Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Bioinformatics ; 34(15): 2666-2669, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29547981

ABSTRACT

Summary: Here we describe NanoPack, a set of tools developed for visualization and processing of long-read sequencing data from Oxford Nanopore Technologies and Pacific Biosciences. Availability and implementation: The NanoPack tools are written in Python3 and released under the GNU GPL3.0 License. The source code can be found at https://github.com/wdecoster/nanopack, together with links to separate scripts and their documentation. The scripts are compatible with Linux, Mac OS and the MS Windows 10 subsystem for Linux and are available as a graphical user interface, a web service at http://nanoplot.bioinf.be and command line tools. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Software , Escherichia coli/genetics
2.
Annu Rev Neurosci ; 33: 71-88, 2010.
Article in English | MEDLINE | ID: mdl-20415586

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a neurodegenerative condition that predominantly affects behavior, social awareness, and language. It is characterized by extensive heterogeneity at the clinical, pathological, and genetic levels. Recognition of these levels of heterogeneity is important for proper disease management. The identification of progranulin and TDP-43 as key proteins in a significant proportion of FTLD patients has provided the impetus for a wealth of studies probing their role in neurodegeneration. This review highlights the most recent developments and future directions in this field and puts them in perspective of the novel insights into the neurodegenerative process, which have been gained from related disorders, e.g., the role of FUS in amyotrophic lateral sclerosis.


Subject(s)
Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Signal Transduction/genetics , Frontotemporal Lobar Degeneration/etiology , Genetic Predisposition to Disease/genetics , Humans , Nerve Degeneration/etiology , Phosphorylation/genetics , tau Proteins/genetics , tau Proteins/metabolism
3.
Hum Mutat ; 38(3): 297-309, 2017 03.
Article in English | MEDLINE | ID: mdl-28008748

ABSTRACT

We investigated the mutation spectrum of the TANK-Binding Kinase 1 (TBK1) gene and its associated phenotypic spectrum by exonic resequencing of TBK1 in a cohort of 2,538 patients with frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), or FTD plus ALS, ascertained within the European Early-Onset Dementia Consortium. We assessed pathogenicity of predicted protein-truncating mutations by measuring loss of RNA expression. Functional effect of in-frame amino acid deletions and missense mutations was further explored in vivo on protein level and in vitro by an NFκB-induced luciferase reporter assay and measuring phosphorylated TBK1. The protein-truncating mutations led to the loss of transcript through nonsense-mediated mRNA decay. For the in-frame amino acid deletions, we demonstrated loss of TBK1 or phosphorylated TBK1 protein. An important fraction of the missense mutations compromised NFκB activation indicating that at least some functions of TBK1 are lost. Although missense mutations were also present in controls, over three times more mutations affecting TBK1 functioning were found in the mutation fraction observed in patients only, suggesting high-risk alleles (P = 0.03). Total mutation frequency for confirmed TBK1 LoF mutations in the European cohort was 0.7%, with frequencies in the clinical subgroups of 0.4% in FTD, 1.3% in ALS, and 3.6% in FTD-ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Protein Serine-Threonine Kinases/genetics , White People/genetics , Aged , Alleles , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/epidemiology , Case-Control Studies , Cohort Studies , Enzyme Activation , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/epidemiology , Genetic Association Studies , Heterozygote , Humans , Male , Middle Aged , Mutation , NF-kappa B/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Sequence Deletion
4.
Brain ; 139(Pt 2): 452-67, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26674655

ABSTRACT

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.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Heterozygote , Intercellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Adult , Aged , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/epidemiology , Belgium/epidemiology , C9orf72 Protein , Cohort Studies , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/epidemiology , Humans , Male , Middle Aged , Mutation/genetics , Pedigree , Progranulins
5.
Proc Natl Acad Sci U S A ; 110(13): 4986-91, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23457265

ABSTRACT

Mutations in the Tar DNA binding protein of 43 kDa (TDP-43; TARDBP) are associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43(+) inclusions (FTLD-TDP). To determine the physiological function of TDP-43, we knocked out zebrafish Tardbp and its paralogue Tardbp (TAR DNA binding protein-like), which lacks the glycine-rich domain where ALS- and FTLD-TDP-associated mutations cluster. tardbp mutants show no phenotype, a result of compensation by a unique splice variant of tardbpl that additionally contains a C-terminal elongation highly homologous to the glycine-rich domain of tardbp. Double-homozygous mutants of tardbp and tardbpl show muscle degeneration, strongly reduced blood circulation, mispatterning of vessels, impaired spinal motor neuron axon outgrowth, and early death. In double mutants the muscle-specific actin binding protein Filamin Ca is up-regulated. Strikingly, Filamin C is similarly increased in the frontal cortex of FTLD-TDP patients, suggesting aberrant expression in smooth muscle cells and TDP-43 loss-of-function as one underlying disease mechanism.


Subject(s)
Axons/metabolism , DNA-Binding Proteins , Motor Neurons/metabolism , Muscular Atrophy/metabolism , Mutation , Vascular Diseases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Contractile Proteins/genetics , Contractile Proteins/metabolism , Filamins , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Motor Neurons/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Protein Structure, Tertiary , Vascular Diseases/genetics , Vascular Diseases/pathology , Zebrafish Proteins/genetics
6.
Acta Neuropathol ; 127(3): 407-18, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24442578

ABSTRACT

Hexanucleotide repeat expansions in chromosome 9 open reading frame 72 (C9orf72) have recently been linked to frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis, and may be the most common genetic cause of both neurodegenerative diseases. Genetic variants at TMEM106B influence risk for the most common neuropathological subtype of FTLD, characterized by inclusions of TAR DNA-binding protein of 43 kDa (FTLD-TDP). Previous reports have shown that TMEM106B is a genetic modifier of FTLD-TDP caused by progranulin (GRN) mutations, with the major (risk) allele of rs1990622 associating with earlier age at onset of disease. Here, we report that rs1990622 genotype affects age at death in a single-site discovery cohort of FTLD patients with C9orf72 expansions (n = 14), with the major allele correlated with later age at death (p = 0.024). We replicate this modifier effect in a 30-site international neuropathological cohort of FTLD-TDP patients with C9orf72 expansions (n = 75), again finding that the major allele associates with later age at death (p = 0.016), as well as later age at onset (p = 0.019). In contrast, TMEM106B genotype does not affect age at onset or death in 241 FTLD-TDP cases negative for GRN mutations or C9orf72 expansions. Thus, TMEM106B is a genetic modifier of FTLD with C9orf72 expansions. Intriguingly, the genotype that confers increased risk for developing FTLD-TDP (major, or T, allele of rs1990622) is associated with later age at onset and death in C9orf72 expansion carriers, providing an example of sign epistasis in human neurodegenerative disease.


Subject(s)
Frontotemporal Lobar Degeneration/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Proteins/genetics , Adult , Age Factors , Age of Onset , Aged , Aged, 80 and over , Alleles , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , C9orf72 Protein , Cohort Studies , DNA Repeat Expansion , Female , Frontotemporal Lobar Degeneration/blood , Frontotemporal Lobar Degeneration/mortality , Genetic Predisposition to Disease , Genotype , Heterozygote , Humans , Intercellular Signaling Peptides and Proteins/blood , Male , Middle Aged , Polymorphism, Single Nucleotide , Progranulins
7.
Trends Genet ; 26(2): 84-93, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20080314

ABSTRACT

The recent discoveries in genome-wide association studies (GWAS) of novel susceptibility loci (CLU, CR1 and PICALM) for Alzheimer's disease (AD) have elicited considerable interest in the AD community. But what are the implications of these purely epidemiological findings for our understanding of disease etiology and patient care? In this review, we attempt to place these findings in the context of current and future AD genetics research. CLU, CR1 and PICALM support existing hypotheses about the amyloid, lipid, chaperone and chronic inflammatory pathways in AD pathogenesis. We discuss how these and future findings can be translated into efforts to ameliorate patient care by genetic profiling for risk prediction and pharmacogenetics and by guiding drug development.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Genetic Predisposition to Disease , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Progression , Drug Design , Gene Expression Profiling , Humans , Risk Factors
8.
Acta Neuropathol ; 125(3): 413-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23381195

ABSTRACT

Genetic analysis revealed the hexanucleotide repeat expansion GGGGCC within the regulatory region of the gene C9orf72 as the most common cause of familial amyotrophic lateral sclerosis and the second most common cause of frontotemporal lobar degeneration. Since repeat expansions might cause RNA toxicity via sequestration of RNA-binding proteins, we searched for proteins capable of binding to GGGGCC repeats. In vitro-transcribed biotinylated RNA containing hexanucleotide GGGGCC or, as control, AAAACC repeats were incubated with nuclear protein extracts. Using stringent filtering protocols 20 RNA-binding proteins with a variety of different functions in RNA metabolism, translation and transport were identified. A subset of these proteins was further investigated by immunohistochemistry in human autopsy brains. This revealed that hnRNP A3 formed neuronal cytoplasmic and intranuclear inclusions in the hippocampus of patients with C9orf72 repeat extensions. Confocal microcopy showed that these inclusions belong to the group of the so far enigmatic p62-positive/TDP-43 negative inclusions characteristically seen in autopsy cases of diseased C9orf72 repeat expansion carriers. Thus, we have identified one protein component of these pathognomonic inclusions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Hippocampus/pathology , Inclusion Bodies/metabolism , Mutation/genetics , Proteins/genetics , Repetitive Sequences, Nucleic Acid/physiology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein , Chromatography, High Pressure Liquid , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Inclusion Bodies/pathology , Mass Spectrometry , RNA, Small Interfering/metabolism , Sequestosome-1 Protein , Transfection
9.
Acta Neuropathol ; 126(6): 881-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24132570

ABSTRACT

Massive GGGGCC repeat expansion in the first intron of the gene C9orf72 is the most common known cause of familial frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Despite its intronic localization and lack of an ATG start codon, the repeat region is translated in all three reading frames into aggregating dipeptide-repeat (DPR) proteins, poly-(Gly-Ala), poly-(Gly-Pro) and poly-(Gly-Arg). We took an antibody-based approach to further validate the translation of DPR proteins. To test whether the antisense repeat RNA transcript is also translated, we raised antibodies against the predicted products, poly-(Ala-Pro) and poly-(Pro-Arg). Both antibodies stained p62-positive neuronal cytoplasmic inclusions throughout the cerebellum and hippocampus indicating that not only sense but also antisense strand repeats are translated into DPR proteins in the absence of ATG start codons. Protein products of both strands co-aggregate suggesting concurrent translation of both strands. Moreover, an antibody targeting the putative carboxyl terminus of DPR proteins can detect inclusion pathology in C9orf72 repeat expansion carriers suggesting that the non-ATG translation continues through the entire repeat and beyond. A highly sensitive monoclonal antibody against poly-(Gly-Arg), visualized abundant inclusion pathology in all cortical regions and some inclusions also in motoneurons. Together, our data show that the GGGGCC repeat is bidirectionally translated into five distinct DPR proteins that co-aggregate in the characteristic p62-positive TDP-43 negative inclusions found in FTLD/ALS cases with C9orf72 repeat expansion. Novel monoclonal antibodies against poly-(Gly-Arg) will facilitate pathological diagnosis of C9orf72 FTLD/ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Brain/metabolism , Frontotemporal Lobar Degeneration/diagnosis , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein , DNA Repeat Expansion , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Humans , Protein Biosynthesis , Proteins/metabolism
10.
J Neurosci ; 31(5): 1885-94, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21289198

ABSTRACT

Numerous loss-of-function mutations in the progranulin (GRN) gene cause frontotemporal lobar degeneration with ubiquitin and TAR-DNA binding protein 43-positive inclusions by reduced production and secretion of GRN. Consistent with the observation that GRN has neurotrophic properties, pharmacological stimulation of GRN production is a promising approach to rescue GRN haploinsufficiency and prevent disease progression. We therefore searched for compounds capable of selectively increasing GRN levels. Here, we demonstrate that four independent and highly selective inhibitors of vacuolar ATPase (bafilomycin A1, concanamycin A, archazolid B, and apicularen A) significantly elevate intracellular and secreted GRN. Furthermore, clinically used alkalizing drugs, including chloroquine, bepridil, and amiodarone, similarly stimulate GRN production. Elevation of GRN levels occurs via a translational mechanism independent of lysosomal degradation, autophagy, or endocytosis. Importantly, alkalizing reagents rescue GRN deficiency in organotypic cortical slice cultures from a mouse model for GRN deficiency and in primary cells derived from human patients with GRN loss-of-function mutations. Thus, alkalizing reagents, specifically those already used in humans for other applications, and vacuolar ATPase inhibitors may be therapeutically used to prevent GRN-dependent neurodegeneration.


Subject(s)
Alkalies/pharmacology , Cerebral Cortex/metabolism , Fibroblasts/metabolism , Frontotemporal Lobar Degeneration/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Amiodarone/pharmacology , Animals , Animals, Newborn , Autophagy-Related Protein 5 , Bepridil/pharmacology , Blotting, Northern , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Chloroquine/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Frontotemporal Lobar Degeneration/drug therapy , Frontotemporal Lobar Degeneration/genetics , Granulins , HEK293 Cells , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Macrolides/pharmacology , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mutation , Neurons/drug effects , Progranulins , RNA, Messenger/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology
11.
Hum Mutat ; 33(9): 1340-4, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22581678

ABSTRACT

The Alzheimer disease and frontotemporal dementia (AD&FTLD) and Parkinson disease (PD) Mutation Databases make available curated information of sequence variations in genes causing Mendelian forms of the most common neurodegenerative brain disease AD, frontotemporal lobar degeneration (FTLD), and PD. They are established resources for clinical geneticists, neurologists, and researchers in need of comprehensive, referenced genetic, epidemiologic, clinical, neuropathological, and/or cell biological information of specific gene mutations in these diseases. In addition, the aggregate analysis of all information available in the databases provides unique opportunities to extract mutation characteristics and genotype-phenotype correlations, which would be otherwise unnoticed and unexplored. Such analyses revealed that 61.4% of mutations are private to one single family, while only 5.7% of mutations occur in 10 or more families. The five mutations with most frequent independent observations occur in 21% of AD, 43% of FTLD, and 48% of PD families recorded in the Mutation Databases, respectively. Although these figures are inevitably biased by a publishing policy favoring novel mutations, they probably also reflect the occurrence of multiple rare and few relatively common mutations in the inherited forms of these diseases. Finally, with the exception of the PD genes PARK2 and PINK1, all other genes are associated with more than one clinical diagnosis or characteristics thereof.


Subject(s)
Databases, Genetic , Genetic Loci , Mutation , Neurodegenerative Diseases/genetics , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Frontotemporal Lobar Degeneration/diagnosis , Frontotemporal Lobar Degeneration/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genome, Human , Humans , Neurodegenerative Diseases/diagnosis , Parkinson Disease/diagnosis , Parkinson Disease/genetics
12.
Acta Neuropathol ; 124(3): 353-72, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22890575

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a heterogeneous group of disorders characterized by disturbances of behavior and personality and different types of language impairment with or without concomitant features of motor neuron disease or parkinsonism. FTLD is characterized by atrophy of the frontal and anterior temporal brain lobes. Detailed neuropathological studies have elicited proteinopathies defined by inclusions of hyperphosphorylated microtubule-associated protein tau, TAR DNA-binding protein TDP-43, fused-in-sarcoma or yet unidentified proteins in affected brain regions. Rather than the type of proteinopathy, the site of neurodegeneration correlates relatively well with the clinical presentation of FTLD. Molecular genetic studies identified five disease genes, of which the gene encoding the tau protein (MAPT), the growth factor precursor gene granulin (GRN), and C9orf72 with unknown function are most frequently mutated. Rare mutations were also identified in the genes encoding valosin-containing protein (VCP) and charged multivesicular body protein 2B (CHMP2B). These genes are good markers to distinguish underlying neuropathological phenotypes. Due to the complex landscape of FTLD diseases, combined characterization of clinical, imaging, biological and genetic biomarkers is essential to establish a detailed diagnosis. Although major progress has been made in FTLD research in recent years, further studies are needed to completely map out and correlate the clinical, pathological and genetic entities, and to understand the underlying disease mechanisms. In this review, we summarize the current state of the rapidly progressing field of genetic, neuropathological and clinical research of this intriguing condition.


Subject(s)
Frontal Lobe/pathology , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , tau Proteins/genetics , Atrophy , Genetic Predisposition to Disease , Humans , Mutation
14.
Brain ; 134(Pt 3): 808-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21354975

ABSTRACT

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.


Subject(s)
Frontotemporal Lobar Degeneration/diagnosis , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Age of Onset , Aged , Cohort Studies , Female , Frontal Lobe/pathology , Frontotemporal Lobar Degeneration/pathology , Gene Expression Regulation/physiology , Gene Frequency , Genome-Wide Association Study , Genotype , Humans , Linkage Disequilibrium , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged
15.
Nature ; 442(7105): 920-4, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16862115

ABSTRACT

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.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Dementia/genetics , Frontal Lobe/physiopathology , Intercellular Signaling Peptides and Proteins/deficiency , Mutation/genetics , Temporal Lobe/physiopathology , Ubiquitin/metabolism , Belgium , DNA Mutational Analysis , Dementia/physiopathology , Frontal Lobe/metabolism , Genetic Linkage/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Physical Chromosome Mapping , Progranulins , RNA Splice Sites/genetics , Temporal Lobe/metabolism
16.
Neurogenetics ; 12(3): 169-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21630033

ABSTRACT

The rate of DNA variation discovery has accelerated the need to collate, store and interpret the data in a standardised coherent way and is becoming a critical step in maximising the impact of discovery on the understanding and treatment of human disease. This particularly applies to the field of neurology as neurological function is impaired in many human disorders. Furthermore, the field of neurogenetics has been proven to show remarkably complex genotype-to-phenotype relationships. To facilitate the collection of DNA sequence variation pertaining to neurogenetic disorders, we have initiated the "Neurogenetics Consortium" under the umbrella of the Human Variome Project. The Consortium's founding group consisted of basic researchers, clinicians, informaticians and database creators. This report outlines the strategic aims established at the preliminary meetings of the Neurogenetics Consortium and calls for the involvement of the wider neurogenetic community in enabling the development of this important resource.


Subject(s)
Databases, Genetic/standards , Genetic Variation , Genetics, Medical/organization & administration , International Cooperation , Nervous System/metabolism , Algorithms , Congresses as Topic , Genetic Variation/physiology , Genetics, Medical/standards , Human Genome Project/organization & administration , Humans , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Research Report
17.
Trends Genet ; 24(4): 186-94, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18328591

ABSTRACT

Frontotemporal lobar degeneration (FTLD) represents a collection of neurodegenerative diseases of frontal and temporal brain regions. It has long been associated with mutations in microtubule-associated protein tau (MAPT), and more recently with loss-of-function mutations in progranulin (PGRN). Phenotypes of PGRN and MAPT mutation carriers overlap, although disease onset in PGRN carriers is a decade later. Mutations in PGRN might influence susceptibility to a wider range of neurodegenerative diseases including Alzheimer and Parkinson diseases. The recent demonstration that mutations in PGRN result in FTLD provided a novel entrance point to the molecular mechanisms leading to this disorder. The high variability in onset age and age-dependent penetrance suggests that the PGRN pathway is highly susceptible to modulating factors that might be exploited to delay the disease processes.


Subject(s)
Dementia/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Amino Acid Sequence , Dementia/genetics , Disease Susceptibility , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Molecular Sequence Data , Mutation, Missense/genetics , Phenotype , Progranulins
18.
Hum Mutat ; 31(7): 763-80, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20506312

ABSTRACT

To date, molecular genetic analyses have identified over 500 distinct DNA variants in five disease genes associated with familial Parkinson disease; alpha-synuclein (SNCA), parkin (PARK2), PTEN-induced putative kinase 1 (PINK1), DJ-1 (PARK7), and Leucine-rich repeat kinase 2 (LRRK2). These genetic variants include approximately 82% simple mutations and approximately 18% copy number variations. Some mutation subtypes are likely underestimated because only few studies reported extensive mutation analyses of all five genes, by both exonic sequencing and dosage analyses. Here we present an update of all mutations published to date in the literature, systematically organized in a novel mutation database (http://www.molgen.ua.ac.be/PDmutDB). In addition, we address the biological relevance of putative pathogenic mutations. This review emphasizes the need for comprehensive genetic screening of Parkinson patients followed by an insightful study of the functional relevance of observed genetic variants. Moreover, while capturing existing data from the literature it became apparent that several of the five Parkinson genes were also contributing to the genetic etiology of other Lewy Body Diseases and Parkinson-plus syndromes, indicating that mutation screening is recommendable in these patient groups.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Mutation , Oncogene Proteins/genetics , Parkinson Disease/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , alpha-Synuclein/genetics , Databases, Genetic , Genetic Predisposition to Disease , Genetic Testing , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease/diagnosis , Protein Deglycase DJ-1
19.
Ann Neurol ; 65(5): 603-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19288468

ABSTRACT

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.


Subject(s)
Biomarkers/blood , Dementia/blood , Intercellular Signaling Peptides and Proteins/blood , Aged , Aged, 80 and over , Arginine/genetics , Cysteine/genetics , Dementia/genetics , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Mutation, Missense/genetics , Progranulins
20.
Trends Genet ; 21(12): 664-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16221505

ABSTRACT

In contrast to the common and genetically complex senile form of Alzheimer's disease (AD), the molecular genetic dissection of inherited presenile dementias has given important mechanistic insights into the pathogenesis of degenerative brain disease. Here, we focus on recent genotype-phenotype correlative studies in presenile AD and the frontotemporal dementia (FTD) complex of disorders. Together, these studies suggest that AD and FTD are linked in a genetic spectrum of presenile degenerative brain disorders in which tau appears to be the central player.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/physiology , Humans , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Phenotype , Presenilin-1 , tau Proteins
SELECTION OF CITATIONS
SEARCH DETAIL