Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; : 106556, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38851544

ABSTRACT

Mutation of the ATL1 gene is one of the most common causes of hereditary spastic paraplegia (HSP), a group of genetic neurodegenerative conditions characterised by distal axonal degeneration of the corticospinal tract axons. Atlastin-1, the protein encoded by ATL1, is one of three mammalian atlastins, which are homologous dynamin-like GTPases that control endoplasmic reticulum (ER) morphology by fusing tubules to form the three-way junctions that characterise ER networks. However, it is not clear whether atlastin-1 is required for correct ER morphology in human neurons and if so what the functional consequences of lack of atlastin-1 are. Using CRISPR-inhibition we generated human cortical neurons lacking atlastin-1. We demonstrate that ER morphology was altered in these neurons, with a reduced number of three-way junctions. Neurons lacking atlastin-1 had longer endosomal tubules, suggestive of defective tubule fission. This was accompanied by reduced lysosomal proteolytic capacity. As well as demonstrating that atlastin-1 is required for correct ER morphology in human neurons, our results indicate that lack of a classical ER-shaping protein such as atlastin-1 may cause altered endosomal tubulation and lysosomal proteolytic dysfunction. Furthermore, they strengthen the idea that defective lysosome function contributes to the pathogenesis of a broad group of HSPs, including those where the primary localisation of the protein involved is not at the endolysosomal system.

2.
medRxiv ; 2024 May 04.
Article in English | MEDLINE | ID: mdl-38746462

ABSTRACT

Solve-RD is a pan-European rare disease (RD) research program that aims to identify disease-causing genetic variants in previously undiagnosed RD families. We utilised 10-fold coverage HiFi long-read sequencing (LRS) for detecting causative structural variants (SVs), single nucleotide variants (SNVs), insertion-deletions (InDels), and short tandem repeat (STR) expansions in extensively studied RD families without clear molecular diagnoses. Our cohort includes 293 individuals from 114 genetically undiagnosed RD families selected by European Rare Disease Network (ERN) experts. Of these, 21 families were affected by so-called 'unsolvable' syndromes for which genetic causes remain unknown, and 93 families with at least one individual affected by a rare neurological, neuromuscular, or epilepsy disorder without genetic diagnosis despite extensive prior testing. Clinical interpretation and orthogonal validation of variants in known disease genes yielded thirteen novel genetic diagnoses due to de novo and rare inherited SNVs, InDels, SVs, and STR expansions. In an additional four families, we identified a candidate disease-causing SV affecting several genes including an MCF2 / FGF13 fusion and PSMA3 deletion. However, no common genetic cause was identified in any of the 'unsolvable' syndromes. Taken together, we found (likely) disease-causing genetic variants in 13.0% of previously unsolved families and additional candidate disease-causing SVs in another 4.3% of these families. In conclusion, our results demonstrate the added value of HiFi long-read genome sequencing in undiagnosed rare diseases.

3.
Am J Hum Genet ; 110(8): 1343-1355, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541188

ABSTRACT

Despite significant progress in unraveling the genetic causes of neurodevelopmental disorders (NDDs), a substantial proportion of individuals with NDDs remain without a genetic diagnosis after microarray and/or exome sequencing. Here, we aimed to assess the power of short-read genome sequencing (GS), complemented with long-read GS, to identify causal variants in participants with NDD from the National Institute for Health and Care Research (NIHR) BioResource project. Short-read GS was conducted on 692 individuals (489 affected and 203 unaffected relatives) from 465 families. Additionally, long-read GS was performed on five affected individuals who had structural variants (SVs) in technically challenging regions, had complex SVs, or required distal variant phasing. Causal variants were identified in 36% of affected individuals (177/489), and a further 23% (112/489) had a variant of uncertain significance after multiple rounds of re-analysis. Among all reported variants, 88% (333/380) were coding nuclear SNVs or insertions and deletions (indels), and the remainder were SVs, non-coding variants, and mitochondrial variants. Furthermore, long-read GS facilitated the resolution of challenging SVs and invalidated variants of difficult interpretation from short-read GS. This study demonstrates the value of short-read GS, complemented with long-read GS, in investigating the genetic causes of NDDs. GS provides a comprehensive and unbiased method of identifying all types of variants throughout the nuclear and mitochondrial genomes in individuals with NDD.


Subject(s)
Genome, Human , Neurodevelopmental Disorders , Humans , Genome, Human/genetics , Chromosome Mapping , Base Sequence , INDEL Mutation , Neurodevelopmental Disorders/genetics
4.
Handb Clin Neurol ; 194: 79-98, 2023.
Article in English | MEDLINE | ID: mdl-36813322

ABSTRACT

Degenerative ataxias and hereditary spastic paraplegias (HSPs) form a continuous, often overlapping disease spectrum sharing not only phenotypic features and underlying genes, but also cellular pathways and disease mechanisms. Mitochondrial metabolism presents a major molecular theme underlying both multiple ataxias and HSPs, thus indicating a heightened vulnerability of Purkinje cells, spinocerebellar tracts, and motor neurons to mitochondrial dysfunction, which is of particular interest for translational approaches. Mitochondrial dysfunction might be the primary (upstream) or secondary (downstream) result of a genetic defect, with underlying genetic defects in nuclear-encoded genes being much more frequent than in mtDNA genes in both, ataxias and HSPs. Here, we outline the substantial number of ataxias, spastic ataxias and HSPs caused by mutated genes implicated in (primary or secondary) mitochondrial dysfunction, highlighting several key "mitochondrial" ataxias and HSPs which are of particular interest for their frequency, pathogenesis and translational opportunities. We then showcase prototypic mitochondrial mechanisms by which disruption of these ataxia and HSP genes contributes to Purkinje cells or corticospinal neuron dysfunction, thus elucidating hypotheses on Purkinje cells and corticospinal neuron vulnerability to mitochondrial dysfunction.


Subject(s)
Mitochondrial Diseases , Spastic Paraplegia, Hereditary , Spinocerebellar Ataxias , Humans , Ataxia , Spinocerebellar Ataxias/genetics , Spastic Paraplegia, Hereditary/genetics , Paraplegia , Mutation
5.
Mol Biol Cell ; 33(12): ar102, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35976706

ABSTRACT

The adaptor protein complex-4 or AP-4 is known to mediate autophagosome maturation through regulating sorting of transmembrane cargo such as ATG9A at the Golgi. There is a need to understand AP-4 function in neurons, as mutations in any of its four subunits cause a complex form of hereditary spastic paraplegia (HSP) with intellectual disability. While AP-4 has been implicated in regulating trafficking and distribution of cargo such as ATG9A and APP, little is known about its effect on neuronal lysosomal protein traffic, lysosome biogenesis, and function. In this study, we demonstrate that in human iPSC-derived neurons AP-4 regulates lysosome composition, function, and transport via regulating the export of critical lysosomal receptors, including Sortilin 1, from the trans-Golgi network to endo-lysosomes. Additionally, loss of AP-4 causes endo-lysosomes to stall and build up in axonal swellings potentially through reduced recruitment of retrograde transport machinery to the organelle. These findings of axonal lysosome buildup are highly reminiscent of those observed in Alzheimer's disease as well as in neurons modeling the most common form of HSP, caused by spastin mutations. Our findings implicate AP-4 as a critical regulator of neuronal lysosome biogenesis and altered lysosome function and axonal endo-lysosome transport as an underlying defect in AP-4-deficient HSP. Additionally, our results also demonstrate the utility of the human i3Neuronal model system in investigating neuronal phenotypes observed in AP-4-deficient mice and/or the human AP-4 deficiency syndrome.


Subject(s)
Adaptor Protein Complex 4 , Spastic Paraplegia, Hereditary , Adaptor Protein Complex 4/metabolism , Animals , Humans , Lysosomes/metabolism , Mice , Neurons/metabolism , Protein Transport , Spastic Paraplegia, Hereditary/metabolism , Spastin/metabolism , trans-Golgi Network/metabolism
6.
Am J Med Genet A ; 185(11): 3446-3458, 2021 11.
Article in English | MEDLINE | ID: mdl-34436830

ABSTRACT

The study aimed at widening the clinical and genetic spectrum of ASXL3-related syndrome, a neurodevelopmental disorder, caused by truncating variants in the ASXL3 gene. In this international collaborative study, we have undertaken a detailed clinical and molecular analysis of 45 previously unpublished individuals with ASXL3-related syndrome, as well as a review of all previously published individuals. We have reviewed the rather limited functional characterization of pathogenic variants in ASXL3 and discuss current understanding of the consequences of the different ASXL3 variants. In this comprehensive analysis of ASXL3-related syndrome, we define its natural history and clinical evolution occurring with age. We report familial ASXL3 pathogenic variants, characterize the phenotype in mildly affected individuals and discuss nonpenetrance. We also discuss the role of missense variants in ASXL3. We delineate a variable but consistent phenotype. The most characteristic features are neurodevelopmental delay with consistently limited speech, significant neuro-behavioral issues, hypotonia, and feeding difficulties. Distinctive features include downslanting palpebral fissures, hypertelorism, tubular nose with a prominent nasal bridge, and low-hanging columella. The presented data will inform clinical management of individuals with ASXL3-related syndrome and improve interpretation of new ASXL3 sequence variants.


Subject(s)
Developmental Disabilities/genetics , Genetic Predisposition to Disease , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics , Adolescent , Adult , Child , Child, Preschool , Developmental Disabilities/epidemiology , Developmental Disabilities/physiopathology , Female , Genetic Variation/genetics , Humans , Hypertelorism/genetics , Hypertelorism/physiopathology , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Male , Muscle Hypotonia/genetics , Muscle Hypotonia/physiopathology , Mutation/genetics , Neurodevelopmental Disorders/epidemiology , Neurodevelopmental Disorders/physiopathology , Phenotype , Young Adult
7.
Neurology ; 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33883237

ABSTRACT

OBJECTIVE: To explore the possibilities of radioligands against the mitochondrial outer membrane protein TSPO as biomarkers for mitochondrial disease, we performed PET (PET)-MR brain imaging with [11C]PK11195 in 14 patients with genetically confirmed mitochondrial disease and 33 matched controls. METHODS: A case-control study of PET-MR imaging with the TSPO radioligand [11C]PK11195. RESULTS: Forty-six percent of symptomatic patients had volumes of abnormal radiotracer binding greater than the 95th percentile in controls. [11C]PK11195 binding was generally greater in grey matter and significantly decreased in white matter. This was most striking in patients with nuclear TYMP or mitochondrial m.3243A>G MT-TL1 mutations, in keeping with differences in mitochondrial density seen post mortem. Some regional binding patterns corresponded to clinical presentation and underlying mutation, even in the absence of structural changes on MRI. This was most obvious for the cerebellum, where patients with ataxia had decreased binding in the cerebellar cortex, but not necessarily volume loss. Overall, there was a positive correlation between aberrant [11C]PK11195 binding and clinical severity. CONCLUSION: These findings endorse the use of PET imaging with TSPO radioligands as a non-invasive in vivo biomarker of mitochondrial pathology. CLASSIFICATION OF EVIDENCE: This study provides Class III evidence that PET-MR brain imaging with TSPO radioligands identifies mitochondrial pathology.

8.
Brain Commun ; 3(1): fcab002, 2021.
Article in English | MEDLINE | ID: mdl-33634263

ABSTRACT

A subset of individuals diagnosed with cerebral palsy will have an underlying genetic diagnosis. Previously, a missense variant in GAD1 was described as a candidate mutation in a single family diagnosed with autosomal recessive spastic cerebral palsy-1 (CPSQ1; OMIM 603513). Following the ascertainment of a further branch of the CPSQ1 kindred, we found that the previously reported GAD1 variant did not segregate with the neurological disease phenotype in the recently ascertained branch of the kindred. Following genetic linkage studies to map autozygous regions and whole-exome sequencing, a missense variant (c.527 T > C; p. Leu176Pro, rs773333490) in the HPDL gene was detected and found to segregate with disease status in both branches of the kindred. HPDL encodes a 371-amino acid protein (4-Hydroxyphenylpyruvate Dioxygenase Like) that localizes to mitochondria but whose function is uncertain. Recently, biallelic loss of function variants and missense substitution-causing variants in HPDL were reported to cause a childhood onset progressive spastic movement disorder with a variable presentation. These findings suggest that HPDL-related neurological disease may mimic spastic cerebral palsy and that GAD1 should not be included in diagnostic gene panels for inherited cerebral palsy.

9.
Nat Commun ; 11(1): 5614, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154382

ABSTRACT

Adult mammalian central nervous system axons have intrinsically poor regenerative capacity, so axonal injury has permanent consequences. One approach to enhancing regeneration is to increase the axonal supply of growth molecules and organelles. We achieved this by expressing the adaptor molecule Protrudin which is normally found at low levels in non-regenerative neurons. Elevated Protrudin expression enabled robust central nervous system regeneration both in vitro in primary cortical neurons and in vivo in the injured adult optic nerve. Protrudin overexpression facilitated the accumulation of endoplasmic reticulum, integrins and Rab11 endosomes in the distal axon, whilst removing Protrudin's endoplasmic reticulum localization, kinesin-binding or phosphoinositide-binding properties abrogated the regenerative effects. These results demonstrate that Protrudin promotes regeneration by functioning as a scaffold to link axonal organelles, motors and membranes, establishing important roles for these cellular components in mediating regeneration in the adult central nervous system.


Subject(s)
Axons/physiology , Central Nervous System/physiology , Endoplasmic Reticulum/metabolism , Nerve Regeneration , Vesicular Transport Proteins/metabolism , Animals , Axons/metabolism , Cells, Cultured , Endoplasmic Reticulum/genetics , Endosomes/metabolism , Female , Humans , Integrins/metabolism , Mice , Mice, Inbred C57BL , Mutation , Nerve Regeneration/drug effects , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Phosphorylation , Protein Domains , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/physiology , Vesicular Transport Proteins/administration & dosage , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
10.
Am J Hum Genet ; 107(6): 1129-1148, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33186545

ABSTRACT

The endosomal sorting complexes required for transport (ESCRTs) are essential for multiple membrane modeling and membrane-independent cellular processes. Here we describe six unrelated individuals with de novo missense variants affecting the ATPase domain of VPS4A, a critical enzyme regulating ESCRT function. Probands had structural brain abnormalities, severe neurodevelopmental delay, cataracts, growth impairment, and anemia. In cultured cells, overexpression of VPS4A mutants caused enlarged endosomal vacuoles resembling those induced by expression of known dominant-negative ATPase-defective forms of VPS4A. Proband-derived fibroblasts had enlarged endosomal structures with abnormal accumulation of the ESCRT protein IST1 on the limiting membrane. VPS4A function was also required for normal endosomal morphology and IST1 localization in iPSC-derived human neurons. Mutations affected other ESCRT-dependent cellular processes, including regulation of centrosome number, primary cilium morphology, nuclear membrane morphology, chromosome segregation, mitotic spindle formation, and cell cycle progression. We thus characterize a distinct multisystem disorder caused by mutations affecting VPS4A and demonstrate that its normal function is required for multiple human developmental and cellular processes.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Mutation, Missense , Neurodevelopmental Disorders/genetics , Vacuolar Proton-Translocating ATPases/genetics , Alleles , Animals , Brain/abnormalities , Cell Cycle , Centrosome/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , Genomics , HEK293 Cells , HeLa Cells , Humans , Mice , Neurons/metabolism , Protein Domains , Protein Transport , Spindle Apparatus/metabolism
11.
Eur J Hum Genet ; 28(12): 1763-1768, 2020 12.
Article in English | MEDLINE | ID: mdl-32934340

ABSTRACT

Hereditary spastic paraplegia (HSP) is a group of heterogeneous inherited degenerative disorders characterized by lower limb spasticity. Fifty percent of HSP patients remain yet genetically undiagnosed. The 100,000 Genomes Project (100KGP) is a large UK-wide initiative to provide genetic diagnosis to previously undiagnosed patients and families with rare conditions. Over 400 HSP families were recruited to the 100KGP. In order to obtain genetic diagnoses, gene-based burden testing was carried out for rare, predicted pathogenic variants using candidate variants from the Exomiser analysis of the genome sequencing data. A significant gene-disease association was identified for UBAP1 and HSP. Three protein truncating variants were identified in 13 patients from 7 families. All patients presented with juvenile form of pure HSP, with median age at onset 10 years, showing autosomal dominant inheritance or de novo occurrence. Additional clinical features included parkinsonism and learning difficulties, but their association with UBAP1 needs to be established.


Subject(s)
Carrier Proteins/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Mutation , Phenotype , Spastic Paraplegia, Hereditary/pathology
12.
Cell Mol Life Sci ; 77(13): 2641-2658, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31587092

ABSTRACT

Mutations in the gene encoding the microtubule severing ATPase spastin are the most frequent cause of hereditary spastic paraplegia, a genetic condition characterised by length-dependent axonal degeneration. Here, we show that HeLa cells lacking spastin and embryonic fibroblasts from a spastin knock-in mouse model become highly polarised and develop cellular protrusions. In HeLa cells, this phenotype was rescued by wild-type spastin, but not by forms unable to sever microtubules or interact with endosomal ESCRT-III proteins. Cells lacking the spastin-interacting ESCRT-III-associated proteins IST1 or CHMP1B also developed protrusions. The protrusion phenotype required protrudin, a RAB-interacting protein that interacts with spastin and localises to ER-endosome contact sites, where it promotes KIF5-dependent endosomal motility to protrusions. Consistent with this, the protrusion phenotype in cells lacking spastin also required KIF5. Lack or mutation of spastin resulted in functional consequences for receptor traffic of a pathway implicated in HSP, as Bone Morphogenetic Protein receptor distribution became polarised. Our results, therefore, identify a novel role for ESCRT-III proteins and spastin in regulating polarised membrane traffic.


Subject(s)
Cell Surface Extensions/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Spastin/metabolism , Animals , Bone Morphogenetic Protein Receptors/metabolism , Cell Membrane/metabolism , Cell Polarity , Cell Surface Extensions/ultrastructure , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knock-In Techniques , HeLa Cells , Humans , Kinesins/physiology , Mice , Protein Transport , Spastic Paraplegia, Hereditary/genetics , Spastin/genetics , Vesicular Transport Proteins/physiology
13.
Front Neurosci ; 13: 1179, 2019.
Article in English | MEDLINE | ID: mdl-31787869

ABSTRACT

The hereditary spastic paraplegias (HSPs) are genetic motor neuron diseases characterized by progressive degeneration of corticospinal tract axons. Mutations in SPAST, encoding the microtubule-severing ATPase spastin, are the most common causes of HSP. The broad SPAST mutational spectrum indicates a haploinsufficiency pathogenic mechanism in most cases. Most missense mutations cluster in the ATPase domain, where they disrupt the protein's ability to sever microtubules. However, several putative missense mutations in the protein's microtubule interacting and trafficking (MIT) domain have also been described, but the pathogenicity of these mutations has not been verified with functional studies. Spastin promotes endosomal tubule fission, and defects in this lead to lysosomal enzyme mistrafficking and downstream lysosomal abnormalities. We investigated the function of three disease-associated spastin MIT mutants and found that none was able to promote normal endosomal tubule fission, lysosomal enzyme receptor trafficking, or lysosomal morphology. One of the mutations affected recruitment of spastin to endosomes, a property that requires the canonical function of the MIT domain in binding endosomal sorting complex required for transport (ESCRT)-III proteins. However, the other mutants did not affect spastin's endosomal recruitment, raising the possibility of pathologically important non-canonical roles for the MIT domain. In conclusion, we demonstrate that spastin MIT mutants cause functional abnormalities related to the pathogenesis of HSP. These mutations do not directly affect spastin's microtubule-severing capacity, and so we identify a new molecular pathological mechanism by which spastin mutations may cause disease.

14.
Lancet Neurol ; 18(12): 1136-1146, 2019 12.
Article in English | MEDLINE | ID: mdl-31377012

ABSTRACT

Hereditary spastic paraplegia (HSP) describes a heterogeneous group of genetic neurodegenerative diseases characterised by progressive spasticity of the lower limbs. The pathogenic mechanism, associated clinical features, and imaging abnormalities vary substantially according to the affected gene and differentiating HSP from other genetic diseases associated with spasticity can be challenging. Next generation sequencing-based gene panels are now widely available but have limitations and a molecular diagnosis is not made in most suspected cases. Symptomatic management continues to evolve but with a greater understanding of the pathophysiological basis of individual HSP subtypes there are emerging opportunities to provide targeted molecular therapies and personalised medicine.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Therapy/methods , Precision Medicine/methods , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/therapy , Genetic Therapy/trends , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/trends , Humans , Mutation/genetics , Precision Medicine/trends , Spastic Paraplegia, Hereditary/diagnostic imaging
15.
Development ; 145(17)2018 09 12.
Article in English | MEDLINE | ID: mdl-30082270

ABSTRACT

Functional analyses of genes responsible for neurodegenerative disorders have unveiled crucial links between neurodegenerative processes and key developmental signalling pathways. Mutations in SPG4-encoding spastin cause hereditary spastic paraplegia (HSP). Spastin is involved in diverse cellular processes that couple microtubule severing to membrane remodelling. Two main spastin isoforms are synthesised from alternative translational start sites (M1 and M87). However, their specific roles in neuronal development and homeostasis remain largely unknown. To selectively unravel their neuronal function, we blocked spastin synthesis from each initiation codon during zebrafish development and performed rescue analyses. The knockdown of each isoform led to different motor neuron and locomotion defects, which were not rescued by the selective expression of the other isoform. Notably, both morphant neuronal phenotypes were observed in a CRISPR/Cas9 spastin mutant. We next showed that M1 spastin, together with HSP proteins atlastin 1 and NIPA1, drives motor axon targeting by repressing BMP signalling, whereas M87 spastin acts downstream of neuropilin 1 to control motor neuron migration. Our data therefore suggest that defective BMP and neuropilin 1 signalling may contribute to the motor phenotype in a vertebrate model of spastin depletion.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Motor Neurons/cytology , Neuropilin-1/metabolism , Spastin/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Axons/metabolism , COS Cells , CRISPR-Cas Systems/genetics , Cell Line , Cell Movement/genetics , Chlorocebus aethiops , GTP-Binding Proteins/metabolism , Gene Knockout Techniques , Humans , Membrane Proteins/metabolism , Protein Isoforms/genetics , Spastic Paraplegia, Hereditary/genetics , Spastin/biosynthesis , Zebrafish Proteins/biosynthesis
16.
Brain ; 141(5): 1286-1299, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29481671

ABSTRACT

Many genetic neurological disorders exhibit variable expression within affected families, often exemplified by variations in disease age at onset. Epistatic effects (i.e. effects of modifier genes on the disease gene) may underlie this variation, but the mechanistic basis for such epistatic interactions is rarely understood. Here we report a novel epistatic interaction between SPAST and the contiguous gene DPY30, which modifies age at onset in hereditary spastic paraplegia, a genetic axonopathy. We found that patients with hereditary spastic paraplegia caused by genomic deletions of SPAST that extended into DPY30 had a significantly younger age at onset. We show that, like spastin, the protein encoded by SPAST, the DPY30 protein controls endosomal tubule fission, traffic of mannose 6-phosphate receptors from endosomes to the Golgi, and lysosomal ultrastructural morphology. We propose that additive effects on this pathway explain the reduced age at onset of hereditary spastic paraplegia in patients who are haploinsufficient for both genes.


Subject(s)
Epistasis, Genetic/genetics , Mutation/genetics , Nuclear Proteins/genetics , Spastic Paraplegia, Hereditary/genetics , Spastin/genetics , Adult , Age of Onset , CD8 Antigens/genetics , CD8 Antigens/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells/metabolism , HeLa Cells/ultrastructure , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomal-Associated Membrane Protein 1/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Nuclear Proteins/metabolism , Nuclear Proteins/ultrastructure , Protein Transport/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Cell Biol ; 216(5): 1337-1355, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28389476

ABSTRACT

Contacts between endosomes and the endoplasmic reticulum (ER) promote endosomal tubule fission, but the mechanisms involved and consequences of tubule fission failure are incompletely understood. We found that interaction between the microtubule-severing enzyme spastin and the ESCRT protein IST1 at ER-endosome contacts drives endosomal tubule fission. Failure of fission caused defective sorting of mannose 6-phosphate receptor, with consequently disrupted lysosomal enzyme trafficking and abnormal lysosomal morphology, including in mouse primary neurons and human stem cell-derived neurons. Consistent with a role for ER-mediated endosomal tubule fission in lysosome function, similar lysosomal abnormalities were seen in cellular models lacking the WASH complex component strumpellin or the ER morphogen REEP1. Mutations in spastin, strumpellin, or REEP1 cause hereditary spastic paraplegia (HSP), a disease characterized by axonal degeneration. Our results implicate failure of the ER-endosome contact process in axonopathy and suggest that coupling of ER-mediated endosomal tubule fission to lysosome function links different classes of HSP proteins, previously considered functionally distinct, into a unifying pathway for axonal degeneration.


Subject(s)
Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Spastic Paraplegia, Hereditary/metabolism , Adult , Animals , Cells, Cultured , Female , HeLa Cells , Humans , Male , Mice , Middle Aged
18.
Am J Hum Genet ; 100(2): 364-370, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157540

ABSTRACT

SPG23 is an autosomal-recessive neurodegenerative subtype of lower limb spastic paraparesis with additional diffuse skin and hair dyspigmentation at birth followed by further patchy pigment loss during childhood. Previously, genome-wide linkage in an Arab-Israeli pedigree mapped the gene to an approximately 25 cM locus on chromosome 1q24-q32. By using whole-exome sequencing in a further Palestinian-Jordanian SPG23 pedigree, we identified a complex homozygous 4-kb deletion/20-bp insertion in DSTYK (dual serine-threonine and tyrosine protein kinase) in all four affected family members. DSTYK is located within the established linkage region and we also found the same mutation in the previously reported pedigree and another Israeli pedigree (total of ten affected individuals from three different families). The mutation removes the last two exons and part of the 3' UTR of DSTYK. Skin biopsies revealed reduced DSTYK protein levels along with focal loss of melanocytes. Ultrastructurally, swollen mitochondria and cytoplasmic vacuoles were also noted in remaining melanocytes and some keratinocytes and fibroblasts. Cultured keratinocytes and fibroblasts from an affected individual, as well as knockdown of Dstyk in mouse melanocytes, keratinocytes, and fibroblasts, were associated with increased cell death after ultraviolet irradiation. Keratinocytes from an affected individual showed loss of kinase activity upon stimulation with fibroblast growth factor. Previously, dominant mutations in DSTYK were implicated in congenital urological developmental disorders, but our study identifies different phenotypic consequences for a recurrent autosomal-recessive deletion mutation in revealing the genetic basis of SPG23.


Subject(s)
Pigmentation Disorders/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Deletion , Spastic Paraplegia, Hereditary/genetics , Vitiligo/genetics , Amino Acid Sequence , Animals , Apoptosis/genetics , Asian People/genetics , Chromosomes, Human, Pair 1/genetics , Exons , Facies , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Linkage , Genetic Loci , Genome-Wide Association Study , Homozygote , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Melanocytes/cytology , Melanocytes/metabolism , Mice , NIH 3T3 Cells , Pedigree , Pigmentation Disorders/diagnosis , Spastic Paraplegia, Hereditary/diagnosis , Vitiligo/diagnosis , Young Adult
19.
PLoS One ; 11(12): e0168294, 2016.
Article in English | MEDLINE | ID: mdl-28006827

ABSTRACT

Recycling of cargos from early endosomes requires regulation of endosomal tubule formation and fission. This regulation is disrupted in cells depleted of the microtubule severing enzyme spastin, causing elongation of endosomal tubules and mis-trafficking of recycling endosomal cargos such as the transferrin receptor. Spastin is encoded by SPAST, mutations in which are the most frequent cause of autosomal dominant hereditary spastic paraplegia, a condition characterised by a progressive loss of lower limb function resulting from upper motor neuron axonopathy. Investigation of molecular factors involved in endosomal tubule regulation is hindered by the need for manual counting of endosomal tubules. We report here the development of an open source automated system for the quantification of endosomal tubules, using ImageJ and R. We validate the method in cells depleted of spastin and its binding partner IST1. The additional speed and reproducibility of this system compared with manual counting makes feasible screens of candidates to further understand the mechanisms of endosomal tubule formation and fission.


Subject(s)
Adenosine Triphosphatases/metabolism , Endosomes/metabolism , Fibroblasts/metabolism , Image Processing, Computer-Assisted/methods , Lung/metabolism , Microscopy, Fluorescence/methods , Microtubules/metabolism , Automation , Cells, Cultured , Fibroblasts/cytology , Fluorescent Antibody Technique , HeLa Cells , Humans , Lung/cytology , Spastin
20.
PLoS One ; 11(3): e0152413, 2016.
Article in English | MEDLINE | ID: mdl-27019090

ABSTRACT

The hereditary spastic paraplegias (HSPs) are genetic conditions in which there is progressive axonal degeneration in the corticospinal tract. Autosomal dominant mutations, including nonsense, frameshift and missense changes, in the gene encoding the microtubule severing ATPase spastin are the most common cause of HSP in North America and northern Europe. In this study we report quantitative gait analysis using a motorized treadmill system, carried out on mice knocked-in for a disease-associated mutation affecting a critical residue in the Walker A motif of the spastin ATPase domain. At 4 months and at one year of age homozygous mutant mice had a number of abnormal gait parameters, including in stride length and stride duration, compared to heterozygous and wild-type littermates. Gait parameters in heterozygous animals did not differ from wild-type littermates. We conclude that quantitative gait analysis using the DigiGait system sensitively detects motor abnormalities in a hereditary spastic paraplegia model, and would be a useful method for analyzing the effects of pharmacological treatments for HSP.


Subject(s)
Adenosine Triphosphatases/genetics , Gait/physiology , Spastic Paraplegia, Hereditary/physiopathology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Analysis of Variance , Animals , Axons/pathology , Cells, Cultured , Disease Models, Animal , Female , Gene Knock-In Techniques , Genotype , Heterozygote , Homozygote , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mutation , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Phenotype , Spastic Paraplegia, Hereditary/genetics , Spastin
SELECTION OF CITATIONS
SEARCH DETAIL
...