Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Publication year range
1.
J Hum Genet ; 69(5): 187-196, 2024 May.
Article in English | MEDLINE | ID: mdl-38355957

ABSTRACT

We report the cases of two Spanish pediatric patients with hypotonia, muscle weakness and feeding difficulties at birth. Whole-exome sequencing (WES) uncovered two new homozygous VAMP1 (Vesicle Associated Membrane Protein 1) splicing variants, NM_014231.5:c.129+5 G > A in the boy patient (P1) and c.341-24_341-16delinsAGAAAA in the girl patient (P2). This gene encodes the vesicle-associated membrane protein 1 (VAMP1) that is a component of a protein complex involved in the fusion of synaptic vesicles with the presynaptic membrane. VAMP1 has a highly variable C-terminus generated by alternative splicing that gives rise to three main isoforms (A, B and D), being VAMP1A the only isoform expressed in the nervous system. In order to assess the pathogenicity of these variants, expression experiments of RNA for VAMP1 were carried out. The c.129+5 G > A and c.341-24_341-16delinsAGAAAA variants induced aberrant splicing events resulting in the deletion of exon 2 (r.5_131del; p.Ser2TrpfsTer7) in the three isoforms in the first case, and the retention of the last 14 nucleotides of the 3' of intron 4 (r.340_341ins341-14_341-1; p.Ile114AsnfsTer77) in the VAMP1A isoform in the second case. Pathogenic VAMP1 variants have been associated with autosomal dominant spastic ataxia 1 (SPAX1) and with autosomal recessive presynaptic congenital myasthenic syndrome (CMS). Our patients share the clinical manifestations of CMS patients with two important differences: they do not show the typical electrophysiological pattern that suggests pathology of pre-synaptic neuromuscular junction, and their muscular biopsies present hypertrophic fibers type 1. In conclusion, our data expand both genetic and phenotypic spectrum associated with VAMP1 variants.


Subject(s)
Homozygote , Myasthenic Syndromes, Congenital , Phenotype , Vesicle-Associated Membrane Protein 1 , Female , Humans , Male , Alternative Splicing/genetics , Exome Sequencing , Mutation , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Protein Isoforms/genetics , RNA Splicing/genetics , Vesicle-Associated Membrane Protein 1/genetics , Infant , Child, Preschool
2.
Neurogenetics ; 24(3): 189-200, 2023 07.
Article in English | MEDLINE | ID: mdl-37231228

ABSTRACT

Congenital myasthenic syndromes are inherited disorders caused by mutation in components of the neuromuscular junction and manifest early in life. Mutations in COLQ gene result in congenital myasthenic syndrome. Here, we present the analysis of data from 209 patients from 195 unrelated families highlighting genotype-phenotype correlation. In addition, we describe a COLQ homozygous variant a new patient and discuss it utilizing the Phyre2 and I-TASSER programs. Clinical, molecular genetics, imaging (MRI), and electrodiagnostic (EEG, EMG/NCS) evaluations were performed. Our data showed 89 pathogenic/likely pathogenic variants including 35 missenses, 21 indels, 14 nonsense, 14 splicing, and 5 large deletions variants. Eight common variants were responsible for 48.46% of those. Weakness in proximal muscles, hypotonia, and generalized weakness were detected in all individuals tested. Apart from the weakness, extensive clinical heterogeneity was noted among patients with COLQ-related patients based on their genotypes-those with variants affecting the splice site exhibited more severe clinical features while those with missense variants displayed milder phenotypes, suggesting the role of differential splice variants in multiple functions within the muscle. Analyses and descriptions of these COLQ variants may be helpful in clinical trial readiness and potential development of novel therapies in the setting of established structure-function relationships.


Subject(s)
Myasthenic Syndromes, Congenital , Humans , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Mutation, Missense , Mutation , Genotype , Phenotype , Acetylcholinesterase/genetics , Collagen/genetics , Muscle Proteins/genetics
3.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835142

ABSTRACT

Congenital myasthenic syndromes (CMS) are a heterogeneous group of disorders characterized by impaired neuromuscular signal transmission due to germline pathogenic variants in genes expressed at the neuromuscular junction (NMJ). A total of 35 genes have been reported in CMS (AGRN, ALG14, ALG2, CHAT, CHD8, CHRNA1, CHRNB1, CHRND, CHRNE, CHRNG, COL13A1, COLQ, DOK7, DPAGT1, GFPT1, GMPPB, LAMA5, LAMB2, LRP4, MUSK, MYO9A, PLEC, PREPL, PURA, RAPSN, RPH3A, SCN4A, SLC18A3, SLC25A1, SLC5A7, SNAP25, SYT2, TOR1AIP1, UNC13A, VAMP1). The 35 genes can be classified into 14 groups according to the pathomechanical, clinical, and therapeutic features of CMS patients. Measurement of compound muscle action potentials elicited by repetitive nerve stimulation is required to diagnose CMS. Clinical and electrophysiological features are not sufficient to identify a defective molecule, and genetic studies are always required for accurate diagnosis. From a pharmacological point of view, cholinesterase inhibitors are effective in most groups of CMS, but are contraindicated in some groups of CMS. Similarly, ephedrine, salbutamol (albuterol), amifampridine are effective in most but not all groups of CMS. This review extensively covers pathomechanical and clinical features of CMS by citing 442 relevant articles.


Subject(s)
Myasthenic Syndromes, Congenital , Symporters , Humans , Albuterol , Amifampridine , Cholinesterase Inhibitors , Mitochondrial Proteins/genetics , Mutation , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , NAV1.4 Voltage-Gated Sodium Channel/genetics , Neuromuscular Junction/pathology , Receptors, Cholinergic/genetics , Symporters/genetics , Synaptic Transmission
4.
J Cell Mol Med ; 26(14): 3828-3836, 2022 07.
Article in English | MEDLINE | ID: mdl-35670010

ABSTRACT

Congenital myasthenic syndrome (CMS) encompasses a heterogeneous group of inherited disorders affecting nerve transmission across the neuromuscular junction. The aim of this study was to characterize the clinical, physiological, pathohistological and genetic features of nine unrelated Chinese patients with CMS from a single neuromuscular centre. A total of nine patients aged from neonates to 34 years were enrolled who exhibited initial symptoms. Physical examinations revealed that all patients exhibited muscle weakness. Muscle biopsies demonstrated multiple myopathological changes, including increased fibre size variation, myofibrillar network disarray, necrosis, myofiber grouping, regeneration, fibre atrophy and angular fibres. Genetic testing revealed six different mutated genes, including AGRN (2/9), CHRNE (1/9), GFPT1 (1/9), GMPPB (1/9), PLEC (3/9) and SCN4A (1/9). In addition, patients exhibited differential responses to pharmacological treatment. Prompt utilization of genetic testing will identify novel variants and expand our understanding of the phenotype of this rare syndrome. Our findings contribute to the clinical, pathohistological and genetic spectrum of congenital myasthenic syndrome in China.


Subject(s)
Myasthenic Syndromes, Congenital , Atrophy , Biopsy , Humans , Mutation/genetics , Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , NAV1.4 Voltage-Gated Sodium Channel/genetics , Phenotype , Synaptic Transmission
5.
Hum Mol Genet ; 29(14): 2325-2336, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32543656

ABSTRACT

Congenital myasthenic syndromes (CMS) are characterized by fatigable muscle weakness resulting from impaired neuromuscular transmission. ß2-adrenergic agonists are an effective treatment for DOK7-CMS. DOK7 is a component within the AGRN-LRP4-MUSK-DOK7 signalling pathway that is key for the formation and maintenance of the synaptic structure of the neuromuscular junction (NMJ). The precise mechanism of action of ß2-adrenergic agonists at the NMJ is not fully understood. In this study, we investigated whether ß2-adrenergic agonists improve both neurotransmission and structural integrity of the NMJ in a mouse model of DOK7-CMS. Ex-vivo electrophysiological techniques and microscopy of the NMJ were used to study the effect of salbutamol, a ß2-adrenergic agonist, on synaptic structure and function. DOK7-CMS model mice displayed a severe phenotype with reduced weight gain and perinatal lethality. Salbutamol treatment improved weight gain and survival in DOK7 myasthenic mice. Model animals had fewer active NMJs, detectable by endplate recordings, compared with age-matched wild-type littermates. Salbutamol treatment increased the number of detectable NMJs during endplate recording. Correspondingly, model mice had fewer acetylcholine receptor-stained NMJs detected by fluorescent labelling, but following salbutamol treatment an increased number were detectable. The data demonstrate that salbutamol can prolong survival and increase NMJ number in a severe model of DOK7-CMS.


Subject(s)
Albuterol/pharmacology , Muscle Proteins/genetics , Myasthenic Syndromes, Congenital/drug therapy , Neuromuscular Junction/drug effects , Animals , Disease Models, Animal , Female , Humans , Mice , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Neuromuscular Junction/metabolism , Pregnancy , Receptors, Cholinergic/genetics , Signal Transduction/drug effects , Synaptic Transmission/drug effects
6.
Eur J Neurol ; 29(3): 833-842, 2022 03.
Article in English | MEDLINE | ID: mdl-34749429

ABSTRACT

OBJECTIVES: To present phenotype features of a large cohort of congenital myasthenic syndromes (CMS) and correlate them with their molecular diagnosis. METHODS: Suspected CMS patients were divided into three groups: group A (limb, bulbar or axial weakness, with or without ocular impairment, and all the following: clinical fatigability, electrophysiology compatible with neuromuscular junction involvement and anticholinesterase agents response), group B (limb, bulbar or axial weakness, with or without ocular impairment, and at least one of additional characteristics noted in group A) and group C (pure ocular syndrome). Individual clinical findings and the clinical groups were compared between the group with a confirmed molecular diagnosis of CMS and the group without molecular diagnosis or with a non-CMS molecular diagnosis. RESULTS: Seventy-nine patients (68 families) were included in the cohort: 48 in group A, 23 in group B and 8 in group C. Fifty-one were considered confirmed CMS (30 CHRNE, 5 RAPSN, 4 COL13A1, 3 DOK7, 3 COLQ, 2 GFPT1, 1 CHAT, 1 SCN4A, 1 GMPPB, 1 CHRNA1), 7 probable CMS, 5 non-CMS and 16 unsolved. The chance of a confirmed molecular diagnosis of CMS was significantly higher for group A and lower for group C. Some individual clinical features, alterations on biopsy and electrophysiology enhanced specificity for CMS. Muscle imaging showed at least mild alterations in the majority of confirmed cases, with preferential involvement of soleus, especially in CHRNE CMS. CONCLUSIONS: Stricter clinical criteria increase the chance of confirming a CMS diagnosis, but may lose sensitivity, especially for some specific genes.


Subject(s)
Myasthenic Syndromes, Congenital , Biopsy , Cohort Studies , Humans , Muscle, Skeletal/pathology , Mutation , Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , NAV1.4 Voltage-Gated Sodium Channel/genetics , Phenotype
7.
Neurogenetics ; 22(4): 271-285, 2021 10.
Article in English | MEDLINE | ID: mdl-34333724

ABSTRACT

Twelve patients from seven unrelated South Indian families with a limb-girdle muscular dystrophy-congenital myasthenic syndrome (LGMD/CMS) phenotype and recessive inheritance underwent deep clinical phenotyping, electrophysiological evaluation, muscle histopathology, and next-generation sequencing/Sanger sequencing-based identification of the genetic defect. Homozygosity mapping was performed using high-throughput genome-wide genotyping for mapping the mutation and to evaluate the founder effect. The age of disease onset among patients ranged from childhood to 40 years of age. The key clinical manifestations observed were progressive fatigable limb-girdle weakness, muscle hypertrophy/atrophy, and preferential weakness in a dystrophic pattern. The ages at last follow-up ranged from 30 to 64 years; nine were independently ambulant, two required assistance, and one was wheelchair-bound. Lower limb muscle MRI showed varying degrees of fat replacement in the glutei, hamstrings, anterior leg muscles, and medial gastrocnemius. All patients showed significant decrement on repetitive nerve stimulation (RNS). Muscle biopsy in 7 patients revealed varying degrees of dystrophic and neurogenic changes. Treatment with pyridostigmine and/or salbutamol resulted in variable improvement in 10 patients. Genetic analysis showed an identical homozygous GMPPB mutation c.1000G > A (p.Asp334Asn) in all affected patients. A region of homozygosity (6Mbp) was observed flanking the c.1000G > A change in carrier chromosomes. This study identifies c.1000G > A in GMPPB as a common founder mutation in an ethnic community of South Indian descent with milder yet variable degree of clinical presentation of GMPPB-associated LGMD-CMS.


Subject(s)
Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Nucleotidyltransferases/genetics , Adult , Child , Female , Genetic Testing/methods , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Muscles/pathology , Mutation/genetics , Phenotype
8.
Am J Med Genet A ; 185(3): 827-835, 2021 03.
Article in English | MEDLINE | ID: mdl-33296147

ABSTRACT

CHRNB1 encodes the ß subunit of the acetylcholine receptor (AChR) at the neuromuscular junction. Inherited defects in the neuromuscular junction can lead to congenital myasthenia syndrome (CMS), a clinically and genetically heterogeneous group of disorders which includes fetal akinesia deformation sequence (FADS) on the severe end of the spectrum. Here, we report two unrelated families with biallelic CHRNB1 variants, and in each family, one child presented with lethal FADS. We contrast the diagnostic odysseys in the two families, one of which lasted 16 years while the other, utilizing rapid exome sequencing, led to specific treatment in the first 2 weeks of life. Furthermore, we note that CHRNB1 variants may be under-recognized because in both families, one of the variants is a single exon deletion that has been previously described but may not easily be detected in clinically available genetic testing.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Mutation , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Receptors, Nicotinic/genetics , Adult , Female , Humans , Infant, Newborn , Male , Pedigree , Prognosis , Retrospective Studies
9.
Molecules ; 26(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652901

ABSTRACT

Slow-channel congenital myasthenic syndromes (SCCMSs) are rare genetic diseases caused by mutations in muscle nicotinic acetylcholine receptor (nAChR) subunits. Most of the known SCCMS-associated mutations localize at the transmembrane region near the ion pore. Only two SCCMS point mutations are at the extracellular domains near the acetylcholine binding site, α1(G153S) being one of them. In this work, a combination of molecular dynamics, targeted mutagenesis, fluorescent Ca2+ imaging and patch-clamp electrophysiology has been applied to G153S mutant muscle nAChR to investigate the role of hydrogen bonds formed by Ser 153 with C-loop residues near the acetylcholine-binding site. Introduction of L199T mutation to the C-loop in the vicinity of Ser 153 changed hydrogen bonds distribution, decreased acetylcholine potency (EC50 2607 vs. 146 nM) of the double mutant and decay kinetics of acetylcholine-evoked cytoplasmic Ca2+ rise (τ 14.2 ± 0.3 vs. 34.0 ± 0.4 s). These results shed light on molecular mechanisms of nAChR activation-desensitization and on the involvement of such mechanisms in channelopathy genesis.


Subject(s)
Acetylcholine/genetics , Amino Acid Sequence/genetics , Myasthenic Syndromes, Congenital/genetics , Receptors, Nicotinic/genetics , Acetylcholine/metabolism , Binding Sites/genetics , Calcium/metabolism , Humans , Kinetics , Myasthenic Syndromes, Congenital/pathology , Patch-Clamp Techniques , Point Mutation/genetics , Protein Binding/genetics
10.
Hum Mol Genet ; 27(8): 1434-1446, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29462312

ABSTRACT

Congenital myasthenic syndromes (CMS) are a group of rare, inherited disorders characterized by compromised function of the neuromuscular junction, manifesting with fatigable muscle weakness. Mutations in MYO9A were previously identified as causative for CMS but the precise pathomechanism remained to be characterized. On the basis of the role of MYO9A as an actin-based molecular motor and as a negative regulator of RhoA, we hypothesized that loss of MYO9A may affect the neuronal cytoskeleton, leading to impaired intracellular transport. To investigate this, we used MYO9A-depleted NSC-34 cells (mouse motor neuron-derived cells), revealing altered expression of a number of cytoskeletal proteins important for neuron structure and intracellular transport. On the basis of these findings, the effect on protein transport was determined using a vesicular recycling assay which revealed impaired recycling of a neuronal growth factor receptor. In addition, an unbiased approach utilizing proteomic profiling of the secretome revealed a key role for defective intracellular transport affecting proper protein secretion in the pathophysiology of MYO9A-related CMS. This also led to the identification of agrin as being affected by the defective transport. Zebrafish with reduced MYO9A orthologue expression were treated with an artificial agrin compound, ameliorating defects in neurite extension and improving motility. In summary, loss of MYO9A affects the neuronal cytoskeleton and leads to impaired transport of proteins, including agrin, which may provide a new and unexpected treatment option.


Subject(s)
Agrin/metabolism , Motor Neurons/metabolism , Muscle Weakness/genetics , Myasthenic Syndromes, Congenital/genetics , Myosins/genetics , Nerve Growth Factor/genetics , Neuromuscular Junction/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/genetics , Actins/metabolism , Agrin/genetics , Amides , Animals , Cell Movement , Disease Models, Animal , Embryo, Nonmammalian , Enzyme Inhibitors , Gene Expression Regulation , Humans , Intermediate Filaments/genetics , Intermediate Filaments/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Motor Neurons/ultrastructure , Muscle Weakness/metabolism , Muscle Weakness/pathology , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/pathology , Myosins/deficiency , Nerve Growth Factor/metabolism , Neuromuscular Junction/ultrastructure , Protein Transport , Pyridines , Tubulin/genetics , Tubulin/metabolism , Zebrafish , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
11.
BMC Med Genet ; 21(1): 80, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32295525

ABSTRACT

BACKGROUND: Pierson syndrome (PS) is a rare autosomal recessive disorder, characterized by congenital nephrotic syndrome and microcoria. Advances in renal replacement therapies have extended the lifespan of patients, whereas the full clinical spectrum of PS in infancy and beyond remains elusive. CASE PRESENTATION: We present the case of a 12-month-old boy with PS, manifesting as the bilateral microcoria and congenital nephrotic syndrome. He was born without asphyxia, and was neurologically intact from birth through the neonatal period. Generalized muscle weakness and hypotonia were recognized from 3 months of age. The infant showed recurrent vomiting at age 5 months of age, and was diagnosed with gastroesophageal reflux and intestinal malrotation. Despite the successful surgical treatment, vomiting persisted and led to severely impaired growth. Tulobuterol treatment was effective in reducing the frequency of vomiting. Targeted sequencing confirmed that he had a compound heterozygous mutation in LAMB2 (NM_002292.3: p.Arg550X and p.Glu1507X). A search of the relevant literature identified 19 patients with severe neuro-muscular phenotypes. Among these, only 8 survived the first 12 months of life, and one had feeding difficulty with similar gastrointestinal problems. CONCLUSIONS: This report demonstrated that severe neurological deficits and gastrointestinal dysfunction may emerge in PS patients after the first few months of life.


Subject(s)
Abnormalities, Multiple/genetics , Laminin/genetics , Myasthenic Syndromes, Congenital/genetics , Nephrotic Syndrome/genetics , Pupil Disorders/genetics , Abnormalities, Multiple/pathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Humans , Infant , Male , Mutation , Myasthenic Syndromes, Congenital/pathology , Nephrotic Syndrome/pathology , Phenotype , Pupil Disorders/pathology
12.
J Hum Genet ; 65(4): 355-362, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31959872

ABSTRACT

Null variants in LAMB2 cause Pierson syndrome (PS), a severe congenital nephrotic syndrome with ocular and neurological defects. Patients' kidney specimens show complete negativity for laminin ß2 expression on glomerular basement membrane (GBM). In contrast, missense variants outside the laminin N-terminal (LN) domain in LAMB2 lead to milder phenotypes. However, we experienced cases not showing these typical genotype-phenotype correlations. In this paper, we report six PS patients: four with mild phenotypes and two with severe phenotypes. We conducted molecular studies including protein expression and transcript analyses. The results revealed that three of the four cases with milder phenotypes had missense variants located outside the LN domain and one of the two severe PS cases had a homozygous missense variant located in the LN domain; these variant positions could explain their phenotypes. However, one mild case possessed a splicing site variant (c.3797 + 5G>A) that should be associated with a severe phenotype. Upon transcript analysis, this variant generated some differently sized transcripts, including completely normal transcript, which could have conferred the milder phenotype. In one severe case, we detected the single-nucleotide substitution of c.4616G>A located outside the LN domain, which should be associated with a milder phenotype. However, we detected aberrant splicing caused by the creation of a novel splice site by this single-base substitution. These are novel mechanisms leading to an atypical genotype-phenotype correlation. In addition, all four cases with milder phenotypes showed laminin ß2 expression on GBM. We identified novel mechanisms leading to atypical genotype-phenotype correlation in PS.


Subject(s)
Glomerular Basement Membrane , Laminin , Mutation, Missense , Myasthenic Syndromes, Congenital , Nephrotic Syndrome , Pupil Disorders , RNA Splicing , Amino Acid Substitution , Child , Child, Preschool , Female , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/pathology , Humans , Infant , Laminin/biosynthesis , Laminin/genetics , Male , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/pathology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Protein Domains , Pupil Disorders/genetics , Pupil Disorders/metabolism , Pupil Disorders/pathology
13.
Neuropathol Appl Neurobiol ; 46(6): 588-601, 2020 10.
Article in English | MEDLINE | ID: mdl-32267004

ABSTRACT

AIMS: Congenital myasthenic syndromes (CMS) are characterized by muscle weakness, ptosis and episodic apnoea. Mutations affect integral protein components of the neuromuscular junction (NMJ). Here we searched for the genetic basis of CMS in female monozygotic twins. METHODS: We employed whole-exome sequencing for mutation detection and Sanger sequencing for segregation analysis. Immunohistology was done with antibodies against CHD8, rapsyn, ß-catenin (ßCAT) and golgin on fi-bro-blasts, human and mouse muscle. We recorded superresolution images of the NMJ using 3D-structured illumination microscopy. RESULTS: We discovered a spontaneous missense mutation in CHD8 [chr14:g.21,884,051G>A, GRCh37.p11 | c.1732C>T, NM_00117062 | p.(R578C)], the gene encoding chromodomain helicase DNA-binding protein 8. This is the first missense mutation affecting Duplin, the short 110 kDa isoform of CHD8. It is known that CHD8/Duplin negatively regulates ßCAT signalling in the WNT pathway and plays a role in chromatin remodelling. Inactivating CHD8 mutations are associated with autism spectrum disorder and intellectual disability in combination with facial dysmorphism, overgrowth and macrocephalus. No muscle-specific phenotype has been reported to date. Co-immunostaining with rapsyn on human and mouse muscle revealed a strong presence of CHD8 at the NMJ being located towards the sarcoplasmic side of the rapsyn cluster, where it co-localizes with ßCAT. CONCLUSION: We hypothesize CHD8 to have a role in the maintenance of the structural integrity and function of the NMJ. Both patients benefited from treatment with 3,4-diaminopyridine, a reversible blocker of voltage-gated potassium channels at the nerve terminal that prolongs the action potential and increases acetylcholine release.


Subject(s)
DNA-Binding Proteins/genetics , Mutation, Missense/genetics , Myasthenic Syndromes, Congenital/genetics , Transcription Factors/genetics , Adolescent , Female , Humans , Immunohistochemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myasthenic Syndromes, Congenital/pathology , Neuromuscular Junction/pathology , Twins, Monozygotic , Exome Sequencing
14.
Clin Genet ; 97(4): 634-638, 2020 04.
Article in English | MEDLINE | ID: mdl-31730230

ABSTRACT

We present a case of lethal fetal akinesia deformation sequence (FADS) caused by a frameshift variant in trans with a 148 kbp deletion encompassing 3-36 exons of AGRN. Pathogenic variants in AGRN have been described in families with a form of congenital myasthenic syndrome (CMS), manifesting in the early childhood with variable fatigable muscle weakness. To the best of our knowledge, this is the first case of FADS caused by defects in AGRN gene. FADS has been reported to be caused by pathogenic variants in genes previously associated with CMS including these involved in endplate development and maintenance: MuSK, DOK7, and RAPSN. FADS seems to be the most severe form of CMS. None of the reported in the literature CMS cases associated with AGRN had two null variants, like the case presented herein. This indicates a strong genotype-phenotype correlation.


Subject(s)
Agrin/genetics , Arthrogryposis/genetics , Genes, Lethal/genetics , Myasthenic Syndromes, Congenital/genetics , Adult , Arthrogryposis/diagnostic imaging , Arthrogryposis/pathology , Child , Female , Fetus/diagnostic imaging , Fetus/pathology , Humans , Male , Mutation/genetics , Myasthenic Syndromes, Congenital/pathology , Pedigree , Pregnancy
15.
Am J Med Genet A ; 182(7): 1744-1749, 2020 07.
Article in English | MEDLINE | ID: mdl-32250532

ABSTRACT

Defects in the gene encoding synaptotagmin 2 (SYT2) have been linked to a presynaptic congenital myasthenic syndrome (CMS) and motor neuropathies. However, to date only dominant forms of the disease have been described. We report here a consanguineous patient with a severe recessive form of presynaptic CMS and denervation atrophy caused by the homozygous mutation c.1191delG, p.Arg397Serfs*37 in SYT2. The affected 2-year-old girl had profound weakness and areflexia with moderate bulbar deficit. Repetitive nerve stimulation revealed an extreme reduction of compound muscle action potential amplitudes at rest, with a striking facilitation followed by a progressive decline at fast stimulation rates. These findings were reminiscent, but not identical to those seen in the Lambert-Eaton myasthenic syndrome. 3,4 diaminopyridine and pyridostigmine were effective to ameliorate muscle fatigue, but albuterol was ineffective. Modeling of the mutation using the rat Syt1 C2B x-ray structure revealed that Arg397Serfs*37 disrupts a highly conserved amino acid sequence at the bottom face of the C2B domain not directly involved in calcium binding, but crucial for synaptotagmin-SNARE interaction and exocytosis. Thus, this report describes a recessive form of synaptotagmin 2-CMS and highlights the importance of the synaptotagmin C-terminal on synaptic vesicle fusion and exocytosis.


Subject(s)
Genetic Predisposition to Disease , Myasthenic Syndromes, Congenital/genetics , Synaptotagmin II/genetics , Amino Acid Sequence/genetics , Child, Preschool , Female , Genes, Recessive/genetics , Humans , Mutation , Myasthenic Syndromes, Congenital/pathology
16.
Am J Med Genet A ; 182(10): 2272-2283, 2020 10.
Article in English | MEDLINE | ID: mdl-32776697

ABSTRACT

Synaptotagmins are integral synaptic vesicle membrane proteins that function as calcium sensors and regulate neurotransmitter release at the presynaptic nerve terminal. Synaptotagmin-2 (SYT2), is the major isoform expressed at the neuromuscular junction. Recently, dominant missense variants in SYT2 have been reported as a rare cause of distal motor neuropathy and myasthenic syndrome, manifesting with stable or slowly progressive distal weakness of variable severity along with presynaptic NMJ impairment. These variants are thought to have a dominant-negative effect on synaptic vesicle exocytosis, although the precise pathomechanism remains to be elucidated. Here we report seven patients of five families, with biallelic loss of function variants in SYT2, clinically manifesting with a remarkably consistent phenotype of severe congenital onset hypotonia and weakness, with variable degrees of respiratory involvement. Electrodiagnostic findings were consistent with a presynaptic congenital myasthenic syndrome (CMS) in some. Treatment with an acetylcholinesterase inhibitor pursued in three patients showed clinical improvement with increased strength and function. This series further establishes SYT2 as a CMS-disease gene and expands its clinical and genetic spectrum to include recessive loss-of-function variants, manifesting as a severe congenital onset presynaptic CMS with potential treatment implications.


Subject(s)
Genetic Predisposition to Disease , Muscle Hypotonia/genetics , Myasthenic Syndromes, Congenital/genetics , Synaptotagmin II/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Male , Muscle Hypotonia/complications , Muscle Hypotonia/pathology , Muscle Weakness/genetics , Muscle Weakness/pathology , Mutation, Missense/genetics , Myasthenic Syndromes, Congenital/complications , Myasthenic Syndromes, Congenital/pathology , Pedigree , Phenotype , Synaptic Transmission/genetics
17.
BMC Nephrol ; 21(1): 363, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32838745

ABSTRACT

BACKGROUND: Congenital nephrotic syndrome (CNS) and infantile nephrotic syndrome (INS) cause substantial morbidity and mortality. In Japan, there is a lack of knowledge regarding the characteristics of CNS and INS. This study aimed to clarify the characteristics of CNS and INS in Japan. METHODS: This cross-sectional nationwide survey obtained data from 44 institutions in Japan managing 92 patients with CNS or INS, by means of two survey questionnaires sent by postal mail. Patients aged < 16 years by 1 April 2015, with a diagnosis of CNS or INS, were included in this study. The primary outcome was end-stage kidney disease. RESULTS: A total of 83 patients with CNS or INS were analyzed. The most frequent disease type was non-Finnish (60.2%); 33 patients (39.8%) had Finnish type. Among those with non-Finnish-type disease, 26 had no syndrome and 24 had a syndrome, of which the most frequent was Denys-Drash syndrome (70.8%). Patients with non-Finnish-type disease with syndrome showed the earliest progression to end-stage kidney disease compared with the other two groups, whereas patients with non-Finnish-type disease without syndrome progressed more slowly compared with the other two groups. In the Finnish-type group, the disease was diagnosed the earliest; a large placenta was reported more frequently; genetic testing was more frequently performed (93.8%); mental retardation was the most frequent extra-renal symptom (21.2%); and thrombosis and infection were more frequent compared with the other groups. Patients with non-Finnish-type disease with syndrome had a higher frequency of positive extra-renal symptoms (79.2%), the most common being urogenital symptoms (54.2%). Treatment with steroids and immunosuppressants was more frequent among patients with non-Finnish-type disease without syndrome. Two patients with non-Finnish-type disease without syndrome achieved complete remission. In all groups, unilateral nephrectomy was performed more often than bilateral nephrectomy and peritoneal dialysis was the most common renal replacement therapy. CONCLUSIONS: The present epidemiological survey sheds light on the characteristics of children with CNS and INS in Japan. A high proportion of patients underwent genetic examination, and patient management was in accord with current treatment recommendations and practices. TRIAL REGISTRATION: Not applicable.


Subject(s)
Intellectual Disability/physiopathology , Kidney Failure, Chronic/physiopathology , Nephrotic Syndrome/physiopathology , Adolescent , Child , Child, Preschool , Denys-Drash Syndrome/pathology , Denys-Drash Syndrome/physiopathology , Disease Progression , Female , Genetic Testing , Glucocorticoids/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Infant , Infant, Newborn , Japan , Kaplan-Meier Estimate , Kidney Failure, Chronic/therapy , Male , Myasthenic Syndromes, Congenital/pathology , Myasthenic Syndromes, Congenital/physiopathology , Nephrectomy , Nephrotic Syndrome/congenital , Nephrotic Syndrome/pathology , Nephrotic Syndrome/therapy , Organ Size , Placenta/pathology , Pregnancy , Pupil Disorders/pathology , Pupil Disorders/physiopathology , Renal Replacement Therapy , Surveys and Questionnaires , Syndrome
18.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(5): 551-554, 2020 May 10.
Article in Zh | MEDLINE | ID: mdl-32335884

ABSTRACT

OBJECTIVE: To explore the genetic basis for a female patient featuring unstable head upright and hypotonia of limbs. METHODS: The child was examined clinically. Peripheral blood samples of the child, her parents and siblings were collected. Genomic DNA was extracted and subjected to next generation sequencing (NGS). Suspected variant was verified by Sanger sequencing and bioinformatic analysis. RESULTS: DNA sequencing found that the patient has carried a de novo heterozygous c.354C>A (p.N118K) variant of the CHRND gene, which was not found in her parents and sibling. Bioinformatics analysis predicted that the variant was likely to be pathogenic. Literature review suggested that the phenotype of the patient was very similar to previously reported ones. CONCLUSION: The child was diagnosed with slow-channel congenital myasthenic syndrome (SCCMS) type 3A caused by heterozygous variant of the CHRND gene. NGS has provided a powerful tool for the diagnosis of such disorders.


Subject(s)
Myasthenic Syndromes, Congenital , Receptors, Cholinergic , Child , Female , Genetic Testing , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Mutation , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Receptors, Cholinergic/genetics
19.
Curr Opin Neurol ; 32(5): 764-770, 2019 10.
Article in English | MEDLINE | ID: mdl-31306211

ABSTRACT

PURPOSE OF REVIEW: Amyotrophic lateral sclerosis (ALS) is a degenerative motor neuron disease with a strong neuroinflammatory component. This review summarizes how the connection between neurodegeneration and the immune system is strengthened by new discoveries from ALS genetics and the analysis of subpopulations of immune cells in ALS. RECENT FINDINGS: Recent genes identified in ALS encode for proteins with direct immune roles, which when mutated lead to deregulation of immune functions, potentially influencing the disease. Although neuroinflammation in the central nervous system (CNS) of ALS patients has been well documented, new evidence suggests also direct malfunctions of immune cells in the CNS and at the periphery. Although CD4+ T-regulatory lymphocytes are protective in ALS, their number and function are altered over the disease course. CD8+ T cells are detrimental for motor neurons in the CNS but show some protective roles at the periphery. Similarly, the presence of mast cells in muscles of ALS models and patients and impairments of monocyte functions reveal potential new players in ALS disease progression. SUMMARY: Although motor neuron degeneration is considered the prime event in ALS, dysfunctions in immune processes can impact the disease, highlighting that targeting specific immune components is a strategy for developing biomarkers and ultimately new drugs.


Subject(s)
Myasthenic Syndromes, Congenital , Animals , Humans , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/pathology , Myasthenic Syndromes, Congenital/physiopathology
20.
Muscle Nerve ; 60(6): 790-800, 2019 12.
Article in English | MEDLINE | ID: mdl-31531871

ABSTRACT

INTRODUCTION: Reduced expression of the vesicular acetylcholine transporter (VAChT) leads to changes in the distribution and shape of synaptic vesicles (SVs) at neuromuscular junctions (NMJs), suggesting vesicular acetylcholine (ACh) as a key component of synaptic structure and function. It is poorly understood how long-term changes in cholinergic transmission contribute to age- and disease-related degeneration in the motor system. METHODS: In this study we performed confocal imaging, electrophysiology, electron microscopy, and analyses of respiratory mechanics of the diaphragm NMJ components in 12-month-old wild-type (WT) and VAChTKDHOM mice. RESULTS: Diaphragms of NMJs of the VAChTKDHOM mice were similar to those in WT mice in number, colocalization, and fragmentation of pre-/postsynaptic components. However, they had increased spontaneous SV exocytosis, miniature endplate potential frequency, and diminished MEPP amplitude. No impairment in respiratory mechanics at rest was observed, probably due to the large neurotransmission safety factor of the diaphragm. DISCUSSION: The present findings help us to understand the consequences of reduced ACh release at the NMJs during aging.


Subject(s)
Aging/pathology , Diaphragm/ultrastructure , Myasthenic Syndromes, Congenital/pathology , Neuromuscular Junction/ultrastructure , Synaptic Vesicles/ultrastructure , Acetylcholine/metabolism , Aging/metabolism , Animals , Diaphragm/metabolism , Diaphragm/physiopathology , Disease Models, Animal , Endocytosis , Excitatory Postsynaptic Potentials/physiology , Exocytosis , Gene Knockdown Techniques , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Motor Endplate , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/physiopathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiopathology , Respiratory Mechanics/physiology , Synaptic Transmission , Synaptic Vesicles/metabolism , Vesicular Acetylcholine Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL