. The level of nuclear Yes-associated protein (YAP) is remarkably decreased in such neurons under AD pathology due to the sequestration into cytoplasmic amyloid beta (Aß) aggregates, supporting the feature of YAP-dependent necrosis. Suppression of early-stage neuronal death by AAV-YAPdeltaC reduces the later-stage extracellular Aß burden and cognitive impairment, suggesting that preclinical/prodromal YAP-dependent neuronal necrosis represents a target for AD therapeutics.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Protéines du cycle cellulaire/métabolisme , Facteurs de transcription/métabolisme , Maladie d'Alzheimer/liquide cérébrospinal , Peptides bêta-amyloïdes/métabolisme , Animaux , Noyau de la cellule/métabolisme , Dysfonctionnement cognitif/liquide cérébrospinal , Dysfonctionnement cognitif/anatomopathologie , Simulation numérique , Modèles animaux de maladie humaine , Réticulum endoplasmique/anatomopathologie , Réticulum endoplasmique/ultrastructure , Femelle , Protéine HMGB1/liquide cérébrospinal , Humains , Cellules souches pluripotentes induites/métabolisme , Lysophospholipides/métabolisme , Mâle , Souris transgéniques , Nécrose , Neurones/métabolisme , Neurones/anatomopathologie , Transduction du signal , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Imagerie accélérée , Protéines de signalisation YAP
7.
J Neurosci ; 39(4): 678-691, 2019 01 23.
Article de Anglais | MEDLINE | ID: mdl-30504273

RÉSUMÉ

The actin cytoskeleton is crucial for neuronal migration in the mammalian developing cerebral cortex. The adaptor protein Drebrin-like (Dbnl) plays important roles in reorganization of the actin cytoskeleton, dendrite formation, and endocytosis by interacting with F-actin, cobl, and dynamin. Although Dbnl is known to be expressed in the brain, the functions of this molecule during brain development are largely unknown. In this study, to examine the roles of Dbnl in the developing cerebral cortex, we conducted experiments using mice of both sexes with knockdown of Dbnl, effected by in utero electroporation, in the migrating neurons of the embryonic cortex. Time-lapse imaging of the Dbnl-knockdown neurons revealed that the presence of Dbnl is a prerequisite for appropriate formation of processes in the multipolar neurons in the multipolar cell accumulation zone or the deep part of the subventricular zone, and for neuronal polarization and entry into the cortical plate. We found that Dbnl knockdown decreased the amount of N-cadherin protein expressed on the plasma membrane of the cortical neurons. The defect in neuronal migration caused by Dbnl knockdown was rescued by moderate overexpression of N-cadherin and αN-catenin or by transfection of the phospho-mimic form (Y337E, Y347E), but not the phospho-resistant form (Y337F, Y347F), of Dbnl. These results suggest that Dbnl controls neuronal migration, neuronal multipolar morphology, and cell polarity in the developing cerebral cortex via regulating N-cadherin expression.SIGNIFICANCE STATEMENT Disruption of neuronal migration can cause neuronal disorders, such as lissencephaly and subcortical band heterotopia. During cerebral cortical development, the actin cytoskeleton plays a key role in neuronal migration; however, the mechanisms of regulation of neuronal migration by the actin cytoskeleton still remain unclear. Herein, we report that the novel protein Dbnl, an actin-binding protein, controls multiple events during neuronal migration in the developing mouse cerebral cortex. We also showed that this regulation is mediated by phosphorylation of Dbnl at tyrosine residues 337 and 347 and αN-catenin/N-cadherin, suggesting that the Dbnl-αN-catenin/N-cadherin pathway is important for neuronal migration in the developing cortex.


Sujet(s)
Cadhérines/biosynthèse , Mouvement cellulaire/physiologie , Cortex cérébral/croissance et développement , Cortex cérébral/physiologie , Protéines des microfilaments/physiologie , Neurones/physiologie , Domaine d'homologie SRC/physiologie , Animaux , Cadhérines/génétique , Membrane cellulaire/métabolisme , Cortex cérébral/embryologie , Femelle , Régulation de l'expression des gènes au cours du développement , Techniques de knock-down de gènes , Ventricules latéraux/cytologie , Ventricules latéraux/métabolisme , Mâle , Souris , Souris de lignée ICR , Protéines des microfilaments/biosynthèse , Protéines des microfilaments/génétique , Neurones/ultrastructure , Grossesse , Domaine d'homologie SRC/génétique
8.
Mol Psychiatry ; 23(10): 2090-2110, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30283027

RÉSUMÉ

Early-phase pathologies of Alzheimer's disease (AD) are attracting much attention after clinical trials of drugs designed to remove beta-amyloid (Aß) aggregates failed to recover memory and cognitive function in symptomatic AD patients. Here, we show that phosphorylation of serine/arginine repetitive matrix 2 (SRRM2) at Ser1068, which is observed in the brains of early phase AD mouse models and postmortem end-stage AD patients, prevents its nuclear translocation by inhibiting interaction with T-complex protein subunit α. SRRM2 deficiency in neurons destabilized polyglutamine binding protein 1 (PQBP1), a causative gene for intellectual disability (ID), greatly affecting the splicing patterns of synapse-related genes, as demonstrated in a newly generated PQBP1-conditional knockout model. PQBP1 and SRRM2 were downregulated in cortical neurons of human AD patients and mouse AD models, and the AAV-PQBP1 vector recovered RNA splicing, the synapse phenotype, and the cognitive decline in the two mouse models. Finally, the kinases responsible for the phosphorylation of SRRM2 at Ser1068 were identified as ERK1/2 (MAPK3/1). These results collectively reveal a new aspect of AD pathology in which a phosphorylation signal affecting RNA splicing and synapse integrity precedes the formation of extracellular Aß aggregates and may progress in parallel with tau phosphorylation.


Sujet(s)
Maladie d'Alzheimer/génétique , Protéines de transport/génétique , Protéines nucléaires/génétique , Protéines de liaison à l'ARN/génétique , Transport nucléaire actif , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Animaux , Encéphale/métabolisme , Protéines de transport/métabolisme , Cognition , Protéines de liaison à l'ADN , Modèles animaux de maladie humaine , Humains , Cellules souches pluripotentes induites , Déficience intellectuelle/génétique , Système de signalisation des MAP kinases , Souris , Souris knockout , Souris transgéniques , Mitogen-Activated Protein Kinases/métabolisme , Neurones/métabolisme , Protéines nucléaires/métabolisme , Phosphorylation , Culture de cellules primaires , Épissage des ARN , Protéines de liaison à l'ARN/métabolisme , Protéines tau/métabolisme
9.
eNeuro ; 5(4)2018.
Article de Anglais | MEDLINE | ID: mdl-30225354

RÉSUMÉ

Phosphorylation of myristoylated alanine-rich C kinase substrate (MARCKS) reflects neurite degeneration at the early stage of Alzheimer's disease (AD), before extracellular Aß aggregates are histologically detectable. Here, we demonstrate that similar changes in MARCKS occur in Parkinson's disease (PD) and dementia with Lewy bodies (DLB) pathologies in both mouse models and human patients. The increase in the level of pSer46-MARCKS began before α-synuclein aggregate formation, at a time when human α-Syn-BAC-Tg/GBA-hetero-KO mice exhibited no symptoms, and was sustained during aging, consistent with the pattern in human postmortem brains. The results strongly imply a common mechanism of pre-aggregation neurite degeneration in AD and PD/DLB pathologies.


Sujet(s)
Vieillissement/métabolisme , Maladie d'Alzheimer , Maladie à corps de Lewy , Protéine myristoylée riche en alanine et substrat de la kinase C/métabolisme , Neurites/anatomopathologie , Maladie de Parkinson , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Femelle , Humains , Maladie à corps de Lewy/métabolisme , Maladie à corps de Lewy/anatomopathologie , Mâle , Souris , Souris transgéniques , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Phosphorylation
10.
Nat Commun ; 9(1): 433, 2018 01 30.
Article de Anglais | MEDLINE | ID: mdl-29382817

RÉSUMÉ

Mutations in the progranulin (PGRN) gene cause a tau pathology-negative and TDP43 pathology-positive form of frontotemporal lobar degeneration (FTLD-TDP). We generated a knock-in mouse harboring the R504X mutation (PGRN-KI). Phosphoproteomic analysis of this model revealed activation of signaling pathways connecting PKC and MAPK to tau prior to TDP43 aggregation and cognitive impairments, and identified PKCα as the kinase responsible for the early-stage tau phosphorylation at Ser203. Disinhibition of Gas6 binding to Tyro3 due to PGRN reduction results in activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, mislocalization of tau to dendritic spines, and spine loss. Administration of a PKC inhibitor, B-Raf inhibitor, or knockdown of molecules in the Gas6-Tyro3-tau axis rescues spine loss and cognitive impairment of PGRN-KI mice. Collectively, these results suggest that targeting of early-stage and aggregation-independent tau signaling represents a promising therapeutic strategy for this disease.


Sujet(s)
Dégénérescence lobaire frontotemporale/étiologie , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Protéines tau/métabolisme , Animaux , Modèles animaux de maladie humaine , Dégénérescence lobaire frontotemporale/métabolisme , Techniques de knock-in de gènes , Granulines , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Phénotype , Phospholipase C gamma/métabolisme , Phosphorylation , Progranulines , Protein kinase C-alpha/métabolisme , Protéines proto-oncogènes B-raf/métabolisme
11.
Thyroid ; 28(3): 395-406, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29415629

RÉSUMÉ

BACKGROUND: Thyroid hormones are essential for normal development of the central nervous system (CNS). Experimental rodents have shown that even a subtle thyroid hormone insufficiency in circulating maternal thyroid hormones during pregnancy may adversely affect neurodevelopment in offspring, resulting in irreversible cognitive deficits. This may be due to the persistent reduced expression of the hippocampal brain-derived neurotrophic factor gene Bdnf, which plays a crucial role in CNS development. However, the underlying molecular mechanisms remain unclear. METHODS: Thiamazole (MMI; 0.025% [w/v]) was administered to dams from two weeks prior to conception until delivery, which succeeded in inducing mild maternal hypothyroxinemia during pregnancy. Serum thyroid hormone and thyrotropin levels of the offspring derived from dams with mild maternal hypothyroxinemia (M offspring) and the control offspring (C offspring) were measured. At 70 days after birth, several behavior tests were performed on the offspring. Gene expression and DNA methylation status were also evaluated in the promoter region of Bdnf exon IV, which is largely responsible for neural activity-dependent Bdnf gene expression, in the hippocampus of the offspring at day 28 and day 70. RESULTS: No significant differences in serum thyroid hormone or thyrotropin levels were found between M and C offspring at day 28 and day 70. M offspring showed an impaired learning capacity in the behavior tests. Hippocampal steady-state Bdnf exon IV expression was significantly weaker in M offspring than it was in C offspring at day 28. At day 70, hippocampal Bdnf exon IV expression at the basal level was comparable between M and C offspring. However, it was significantly weaker in M offspring than in C offspring after the behavior tests. Persistent DNA hypermethylation was also found in the promoter region of Bdnf exon IV in the hippocampus of M offspring compared to that of C offspring, which may cause the attenuation of Bdnf exon IV expression in M offspring. CONCLUSIONS: Mild maternal hypothyroxinemia induces persistent DNA hypermethylation in Bdnf exon IV in offspring as epigenetic memory, which may result in long-term cognitive disorders.


Sujet(s)
Facteur neurotrophique dérivé du cerveau/métabolisme , Méthylation de l'ADN , Hippocampe/métabolisme , Hypothyroïdie/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Thyroxine/sang , Animaux , Comportement animal/physiologie , Facteur neurotrophique dérivé du cerveau/génétique , Femelle , Hypothyroïdie/génétique , Apprentissage du labyrinthe/physiologie , Souris , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/génétique , Test du rotarod
12.
Magn Reson Med Sci ; 17(2): 138-144, 2018 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-29213008

RÉSUMÉ

PURPOSE: Diffusional kurtosis imaging (DKI) enables sensitive measurement of tissue microstructure by quantifying the non-Gaussian diffusion of water. Although DKI is widely applied in many situations, histological correlation with DKI analysis is lacking. The purpose of this study was to determine the relationship between DKI metrics and neurite density measured using confocal microscopy of a cleared mouse brain. METHODS: One thy-1 yellow fluorescent protein 16 mouse was deeply anesthetized and perfusion fixation was performed. The brain was carefully dissected out and whole-brain MRI was performed using a 7T animal MRI system. DKI and diffusion tensor imaging (DTI) data were obtained. After the MRI scan, brain sections were prepared and then cleared using aminoalcohols (CUBIC). Confocal microscopy was performed using a two-photon confocal microscope with a laser. Forty-eight ROIs were set on the caudate putamen, seven ROIs on the anterior commissure, and seven ROIs on the ventral hippocampal commissure on the confocal microscopic image and a corresponding MR image. In each ROI, histological neurite density and the metrics of DKI and DTI were calculated. The correlations between diffusion metrics and neurite density were analyzed using Pearson correlation coefficient analysis. RESULTS: Mean kurtosis (MK) (P = 5.2 × 10-9, r = 0.73) and radial kurtosis (P = 2.3 × 10-9, r = 0.74) strongly correlated with neurite density in the caudate putamen. The correlation between fractional anisotropy (FA) and neurite density was moderate (P = 0.0030, r = 0.42). In the anterior commissure and the ventral hippocampal commissure, neurite density and FA are very strongly correlated (P = 1.3 × 10-5, r = 0.90). MK in these areas were very high value and showed no significant correlation (P = 0.48). CONCLUSION: DKI accurately reflected neurite density in the area with crossing fibers, potentially allowing evaluation of complex microstructures.


Sujet(s)
Encéphale , Imagerie par tenseur de diffusion/méthodes , Microscopie confocale/méthodes , Neurites/composition chimique , Animaux , Anisotropie , Encéphale/cytologie , Encéphale/anatomopathologie , Diffusion , Souris , Eau
13.
Nat Commun ; 8(1): 1864, 2017 11 30.
Article de Anglais | MEDLINE | ID: mdl-29192206

RÉSUMÉ

YAP and its neuronal isoform YAPdeltaC are implicated in various cellular functions. We found that expression of YAPdeltaC during development, but not adulthood, rescued neurodegeneration phenotypes of mutant ataxin-1 knock-in (Atxn1-KI) mice. YAP/YAPdeltaC interacted with RORα via the second WW domain and served as co-activators of its transcriptional activity. YAP/YAPdeltaC formed a transcriptional complex with RORα on cis-elements of target genes and regulated their expression. Both normal and mutant Atxn1 interacted with YAP/YAPdeltaC, but only mutant Atxn1 depleted YAP/YAPdeltaC from the RORα complex to suppress transcription on short timescales. Over longer periods, mutant Atxn1 also decreased RORα in vivo. Genetic supplementation of YAPdeltaC restored the RORα and YAP/YAPdeltaC levels, recovered YAP/YAPdeltaC in the RORα complex and normalized target gene transcription in Atxn1-KI mice in vivo. Collectively, our data suggest that functional impairment of YAP/YAPdeltaC by mutant Atxn1 during development determines the adult pathology of SCA1 by suppressing RORα-mediated transcription.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Ataxine-1/génétique , Cervelet/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Neurones/métabolisme , Membre-1 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Phosphoprotéines/métabolisme , Ataxies spinocérébelleuses/génétique , Animaux , Protéines du cycle cellulaire , Cervelet/cytologie , Modèles animaux de maladie humaine , Techniques de knock-in de gènes , Mâle , Souris , Phénotype , Isoformes de protéines , Test du rotarod , Ataxies spinocérébelleuses/physiopathologie , Protéines de signalisation YAP
14.
Acta Radiol Open ; 6(4): 2058460117703816, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28491462

RÉSUMÉ

BACKGROUND: Neurite orientation dispersion and density imaging (NODDI) is a diffusion magnetic resonance imaging (MRI) technique with the potential to visualize the microstructure of the brain. Revolutionary histological methods to render the mouse brain transparent have recently been developed, but verification of NODDI by these methods has not been reported. PURPOSE: To confirm the concordance of NODDI with histology in terms of density and orientation dispersion of neurites of the brain. MATERIAL AND METHODS: Whole brain diffusion MRI of a thy-1 yellow fluorescent protein mouse was acquired with a 7-T MRI scanner, after which transparent brain sections were created from the same mouse. NODDI parameters calculated from the MR images, including the intracellular volume fraction (Vic) and the orientation dispersion index (ODI), were compared with histological findings. Neurite density, Vic, and ODI were compared between areas of the anterior commissure and the hippocampus containing crossing fibers (crossing areas) and parallel fibers (parallel areas), and the correlation between fiber density and Vic was assessed. RESULTS: The ODI was significantly higher in the crossing area compared to the parallel area in both the anterior commissure and the hippocampus (P = 0.0247, P = 0.00022, respectively). Neurite density showed a similar tendency, but was significantly different only in the hippocampus (P = 7.91E-07). There was no significant correlation between neurite density and Vic. CONCLUSION: NODDI was verified by histology for quantification of the orientation dispersion of neurites. These results indicate that the ODI is a suitable index for understanding the microstructure of the brain in vivo.

15.
Acta Neuropathol Commun ; 5(1): 19, 2017 03 08.
Article de Anglais | MEDLINE | ID: mdl-28274274

RÉSUMÉ

We previously reported transcriptional repression-induced atypical cell death of neuron (TRIAD), a new type of necrosis that is mainly regulated by Hippo pathway signaling and distinct from necroptosis regulated by RIP1/3 pathway. Here, we examined the ultrastructural and biochemical features of neuronal cell death in the brains of human HD patients in parallel with the similar analyses using mutant Htt-knock-in (Htt-KI) mice. LATS1 kinase, the critical regulator and marker of TRIAD, is actually activated in cortical neurons of postmortem human HD and of Htt-KI mouse brains, while apoptosis promoter kinase Plk1 was inactivated in human HD brains. Expression levels of YAP/YAPdeltaC were decreased in cortical neurons of human HD brains. Ultra-structural analyses revealed extreme enlargement of endoplasmic reticulum (ER), which characterizes TRIAD, in cortical neurons of human HD and those of Htt-KI mice. These biochemical and morphological results support that TRIAD occurs in human and mouse neurons under the HD pathology.


Sujet(s)
Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Nécrose/métabolisme , Nécrose/anatomopathologie , Neurones/métabolisme , Neurones/ultrastructure , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Apoptose/physiologie , Encéphale/métabolisme , Encéphale/ultrastructure , Protéines du cycle cellulaire/métabolisme , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/ultrastructure , Techniques de knock-in de gènes , Humains , Souris de souche-129 , Souris de lignée C57BL , Souris transgéniques , Phosphoprotéines/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/métabolisme , Facteurs de transcription , Protéines de signalisation YAP ,
16.
Sci Rep ; 6: 33861, 2016 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-27653664

RÉSUMÉ

We identified drug seeds for treating Huntington's disease (HD) by combining in vitro single molecule fluorescence spectroscopy, in silico molecular docking simulations, and in vivo fly and mouse HD models to screen for inhibitors of abnormal interactions between mutant Htt and physiological Ku70, an essential DNA damage repair protein in neurons whose function is known to be impaired by mutant Htt. From 19,468 and 3,010,321 chemicals in actual and virtual libraries, fifty-six chemicals were selected from combined in vitro-in silico screens; six of these were further confirmed to have an in vivo effect on lifespan in a fly HD model, and two chemicals exerted an in vivo effect on the lifespan, body weight and motor function in a mouse HD model. Two oligopeptides, hepta-histidine (7H) and Angiotensin III, rescued the morphological abnormalities of primary neurons differentiated from iPS cells of human HD patients. For these selected drug seeds, we proposed a possible common structure. Unexpectedly, the selected chemicals enhanced rather than inhibited Htt aggregation, as indicated by dynamic light scattering analysis. Taken together, these integrated screens revealed a new pathway for the molecular targeted therapy of HD.

17.
Sci Rep ; 6: 31895, 2016 08 25.
Article de Anglais | MEDLINE | ID: mdl-27557632

RÉSUMÉ

Alzheimer's disease (AD) is the most common neurodegenerative disease, but it remains an intractable condition. Its pathogenesis is predominantly attributed to the aggregation and transmission of two molecules, Aß and tau; however, other pathological mechanisms are possible. Here, we reveal that phosphorylation of MARCKS, a submembrane protein that regulates the stability of the actin network, occurs at Ser46 prior to aggregation of Aß and is sustained throughout the course of AD in human and mouse brains. Furthermore, HMGB1 released from necrotic or hyperexcitatory neurons binds to TLR4, triggers the specific phosphorylation of MARCKS via MAP kinases, and induces neurite degeneration, the classical hallmark of AD pathology. Subcutaneous injection of a newly developed monoclonal antibody against HMGB1 strongly inhibits neurite degeneration even in the presence of Aß plaques and completely recovers cognitive impairment in a mouse model. HMGB1 and Aß mutually affect polymerization of the other molecule, and the therapeutic effects of the anti-HMGB1 monoclonal antibody are mediated by Aß-dependent and Aß-independent mechanisms. We propose that HMGB1 is a critical pathogenic molecule promoting AD pathology in parallel with Aß and tau and a new key molecular target of preclinical antibody therapy to delay the onset of AD.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Protéine HMGB1/métabolisme , Protéine myristoylée riche en alanine et substrat de la kinase C/métabolisme , Neurites/anatomopathologie , Récepteur de type Toll-4/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Animaux , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/pharmacologie , Modèles animaux de maladie humaine , Protéine HMGB1/antagonistes et inhibiteurs , Humains , Souris , Mitogen-Activated Protein Kinases/métabolisme , Thérapie moléculaire ciblée , Protéine myristoylée riche en alanine et substrat de la kinase C/composition chimique , Neurites/effets des médicaments et des substances chimiques , Neurites/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Sérine/métabolisme , Protéines tau/métabolisme
19.
Hum Mol Genet ; 25(21): 4749-4770, 2016 11 01.
Article de Anglais | MEDLINE | ID: mdl-28171658

RÉSUMÉ

Neuronal cell death in neurodegenerative diseases is not fully understood. Here we report that mutant huntingtin (Htt), a causative gene product of Huntington's diseases (HD) selectively induces a new form of necrotic cell death, in which endoplasmic reticulum (ER) enlarges and cell body asymmetrically balloons and finally ruptures. Pharmacological and genetic analyses revealed that the necrotic cell death is distinct from the RIP1/3 pathway-dependent necroptosis, but mediated by a functional deficiency of TEAD/YAP-dependent transcription. In addition, we revealed that a cell cycle regulator, Plk1, switches the balance between TEAD/YAP-dependent necrosis and p73/YAP-dependent apoptosis by shifting the interaction partner of YAP from TEAD to p73 through YAP phosphorylation at Thr77. In vivo ER imaging with two-photon microscopy detects similar ER enlargement, and viral vector-mediated delivery of YAP as well as chemical inhibitors of the Hippo pathway such as S1P recover the ER instability and necrosis in HD model mice. Intriguingly S1P completely stops the decline of motor function of HD model mice even after the onset of symptom. Collectively, we suggest approaches targeting the signalling pathway of TEAD/YAP-transcription-dependent necrosis (TRIAD) could lead to a therapeutic development against HD.


Sujet(s)
Protéine huntingtine/génétique , Protéine huntingtine/métabolisme , Nécrose/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Protéines du cycle cellulaire , Mort cellulaire , Protéines de liaison à l'ADN/génétique , Réticulum endoplasmique/métabolisme , Humains , Maladie de Huntington/métabolisme , Souris , Souris de lignée C57BL , Nécrose/génétique , Neurones/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Phosphoprotéines/métabolisme , Phosphorylation , Culture de cellules primaires , Liaison aux protéines , Transduction du signal , Transactivateurs/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
20.
Hum Mol Genet ; 25(20): 4432-4447, 2016 10 15.
Article de Anglais | MEDLINE | ID: mdl-28173122

RÉSUMÉ

DNA damage and repair is a critical domain of many neurodegenerative diseases. In this study, we focused on RpA1, a candidate key molecule in polyQ disease pathologies, and tested the therapeutic effect of adeno-associated virus (AAV) vector expressing RpA1 on mutant Ataxin-1 knock-in (Atxn1-KI) mice. We found significant effects on motor functions, normalized DNA damage markers (γH2AX and 53BP1), and improved Purkinje cell morphology; effects that lasted for 50 weeks following AAV-RpA1 infection. In addition, we confirmed that AAV-RpA1 indirectly recovered multiple cellular functions such as RNA splicing, transcription and cell cycle as well as abnormal morphology of dendrite and dendritic spine of Purkinje cells in Atxn1-KI mice. All these results suggested a possibility of gene therapy with RpA1 for SCA1.


Sujet(s)
Ataxine-1/génétique , Réparation de l'ADN , Mutation , Protéine A de réplication/métabolisme , Ataxies spinocérébelleuses/métabolisme , Animaux , Cycle cellulaire , ADN/métabolisme , Altération de l'ADN , Dependovirus , Modèles animaux de maladie humaine , Techniques de knock-in de gènes , Thérapie génétique , Souris , Cellules de Purkinje/métabolisme , Cellules de Purkinje/anatomopathologie , Cellules de Purkinje/physiologie , ARN/métabolisme , Épissage des ARN , Ataxies spinocérébelleuses/génétique , Ataxies spinocérébelleuses/anatomopathologie , Ataxies spinocérébelleuses/physiopathologie , Transcription génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...