Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 347
Filtrer
1.
Nat Commun ; 15(1): 8195, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39294122

RÉSUMÉ

Progressive supranuclear palsy (PSP) is an incurable neurodegenerative disease characterized by 4-repeat (0N/4R)-Tau protein accumulation in CNS neurons. We generated transgenic zebrafish expressing human 0N/4R-Tau to investigate PSP pathophysiology. Tau zebrafish replicated multiple features of PSP, including: decreased survival; hypokinesia; impaired optokinetic responses; neurodegeneration; neuroinflammation; synapse loss; and Tau hyperphosphorylation, misfolding, mislocalization, insolubility, truncation, and oligomerization. Using automated assays, we screened 147 small molecules for activity in rescuing neurological deficits in Tau zebrafish. (+)JQ1, a bromodomain inhibitor, improved hypokinesia, survival, microgliosis, and brain synapse elimination. A heterozygous brd4+/- mutant reducing expression of the bromodomain protein Brd4 similarly rescued these phenotypes. Microglial phagocytosis of synaptic material was decreased by (+)JQ1 in both Tau zebrafish and rat primary cortical cultures. Microglia in human PSP brains expressed Brd4. Our findings implicate Brd4 as a regulator of microglial synaptic elimination in tauopathy and provide an unbiased approach for identifying mechanisms and therapeutic targets in PSP.


Sujet(s)
Animal génétiquement modifié , Modèles animaux de maladie humaine , Microglie , Paralysie supranucléaire progressive , Synapses , Facteurs de transcription , Danio zébré , Protéines tau , Animaux , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines tau/métabolisme , Protéines tau/génétique , Microglie/métabolisme , Microglie/anatomopathologie , Synapses/métabolisme , Paralysie supranucléaire progressive/métabolisme , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Azépines/pharmacologie , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Triazoles/pharmacologie , Rats , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Encéphale/métabolisme , Encéphale/anatomopathologie , Phagocytose , Neurones/métabolisme , Protéines contenant un bromodomaine , Protéines du cycle cellulaire
2.
Nat Commun ; 15(1): 7880, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251599

RÉSUMÉ

Progressive supranuclear palsy (PSP), a rare Parkinsonian disorder, is characterized by problems with movement, balance, and cognition. PSP differs from Alzheimer's disease (AD) and other diseases, displaying abnormal microtubule-associated protein tau by both neuronal and glial cell pathologies. Genetic contributors may mediate these differences; however, the genetics of PSP remain underexplored. Here we conduct the largest genome-wide association study (GWAS) of PSP which includes 2779 cases (2595 neuropathologically-confirmed) and 5584 controls and identify six independent PSP susceptibility loci with genome-wide significant (P < 5 × 10-8) associations, including five known (MAPT, MOBP, STX6, RUNX2, SLCO1A2) and one novel locus (C4A). Integration with cell type-specific epigenomic annotations reveal an oligodendrocytic signature that might distinguish PSP from AD and Parkinson's disease in subsequent studies. Candidate PSP risk gene prioritization using expression quantitative trait loci (eQTLs) identifies oligodendrocyte-specific effects on gene expression in half of the genome-wide significant loci, and an association with C4A expression in brain tissue, which may be driven by increased C4A copy number. Finally, histological studies demonstrate tau aggregates in oligodendrocytes that colocalize with C4 (complement) deposition. Integrating GWAS with functional studies, epigenomic and eQTL analyses, we identify potential causal roles for variation in MOBP, STX6, RUNX2, SLCO1A2, and C4A in PSP pathogenesis.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Locus de caractère quantitatif , Paralysie supranucléaire progressive , Protéines tau , Humains , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/métabolisme , Sujet âgé , Mâle , Femelle , Protéines tau/génétique , Protéines tau/métabolisme , Transcriptome , Polymorphisme de nucléotide simple , Névroglie/métabolisme , Névroglie/anatomopathologie , Sujet âgé de 80 ans ou plus , Oligodendroglie/métabolisme , Oligodendroglie/anatomopathologie , Adulte d'âge moyen , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Études cas-témoins , Protéines de la myéline
3.
Cell ; 187(20): 5753-5774.e28, 2024 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-39265576

RÉSUMÉ

The development of successful therapeutics for dementias requires an understanding of their shared and distinct molecular features in the human brain. We performed single-nuclear RNA-seq and ATAC-seq in Alzheimer's disease (AD), frontotemporal dementia (FTD), and progressive supranuclear palsy (PSP), analyzing 41 participants and ∼1 million cells (RNA + ATAC) from three brain regions varying in vulnerability and pathological burden. We identify 32 shared, disease-associated cell types and 14 that are disease specific. Disease-specific cell states represent glial-immune mechanisms and selective neuronal vulnerability impacting layer 5 intratelencephalic neurons in AD, layer 2/3 intratelencephalic neurons in FTD, and layer 5/6 near-projection neurons in PSP. We identify disease-associated gene regulatory networks and cells impacted by causal genetic risk, which differ by disorder. These data illustrate the heterogeneous spectrum of glial and neuronal compositional and gene expression alterations in different dementias and identify therapeutic targets by revealing shared and disease-specific cell states.


Sujet(s)
Maladie d'Alzheimer , Démence frontotemporale , Réseaux de régulation génique , Génomique , Neurones , Analyse sur cellule unique , Paralysie supranucléaire progressive , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/métabolisme , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Démence frontotemporale/métabolisme , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/métabolisme , Paralysie supranucléaire progressive/anatomopathologie , Génomique/méthodes , Neurones/métabolisme , Neurones/anatomopathologie , Sujet âgé , Mâle , Femelle , Encéphale/métabolisme , Encéphale/anatomopathologie , Démence/génétique , Démence/anatomopathologie , Démence/métabolisme , Névroglie/métabolisme , Névroglie/anatomopathologie , Sujet âgé de 80 ans ou plus , Adulte d'âge moyen , RNA-Seq
4.
Orphanet J Rare Dis ; 19(1): 312, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-39183347

RÉSUMÉ

INTRODUCTION: We present a systematic review of phenotypes of McLeod syndrome (MLS) and a case of a 73-year-old female carrier of the genetic alteration leading to MLS with the typical progressive supranuclear palsy (PSP) phenotype. METHODS: To facilitate clinical reasoning and enable targeted diagnosis, we conducted a systematic review of the papers describing symptomatic cases of confirmed McLeod syndrome. This review follows the PRISMA 2020 statement: an updated guideline for reporting systematic reviews (Page et al in Syst Rev 10(1):89, 2021). RESULTS: The average onset of MLS was at 40.2 years of age with chorea (46%), seizures and psychiatric changes (each 13%). Very common are weakened Kell antigen (100%), changes in muscle biopsy (100%), genetic alterations in XK (100%), elevated creatine kinase (97%), acanthocytes (96%), MRI changes (95%), chorea (84%) and hyporeflexia (82%). CONCLUSION: This review of 65 males and 11 females gives a concise overview of clinical phenotypes in MLS, reinforcing our view that this female patient had PSP independent of MLS carrier status. This report highlights the pitfalls of anchoring in medical decision-making, particularly the possible diagnostic bias of a known genetic carrier status of a very rare disease.


Sujet(s)
Neuroacanthocytose , Paralysie supranucléaire progressive , Sujet âgé , Femelle , Humains , Mâle , Neuroacanthocytose/génétique , Neuroacanthocytose/anatomopathologie , Neuroacanthocytose/diagnostic , Phénotype , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie
5.
Acta Neuropathol Commun ; 12(1): 135, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154163

RÉSUMÉ

Progressive supranuclear palsy (PSP) is a neurodegenerative movement and cognitive disorder characterized by abnormal accumulation of the microtubule-associated protein tau in the brain. Biochemically, inclusions in PSP are enriched for tau proteoforms with four microtubule-binding domain repeats (4R), an isoform that arises from alternative tau pre-mRNA splicing. While preferential aggregation and reduced degradation of 4R tau protein is thought to play a role in inclusion formation and toxicity, an alternative hypothesis is that altered expression of tau mRNA isoforms plays a causal role. This stems from the observation that PSP is associated with common variation in the tau gene (MAPT) at the 17q21.31 locus which contains low copy number repeats flanking a large recurrent genomic inversion. The complex genomic structural changes at the locus give rise to two dominant haplotypes, termed H1 and H2, that have the potential to markedly influence gene expression. Here, we explored haplotype-dependent differences in gene expression using a bulk RNA-seq dataset derived from human post-mortem brain tissue from PSP (n = 84) and controls (n = 77) using a rigorous computational pipeline, including alternative pre-mRNA splicing. We found 3579 differentially expressed genes in the temporal cortex and 10,011 in the cerebellum. We also found 7214 differential splicing events in the temporal cortex and 18,802 in the cerebellum. In the cerebellum, total tau mRNA levels and the proportion of transcripts encoding 4R tau were significantly increased in PSP compared to controls. In the temporal cortex, the proportion of reads that expressed 4R tau was increased in cases compared to controls. 4R tau mRNA levels were significantly associated with the H1 haplotype in the temporal cortex. Further, we observed a marked haplotype-dependent difference in KANSL1 expression that was strongly associated with H1 in both brain regions. These findings support the hypothesis that sporadic PSP is associated with haplotype-dependent increases in 4R tau mRNA that might play a causal role in this disorder.


Sujet(s)
Haplotypes , Paralysie supranucléaire progressive , Transcriptome , Protéines tau , Humains , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/métabolisme , Protéines tau/génétique , Protéines tau/métabolisme , Sujet âgé , Mâle , Femelle , Sujet âgé de 80 ans ou plus , Encéphale/métabolisme , Encéphale/anatomopathologie , Adulte d'âge moyen
6.
Acta Neuropathol Commun ; 12(1): 132, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39138580

RÉSUMÉ

Progressive supranuclear palsy (PSP) is a neurodegenerative disease characterized by 4R tau deposition in neurons as well as in astrocytes and oligodendrocytes. While astrocytic tau deposits are rarely observed in normal aging (so-called aging-related tau astrogliopathy, ARTAG) and Alzheimer's disease (AD), astrocytic tau in the form of tufted astrocytes is a pathognomonic hallmark of PSP. Classical biochemical experiments emphasized tau synthesis in neurons in the central nervous system, suggesting that astrocytic tau inclusions might be derived from uptake of extracellular neuronal-derived tau. However, recent single-nucleus RNAseq experiments highlight the fact that MAPT, the gene encoding tau, is also expressed by astrocytes, albeit in lower amounts. We, therefore, revisited the question of whether astrocyte-driven expression of tau might contribute to astrocytic tau aggregates in PSP by performing fluorescent in situ hybridization/immunohistochemical co-localization in human postmortem brain specimens from individuals with PSP and AD with ARTAG as well as normal controls. We find that, in PSP but not in AD, tau-immunoreactive astrocytes have higher levels of MAPT mRNA compared to astrocytes that do not have tau aggregates. These results suggest that astrocytic responses in PSP are unique to this tauopathy and support the possibility that fundamental changes in PSP astrocyte-endogenous mRNA biology contribute to increased synthesis of tau protein and underlies the formation of the astrocytic tau deposits characteristic of PSP.


Sujet(s)
Astrocytes , Paralysie supranucléaire progressive , Protéines tau , Paralysie supranucléaire progressive/métabolisme , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/génétique , Protéines tau/métabolisme , Protéines tau/génétique , Humains , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Sujet âgé , Mâle , Sujet âgé de 80 ans ou plus , Femelle , Adulte d'âge moyen , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Transcription génétique , Encéphale/métabolisme , Encéphale/anatomopathologie
7.
Mol Neurodegener ; 19(1): 61, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152475

RÉSUMÉ

BACKGROUND: Progressive supranuclear palsy (PSP) is a rare neurodegenerative disease characterized by the accumulation of aggregated tau proteins in astrocytes, neurons, and oligodendrocytes. Previous genome-wide association studies for PSP were based on genotype array, therefore, were inadequate for the analysis of rare variants as well as larger mutations, such as small insertions/deletions (indels) and structural variants (SVs). METHOD: In this study, we performed whole genome sequencing (WGS) and conducted association analysis for single nucleotide variants (SNVs), indels, and SVs, in a cohort of 1,718 cases and 2,944 controls of European ancestry. Of the 1,718 PSP individuals, 1,441 were autopsy-confirmed and 277 were clinically diagnosed. RESULTS: Our analysis of common SNVs and indels confirmed known genetic loci at MAPT, MOBP, STX6, SLCO1A2, DUSP10, and SP1, and further uncovered novel signals in APOE, FCHO1/MAP1S, KIF13A, TRIM24, TNXB, and ELOVL1. Notably, in contrast to Alzheimer's disease (AD), we observed the APOE ε2 allele to be the risk allele in PSP. Analysis of rare SNVs and indels identified significant association in ZNF592 and further gene network analysis identified a module of neuronal genes dysregulated in PSP. Moreover, seven common SVs associated with PSP were observed in the H1/H2 haplotype region (17q21.31) and other loci, including IGH, PCMT1, CYP2A13, and SMCP. In the H1/H2 haplotype region, there is a burden of rare deletions and duplications (P = 6.73 × 10-3) in PSP. CONCLUSIONS: Through WGS, we significantly enhanced our understanding of the genetic basis of PSP, providing new targets for exploring disease mechanisms and therapeutic interventions.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Polymorphisme de nucléotide simple , Paralysie supranucléaire progressive , Séquençage du génome entier , Humains , Paralysie supranucléaire progressive/génétique , Prédisposition génétique à une maladie/génétique , Mâle , Femelle , Sujet âgé , Polymorphisme de nucléotide simple/génétique , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus
8.
CNS Neurosci Ther ; 30(8): e14901, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39097922

RÉSUMÉ

BACKGROUND: It has been demonstrated that progressive supranuclear palsy (PSP) correlates with structural abnormalities in several distinct regions of the brain. However, whether there are changes in the morphological similarity network (MSN) and the relationship between changes in brain structure and gene expression remain largely unknown. METHODS: We used two independent cohorts (discovery dataset: PSP: 51, healthy controls (HC): 82; replication dataset: PSP: 53, HC: 55) for MSN analysis and comparing the longitudinal changes in the MSN of PSP. Then, we applied partial least squares regression to determine the relationships between changes in MSN and spatial transcriptional features and identified specific genes associated with MSN differences in PSP. We further investigated the biological processes enriched in PSP-associated genes and the cellular characteristics of these genes, and finally, we performed an exploratory analysis of the relationship between MSN changes and neurotransmitter receptors. RESULTS: We found that the MSN in PSP patients was mainly decreased in the frontal and temporal cortex but increased in the occipital cortical region. This difference is replicable. In longitudinal studies, MSN differences are mainly manifested in the frontal and parietal regions. Furthermore, the expression pattern associated with MSN changes in PSP involves genes implicated in astrocytes and excitatory and inhibitory neurons and is functionally enriched in neuron-specific biological processes related to synaptic signaling. Finally, we found that the changes in MSN were mainly negatively correlated with the levels of serotonin, norepinephrine, and opioid receptors. CONCLUSIONS: These results have enhanced our understanding of the microscale genetic and cellular mechanisms responsible for large-scale morphological abnormalities in PSP patients, suggesting potential targets for future therapeutic trials.


Sujet(s)
Paralysie supranucléaire progressive , Humains , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/métabolisme , Femelle , Mâle , Sujet âgé , Adulte d'âge moyen , Cortex cérébral/métabolisme , Cortex cérébral/anatomopathologie , Études de cohortes , Études longitudinales
9.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39125813

RÉSUMÉ

Progressive supranuclear palsy (PSP) is a rare, neurodegenerative movement disorder. Together with multiple system atrophy (MSA), Dementia with Lewy bodies (DLB), and corticobasal degeneration (CBD), PSP forms a group of atypical parkinsonisms. The latest diagnostic criteria, published in 2017 by the Movement Disorders Society, classify PSP diagnosis into defined, probable, and possible categories based on clinical examination. However, no single test is specific and sensitive for this disease. Microribonucleic acids (miRNAs) are promising molecules, particularly in the case of diseases that lack appropriate diagnostic and treatment tools, which supports exploring their role in PSP. We aimed to systematically review the current knowledge about the role of miRNAs in PSP. This study was registered in the Open Science Framework Registry, and the protocol is available online. Primary original studies, both clinical and preclinical, written in English and assessing miRNAs in PSP were included. Systematic reviews, meta-analyses, reviews, case reports, letters to editors, commentaries, conference abstracts, guidelines/statements, expert opinions, preprints, and book chapters were excluded. The following five databases were searched: Embase, Medline Ultimate, PubMed, Scopus, and Web of Science. Each database was last searched on 18 June 2024. Eventually, nine original studies relevant to the discussed area were included. The risk of bias was not assessed. The selected research suggests that miRNAs may be considered promising biomarkers in PSP. However, the exact involvement of miRNAs in the pathogenesis of PSP is still to be determined. Several microRNAs were found to be dysregulated in patients with PSP. This applies to both brain tissue and fluids like cerebrospinal fluid CSF or blood. Several miRNAs were found that could potentially be helpful in differentiating among PSP patients, PD patients, and healthy individuals. Although some correlations and alterations have already been found, this field requires much more research. MicroRNAs are exciting and promising small molecules, and their investigation into many diseases, including PSP, may lead to significant discoveries.


Sujet(s)
microARN , Paralysie supranucléaire progressive , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/diagnostic , Humains , microARN/génétique , Marqueurs biologiques
10.
Acta Neuropathol ; 148(1): 4, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38995454

RÉSUMÉ

Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by neuronal loss and gliosis, with oligodendroglial cytoplasmic inclusions (GCIs) containing α-synuclein being the primary pathological hallmark. Clinical presentations of MSA overlap with other parkinsonian disorders, such as Parkinson's disease (PD), dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP), posing challenges in early diagnosis. Numerous studies have reported alterations in DNA methylation in neurodegenerative diseases, with candidate loci being identified in various parkinsonian disorders including MSA, PD, and PSP. Although MSA and PSP present with substantial white matter pathology, alterations in white matter have also been reported in PD. However, studies comparing the DNA methylation architectures of white matter in these diseases are lacking. We therefore aimed to investigate genome-wide DNA methylation patterns in the frontal lobe white matter of individuals with MSA (n = 17), PD (n = 17), and PSP (n = 16) along with controls (n = 15) using the Illumina EPIC array, to identify shared and disease-specific DNA methylation alterations. Genome-wide DNA methylation profiling of frontal lobe white matter in the three parkinsonian disorders revealed substantial commonalities in DNA methylation alterations in MSA, PD, and PSP. We further used weighted gene correlation network analysis to identify disease-associated co-methylation signatures and identified dysregulation in processes relating to Wnt signaling, signal transduction, endoplasmic reticulum stress, mitochondrial processes, RNA interference, and endosomal transport to be shared between these parkinsonian disorders. Our overall analysis points toward more similarities in DNA methylation patterns between MSA and PD, both synucleinopathies, compared to that between MSA and PD with PSP, which is a tauopathy. Our results also highlight several shared DNA methylation changes and pathways indicative of converging molecular mechanisms in the white matter contributing toward neurodegeneration in all three parkinsonian disorders.


Sujet(s)
Méthylation de l'ADN , Lobe frontal , Atrophie multisystématisée , Maladie de Parkinson , Paralysie supranucléaire progressive , Substance blanche , Humains , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Méthylation de l'ADN/génétique , Atrophie multisystématisée/génétique , Atrophie multisystématisée/anatomopathologie , Substance blanche/anatomopathologie , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Sujet âgé , Femelle , Mâle , Lobe frontal/anatomopathologie , Lobe frontal/métabolisme , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus
11.
Mov Disord Clin Pract ; 11(8): 1030-1034, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38881158

RÉSUMÉ

BACKGROUND: Heterozygous mutations in GBA1 gene are known as most common genetic risk factor for Parkinson's disease (PD). However, role of GBA1 mutations in non-α-synuclein disorders is unclear. CASES: Case index, 76 year-old woman referred to our movement disorders outpatient clinic for 2-year history of gait impairment, falls and motor slowness, with partial response to levodopa. Clinical and instrumental examinations were consistent with Progressive Supranuclear Palsy-Corticobasal Syndrome (PSP-CBS). Case 2 is older sister reporting depressive symptoms; however, she had dementia (MMSE 18/30), gait apraxia and vertical supranuclear gaze palsy (VSNGP). Case 3 is her deceased older sister who had been diagnosed with Corticobasal Syndrome (CBS). Case 4, older brother had been diagnosed with Parkinson's disease-dementia (PDD) with good response to levodopa. Two affected living siblings harboring same genetic variant. CONCLUSIONS: To our knowledge, this is the first family showing such intrafamilial variability ranging from CBS to PDD to dementia.


Sujet(s)
Glucosylceramidase , Maladies neurodégénératives , Humains , Glucosylceramidase/génétique , Femelle , Sujet âgé , Mâle , Maladies neurodégénératives/génétique , Maladies neurodégénératives/diagnostic , Italie , Maladie de Parkinson/génétique , Maladie de Parkinson/complications , Mutation , Paralysie supranucléaire progressive/génétique
12.
Cell ; 187(10): 2446-2464.e22, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38582079

RÉSUMÉ

Tauopathies are age-associated neurodegenerative diseases whose mechanistic underpinnings remain elusive, partially due to a lack of appropriate human models. Here, we engineered human induced pluripotent stem cell (hiPSC)-derived neuronal lines to express 4R Tau and 4R Tau carrying the P301S MAPT mutation when differentiated into neurons. 4R-P301S neurons display progressive Tau inclusions upon seeding with Tau fibrils and recapitulate features of tauopathy phenotypes including shared transcriptomic signatures, autophagic body accumulation, and reduced neuronal activity. A CRISPRi screen of genes associated with Tau pathobiology identified over 500 genetic modifiers of seeding-induced Tau propagation, including retromer VPS29 and genes in the UFMylation cascade. In progressive supranuclear palsy (PSP) and Alzheimer's Disease (AD) brains, the UFMylation cascade is altered in neurofibrillary-tangle-bearing neurons. Inhibiting the UFMylation cascade in vitro and in vivo suppressed seeding-induced Tau propagation. This model provides a robust platform to identify novel therapeutic strategies for 4R tauopathy.


Sujet(s)
Cellules souches pluripotentes induites , Neurones , Tauopathies , Protéines tau , Humains , Cellules souches pluripotentes induites/métabolisme , Protéines tau/métabolisme , Tauopathies/métabolisme , Tauopathies/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Animaux , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Encéphale/métabolisme , Encéphale/anatomopathologie , Paralysie supranucléaire progressive/métabolisme , Paralysie supranucléaire progressive/anatomopathologie , Paralysie supranucléaire progressive/génétique , Différenciation cellulaire , Mutation , Autophagie
13.
Mov Disord Clin Pract ; 11(6): 720-727, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38605589

RÉSUMÉ

BACKGROUND: MAPT is a causative gene in frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), a hereditary degenerative disease with various clinical manifestations, including progressive supranuclear palsy, corticobasal syndrome, Parkinson's disease, and frontotemporal dementia. OBJECTIVES: To analyze genetically, biochemically, and pathologically multiple members of two families who exhibited various phenotypes of the disease. METHODS: Genetic analysis included linkage analysis, homozygosity haplotyping, and exome sequencing. We conducted tau protein microtubule polymerization assay, heparin-induced tau aggregation, and western blotting with brain lysate from an autopsy case. We also evaluated abnormal tau aggregation by using anti-tau antibody and PM-PBB3. RESULTS: We identified a variant, c.896_897insACA, p.K298_H299insQ, in the MAPT gene of affected patients. Similar to previous reports, most patients presented with atypical parkinsonism. Biochemical analysis revealed that the mutant tau protein had a reduced ability to polymerize microtubules and formed abnormal fibrous aggregates. Pathological study revealed frontotemporal lobe atrophy, midbrain atrophy, depigmentation of the substantia nigra, and four-repeat tau-positive inclusions in the hippocampus, brainstem, and spinal cord neurons. The inclusion bodies also stained positively with PM-PBB3. CONCLUSIONS: This study confirmed that the insACA mutation caused FTDP-17. The affected patients showed symptoms resembling Parkinson's disease initially and symptoms of progressive supranuclear palsy later. Despite the initial clinical diagnosis of frontotemporal dementia in the autopsy case, the spread of lesions could explain the process of progressive supranuclear palsy. The study of more cases in the future will help clarify the common pathogenesis of MAPT mutations or specific pathogeneses of each mutation.


Sujet(s)
Démence frontotemporale , Mutation , Protéines tau , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Encéphale/anatomopathologie , Encéphale/métabolisme , Chromosomes humains de la paire 17/génétique , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Démence frontotemporale/métabolisme , Démence frontotemporale/diagnostic , Syndromes parkinsoniens/génétique , Syndromes parkinsoniens/anatomopathologie , Syndromes parkinsoniens/métabolisme , Pedigree , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Protéines tau/génétique , Protéines tau/métabolisme
14.
Clin Neurol Neurosurg ; 240: 108261, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38599043

RÉSUMÉ

This report presents a case of Alexander disease showing clinical characteristics mimicking progressive supranuclear palsy (PSP). A 67-year-old woman complaining of motor disturbance exhibited severe atrophy of medulla, spinal cord, and midbrain tegmentum, as well as periventricular hyperintensity on cerebral MRI. Genetic analysis identified a novel in-frame deletion/insertion mutation in the exon 3 of the GFAP gene. Interestingly, neurological findings and decreased striatal uptake in dopamine transporter SPECT were suggestive of PSP. A novel GFAP gene mutation found in the present case may cause the unique clinical phenotype, which should be differentiated from PSP.


Sujet(s)
Maladie d'Alexander , Protéine gliofibrillaire acide , Imagerie par résonance magnétique , Paralysie supranucléaire progressive , Humains , Maladie d'Alexander/génétique , Maladie d'Alexander/imagerie diagnostique , Maladie d'Alexander/diagnostic , Femelle , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/imagerie diagnostique , Sujet âgé , Protéine gliofibrillaire acide/génétique , Diagnostic différentiel , Tomographie par émission monophotonique , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Mutagenèse par insertion/génétique
15.
Cogn Behav Neurol ; 37(1): 3-12, 2024 03 01.
Article de Anglais | MEDLINE | ID: mdl-38498721

RÉSUMÉ

We present a review of the definition, classification, and epidemiology of primary progressive aphasia (PPA); an update of the taxonomy of the clinical syndrome of PPA; and recent advances in the neuroanatomy, pathology, and genetics of PPA, as well as the search for biomarkers and treatment. PPA studies that have contributed to concepts of language organization and disease propagation in neurodegeneration are also reviewed. In addition, the issues of heterogeneity versus the relationships of the clinical phenotypes and their relationship to biological, pathological, and genetic advances are discussed, as is PPA's relationship to other conditions such as frontotemporal dementia, corticobasal degeneration, progressive supranuclear palsy, Pick disease, and amyotrophic lateral sclerosis. Arguments are presented in favor of considering these conditions as one entity versus many.


Sujet(s)
Aphasie progressive primaire , Démence frontotemporale , Paralysie supranucléaire progressive , Humains , Démence frontotemporale/diagnostic , Démence frontotemporale/génétique , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Syndrome
16.
Brain Pathol ; 34(4): e13250, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38418081

RÉSUMÉ

Previous studies have suggested a relationship between the number of CAG triplet repeats in the HTT gene and neurodegenerative diseases not related to Huntington's disease (HD). This study seeks to investigate whether the number of CAG repeats of HTT is associated with the risk of developing certain tauopathies and its influence as a modulator of the clinical and neuropathological phenotype. Additionally, it aims to evaluate the potential of polyglutamine staining as a neuropathological screening. We genotyped the HTT gene CAG repeat number and APOE-ℰ isoforms in a cohort of patients with neuropathological diagnoses of tauopathies (n=588), including 34 corticobasal degeneration (CBD), 98 progressive supranuclear palsy (PSP) and 456 Alzheimer's disease (AD). Furthermore, we genotyped a control group of 1070 patients, of whom 44 were neuropathologic controls. We identified significant differences in the number of patients with pathological HTT expansions in the CBD group (2.7%) and PSP group (3.2%) compared to control subjects (0.2%). A significant increase in the size of the HTT CAG repeats was found in the AD compared to the control group, influenced by the presence of the Apoliprotein E (APOE)-ℰ4 isoform. Post-mortem assessments uncovered tauopathy pathology with positive polyglutamine aggregates, with a slight predominance in the neostriatum for PSP and CBD cases and somewhat greater limbic involvement in the AD case. Our results indicated a link between HTT CAG repeat expansion with other non-HD pathology, suggesting they could share common neurodegenerative pathways. These findings support that genetic or histological screening for HTT repeat expansions should be considered in tauopathies.


Sujet(s)
Protéine huntingtine , Tauopathies , Humains , Mâle , Femelle , Sujet âgé , Tauopathies/génétique , Tauopathies/anatomopathologie , Adulte d'âge moyen , Protéine huntingtine/génétique , Sujet âgé de 80 ans ou plus , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/anatomopathologie , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Répétitions de trinucléotides/génétique , Encéphale/anatomopathologie , Expansion de trinucléotide répété/génétique , Génotype , Dégénérescence corticobasale/génétique , Dégénérescence corticobasale/anatomopathologie , Peptides
18.
Parkinsonism Relat Disord ; 119: 105962, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38134678

RÉSUMÉ

INTRODUCTION: Progressive supranuclear palsy (PSP) is an atypical parkinsonism caused by the intracerebral aggregation of the microtubule-associated protein tau (MAPT) which is encoded by MAPT gene. Although PSP is a sporadic disease, MAPT mutations have been reported in rare cases. METHODS: Among 190 patients with PSP who were recruited by the Neurodegenerative Research Group at Mayo Clinic during 2009-2023, we identified two patients who fulfilled diagnostic criteria for PSP-Richardson's syndrome (PSP-RS) and harbor novel MAPT mutations. To better investigate the potential effects of these mutations, we compared the clinical, and neuroimaging characteristics of these two patients to 20 randomly selected patients with PSP-RS without a MAPT mutation. RESULTS: MAPT c.1024G > A, p. Glu342Lys, and MAPT c.1217 G > A, p. Arg406Gln mutations were found in 2 men who developed PSP-RS with atypical features at the ages of 60 and 62 years, respectively. Glu342Lys mutation was associated with features resembling alpha-synucleinopathies (autonomic dysfunction, dream enactment behavior), while both mutations were associated with features suggestive of Alzheimer's disease with poorer performance on tests of episodic memory. Comparison of 18F-flortaucipir uptake between the two MAPT mutation cases with 20 patients without a mutation revealed increased signal on flortaucipir-PET in bilateral medial temporal lobe regions (amygdala, entorhinal cortices, hippocampus, parahippocampus) but not in PSP-related regions (globus pallidum, midbrain, superior frontal cortex and dentate nucleus of the cerebellum). CONCLUSION: Glu342Lys and Arg406Gln mutations appear to modify the PSP-RS phenotype by targeting the medial temporal lobe regions resulting in more memory loss and greater flortaucipir uptake.


Sujet(s)
Syndromes parkinsoniens , Paralysie supranucléaire progressive , Mâle , Humains , Paralysie supranucléaire progressive/imagerie diagnostique , Paralysie supranucléaire progressive/génétique , Protéines tau/génétique , Protéines tau/métabolisme , Mutation/génétique , Neuroimagerie , Syndromes parkinsoniens/imagerie diagnostique , Syndromes parkinsoniens/génétique , Phénotype
19.
J Alzheimers Dis ; 96(1): 57-64, 2023.
Article de Anglais | MEDLINE | ID: mdl-37742642

RÉSUMÉ

The first primary age-related tauopathy (PART) genome-wide association study confirmed significant associations of Alzheimer's disease (AD) and progressive supranuclear palsy (PSP) genetic variants with PART, and highlighted a novel genetic variant rs56405341. Here, we perform a comprehensive analysis of rs56405341. We found that rs56405341 was significantly associated with C4orf33 mRNA expression, but not JADE1 mRNA expression in multiple brain tissues. C4orf33 was mainly expressed in cerebellar hemisphere and cerebellum, and JADE1 was mainly expressed in thyroid, and coronary artery. Meanwhile, we found significantly downregulated C4orf33 expression both AD and PSP compared with normal controls, respectively.


Sujet(s)
Maladie d'Alzheimer , Paralysie supranucléaire progressive , Tauopathies , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Paralysie supranucléaire progressive/génétique , Paralysie supranucléaire progressive/métabolisme , Étude d'association pangénomique , ARN messager
20.
Cells ; 12(14)2023 07 24.
Article de Anglais | MEDLINE | ID: mdl-37508584

RÉSUMÉ

Frontotemporal lobar degeneration (FTLD) includes a heterogeneous group of disorders pathologically characterized by the degeneration of the frontal and temporal lobes. In addition to major genetic contributors of FTLD such as mutations in MAPT, GRN, and C9orf72, recent work has identified several epigenetic modifications including significant differential DNA methylation in DLX1, and OTUD4 loci. As aging remains one of the major risk factors for FTLD, we investigated the presence of accelerated epigenetic aging in FTLD compared to controls. We calculated epigenetic age in both peripheral blood and brain tissues of multiple FTLD subtypes using several DNA methylation clocks, i.e., DNAmClockMulti, DNAmClockHannum, DNAmClockCortical, GrimAge, and PhenoAge, and determined age acceleration and its association with different cellular proportions and clinical traits. Significant epigenetic age acceleration was observed in the peripheral blood of both frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP) patients compared to controls with DNAmClockHannum, even after accounting for confounding factors. A similar trend was observed with both DNAmClockMulti and DNAmClockCortical in post-mortem frontal cortex tissue of PSP patients and in FTLD cases harboring GRN mutations. Our findings support that increased epigenetic age acceleration in the peripheral blood could be an indicator for PSP and to a smaller extent, FTD.


Sujet(s)
Démence frontotemporale , Dégénérescence lobaire frontotemporale , Paralysie supranucléaire progressive , Humains , Dégénérescence lobaire frontotemporale/génétique , Encéphale , Paralysie supranucléaire progressive/génétique , Mutation/génétique , Ubiquitin-specific proteases
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE