Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Brain ; 146(5): 1831-1843, 2023 05 02.
Article in English | MEDLINE | ID: mdl-36227727

ABSTRACT

Instability of simple DNA repeats has been known as a common cause of hereditary ataxias for over 20 years. Routine genetic diagnostics of these phenotypically similar diseases still rely on an iterative workflow for quantification of repeat units by PCR-based methods of limited precision. We established and validated clinical nanopore Cas9-targeted sequencing, an amplification-free method for simultaneous analysis of 10 repeat loci associated with clinically overlapping hereditary ataxias. The method combines target enrichment by CRISPR-Cas9, Oxford Nanopore long-read sequencing and a bioinformatics pipeline using the tools STRique and Megalodon for parallel detection of length, sequence, methylation and composition of the repeat loci. Clinical nanopore Cas9-targeted sequencing allowed for the precise and parallel analysis of 10 repeat loci associated with adult-onset ataxia and revealed additional parameter such as FMR1 promotor methylation and repeat sequence required for diagnosis at the same time. Using clinical nanopore Cas9-targeted sequencing we analysed 100 clinical samples of undiagnosed ataxia patients and identified causative repeat expansions in 28 patients. Parallel repeat analysis enabled a molecular diagnosis of ataxias independent of preconceptions on the basis of clinical presentation. Biallelic expansions within RFC1 were identified as the most frequent cause of ataxia. We characterized the RFC1 repeat composition of all patients and identified a novel repeat motif, AGGGG. Our results highlight the power of clinical nanopore Cas9-targeted sequencing as a readily expandable workflow for the in-depth analysis and diagnosis of phenotypically overlapping repeat expansion disorders.


Subject(s)
Cerebellar Ataxia , Spinocerebellar Degenerations , Adult , Humans , Ataxia/genetics , Cerebellar Ataxia/genetics , Computational Biology , High-Throughput Nucleotide Sequencing , Fragile X Mental Retardation Protein
2.
Brain ; 146(4): 1388-1402, 2023 04 19.
Article in English | MEDLINE | ID: mdl-36100962

ABSTRACT

Genetic diagnosis of facioscapulohumeral muscular dystrophy (FSHD) remains a challenge in clinical practice as it cannot be detected by standard sequencing methods despite being the third most common muscular dystrophy. The conventional diagnostic strategy addresses the known genetic parameters of FSHD: the required presence of a permissive haplotype, a size reduction of the D4Z4 repeat of chromosome 4q35 (defining FSHD1) or a pathogenic variant in an epigenetic suppressor gene (consistent with FSHD2). Incomplete penetrance and epistatic effects of the underlying genetic parameters as well as epigenetic parameters (D4Z4 methylation) pose challenges to diagnostic accuracy and hinder prediction of clinical severity. In order to circumvent the known limitations of conventional diagnostics and to complement genetic parameters with epigenetic ones, we developed and validated a multistage diagnostic workflow that consists of a haplotype analysis and a high-throughput methylation profile analysis (FSHD-MPA). FSHD-MPA determines the average global methylation level of the D4Z4 repeat array as well as the regional methylation of the most distal repeat unit by combining bisulphite conversion with next-generation sequencing and a bioinformatics pipeline and uses these as diagnostic parameters. We applied the diagnostic workflow to a cohort of 148 patients and compared the epigenetic parameters based on FSHD-MPA to genetic parameters of conventional genetic testing. In addition, we studied the correlation of repeat length and methylation level within the most distal repeat unit with age-corrected clinical severity and age at disease onset in FSHD patients. The results of our study show that FSHD-MPA is a powerful tool to accurately determine the epigenetic parameters of FSHD, allowing discrimination between FSHD patients and healthy individuals, while simultaneously distinguishing FSHD1 and FSHD2. The strong correlation between methylation level and clinical severity indicates that the methylation level determined by FSHD-MPA accounts for differences in disease severity among individuals with similar genetic parameters. Thus, our findings further confirm that epigenetic parameters rather than genetic parameters represent FSHD disease status and may serve as a valuable biomarker for disease status.


Subject(s)
Muscular Dystrophy, Facioscapulohumeral , Humans , Muscular Dystrophy, Facioscapulohumeral/diagnosis , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , DNA Methylation/genetics , Haplotypes , Chromosomes, Human, Pair 4/genetics
3.
J Med Genet ; 59(7): 697-705, 2022 07.
Article in English | MEDLINE | ID: mdl-34321323

ABSTRACT

BACKGROUND: O'Donnell-Luria-Rodan syndrome (ODLURO) is an autosomal-dominant neurodevelopmental disorder caused by pathogenic, mostly truncating variants in KMT2E. It was first described by O'Donnell-Luria et al in 2019 in a cohort of 38 patients. Clinical features encompass macrocephaly, mild intellectual disability (ID), autism spectrum disorder (ASD) susceptibility and seizure susceptibility. METHODS: Affected individuals were ascertained at paediatric and genetic centres in various countries by diagnostic chromosome microarray or exome/genome sequencing. Patients were collected into a case cohort and were systematically phenotyped where possible. RESULTS: We report 18 additional patients from 17 families with genetically confirmed ODLURO. We identified 15 different heterozygous likely pathogenic or pathogenic sequence variants (14 novel) and two partial microdeletions of KMT2E. We confirm and refine the phenotypic spectrum of the KMT2E-related neurodevelopmental disorder, especially concerning cognitive development, with rather mild ID and macrocephaly with subtle facial features in most patients. We observe a high prevalence of ASD in our cohort (41%), while seizures are present in only two patients. We extend the phenotypic spectrum by sleep disturbances. CONCLUSION: Our study, bringing the total of known patients with ODLURO to more than 60 within 2 years of the first publication, suggests an unexpectedly high relative frequency of this syndrome worldwide. It seems likely that ODLURO, although just recently described, is among the more common single-gene aetiologies of neurodevelopmental delay and ASD. We present the second systematic case series of patients with ODLURO, further refining the mutational and phenotypic spectrum of this not-so-rare syndrome.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Megalencephaly , Neurodevelopmental Disorders , Autism Spectrum Disorder/genetics , Child , Humans , Intellectual Disability/diagnosis , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Seizures/epidemiology , Seizures/genetics , Syndrome , Exome Sequencing
4.
Hum Mutat ; 43(4): 477-486, 2022 04.
Article in English | MEDLINE | ID: mdl-35112411

ABSTRACT

The synthesis of cytochrome c oxidase 2 (SCO2 ) gene encodes for a mitochondrial located metallochaperone essential for the synthesis of the cytochrome c oxidase (COX) subunit 2. Recessive mutations in SCO2 have been reported in several cases with fatal infantile cardioencephalomyopathy with COX deficiency and in only four cases with axonal neuropathy. Here, we identified a homozygous pathogenic variant (c.361G > C; p.[Gly121Arg]) in SCO2 in two brothers with isolated axonal motor neuropathy. To address pathogenicity of the amino acid substitution, biochemical studies were performed and revealed increased level of the mutant SCO2 -protein and dysregulation of COX subunits in leukocytes and moreover unraveled decrease of proteins involved in the manifestation of neuropathies. Hence, our combined data strengthen the concept of SCO2 being causative for a very rare form of axonal neuropathy, expand its molecular genetic spectrum and provide first biochemical insights into the underlying pathophysiology.


Subject(s)
Charcot-Marie-Tooth Disease , Carrier Proteins/genetics , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones/genetics , Mutation , Oxidoreductases/genetics , Oxidoreductases/metabolism , Siblings
5.
Pract Neurol ; 22(6): 518-520, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35907633

ABSTRACT

A man in his 20s gave a 9-year history of recurrent muscle pain and weakness, occurring mostly after exercise, and lasting for up to 2 days. There had been one episode of severe rhabdomyolysis after cold exposure. He also had longstanding hypokalaemia, which was key to his correct diagnosis but was not followed. This case highlights the importance of an appropriately methodical investigation of weak hypokalaemic patients, and the relevance of hypokalaemia as a cause of neuromuscular symptoms not related to muscular channelopathies.


Subject(s)
Hypokalemia , Rhabdomyolysis , Male , Humans , Hypokalemia/complications , Muscle Weakness/etiology , Rhabdomyolysis/etiology , Paresis
6.
Neuropediatrics ; 52(2): 123-125, 2021 04.
Article in English | MEDLINE | ID: mdl-33086386

ABSTRACT

Episodic encephalopathy due to mutations in the thiamine pyrophosphokinase 1 (TPK1) gene is a rare autosomal recessive metabolic disorder. Patients reported so far have onset in early childhood of acute encephalopathic episodes, which result in a progressive neurologic dysfunction including ataxia, dystonia, and spasticity. Here, we report the case of an infant with TPK1 deficiency (compound heterozygosity for two previously described pathogenic variants) presenting with two encephalopathic episodes and clinical stabilization under oral thiamine and biotin supplementation. In contrast to other reported cases, our patient showed an almost normal psychomotor development, which might be due to an early diagnosis and subsequent therapy.


Subject(s)
Brain Diseases, Metabolic, Inborn/diet therapy , Thiamin Pyrophosphokinase/deficiency , Thiamine/pharmacology , Vitamin B Complex/pharmacology , Biotin/administration & dosage , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/physiopathology , Dietary Supplements , Humans , Infant , Thiamine/administration & dosage , Vitamin B Complex/administration & dosage
7.
Neuropediatrics ; 52(2): 92-97, 2021 04.
Article in English | MEDLINE | ID: mdl-33086385

ABSTRACT

INTRODUCTION: Next generation sequencing (NGS) with customized gene panels is a helpful tool to identify monogenic epilepsy syndromes. The number of genes tested within a customized panel may vary greatly. The aim of the present study was to compare the diagnostic yield of small (<25 kb) and large (>25 kb) customized epilepsy panels. METHODS: This retrospective cohort study investigated data of 190 patients of 18 years or younger, with the diagnosis of an epilepsy of unknown etiology who underwent NGS using customized gene panels. Small (<25 kb) and large (>25 kb) panels were compared regarding the distribution of benign/likely benign and pathogenic/likely pathogenic variants and variants of unclear significance. In addition, differences of the diagnostic yield with respect to epilepsy severity, i.e., developmental and epileptic encephalopathy [DEE] vs. non-DEE, were analyzed. RESULTS: The diagnostic yield defined as pathogenic or likely pathogenic variants in large panels was significantly increased (29% [n = 14/48] vs. 13% [n = 18/142], p = 0.0198) compared with smaller panels. In non-DEE patients the increase of the diagnostic yield in large panels was significant(35% n = 6/17 vs. 13% n = 12/94, p = 0.0378), which was not true for DEE patients. DISCUSSION: This study indicates that large panels are superior for pediatric patients with epilepsy forms without encephalopathy (non-DEE). For patients suffering from DEE small panels of a maximum of 10 genes seem to be sufficient. The proportion of unclear findings increases with rising panel sizes. CONCLUSION: Customized epilepsy panels of >25 kb compared with smaller panels show a significant higher diagnostic yield in patients with epilepsy especially in non-DEE patients.


Subject(s)
Epileptic Syndromes/diagnosis , Epileptic Syndromes/genetics , High-Throughput Nucleotide Sequencing/standards , Adolescent , Child , Child, Preschool , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Male , Retrospective Studies
8.
Hum Mutat ; 41(5): 1025-1032, 2020 05.
Article in English | MEDLINE | ID: mdl-32048431

ABSTRACT

As comprehensive sequencing technologies gain widespread use, questions about so-called secondary findings (SF) require urgent consideration. The American College of Medical Genetics and Genomics has recommended to report SF in 59 genes (ACMG SF v2.0) including four actionable genes associated with inherited primary arrhythmia syndromes (IPAS) such as catecholaminergic polymorphic ventricular tachycardia, long QT syndrome, and Brugada syndrome. Databases provide conflicting results for the purpose of identifying pathogenic variants in SF associated with IPAS at a level of sufficient evidence for clinical return. As IPAS account for a significant proportion of sudden cardiac deaths (SCD) in young and apparently healthy individuals, variant interpretation has a great impact on diagnosis and prevention of disease. Of 6381 individuals, 0.4% carry pathogenic variants in one of the four actionable genes related to IPAS: RYR2, KCNQ1, KCNH2, and SCN5A. Comparison of the databases ClinVar, Leiden Open-source Variant Database, and Human Gene Mutation Database showed impactful differences (0.2% to 1.3%) in variant interpretation improvable by expert-curation depending on database and classification system used. These data further highlight the need for international consensus regarding the variant interpretation, and subsequently management of SF in particular with regard to treatable arrhythmic disorders with increased risk of SCD.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Alleles , Databases, Genetic , Female , Genetic Association Studies/methods , Genetic Testing , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Male , Phenotype , Syndrome
9.
J Peripher Nerv Syst ; 25(2): 112-116, 2020 06.
Article in English | MEDLINE | ID: mdl-32077159

ABSTRACT

PHARC syndrome is a rare neurodegenerative disorder caused by mutations in the ABHD12 gene. It is a genetically heterogeneous and clinically variable disease, which is characterized by demyelinating polyneuropathy, hearing loss, cerebellar ataxia, retinitis pigmentosa, and early-onset cataract and can easily be misdiagnosed as other neurologic disorders with a similar clinical picture, such as Charcot-Marie-Tooth disease and Refsum disease. We describe the genotype-phenotype correlation of two siblings with a novel genotype underlying PHARC syndrome. The genotype was identified using next-generation sequencing. We examined both patients by means of thorough history taking and clinical examination, nerve conduction studies (NCS), brain imaging, and optical coherence tomography to establish a genotype-phenotype correlation. We identified a novel homozygous point mutation (c.784C > T, p.Arg262*) in the ABHD12 gene. This mutation was detected in both siblings, who had bilateral hearing loss and cataracts, signs of cerebellar ataxia, and neuropathy with a primarily demyelinating pattern in NCS. In one case, retinitis pigmentosa was also evident. As PHARC syndrome is a rare autosomal recessive disorder, our findings highlight the importance of an interdisciplinary diagnostic workup with clinical and molecular genetic testing to avoid a misdiagnosis as Charcot-Marie-Tooth disease or Refsum disease.


Subject(s)
Ataxia/genetics , Ataxia/physiopathology , Cataract/genetics , Cataract/physiopathology , Monoacylglycerol Lipases/genetics , Polyneuropathies/genetics , Polyneuropathies/physiopathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/physiopathology , Adult , Ataxia/diagnosis , Cataract/diagnosis , Genetic Association Studies , Humans , Male , Polyneuropathies/diagnosis , Retinitis Pigmentosa/diagnosis , Siblings , Young Adult
10.
Neuropediatrics ; 51(1): 49-52, 2020 02.
Article in English | MEDLINE | ID: mdl-31541998

ABSTRACT

Nicolaides-Baraitser syndrome (NCBRS) is a rare disease caused by a mutation in the SMARCA2 gene. Clinical features include craniofacial dysmorphia and abnormalities of the limbs, as well as intellectual disorder and often epilepsy. Hepatotoxicity is a rare complication of the therapy with valproic acid (VPA) and a mutation of the polymerase γ (POLG) might lead to a higher sensitivity for liver hepatotoxicity. We present a patient with the coincidence of two rare diseases, the NCBRS and additionally a POLG1 mutation in combination with a liver hepatotoxicity. The co-occurrence in children for two different genetic diseases is discussed with the help of literature review.


Subject(s)
Anticonvulsants/adverse effects , Chemical and Drug Induced Liver Injury , DNA Polymerase gamma/genetics , Facies , Foot Deformities, Congenital , Hypotrichosis , Intellectual Disability , Transcription Factors/genetics , Valproic Acid/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Child , Female , Foot Deformities, Congenital/drug therapy , Foot Deformities, Congenital/genetics , Humans , Hypotrichosis/drug therapy , Hypotrichosis/genetics , Intellectual Disability/drug therapy , Intellectual Disability/genetics
11.
Mol Cell Probes ; 44: 14-20, 2019 04.
Article in English | MEDLINE | ID: mdl-30682426

ABSTRACT

The heterooctameric mitochondrial trifunctional protein (MTP), composed of four α- and ß-subunits harbours three enzymes that each perform a different function in mitochondrial fatty acid ß-oxidation. Pathogenic variants in the MTP genes (HADHA and HADHB) cause MTP deficiency, a rare autosomal recessive metabolic disorder characterized by phenotypic heterogeneity ranging from severe, early-onset, cardiac disease to milder, later-onset, myopathy and neuropathy. Since metabolic myopathies and neuropathies are a group of rare genetic disorders and their associated muscle symptoms may be subtle, the diagnosis is often delayed. Here we evaluated data of 161 patients with myopathy and 242 patients with neuropathy via next generation sequencing (NGS) and report the diagnostic yield in three patients of this cohort by the detection of disease-causing variants in the HADHA or HADHB gene. The mitigated phenotypes of this treatable disease were missed by the newborn screening, highlighting the importance of phenotype-based NGS analysis in patients with rare and clinically very variable disorders such as MTP deficiency.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mutation/genetics , Adolescent , Cardiomyopathies/genetics , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Lipid Metabolism, Inborn Errors/genetics , Male , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein/deficiency , Mitochondrial Trifunctional Protein/genetics , Nervous System Diseases/genetics , Phenotype , Rhabdomyolysis/genetics , Syndrome
13.
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
14.
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
15.
Neuropediatrics ; 48(2): 111-114, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28024309

ABSTRACT

Deficiency of adenosine deaminase 2 (ADA2) due to homozygous or compound heterozygous mutations in the cat eye syndrome chromosome region, candidate 1 (CECR1) gene causes an autoimmune phenotype with systemic vasculitis affecting the skin, inner organs, and the central nervous system. Typically, stroke has been reported to follow systemic inflammatory disease and predominantly affects posterior and central brain areas. Here, we describe one of the rare patients in whom acute mesencephalic stroke preceded systemic inflammation and presented as initial clinical symptom. Symptoms typical for ADA2 deficiency such as fever, livedo racemosa, abdominal colics, arthralgias, and Raynaud phenomenon were observed later. Moreover, angiography of cerebral arteries did not reveal typical vasculitic findings supporting the hypothesis that alternative mechanism of vascular occlusion might have caused the stroke. ADA2 deficiency should be considered in patients with childhood stroke despite the absence of systemic inflammation and cerebral vasculitis.


Subject(s)
Adenosine Deaminase/deficiency , Brain Ischemia/etiology , Inflammation/complications , Intercellular Signaling Peptides and Proteins/deficiency , Stroke/etiology , Adenosine Deaminase/genetics , Brain/diagnostic imaging , Brain Ischemia/diagnosis , Brain Ischemia/enzymology , Brain Ischemia/genetics , Diagnosis, Differential , Hand/pathology , Humans , Infant , Inflammation/diagnosis , Inflammation/enzymology , Inflammation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Leg/pathology , Male , Mutation, Missense , Stroke/diagnosis , Stroke/enzymology , Stroke/genetics
16.
Muscle Nerve ; 54(2): 328-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27015314

ABSTRACT

INTRODUCTION: Neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP) is caused by m.8993T>G/C mutations in the mitochondrial adenosine triphosphate synthase subunit 6 gene (MT-ATP6). Traditionally, heteroplasmy levels between 70% and 90% lead to NARP, and >90% result in Leigh syndrome. METHODS: In this study we report a 30-year-old man with NARP and m.8993T>G in MT-ATP6. RESULTS: Although the patient carried the mutation in homoplasmic state in blood with similarly high levels in urine (94%) and buccal swab (92%), he presented with NARP and not the expected, more severe Leigh phenotype. The mutation could not be detected in any of the 3 analyzed tissues of the mother, indicating a large genetic shift between mother and offspring. Nerve biopsy revealed peculiar endoneurial Schwann cell nuclear accumulations, clusters of concentrically arranged Schwann cells devoid of myelinated axons, and degenerated mitochondria. CONCLUSIONS: We emphasize the phenotypic variability of the m.8993T>G MT-ATP6 mutation and the need for caution in predictive counseling in such patients. Muscle Nerve 54: 328-333, 2016.


Subject(s)
Ataxia/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Muscle Weakness/genetics , Retinitis Pigmentosa/genetics , Adult , Ataxia/complications , Humans , Magnetic Resonance Imaging , Male , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/diagnostic imaging , Muscle Weakness/complications , Mutation/genetics , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/diagnostic imaging , Sural Nerve/pathology , Sural Nerve/ultrastructure
17.
J Inherit Metab Dis ; 39(3): 427-436, 2016 05.
Article in English | MEDLINE | ID: mdl-27053192

ABSTRACT

Liver failure is a heterogeneous condition which may be fatal and the primary cause is frequently unknown. We investigated mitochondrial oxidative phosphorylation in patients undergoing liver transplantation. We studied 45 patients who had liver transplantation due to a variety of clinical presentations. Blue native polyacrylamide gel electrophoresis with immunodetection of respiratory chain complexes I-V, biochemical activity of respiratory chain complexes II and IV and quantification of mitochondrial DNA (mtDNA) copy number were investigated in liver tissue collected from the explanted liver during transplantation. Abnormal mitochondrial function was frequently present in this cohort: ten of 40 patients (25 %) had a defect of one or more respiratory chain enzyme complexes on blue native gels, 20 patients (44 %) had low activity of complex II and/or IV and ten (22 %) had a reduced mtDNA copy number. Combined respiratory chain deficiency and reduced numbers of mitochondria were detected in all three patients with acute liver failure. Low complex IV activity in biliary atresia and complex II defects in cirrhosis were common findings. All six patients diagnosed with liver tumours showed variable alterations in mitochondrial function, probably due to the heterogeneity of the presenting tumour. In conclusion, mitochondrial dysfunction is common in severe liver failure in non-mitochondrial conditions. Therefore, in contrast to the common practice detection of respiratory chain abnormalities in liver should not restrict the inclusion of patients for liver transplantation. Furthermore, improving mitochondrial function may be targeted as part of a complex therapy approach in different forms of liver diseases.


Subject(s)
Liver Failure/pathology , Liver/pathology , Mitochondria/pathology , Mitochondrial Diseases/pathology , Adolescent , Adult , Biliary Atresia/metabolism , Biliary Atresia/pathology , Child , Child, Preschool , DNA, Mitochondrial/metabolism , Electron Transport/physiology , Electron Transport Complex IV/metabolism , Female , Humans , Infant , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Failure/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Transplantation/methods , Male , Middle Aged , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Oxidative Phosphorylation , Young Adult
18.
Mol Cell Probes ; 29(5): 319-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26327357

ABSTRACT

Mutations in the DARS2 gene are known to cause leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), a rare autosomal recessive neurological disorder. It was originally described as juvenile-onset slowly progressive ataxia and spasticity, but recent reports suggest a broader clinical spectrum. Most patients were found to carry compound heterozygous DARS2 mutations, and only very few patients with homozygous mutations have been described so far. We present here an 8-month-old boy carrying a homozygous missense mutation in DARS2 who clinically showed severe neurological deterioration after a respiratory tract infection, followed by an almost complete remission of symptoms. This report further extends the knowledge about the clinical and molecular genetic spectrum of LBSL.


Subject(s)
Aspartate-tRNA Ligase/genetics , Leukoencephalopathies/genetics , Mutation, Missense , Age of Onset , Genetic Predisposition to Disease , Homozygote , Humans , Infant , Leukoencephalopathies/diagnosis , Male , Pedigree , Sequence Analysis, DNA
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL