Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Infect Dis ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195164

RESUMEN

The varicella-zoster virus (VZV) infects over 95% of the population. VZV reactivation causes herpes zoster (HZ), known as shingles, primarily affecting the elderly and immunocompromised individuals. However, HZ can also occur in otherwise healthy individuals. We analyzed the immune signature and risk profile in HZ patients using a genome-wide association study across different UK Biobank HZ cohorts. Additionally, we conducted one of the largest HZ HLA association studies to date, coupled with transcriptomic analysis of pathways underlying HZ susceptibility. Our findings highlight the significance of the MHC locus for HZ development, identifying five protective and four risk HLA alleles. This demonstrates that HZ susceptibility is largely governed by variations in the MHC. Furthermore, functional analyses revealed the upregulation of type I interferon and adaptive immune responses. These findings provide fresh molecular insights into the pathophysiology and the activation of innate and adaptive immune responses triggered by symptomatic VZV reactivation.

2.
Acta Neuropathol ; 137(6): 901-918, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30874922

RESUMEN

Emerging evidence suggested a converging mechanism in neurodegenerative brain diseases (NBD) involving early neuronal network dysfunctions and alterations in the homeostasis of neuronal firing as culprits of neurodegeneration. In this study, we used paired-end short-read and direct long-read whole genome sequencing to investigate an unresolved autosomal dominant dementia family significantly linked to 7q36. We identified and validated a chromosomal inversion of ca. 4 Mb, segregating on the disease haplotype and disrupting the coding sequence of dipeptidyl-peptidase 6 gene (DPP6). DPP6 resequencing identified significantly more rare variants-nonsense, frameshift, and missense-in early-onset Alzheimer's disease (EOAD, p value = 0.03, OR = 2.21 95% CI 1.05-4.82) and frontotemporal dementia (FTD, p = 0.006, OR = 2.59, 95% CI 1.28-5.49) patient cohorts. DPP6 is a type II transmembrane protein with a highly structured extracellular domain and is mainly expressed in brain, where it binds to the potassium channel Kv4.2 enhancing its expression, regulating its gating properties and controlling the dendritic excitability of hippocampal neurons. Using in vitro modeling, we showed that the missense variants found in patients destabilize DPP6 and reduce its membrane expression (p < 0.001 and p < 0.0001) leading to a loss of protein. Reduced DPP6 and/or Kv4.2 expression was also detected in brain tissue of missense variant carriers. Loss of DPP6 is known to cause neuronal hyperexcitability and behavioral alterations in Dpp6-KO mice. Taken together, the results of our genomic, genetic, expression and modeling analyses, provided direct evidence supporting the involvement of DPP6 loss in dementia. We propose that loss of function variants have a higher penetrance and disease impact, whereas the missense variants have a variable risk contribution to disease that can vary from high to low penetrance. Our findings of DPP6, as novel gene in dementia, strengthen the involvement of neuronal hyperexcitability and alteration in the homeostasis of neuronal firing as a disease mechanism to further investigate.


Asunto(s)
Inversión Cromosómica , Demencia/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/deficiencia , Mutación , Proteínas del Tejido Nervioso/deficiencia , Enfermedades Neurodegenerativas/genética , Neuronas/fisiología , Canales de Potasio/deficiencia , Potenciales de Acción/fisiología , Adulto , Anciano , Cromosomas Humanos Par 7/genética , Demencia/fisiopatología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Femenino , Genes Dominantes , Homeostasis , Humanos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Penetrancia , Polimorfismo de Nucleótido Simple , Canales de Potasio/genética , Canales de Potasio/fisiología , Estabilidad Proteica , Transporte de Proteínas , Transmisión Sináptica , Secuenciación Completa del Genoma
3.
Brain ; 139(Pt 2): 452-67, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26674655

RESUMEN

We identified in a cohort of patients with frontotemporal dementia (n = 481) or amyotrophic lateral sclerosis (n = 147), 10 index patients carrying a TBK1 loss of function mutation reducing TBK1 expression by 50%. Here, we describe the clinical and pathological characteristics of the 10 index patients and six of their affected relatives carrying a TBK1 mutation. Six TBK1 carriers were diagnosed with frontotemporal dementia, seven with amyotrophic lateral sclerosis, one with both clinical phenotypes and two with dementia unspecified. The mean age at onset of all 16 TBK1 carriers was 62.1 ± 8.9 years (range 41-73) with a mean disease duration of 4.7 ± 4.5 years (range 1-13). TBK1 carriers with amyotrophic lateral sclerosis had shorter disease duration than carriers with frontotemporal dementia. Six of seven TBK1 carriers were diagnosed with the behavioural variant of frontotemporal dementia, presenting predominantly as disinhibition. Memory loss was an important associated symptom in the initial phase of the disease in all but one of the carriers with frontotemporal dementia. Three of the patients with amyotrophic lateral sclerosis exhibited pronounced upper motor neuron symptoms. Overall, neuroimaging displayed widespread atrophy, both symmetric and asymmetric. Brain perfusion single-photon emission computed tomography or fluorodeoxyglucose-positron emission tomography showed asymmetric and predominantly frontotemporal involvement. Neuropathology in two patients demonstrated TDP-43 type B pathology. Further, we compared genotype-phenotype data of TBK1 carriers with frontotemporal dementia (n = 7), with those of frontotemporal dementia patients with a C9orf72 repeat expansion (n = 65) or a GRN mutation (n = 52) and with frontotemporal dementia patients (n = 259) negative for mutations in currently known causal genes. TBK1 carriers with frontotemporal dementia had a later age at onset (63.3 years) than C9orf72 carriers (54.3 years) (P = 0.019). In clear contrast with TBK1 carriers, GRN carriers were more often diagnosed with the language variant than the behavioural variant, and presented in case of the diagnosis of behavioural variant, more often than TBK1 carriers with apathy as the predominant characteristic (P = 0.004). Also, TBK1 carriers exhibited more often extrapyramidal symptoms than C9orf72 carriers (P = 0.038). In conclusion, our study identified clinical differences between the TBK1, C9orf72 and GRN carriers, which allows us to formulate guidelines for genetic diagnosis. After a negative result for C9orf72, patients with both frontotemporal dementia and amyotrophic lateral sclerosis should be tested first for mutations in TBK1. Specifically in frontotemporal dementia patients with early memory difficulties, a relatively late age at onset or extrapyramidal symptoms, screening for TBK1 mutations should be considered.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Demencia Frontotemporal/genética , Heterocigoto , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas/genética , Adulto , Anciano , Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/epidemiología , Bélgica/epidemiología , Proteína C9orf72 , Estudios de Cohortes , Femenino , Demencia Frontotemporal/diagnóstico , Demencia Frontotemporal/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje , Progranulinas
4.
Acta Neuropathol ; 128(3): 397-410, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24899140

RESUMEN

Mutations in the gene coding for Sequestosome 1 (SQSTM1) have been genetically associated with amyotrophic lateral sclerosis (ALS) and Paget disease of bone. In the present study, we analyzed the SQSTM1 coding sequence for mutations in an extended cohort of 1,808 patients with frontotemporal lobar degeneration (FTLD), ascertained within the European Early-Onset Dementia consortium. As control dataset, we sequenced 1,625 European control individuals and analyzed whole-exome sequence data of 2,274 German individuals (total n = 3,899). Association of rare SQSTM1 mutations was calculated in a meta-analysis of 4,332 FTLD and 10,240 control alleles. We identified 25 coding variants in FTLD patients of which 10 have not been described. Fifteen mutations were absent in the control individuals (carrier frequency <0.00026) whilst the others were rare in both patients and control individuals. When pooling all variants with a minor allele frequency <0.01, an overall frequency of 3.2 % was calculated in patients. Rare variant association analysis between patients and controls showed no difference over the whole protein, but suggested that rare mutations clustering in the UBA domain of SQSTM1 may influence disease susceptibility by doubling the risk for FTLD (RR = 2.18 [95 % CI 1.24-3.85]; corrected p value = 0.042). Detailed histopathology demonstrated that mutations in SQSTM1 associate with widespread neuronal and glial phospho-TDP-43 pathology. With this study, we provide further evidence for a putative role of rare mutations in SQSTM1 in the genetic etiology of FTLD and showed that, comparable to other FTLD/ALS genes, SQSTM1 mutations are associated with TDP-43 pathology.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Degeneración Lobar Frontotemporal/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Anciano , Anciano de 80 o más Años , Esclerosis Amiotrófica Lateral , Animales , Estudios de Cohortes , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Europa (Continente) , Femenino , Degeneración Lobar Frontotemporal/patología , Humanos , Cooperación Internacional , Masculino , Metaanálisis como Asunto , Persona de Mediana Edad , Proteína Sequestosoma-1
5.
Cell Rep ; 43(4): 114062, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38588339

RESUMEN

The role of T cell receptor (TCR) diversity in infectious disease susceptibility is not well understood. We use a systems immunology approach on three cohorts of herpes zoster (HZ) patients and controls to investigate whether TCR diversity against varicella-zoster virus (VZV) influences the risk of HZ. We show that CD4+ T cell TCR diversity against VZV glycoprotein E (gE) and immediate early 63 protein (IE63) after 1-week culture is more restricted in HZ patients. Single-cell RNA and TCR sequencing of VZV-specific T cells shows that T cell activation pathways are significantly decreased after stimulation with VZV peptides in convalescent HZ patients. TCR clustering indicates that TCRs from HZ patients co-cluster more often together than TCRs from controls. Collectively, our results suggest that not only lower VZV-specific TCR diversity but also reduced functional TCR affinity for VZV-specific proteins in HZ patients leads to lower T cell activation and consequently affects the susceptibility for viral reactivation.


Asunto(s)
Herpes Zóster , Herpesvirus Humano 3 , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T , Humanos , Herpes Zóster/inmunología , Herpes Zóster/virología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Activación de Linfocitos/inmunología , Herpesvirus Humano 3/inmunología , Femenino , Persona de Mediana Edad , Masculino , Linfocitos T CD4-Positivos/inmunología , Anciano , Adulto , Epítopos de Linfocito T/inmunología
6.
Mov Disord ; 27(11): 1451-6, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22976901

RESUMEN

BACKGROUND: Autosomal dominant dopa-responsive dystonia (AD-DRD) is caused by a biochemical defect primarily resulting from guanosine triphosphate cyclohydrolase 1 gene (GCH1) mutations. Few families have been reported without mutations in GCH1. METHODS: Genome-wide linkage analysis and positional cloning to identify the genetic defect in a Belgian AD-DRD family was carried out. RESULTS AND CONCLUSION: In this study, we report on the identification and characterization of a novel 24-kb deletion spanning exon 1 and the 5' regulatory region of GCH1 causing a wide spectrum of motor and nonmotor symptoms in a large Belgian AD-DRD family. This large-scale deletion of regulatory sequences leads to decreased GCH1 activity in all carriers, most probably resulting from allelic loss of transcription. We mapped the breakpoints of this deletion to the nucleotide level, allowing the development of a straightforward polymerase chain reaction assay for fast, efficient detection of this large deletion, which will prove valuable for preimplantation genetic diagnosis.


Asunto(s)
Distonía/genética , GTP Ciclohidrolasa/genética , Regiones Promotoras Genéticas/genética , Eliminación de Secuencia/genética , Adulto , Bélgica , Mapeo Cromosómico , Dopaminérgicos/uso terapéutico , Distonía/tratamiento farmacológico , Distonía/etiología , Salud de la Familia , Femenino , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Humanos , Levodopa/uso terapéutico , Masculino , Persona de Mediana Edad , Adulto Joven
7.
Brain ; 134(Pt 3): 808-15, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21354975

RESUMEN

In a genome-wide association study of frontotemporal lobar degeneration with pathological inclusions of TAR DNA-binding protein, significant association was obtained with three single nucleotide polymorphisms at 7p21.3, in a region encompassing the gene TMEM106B. This study also suggested a potential modifying effect of TMEM106B on disease since the association was strongest in progranulin mutation carriers. Further, the risk effect seemed to correlate with increased TMEM106B expression in patients. In the present study, we sought to replicate these three findings using an independent Flanders-Belgian cohort of primarily clinically diagnosed patients with frontotemporal lobar degeneration (n = 288). We were able to confirm the association with TMEM106B with a P-value of 0.008 for rs1990622, the top marker from the genome-wide association study [odds ratio 0.75 (95% confidence interval 0.61-0.93)]. Further, high-density single nucleotide polymorphism mapping suggested that the association was solely driven by the gene TMEM106B. Homozygous carriers of the TMEM106B protective alleles had a 50% reduced risk of developing frontotemporal lobar degeneration. However, we were unable to detect a modifying effect of the TMEM106B single nucleotide polymorphisms on onset age in progranulin mutation carriers belonging to an extended, clinical and pathological well-documented founder family segregating a progranulin null mutation. Also, we could not observe significant differences in messenger RNA expression between patients and control individuals in lymphoblast cell lines and in brain frontal cortex. In conclusion, we replicated the genetic TMEM106B association in a primarily clinically diagnosed cohort of patients with frontotemporal lobar degeneration from Flanders-Belgium. Additional studies are needed to unravel the molecular role of TMEM106B in disease onset and pathogenesis.


Asunto(s)
Degeneración Lobar Frontotemporal/diagnóstico , Degeneración Lobar Frontotemporal/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple/genética , Edad de Inicio , Anciano , Estudios de Cohortes , Femenino , Lóbulo Frontal/patología , Degeneración Lobar Frontotemporal/patología , Regulación de la Expresión Génica/fisiología , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Desequilibrio de Ligamiento , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Persona de Mediana Edad
8.
Nature ; 442(7105): 920-4, 2006 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-16862115

RESUMEN

Frontotemporal dementia (FTD) with ubiquitin-immunoreactive neuronal inclusions (both cytoplasmic and nuclear) of unknown nature has been linked to a chromosome 17q21 region (FTDU-17) containing MAPT (microtubule-associated protein tau). FTDU-17 patients have consistently been shown to lack a tau-immunoreactive pathology, a feature characteristic of FTD with parkinsonism linked to mutations in MAPT (FTDP-17). Furthermore, in FTDU-17 patients, mutations in MAPT and genomic rearrangements in the MAPT region have been excluded by both genomic sequencing and fluorescence in situ hybridization on mechanically stretched chromosomes. Here we demonstrate that FTDU-17 is caused by mutations in the gene coding for progranulin (PGRN), a growth factor involved in multiple physiological and pathological processes including tumorigenesis. Besides the production of truncated PGRN proteins due to premature stop codons, we identified a mutation within the splice donor site of intron 0 (IVS0 + 5G > C), indicating loss of the mutant transcript by nuclear degradation. The finding was made within an extensively documented Belgian FTDU-17 founder family. Transcript and protein analyses confirmed the absence of the mutant allele and a reduction in the expression of PGRN. We also identified a mutation (c.3G > A) in the Met1 translation initiation codon, indicating loss of PGRN due to lack of translation of the mutant allele. Our data provide evidence that PGRN haploinsufficiency leads to neurodegeneration because of reduced PGRN-mediated neuronal survival. Furthermore, in a Belgian series of familial FTD patients, PGRN mutations were 3.5 times more frequent than mutations in MAPT, underscoring a principal involvement of PGRN in FTD pathogenesis.


Asunto(s)
Cromosomas Humanos Par 17/genética , Demencia/genética , Lóbulo Frontal/fisiopatología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Mutación/genética , Lóbulo Temporal/fisiopatología , Ubiquitina/metabolismo , Bélgica , Análisis Mutacional de ADN , Demencia/fisiopatología , Lóbulo Frontal/metabolismo , Ligamiento Genético/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Mapeo Físico de Cromosoma , Progranulinas , Sitios de Empalme de ARN/genética , Lóbulo Temporal/metabolismo
9.
Hum Mol Genet ; 17(2): 313-22, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17956895

RESUMEN

The charged multivesicular body protein 2B gene (CHMP2B) was recently associated with frontotemporal lobar degeneration (FTLD) linked to chromosome 3 in a Danish FTLD family (FTD-3). In this family, a mutation in the acceptor splice site of exon 6 produced two aberrant transcripts predicting two C-truncated CHMP2B proteins due to a read through of intron 5 (p.Met178ValfsX2) and a cryptic splicing event within exon 6 (p.Met178LeufsX30). Extensive mutation analysis of CHMP2B in Belgian patients (N = 146) identified one nonsense mutation in exon 5 (c.493C>T) in a familial FTLD patient, predicting a C-truncated protein p.Gln165X analogous to the Danish mutant proteins. Overexpression of Belgian p.Gln165X in human neuroblastoma SK-N-SH cells showed the formation of large, aberrant endosomal structures that were highly similar to those observed for Danish p.Met178ValfsX2. Together, these data suggest that C-truncating mutations in CHMP2B might underlie the pathogenic mechanism in FTLD by disturbing endosome function. We also describe a missense mutation in exon 5 of CHMP2B (p.Asn143Ser) in a familial patient with cortical basal degeneration. However, the pathogenic character of this mutation remains elusive.


Asunto(s)
Demencia/genética , Proteínas del Tejido Nervioso/genética , Línea Celular Tumoral , Análisis Mutacional de ADN , Complejos de Clasificación Endosomal Requeridos para el Transporte , Endosomas/metabolismo , Femenino , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Mutación , Mutación Missense , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Linaje , Transfección
10.
Ann Neurol ; 65(5): 603-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19288468

RESUMEN

OBJECTIVE: Mutations that lead to a loss of progranulin (PGRN) explain a considerable portion of the occurrence of frontotemporal lobar degeneration. We tested a biomarker allowing rapid detection of a loss of PGRN. METHODS: We used an enzyme-linked immunosorbent assay to measure in serum the PGRN protein levels of six affected and eight unaffected carriers from within an extended Belgian founder family segregating the null mutation IVS1+5G>C. Further, we measured serum PGRN levels in 2 patients with another null mutation (a Met1 and a frameshift mutation), in 4 patients carrying a predicted pathogenic missense mutation and in 5 patients carrying a benign missense polymorphism, in 9 unaffected noncarrier relatives, and in 22 community controls. RESULTS: Serum PGRN levels were reduced in both affected and unaffected null mutation carriers compared with noncarrier relatives (p(exact) < 0.0001), and allowed perfect discrimination between carriers and noncarriers (sensitivity: 1.0; 1 - specificity: 0.0). Serum PGRN levels in Cys139Arg and Arg564Cys mutation carriers were significantly lower than in controls, but greater than in null mutation carriers, fitting the hypothesis of partial loss of function caused by these missense mutations. As expected, levels for carriers of benign missense polymorphisms were not significantly different from controls. INTERPRETATION: Our results indicate that the serum PGRN level is a reliable biomarker for diagnosing and early detection of frontotemporal lobar degeneration caused by PGRN null mutations, and provided the first in vivo evidence that at least some missense mutations in PGRN may lead to a (partial) loss of PGRN.


Asunto(s)
Biomarcadores/sangre , Demencia/sangre , Péptidos y Proteínas de Señalización Intercelular/sangre , Anciano , Anciano de 80 o más Años , Arginina/genética , Cisteína/genética , Demencia/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Persona de Mediana Edad , Mutación Missense/genética , Progranulinas
11.
Hum Mutat ; 29(6): 832-40, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18401856

RESUMEN

In one genetic study, the high temperature requirement A2 (HTRA2) mitochondrial protein has been associated with increased risk for sporadic Parkinson disease (PD). One missense mutation, p.Gly399Ser, in its C-terminal PDZ domain (from the initial letters of the postsynaptic density 95, PSD-95; discs large; and zonula occludens-1, ZO-1 proteins [Kennedy, 1995]) resulted in defective protease activation, and induced mitochondrial dysfunction when overexpressed in stably transfected cells. Here we examined the contribution of genetic variability in HTRA2 to PD risk in an extended series of 266 Belgian PD patients and 273 control individuals. Mutation analysis identified a novel p.Arg404Trp mutation within the PDZ domain predicted to freeze HTRA2 in an inactive form. Moreover, we identified six patient-specific variants in 5' and 3' regulatory regions that might affect HTRA2 expression as supported by data of luciferase reporter gene analyses. Our study confirms a role of the HTRA2 mitochondrial protein in PD susceptibility through mutations in its functional PDZ domain. In addition, it extends the HTRA2 mutation spectrum to functional variants possibly affecting transcriptional activity. The latter underpins a previously unrecognized role for altered HTRA2 expression as a risk factor relevant to parkinsonian neurodegeneration.


Asunto(s)
Proteínas Mitocondriales/genética , Enfermedad de Parkinson/genética , Serina Endopeptidasas/genética , Secuencia de Aminoácidos , Animales , Bélgica , Estudios de Casos y Controles , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Masculino , Mitocondrias , Proteínas Mitocondriales/química , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Alineación de Secuencia , Serina Endopeptidasas/química
12.
Eur J Hum Genet ; 16(4): 471-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18197194

RESUMEN

We determined the prevalence of mutations in two major functional domains of the leucine-rich repeat kinase 2 gene (LRRK2) in Belgian Parkinson's disease (PD) patients (N=304) of which 18.1% were familial PD patients. Ten patients were heterozygous for five different missense mutations (3.29%) of whom six carried the same mutation p.R1441C (1.97%). All six p.R1441C carriers were familial PD patients explaining 10.7% of familial PD in the Belgian patient group. Moreover, they shared a common disease haplotype of 21 consecutive markers in a region of 438 kb, suggesting that they are distant descendants of a single common ancestor. Clinically, p.R1441C carriers had typical levodopa-responsive parkinsonism with tremor as the most common presenting feature. Their age at onset was highly variable and ranged from 39 to 73 years, suggesting the influence of modifying factors. The remaining four patients were heterozygous each for a novel missense mutation located in the Roc or kinase domain. The pathogenic nature of these mutations remains to be determined, though we have genetic evidence that at least some represent rare but benign variants rather than causal mutations. The latter observation indicates that prudence is needed in diagnostic testing of LRRK2 in PD patients. Functional data should underlie a conclusion on the pathogenic nature of some mutations that have not been conclusively linked to disease.


Asunto(s)
Efecto Fundador , Mutación/genética , Enfermedad de Parkinson/genética , Proteínas Serina-Treonina Quinasas/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Secuencia de Aminoácidos , Bélgica , Femenino , Haplotipos , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Proteínas Serina-Treonina Quinasas/química , Alineación de Secuencia
13.
Brain ; 130(Pt 9): 2277-91, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17681982

RESUMEN

Dementia with Lewy bodies (DLB) represents the second most frequent type of neurodegenerative dementia in the elderly. Although most patients have sporadic DLB, a limited number of DLB families have been described, suggesting that genetic factors may contribute to DLB pathogenesis. Here, we describe a three-generation Belgian family with prominent dementia and parkinsonism, consistent with a diagnosis of DLB, that was autopsy confirmed for the index patient. In a genome-wide scan and subsequent finemapping of candidate loci we obtained significant linkage to 2q35-q36 (Z = 3.01 at D2S1242). Segregation analysis defined a candidate region of 9.2 Mb between D2S433 and chr2q36.3-8, adjacent to the previously reported PARK11 locus. In addition, haplotype sharing studies in another DLB family of close geographical origin with similar clinical and neuropathological features highlighted the specificity of a 2q35-q36 haplotype harbouring a pathogenic mutation that causes DLB in the Belgian family. So far, extensive sequence analysis of five candidate genes within the 2q35-q36 region has not revealed a disease-causing mutation. Together, our data re-emphasize the genetic heterogeneity of DLB, and strongly support the existence of a gene for familial DLB on 2q35-q36. Once identified this will be the first novel causal gene for DLB and can be expected to open new avenues for biological studies of the disease process.


Asunto(s)
Heterogeneidad Genética , Enfermedad por Cuerpos de Lewy/genética , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Cromosomas Humanos Par 2/genética , Análisis Mutacional de ADN/métodos , Femenino , Ligamiento Genético , Genotipo , Humanos , Enfermedad por Cuerpos de Lewy/patología , Escala de Lod , Masculino , Persona de Mediana Edad , Mutación , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Linaje
14.
Neurobiol Aging ; 67: 84-94, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29653316

RESUMEN

We previously reported a granulin (GRN) null mutation, originating from a common founder, in multiple Belgian families with frontotemporal dementia. Here, we used data of a 10-year follow-up study to describe in detail the clinical heterogeneity observed in this extended founder pedigree. We identified 85 patients and 40 unaffected mutation carriers, belonging to 29 branches of the founder pedigree. Most patients (74.4%) were diagnosed with frontotemporal dementia, while others had a clinical diagnosis of unspecified dementia, Alzheimer's dementia or Parkinson's disease. The observed clinical heterogeneity can guide clinical diagnosis, genetic testing, and counseling of mutation carriers. Onset of initial symptomatology is highly variable, ranging from age 45 to 80 years. Analysis of known modifiers, suggested effects of GRN rs5848, microtubule-associated protein tau H1/H2, and chromosome 9 open reading frame 72 G4C2 repeat length on onset age but explained only a minor fraction of the variability. Contrary, the extended GRN founder family is a valuable source for identifying other onset age modifiers based on exome or genome sequences. These modifiers might be interesting targets for developing disease-modifying therapies.


Asunto(s)
Demencia Frontotemporal/genética , Estudios de Asociación Genética , Péptidos y Proteínas de Señalización Intercelular/genética , Mutación con Pérdida de Función , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Bélgica , Dimetilhidrazinas , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Linaje , Progranulinas , Propionatos
15.
Neurobiol Aging ; 66: 181.e3-181.e10, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29555433

RESUMEN

We investigated the genetic role of sortilin (SORT1) in frontotemporal dementia (FTD). SORT1 is the neuronal receptor for granulin, encoded by the progranulin gene (GRN), a major causal gene for inherited FTD. In Belgian cohorts of 636 FTD patients and 1066 unaffected control individuals, we identified 5 patient-only nonsynonymous rare variants in SORT1. Rare variant burden analysis showed a significant increase in rare coding variants in patients compared to control individuals (p = 0.04), particularly in the ß-propeller domain (p = 0.04), with 2 rare variants located in the predicted binding site for GRN (p = 0.001). We extended these observations by analyzing 3 independent patient/control cohorts sampled in Spain, Italy, and Portugal by partners of the European Early-Onset Dementia Consortium, together with 1155 FTD patients and 1161 control persons. An additional 7 patient-only nonsynonymous variants were observed in SORT1 in European patients. Meta-analysis of the rare nonsynonymous variants in the Belgian and European patient/control cohorts revealed a significant enrichment in FTD patients (p = 0.006), establishing SORT1 as a genetic risk factor for FTD.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Estudios de Asociación Genética , Variación Genética/genética , Proteínas Adaptadoras del Transporte Vesicular/química , Anciano , Bélgica , Sitios de Unión , Estudios de Cohortes , Europa (Continente) , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Persona de Mediana Edad , Progranulinas , Unión Proteica , Dominios Proteicos , Riesgo
16.
Hum Mutat ; 28(4): 416, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17345602

RESUMEN

Null mutations in the progranulin gene (GRN, PGRN) were recently identified as the causal mechanism underlying frontotemporal dementia (FTD) with ubiquitin-positive brain pathology linked to chromosome 17 (FTDU-17). In a Belgian and French FTD series comprising 332 patients, we reported 13 PGRN null mutations which were mainly nonsense and frameshift mutations resulting in premature stop codons. Here we report in the same patient series three missense mutations of which two (c.743C>T, p.Pro248Leu and c.1294C>T, p.Arg432Cys) were predicted in silico to severely affect protein folding and/or processing leading to PGRN protein haploinsufficiency. In addition, we observed three sequence variations in the 5' regulatory region that might potentially affect PGRN transcription activity. Our findings extend the mutation spectrum in PGRN leading to loss of functional PGRN as the basis for FTD.


Asunto(s)
Demencia/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Mutación Missense , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Estudios de Casos y Controles , Secuencia Conservada , Análisis Mutacional de ADN , Demencia/metabolismo , Demencia/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Datos de Secuencia Molecular , Progranulinas , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína
17.
Arch Neurol ; 64(10): 1436-46, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17923627

RESUMEN

BACKGROUND: Progranulin gene (PGRN) haploinsufficiency was recently associated with ubiquitin-positive frontotemporal lobar degeneration linked to chromosome 17q21 (FTLDU-17). OBJECTIVE: To assess whether PGRN genetic variability contributed to other common neurodegenerative brain diseases, such as Alzheimer disease (AD) or Parkinson disease (PD). DESIGN: Mutation analysis of PGRN. SETTING: Memory Clinic of the Middelheim General Hospital. Patients We analyzed 666 Belgian patients with AD and 255 with PD. MAIN OUTCOME MEASURES: Results of PGRN sequencing, PGRN transcript analysis, short tandem repeat genotyping, and neuropathologic analysis. RESULTS: We identified 2 patients with AD and 1 patient with PD who carried the null mutation IVS0 + 5G>C, which we reported earlier in an extensively characterized Belgian founder family, DR8, segregating FTLDU. Postmortem pathologic diagnosis of the patient with PD revealed both FTLDU and Lewy body pathologic features. In addition, we identified in PGRN only 1 other null mutation, the nonsense mutation p.Arg535X, in 1 patient with probable AD. However, in vitro analysis predicted a PGRN C-truncated protein, although it remains to be elucidated if this shortened transcript leads to haploinsufficiency. CONCLUSIONS: Our mutation data indicated that null mutations are rare in patients with AD (3/666 = 0.45%) and PD (1/255 = 0.39%). Also, AD and PD clinical diagnoses in patients who carry PGRN null mutations likely result from etiologic heterogeneity rather than PGRN haploinsufficiency.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/genética , Factores de Edad , Anciano , Enfermedad de Alzheimer/epidemiología , Autopsia , Bélgica/epidemiología , Encéfalo/patología , Cromosomas Humanos Par 17/genética , Codón sin Sentido/genética , Análisis Mutacional de ADN , Femenino , Efecto Fundador , Variación Genética , Genotipo , Heterocigoto , Humanos , Inmunohistoquímica , Enfermedad por Cuerpos de Lewy/patología , Masculino , Persona de Mediana Edad , Mutación , Enfermedad de Parkinson/epidemiología , Linaje , Progranulinas , Secuencias Repetidas en Tándem/genética , Ubiquitina/metabolismo
18.
Brain ; 129(Pt 4): 841-52, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16495329

RESUMEN

Among patients with frontotemporal lobar degeneration (FTLD), the respective frequencies of dominant 17q21-linked tau-negative FTLD (with unidentified molecular defect) and 17q21-linked tau-positive FTLD (due to MAPT mutations) remain unknown. Here, in a series of 98 genealogically unrelated Belgian FTLD patients, we identified an ancestral 8 cM MAPT containing haplotype in two patients belonging to multiplex families DR2 and DR8, without demonstrable MAPT mutations, in which FTLD was conclusively linked to 17q21 [maximum summed log of the odds (LOD) score of 5.28 at D17S931]. Interestingly, the same DR2-DR8 ancestral haplotype was observed in five additional familial FTLD patients, indicative of a founder effect. In the FTLD series, the DR2-DR8 ancestral haplotype explained 7% (7 out of 98) of FTLD and 17% (7 out of 42) of familial FTLD and was seven times more frequent than MAPT mutations (1 out of 98 or 1%). Clinically, DR2-DR8 haplotype carriers presented with FTLD often characterized by language impairment, and in one carrier the neuropathological diagnosis was FTLD with rare tau-negative ubiquitin-positive inclusions. Together, these results strongly suggest that the DR2-DR8 founder haplotype at 17q21 harbours a tau-negative FTLD causing mutation that is a much more frequent cause of FTLD in Belgium than MAPT mutations.


Asunto(s)
Cromosomas Humanos Par 17/genética , Demencia/genética , Mutación , Proteínas tau/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bélgica , Mapeo Cromosómico , Análisis Mutacional de ADN/métodos , Demencia/metabolismo , Demencia/patología , Femenino , Efecto Fundador , Predisposición Genética a la Enfermedad , Antígenos HLA-DR/genética , Subtipos Serológicos HLA-DR , Antígeno HLA-DR2/genética , Haplotipos , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Linaje , Estudios Prospectivos , Ubiquitina/análisis , Proteínas tau/análisis
19.
JAMA Neurol ; 74(4): 445-452, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28192553

RESUMEN

Importance: Patients carrying a C9orf72 repeat expansion leading to frontotemporal dementia and/or amyotrophic lateral sclerosis have highly variable ages at onset of disease, suggesting the presence of modifying factors. Objective: To provide clinical-based evidence for disease anticipation in families carrying a C9orf72 repeat expansion by analyzing age at onset, disease duration, and age at death in successive generations. Design, Setting, and Participants: This cohort study was performed from June 16, 2000, to June 1, 2016, in 36 extended Belgian families in which a C9orf72 repeat expansion was segregating. The generational effect on age at onset, disease duration, and age at death was estimated using a mixed effects Cox proportional hazards regression model, including random-effects terms for within-family correlation and kinship. Time until disease onset or last examination, time from disease onset until death or last examination, or age at death was collected for for 244 individuals (132 proven or obligate C9orf72 carriers), of whom 147 were clinically affected (89 proven or obligate C9orf72 carriers). Main Outcomes and Measures: Generational effect on age at onset, disease duration, and age at death. Results: Among the 111 individuals with age at onset available (66 men and 45 women; mean [SD] age, 57.2 [9.1] years), the mean (SD) age at onset per generation (from earliest-born to latest-born generation) was 62.5 (8.3), 57.1 (8.2), 54.6 (10.2), and 49.3 (7.5) years. Censored regression analysis on all affected and unaffected at-risk relatives confirmed a decrease in age at onset in successive generations (P < .001). No generational effect was observed for disease duration or age at death. Conclusions and Relevance: The clinical data provide supportive evidence for the occurrence of disease anticipation in families carrying a C9orf72 repeat expansion by means of a decrease in age at onset across successive generations. This finding may help clinicians decide from which age onward it may be relevant to clinically follow presymptomatic individuals who carry a C9orf72 repeat expansion.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Expansión de las Repeticiones de ADN/genética , Demencia Frontotemporal/genética , Linaje , Proteínas/genética , Edad de Inicio , Anciano , Proteína C9orf72 , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales
20.
Neurosci Lett ; 629: 160-164, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27397011

RESUMEN

OBJECTIVE: To investigate the frequency of glucocerebrosidase (GBA) mutations in a Flanders-Belgian Parkinson's disease (PD) patient cohort and to assess genotype-phenotype correlations. METHODS: We performed an in-depth sequencing of all coding exons of GBA in 266 clinically well-characterized PD patients and 536 healthy control individuals. RESULTS: We identified rare, heterozygous GBA mutations in 12 PD patients (4.5%) and in 2 healthy control individuals (0.37%), confirming the genetic association of GBA mutations with PD in the Flanders-Belgian population (p<0.001). The patient carriers had a more severe Unified Parkinson's Disease Rating Scale (UPDRS) motor score than non-carriers. Also, GBA mutation status was a significant, independent predictor for the presence of dementia (OR=12.43, 95% CI: 2.27-68.14. p=0.004). Genetic association of PD with the common p.E326K and p.T369M variants in GBA was absent. CONCLUSION: In our Flanders-Belgian cohort, carrier status of a heterozygous GBA mutation was a strong genetic risk factor for PD. The GBA mutation frequency of 4.5% is comparable to previously reported data in other European PD patient cohorts. Furthermore, our clinical data suggest a more severe motor phenotype and a strong predisposition to dementia in GBA mutation carriers.


Asunto(s)
Demencia/genética , Predisposición Genética a la Enfermedad , Glucosilceramidasa/genética , Enfermedad de Parkinson/genética , Anciano , Bélgica , Estudios de Cohortes , Demencia/complicaciones , Exones , Femenino , Estudios de Asociación Genética , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación , Enfermedad de Parkinson/complicaciones , Fenotipo , Factores de Riesgo , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA