Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Neurochem Res ; 45(7): 1711-1728, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32361798

ABSTRACT

Healthy brain function is mediated by several complementary signalling pathways, many of which are driven by extracellular vesicles (EVs). EVs are heterogeneous in both size and cargo and are constitutively released from cells into the extracellular milieu. They are subsequently trafficked to recipient cells, whereupon their entry can modify the cellular phenotype. Here, in order to further analyse the mRNA and protein cargo of neuronal EVs, we isolated EVs by size exclusion chromatography from human induced pluripotent stem cell (iPSC)-derived neurons. Electron microscopy and dynamic light scattering revealed that the isolated EVs had a diameter of 30-100 nm. Transcriptomic and proteomics analyses of the EVs and neurons identified key molecules enriched in the EVs involved in cell surface interaction (integrins and collagens), internalisation pathways (clathrin- and caveolin-dependent), downstream signalling pathways (phospholipases, integrin-linked kinase and MAPKs), and long-term impacts on cellular development and maintenance. Overall, we show that key signalling networks and mechanisms are enriched in EVs isolated from human iPSC-derived neurons.


Subject(s)
Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Gene Regulatory Networks/physiology , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Transcription, Genetic/physiology , Humans
2.
Biosci Rep ; 40(4)2020 04 30.
Article in English | MEDLINE | ID: mdl-32301481

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative condition, of which one of the cardinal pathological hallmarks is the extracellular accumulation of amyloid ß (Aß) peptides. These peptides are generated via proteolysis of the amyloid precursor protein (APP), in a manner dependent on the ß-secretase, BACE1 and the multicomponent γ-secretase complex. Recent data also suggest a contributory role in AD of transactive response DNA binding protein 43 (TDP-43). There is little insight into a possible mechanism linking TDP-43 and APP processing. To this end, we used cultured human neuronal cells to investigate the ability of TDP-43 to interact with APP and modulate its proteolytic processing. Immunocytochemistry showed TDP-43 to be spatially segregated from both the extranuclear APP holoprotein and its nuclear C-terminal fragment. The latter (APP intracellular domain) was shown to predominantly localise to nucleoli, from which TDP-43 was excluded. Furthermore, neither overexpression of each of the APP isoforms nor siRNA-mediated knockdown of APP had any effect on TDP-43 expression. Doxycycline-stimulated overexpression of TDP-43 was explored in an inducible cell line. Overexpression of TDP-43 had no effect on expression of the APP holoprotein, nor any of the key proteins involved in its proteolysis. Furthermore, increased TDP-43 expression had no effect on BACE1 enzymatic activity or immunoreactivity of Aß1-40, Aß1-42 or the Aß1-40:Aß1-42 ratio. Also, siRNA-mediated knockdown of TDP-43 had no effect on BACE1 immunoreactivity. Taken together, these data indicate that TDP-43 function and/or dysfunction in AD is likely independent from dysregulation of APP expression and proteolytic processing and Aß generation.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , DNA-Binding Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Aspartic Acid Endopeptidases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Humans , Neurons/cytology , Neurons/metabolism , Proteolysis , RNA, Small Interfering/metabolism
3.
Neurobiol Aging ; 84: 235.e1-235.e8, 2019 12.
Article in English | MEDLINE | ID: mdl-31676125

ABSTRACT

A C9orf72 repeat expansion is the most common cause of both frontotemporal dementia and motor neuron disease. The expansion is translated to produce dipeptide repeat proteins (DPRs), which are toxic in vivo and in vitro. However, the mechanisms underlying DPR toxicity remain unclear. Mouse models which express DPRs at repeat lengths found in human disease are urgently required to investigate this. We aimed to generate transgenic mice expressing DPRs at repeat lengths of >1000 using alternative codon sequences, to reduce the repetitive nature of the insert. We found that although these inserts did integrate into the mouse genome, the alternative codon sequences did not protect from instability between generations. Our findings suggest that stable integration of long DPR sequences may not be possible. Administration of viral vectors after birth may be a more effective delivery method for long repeats.


Subject(s)
C9orf72 Protein , Clustered Regularly Interspaced Short Palindromic Repeats , Animals , Mice , Mice, Transgenic , Trinucleotide Repeat Expansion
4.
Hum Mol Genet ; 27(10): 1754-1762, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29528390

ABSTRACT

Large expansions of hexanucleotide GGGGCC (G4C2) repeats (hundreds to thousands) in the first intron of the chromosome 9 open reading frame 72 (C9orf72) locus are the strongest known genetic factor associated with amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Different hypotheses exist about the underlying disease mechanism including loss of function by haploinsufficiency, toxicity arising as a result of RNA or dipeptide repeats (DPRs). Five different DPRs are produced by repeat-associated non-ATG-initiated translation of the G4C2 repeats. Though earlier studies have indicated toxicity of the DPRs in worms, flies, primary cultured cells and cell lines, the effect of expressing DPRs of amyotrophic lateral sclerosis-relevant length has not been tested on motor behaviour in vertebrate models. In this study, by expressing constructs with alternate codons encoding different lengths of each DPR (40, 200 and 1000) in the vertebrate zebrafish model, the GR DPR was found to lead to the greatest developmental lethality and morphological defects, and GA, the least. However, expressing 1000 repeats of any DPR, including the 'non-toxic' GA DPR led to locomotor defects. Based on these observations, a transgenic line stably expressing 100 GR repeats was generated to allow specific regional and temporal expression of GR repeats in vivo. Expression of GR DPRs ubiquitously resulted in severe morphological defects and reduced swimming. However, when expressed specifically in motor neurons, the developmental defects were significantly reduced, but the swimming phenotype persisted, suggesting that GR DPRs have a toxic effect on motor neuron function. This was validated by the reduction in motor neuron length even in already formed motor neurons when GR was expressed in these. Hence, the expression of C9orf72-associated DPRs can cause significant motor deficits in vertebrates.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , DNA Repeat Expansion/genetics , Frontotemporal Lobar Degeneration/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Animals, Genetically Modified/genetics , Dipeptides/genetics , Disease Models, Animal , Frontotemporal Lobar Degeneration/physiopathology , Gene Expression Regulation , Humans , Locomotion/genetics , Locomotion/physiology , Motor Neurons/pathology , Motor Neurons/physiology , Zebrafish/genetics
5.
Hum Mol Genet ; 25(23): 5069-5082, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27798094

ABSTRACT

C9orf72 expansions are the most common genetic cause of FTLD and MND identified to date. Although being intronic, the expansion is translated into five different dipeptide repeat proteins (DPRs) that accumulate within patients' neurons. Attempts have been made to model DPRs in cell and animals. However, the majority of these use DPRs repeat numbers much shorter than those observed in patients. To address this we have generated a selection of DPR expression constructs with repeat numbers in excess of 1000 repeats, matching what is seen in patients. Small and larger DPRs produce inclusions with similar morphology but different cellular effects. We demonstrate a length dependent effect using electrophysiology with a phenotype only occurring with the longest DPRs. These data highlight the importance of using physiologically relevant repeat numbers when modelling DPRs.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Dipeptides/genetics , Frontotemporal Lobar Degeneration/genetics , Proteins/genetics , Amyotrophic Lateral Sclerosis/physiopathology , C9orf72 Protein , DNA Repeat Expansion/genetics , Dipeptides/metabolism , Electrophysiological Phenomena , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/physiopathology , Humans , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Introns/genetics , Neurons/metabolism , Neurons/pathology , Protein Aggregates/genetics , Protein Aggregates/physiology , Proteins/metabolism
6.
Eur J Neurosci ; 44(5): 2214-25, 2016 09.
Article in English | MEDLINE | ID: mdl-27307215

ABSTRACT

Mutations in progranulin (PGRN) have been linked to two neurodegenerative disorders, heterozygote mutations with frontotemporal lobar degeneration (FTLD) and homozygote mutations with neuronal ceroid lipofuscinosis (NCL). Human PGRN is 593aa secreted growth factor, made up of seven and a half repeats of a highly conserved granulin motif that is cleaved to produce the granulin peptides A-G and paragranulin. While it is thought that PGRN protects against neurodegeneration through its role in inflammation and tissue repair, the role of PGRN and granulins in the nervous system is currently unclear. To better understand this, we prepared recombinant PGRN, granulin A-F and paragranulin, and used these to treat differentiated neuronal SH-SY5Y cells. Using RNA sequencing and bioinformatics techniques we investigated the functional effects of PGRN and the individual granulins upon the transcriptome. For PGRN treatment we show that the main effect of short-duration treatments is the down-regulation of transcripts, supporting that signalling pathway induction appears to be dominant effect. Gene ontology analysis, however, also supports the regulation of biological processes such as the spliceosome and proteasome in response to PGRN treatment, as well as the lysosomal pathway constituents such as CHMP1A, further supporting the role of PGRN in lysosomal function. We also show that the response to granulin treatments involves the regulation of numerous non-coding RNA's, and the granulins cluster into groups of similar activity on the basis of expression profile with paragranulin and PGRN having similar expression profiles, while granulins B, D, E and G appear more similar.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Transcriptome , Cell Line, Tumor , Down-Regulation , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomes/metabolism , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/pharmacology , Progranulins , Proteolysis , RNA Splicing , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
7.
Alzheimers Dement ; 12(8): 862-71, 2016 08.
Article in English | MEDLINE | ID: mdl-26993346

ABSTRACT

INTRODUCTION: The genetics underlying posterior cortical atrophy (PCA), typically a rare variant of Alzheimer's disease (AD), remain uncertain. METHODS: We genotyped 302 PCA patients from 11 centers, calculated risk at 24 loci for AD/DLB and performed an exploratory genome-wide association study. RESULTS: We confirm that variation in/near APOE/TOMM40 (P = 6 × 10(-14)) alters PCA risk, but with smaller effect than for typical AD (PCA: odds ratio [OR] = 2.03, typical AD: OR = 2.83, P = .0007). We found evidence for risk in/near CR1 (P = 7 × 10(-4)), ABCA7 (P = .02) and BIN1 (P = .04). ORs at variants near INPP5D and NME8 did not overlap between PCA and typical AD. Exploratory genome-wide association studies confirmed APOE and identified three novel loci: rs76854344 near CNTNAP5 (P = 8 × 10(-10) OR = 1.9 [1.5-2.3]); rs72907046 near FAM46A (P = 1 × 10(-9) OR = 3.2 [2.1-4.9]); and rs2525776 near SEMA3C (P = 1 × 10(-8), OR = 3.3 [2.1-5.1]). DISCUSSION: We provide evidence for genetic risk factors specifically related to PCA. We identify three candidate loci that, if replicated, may provide insights into selective vulnerability and phenotypic diversity in AD.


Subject(s)
Alzheimer Disease/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/pathology , Genetic Predisposition to Disease/genetics , Proteins/genetics , Semaphorins/genetics , Age Factors , Aged , Alzheimer Disease/complications , Apolipoproteins E/genetics , Atrophy/etiology , Female , Genetic Association Studies , Humans , Male , Membrane Transport Proteins/genetics , Middle Aged , Mitochondrial Precursor Protein Import Complex Proteins , Polymorphism, Single Nucleotide/genetics , Polynucleotide Adenylyltransferase , Receptors, Complement 3b/genetics , Risk Factors
8.
Neurobiol Aging ; 36(3): 1601.e1-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25595499

ABSTRACT

Frontotemporal lobar degeneration is a highly familial disease and the most common known genetic cause is the repeat expansion mutation in the gene C9orf72. We have identified 2 brothers with an expansion mutation in C9orf72 using Southern blotting that is undetectable using repeat-primed polymerase chain reaction. Sequencing using high concentrations of DNA denaturants of a bacterial artificial chromosome clone obtained from one of the brothers identified a 10-base pair deletion adjacent to the expansion that presumably confers strong secondary structure that interferes with the genotyping. Using an alternative method, we have identified missed expansion carriers in our cohort, and this number has increased by approximately 25%. This observation has important implications for patients undergoing genetic testing for C9orf72.


Subject(s)
DNA Repeat Expansion/genetics , Frontotemporal Lobar Degeneration/genetics , Gene Deletion , Heterozygote , Mutation/genetics , Proteins/genetics , Adult , Aged , Aged, 80 and over , C9orf72 Protein , Cohort Studies , Female , Frontotemporal Lobar Degeneration/epidemiology , Genetic Testing/methods , Humans , Male , Middle Aged , Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods , Young Adult
9.
Neurobiol Aging ; 36(3): 1603.e5-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25433461

ABSTRACT

Mutations in the gene p62/SQSTM1 have been reported as a relatively rare cause of frontotemporal lobar degeneration (FTLD). To establish whether this was the case for cases of FTLD from the United Kingdom, we sequenced the sequenced the entire open reading frame of this gene in a large cohort of patients. We identified 3 novel mutations in p62/SQSTM1 in 4 patients. One of these was a premature stop codon that removed the last 101 amino acids of the protein that presumably has a negative effect on protein function. Another mutation was also found in a case with a repeat expansion mutation in C9orf72 confirmed by Southern blot. These findings confirm a role of p62/SQSTM1 as a cause of FTLD.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Frontotemporal Lobar Degeneration/genetics , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Mutation/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adult , Aged , Aged, 80 and over , Amino Acids/genetics , C9orf72 Protein , Codon, Nonsense/genetics , Cohort Studies , DNA Repeat Expansion/genetics , Female , Humans , Male , Middle Aged , Open Reading Frames/genetics , Proteins/genetics , Sequestosome-1 Protein , United Kingdom
10.
Neurobiol Aging ; 36(3): 1603.e1-4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25435337

ABSTRACT

We have measured plasma progranulin and interleukin-6 in 230 patients with frontotemporal lobar degeneration (FTLD), 104 patients with Alzheimer's disease, and 161 control subjects. We have replicated previous findings of decreased levels of progranulin protein in FTLD because of mutations in GRN and show this is not observed in FTLD cases because of other causes. interleukin-6 levels were increased in FTLD overall, but these did not discriminate between clinical and genetic subtypes.


Subject(s)
Frontotemporal Lobar Degeneration/diagnosis , Intercellular Signaling Peptides and Proteins/blood , Interleukin-6/blood , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Biomarkers/blood , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Progranulins
11.
Exp Neurol ; 262 Pt B: 84-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24915640

ABSTRACT

One of the most interesting findings in the field of neurodegeneration in recent years is tfche discovery of a genetic mutation in the C9orf72 gene, the most common mutation found to be causative of sporadic and familial frontotemporal lobar degeneration (FTLD), amyotrophic lateral sclerosis (ALS) and concomitant FTD-ALS (DeJesus-Hernandez et al., 2011b; Renton et al., 2011). While clinical and molecular data, such as the identification of TDP-43 being a common pathological protein (Neumann et al., 2006) have hinted at such a link for years, the identification of what was formally known as "the chromosome 9 FTLD-ALS gene" has provided a foundation for better understanding of the relationship between the two. Indeed, it is now recognized that ALS and FTLD-TDP represent a disease spectrum. In this review, we will discuss the current genetic and pathological features of the FTLD-ALS spectrum.


Subject(s)
Amyotrophic Lateral Sclerosis , DNA-Binding Proteins/genetics , Frontotemporal Dementia , Mutation/genetics , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/complications , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Humans
12.
Neurobiol Aging ; 33(8): 1846.e5-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22410647

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a highly familial neurodegenerative disease. It has recently been shown that the most common genetic cause of FTLD and amyotrophic lateral sclerosis (ALS) is a hexanucleotide repeat expansion in C9ORF72. To investigate whether this expansion was specific to the FTLD/ALS disease spectrum, we genotyped the hexanucleotide repeat region of C9ORF72 in a large cohort of patients with Alzheimer's disease (AD). A normal range of repeats was found in all cases. We conclude that the hexanucleotide repeat expansion is specific to the FTLD/ALS disease spectrum.


Subject(s)
Alzheimer Disease/genetics , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease/genetics , Nucleotides/genetics , Proteins/genetics , Repetitive Sequences, Nucleic Acid/genetics , Adult , Aged , Alzheimer Disease/epidemiology , C9orf72 Protein , Comorbidity , Female , Frontotemporal Lobar Degeneration/epidemiology , Genetic Markers/genetics , Humans , Male , Middle Aged , Prevalence , Risk Factors , United Kingdom/epidemiology
13.
Brain ; 135(Pt 3): 693-708, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22300873

ABSTRACT

The identification of a hexanucleotide repeat expansion in the C9ORF72 gene as the cause of chromosome 9-linked frontotemporal dementia and motor neuron disease offers the opportunity for greater understanding of the relationship between these disorders and other clinical forms of frontotemporal lobar degeneration. In this study, we screened a cohort of 398 patients with frontotemporal dementia, progressive non-fluent aphasia, semantic dementia or mixture of these syndromes for mutations in the C9ORF72 gene. Motor neuron disease was present in 55 patients (14%). We identified 32 patients with C9ORF72 mutations, representing 8% of the cohort. The patients' clinical phenotype at presentation varied: nine patients had frontotemporal dementia with motor neuron disease, 19 had frontotemporal dementia alone, one had mixed semantic dementia with frontal features and three had progressive non-fluent aphasia. There was, as expected, a significant association between C9ORF72 mutations and presence of motor neuron disease. Nevertheless, 46 patients, including 22 familial, had motor neuron disease but no mutation in C9ORF72. Thirty-eight per cent of the patients with C9ORF72 mutations presented with psychosis, with a further 28% exhibiting paranoid, deluded or irrational thinking, whereas <4% of non-mutation bearers presented similarly. The presence of psychosis dramatically increased the odds that patients carried the mutation. Mutation bearers showed a low incidence of motor stereotypies, and relatively high incidence of complex repetitive behaviours, largely linked to patients' delusions. They also showed a lower incidence of acquired sweet food preference than patients without C9ORF72 mutations. Post-mortem pathology in five patients revealed transactive response DNA-binding protein 43 pathology, type A in one patient and type B in three. However, one patient had corticobasal degeneration pathology. The findings indicate that C9ORF72 mutations cause some but not all cases of frontotemporal dementia with motor neuron disease. Other mutations remain to be discovered. C9ORF72 mutations are associated with variable clinical presentations and pathology. Nevertheless, the findings highlight a powerful association between C9ORF72 mutations and psychosis and suggest that the behavioural characteristics of patients with C9ORF72 mutations are qualitatively distinct. Mutations in the C9ORF72 gene may be a major cause not only of frontotemporal dementia with motor neuron disease but also of late onset psychosis.


Subject(s)
Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Proteins/genetics , Adult , Age of Onset , Aged , Autopsy , Behavior/physiology , Brain/pathology , C9orf72 Protein , Cerebellum/pathology , Cognition Disorders/etiology , Cognition Disorders/genetics , Cognition Disorders/psychology , Cohort Studies , DNA/genetics , DNA-Binding Proteins/genetics , Demography , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Magnetic Resonance Imaging , Male , Medulla Oblongata/pathology , Middle Aged , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Mutation/genetics , Neuropsychological Tests , Psychotic Disorders/etiology , Psychotic Disorders/psychology , Spinal Cord/pathology
14.
Neurobiol Aging ; 33(2): 425.e1-2, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21074902

ABSTRACT

Frontotemporal lobar degeneration (FTLD) can occur jointly with amyotrophic lateral sclerosis (ALS), and these 2 conditions share a genetic risk factor on chromosome 9. It has been reported that mutations in optineurin (OPTN) can cause ALS. Therefore, we sequenced OPTN in 371 FTLD cases but no mutations were detected, suggesting changes in OPTN do not cause FTLD.


Subject(s)
Frontotemporal Lobar Degeneration/epidemiology , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factor TFIIIA/genetics , Aged , Aged, 80 and over , Cell Cycle Proteins , Female , Humans , Male , Membrane Transport Proteins , Prevalence , Risk Assessment , Risk Factors , Tissue Distribution , United Kingdom/epidemiology
15.
Acta Neuropathol ; 122(6): 703-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21968532

ABSTRACT

TDP-43 immunoreactive (TDP-43-ir) pathological changes were investigated in the temporal cortex and hippocampus of 11 patients with autosomal dominant familial forms of Alzheimer's disease (FAD), 169 patients with sporadic AD [85 with early onset disease (EOAD) (i.e before 65 years of age), and 84 with late onset after this age (LOAD)], 50 individuals with Down's Syndrome (DS) and 5 patients with primary hippocampal sclerosis (HS). TDP-43-ir pathological changes were present, overall, in 34/180 of AD cases. They were present in 1/11 (9%) FAD, and 9/85 (10%) EOAD patients but were significantly more common (p = 0.003) in LOAD where 24/84 (29%) patients showed such changes. There were no demographic differences, other than onset age, between AD patients with or without TDP-43-ir pathological changes. Double immunolabelling indicated that these TDP-43-ir inclusions were frequently ubiquitinated, but were only rarely AT8 (tau) immunoreactive. Only 3 elderly DS individuals and 4/5 cases of primary HS showed similar changes. Overall, 21.7% of AD cases and 6% DS cases showed hippocampal sclerosis (HS). However, only 9% FAD cases and 16% EOAD cases showed HS, but 29% LOAD cases showed HS. The proportion of EOAD cases with both TDP-43 pathology and HS tended to be greater than those in LOAD, where nearly half of all the cases with TDP-43 pathology did not show HS. The presence of TDP-43-ir changes in AD and DS may therefore be a secondary phenomenon, relating more to ageing than to AD itself. Nevertheless, a challenge to such an interpretation comes from the finding in AD of a strong relationship between TDP-43 pathology and cognitive phenotype. Patients with TDP-43 pathology were significantly more likely to present with an amnestic syndrome than those without (p < 0.0001), in keeping with pathological changes in medial temporal lobe structures. HS was also associated more commonly with an amnestic presentation (p < 0.005), but this association disappeared when TDP-43-positive cases were excluded from the analysis. TDP-43 may, after all, be integral to the pathology of AD, and to some extent determine the clinical phenotype present.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , DNA-Binding Proteins/metabolism , Down Syndrome/pathology , Hippocampus/pathology , Phenotype , Adult , Aged , Aged, 80 and over , Aging/metabolism , Alzheimer Disease/metabolism , Apolipoproteins E/genetics , Autopsy , Cognition , Cohort Studies , Down Syndrome/metabolism , Female , Genotype , Hippocampus/metabolism , Humans , Male , Middle Aged , Retrospective Studies , Sclerosis , tau Proteins/metabolism
16.
Arch Neurol ; 68(4): 488-97, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21482928

ABSTRACT

OBJECTIVE: To assess the relative frequency of unique mutations and their associated characteristics in 97 individuals with mutations in progranulin (GRN), an important cause of frontotemporal lobar degeneration (FTLD). PARTICIPANTS AND DESIGN: A 46-site International Frontotemporal Lobar Degeneration Collaboration was formed to collect cases of FTLD with TAR DNA-binding protein of 43-kDa (TDP-43)-positive inclusions (FTLD-TDP). We identified 97 individuals with FTLD-TDP with pathogenic GRN mutations (GRN+ FTLD-TDP), assessed their genetic and clinical characteristics, and compared them with 453 patients with FTLD-TDP in which GRN mutations were excluded (GRN- FTLD-TDP). No patients were known to be related. Neuropathologic characteristics were confirmed as FTLD-TDP in 79 of the 97 GRN+ FTLD-TDP cases and all of the GRN- FTLD-TDP cases. RESULTS: Age at onset of FTLD was younger in patients with GRN+ FTLD-TDP vs GRN- FTLD-TDP (median, 58.0 vs 61.0 years; P < .001), as was age at death (median, 65.5 vs 69.0 years; P < .001). Concomitant motor neuron disease was much less common in GRN+ FTLD-TDP vs GRN- FTLD-TDP (5.4% vs 26.3%; P < .001). Fifty different GRN mutations were observed, including 2 novel mutations: c.139delG (p.D47TfsX7) and c.378C>A (p.C126X). The 2 most common GRN mutations were c.1477C>T (p.R493X, found in 18 patients, representing 18.6% of GRN cases) and c.26C>A (p.A9D, found in 6 patients, representing 6.2% of cases). Patients with the c.1477C>T mutation shared a haplotype on chromosome 17; clinically, they resembled patients with other GRN mutations. Patients with the c.26C>A mutation appeared to have a younger age at onset of FTLD and at death and more parkinsonian features than those with other GRN mutations. CONCLUSION: GRN+ FTLD-TDP differs in key features from GRN- FTLD-TDP.


Subject(s)
Frontotemporal Lobar Degeneration/diagnosis , Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mutation/genetics , Protein Precursors/genetics , Aged , Female , Genetic Markers/genetics , Humans , Male , Middle Aged , Parkinsonian Disorders/genetics , Progranulins
17.
Acta Neuropathol ; 122(1): 99-110, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21424531

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is clinically, pathologically and genetically heterogeneous. Recent descriptions of a pathological sub-type that is ubiquitin positive, TDP-43 negative and immunostains positive for the Fused in Sarcoma protein (FUS) raises the question whether it is associated with a distinct clinical phenotype identifiable on clinical grounds, and whether mutations in the Fused in Sarcoma gene (FUS) might also be associated with FTLD. Examination of a pathological series of 118 cases of FTLD from two centres, showing tau-negative, ubiquitin-positive pathology, revealed FUS pathology in five patients, four classified as atypical FTLD with ubiquitin inclusions (aFTLD-U), and one as neuronal intermediate filament inclusion disease (NIFID). The aFTLD-U cases had youthful onset (22-46 years), an absence of strong family history, a behavioural syndrome consistent with frontotemporal dementia (FTD) and severe caudate atrophy. Their cognitive/behavioural profile was distinct, characterised by prominent obsessionality, repetitive behaviours and rituals, social withdrawal and lack of engagement, hyperorality with pica, and marked stimulus-bound behaviour including utilisation behaviour. They conformed to the rare behavioural sub-type of FTD identified previously by us as the "stereotypic" form, and linked to striatal pathology. Cognitive evaluation revealed executive deficits in keeping with subcortical-frontal dysfunction, but no cortical deficits in language, perceptuospatial skills or praxis. The patient with NIFID was older and exhibited aphasia and dyspraxia. No patient had clinical evidence of motor neurone disease during life, or a mutation in the FUS gene. In the complementary clinical study of 312 patients with clinical syndromes of FTLD, genetic analysis revealed a 6 bp deletion in FUS in 3 patients, of questionable significance. One presented a prototypical picture of FTD, another expressive language disorder, and the third semantic dementia. None showed the early onset age or distinctive 'stereotypic' picture of patients with aFTLD-U. We conclude that aFTLD-U is associated with a distinct clinical form of frontotemporal dementia, potentially allowing identification of such patients in life with a high degree of precision. Whether mutations in the FUS gene cause some cases of FTLD remains unresolved.


Subject(s)
Frontotemporal Dementia/epidemiology , Frontotemporal Dementia/genetics , Frontotemporal Lobar Degeneration/epidemiology , Frontotemporal Lobar Degeneration/genetics , Mutation/genetics , RNA-Binding Protein FUS/genetics , Adult , Brain/metabolism , Brain/pathology , Cognition Disorders/epidemiology , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Comorbidity , DNA-Binding Proteins/metabolism , Female , Frontotemporal Dementia/physiopathology , Frontotemporal Lobar Degeneration/physiopathology , Gene Deletion , Humans , Male , Mental Disorders/epidemiology , Mental Disorders/physiopathology , Mental Disorders/psychology , Middle Aged , Phenotype , RNA-Binding Protein FUS/metabolism , Ubiquitin/metabolism
18.
Neurobiol Aging ; 32(4): 754-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-19446372

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is a highly familial neurodegenerative disease. It has been claimed that homozygosity of the SNP rs5848 located in the 3'UTR of progranulin increases risk for FTLD. We have attempted to replicate the association of rs5848 in three independent FTLD cohorts. No association of rs5848 with FTLD was observed in any individual cohort nor was any observed when the data was combined. These data argue that rs5848 is not a risk factor for FTLD.


Subject(s)
3' Untranslated Regions/genetics , Frontotemporal Lobar Degeneration/genetics , Intercellular Signaling Peptides and Proteins/genetics , Alleles , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Polymorphism, Single Nucleotide , Progranulins
19.
Acta Neuropathol ; 120(1): 33-41, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20490813

ABSTRACT

Through an international consortium, we have collected 37 tau- and TAR DNA-binding protein 43 (TDP-43)-negative frontotemporal lobar degeneration (FTLD) cases, and present here the first comprehensive analysis of these cases in terms of neuropathology, genetics, demographics and clinical data. 92% (34/37) had fused in sarcoma (FUS) protein pathology, indicating that FTLD-FUS is an important FTLD subtype. This FTLD-FUS collection specifically focussed on aFTLD-U cases, one of three recently defined subtypes of FTLD-FUS. The aFTLD-U subtype of FTLD-FUS is characterised clinically by behavioural variant frontotemporal dementia (bvFTD) and has a particularly young age of onset with a mean of 41 years. Further, this subtype had a high prevalence of psychotic symptoms (36% of cases) and low prevalence of motor symptoms (3% of cases). We did not find FUS mutations in any aFTLD-U case. To date, the only subtype of cases reported to have ubiquitin-positive but tau-, TDP-43- and FUS-negative pathology, termed FTLD-UPS, is the result of charged multivesicular body protein 2B gene (CHMP2B) mutation. We identified three FTLD-UPS cases, which are negative for CHMP2B mutation, suggesting that the full complement of FTLD pathologies is yet to be elucidated.


Subject(s)
Frontotemporal Lobar Degeneration/epidemiology , Frontotemporal Lobar Degeneration/metabolism , RNA-Binding Protein FUS/metabolism , Adult , Age of Onset , DNA-Binding Proteins/metabolism , Dyskinesias/epidemiology , Female , Frontal Lobe/metabolism , Frontotemporal Lobar Degeneration/genetics , Hippocampus/metabolism , Humans , Male , Mental Disorders/epidemiology , Middle Aged , Mutation , Prevalence , RNA-Binding Protein FUS/genetics , Sequence Analysis, DNA , tau Proteins/metabolism
20.
Nat Genet ; 42(3): 234-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20154673

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is the second most common cause of presenile dementia. The predominant neuropathology is FTLD with TAR DNA-binding protein (TDP-43) inclusions (FTLD-TDP). FTLD-TDP is frequently familial, resulting from mutations in GRN (which encodes progranulin). We assembled an international collaboration to identify susceptibility loci for FTLD-TDP through a genome-wide association study of 515 individuals with FTLD-TDP. We found that FTLD-TDP associates with multiple SNPs mapping to a single linkage disequilibrium block on 7p21 that contains TMEM106B. Three SNPs retained genome-wide significance following Bonferroni correction (top SNP rs1990622, P = 1.08 x 10(-11); odds ratio, minor allele (C) 0.61, 95% CI 0.53-0.71). The association replicated in 89 FTLD-TDP cases (rs1990622; P = 2 x 10(-4)). TMEM106B variants may confer risk of FTLD-TDP by increasing TMEM106B expression. TMEM106B variants also contribute to genetic risk for FTLD-TDP in individuals with mutations in GRN. Our data implicate variants in TMEM106B as a strong risk factor for FTLD-TDP, suggesting an underlying pathogenic mechanism.


Subject(s)
Chromosomes, Human, Pair 7 , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Inclusion Bodies/metabolism , Polymorphism, Single Nucleotide , Case-Control Studies , Frontotemporal Lobar Degeneration/metabolism , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Inclusion Bodies/genetics , Intercellular Signaling Peptides and Proteins/genetics , Linkage Disequilibrium , Membrane Proteins/genetics , Progranulins
SELECTION OF CITATIONS
SEARCH DETAIL
...