Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 109(3): 518-532, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35108495

ABSTRACT

Cell adhesion molecules are membrane-bound proteins predominantly expressed in the central nervous system along principal axonal pathways with key roles in nervous system development, neural cell differentiation and migration, axonal growth and guidance, myelination, and synapse formation. Here, we describe ten affected individuals with bi-allelic variants in the neuronal cell adhesion molecule NRCAM that lead to a neurodevelopmental syndrome of varying severity; the individuals are from eight families. This syndrome is characterized by developmental delay/intellectual disability, hypotonia, peripheral neuropathy, and/or spasticity. Computational analyses of NRCAM variants, many of which cluster in the third fibronectin type III (Fn-III) domain, strongly suggest a deleterious effect on NRCAM structure and function, including possible disruption of its interactions with other proteins. These findings are corroborated by previous in vitro studies of murine Nrcam-deficient cells, revealing abnormal neurite outgrowth, synaptogenesis, and formation of nodes of Ranvier on myelinated axons. Our studies on zebrafish nrcamaΔ mutants lacking the third Fn-III domain revealed that mutant larvae displayed significantly altered swimming behavior compared to wild-type larvae (p < 0.03). Moreover, nrcamaΔ mutants displayed a trend toward increased amounts of α-tubulin fibers in the dorsal telencephalon, demonstrating an alteration in white matter tracts and projections. Taken together, our study provides evidence that NRCAM disruption causes a variable form of a neurodevelopmental disorder and broadens the knowledge on the growing role of the cell adhesion molecule family in the nervous system.


Subject(s)
Neurodevelopmental Disorders , Peripheral Nervous System Diseases , Animals , Axons/metabolism , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules, Neuronal , Humans , Mice , Muscle Hypotonia/genetics , Muscle Hypotonia/metabolism , Muscle Spasticity/metabolism , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Zebrafish/genetics , Zebrafish/metabolism
2.
Genet Med ; 26(6): 101117, 2024 06.
Article in English | MEDLINE | ID: mdl-38459834

ABSTRACT

PURPOSE: We describe 3 families with Charcot-Marie-Tooth neuropathy (CMT), harboring a homozygous NDUFS6 NM_004553.6:c.309+5G>A variant previously linked to fatal Leigh syndrome. We aimed to characterize clinically and molecularly the newly identified patients and understand the mechanism underlying their milder phenotype. METHODS: The patients underwent extensive clinical examinations. Exome sequencing was done in 4 affected individuals. The functional effect of the c.309+5G>A variant was investigated in patient-derived EBV-transformed lymphoblasts at the complementary DNA, protein, and mitochondrial level. Alternative splicing was evaluated using complementary DNA long-read sequencing. RESULTS: All patients presented with early-onset, slowly progressive axonal CMT, and nystagmus; some exhibited additional central nervous system symptoms. The c.309+5G>A substitution caused the expression of aberrantly spliced transcripts and negligible levels of the canonical transcript. Immunoblotting showed reduced levels of mutant isoforms. No detectable defects in mitochondrial complex stability or bioenergetics were found. CONCLUSION: We expand the clinical spectrum of NDUFS6-related mitochondrial disorders to include axonal CMT, emphasizing the clinical and pathophysiologic overlap between these 2 clinical entities. This work demonstrates the critical role that alternative splicing may play in modulating the severity of a genetic disorder, emphasizing the need for careful consideration when interpreting splice variants and their implications on disease prognosis.


Subject(s)
Alternative Splicing , Charcot-Marie-Tooth Disease , Mitochondrial Diseases , Humans , Alternative Splicing/genetics , Male , Female , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Child , NADH Dehydrogenase/genetics , Pedigree , Mutation/genetics , Phenotype , Exome Sequencing , Leigh Disease/genetics , Leigh Disease/pathology , Mitochondria/genetics , Mitochondria/pathology , Electron Transport Complex I/genetics , Adult , Child, Preschool , Adolescent
3.
Molecules ; 29(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731451

ABSTRACT

A novel second-generation blue fluorescent polyamidoamine dendrimer peripherally modified with sixteen 4-N,N-dimethylaninoethyloxy-1,8-naphthalimide units was synthesized. Its basic photophysical characteristics were investigated in organic solvents of different polarity. It was found that in these solvents, the dendrimer is colorless and emitted blue fluorescence with different intensities depending on their polarity. The effect of the pH of the medium on the fluorescence intensity was investigated and it was found that in the acidic medium, the fluorescence is intense and is quenched in the alkaline medium. The ability of the dendrimer to detect metal ions (Pb2+, Zn2+, Mg2+, Sn2+, Ba2+, Ni2+, Sn2+, Mn2+, Co2+, Fe3+, and Al3+) was also investigated, and it was found that in the presence of Fe3+, the fluorescent intensity was amplified more than 66 times. The antimicrobial activity of the new compound has been tested in vitro against Gram-positive B. cereus and Gram-negative P. aeruginosa. The tests were performed in the dark and after irradiation with visible light. The antimicrobial activity of the compound enhanced after light irradiation and B. cereus was found slightly more sensitive than P. aeruginosa. The increase in antimicrobial activity after light irradiation is due to the generation of singlet oxygen particles, which attack bacterial cell membranes.


Subject(s)
Dendrimers , Microbial Sensitivity Tests , Naphthalimides , Polyamines , Naphthalimides/chemistry , Naphthalimides/pharmacology , Dendrimers/chemistry , Dendrimers/pharmacology , Polyamines/chemistry , Polyamines/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Fluorescence , Pseudomonas aeruginosa/drug effects , Hydrogen-Ion Concentration , Bacillus cereus/drug effects , Light , Fluorescent Dyes/chemistry , Spectrometry, Fluorescence
4.
Int J Mol Sci ; 24(22)2023 Nov 09.
Article in English | MEDLINE | ID: mdl-38003325

ABSTRACT

Investigating the impact of disease-causing mutations, their affected pathways, and/or potential therapeutic strategies using disease modeling often requires the generation of different in vivo and in cellulo models. To date, several approaches have been established to induce transgene expression in a controlled manner in different model systems. Several rounds of subcloning are, however, required, depending on the model organism used, thus bringing labor-intensive experiments into the technical approach and analysis comparison. The GeneSwitch™ technology is an adapted version of the classical UAS-GAL4 inducible system, allowing the spatial and temporal modulation of transgene expression. It consists of three components: a plasmid encoding for the chimeric regulatory pSwitch protein, Mifepristone as an inducer, and an inducible plasmid. While the pSwitch-containing first plasmid can be used both in vivo and in cellulo, the inducible second plasmid can only be used in cellulo. This requires a specific subcloning strategy of the inducible plasmid tailored to the model organism used. To avoid this step and unify gene expression in the transgenic models generated, we replaced the backbone vector with standard pUAS-attB plasmid for both plasmids containing either the chimeric GeneSwitch™ cDNA sequence or the transgene cDNA sequence. We optimized this adapted system to regulate transgene expression in several mammalian cell lines. Moreover, we took advantage of this new system to generate unified cellular and fruit fly models for YARS1-induced Charco-Marie-Tooth neuropathy (CMT). These new models displayed the expected CMT-like phenotypes. In the N2a neuroblastoma cells expressing YARS1 transgenes, we observed the typical "teardrop" distribution of the synthetase that was perturbed when expressing the YARS1CMT mutation. In flies, the ubiquitous expression of YARS1CMT induced dose-dependent developmental lethality and pan-neuronal expression caused locomotor deficit, while expression of the wild-type allele was harmless. Our proof-of-concept disease modeling studies support the efficacy of the adapted transgenesis system as a powerful tool allowing the design of studies with optimal data comparability.


Subject(s)
Charcot-Marie-Tooth Disease , Tyrosine-tRNA Ligase , Animals , DNA, Complementary/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Drosophila/genetics , Mutation , Neurons/metabolism , Tyrosine-tRNA Ligase/metabolism , Disease Models, Animal , Mammals/genetics
5.
Am J Hum Genet ; 104(4): 767-773, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30929741

ABSTRACT

The diagnostic gap for rare neurodegenerative diseases is still considerable, despite continuous advances in gene identification. Many novel Mendelian genes have only been identified in a few families worldwide. Here we report the identification of an autosomal-dominant gene for hereditary spastic paraplegia (HSP) in 10 families that are of diverse geographic origin and whose affected members all carry unique truncating changes in a circumscript region of UBAP1 (ubiquitin-associated protein 1). HSP is a neurodegenerative disease characterized by progressive lower-limb spasticity and weakness, as well as frequent bladder dysfunction. At least 40% of affected persons are currently undiagnosed after exome sequencing. We identified pathological truncating variants in UBAP1 in affected persons from Iran, USA, Germany, Canada, Spain, and Bulgarian Roma. The genetic support ranges from linkage in the largest family (LOD = 8.3) to three confirmed de novo mutations. We show that mRNA in the fibroblasts of affected individuals escapes nonsense-mediated decay and thus leads to the expression of truncated proteins; in addition, concentrations of the full-length protein are reduced in comparison to those in controls. This suggests either a dominant-negative effect or haploinsufficiency. UBAP1 links endosomal trafficking to the ubiquitination machinery pathways that have been previously implicated in HSPs, and UBAP1 provides a bridge toward a more unified pathophysiology.


Subject(s)
Carrier Proteins/genetics , Mutation , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Databases, Factual , Disease Models, Animal , Endosomes/metabolism , Family Health , Female , Fibroblasts/metabolism , Genes, Dominant , Genetic Linkage , Genetic Predisposition to Disease , Genomics , HEK293 Cells , Haploinsufficiency , Humans , Male , Middle Aged , Pedigree , Protein Isoforms , Young Adult , Zebrafish
6.
Nature ; 526(7575): 710-4, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26503042

ABSTRACT

Selective neuronal loss is a hallmark of neurodegenerative diseases, which, counterintuitively, are often caused by mutations in widely expressed genes. Charcot-Marie-Tooth (CMT) diseases are the most common hereditary peripheral neuropathies, for which there are no effective therapies. A subtype of these diseases--CMT type 2D (CMT2D)--is caused by dominant mutations in GARS, encoding the ubiquitously expressed enzyme glycyl-transfer RNA (tRNA) synthetase (GlyRS). Despite the broad requirement of GlyRS for protein biosynthesis in all cells, mutations in this gene cause a selective degeneration of peripheral axons, leading to deficits in distal motor function. How mutations in GlyRS (GlyRS(CMT2D)) are linked to motor neuron vulnerability has remained elusive. Here we report that GlyRS(CMT2D) acquires a neomorphic binding activity that directly antagonizes an essential signalling pathway for motor neuron survival. We find that CMT2D mutations alter the conformation of GlyRS, enabling GlyRS(CMT2D) to bind the neuropilin 1 (Nrp1) receptor. This aberrant interaction competitively interferes with the binding of the cognate ligand vascular endothelial growth factor (VEGF) to Nrp1. Genetic reduction of Nrp1 in mice worsens CMT2D symptoms, whereas enhanced expression of VEGF improves motor function. These findings link the selective pathology of CMT2D to the neomorphic binding activity of GlyRS(CMT2D) that antagonizes the VEGF-Nrp1 interaction, and indicate that the VEGF-Nrp1 signalling axis is an actionable target for treating CMT2D.


Subject(s)
Binding, Competitive , Charcot-Marie-Tooth Disease/metabolism , Glycine-tRNA Ligase/metabolism , Animals , Axons/enzymology , Axons/metabolism , Axons/pathology , Cell Line , Cell Survival , Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Female , Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/genetics , Ligands , Male , Mice , Models, Molecular , Motor Neurons/enzymology , Motor Neurons/metabolism , Motor Neurons/pathology , Motor Skills/drug effects , Mutation/genetics , Neuropilin-1/deficiency , Neuropilin-1/genetics , Neuropilin-1/metabolism , Protein Binding , Protein Multimerization , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/therapeutic use
7.
J Membr Biol ; 253(3): 247-256, 2020 06.
Article in English | MEDLINE | ID: mdl-32393995

ABSTRACT

The increasing resistance of many pathogens to most of the common antimicrobials requires the development of new substances with more effective antimicrobial properties. In the present work, we investigated the mechanism of the antimicrobial activity of novel water soluble ammonium quaternary benzanthrone (Compound B) on model membranes, composed of dipalmitoylphosphatidylcholine, 1-palmitoyl-2-oleoylphosphatidylcholine, dipalmitoylphosphatidylglycerol, 1-palmitoyl-2-oleoylphosphatidylglycerol, and dipalmitoylphosphatidylethanolamine (DPPE). The lipids were chosen to represent a model of a bacterial membrane. The changes in surface pressure of the model membranes, before and after the addition of Compound B, were studied by the Langmuir's monolayer method, and the compressional modulus for each monolayer was determined. In addition, the surface morphology of the lipid monolayers before and after injection of Compound B was monitored by Brewster Angle Microscopy. The results showed that Compound B penetrated all the monolayers studied. The most noticeable effects were found with the negatively charged phosphatidylglycerols and with DPPE leading to the conclusion that the electrostatic interactions between the compound and the lipid head groups and the possible formation of hydrogen bonds between the amino group of the ethanolamine and the keto groups in the structure of Compound B are of great importance. In addition, the penetration ability of the benzoquinone with all phospholipids studied was stable even at higher values of the surface pressure, i.e. thicker monolayers, due to the hydrophobic interaction, which plays also an important role for the antimicrobial activity of Compound B.


Subject(s)
Ammonium Compounds , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Benz(a)Anthracenes/chemistry , Benz(a)Anthracenes/pharmacology , Ammonium Compounds/chemistry , Benz(a)Anthracenes/chemical synthesis , Membranes, Artificial , Molecular Structure , Phosphatidylglycerols/chemistry , Phospholipids/chemistry , Solubility , Surface Properties , Water/chemistry
8.
Neurogenetics ; 20(3): 117-127, 2019 08.
Article in English | MEDLINE | ID: mdl-31011849

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a form of inherited peripheral neuropathy that affects motor and sensory neurons. To identify the causative gene in a consanguineous family with autosomal recessive CMT (AR-CMT), we employed a combination of linkage analysis and whole exome sequencing. After excluding known AR-CMT genes, genome-wide linkage analysis mapped the disease locus to a 7.48-Mb interval on chromosome 14q32.11-q32.33, flanked by the markers rs2124843 and rs4983409. Whole exome sequencing identified two non-synonymous variants (p.T40P and p.H915Y) in the AHNAK2 gene that segregated with the disease in the family. Pathogenic predictions indicated that p.T40P is the likely causative allele. Analysis of AHNAK2 expression in the AR-CMT patient fibroblasts showed significantly reduced mRNA and protein levels. AHNAK2 binds directly to periaxin which is encoded by the PRX gene, and PRX mutations are associated with another form of AR-CMT (CMT4F). The altered expression of mutant AHNAK2 may disrupt the AHNAK2-PRX interaction in which one of its known functions is to regulate myelination.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Adolescent , Alleles , Biopsy , Chromosome Mapping , Consanguinity , Family Health , Female , Fibroblasts/metabolism , Genes, Recessive , Genetic Linkage , Genetic Markers , Haplotypes , Humans , Lod Score , Loss of Heterozygosity , Malaysia , Male , Mutation, Missense , Neurons/metabolism , Pedigree , Exome Sequencing
9.
J Neurol Neurosurg Psychiatry ; 90(10): 1171-1179, 2019 10.
Article in English | MEDLINE | ID: mdl-31167812

ABSTRACT

BACKGROUND: Inherited peripheral neuropathies (IPNs) represent a broad group of genetically and clinically heterogeneous disorders, including axonal Charcot-Marie-Tooth type 2 (CMT2) and hereditary motor neuropathy (HMN). Approximately 60%-70% of cases with HMN/CMT2 still remain without a genetic diagnosis. Interestingly, mutations in HMN/CMT2 genes may also be responsible for motor neuron disorders or other neuromuscular diseases, suggesting a broad phenotypic spectrum of clinically and genetically related conditions. Thus, it is of paramount importance to identify novel causative variants in HMN/CMT2 patients to better predict clinical outcome and progression. METHODS: We designed a collaborative study for the identification of variants responsible for HMN/CMT2. We collected 15 HMN/CMT2 families with evidence for autosomal recessive inheritance, who had tested negative for mutations in 94 known IPN genes, who underwent whole-exome sequencing (WES) analyses. Candidate genes identified by WES were sequenced in an additional cohort of 167 familial or sporadic HMN/CMT2 patients using next-generation sequencing (NGS) panel analysis. RESULTS: Bioinformatic analyses led to the identification of novel or very rare variants in genes, which have not been previously associated with HMN/CMT2 (ARHGEF28, KBTBD13, AGRN and GNE); in genes previously associated with HMN/CMT2 but in combination with different clinical phenotypes (VRK1 and PNKP), and in the SIGMAR1 gene, which has been linked to HMN/CMT2 in only a few cases. These findings were further validated by Sanger sequencing, segregation analyses and functional studies. CONCLUSIONS: These results demonstrate the broad spectrum of clinical phenotypes that can be associated with a specific disease gene, as well as the complexity of the pathogenesis of neuromuscular disorders.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Muscular Atrophy, Spinal/genetics , Adult , Aged , Agrin/genetics , Charcot-Marie-Tooth Disease/physiopathology , Computational Biology , DNA Repair Enzymes/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Multienzyme Complexes/genetics , Muscle Proteins/genetics , Muscular Atrophy, Spinal/physiopathology , Pedigree , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, sigma/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Exome Sequencing , Sigma-1 Receptor
10.
Nucleic Acids Res ; 45(13): 8091-8104, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28531329

ABSTRACT

While having multiple aminoacyl-tRNA synthetases implicated in Charcot-Marie-Tooth (CMT) disease suggests a common mechanism, a defect in enzymatic activity is not shared among the CMT-causing mutants. Protein misfolding is a common hypothesis underlying the development of many neurological diseases. Its process usually involves an initial reduction in protein stability and then the subsequent oligomerization and aggregation. Here, we study the structural effect of three CMT-causing mutations in tyrosyl-tRNA synthetase (TyrRS or YARS). Through various approaches, we found that the mutations do not induce changes in protein secondary structures, or shared effects on oligomerization state and stability. However, all mutations provide access to a surface masked in the wild-type enzyme, and that access correlates with protein misinteraction. With recent data on another CMT-linked tRNA synthetase, we suggest that an inherent plasticity, engendering the formation of alternative stable conformations capable of aberrant interactions, links the tRNA synthetase family to CMT.


Subject(s)
Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Tyrosine-tRNA Ligase/chemistry , Tyrosine-tRNA Ligase/metabolism , Amino Acid Substitution , Crystallography, X-Ray , Deuterium Exchange Measurement , Enzyme Stability/genetics , Humans , Kinetics , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Conformation , Protein Folding , Protein Multimerization/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Scattering, Small Angle , Tripartite Motif-Containing Protein 28 , Tyrosine-tRNA Ligase/genetics , X-Ray Diffraction
11.
Am J Hum Genet ; 96(5): 808-15, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25865495

ABSTRACT

GAT-1, encoded by SLC6A1, is one of the major gamma-aminobutyric acid (GABA) transporters in the brain and is responsible for re-uptake of GABA from the synapse. In this study, targeted resequencing of 644 individuals with epileptic encephalopathies led to the identification of six SLC6A1 mutations in seven individuals, all of whom have epilepsy with myoclonic-atonic seizures (MAE). We describe two truncations and four missense alterations, all of which most likely lead to loss of function of GAT-1 and thus reduced GABA re-uptake from the synapse. These individuals share many of the electrophysiological properties of Gat1-deficient mice, including spontaneous spike-wave discharges. Overall, pathogenic mutations occurred in 6/160 individuals with MAE, accounting for ~4% of unsolved MAE cases.


Subject(s)
Epilepsies, Myoclonic/genetics , Epilepsy, Generalized/genetics , GABA Plasma Membrane Transport Proteins/genetics , Animals , Epilepsies, Myoclonic/pathology , Epilepsy, Generalized/pathology , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Mutation
12.
Brain ; 140(4): 868-877, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28007994

ABSTRACT

Recessive mutations in the gene encoding the histidine triad nucleotide binding protein 1 (HINT1) were recently shown to cause a motor-predominant Charcot-Marie-Tooth neuropathy. About 80% of the patients exhibit neuromyotonia, a striking clinical and electrophysiological hallmark that can help to distinguish this disease and to guide diagnostic screening. HINT1 neuropathy has worldwide distribution and is particularly prevalent in populations inhabiting central and south-eastern Europe. With 12 different mutations identified in more than 60 families, it ranks among the most common subtypes of axonal Charcot-Marie-Tooth neuropathy. This article provides an overview of the present knowledge on HINT1 neuropathy with the aim to increase awareness and spur interest among clinicians and researchers in the field. We propose diagnostic guidelines to recognize and differentiate this entity and suggest treatment strategies to manage common symptoms. As a recent player in the field of hereditary neuropathies, the role of HINT1 in peripheral nerves is unknown and the underlying disease mechanisms are unexplored. We provide a comprehensive overview of the structural and functional characteristics of the HINT1 protein that may guide further studies into the molecular aetiology and treatment strategies of this peculiar Charcot-Marie-Tooth subtype.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Isaacs Syndrome/genetics , Myotonia/genetics , Nerve Tissue Proteins/genetics , Peripheral Nervous System Diseases/genetics , Charcot-Marie-Tooth Disease/epidemiology , Charcot-Marie-Tooth Disease/pathology , Hereditary Sensory and Motor Neuropathy/epidemiology , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Isaacs Syndrome/epidemiology , Isaacs Syndrome/pathology , Myotonia/epidemiology , Myotonia/pathology , Peripheral Nervous System Diseases/epidemiology , Peripheral Nervous System Diseases/pathology
13.
Brain ; 140(2): 287-305, 2017 02.
Article in English | MEDLINE | ID: mdl-28137957

ABSTRACT

Hereditary spastic paraplegias are heterogeneous neurodegenerative disorders characterized by progressive spasticity of the lower limbs due to degeneration of the corticospinal motor neurons. In a Bulgarian family with three siblings affected by complicated hereditary spastic paraplegia, we performed whole exome sequencing and homozygosity mapping and identified a homozygous p.Thr512Ile (c.1535C > T) mutation in ATP13A2. Molecular defects in this gene have been causally associated with Kufor-Rakeb syndrome (#606693), an autosomal recessive form of juvenile-onset parkinsonism, and neuronal ceroid lipofuscinosis (#606693), a neurodegenerative disorder characterized by the intracellular accumulation of autofluorescent lipopigments. Further analysis of 795 index cases with hereditary spastic paraplegia and related disorders revealed two additional families carrying truncating biallelic mutations in ATP13A2. ATP13A2 is a lysosomal P5-type transport ATPase, the activity of which critically depends on catalytic autophosphorylation. Our biochemical and immunocytochemical experiments in COS-1 and HeLa cells and patient-derived fibroblasts demonstrated that the hereditary spastic paraplegia-associated mutations, similarly to the ones causing Kufor-Rakeb syndrome and neuronal ceroid lipofuscinosis, cause loss of ATP13A2 function due to transcript or protein instability and abnormal intracellular localization of the mutant proteins, ultimately impairing the lysosomal and mitochondrial function. Moreover, we provide the first biochemical evidence that disease-causing mutations can affect the catalytic autophosphorylation activity of ATP13A2. Our study adds complicated hereditary spastic paraplegia (SPG78) to the clinical continuum of ATP13A2-associated neurological disorders, which are commonly hallmarked by lysosomal and mitochondrial dysfunction. The disease presentation in our patients with hereditary spastic paraplegia was dominated by an adult-onset lower-limb predominant spastic paraparesis. Cognitive impairment was present in most of the cases and ranged from very mild deficits to advanced dementia with fronto-temporal characteristics. Nerve conduction studies revealed involvement of the peripheral motor and sensory nerves. Only one of five patients with hereditary spastic paraplegia showed clinical indication of extrapyramidal involvement in the form of subtle bradykinesia and slight resting tremor. Neuroimaging cranial investigations revealed pronounced vermian and hemispheric cerebellar atrophy. Notably, reduced striatal dopamine was apparent in the brain of one of the patients, who had no clinical signs or symptoms of extrapyramidal involvement.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation/genetics , Proton-Translocating ATPases/genetics , Spastic Paraplegia, Hereditary/genetics , Adult , Animals , Cells, Cultured/cytology , Cells, Cultured/ultrastructure , Chlorocebus aethiops , Cognition Disorders/etiology , Cognition Disorders/genetics , Enzyme Inhibitors/pharmacology , Family Health , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Leupeptins/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Mental Disorders/etiology , Mental Disorders/genetics , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Neuropsychological Tests , Psychiatric Status Rating Scales , Spastic Paraplegia, Hereditary/complications , Spastic Paraplegia, Hereditary/diagnostic imaging
14.
Brain ; 140(8): 2093-2103, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28633435

ABSTRACT

Defects in mRNA export from the nucleus have been linked to various neurodegenerative disorders. We report mutations in the gene MCM3AP, encoding the germinal center associated nuclear protein (GANP), in nine affected individuals from five unrelated families. The variants were associated with severe childhood onset primarily axonal (four families) or demyelinating (one family) Charcot-Marie-Tooth neuropathy. Mild to moderate intellectual disability was present in seven of nine affected individuals. The affected individuals were either compound heterozygous or homozygous for different MCM3AP variants, which were predicted to cause depletion of GANP or affect conserved amino acids with likely importance for its function. Accordingly, fibroblasts of affected individuals from one family demonstrated severe depletion of GANP. GANP has been described to function as an mRNA export factor, and to suppress TDP-43-mediated motor neuron degeneration in flies. Thus our results suggest defective mRNA export from nucleus as a potential pathogenic mechanism of axonal degeneration in these patients. The identification of MCM3AP variants in affected individuals from multiple centres establishes it as a disease gene for childhood-onset recessively inherited Charcot-Marie-Tooth neuropathy with intellectual disability.


Subject(s)
Acetyltransferases/genetics , Charcot-Marie-Tooth Disease/genetics , Genetic Predisposition to Disease/genetics , Intellectual Disability/genetics , Intracellular Signaling Peptides and Proteins/genetics , Acetyltransferases/metabolism , Adolescent , Adult , Cells, Cultured , Charcot-Marie-Tooth Disease/complications , Child , Child, Preschool , Female , Fibroblasts/metabolism , Humans , Intellectual Disability/complications , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mutation , Pedigree , Young Adult
15.
Hum Mutat ; 38(3): 297-309, 2017 03.
Article in English | MEDLINE | ID: mdl-28008748

ABSTRACT

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


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Protein Serine-Threonine Kinases/genetics , White People/genetics , Aged , Alleles , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/epidemiology , Case-Control Studies , Cohort Studies , Enzyme Activation , Female , Frontotemporal Dementia/diagnosis , Frontotemporal Dementia/epidemiology , Genetic Association Studies , Heterozygote , Humans , Male , Middle Aged , Mutation , NF-kappa B/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Sequence Deletion
16.
Am J Hum Genet ; 95(5): 590-601, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25439726

ABSTRACT

Using a combination of exome sequencing and linkage analysis, we investigated an English family with two affected siblings in their 40s with recessive Charcot-Marie Tooth disease type 2 (CMT2). Compound heterozygous mutations in the immunoglobulin-helicase-µ-binding protein 2 (IGHMBP2) gene were identified. Further sequencing revealed a total of 11 CMT2 families with recessively inherited IGHMBP2 gene mutations. IGHMBP2 mutations usually lead to spinal muscular atrophy with respiratory distress type 1 (SMARD1), where most infants die before 1 year of age. The individuals with CMT2 described here, have slowly progressive weakness, wasting and sensory loss, with an axonal neuropathy typical of CMT2, but no significant respiratory compromise. Segregating IGHMBP2 mutations in CMT2 were mainly loss-of-function nonsense in the 5' region of the gene in combination with a truncating frameshift, missense, or homozygous frameshift mutations in the last exon. Mutations in CMT2 were predicted to be less aggressive as compared to those in SMARD1, and fibroblast and lymphoblast studies indicate that the IGHMBP2 protein levels are significantly higher in CMT2 than SMARD1, but lower than controls, suggesting that the clinical phenotype differences are related to the IGHMBP2 protein levels.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Exome/genetics , Models, Molecular , Mutation, Missense/genetics , Phenotype , Adult , Base Sequence , Charcot-Marie-Tooth Disease/pathology , Chromosome Mapping , Female , Haplotypes/genetics , Humans , Molecular Sequence Data , Pedigree , Protein Interaction Mapping , Sequence Analysis, DNA , Sural Nerve/pathology
18.
Ann Neurol ; 80(6): 823-833, 2016 12.
Article in English | MEDLINE | ID: mdl-27686364

ABSTRACT

OBJECTIVE: To identify the unknown genetic cause in a large pedigree previously classified with a distinct form of axonal Charcot-Marie-Tooth disease type 2G (CMT2G) and to explore its transcriptional consequences. METHODS: Clinical reevaluation of the pedigree was performed, followed by linkage analysis with the redefined disease statuses, and whole genome and exome sequencing. The impact of the mutation was investigated by immunoblotting and transcriptome sequencing. RESULTS: Thirteen affected individuals over 3 generations displayed mild and quiescent lower-limb axonal sensorimotor neuropathy. Magnetic resonance imaging (MRI) of lower-limb musculature systematically showed fatty atrophy in clinical and subclinical mutation carriers. We redefined the disease-linked region to chr9q31.3-q34.2 and subsequently identified a novel missense variant in the E3 ubiquitin-protein ligase LRSAM1 (p.Cys694Tyr). Unlike previous reports, we demonstrated in patients' lymphoblasts that the mutation does not influence overall protein levels of LRSAM1, nor of its ubiquitylation target TSG101. The mutation is associated with several transcriptional changes, including a significant upregulation of another E3 ubiquitin-protein ligase, NEDD4L, and of TNFRSF21, a key regulator of axonal degeneration. INTERPRETATION: Our findings demonstrate that the isolated genetic entity CMT2G is caused by a missense mutation in LRSAM1 and should be reclassified as CMT2P. MRI of lower-limb musculature can be used to detect minimal signs of the disease. Transcriptome analysis of patients' cells highlights novel molecular players associated with LRSAM1 dysfunction, and reveals pathways and therapeutic targets shared with amyotrophic lateral sclerosis and Alzheimer disease. Ann Neurol 2016;80:823-833.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Aged, 80 and over , Female , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Mutation, Missense , Nedd4 Ubiquitin Protein Ligases , Neural Conduction/genetics , Neural Conduction/physiology , Pedigree , Up-Regulation
19.
Brain ; 139(Pt 6): 1649-56, 2016 06.
Article in English | MEDLINE | ID: mdl-27009151

ABSTRACT

We performed whole exome sequencing on a patient with Charcot-Marie-Tooth disease type 1 and identified a de novo mutation in PMP2, the gene that encodes the myelin P2 protein. This mutation (p.Ile52Thr) was passed from the proband to his one affected son, and segregates with clinical and electrophysiological evidence of demyelinating neuropathy. We then screened a cohort of 136 European probands with uncharacterized genetic cause of Charcot-Marie-Tooth disease and identified another family with Charcot-Marie-Tooth disease type 1 that has a mutation affecting an adjacent amino acid (p.Thr51Pro), which segregates with disease. Our genetic and clinical findings in these kindred demonstrate that dominant PMP2 mutations cause Charcot-Marie-Tooth disease type 1.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Myelin P2 Protein/genetics , Adolescent , Exome/genetics , Female , Genetic Predisposition to Disease/genetics , Haplotypes , Humans , Male , Middle Aged , Mutation , Neural Conduction/genetics , Pedigree , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL