Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28
Filtrer
1.
Brain Commun ; 5(1): fcac310, 2023.
Article de Anglais | MEDLINE | ID: mdl-36694576

RÉSUMÉ

Genetic frontotemporal lobar degeneration caused by autosomal dominant gene mutations provides an opportunity for targeted drug development in a highly complex and clinically heterogeneous dementia. These neurodegenerative disorders can affect adults in their middle years, progress quickly relative to other dementias, are uniformly fatal and have no approved disease-modifying treatments. Frontotemporal dementia, caused by mutations in the GRN gene which encodes the protein progranulin, is an active area of interventional drug trials that are testing multiple strategies to restore progranulin protein deficiency. These and other trials are also examining neurofilament light as a potential biomarker of disease activity and disease progression and as a therapeutic endpoint based on the assumption that cerebrospinal fluid and blood neurofilament light levels are a surrogate for neuroaxonal damage. Reports from genetic frontotemporal dementia longitudinal studies indicate that elevated concentrations of blood neurofilament light reflect disease severity and are associated with faster brain atrophy. To better inform patient stratification and treatment response in current and upcoming clinical trials, a more nuanced interpretation of neurofilament light as a biomarker of neurodegeneration is now required, one that takes into account its relationship to other pathophysiological and topographic biomarkers of disease progression from early presymptomatic to later clinically symptomatic stages.

2.
Brain Pathol ; 31(3): e12945, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33709463

RÉSUMÉ

TMEM106B has been recently implicated in multiple neurodegenerative diseases. Here, Rademakers et al. report a late-onset cerebellar Purkinje cell loss and progressive decline in motor function and gait deficits in a conventional Tmem106b-/- mouse model. By using high-power microscopy and bulk RNA sequencing, the authors further identify lysosomal and immune dysfunction as potential underlying mechanisms of the Purkinje cell loss.


Sujet(s)
Cellules de Purkinje , Animaux , Modèles animaux de maladie humaine , Souris
3.
Brain ; 143(6): 1905-1919, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32504082

RÉSUMÉ

Genetic variants that define two distinct haplotypes at the TMEM106B locus have been implicated in multiple neurodegenerative diseases and in healthy brain ageing. In frontotemporal dementia (FTD), the high expressing TMEM106B risk haplotype was shown to increase susceptibility for FTD with TDP-43 inclusions (FTD-TDP) and to modify disease penetrance in progranulin mutation carriers (FTD-GRN). To elucidate the biological function of TMEM106B and determine whether lowering TMEM106B may be a viable therapeutic strategy, we performed brain transcriptomic analyses in 8-month-old animals from our recently developed Tmem106b-/- mouse model. We included 10 Tmem106b+/+ (wild-type), 10 Tmem106b+/- and 10 Tmem106-/- mice. The most differentially expressed genes (153 downregulated and 60 upregulated) were identified between Tmem106b-/- and wild-type animals, with an enrichment for genes implicated in myelination-related cellular processes including axon ensheathment and oligodendrocyte differentiation. Co-expression analysis also revealed that the most downregulated group of correlated genes was enriched for myelination-related processes. We further detected a significant loss of OLIG2-positive cells in the corpus callosum of Tmem106b-/- mice, which was present already in young animals (21 days) and persisted until old age (23 months), without worsening. Quantitative polymerase chain reaction revealed a reduction of differentiated but not undifferentiated oligodendrocytes cellular markers. While no obvious changes in myelin were observed at the ultrastructure levels in unchallenged animals, treatment with cuprizone revealed that Tmem106b-/- mice are more susceptible to cuprizone-induced demyelination and have a reduced capacity to remyelinate, a finding which we were able to replicate in a newly generated Tmem106b CRISPR/cas9 knock-out mouse model. Finally, using a TMEM106B HeLa knock-out cell line and primary cultured oligodendrocytes, we determined that loss of TMEM106B leads to abnormalities in the distribution of lysosomes and PLP1. Together these findings reveal an important function for TMEM106B in myelination with possible consequences for therapeutic strategies aimed at lowering TMEM106B levels.


Sujet(s)
Démence frontotemporale/génétique , Démence frontotemporale/thérapie , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Animaux , Protéines de liaison à l'ADN/métabolisme , Femelle , Expression des gènes/génétique , Haplotypes , Cellules HeLa , Humains , Protéines et peptides de signalisation intercellulaire/métabolisme , Mâle , Protéines membranaires/métabolisme , Souris de lignée C57BL , Souris knockout , Mutation/génétique , Neurofibres myélinisées/anatomopathologie , Protéines de tissu nerveux/métabolisme , Polymorphisme de nucléotide simple/génétique , Transcriptome/génétique
4.
Mol Neurodegener ; 13(1): 32, 2018 06 22.
Article de Anglais | MEDLINE | ID: mdl-29929528

RÉSUMÉ

BACKGROUND: Loss of function mutations in progranulin (GRN) are a major cause of frontotemporal dementia (FTD). Progranulin is a secreted glycoprotein that localizes to lysosomes and is critical for proper lysosomal function. Heterozygous GRN mutation carriers develop FTD with TDP-43 pathology and exhibit signs of lysosomal dysfunction in the brain, with increased levels of lysosomal proteins and lipofuscin accumulation. Homozygous GRN mutation carriers develop neuronal ceroid lipofuscinosis (NCL), an earlier-onset lysosomal storage disorder caused by severe lysosomal dysfunction. Multiple genome-wide association studies have shown that risk of FTD in GRN mutation carriers is modified by polymorphisms in TMEM106B, which encodes a lysosomal membrane protein. Risk alleles of TMEM106B may increase TMEM106B levels through a variety of mechanisms. Brains from FTD patients with GRN mutations exhibit increased TMEM106B expression, and protective TMEM106B polymorphisms are associated with decreased TMEM106B expression. Together, these data raise the possibility that reduction of TMEM106B levels may protect against the pathogenic effects of progranulin haploinsufficiency. METHODS: We crossed Tmem106b +/- mice with Grn +/- mice, which model the progranulin haploinsufficiency of GRN mutation carriers and develop age-dependent social deficits and lysosomal abnormalities in the brain. We tested whether partial Tmem106b reduction could normalize the social deficits and lysosomal abnormalities of Grn +/- mice. RESULTS: Partial reduction of Tmem106b levels did not correct the social deficits of Grn +/- mice. Tmem106b reduction also failed to normalize most lysosomal abnormalities of Grn +/- mice, except for ß-glucuronidase activity, which was suppressed by Tmem106b reduction and increased by progranulin insufficiency. CONCLUSIONS: These data do not support the hypothesis that Tmem106b reduction protects against the pathogenic effects of progranulin haploinsufficiency, but do show that Tmem106b reduction normalizes some lysosomal phenotypes in Grn +/- mice.


Sujet(s)
Démence frontotemporale/génétique , Protéines et peptides de signalisation intercellulaire/génétique , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Animaux , Démence frontotemporale/anatomopathologie , Granulines , Haploinsuffisance , Hétérozygote , Souris , Souches mutantes de souris , Mutation , Polymorphisme de nucléotide simple , Progranulines
5.
Acta Neuropathol Commun ; 6(1): 42, 2018 05 31.
Article de Anglais | MEDLINE | ID: mdl-29855382

RÉSUMÉ

Loss-of-function mutations in progranulin (GRN) and a non-coding (GGGGCC)n hexanucleotide repeat expansions in C9ORF72 are the two most common genetic causes of frontotemporal lobar degeneration with aggregates of TAR DNA binding protein 43 (FTLD-TDP). TMEM106B encodes a type II transmembrane protein with unknown function. Genetic variants in TMEM106B associated with reduced TMEM106B levels have been identified as disease modifiers in individuals with GRN mutations and C9ORF72 expansions. Recently, loss of Tmem106b has been reported to protect the FTLD-like phenotypes in Grn-/- mice. Here, we generated Tmem106b-/- mice and examined whether loss of Tmem106b could rescue FTLD-like phenotypes in an AAV mouse model of C9ORF72-repeat induced toxicity. Our results showed that neither partial nor complete loss of Tmem106b was able to rescue behavioral deficits induced by the expression of (GGGGCC)66 repeats (66R). Loss of Tmem106b also failed to ameliorate 66R-induced RNA foci, dipeptide repeat protein formation and pTDP-43 pathological burden. We further found that complete loss of Tmem106b increased astrogliosis, even in the absence of 66R, and failed to rescue 66R-induced neuronal cell loss, whereas partial loss of Tmem106b significantly rescued the neuronal cell loss but not neuroinflammation induced by 66R. Finally, we showed that overexpression of 66R did not alter expression of Tmem106b and other lysosomal genes in vivo, and subsequent analyses in vitro found that transiently knocking down C9ORF72, but not overexpression of 66R, significantly increased TMEM106B and other lysosomal proteins. In summary, reducing Tmem106b levels failed to rescue FTLD-like phenotypes in a mouse model mimicking the toxic gain-of-functions associated with overexpression of 66R. Combined with the observation that loss of C9ORF72 and not 66R overexpression was associated with increased levels of TMEM106B, this work suggests that the protective TMEM106B haplotype may exert its effect in expansion carriers by counteracting lysosomal dysfunction resulting from a loss of C9ORF72.


Sujet(s)
Protéine C9orf72/génétique , Expansion de séquence répétée de l'ADN/génétique , Dégénérescence lobaire frontotemporale/génétique , Dégénérescence lobaire frontotemporale/thérapie , Régulation de l'expression des gènes/génétique , Protéines membranaires/déficit , Protéines suppresseurs de tumeurs/déficit , Animaux , Protéine C9orf72/métabolisme , Lignée de cellules transformées , Conditionnement psychologique/physiologie , Modèles animaux de maladie humaine , Comportement d'exploration , Peur/psychologie , Dégénérescence lobaire frontotemporale/psychologie , Vecteurs génétiques/génétique , Vecteurs génétiques/métabolisme , Glycérophosphate , Humains , Relations interpersonnelles , Protéines membranaires/génétique , Souris , Souris de lignée C57BL , Souris knockout , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction génétique , Protéines suppresseurs de tumeurs/génétique
6.
Acta Neuropathol Commun ; 5(1): 96, 2017 Dec 07.
Article de Anglais | MEDLINE | ID: mdl-29216908

RÉSUMÉ

Mutations in the stress granule protein T-cell restricted intracellular antigen 1 (TIA1) were recently shown to cause amyotrophic lateral sclerosis (ALS) with or without frontotemporal dementia (FTD). Here, we provide detailed clinical and neuropathological descriptions of nine cases with TIA1 mutations, together with comparisons to sporadic ALS (sALS) and ALS due to repeat expansions in C9orf72 (C9orf72+). All nine patients with confirmed mutations in TIA1 were female. The clinical phenotype was heterogeneous with a range in the age at onset from late twenties to the eighth decade (mean = 60 years) and disease duration from one to 6 years (mean = 3 years). Initial presentation was either focal weakness or language impairment. All affected individuals received a final diagnosis of ALS with or without FTD. No psychosis or parkinsonism was described. Neuropathological examination on five patients found typical features of ALS and frontotemporal lobar degeneration (FTLD-TDP, type B) with anatomically widespread TDP-43 proteinopathy. In contrast to C9orf72+ cases, caudate atrophy and hippocampal sclerosis were not prominent. Detailed evaluation of the pyramidal motor system found a similar degree of neurodegeneration and TDP-43 pathology as in sALS and C9orf72+ cases; however, cases with TIA1 mutations had increased numbers of lower motor neurons containing round eosinophilic and Lewy body-like inclusions on HE stain and round compact cytoplasmic inclusions with TDP-43 immunohistochemistry. Immunohistochemistry and immunofluorescence failed to demonstrate any labeling of inclusions with antibodies against TIA1. In summary, our TIA1 mutation carriers developed ALS with or without FTD, with a wide range in age at onset, but without other neurological or psychiatric features. The neuropathology was characterized by widespread TDP-43 pathology, but a more restricted pattern of neurodegeneration than C9orf72+ cases. Increased numbers of round eosinophilic and Lewy-body like inclusions in lower motor neurons may be a distinctive feature of ALS caused by TIA1 mutations.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Mutation/génétique , Antigène intracellulaire-1 des lymphocytes T/génétique , Adulte , Sujet âgé , Sclérose latérale amyotrophique/complications , Autopsie , Protéine C9orf72/génétique , Protéines de liaison à l'ADN/métabolisme , Santé de la famille , Femelle , Démence frontotemporale/complications , Humains , Mâle , Adulte d'âge moyen , Neuropathologie
7.
Neuron ; 95(4): 808-816.e9, 2017 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-28817800

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are age-related neurodegenerative disorders with shared genetic etiologies and overlapping clinical and pathological features. Here we studied a novel ALS/FTD family and identified the P362L mutation in the low-complexity domain (LCD) of T cell-restricted intracellular antigen-1 (TIA1). Subsequent genetic association analyses showed an increased burden of TIA1 LCD mutations in ALS patients compared to controls (p = 8.7 × 10-6). Postmortem neuropathology of five TIA1 mutations carriers showed a consistent pathological signature with numerous round, hyaline, TAR DNA-binding protein 43 (TDP-43)-positive inclusions. TIA1 mutations significantly increased the propensity of TIA1 protein to undergo phase transition. In live cells, TIA1 mutations delayed stress granule (SG) disassembly and promoted the accumulation of non-dynamic SGs that harbored TDP-43. Moreover, TDP-43 in SGs became less mobile and insoluble. The identification of TIA1 mutations in ALS/FTD reinforces the importance of RNA metabolism and SG dynamics in ALS/FTD pathogenesis.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Mutation/génétique , Protéines de liaison au poly(A)/génétique , Adulte , Sujet âgé , Protéines de liaison à l'ADN/métabolisme , Santé de la famille , Femelle , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Cellules HeLa , Ribonucléoprotéine nucléaire hétérogène A1 , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/métabolisme , Humains , Mâle , Microscopie confocale , Adulte d'âge moyen , Protéine FUS de liaison à l'ARN/métabolisme , Stress physiologique/physiologie , Antigène intracellulaire-1 des lymphocytes T , Facteurs temps , Transfection
8.
Neurol Genet ; 3(4): e161, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28660252

RÉSUMÉ

OBJECTIVE: We performed a genome-wide brain expression study to reveal the underpinnings of diseases linked to a repeat expansion in chromosome 9 open reading frame 72 (C9ORF72). METHODS: The genome-wide expression profile was investigated in brain tissue obtained from C9ORF72 expansion carriers (n = 32), patients without this expansion (n = 30), and controls (n = 20). Using quantitative real-time PCR, findings were confirmed in our entire pathologic cohort of expansion carriers (n = 56) as well as nonexpansion carriers (n = 31) and controls (n = 20). RESULTS: Our findings were most profound in the cerebellum, where we identified 40 differentially expressed genes, when comparing expansion carriers to patients without this expansion, including 22 genes that have a homeobox (e.g., HOX genes) and/or are located within the HOX gene cluster (top hit: homeobox A5 [HOXA5]). In addition to the upregulation of multiple homeobox genes that play a vital role in neuronal development, we noticed an upregulation of transthyretin (TTR), an extracellular protein that is thought to be involved in neuroprotection. Pathway analysis aligned with these findings and revealed enrichment for gene ontology processes involved in (anatomic) development (e.g., organ morphogenesis). Additional analyses uncovered that HOXA5 and TTR levels are associated with C9ORF72 variant 2 levels as well as with intron-containing transcript levels, and thus, disease-related changes in those transcripts may have triggered the upregulation of HOXA5 and TTR. CONCLUSIONS: In conclusion, our identification of genes involved in developmental processes and neuroprotection sheds light on potential compensatory mechanisms influencing the occurrence, presentation, and/or progression of C9ORF72-related diseases.

9.
Exp Gerontol ; 84: 96-100, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27612602

RÉSUMÉ

Mutations in progranulin are a major cause of frontotemporal lobe degeneration (FTLD). Hence, plasma progranulin is an attractive biomarker in FTLD but poorly reflects levels in cerebrospinal fluid (CSF), suggesting tissue-specific regulation of progranulin levels. Sortilin was recently identified as a progranulin scavenger receptor that destines it for lysosomal degradation. Proteolysis or alternative splicing generates soluble sortilin variants that retain progranulin binding and potentially functions as a decoy receptor. In the present study, we analyzed soluble sortilin and progranulin in plasma and CSF in 341 aging individuals. We found that soluble sortilin exists in CSF in ten-fold molar excess compared to progranulin and observed a highly significant positive correlation between soluble sortilin and progranulin levels in CSF but not in plasma. However, carriers of the minor allele of SNP rs646776 in SORT1 encoding sortilin displayed significantly increased soluble sortilin and reduced progranulin specifically in plasma but not in CSF. Taken together, our findings suggest that soluble sortilin may affect progranulin levels in both a tissue-specific and genotype-dependent manner.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/génétique , Vieillissement/génétique , Protéines et peptides de signalisation intercellulaire/liquide cérébrospinal , Protéines adaptatrices du transport vésiculaire/sang , Protéines adaptatrices du transport vésiculaire/liquide cérébrospinal , Sujet âgé , Sujet âgé de 80 ans ou plus , Vieillissement/sang , Vieillissement/liquide cérébrospinal , Marqueurs biologiques/sang , Marqueurs biologiques/liquide cérébrospinal , Danemark , Femelle , Dégénérescence lobaire frontotemporale/génétique , Génotype , Humains , Modèles linéaires , Mâle , Mutation , Polymorphisme de nucléotide simple , Progranulines
10.
Acta Neuropathol ; 132(5): 639-651, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27543298

RÉSUMÉ

Frontotemporal lobar degeneration is a neurodegenerative disorder affecting over 50,000 people in the United States alone. The most common pathological subtype of FTLD is the presence of ubiquitinated TAR DNA binding protein 43 (TDP-43) accumulations in frontal and temporal brain regions at autopsy. While some cases of FTLD-TDP can be attributed to the inheritance of disease-causing mutations, the majority of cases arise with no known genetic cause. In 2010, the first genome-wide association study was conducted in patients with FTLD-TDP to determine potential genetic risk factors for this homogenous subgroup of dementia patients, leading to the identification of the TMEM106B locus on chromosome 7. In this manuscript, we review the initial discovery and replication studies describing TMEM106B variants as disease risk factors and modifiers in TDP-43 proteinopathies, such as FTLD-TDP caused by progranulin (GRN) or chromosome 9 open reading frame 72 (C9orf72) mutations, as well as Alzheimer's disease and hippocampal sclerosis. We further summarize what is currently known about the previously uncharacterized TMEM106B protein and its role as a potential regulator of lysosomal function, and we discuss how modifying TMEM106B levels might uncover promising therapeutic strategies for individuals suffering from TDP-43 proteinopathy.


Sujet(s)
Démence frontotemporale/génétique , Prédisposition génétique à une maladie/génétique , Variation génétique/génétique , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Chromosomes humains de la paire 7/génétique , Protéines de liaison à l'ADN/génétique , Génotype , Humains , Facteurs de risque
11.
Nat Commun ; 7: 11992, 2016 06 30.
Article de Anglais | MEDLINE | ID: mdl-27356620

RÉSUMÉ

Progranulin (GRN) loss-of-function mutations leading to progranulin protein (PGRN) haploinsufficiency are prevalent genetic causes of frontotemporal dementia. Reports also indicated PGRN-mediated neuroprotection in models of Alzheimer's and Parkinson's disease; thus, increasing PGRN levels is a promising therapeutic for multiple disorders. To uncover novel PGRN regulators, we linked whole-genome sequence data from 920 individuals with plasma PGRN levels and identified the prosaposin (PSAP) locus as a new locus significantly associated with plasma PGRN levels. Here we show that both PSAP reduction and overexpression lead to significantly elevated extracellular PGRN levels. Intriguingly, PSAP knockdown increases PGRN monomers, whereas PSAP overexpression increases PGRN oligomers, partly through a protein-protein interaction. PSAP-induced changes in PGRN levels and oligomerization replicate in human-derived fibroblasts obtained from a GRN mutation carrier, further supporting PSAP as a potential PGRN-related therapeutic target. Future studies should focus on addressing the relevance and cellular mechanism by which PGRN oligomeric species provide neuroprotection.


Sujet(s)
Démence frontotemporale/génétique , Protéines et peptides de signalisation intercellulaire/génétique , Saposines/génétique , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Animaux , Démence frontotemporale/métabolisme , Techniques de knock-down de gènes , Haploinsuffisance , Cellules HeLa , Humains , Protéines et peptides de signalisation intercellulaire/métabolisme , Souris , Souris knockout , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Polymorphisme de nucléotide simple , Progranulines , Cartes d'interactions protéiques
13.
Acta Neuropathol ; 130(1): 77-92, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25943890

RÉSUMÉ

Frontotemporal lobar degeneration with TAR DNA-binding protein 43 inclusions (FTLD-TDP) is the most common pathology associated with frontotemporal dementia (FTD). Repeat expansions in chromosome 9 open reading frame 72 (C9ORF72) and mutations in progranulin (GRN) are the major known genetic causes of FTLD-TDP; however, the genetic etiology in the majority of FTLD-TDP remains unexplained. In this study, we performed whole-genome sequencing in 104 pathologically confirmed FTLD-TDP patients from the Mayo Clinic brain bank negative for C9ORF72 and GRN mutations and report on the contribution of rare single nucleotide and copy number variants in 21 known neurodegenerative disease genes. Interestingly, we identified 5 patients (4.8 %) with variants in optineurin (OPTN) and TANK-binding kinase 1 (TBK1) that are predicted to be highly pathogenic, including two double mutants. Case A was a compound heterozygote for mutations in OPTN, carrying the p.Q235* nonsense and p.A481V missense mutation in trans, while case B carried a deletion of OPTN exons 13-15 (p.Gly538Glufs*27) and a loss-of-function mutation (p.Arg117*) in TBK1. Cases C-E carried heterozygous missense mutations in TBK1, including the p.Glu696Lys mutation which was previously reported in two amyotrophic lateral sclerosis (ALS) patients and is located in the OPTN binding domain. Quantitative mRNA expression and protein analysis in cerebellar tissue showed a striking reduction of OPTN and/or TBK1 expression in 4 out of 5 patients supporting pathogenicity in these specific patients and suggesting a loss-of-function disease mechanism. Importantly, neuropathologic examination showed FTLD-TDP type A in the absence of motor neuron disease in 3 pathogenic mutation carriers. In conclusion, we highlight TBK1 as an important cause of pure FTLD-TDP, identify the first OPTN mutations in FTLD-TDP, and suggest a potential oligogenic basis for at least a subset of FTLD-TDP patients. Our data further add to the growing body of evidence linking ALS and FTD and suggest a key role for the OPTN/TBK1 pathway in these diseases.


Sujet(s)
Dégénérescence lobaire frontotemporale/génétique , Mutation , Protein-Serine-Threonine Kinases/génétique , Facteur de transcription TFIIIA/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Technique de Western , Encéphale/métabolisme , Encéphale/anatomopathologie , Protéines du cycle cellulaire , Études de cohortes , Variations de nombre de copies de segment d'ADN , Femelle , Dégénérescence lobaire frontotemporale/métabolisme , Dégénérescence lobaire frontotemporale/anatomopathologie , Expression des gènes , Humains , Immunohistochimie , Mâle , Protéines de transport membranaire , Maladies du motoneurone/génétique , Maladies du motoneurone/métabolisme , Maladies du motoneurone/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , ARN messager/métabolisme , Analyse de séquence d'ADN , Facteur de transcription TFIIIA/métabolisme
14.
Neurology ; 82(21): 1871-8, 2014 May 27.
Article de Anglais | MEDLINE | ID: mdl-24771538

RÉSUMÉ

OBJECTIVE: We aimed to investigate the relationship between plasma and CSF progranulin (PGRN) levels. METHODS: Plasma and CSF PGRN were measured in a cohort of 345 subjects from the Mayo Clinic Study of Aging by ELISA. Single nucleotide polymorphism genotyping was performed using TaqMan assays. Associations between PGRN and sex, age at sample collection, diagnosis, single nucleotide polymorphism genotypes (GRN, SORT1, and APOE), and Pittsburgh compound B score were explored separately in CSF and plasma using single variable linear regression models. Pearson partial correlation coefficient was used to estimate the correlation of PGRN in CSF and plasma. RESULTS: Plasma (p = 0.0031) and CSF (p = 0.0044) PGRN significantly increased with age, whereas plasma PGRN levels were 7% lower (p = 0.0025) and CSF PGRN levels 5% higher (p = 0.0024) in male compared with female participants. Correcting for age and sex, higher plasma PGRN was associated with higher CSF PGRN (partial r = 0.17, p = 0.004). In plasma, both rs5848 (GRN; p = 0.002) and rs646776 (SORT1; p = 3.56E-7) were associated with PGRN, while only rs5848 showed highly significant association in CSF (p = 5.59E-14). Age, sex, rs5848 genotype, and plasma PGRN together accounted for only 18% of the variability observed in CSF PGRN. CONCLUSIONS: While some correlation exists between plasma and CSF PGRN, age, sex, and genetic factors differently affect PGRN levels. Therefore, caution should be taken when using plasma PGRN to predict PGRN changes in the brain. These findings further highlight that plasma PGRN levels may not accurately predict clinical features or response to future frontotemporal lobar degeneration therapies.


Sujet(s)
Protéines et peptides de signalisation intercellulaire/analyse , Protéines et peptides de signalisation intercellulaire/génétique , Polymorphisme de nucléotide simple , Facteurs âges , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Études d'associations génétiques , Humains , Protéines et peptides de signalisation intercellulaire/sang , Protéines et peptides de signalisation intercellulaire/liquide cérébrospinal , Mâle , Progranulines , Facteurs sexuels
15.
Acta Neuropathol ; 127(3): 397-406, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24385136

RÉSUMÉ

Variants in transmembrane protein 106 B (TMEM106B) modify the disease penetrance of frontotemporal dementia (FTD) in carriers of progranulin (GRN) mutations. We investigated whether TMEM106B is also a genetic modifier of disease in carriers of chromosome 9 open reading frame 72 (C9ORF72) expansions. We assessed the genotype of 325 C9ORF72 expansion carriers (cohort 1), 586 FTD patients lacking C9ORF72 expansions [with or without motor neuron disease (MND); cohort 2], and a total of 1,302 controls for TMEM106B variants (rs3173615 and rs1990622) using MassArray iPLEX and Taqman genotyping assays. For our primary analysis, we focused on functional variant rs3173615, and employed a recessive genotypic model. In cohort 1, patients with C9ORF72 expansions showed a significantly reduced frequency of carriers homozygous for the minor allele as compared to controls [11.9 vs. 19.1 %, odds ratio (OR) 0.57, p = 0.014; same direction as carriers of GRN mutations]. The strongest evidence was provided by FTD patients (OR 0.33, p = 0.009) followed by FTD/MND patients (OR 0.38, p = 0.017), whereas no significant difference was observed in MND patients (OR 0.85, p = 0.55). In cohort 2, the frequency of carriers homozygous for the minor allele was not significantly reduced in patients as compared to controls (OR 0.77, p = 0.079); however, a significant reduction was observed when focusing on those patients with frontotemporal lobar degeneration and TAR DNA-binding protein 43 inclusions (FTLD-TDP; OR 0.26, p < 0.001). Our study identifies TMEM106B as the first genetic factor modifying disease presentation in C9ORF72 expansion carriers. Homozygosity for the minor allele protects carriers from developing FTD, but not from developing MND; similar effects are seen in FTLD-TDP patients with yet unknown genetic causes. These new findings show that the protective effects of TMEM106B are not confined to carriers of GRN mutations and might be relevant for prognostic testing, and as a promising therapeutic target for the entire spectrum of FTLD-TDP.


Sujet(s)
Démence frontotemporale/génétique , Protéines membranaires/génétique , Protéines de tissu nerveux/génétique , Protéines/génétique , Adulte , Âge de début , Sujet âgé , Sujet âgé de 80 ans ou plus , Allèles , Protéine C9orf72 , Études de cohortes , Expansion de séquence répétée de l'ADN , Protéines de liaison à l'ADN/métabolisme , Femelle , Démence frontotemporale/complications , Démence frontotemporale/métabolisme , Prédisposition génétique à une maladie , Génotype , Hétérozygote , Humains , Mâle , Adulte d'âge moyen , Modèles génétiques , Maladies du motoneurone/complications , Maladies du motoneurone/génétique , Maladies du motoneurone/métabolisme , Polymorphisme de nucléotide simple
16.
Parkinsonism Relat Disord ; 19(10): 869-77, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-23787135

RÉSUMÉ

Atypical Parkinsonism associated with white matter pathology has been described in cerebrovascular diseases, mitochondrial cytopathies, osmotic demyelinating disorders, leukoencephalopathies leukodystrophies, and others. Hereditary diffuse leukoencephalopathy with spheroids (HDLS) is an autosomal dominant disorder with symptomatic onset in midlife and death within a few years after symptom onset. Neuroimaging reveals cerebral white matter lesions that are pathologically characterized by non-inflammatory myelin loss, reactive astrocytosis, and axonal spheroids. Most cases are caused by mutations in the colony-stimulating factor 1 receptor (CSF1R) gene. We studied neuropathologically verified HDLS patients with CSF1R mutations to assess parkinsonian features. Ten families were evaluated with 16 affected individuals. During the course of the illness, all patients had at least some degree of bradykinesia. Fifteen patients had postural instability, and seven had rigidity. Two patients initially presented with parkinsonian gait and asymmetrical bradykinesia. These two patients and two others exhibited bradykinesia, rigidity, postural instability, and tremor (two with resting) early in the course of the illness. Levodopa/carbidopa therapy in these four patients provided no benefit, and the remaining 12 patients were not treated. The mean age of onset for all patients was about 45 years (range, 18-71) and the mean disease duration was approximately six years (range, 3-11). We also reviewed HDLS patients published prior to the CSF1R discovery for the presence of parkinsonian features. Out of 50 patients, 37 had gait impairments, 8 rigidity, 7 bradykinesia, and 5 resting tremor. Our report emphasizes the presence of atypical Parkinsonism in HDLS due to CSF1R mutations.


Sujet(s)
Gliose/congénital , Leucoencéphalopathies/génétique , Leucoencéphalopathies/anatomopathologie , Mutation/génétique , Mutation/physiologie , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Récepteur du facteur de stimulation des colonies de macrophages/génétique , Adolescent , Adulte , Âge de début , Sujet âgé , Antiparkinsoniens/usage thérapeutique , Biobanques , Encéphale/anatomopathologie , Famille , Femelle , Troubles neurologiques de la marche/étiologie , Gliose/complications , Gliose/génétique , Gliose/anatomopathologie , Humains , Hypocinésie/étiologie , Traitement d'image par ordinateur , Leucoencéphalopathies/complications , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Raideur musculaire/étiologie , Raideur musculaire/physiopathologie , Neuroimagerie , Maladie de Parkinson/étiologie , Récepteur du facteur de stimulation des colonies de macrophages/physiologie , Tremblement/étiologie , Royaume-Uni , Jeune adulte
17.
J Neurochem ; 126(6): 781-91, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23742080

RÉSUMÉ

Frontotemporal lobar degeneration (FTLD) is the second leading cause of dementia in individuals under age 65. In many patients, the predominant pathology includes neuronal cytoplasmic or intranuclear inclusions of ubiquitinated TAR DNA binding protein 43 (FTLD-TDP). Recently, a genome-wide association study identified the first FTLD-TDP genetic risk factor, in which variants in and around the TMEM106B gene (top SNP rs1990622) were significantly associated with FTLD-TDP risk. Intriguingly, the most significant association was in FTLD-TDP patients carrying progranulin (GRN) mutations. Here, we investigated to what extent the coding variant, rs3173615 (p.T185S) in linkage disequilibrium with rs1990622, affects progranulin protein (PGRN) biology and transmembrane protein 106 B (TMEM106B) regulation. First, we confirmed the association of TMEM106B variants with FTLD-TDP in a new cohort of GRN mutation carriers. We next generated and characterized a TMEM106B-specific antibody for investigation of this protein. Enzyme-linked immunoassay analysis of progranulin protein levels showed similar effects upon T185 and S185 TMEM106B over-expression. However, over-expression of T185 consistently led to higher TMEM106B protein levels than S185. Cycloheximide treatment experiments revealed that S185 degrades faster than T185 TMEM106B, potentially due to differences in N-glycosylation at residue N183. Together, our results provide a potential mechanism by which TMEM106B variants lead to differences in FTLD-TDP risk. We studied the p.T185S TMEM106B genetic variant previously implicated in frontotemporal dementia with TAR DNA binding protein 43 pathology caused by progranulin mutations. Our cell culture studies provide evidence that the protective S185 isoform is degraded more rapidly than T185 TMEM106B, potentially due to differences in glycosylation. These findings suggest that low TMEM106B levels might protect against FTLD-TDP in these patients.


Sujet(s)
Démence frontotemporale/génétique , Démence frontotemporale/métabolisme , Protéines membranaires/génétique , Protéines membranaires/physiologie , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/physiologie , Sujet âgé , Technique de Western , Cellules cultivées , Études de cohortes , ADN complémentaire/biosynthèse , ADN complémentaire/génétique , Test ELISA , Femelle , Technique d'immunofluorescence , Génotype , Glycosylation , Cellules HeLa , Humains , Immunohistochimie , Protéines et peptides de signalisation intercellulaire/biosynthèse , Protéines et peptides de signalisation intercellulaire/génétique , Isomérie , Lysosomes/métabolisme , Mâle , Mutagenèse/génétique , Mutation/génétique , Polymorphisme génétique/génétique , Polymorphisme de nucléotide simple/génétique , Progranulines , Réaction de polymérisation en chaine en temps réel
18.
Neurology ; 80(11): 1033-40, 2013 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-23408870

RÉSUMÉ

OBJECTIVE: Pigmented orthochromatic leukodystrophy (POLD) and hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS) are rare neurodegenerative disorders characterized by cerebral white matter abnormalities, myelin loss, and axonal swellings. The striking overlap of clinical and pathologic features of these disorders suggested a common pathogenesis; however, no genetic or mechanistic link between POLD and HDLS has been established. Recently, we reported that mutations in the colony-stimulating factor 1 receptor (CSF1R) gene cause HDLS. In this study, we determined whether CSF1R mutations are also a cause of POLD. METHODS: We performed sequencing of CSF1R in 2 pathologically confirmed POLD families. For the largest family (FTD368), a detailed case report was provided and brain samples from 2 affected family members previously diagnosed with POLD were re-evaluated to determine whether they had HDLS features. In vitro functional characterization of wild-type and mutant CSF1R was also performed. RESULTS: We identified CSF1R mutations in both POLD families: in family 5901, we found c.2297T>C (p.M766T), previously reported by us in HDLS family CA1, and in family FTD368, we identified c.2345G>A (p.R782H), recently reported in a biopsy-proven HDLS case. Immunohistochemical examination in family FTD368 showed the typical neuronal and glial findings of HDLS. Functional analyses of CSF1R mutant p.R782H (identified in this study) and p.M875T (previously observed in HDLS), showed a similar loss of CSF1R autophosphorylation of selected tyrosine residues in the kinase domain for both mutations when compared with wild-type CSF1R. CONCLUSIONS: We provide the first genetic and mechanistic evidence that POLD and HDLS are a single clinicopathologic entity.


Sujet(s)
Liaison génétique/génétique , Leucodystrophie à cellules globoïdes/génétique , Leucoencéphalopathies/génétique , Troubles de la pigmentation/génétique , Récepteur du facteur de stimulation des colonies de macrophages/génétique , Femelle , Cellules HeLa , Humains , Leucodystrophie à cellules globoïdes/diagnostic , Leucoencéphalopathies/diagnostic , Adulte d'âge moyen , Mutation , Troubles de la pigmentation/diagnostic
19.
Neurology ; 79(6): 566-74, 2012 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-22843259

RÉSUMÉ

OBJECTIVE: To describe the brain MRI characteristics of hereditary diffuse leukoencephalopathy with spheroids (HDLS) with known mutations in the colony-stimulating factor 1 receptor gene (CSF1R) on chromosome 5. METHODS: We reviewed 20 brain MRI scans of 15 patients with autopsy- or biopsy-verified HDLS and CSF1R mutations. We assessed sagittal T1-, axial T1-, T2-, proton density-weighted and axial fluid-attenuated inversion recovery images for distribution of white matter lesions (WMLs), gray matter involvement, and atrophy. We calculated a severity score based on a point system (0-57) for each MRI scan. RESULTS: Of the patients, 93% (14 of 15) demonstrated localized WMLs with deep and subcortical involvement, whereas one patient revealed generalized WMLs. All WMLs were bilateral but asymmetric and predominantly frontal. Fourteen patients had a rapidly progressive clinical course with an initial MRI mean total severity score of 16.7 points (range 10-33.5). Gray matter pathology and brainstem atrophy were absent, and the corticospinal tracts were involved late in the disease course. There was no enhancement, and there was minimal cerebellar pathology. CONCLUSION: Recognition of the typical MRI patterns of HDLS and the use of an MRI severity score might help during the diagnostic evaluation to characterize the natural history and to monitor potential future treatments. Indicators of rapid disease progression were symptomatic disease onset before 45 years, female sex, WMLs extending beyond the frontal regions, a MRI severity score greater than 15 points, and mutation type of deletion.


Sujet(s)
Encéphale/anatomopathologie , Leucoencéphalopathies/classification , Leucoencéphalopathies/génétique , Leucoencéphalopathies/anatomopathologie , Mutation , Récepteur du facteur de stimulation des colonies de macrophages/génétique , Atrophie/étiologie , Atrophie/anatomopathologie , Femelle , Humains , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen
20.
Alzheimers Res Ther ; 4(1): 4, 2012 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-22277331

RÉSUMÉ

Frontotemporal lobar degeneration (FTLD) is a devastating neurodegenerative disease that is the second most common form of dementia affecting individuals under age 65. The most common pathological subtype, FTLD with transactive response DNA-binding protein with a molecular weight of 43 kDa inclusions (FTLD-TDP), is often caused by autosomal dominant mutations in the progranulin gene (GRN) encoding the progranulin protein (PGRN). GRN pathogenic mutations result in haploinsufficiency, usually by nonsense-mediated decay of the mRNA. Since the discovery of these mutations in 2006, several groups have published data and animal models that provide further insight into the genetic and functional relevance of PGRN in the context of FTLD-TDP. These studies were critical in initiating our understanding of the role of PGRN in neural development, degeneration, synaptic transmission, cell signaling, and behavior. Furthermore, recent publications have now identified the receptors for PGRN, which will hopefully lead to additional therapeutic targets. Additionally, drug screens have been conducted to identify pharmacological regulators of PGRN levels to be used as potential treatments for PGRN haploinsufficiency. Here we review recent literature describing relevant data on GRN genetics, cell culture experiments describing the potential role and regulators of PGRN in the central nervous system, animal models of PGRN deficiency, and potential PGRN-related FTLD therapies that are currently underway. The present review aims to underscore the necessity of further elucidation of PGRN biology in FTLD-related neurodegeneration.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE