Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Publication year range
1.
EMBO J ; 39(23): e105364, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33128823

ABSTRACT

Reversible infantile respiratory chain deficiency (RIRCD) is a rare mitochondrial myopathy leading to severe metabolic disturbances in infants, which recover spontaneously after 6-months of age. RIRCD is associated with the homoplasmic m.14674T>C mitochondrial DNA mutation; however, only ~ 1/100 carriers develop the disease. We studied 27 affected and 15 unaffected individuals from 19 families and found additional heterozygous mutations in nuclear genes interacting with mt-tRNAGlu including EARS2 and TRMU in the majority of affected individuals, but not in healthy carriers of m.14674T>C, supporting a digenic inheritance. Our transcriptomic and proteomic analysis of patient muscle suggests a stepwise mechanism where first, the integrated stress response associated with increased FGF21 and GDF15 expression enhances the metabolism modulated by serine biosynthesis, one carbon metabolism, TCA lipid oxidation and amino acid availability, while in the second step mTOR activation leads to increased mitochondrial biogenesis. Our data suggest that the spontaneous recovery in infants with digenic mutations may be modulated by the above described changes. Similar mechanisms may explain the variable penetrance and tissue specificity of other mtDNA mutations and highlight the potential role of amino acids in improving mitochondrial disease.


Subject(s)
Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/metabolism , Adolescent , Cell Line , DNA, Mitochondrial/genetics , Female , Gene Expression , Humans , Infant , Male , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Pedigree , Proteomics , Quadriceps Muscle/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
2.
Brain ; 145(4): 1507-1518, 2022 05 24.
Article in English | MEDLINE | ID: mdl-34791078

ABSTRACT

Consanguineous marriages have a prevalence rate of 24% in Turkey. These carry an increased risk of autosomal recessive genetic conditions, leading to severe disability or premature death, with a significant health and economic burden. A definitive molecular diagnosis could not be achieved in these children previously, as infrastructures and access to sophisticated diagnostic options were limited. We studied the cause of neurogenetic disease in 246 children from 190 consanguineous families recruited in three Turkish hospitals between 2016 and 2020. All patients underwent deep phenotyping and trio whole exome sequencing, and data were integrated in advanced international bioinformatics platforms. We detected causative variants in 119 known disease genes in 72% of families. Due to overlapping phenotypes 52% of the confirmed genetic diagnoses would have been missed on targeted diagnostic gene panels. Likely pathogenic variants in 27 novel genes in 14% of the families increased the diagnostic yield to 86%. Eighty-two per cent of causative variants (141/172) were homozygous, 11 of which were detected in genes previously only associated with autosomal dominant inheritance. Eight families carried two pathogenic variants in different disease genes. De novo (9.3%), X-linked recessive (5.2%) and compound heterozygous (3.5%) variants were less frequent compared to non-consanguineous populations. This cohort provided a unique opportunity to better understand the genetic characteristics of neurogenetic diseases in a consanguineous population. Contrary to what may be expected, causative variants were often not on the longest run of homozygosity and the diagnostic yield was lower in families with the highest degree of consanguinity, due to the high number of homozygous variants in these patients. Pathway analysis highlighted that protein synthesis/degradation defects and metabolic diseases are the most common pathways underlying paediatric neurogenetic disease. In our cohort 164 families (86%) received a diagnosis, enabling prevention of transmission and targeted treatments in 24 patients (10%). We generated an important body of genomic data with lasting impacts on the health and wellbeing of consanguineous families and economic benefit for the healthcare system in Turkey and elsewhere. We demonstrate that an untargeted next generation sequencing approach is far superior to a more targeted gene panel approach, and can be performed without specialized bioinformatics knowledge by clinicians using established pipelines in populations with high rates of consanguinity.


Subject(s)
Exome , Consanguinity , Exome/genetics , Homozygote , Humans , Mutation , Pedigree , Phenotype , Exome Sequencing
3.
Brain ; 144(5): 1422-1434, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33970200

ABSTRACT

Human 4-hydroxyphenylpyruvate dioxygenase-like (HPDL) is a putative iron-containing non-heme oxygenase of unknown specificity and biological significance. We report 25 families containing 34 individuals with neurological disease associated with biallelic HPDL variants. Phenotypes ranged from juvenile-onset pure hereditary spastic paraplegia to infantile-onset spasticity and global developmental delays, sometimes complicated by episodes of neurological and respiratory decompensation. Variants included bona fide pathogenic truncating changes, although most were missense substitutions. Functionality of variants could not be determined directly as the enzymatic specificity of HPDL is unknown; however, when HPDL missense substitutions were introduced into 4-hydroxyphenylpyruvate dioxygenase (HPPD, an HPDL orthologue), they impaired the ability of HPPD to convert 4-hydroxyphenylpyruvate into homogentisate. Moreover, three additional sets of experiments provided evidence for a role of HPDL in the nervous system and further supported its link to neurological disease: (i) HPDL was expressed in the nervous system and expression increased during neural differentiation; (ii) knockdown of zebrafish hpdl led to abnormal motor behaviour, replicating aspects of the human disease; and (iii) HPDL localized to mitochondria, consistent with mitochondrial disease that is often associated with neurological manifestations. Our findings suggest that biallelic HPDL variants cause a syndrome varying from juvenile-onset pure hereditary spastic paraplegia to infantile-onset spastic tetraplegia associated with global developmental delays.


Subject(s)
Oxygenases/genetics , Spastic Paraplegia, Hereditary/genetics , Animals , Female , Humans , Male , Mice , Mutation , Pedigree , Rats , Zebrafish
4.
Hum Mol Genet ; 28(5): 796-803, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30428046

ABSTRACT

Deoxyguanosine kinase (dGK) is an essential rate-limiting component of the mitochondrial purine nucleotide salvage pathway, encoded by the nuclear gene encoding deoxyguanosine kinase (DGUOK). Mutations in DGUOK lead to mitochondrial DNA (mtDNA) depletion typically in the liver and brain, causing a hepatocerebral phenotype. Previous work has shown that in cultured DGUOK patient cells it is possible to rescue mtDNA depletion by increasing substrate amounts for dGK. In this study we developed a mutant dguok zebrafish (Danio rerio) line using CRISPR/Cas9 mediated mutagenesis; dguok-/- fish have significantly reduced mtDNA levels compared with wild-type (wt) fish. When supplemented with only one purine nucleoside (dGuo), mtDNA copy number in both mutant and wt juvenile animals was significantly reduced, contrasting with previous cell culture studies, possibly because of nucleotide pool imbalance. However, in adult dguok-/- fish we detected a significant increase in liver mtDNA copy number when supplemented with both purine nucleosides. This study further supports the idea that nucleoside supplementation has a potential therapeutic benefit in mtDNA depletion syndromes by substrate enhancement of the purine nucleoside salvage pathway and might improve the liver pathology in patients.


Subject(s)
DNA Copy Number Variations , Dietary Supplements , Mitochondria/drug effects , Mitochondria/genetics , Nucleosides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Zebrafish/genetics , Animals , Gene Expression Profiling , Genes, Mitochondrial , Genotype , Humans , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mutation , Nucleosides/metabolism , Phenotype , Zebrafish/metabolism
5.
Am J Hum Genet ; 102(5): 858-873, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727687

ABSTRACT

The exosome is a conserved multi-protein complex that is essential for correct RNA processing. Recessive variants in exosome components EXOSC3, EXOSC8, and RBM7 cause various constellations of pontocerebellar hypoplasia (PCH), spinal muscular atrophy (SMA), and central nervous system demyelination. Here, we report on four unrelated affected individuals with recessive variants in EXOSC9 and the effect of the variants on the function of the RNA exosome in vitro in affected individuals' fibroblasts and skeletal muscle and in vivo in zebrafish. The clinical presentation was severe, early-onset, progressive SMA-like motor neuronopathy, cerebellar atrophy, and in one affected individual, congenital fractures of the long bones. Three affected individuals of different ethnicity carried the homozygous c.41T>C (p.Leu14Pro) variant, whereas one affected individual was compound heterozygous for c.41T>C (p.Leu14Pro) and c.481C>T (p.Arg161∗). We detected reduced EXOSC9 in fibroblasts and skeletal muscle and observed a reduction of the whole multi-subunit exosome complex on blue-native polyacrylamide gel electrophoresis. RNA sequencing of fibroblasts and skeletal muscle detected significant >2-fold changes in genes involved in neuronal development and cerebellar and motor neuron degeneration, demonstrating the widespread effect of the variants. Morpholino oligonucleotide knockdown and CRISPR/Cas9-mediated mutagenesis of exosc9 in zebrafish recapitulated aspects of the human phenotype, as they have in other zebrafish models of exosomal disease. Specifically, portions of the cerebellum and hindbrain were absent, and motor neurons failed to develop and migrate properly. In summary, we show that variants in EXOSC9 result in a neurological syndrome combining cerebellar atrophy and spinal motoneuronopathy, thus expanding the list of human exosomopathies.


Subject(s)
Cerebellum/pathology , Exosome Multienzyme Ribonuclease Complex/genetics , Exosomes/metabolism , Genetic Variation , Motor Neurons/pathology , RNA-Binding Proteins/genetics , Spinal Cord/pathology , Amino Acid Sequence , Animals , Atrophy , Base Sequence , Cerebellum/diagnostic imaging , Child, Preschool , Exosome Multienzyme Ribonuclease Complex/chemistry , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Knockdown Techniques , Haplotypes/genetics , Humans , Infant , Male , Muscle, Skeletal/metabolism , Pedigree , RNA-Binding Proteins/chemistry , Zebrafish
6.
Hum Mol Genet ; 27(12): 2187-2204, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29648643

ABSTRACT

The nuclear-encoded glycyl-tRNA synthetase gene (GARS) is essential for protein translation in both cytoplasm and mitochondria. In contrast, different genes encode the mitochondrial and cytosolic forms of most other tRNA synthetases. Dominant GARS mutations were described in inherited neuropathies, while recessive mutations cause severe childhood-onset disorders affecting skeletal muscle and heart. The downstream events explaining tissue-specific phenotype-genotype relations remained unclear. We investigated the mitochondrial function of GARS in human cell lines and in the GarsC210R mouse model. Human-induced neuronal progenitor cells (iNPCs) carrying dominant and recessive GARS mutations showed alterations of mitochondrial proteins, which were more prominent in iNPCs with dominant, neuropathy-causing mutations. Although comparative proteomic analysis of iNPCs showed significant changes in mitochondrial respiratory chain complex subunits, assembly genes, Krebs cycle enzymes and transport proteins in both recessive and dominant mutations, proteins involved in fatty acid oxidation were only altered by recessive mutations causing mitochondrial cardiomyopathy. In contrast, significant alterations of the vesicle-associated membrane protein-associated protein B (VAPB) and its downstream pathways such as mitochondrial calcium uptake and autophagy were detected in dominant GARS mutations. The role of VAPB has been supported by similar results in the GarsC210R mice. Our data suggest that altered mitochondria-associated endoplasmic reticulum (ER) membranes (MAM) may be important disease mechanisms leading to neuropathy in this condition.


Subject(s)
Endoplasmic Reticulum/genetics , Glycine-tRNA Ligase/genetics , Mitochondria/genetics , Vesicular Transport Proteins/genetics , Animals , Humans , Mice , Mitochondria/metabolism , Mutation , Neurons/metabolism , Neurons/pathology , Signal Transduction , Stem Cells/metabolism
7.
Hum Mol Genet ; 25(14): 2985-2996, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27193168

ABSTRACT

The exosome complex is the most important RNA processing machinery within the cell. Mutations in its subunits EXOSC8 and EXOSC3 cause pontocerebellar hypoplasia, spinal muscular atrophy (SMA) and central nervous system demyelination. We present a patient with SMA-like phenotype carrying a homozygous mutation in RBM7-a subunit of the nuclear exosome targeting (NEXT) complex-which is known to bind and carry specific subtypes of coding and non-coding RNAs to the exosome. The NEXT complex with other protein complexes is responsible for the substrate specificity of the exosome. We performed RNA-sequencing (RNA-seq) analysis on primary fibroblasts of patients with mutations in EXOSC8 and RBM7 and gene knock-down experiments using zebrafish as a model system. RNA-seq analysis identified significantly altered expression of 62 transcripts shared by the two patient cell lines. Knock-down of rbm7, exosc8 and exosc3 in zebrafish showed a common pattern of defects in motor neurons and cerebellum. Our data indicate that impaired RNA metabolism may underlie the clinical phenotype by fine tuning gene expression which is essential for correct neuronal differentiation.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , Muscular Atrophy, Spinal/genetics , RNA-Binding Proteins/genetics , Animals , Cerebellum/metabolism , Cerebellum/pathology , Disease Models, Animal , Exosomes/genetics , Humans , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Mutation , Sequence Analysis, RNA , Zebrafish/metabolism
8.
Hum Mol Genet ; 24(12): 3418-26, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25765662

ABSTRACT

Mutations in the gene encoding glutamine-fructose-6-phosphate transaminase 1 (GFPT1) cause the neuromuscular disorder limb-girdle congenital myasthenic syndrome (LG-CMS). One recurrent GFPT1 mutation detected in LG-CMS patients is a c.*22C>A transversion in the 3'-untranslated region (UTR). Because this variant does not alter the GFPT1 open reading frame, its pathogenic relevance has not yet been established. We found that GFPT1 protein levels were reduced in myoblast cells of the patients carrying this variant. In silico algorithms predicted that the mutation creates a microRNA target site for miR-206*. Investigation of the expression of this so far unrecognized microRNA confirmed that miR-206* (like its counterpart miR-206) is abundant in skeletal muscle. MiR-206* efficiently reduced the expression of reporter constructs containing the mutated 3'-UTR while no such effect was observed with reporter constructs containing the wild-type 3'-UTR or when a specific anti-miR-206* inhibitor was added. Moreover, anti-miR-206* inhibitor treatment substantially rescued GFPT1 expression levels in patient-derived myoblasts. Our data demonstrate that the c.*22C>A mutation in the GFPT1 gene leads to illegitimate binding of microRNA resulting in reduced protein expression. We confirm that c.*22C>A is a causative mutation and suggest that formation of microRNA target sites might be a relevant pathomechanism in Mendelian disorders. Variants in the 3'-UTRs should be considered in genetic diagnostic procedures.


Subject(s)
3' Untranslated Regions , Binding Sites , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , MicroRNAs/genetics , Mutation , Myasthenic Syndromes, Congenital/genetics , RNA, Messenger/genetics , Animals , Base Sequence , Cell Line , Gene Expression , Gene Expression Profiling , Genes, Reporter , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/chemistry , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Humans , MicroRNAs/chemistry , Muscle Cells/metabolism , Myasthenic Syndromes, Congenital/metabolism , RNA Interference , RNA, Messenger/chemistry
10.
Brain ; 139(Pt 8): 2143-53, 2016 08.
Article in English | MEDLINE | ID: mdl-27259756

ABSTRACT

Congenital myasthenic syndromes are a group of rare and genetically heterogenous disorders resulting from defects in the structure and function of the neuromuscular junction. Patients with congenital myasthenic syndrome exhibit fatigable muscle weakness with a variety of accompanying phenotypes depending on the protein affected. A cohort of patients with a clinical diagnosis of congenital myasthenic syndrome that lacked a genetic diagnosis underwent whole exome sequencing in order to identify genetic causation. Missense biallelic mutations in the MYO9A gene, encoding an unconventional myosin, were identified in two unrelated families. Depletion of MYO9A in NSC-34 cells revealed a direct effect of MYO9A on neuronal branching and axon guidance. Morpholino-mediated knockdown of the two MYO9A orthologues in zebrafish, myo9aa/ab, demonstrated a requirement for MYO9A in the formation of the neuromuscular junction during development. The morphants displayed shortened and abnormally branched motor axons, lack of movement within the chorion and abnormal swimming in response to tactile stimulation. We therefore conclude that MYO9A deficiency may affect the presynaptic motor axon, manifesting in congenital myasthenic syndrome. These results highlight the involvement of unconventional myosins in motor axon functionality, as well as the need to look outside traditional neuromuscular junction-specific proteins for further congenital myasthenic syndrome candidate genes.


Subject(s)
Exome , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/physiopathology , Myosins/genetics , Neuromuscular Junction/metabolism , Animals , Cells, Cultured , Child , Child, Preschool , Cohort Studies , Female , Humans , Male , Mice , Mutation, Missense , Pedigree , Zebrafish Proteins
12.
Brain ; 137(Pt 9): 2429-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24951643

ABSTRACT

Congenital myasthenic syndromes are a clinically and genetically heterogeneous group of rare diseases resulting from impaired neuromuscular transmission. Their clinical hallmark is fatigable muscle weakness associated with a decremental muscle response to repetitive nerve stimulation and frequently related to postsynaptic defects. Distal myopathies form another clinically and genetically heterogeneous group of primary muscle disorders where weakness and atrophy are restricted to distal muscles, at least initially. In both congenital myasthenic syndromes and distal myopathies, a significant number of patients remain genetically undiagnosed. Here, we report five patients from three unrelated families with a strikingly homogenous clinical entity combining congenital myasthenia with distal muscle weakness and atrophy reminiscent of a distal myopathy. MRI and neurophysiological studies were compatible with mild myopathy restricted to distal limb muscles, but decrement (up to 72%) in response to 3 Hz repetitive nerve stimulation pointed towards a neuromuscular transmission defect. Post-exercise increment (up to 285%) was observed in the distal limb muscles in all cases suggesting presynaptic congenital myasthenic syndrome. Immunofluorescence and ultrastructural analyses of muscle end-plate regions showed synaptic remodelling with denervation-reinnervation events. We performed whole-exome sequencing in two kinships and Sanger sequencing in one isolated case and identified five new recessive mutations in the gene encoding agrin. This synaptic proteoglycan with critical function at the neuromuscular junction was previously found mutated in more typical forms of congenital myasthenic syndrome. In our patients, we found two missense mutations residing in the N-terminal agrin domain, which reduced acetylcholine receptors clustering activity of agrin in vitro. Our findings expand the spectrum of congenital myasthenic syndromes due to agrin mutations and show an unexpected correlation between the mutated gene and the associated phenotype. This provides a good rationale for examining patients with apparent distal myopathy for a neuromuscular transmission disorder and agrin mutations.


Subject(s)
Agrin/genetics , Muscle Weakness/genetics , Muscular Atrophy/genetics , Myasthenic Syndromes, Congenital/genetics , Adult , Amino Acid Sequence , Atrophy , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Muscle Weakness/complications , Muscle Weakness/pathology , Muscular Atrophy/complications , Muscular Atrophy/pathology , Myasthenic Syndromes, Congenital/complications , Myasthenic Syndromes, Congenital/pathology , Pedigree
13.
Am J Hum Genet ; 88(2): 162-72, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21310273

ABSTRACT

Neuromuscular junctions (NMJs) are synapses that transmit impulses from motor neurons to skeletal muscle fibers leading to muscle contraction. Study of hereditary disorders of neuromuscular transmission, termed congenital myasthenic syndromes (CMS), has helped elucidate fundamental processes influencing development and function of the nerve-muscle synapse. Using genetic linkage, we find 18 different biallelic mutations in the gene encoding glutamine-fructose-6-phosphate transaminase 1 (GFPT1) in 13 unrelated families with an autosomal recessive CMS. Consistent with these data, downregulation of the GFPT1 ortholog gfpt1 in zebrafish embryos altered muscle fiber morphology and impaired neuromuscular junction development. GFPT1 is the key enzyme of the hexosamine pathway yielding the amino sugar UDP-N-acetylglucosamine, an essential substrate for protein glycosylation. Our findings provide further impetus to study the glycobiology of NMJ and synapses in general.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Hexosamines/metabolism , Mutation/genetics , Myasthenic Syndromes, Congenital/genetics , Signal Transduction , Animals , Blotting, Western , Case-Control Studies , Cells, Cultured , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Genetic Linkage , Glycosylation , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Male , Myasthenic Syndromes, Congenital/pathology , Neuromuscular Junction/physiology , Pedigree , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/physiology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Hum Mol Genet ; 20(24): 4879-90, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21926082

ABSTRACT

Fukutin and fukutin-related protein (FKRP) are involved in the glycosylation of α-dystroglycan, a key receptor for basement membrane proteins. Aberrant α-dystroglycan glycosylation leads to a broad spectrum of disorders, ranging from limb girdle muscular dystrophy to Walker-Warburg syndrome. This is the first study investigating a role of fukutin and FKRP-mediated glycosylation in angiogenesis. Transgenic zebrafish expressing enhanced green fluorescent protein in blood vessels were treated with morpholino antisense oligonucleotides that blocked the expression of fukutin, FKRP and dystroglycan. All morphant fish showed muscle damage and vascular abnormalities at day 1 post-fertilization. Intersegmental vessels of somites failed to reach the dorsal longitudinal anastomosis and in more severe phenotypes retracted further or were in some cases even completely missing. In contrast, the eye vasculature was distorted in both fukutin and FKRP morphants, but not in dystroglycan morphants or control fish. The eye size was also smaller in the fukutin and FKRP morphants when compared with dystroglycan knockdown fish and controls. In general, the fukutin morphant fish had the most severe skeletal muscle and eye phenotype. Our findings suggest that fukutin and FKRP have functions that affect ocular development in zebrafish independently of dystroglycan. Despite anecdotal reports about vascular abnormalities in patients affected by dystroglycanopathies, the clinical relevance of such lesions remains unclear and should be subject to further review and investigations.


Subject(s)
Blood Vessels/abnormalities , Blood Vessels/embryology , Glycosyltransferases/deficiency , Zebrafish Proteins/deficiency , Zebrafish/embryology , Animals , Animals, Genetically Modified , Antibodies/immunology , Blood Vessels/drug effects , Blood Vessels/pathology , Dystroglycans/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Eye/blood supply , Eye/drug effects , Eye/pathology , Glycosyltransferases/metabolism , Models, Animal , Morpholinos/pharmacology , Phalloidine/metabolism , Proto-Oncogene Protein c-fli-1 , Somites/abnormalities , Somites/blood supply , Somites/drug effects , Somites/embryology , Staining and Labeling , Zebrafish/genetics , Zebrafish Proteins/metabolism
15.
Hum Mutat ; 33(10): 1474-84, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22678886

ABSTRACT

Congenital myasthenic syndromes (CMSs) are clinically and genetically heterogeneous disorders characterized by a neuromuscular transmission defect. Even though CMSs are genetic disorders, they are highly treatable, and the appropriate drug treatment depends on the underlying genetic defect. This highlights the importance of genetic testing in CMS. In recent years, the molecular basis of CMS has constantly broadened and disease-associated mutations have been identified in 14 genes encoding proteins of the neuromuscular junction. In the dawn of novel sequencing strategies, we report on our 14-year experience in traditional Sanger-based mutation screening of a large cohort of 680 independent patients with suspected CMS. In total, we identified disease-causing mutations in 299 patients (44%) of patients in various known CMS genes, confirming the high degree of genetic heterogeneity associated with the disease. Apart from four known founder mutations, and a few additional recurrent mutations, the majority of variants are private, found in single families. The impact of previously reported genotype-phenotype correlations on efficiency of genetic testing was analyzed in our population. Taking our experiment into account, we present our algorithm for genetic testing in CMS.


Subject(s)
Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/genetics , Cohort Studies , Female , Genetic Association Studies , Genetic Heterogeneity , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism
16.
Hum Mol Genet ; 19(9): 1726-40, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20147321

ABSTRACT

The small signalling adaptor protein Dok-7 has recently been reported as an essential protein of the neuromuscular junction (NMJ). Mutations resulting in partial loss of Dok-7 activity cause a distinct limb-girdle subtype of the inherited NMJ disorder congenital myasthenic syndromes (CMSs), whereas complete loss of Dok-7 results in a lethal phenotype in both mice and humans. Here we describe the zebrafish orthologue of Dok-7 and study its in vivo function. Dok-7 deficiency leads to motility defects in zebrafish embryos and larvae. The relative importance of Dok-7 at different stages of NMJ development varies; it is crucial for the earliest step, the formation of acetylcholine receptor (AChR) clusters in the middle of the muscle fibre prior to motor neuron contact. At later stages, presence of Dok-7 is not absolutely essential, as focal and non-focal synapses do form when Dok-7 expression is downregulated. These contacts however are smaller than in the wild-type zebrafish, reminiscent of the neuromuscular endplate pathology seen in patients with DOK7 mutations. Intriguingly, we also observed changes in slow muscle fibre arrangement; previously, Dok-7 has not been linked to functions other than postsynaptic AChR clustering. Our results suggest an additional role of Dok-7 in muscle. This role seems to be independent of the muscle-specific tyrosine kinase MuSK, the known binding partner of Dok-7 at the NMJ. Our findings in the zebrafish model contribute to a better understanding of the signalling pathways at the NMJ and the pathomechanisms of DOK7 CMSs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myasthenic Syndromes, Congenital/physiopathology , Neuromuscular Junction/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Components , In Situ Hybridization , Molecular Sequence Data , Muscle, Skeletal/innervation , Neuromuscular Junction/physiopathology , Receptors, Cholinergic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Signal Transduction/genetics , Zebrafish , Zebrafish Proteins/genetics
17.
Life Sci Alliance ; 3(8)2020 08.
Article in English | MEDLINE | ID: mdl-32527837

ABSTRACT

The RNA exosome is a ubiquitously expressed complex of nine core proteins (EXOSC1-9) and associated nucleases responsible for RNA processing and degradation. Mutations in EXOSC3, EXOSC8, EXOSC9, and the exosome cofactor RBM7 cause pontocerebellar hypoplasia and motor neuronopathy. We investigated the consequences of exosome mutations on RNA metabolism and cellular survival in zebrafish and human cell models. We observed that levels of mRNAs encoding p53 and ribosome biogenesis factors are increased in zebrafish lines with homozygous mutations of exosc8 or exosc9, respectively. Consistent with higher p53 levels, mutant zebrafish have a reduced head size, smaller brain, and cerebellum caused by an increased number of apoptotic cells during development. Down-regulation of EXOSC8 and EXOSC9 in human cells leads to p53 protein stabilisation and G2/M cell cycle arrest. Increased p53 transcript levels were also observed in muscle samples from patients with EXOSC9 mutations. Our work provides explanation for the pathogenesis of exosome-related disorders and highlights the link between exosome function, ribosome biogenesis, and p53-dependent signalling. We suggest that exosome-related disorders could be classified as ribosomopathies.


Subject(s)
Cerebellar Diseases/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Ribosomes/metabolism , Adult , Animals , Cell Line, Tumor , Cerebellar Diseases/physiopathology , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/genetics , Female , Homozygote , Humans , Male , Mutation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomes/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
18.
Brain ; 131(Pt 3): 747-59, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18180250

ABSTRACT

Congenital myasthenic syndromes (CMS) are clinically and genetically heterogeneous inherited disorders characterized by impaired neuromuscular transmission. Mutations in the acetylcholinesterase (AChE) collagen-like tail subunit gene (COLQ) cause synaptic basal-lamina associated CMS with end-plate AChE deficiency. Here we present the clinical and molecular genetic findings of 22 COLQ-mutant CMS patients, carrying a total of 20 different COLQ mutations, 11 of them had not previously been reported. Typically, patients with esterase deficiency suffer from a severe, progressive weakness with onset at birth or in early infancy. In addition, patients with a late onset showing a mild course of disease are described. AChE inhibitor therapy, beneficial for other forms of CMS, is of no effect in cases of esterase deficiency. The large cohort of COLQ patients studied here enabled us to define additional clinical presentations associated with COLQ mutations that differ from the 'classical' phenotypes: several patients with disease onset at birth or in early infancy presented an unexpected, mild disease course without significant progression of weakness. Moreover, many patients had clinical features reminiscent of limb-girdle CMS with mutations in the recently discovered DOK7 gene, including sparing of eye movements and a predominantly proximal muscle weakness. There was no long-term objective benefit from esterase inhibitors treatment in COLQ patients. Surprisingly, a short-term beneficial effect was observed in four patients and a Tensilon test was positive in two. Treatment with ephedrine was efficient in all five cases where it was administered. The variability of phenotypes caused by COLQ mutations, the divergence from the previously published classical clinical features and an initial positive response to esterase inhibitors in some patients may obscure AChE deficiency as the molecular cause of the disease and delay the start of appropriate therapy. Moreover, overlap with other CMS subtypes and potentially absence of a repetitive compound muscle action potential should be considered in the diagnosis of COLQ-mutated patients.


Subject(s)
Acetylcholinesterase/genetics , Collagen/genetics , Muscle Proteins/genetics , Mutation , Myasthenic Syndromes, Congenital/genetics , Acetylcholinesterase/deficiency , Action Potentials , Adolescent , Age of Onset , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Biopsy , Child , Child, Preschool , Electric Stimulation , Eye Movements , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Muscle, Skeletal/pathology , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/pathology , Myasthenic Syndromes, Congenital/physiopathology , Phenotype , Treatment Outcome
19.
Brain ; 130(Pt 6): 1497-506, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17439981

ABSTRACT

Dok ('downstream-of-kinase') family of cytoplasmic proteins play a role in signalling downstream of receptor and non-receptor phosphotyrosine kinases. Recently, a skeletal muscle receptor tyrosine kinase (MuSK)-interacting cytoplasmic protein termed Dok-7 has been identified. Subsequently, we and others identified mutations in DOK7 as a cause of congenital myasthenic syndromes (CMS), providing evidence for a crucial role of Dok-7 in maintaining synaptic structure. Here we present clinical and molecular genetic data of 14 patients from 12 independent kinships with 13 different mutations in the DOK7 gene. The clinical picture of CMS with DOK7 mutations is highly variable. The age of onset may vary between birth and the third decade. However, most of the patients display a characteristic 'limb-girdle' pattern of weakness with a waddling gait and ptosis, but without ophthalmoparesis. Respiratory problems were frequent. Patients did not benefit from long-term therapy with esterase inhibitors; some of the patients even worsened. DOK7 mutations have emerged as one of the major genetic defects in CMS. The clinical picture differs significantly from CMS caused by mutations in other genes, such as the acetylcholine receptor (AChR) subunit genes. None of the patients with DOK7 mutations had tubular aggregates in the muscle biopsy, implying that 'limb-girdle myasthenia (LGM) with tubular aggregates' previously described in literature may be a pathogenic entity distinct from CMS caused by DOK7 mutations.


Subject(s)
Muscle Proteins/genetics , Mutation , Myasthenic Syndromes, Congenital/genetics , Adolescent , Adult , Biopsy , Child , Child, Preschool , Cholinesterase Inhibitors/adverse effects , Cholinesterase Inhibitors/therapeutic use , DNA Mutational Analysis/methods , Electric Stimulation , Female , Gait Disorders, Neurologic/genetics , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/pathology , Phenotype , Polymorphism, Restriction Fragment Length , Treatment Failure
20.
Neurology ; 90(21): e1842-e1848, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29720545

ABSTRACT

OBJECTIVE: To describe a patient with a multifocal demyelinating motor neuropathy with onset in childhood and a mutation in phosphatase and tensin homolog (PTEN), a tumor suppressor gene associated with inherited tumor susceptibility conditions, macrocephaly, autism, ataxia, tremor, and epilepsy. Functional implications of this protein have been investigated in Parkinson and Alzheimer diseases. METHODS: We performed whole-exome sequencing in the patient's genomic DNA validated by Sanger sequencing. Immunoblotting, in vitro enzymatic assay, and label-free shotgun proteomic profiling were performed in the patient's fibroblasts. RESULTS: The predominant clinical presentation of the patient was a childhood onset, asymmetric progressive multifocal motor neuropathy. In addition, he presented with macrocephaly, autism spectrum disorder, and skin hamartomas, considered as clinical criteria for PTEN-related hamartoma tumor syndrome. Extensive tumor screening did not detect any malignancies. We detected a novel de novo heterozygous c.269T>C, p.(Phe90Ser) PTEN variant, which was absent in both parents. The pathogenicity of the variant is supported by altered expression of several PTEN-associated proteins involved in tumorigenesis. Moreover, fibroblasts showed a defect in catalytic activity of PTEN against the secondary substrate, phosphatidylinositol 3,4-trisphosphate. In support of our findings, focal hypermyelination leading to peripheral neuropathy has been reported in PTEN-deficient mice. CONCLUSION: We describe a novel phenotype, PTEN-associated multifocal demyelinating motor neuropathy with a skin hamartoma syndrome. A similar mechanism may potentially underlie other forms of Charcot-Marie-Tooth disease with involvement of the phosphatidylinositol pathway.


Subject(s)
Hamartoma/genetics , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Sensory and Motor Neuropathy/genetics , PTEN Phosphohydrolase/genetics , Adult , Genetic Predisposition to Disease , Hamartoma/complications , Hereditary Central Nervous System Demyelinating Diseases/complications , Hereditary Sensory and Motor Neuropathy/complications , Humans , Male , Mutation , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL