Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Blood ; 143(12): 1193-1197, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38237140

ABSTRACT

ABSTRACT: Many patients with antiphospholipid syndrome had decreased ectonucleotidase activity on neutrophils and platelets, which enabled extracellular nucleotides to trigger neutrophil-platelet aggregates. This phenotype was replicated by treating healthy neutrophils and platelets with patient-derived antiphospholipid antibodies or ectonucleotidase inhibitors.


Subject(s)
Antiphospholipid Syndrome , Humans , Neutrophils , Antibodies, Antiphospholipid , Blood Platelets
2.
Acta Neuropathol Commun ; 10(1): 122, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008843

ABSTRACT

Multiple neurotoxic proteinopathies co-exist within vulnerable neuronal populations in all major neurodegenerative diseases. Interactions between these pathologies may modulate disease progression, suggesting they may constitute targets for disease-modifying treatments aiming to slow or halt neurodegeneration. Pairwise interactions between superoxide dismutase 1 (SOD1), TAR DNA-binding protein 43 (TDP-43) and ubiquitin-binding protein 62/sequestosome 1 (p62) proteinopathies have been reported in multiple transgenic cellular and animal models of amyotrophic lateral sclerosis (ALS), however corresponding examination of these relationships in patient tissues is lacking. Further, the coalescence of all three proteinopathies has not been studied in vitro or in vivo to date. These data are essential to guide therapeutic development and enhance the translation of relevant therapies into the clinic. Our group recently profiled SOD1 proteinopathy in post-mortem spinal cord tissues from familial and sporadic ALS cases, demonstrating an abundance of structurally-disordered (dis)SOD1 conformers which become mislocalized within these vulnerable neurons compared with those of aged controls. To explore any relationships between this, and other, ALS-linked proteinopathies, we profiled TDP-43 and p62 within spinal cord motor neurons of the same post-mortem tissue cohort using multiplexed immunofluorescence and immunohistochemistry. We identified distinct patterns of SOD1, TDP43 and p62 co-deposition and subcellular mislocalization between motor neurons of familial and sporadic ALS cases, which we primarily attribute to SOD1 gene status. Our data demonstrate co-deposition of p62 with mutant and wild-type disSOD1 and phosphorylated TDP-43 in familial and sporadic ALS spinal cord motor neurons, consistent with attempts by p62 to mitigate SOD1 and TDP-43 deposition. Wild-type SOD1 and TDP-43 co-deposition was also frequently observed in ALS cases lacking SOD1 mutations. Finally, alterations to the subcellular localization of the three proteins were tightly correlated, suggesting close relationships between the regulatory mechanisms governing the subcellular compartmentalization of these proteins. Our study is the first to profile spatial relationships between SOD1, TDP-43 and p62 pathologies in post-mortem spinal cord motor neurons of ALS patients, previously only studied in vitro. Our findings suggest interactions between these three key ALS-linked proteins are likely to modulate the formation of their respective proteinopathies, and perhaps the rate of motor neuron degeneration, in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Motor Neurons/metabolism , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
3.
Brain ; 145(9): 3108-3130, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35512359

ABSTRACT

Aberrant self-assembly and toxicity of wild-type and mutant superoxide dismutase 1 (SOD1) has been widely examined in silico, in vitro and in transgenic animal models of amyotrophic lateral sclerosis. Detailed examination of the protein in disease-affected tissues from amyotrophic lateral sclerosis patients, however, remains scarce. We used histological, biochemical and analytical techniques to profile alterations to SOD1 protein deposition, subcellular localization, maturation and post-translational modification in post-mortem spinal cord tissues from amyotrophic lateral sclerosis cases and controls. Tissues were dissected into ventral and dorsal spinal cord grey matter to assess the specificity of alterations within regions of motor neuron degeneration. We provide evidence of the mislocalization and accumulation of structurally disordered, immature SOD1 protein conformers in spinal cord motor neurons of SOD1-linked and non-SOD1-linked familial amyotrophic lateral sclerosis cases, and sporadic amyotrophic lateral sclerosis cases, compared with control motor neurons. These changes were collectively associated with instability and mismetallation of enzymatically active SOD1 dimers, as well as alterations to SOD1 post-translational modifications and molecular chaperones governing SOD1 maturation. Atypical changes to SOD1 protein were largely restricted to regions of neurodegeneration in amyotrophic lateral sclerosis cases, and clearly differentiated all forms of amyotrophic lateral sclerosis from controls. Substantial heterogeneity in the presence of these changes was also observed between amyotrophic lateral sclerosis cases. Our data demonstrate that varying forms of SOD1 proteinopathy are a common feature of all forms of amyotrophic lateral sclerosis, and support the presence of one or more convergent biochemical pathways leading to SOD1 proteinopathy in amyotrophic lateral sclerosis. Most of these alterations are specific to regions of neurodegeneration, and may therefore constitute valid targets for therapeutic development.


Subject(s)
Amyotrophic Lateral Sclerosis , Protein Processing, Post-Translational , Superoxide Dismutase-1 , Amyotrophic Lateral Sclerosis/genetics , Humans , Mutation , Spinal Cord/pathology , Superoxide Dismutase-1/genetics
4.
Neurobiol Aging ; 106: 351.e1-351.e6, 2021 10.
Article in English | MEDLINE | ID: mdl-34272080

ABSTRACT

Loss of function (LoF) mutations in Optineurin can cause recessive amyotrophic lateral sclerosis (ALS) with some heterozygous LoF mutations associated with dominant ALS. The molecular mechanisms underlying the variable inheritance pattern associated with OPTN mutations have remained elusive. We identified that affected members of a consanguineous Middle Eastern ALS kindred possessed a novel homozygous p.S174X OPTN mutation. Analysis of these primary fibroblast lines from family members identified that the p.S174X mutation reduces OPTN mRNA expression in an allele-dependent fashion by nonsense mediated decay. Western blotting correlated a reduced expression in heterozygote carriers but a complete absence of OPTN protein in the homozygous carrier. This data suggests that the p.S174X truncation mutation causes recessive ALS through LoF. However, functional analysis detected a significant increase in mitophagy markers TOM20 and COXIV, and higher rates of mitochondrial respiration and ATP levels in heterozygous carriers only. This suggests that heterozygous LoF OPTN mutations may not be causative in a Mendelian manner but may potentially behave as contributory ALS risk factors.


Subject(s)
Alleles , Amyotrophic Lateral Sclerosis/genetics , Cell Cycle Proteins/genetics , Genes, Recessive/genetics , Genetic Association Studies/methods , Loss of Function Mutation/genetics , Membrane Transport Proteins/genetics , Nonsense Mediated mRNA Decay/genetics , Aged , Aged, 80 and over , Consanguinity , Female , Gene Expression/genetics , Heterozygote , Humans , Male , Middle Aged , Middle East , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors
5.
Sci Rep ; 11(1): 13613, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34193962

ABSTRACT

Aberrantly expressed fused in sarcoma (FUS) is a hallmark of FUS-related amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Wildtype FUS localises to synapses and interacts with mitochondrial proteins while mutations have been shown to cause to pathological changes affecting mitochondria, synapses and the neuromuscular junction (NMJ). This indicates a crucial physiological role for FUS in regulating synaptic and mitochondrial function that is currently poorly understood. In this paper we provide evidence that mislocalised cytoplasmic FUS causes mitochondrial and synaptic changes and that FUS plays a vital role in maintaining neuronal health in vitro and in vivo. Overexpressing mutant FUS altered synaptic numbers and neuronal complexity in both primary neurons and zebrafish models. The degree to which FUS was mislocalised led to differences in the synaptic changes which was mirrored by changes in mitochondrial numbers and transport. Furthermore, we showed that FUS co-localises with the mitochondrial tethering protein Syntaphilin (SNPH), and that mutations in FUS affect this relationship. Finally, we demonstrated mutant FUS led to changes in global protein translation. This localisation between FUS and SNPH could explain the synaptic and mitochondrial defects observed leading to global protein translation defects. Importantly, our results support the 'gain-of-function' hypothesis for disease pathogenesis in FUS-related ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Carrier Proteins/metabolism , Mitochondria/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , RNA-Binding Protein FUS/metabolism , Synapses/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Carrier Proteins/genetics , Mitochondria/genetics , Nerve Tissue Proteins/genetics , Neuromuscular Junction/genetics , RNA-Binding Protein FUS/genetics , Rats , Synapses/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Int J Biochem Cell Biol ; 110: 149-153, 2019 05.
Article in English | MEDLINE | ID: mdl-30904737

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a disease with highly heterogenous causes, most of which remain unknown, a multitude of possible disease mechanisms, and no therapy currently available that can halt disease progression. However, recent advances in antisense oligonucleotides have made them a viable option for targeted therapeutics for patients. These molecules offer a method of targeting RNA that is highly specific, adaptable, and does not require viral delivery. Antisense oligonucleotides are therefore being developed for several genetic causes of ALS. Furthermore, biological pathways involved in the pathogenesis of disease also offer tantalizing targets for intervention using antisense oligonucleotides. Here we detail existing and potential targets for antisense oligonucleotides in ALS and briefly examine the requirements for these drugs to reach and be effective in clinic.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Molecular Targeted Therapy/methods , Oligonucleotides, Antisense/genetics , Drug Delivery Systems , Humans , Oligonucleotides, Antisense/chemistry
7.
Neurobiol Aging ; 73: 229.e5-229.e9, 2019 01.
Article in English | MEDLINE | ID: mdl-30348461

ABSTRACT

Analysis of 226 exome-sequenced UK cases of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia identified 2 individuals who harbored a P497H and P506S UBQLN2 mutation, respectively (n = 0.9%). The P506S index case presented with behavioral variant frontotemporal dementia at the age of 54 years then progressed to ALS surviving 3 years. Three sons presented with (1) slowly progressive pure spastic paraplegia with an onset at 25 years and (2) ALS with disease onset of 25 years and survival of 2 years, and (3) ALS presenting symptoms at the age of 26 years, respectively. Analysis of postmortem tissue from the index case revealed frequent neuronal cytoplasmic UBQLN2-positive inclusions in the dentate gyrus and TDP-43-positive neuronal cytoplasmic inclusions in the frontal and temporal cortex and granular cell layer of the dentate gyrus of the hippocampus. Furthermore, a comprehensive analysis of published UBQLN2 mutations demonstrated that only proline-rich domain mutations contribute to a significantly earlier age of onset in male patients (p = 0.0026).


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cell Cycle Proteins/genetics , Frontotemporal Dementia/genetics , Mutation/genetics , Paraplegia/genetics , Ubiquitins/genetics , Adaptor Proteins, Signal Transducing , Adult , Animals , Autophagy-Related Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Dentate Gyrus/metabolism , Disease Progression , Female , Frontal Lobe/metabolism , Humans , Inclusion Bodies/metabolism , Male , Middle Aged , Phenotype , Sex Factors , Temporal Lobe/metabolism , Ubiquitins/metabolism
8.
Neurobiol Aging ; 71: 266.e1-266.e10, 2018 11.
Article in English | MEDLINE | ID: mdl-30033073

ABSTRACT

Mutations in TANK binding kinase 1 (TBK1) have been linked to amyotrophic lateral sclerosis. Some TBK1 variants are nonsense and are predicted to cause disease through haploinsufficiency; however, many other mutations are missense with unknown functional effects. We exome sequenced 699 familial amyotrophic lateral sclerosis patients and identified 16 TBK1 novel or extremely rare protein-changing variants. We characterized a subset of these: p.G217R, p.R357X, and p.C471Y. Here, we show that the p.R357X and p.G217R both abolish the ability of TBK1 to phosphorylate 2 of its kinase targets, IRF3 and optineurin, and to undergo phosphorylation. They both inhibit binding to optineurin and the p.G217R, within the TBK1 kinase domain, reduces homodimerization, essential for TBK1 activation and function. Finally, we show that the proportion of TBK1 that is active (phosphorylated) is reduced in 5 lymphoblastoid cell lines derived from patients harboring heterozygous missense or in-frame deletion TBK1 mutations. We conclude that missense mutations in functional domains of TBK1 impair the binding and phosphorylation of its normal targets, implicating a common loss of function mechanism, analogous to truncation mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Protein Serine-Threonine Kinases/genetics , Cell Cycle Proteins , Codon, Nonsense , Exons , Female , Genetic Association Studies , Humans , Interferon Regulatory Factor-3/genetics , Male , Membrane Transport Proteins , Mutation, Missense , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Transcription Factor TFIIIA/genetics
9.
Hum Mol Genet ; 27(3): 463-474, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29194538

ABSTRACT

FUS (fused in sarcoma) mislocalization and cytoplasmic aggregation are hallmark pathologies in FUS-related amyotrophic lateral sclerosis and frontotemporal dementia. Many of the mechanistic hypotheses have focused on a loss of nuclear function in the FUS-opathies, implicating dysregulated RNA transcription and splicing in driving neurodegeneration. Recent studies describe an additional somato-dendritic localization for FUS in the cerebral cortex implying a regulatory role in mRNA transport and local translation at the synapse. Here, we report that FUS is also abundant at the pre-synaptic terminal of the neuromuscular junction (NMJ), suggesting an important function for this protein at peripheral synapses. We have previously reported dose and age-dependent motor neuron degeneration in transgenic mice overexpressing human wild-type FUS, resulting in a motor phenotype detected by ∼28 days and death by ∼100 days. Now, we report the earliest structural events using electron microscopy and quantitative immunohistochemistry. Mitochondrial abnormalities in the pre-synaptic motor nerve terminals are detected at postnatal day 6, which are more pronounced at P15 and accompanied by a loss of synaptic vesicles and synaptophysin protein coupled with NMJs of a smaller size at a time when there is no detectable motor neuron loss. These changes occur in the presence of abundant FUS and support a peripheral toxic gain of function. This appearance is typical of a 'dying-back' axonopathy, with the earliest manifestation being mitochondrial disruption. These findings support our hypothesis that FUS has an important function at the NMJ, and challenge the 'loss of nuclear function' hypothesis for disease pathogenesis in the FUS-opathies.


Subject(s)
Neuromuscular Junction/metabolism , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cerebral Cortex/metabolism , Disease Models, Animal , Frontotemporal Dementia/metabolism , Humans , Mice , Mice, Transgenic , Motor Neurons , RNA-Binding Protein FUS/genetics , Synapses/metabolism , Synaptophysin/metabolism
10.
Sci Transl Med ; 9(388)2017 05 03.
Article in English | MEDLINE | ID: mdl-28469040

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder. We screened 751 familial ALS patient whole-exome sequences and identified six mutations including p.D40G in the ANXA11 gene in 13 individuals. The p.D40G mutation was absent from 70,000 control whole-exome sequences. This mutation segregated with disease in two kindreds and was present in another two unrelated cases (P = 0.0102), and all mutation carriers shared a common founder haplotype. Annexin A11-positive protein aggregates were abundant in spinal cord motor neurons and hippocampal neuronal axons in an ALS patient carrying the p.D40G mutation. Transfected human embryonic kidney cells expressing ANXA11 with the p.D40G mutation and other N-terminal mutations showed altered binding to calcyclin, and the p.R235Q mutant protein formed insoluble aggregates. We conclude that mutations in ANXA11 are associated with ALS and implicate defective intracellular protein trafficking in disease pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Annexins/genetics , Annexins/metabolism , Human Embryonic Stem Cells/metabolism , Humans , Mutation/genetics , Protein Binding , Protein Transport , S100 Calcium Binding Protein A6/metabolism
12.
Acta Neuropathol ; 134(1): 113-127, 2017 07.
Article in English | MEDLINE | ID: mdl-28527045

ABSTRACT

Neuronal loss in numerous neurodegenerative disorders has been linked to protein aggregation and oxidative stress. Emerging data regarding overlapping proteinopathy in traditionally distinct neurodegenerative diseases suggest that disease-modifying treatments targeting these pathological features may exhibit efficacy across multiple disorders. Here, we describe proteinopathy distinct from classic synucleinopathy, predominantly comprised of the anti-oxidant enzyme superoxide dismutase-1 (SOD1), in the Parkinson's disease brain. Significant expression of this pathology closely reflected the regional pattern of neuronal loss. The protein composition and non-amyloid macrostructure of these novel aggregates closely resembles that of neurotoxic SOD1 deposits in SOD1-associated familial amyotrophic lateral sclerosis (fALS). Consistent with the hypothesis that deposition of protein aggregates in neurodegenerative disorders reflects upstream dysfunction, we demonstrated that SOD1 in the Parkinson's disease brain exhibits evidence of misfolding and metal deficiency, similar to that seen in mutant SOD1 in fALS. Our data suggest common mechanisms of toxic SOD1 aggregation in both disorders and a potential role for SOD1 dysfunction in neuronal loss in the Parkinson's disease brain. This shared restricted proteinopathy highlights the potential translation of therapeutic approaches targeting SOD1 toxicity, already in clinical trials for ALS, into disease-modifying treatments for Parkinson's disease.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , Parkinson Disease/pathology , Superoxide Dismutase-1/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/enzymology , Brain/enzymology , Cell Count , Female , Humans , Immunoblotting , Immunohistochemistry , Lewy Bodies/enzymology , Lewy Bodies/pathology , Male , Microscopy, Fluorescence , Middle Aged , Neurons/enzymology , Neurons/pathology , Parkinson Disease/enzymology , Protein Aggregation, Pathological/enzymology , Protein Aggregation, Pathological/pathology , Protein Folding , Spinal Cord/enzymology , Spinal Cord/pathology
13.
Neuron ; 94(2): 322-336.e5, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28392072

ABSTRACT

Recent progress revealed the complexity of RNA processing and its association to human disorders. Here, we unveil a new facet of this complexity. Complete loss of function of the ubiquitous splicing factor SFPQ affects zebrafish motoneuron differentiation cell autonomously. In addition to its nuclear localization, the protein unexpectedly localizes to motor axons. The cytosolic version of SFPQ abolishes motor axonal defects, rescuing key transcripts, and restores motility in the paralyzed sfpq null mutants, indicating a non-nuclear processing role in motor axons. Novel variants affecting the conserved coiled-coil domain, so far exclusively found in fALS exomes, specifically affect the ability of SFPQ to localize in axons. They broadly rescue morphology and motility in the zebrafish mutant, but alter motor axon morphology, demonstrating functional requirement for axonal SFPQ. Altogether, we uncover the axonal function of the splicing factor SFPQ in motor development and highlight the importance of the coiled-coil domain in this process. VIDEO ABSTRACT.


Subject(s)
Axons/metabolism , Motor Neurons/metabolism , PTB-Associated Splicing Factor/metabolism , RNA Splicing/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , DNA-Binding Proteins/metabolism , Humans , Mice , Motor Cortex/growth & development , PTB-Associated Splicing Factor/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Zebrafish
14.
Neurobiol Aging ; 49: 214.e1-214.e5, 2017 01.
Article in English | MEDLINE | ID: mdl-27480424

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease, which causes progressive and eventually fatal loss of motor function. Here, we describe genetic and pathologic characterization of brain tissue banked from 19 ALS patients over nearly 20 years at the Department of Anatomy and the Centre for Brain Research, University of Auckland, New Zealand. We screened for mutations in SOD1, TARDBP, FUS, and C9ORF72 genes and for neuropathology caused by phosphorylated TDP-43, dipeptide repeats (DPRs), and ubiquilin. We identified 2 cases with C9ORF72 repeat expansions. Both harbored phosphorylated TDP-43 and DPR inclusions. We show that DPR inclusions can incorporate or occur independently of ubiquilin. We also identified 1 case with a UBQLN2 mutation, which showed phosphorylated TDP-43 and characteristic ubiquilin protein inclusions. This is the first study of ALS genetics in New Zealand, adding New Zealand to the growing list of countries in which C9ORF72 repeat expansion and UBQLN2 mutations are detected in ALS cases.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Brain/pathology , C9orf72 Protein/genetics , Cell Cycle Proteins/genetics , DNA Repeat Expansion/genetics , Genetic Association Studies , Mutation/genetics , Ubiquitins/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Autophagy-Related Proteins , Female , Humans , Male , Middle Aged , New Zealand
15.
Nat Genet ; 48(9): 1037-42, 2016 09.
Article in English | MEDLINE | ID: mdl-27455347

ABSTRACT

To identify genetic factors contributing to amyotrophic lateral sclerosis (ALS), we conducted whole-exome analyses of 1,022 index familial ALS (FALS) cases and 7,315 controls. In a new screening strategy, we performed gene-burden analyses trained with established ALS genes and identified a significant association between loss-of-function (LOF) NEK1 variants and FALS risk. Independently, autozygosity mapping for an isolated community in the Netherlands identified a NEK1 p.Arg261His variant as a candidate risk factor. Replication analyses of sporadic ALS (SALS) cases and independent control cohorts confirmed significant disease association for both p.Arg261His (10,589 samples analyzed) and NEK1 LOF variants (3,362 samples analyzed). In total, we observed NEK1 risk variants in nearly 3% of ALS cases. NEK1 has been linked to several cellular functions, including cilia formation, DNA-damage response, microtubule stability, neuronal morphology and axonal polarity. Our results provide new and important insights into ALS etiopathogenesis and genetic etiology.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genetic Predisposition to Disease , Mutation/genetics , NIMA-Related Kinase 1/genetics , Amyotrophic Lateral Sclerosis/epidemiology , Case-Control Studies , Cohort Studies , Exome/genetics , Genetic Association Studies , Humans , Netherlands/epidemiology
16.
Nat Commun ; 7: 11253, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27080313

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are overlapping, fatal neurodegenerative disorders in which the molecular and pathogenic basis remains poorly understood. Ubiquitinated protein aggregates, of which TDP-43 is a major component, are a characteristic pathological feature of most ALS and FTD patients. Here we use genome-wide linkage analysis in a large ALS/FTD kindred to identify a novel disease locus on chromosome 16p13.3. Whole-exome sequencing identified a CCNF missense mutation at this locus. Interrogation of international cohorts identified additional novel CCNF variants in familial and sporadic ALS and FTD. Enrichment of rare protein-altering CCNF variants was evident in a large sporadic ALS replication cohort. CCNF encodes cyclin F, a component of an E3 ubiquitin-protein ligase complex (SCF(Cyclin F)). Expression of mutant CCNF in neuronal cells caused abnormal ubiquitination and accumulation of ubiquitinated proteins, including TDP-43 and a SCF(Cyclin F) substrate. This implicates common mechanisms, linked to protein homeostasis, underlying neuronal degeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cyclins/genetics , Frontotemporal Dementia/genetics , Genetic Predisposition to Disease/genetics , Mutation, Missense , Adult , Aged , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Human, Pair 16/genetics , Family Health , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Pedigree , Sequence Analysis, DNA/methods , Sequence Homology, Amino Acid
17.
Brain ; 139(Pt 5): 1417-32, 2016 05.
Article in English | MEDLINE | ID: mdl-26936937

ABSTRACT

Detergent-resistant, ubiquitinated and hyperphosphorylated Tar DNA binding protein 43 (TDP-43, encoded by TARDBP) neuronal cytoplasmic inclusions are the pathological hallmark in ∼95% of amyotrophic lateral sclerosis and ∼60% of frontotemporal lobar degeneration cases. We sought to explore the role for the heat shock response in the clearance of insoluble TDP-43 in a cellular model of disease and to validate our findings in transgenic mice and human amyotrophic lateral sclerosis tissues. The heat shock response is a stress-responsive protective mechanism regulated by the transcription factor heat shock factor 1 (HSF1), which increases the expression of chaperones that refold damaged misfolded proteins or facilitate their degradation. Here we show that manipulation of the heat shock response by expression of dominant active HSF1 results in a dramatic reduction of insoluble and hyperphosphorylated TDP-43 that enhances cell survival, whereas expression of dominant negative HSF1 leads to enhanced TDP-43 aggregation and hyperphosphorylation. To determine which chaperones were mediating TDP-43 clearance we over-expressed a range of heat shock proteins (HSPs) and identified DNAJB2a (encoded by DNAJB2, and also known as HSJ1a) as a potent anti-aggregation chaperone for TDP-43. DNAJB2a has a J domain, allowing it to interact with HSP70, and ubiquitin interacting motifs, which enable it to engage the degradation of its client proteins. Using functionally deleted DNAJB2a constructs we demonstrated that TDP-43 clearance was J domain-dependent and was not affected by ubiquitin interacting motif deletion or proteasome inhibition. This indicates that TDP-43 is maintained in a soluble state by DNAJB2a, leaving the total levels of TDP-43 unchanged. Additionally, we have demonstrated that the levels of HSF1 and heat shock proteins are significantly reduced in affected neuronal tissues from a TDP-43 transgenic mouse model of amyotrophic lateral sclerosis and patients with sporadic amyotrophic lateral sclerosis. This implies that the HSF1-mediated DNAJB2a/HSP70 heat shock response pathway is compromised in amyotrophic lateral sclerosis. Defective refolding of TDP-43 is predicted to aggravate the TDP-43 proteinopathy. The finding that the pathological accumulation of insoluble TDP-43 can be reduced by the activation of HSF1/HSP pathways presents an exciting opportunity for the development of novel therapeutics.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Heat-Shock Proteins/metabolism , Transcription Factors/physiology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cell Survival/genetics , DNA-Binding Proteins/biosynthesis , HSP40 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/physiology , Heat Shock Transcription Factors , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Transcription Factors/biosynthesis , Ubiquitin/metabolism
18.
Acta Neuropathol Commun ; 3: 62, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26452761

ABSTRACT

INTRODUCTION: Mutations in the FUS gene have been shown to be a rare cause of amyotrophic lateral sclerosis (ALS-FUS) and whilst well documented clinically and genetically there have been relatively few neuropathological studies.Recent work suggested a possible correlation between pathological features such as frequency of basophilic inclusions in neurons and rate of clinical decline, other studies have revealed a discrepancy between the upper motor neuron features detected clinically and the associated pathology. The purpose of this study was to describe the pathological features associated with more recently discovered FUS mutations and reinvestigate those with well recognised mutations in an attempt to correlate the pathology with mutation and/or clinical phenotype. The brains and spinal cords of seven cases of ALS-FUS were examined neuropathologically, including cases with the newly described p.K510E mutation and a case with both a known p.P525L mutation in the FUS gene and a truncating p.Y374X mutation in the TARDBP gene. RESULTS: The neuropathology in all cases revealed basophilic and FUS inclusions in the cord. The density and type of inclusions varied markedly between cases, but did not allow a clear correlation with clinical progression. Only one case showed significant motor cortical pathology despite the upper motor neuron clinical features being evident in 4 patients. The case with both a FUS and TARDBP mutation revealed FUS positive inclusions but no TDP-43 pathology. Instead there were unusual p62 positive, FUS negative neuronal and glial inclusions as well as dot-like neurites. CONCLUSIONS: The study confirms cases of ALS-FUS to be mainly a lower motor neuron disease and to have pathology that does not appear to neatly correlate with clinical features or genetics. Furthermore, the case with both a FUS and TARDBP mutation reveals an intriguing pathological profile which at least in part involves a very unusual staining pattern for the ubiquitin-binding protein p62.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , Mutation/genetics , RNA-Binding Protein FUS/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/metabolism , Brain/pathology , DNA-Binding Proteins/metabolism , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Middle Aged , Motor Neuron Disease , Motor Neurons/metabolism , Sequestosome-1 Protein , Spinal Cord/metabolism , Spinal Cord/pathology , Young Adult
19.
Neurobiol Aging ; 36(10): 2908.e17-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26344877

ABSTRACT

Mutations in CHCHD10 have recently been reported as a cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. To address the genetic contribution of CHCHD10 to ALS, we have screened a cohort of 425 UK ALS ± frontotemporal dementia patients and 576 local controls in all coding exons of CHCHD10 by Sanger sequencing. We identified a previously reported p.P34S variant that is also present in neurologically healthy controls (p = 0.58). Our results suggest that CHCHD10 is not a primary cause of ALS in UK cases.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genetic Association Studies , Genetic Variation/genetics , Mitochondrial Proteins/genetics , Cohort Studies , Exons/genetics , Female , Humans , Male , Open Reading Frames/genetics , Sequence Analysis, DNA/methods , United Kingdom
20.
Acta Neuropathol Commun ; 3: 36, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26108367

ABSTRACT

INTRODUCTION: Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive neurodegenerative disorder, and cytoplasmic inclusions containing transactive response (TAR) DNA binding protein (TDP-43) are present in ~90 % of cases. Here we report detailed pathology in human TDP-43 transgenic mice that recapitulate key features of TDP-43-linked ALS. RESULTS: Expression of human wild-type TDP-43 (TDP-43(WT)) caused no clinical or pathological phenotype, while expression of Q331K mutant (TDP-43(Q331K)) resulted in a non-lethal age-dependent motor phenotype, accompanied by cytoplasmic TDP-43 aggregation, mild neuronal loss, with astroglial and microglial activation in the motor cortex and spinal cord at 24 months. However, co-expression of WT and Q331K mutant (TDP-43(WTxQ331K)) resulted in an extremely aggressive motor phenotype with tremor from 3 weeks and progressive hind-limb paralysis necessitating euthanasia by 8-10 weeks of age. Neuronal loss and reactive gliosis was observed in the spinal cord and layer V region of the cortex, with TDP-43, ubiquitin and p62 cytoplasmic inclusions and an increase in insoluble TDP-43. Nuclear clearance of TDP-43 was not observed in TDP-43(Q331K) mice but was seen in 65 % of aggregate containing spinal cord motor neurons in TDP-43(WTxQ331K) mice. CONCLUSIONS: We hypothesise that cytoplasmic TDP-43(Q331K) aggregates facilitate the recruitment of WT protein in compound animals, which dramatically accelerates neurodegeneration and disease progression. The exploration of disease mechanisms in slow and rapid disease models of TDP-43 proteinopathy will help elucidate novel drug targets and provide a more informative platform for preclinical trials.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cerebral Cortex/pathology , DNA-Binding Proteins/genetics , Mutation/genetics , Neurons/pathology , Age Factors , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Antigens, CD/metabolism , Cytoplasm/metabolism , Cytoplasm/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL