Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Orphanet J Rare Dis ; 19(1): 197, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741100

ABSTRACT

BACKGROUND: Rare diseases are often complex, chronic and many of them life-shortening. In Germany, healthcare for rare diseases is organized in expert centers for rare diseases. Most patients additionally have regional general practicioners and specialists for basic medical care. Thus, collaboration and information exchange between sectors is highly relevant. Our study focuses on the patient and caregiver perspective on intersectoral and interdisciplinary care between local healthcare professionals (HCPs) and centers for rare diseases in Germany. The aims were (1) to investigate patients' and caregivers' general experience of healthcare, (2) to analyse patients' and caregivers' perception of collaboration and cooperation between local healthcare professionals and expert centers for rare diseases and (3) to investigate patients' and caregivers' satisfaction with healthcare in the expert centers for rare diseases. RESULTS: In total 299 individuals of whom 176 were patients and 123 were caregivers to pediatric patients participated in a survey using a questionnaire comprising several instruments and constructs. Fifty participants were additionally interviewed using a semistructured guideline. Most patients reported to receive written information about their care, have a contact person for medical issues and experienced interdisciplinary exchange within the centers for rare diseases. Patients and caregivers in our sample were mainly satisfied with the healthcare in the centers for rare diseases. The qualitative interviews showed a rather mixed picture including experiences of uncoordinated care, low engagement and communication difficulties between professionals of different sectors. Patients reported several factors that influenced the organization and quality of healthcare e.g. engagement and health literacy in patients or engagement of HCPs. CONCLUSIONS: Our findings indicate the high relevance of transferring affected patients to specialized care as fast as possible to provide best medical treatment and increase patient satisfaction. Intersectoral collaboration should exceed written information exchange and should unburden patients of being and feeling responsible for communication between sectors and specialists. Results indicate a lack of inclusion of psychosocial aspects in routine care, which suggests opportunities for necessary improvements.


Subject(s)
Rare Diseases , Humans , Rare Diseases/therapy , Germany , Male , Female , Surveys and Questionnaires , Adult , Middle Aged , Intersectoral Collaboration , Health Personnel/psychology , Delivery of Health Care , Communication , Patient Satisfaction , Young Adult , Caregivers/psychology
2.
Mol Genet Metab ; 140(3): 107675, 2023 11.
Article in English | MEDLINE | ID: mdl-37572574

ABSTRACT

Recessive variants in NDUFAF3 are a known cause of complex I (CI)-related mitochondrial disorders (MDs). The seven patients reported to date exhibited severe neurologic symptoms and lactic acidosis, followed by a fatal course and death during infancy in most cases. We present a 10-year-old patient with a neurodevelopmental disorder, progressive exercise intolerance, dystonia, basal ganglia abnormalities, and elevated lactate concentration in blood. Trio-exome sequencing revealed compound-heterozygosity for a pathogenic splice-site and a likely pathogenic missense variant in NDUFAF3. Spectrophotometric analysis of fibroblast-derived mitochondria demonstrated a relatively mild reduction of CI activity. Complexome analyses revealed severely reduced NDUFAF3 as well as CI in patient fibroblasts. Accumulation of early sub-assemblies of the membrane arm of CI associated with mitochondrial complex I intermediate assembly (MCIA) complex was observed. The most striking additional findings were both the unusual occurrence of free monomeric CI holding MCIA and other assembly factors. Here we discuss our patient in context of genotype, phenotype and metabolite data from previously reported NDUFAF3 cases. With the atypical presentation of our patient, we provide further insight into the phenotypic spectrum of NDUFAF3-related MDs. Complexome analysis in our patient confirms the previously defined role of NDUFAF3 within CI biogenesis, yet adds new aspects regarding the correct timing of both the association of soluble and membrane arm modules and CI-maturation as well as respiratory supercomplex formation.


Subject(s)
Acidosis, Lactic , Mitochondrial Diseases , Humans , Child , Mitochondrial Diseases/genetics , Mitochondria/genetics , Mitochondria/metabolism , Exome Sequencing , Acidosis, Lactic/genetics , Phenotype , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
3.
Mol Genet Metab Rep ; 35: 100977, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37275680

ABSTRACT

We report on liver transplantation in two patients with GSD Ib on treatment with empagliflozin. The use of this SGLT2 inhibitor resulted in a marked decrease of 1,5-anhydroglucitol which has an important role in the development of neutropenia in this condition. As intended, this caused a significant rise of neutrophil numbers. Liver transplantation alone did not produce the desired effect and our observation argues for continuing SGLT2 inhibitor treatment after transplantation.

4.
Brain Commun ; 5(1): fcad017, 2023.
Article in English | MEDLINE | ID: mdl-36793789

ABSTRACT

Superoxide dismutase-1 is a ubiquitously expressed antioxidant enzyme. Mutations in SOD1 can cause amyotrophic lateral sclerosis, probably via a toxic gain-of-function involving protein aggregation and prion-like mechanisms. Recently, homozygosity for loss-of-function mutations in SOD1 has been reported in patients presenting with infantile-onset motor neuron disease. We explored the bodily effects of superoxide dismutase-1 enzymatic deficiency in eight children homozygous for the p.C112Wfs*11 truncating mutation. In addition to physical and imaging examinations, we collected blood, urine and skin fibroblast samples. We used a comprehensive panel of clinically established analyses to assess organ function and analysed oxidative stress markers, antioxidant compounds, and the characteristics of the mutant Superoxide dismutase-1. From around 8 months of age, all patients exhibited progressive signs of both upper and lower motor neuron dysfunction, cerebellar, brain stem, and frontal lobe atrophy and elevated plasma neurofilament concentration indicating ongoing axonal damage. The disease progression seemed to slow down over the following years. The p.C112Wfs*11 gene product is unstable, rapidly degraded and no aggregates were found in fibroblast. Most laboratory tests indicated normal organ integrity and only a few modest deviations were found. The patients displayed anaemia with shortened survival of erythrocytes containing decreased levels of reduced glutathione. A variety of other antioxidants and oxidant damage markers were within normal range. In conclusion, non-neuronal organs in humans show a remarkable tolerance to absence of Superoxide dismutase-1 enzymatic activity. The study highlights the enigmatic specific vulnerability of the motor system to both gain-of-function mutations in SOD1 and loss of the enzyme as in the here depicted infantile superoxide dismutase-1 deficiency syndrome.

5.
Calcif Tissue Int ; 112(3): 308-319, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36414794

ABSTRACT

Hypophosphatasia (HPP) is an inherited, systemic disorder, caused by loss-of-function variants of the ALPL gene encoding the enzyme tissue non-specific alkaline phosphatase (TNSALP). HPP is characterized by low serum TNSALP concentrations associated with defective bone mineralization and increased fracture risk. Dental manifestations have been reported as the exclusive feature (odontohypophosphatasia) and in combination with skeletal complications. Enzyme replacement therapy (asfotase alfa) has been shown to improve respiratory insufficiency and skeletal complications in HPP patients, while its effects on dental status have been understudied to date. In this study, quantitative backscattered electron imaging (qBEI) and histological analysis were performed on teeth from two patients with infantile HPP before and during asfotase alfa treatment and compared to matched healthy control teeth. qBEI and histological methods revealed varying mineralization patterns in cementum and dentin with lower mineralization in HPP. Furthermore, a significantly higher repair cementum thickness was observed in HPP compared to control teeth. Comparison before and during treatment showed minor improvements in mineralization and histological parameters in the patient when normalized to matched control teeth. HPP induces heterogeneous effects on mineralization and morphology of the dental status. Short treatment with asfotase alfa slightly affects mineralization in cementum and dentin. Despite HPP being a rare disease, its mild form occurs at higher prevalence. This study is of high clinical relevance as it expands our knowledge of HPP and dental involvement. Furthermore, it contributes to the understanding of dental tissue treatment, which has hardly been studied so far.


Subject(s)
Calcinosis , Hypophosphatasia , Tooth Demineralization , Humans , Hypophosphatasia/complications , Alkaline Phosphatase/genetics , Calcification, Physiologic , Calcinosis/complications , Tooth Demineralization/complications , Tooth Demineralization/drug therapy
6.
Hum Mol Genet ; 32(6): 917-933, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36190515

ABSTRACT

Maintaining protein lipoylation is vital for cell metabolism. The H-protein encoded by GCSH has a dual role in protein lipoylation required for bioenergetic enzymes including pyruvate dehydrogenase and 2-ketoglutarate dehydrogenase, and in the one-carbon metabolism through its involvement in glycine cleavage enzyme system, intersecting two vital roles for cell survival. Here, we report six patients with biallelic pathogenic variants in GCSH and a broad clinical spectrum ranging from neonatal fatal glycine encephalopathy to an attenuated phenotype of developmental delay, behavioral problems, limited epilepsy and variable movement problems. The mutational spectrum includes one insertion c.293-2_293-1insT, one deletion c.122_(228 + 1_229-1) del, one duplication of exons 4 and 5, one nonsense variant p.Gln76*and four missense p.His57Arg, p.Pro115Leu and p.Thr148Pro and the previously described p.Met1?. Via functional studies in patient's fibroblasts, molecular modeling, expression analysis in GCSH knockdown COS7 cells and yeast, and in vitro protein studies, we demonstrate for the first time that most variants identified in our cohort produced a hypomorphic effect on both mitochondrial activities, protein lipoylation and glycine metabolism, causing combined deficiency, whereas some missense variants affect primarily one function only. The clinical features of the patients reflect the impact of the GCSH changes on any of the two functions analyzed. Our analysis illustrates the complex interplay of functional and clinical impact when pathogenic variants affect a multifunctional protein involved in two metabolic pathways and emphasizes the value of the functional assays to select the treatment and investigate new personalized options.


Subject(s)
Hyperglycinemia, Nonketotic , Humans , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/pathology , Proteins/genetics , Mutation , Exons/genetics , Glycine/genetics , Glycine/metabolism
7.
Genes (Basel) ; 12(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34828390

ABSTRACT

Fanconi-Bickel syndrome (FBS) is a very rare but distinct clinical entity with the combined features of hepatic glycogen storage disease, generalized proximal renal tubular dysfunction with disproportionately severe glucosuria, and impaired galactose tolerance. Here, we report five cases (out of 93 diagnosed in our lab) with pathogenic variants on both GLUT2 (SLC2A2) alleles. They come from 3 families and presented with an exceptionally mild clinical course. This course was correlated to data from old and most recent expression and transport studies in Xenopus oocytes. GLUT2 genotype in patients 1 and 2 was p.[153_4delLI];[P417R] with the first variant exhibiting normal membrane expression and partially retained transport activity (5.8%) for 2-deoxyglucose. In patient 3, the very first GLUT2 variant ever detected (p.V197I) was found, but for the first time it was present in a patient in the homozygous state. This variant had also shown unaffected membrane expression and remarkable residual activity (8%). The genotype in patient 4, p.[153_4delLI];[(E440A)], again included the 2-amino-acid deletion with residual transporter function, and patient 5 is the first found to be homozygous for this variant. Our results provide further evidence for a genotype-phenotype correlation in patients with GLUT2 variants; non-functional variants result in the full picture of FBS while dysfunctional variants may result in milder presentations, even glucosuria only, without other typical signs of FBS.


Subject(s)
Fanconi Syndrome/genetics , Glucose Transporter Type 2/genetics , Mutation , Phenotype , Adolescent , Adult , Animals , Fanconi Syndrome/pathology , Female , Genotype , Glucose/metabolism , Glucose Transporter Type 2/metabolism , Homozygote , Humans , Infant , Male , Pedigree , Xenopus
8.
Clin Genet ; 100(6): 766-770, 2021 12.
Article in English | MEDLINE | ID: mdl-34490615

ABSTRACT

Neurological symptoms are frequent and often a leading feature of childhood-onset mitochondrial disorders (MD) but the exact incidence of MD in unselected neuropediatric patients is unknown. Their early detection is desirable due to a potentially rapid clinical decline and the availability of management options. In 491 children with neurological symptoms, a comprehensive diagnostic work-up including exome sequencing was performed. The success rate in terms of a molecular genetic diagnosis within our cohort was 51%. Disease-causing variants in a mitochondria-associated gene were detected in 12% of solved cases. In order to facilitate the clinical identification of MDs within neuropediatric cohorts, we have created an easy-to-use bedside-tool, the MDC-NP. In our cohort, the MDC-NP predicted disease conditions related to MDs with a sensitivity of 0.83, and a specificity of 0.96.


Subject(s)
Genetic Predisposition to Disease , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/genetics , Nervous System Diseases/epidemiology , Nervous System Diseases/genetics , Age Factors , Alleles , Child , Cohort Studies , Genes, Mitochondrial , Genetic Association Studies , Genotype , Humans , Mitochondrial Diseases/diagnosis , Mutation , Nervous System Diseases/diagnosis , Phenotype , Prevalence , Prognosis
9.
J Inherit Metab Dis ; 44(5): 1272-1287, 2021 09.
Article in English | MEDLINE | ID: mdl-34145613

ABSTRACT

Mevalonic aciduria (MVA) and hyperimmunoglobulinemia D syndrome (MKD/HIDS) are disorders of cholesterol biosynthesis caused by variants in the MVK gene and characterized by increased urinary excretion of mevalonic acid. So far, 30 MVA patients have been reported, suffering from recurrent febrile crises and neurologic impairment. Here, we present an in-depth analysis of the phenotypic spectrum of MVA and provide an in-silico pathogenicity model analysis of MVK missense variants. The phenotypic spectrum of 11 MVA patients (age range 0-51 years) registered in the Unified European Registry for Inherited Metabolic Disorders database was systematically analyzed using terms of the Human Phenotype Ontology. Biochemical, radiological as well as genetic characteristics were investigated. Six of eleven patients have reached adulthood and four have reached adolescence. One of the adolescent patients died at the age of 16 years and one patient died shortly after birth. Symptoms started within the first year of life, including episodic fever, developmental delay, ataxia, and ocular involvement. We also describe a case with absence of symptoms despite massive excretion of mevalonic acid. Pathogenic variants causing MVA cluster within highly conserved regions, which are involved in mevalonate and ATP binding. The phenotype of adult and adolescent MVA patients is more heterogeneous than previously assumed. Outcome varies from an asymptomatic course to early death. MVK variants cluster in functionally important and highly conserved protein domains and show high concordance regarding their expected pathogenicity.


Subject(s)
Mevalonate Kinase Deficiency/pathology , Mevalonic Acid/metabolism , Mutation, Missense , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adolescent , Adult , Disease Progression , Female , Humans , Male , Mevalonate Kinase Deficiency/metabolism , Mevalonic Acid/urine , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Young Adult
10.
J Inherit Metab Dis ; 44(4): 893-902, 2021 07.
Article in English | MEDLINE | ID: mdl-33638202

ABSTRACT

Peripheral neuropathy is a known irreversible long-term complication of long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) and mitochondrial trifunctional protein deficiency (MTPD), two inherited disorders of mitochondrial long-chain fatty acid oxidation. The underlying pathophysiology of neuropathy is still not fully understood. We report electrophysiological studies and neurological findings in a series of 8 LCHAD-deficient and 11 MTP-deficient patients. The median age at time of the study was 8.0 years (0.5-25 years). The overall prevalence of neuropathy was 58% with neuropathic symptoms being slightly more common in MTPD compared to LCHADD (70% vs 50%, respectively). Onset of neuropathy was significantly earlier in MTPD patients compared to LCHADD patients (median age at onset 4.7 vs 15.3 years, respectively, P = .047). In four patients, isolated peripheral neuropathy was the first and only presenting symptom, and in all four the diagnosis was missed by newborn screening. About half of the patients (45.5%) had a sensorimotor neuropathy, while 27.3% showed a pure motor form and another 27.3% an isolated sensory form. Despite early diagnosis by newborn screening and early initiation of therapy, peripheral neuropathy cannot be prevented in all patients with LCHADD/MTPD and has severe impact on the life of affected patients. Electrophysiology classifies LCHADD/MTPD neuropathy as axonal with secondary demyelination. A novel observation is that in patients with acute, fulminant onset of neuropathy, symptoms can be partly reversible. Further studies are needed to elucidate the underlying pathophysiology of axonal damage and possible therapeutic targets.


Subject(s)
Cardiomyopathies/complications , Lipid Metabolism, Inborn Errors/complications , Mitochondrial Myopathies/complications , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/complications , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/etiology , Rhabdomyolysis/complications , Adolescent , Adult , Age Factors , Cardiomyopathies/diagnosis , Cardiomyopathies/pathology , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/pathology , Male , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/pathology , Nervous System Diseases/diagnosis , Nervous System Diseases/pathology , Peripheral Nervous System Diseases/pathology , Phenotype , Rhabdomyolysis/diagnosis , Rhabdomyolysis/pathology , Young Adult
11.
Ophthalmic Genet ; 42(1): 23-27, 2021 02.
Article in English | MEDLINE | ID: mdl-33107778

ABSTRACT

Introduction: LCHADD causes retinopathy associated with low vision, visual field defects, nyctalopia and myopia. We report a retrospective long-term single-center study of 6 LCHADD patients trying to clarify if early diagnosis has an impact on the course and outcome of chorioretinal degeneration. Methods: Long-term follow-up of visual acuity and staging of chorioretinal degeneration by fundus photography, optical coherence tomography (OCT) and autofluorescence (AF) in all six patients. Three patients (2 m/1 f; age 8-14.8 years) were diagnosed by newborn screening, a single patient early within the first year of life and treated promptly while the other two (1 m/1 f; age 23-24 years) were diagnosed later after developing symptoms. All carried HADHA variants; five were homozygous for the common p.E510Q variant, in one from the symptomatically diagnosed group p.[E510Q]; [R291*] was detected. Results: All patients showed retinal alterations, but early diagnosis was associated with a milder phenotype and a longer preservation of visual function. Among symptomatic patients, only one showed mild retinal involvement at the time of diagnosis. Conclusion: Despite the small number our study suggests that early diagnosis does not prevent retinopathy but might contribute to a milder phenotype with retained good visual acuity over time. OCT and AF are reliable non-invasive diagnostic tools to estimate the progression of early-stage retinal changes in LCHADD patients.


Subject(s)
Cardiomyopathies/pathology , Lipid Metabolism, Inborn Errors/pathology , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/deficiency , Mitochondrial Myopathies/pathology , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/pathology , Rhabdomyolysis/pathology , Visual Acuity , Adolescent , Adult , Cardiomyopathies/genetics , Child , Female , Humans , Lipid Metabolism, Inborn Errors/genetics , Male , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein/genetics , Multimodal Imaging , Nervous System Diseases/genetics , Prognosis , Retrospective Studies , Rhabdomyolysis/genetics , Young Adult
13.
Am J Hum Genet ; 105(1): 132-150, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31230720

ABSTRACT

Arthrogryposis is a clinical finding that is present either as a feature of a neuromuscular condition or as part of a systemic disease in over 400 Mendelian conditions. The underlying molecular etiology remains largely unknown because of genetic and phenotypic heterogeneity. We applied exome sequencing (ES) in a cohort of 89 families with the clinical sign of arthrogryposis. Additional molecular techniques including array comparative genomic hybridization (aCGH) and Droplet Digital PCR (ddPCR) were performed on individuals who were found to have pathogenic copy number variants (CNVs) and mosaicism, respectively. A molecular diagnosis was established in 65.2% (58/89) of families. Eleven out of 58 families (19.0%) showed evidence for potential involvement of pathogenic variation at more than one locus, probably driven by absence of heterozygosity (AOH) burden due to identity-by-descent (IBD). RYR3, MYOM2, ERGIC1, SPTBN4, and ABCA7 represent genes, identified in two or more families, for which mutations are probably causative for arthrogryposis. We also provide evidence for the involvement of CNVs in the etiology of arthrogryposis and for the idea that both mono-allelic and bi-allelic variants in the same gene cause either similar or distinct syndromes. We were able to identify the molecular etiology in nine out of 20 families who underwent reanalysis. In summary, our data from family-based ES further delineate the molecular etiology of arthrogryposis, yielded several candidate disease-associated genes, and provide evidence for mutational burden in a biological pathway or network. Our study also highlights the importance of reanalysis of individuals with unsolved diagnoses in conjunction with sequencing extended family members.


Subject(s)
Arthrogryposis/genetics , Arthrogryposis/pathology , DNA Copy Number Variations , Genetic Markers , Genomics/methods , Multifactorial Inheritance/genetics , Mutation , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Connectin/genetics , Female , Gestational Age , Humans , Infant , Infant, Newborn , Male , Mosaicism , Pedigree , Ryanodine Receptor Calcium Release Channel/genetics , Vesicular Transport Proteins/genetics , Exome Sequencing , Young Adult
14.
Dtsch Arztebl Int ; 116(12): 197-204, 2019 03 22.
Article in English | MEDLINE | ID: mdl-31056085

ABSTRACT

BACKGROUND: In developed countries, global developmental disorders are encounter- ed in approximately 1% of all children. The causes are manifold, and no exogenous cause can be identified in about half of the affected children. The parallel investi- gation of the coding sequences of all genes of the affected individual (whole exome sequencing, WES) has developed into a successful diagnostic method for identify- ing the cause of the problem. It is not yet clear, however, when WES should best be used in routine clinical practice in order to exploit the potential of this method to the fullest. METHODS: In an interdisciplinary study, we carried out standardized clinical pheno- typing and a systematic genetic analysis (WES of the index patient and his or her parents, so-called trio WES) in 50 children with developmental disturbances of unclear etiology and with nonspecific neurological manifestations. RESULTS: In 21 children (42% of the collective), we were able to identify the cause of the disorder by demonstrating a mutation in a gene known to be associated with disease. Three of these children subsequently underwent specific treatment. In 22 other children (44%), we detected possibly etiological changes in candidate genes not currently known to be associated with human disease. CONCLUSION: Our detection rate of at least 42% is high in comparison with the results obtained in other studies from Germany and other countries to date and implies that WES can be used to good effect as a differential diagnostic tool in pediatric neurol- ogy. WES should be carried out in both the index patient and his or her parents (trio- WES) and accompanied by close interdisciplinary collaboration of human geneti- cists and pediatricians, by comprehensive and targeted phenotyping (also after the diagnosis is established), and by the meticulous evaluation of all gene variants.


Subject(s)
Exome Sequencing , Child , Germany , Humans
15.
Ann Neurol ; 83(6): 1089-1095, 2018 06.
Article in English | MEDLINE | ID: mdl-29518281

ABSTRACT

VPS13 protein family members VPS13A through VPS13C have been associated with various recessive movement disorders. We describe the first disease association of rare recessive VPS13D variants including frameshift, missense, and partial duplication mutations with a novel complex, hyperkinetic neurological disorder. The clinical features include developmental delay, a childhood onset movement disorder (chorea, dystonia, or tremor), and progressive spastic ataxia or paraparesis. Characteristic brain magnetic resonance imaging shows basal ganglia or diffuse white matter T2 hyperintensities as seen in Leigh syndrome and choreoacanthocytosis. Muscle biopsy in 1 case showed mitochondrial aggregates and lipidosis, suggesting mitochondrial dysfunction. These findings underline the importance of the VPS13 complex in neurological diseases and a possible role in mitochondrial function. Ann Neurol 2018;83:1089-1095.


Subject(s)
Intellectual Disability/genetics , Movement Disorders/genetics , Muscle Spasticity/genetics , Mutation/genetics , Optic Atrophy/genetics , Proteins/genetics , Spinocerebellar Ataxias/genetics , Basal Ganglia/pathology , Brain/pathology , Child , Humans , Leigh Disease/pathology , Magnetic Resonance Imaging/methods , Muscle Spasticity/pathology , Pedigree
16.
Genet Med ; 20(2): 259-268, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28749473

ABSTRACT

PurposeSLC39A8 deficiency is a severe inborn error of metabolism that is caused by impaired function of manganese metabolism in humans. Mutations in SLC39A8 lead to impaired function of the manganese transporter ZIP8 and thus manganese deficiency. Due to the important role of Mn2+ as a cofactor for a variety of enzymes, the resulting phenotype is complex and severe. The manganese-dependence of ß-1,4-galactosyltransferases leads to secondary hypoglycosylation, making SLC39A8 deficiency both a disorder of trace element metabolism and a congenital disorder of glycosylation. Some hypoglycosylation disorders have previously been treated with galactose administration. The development of an effective treatment of the disorder by high-dose manganese substitution aims at correcting biochemical, and hopefully, clinical abnormalities.MethodsTwo SCL39A8 deficient patients were treated with 15 and 20 mg MnSO4/kg bodyweight per day. Glycosylation and blood manganese were monitored closely. In addition, magnetic resonance imaging was performed to detect potential toxic effects of manganese.ResultsAll measured enzyme dysfunctions resolved completely and considerable clinical improvement regarding motor abilities, hearing, and other neurological manifestations was observed.ConclusionHigh-dose manganese substitution was effective in two patients with SLC39A8 deficiency. Close therapy monitoring by glycosylation assays and blood manganese measurements is necessary to prevent manganese toxicity.


Subject(s)
Cation Transport Proteins/deficiency , Genetic Association Studies , Genetic Predisposition to Disease , Alleles , Biomarkers , Dietary Supplements , Electroencephalography , Female , Genetic Association Studies/methods , Glycosylation/drug effects , Humans , Magnetic Resonance Imaging , Manganese/administration & dosage , Manganese/adverse effects , Manganese/therapeutic use , Mutation , Phenotype , Treatment Outcome
17.
Childs Nerv Syst ; 34(3): 581-584, 2018 03.
Article in English | MEDLINE | ID: mdl-29167993

ABSTRACT

Glycogen storage disease (GSD) 0a is a rare congenital metabolic disease with symptoms in infancy and childhood caused by biallelic GYS2 germline variants. A predisposition to cancer has not been described yet. We report here a boy with GSD 0a, who developed a malignant brain tumor at the age of 4.5 years. The tumor was classified as a group 3 medulloblastoma, and the patient died from cancer 27 months after initial tumor diagnosis. This case appears interesting as group 3 medulloblastoma is so far not known to arise in hereditary syndromes and the biology of sporadic group 3 medulloblastoma is largely unknown.


Subject(s)
Cerebellar Neoplasms/diagnostic imaging , Germ-Line Mutation , Glycogen Storage Disease/diagnostic imaging , Medulloblastoma/diagnostic imaging , Cerebellar Neoplasms/complications , Cerebellar Neoplasms/genetics , Child, Preschool , Fatal Outcome , Germ-Line Mutation/genetics , Glycogen Storage Disease/complications , Glycogen Storage Disease/genetics , Humans , Male , Medulloblastoma/complications , Medulloblastoma/genetics
18.
Ann Neurol ; 82(6): 1004-1015, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29205472

ABSTRACT

OBJECTIVE: 3-Methylglutaconic aciduria, dystonia-deafness, hepatopathy, encephalopathy, Leigh-like syndrome (MEGDHEL) syndrome is caused by biallelic variants in SERAC1. METHODS: This multicenter study addressed the course of disease for each organ system. Metabolic, neuroradiological, and genetic findings are reported. RESULTS: Sixty-seven individuals (39 previously unreported) from 59 families were included (age range = 5 days-33.4 years, median age = 9 years). A total of 41 different SERAC1 variants were identified, including 20 that have not been reported before. With the exception of 2 families with a milder phenotype, all affected individuals showed a strikingly homogeneous phenotype and time course. Severe, reversible neonatal liver dysfunction and hypoglycemia were seen in >40% of all cases. Starting at a median age of 6 months, muscular hypotonia (91%) was seen, followed by progressive spasticity (82%, median onset = 15 months) and dystonia (82%, 18 months). The majority of affected individuals never learned to walk (68%). Seventy-nine percent suffered hearing loss, 58% never learned to speak, and nearly all had significant intellectual disability (88%). Magnetic resonance imaging features were accordingly homogenous, with bilateral basal ganglia involvement (98%); the characteristic "putaminal eye" was seen in 53%. The urinary marker 3-methylglutaconic aciduria was present in virtually all patients (98%). Supportive treatment focused on spasticity and drooling, and was effective in the individuals treated; hearing aids or cochlear implants did not improve communication skills. INTERPRETATION: MEGDHEL syndrome is a progressive deafness-dystonia syndrome with frequent and reversible neonatal liver involvement and a strikingly homogenous course of disease. Ann Neurol 2017;82:1004-1015.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Deaf-Blind Disorders/diagnostic imaging , Deaf-Blind Disorders/genetics , Disease Progression , Dystonia/diagnostic imaging , Dystonia/genetics , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Mutation/genetics , Optic Atrophy/diagnostic imaging , Optic Atrophy/genetics , Adolescent , Adult , Amino Acid Sequence , Child , Child, Preschool , Cohort Studies , Deaf-Blind Disorders/therapy , Dystonia/therapy , Female , Humans , Infant , Infant, Newborn , Intellectual Disability/therapy , Male , Optic Atrophy/therapy , Young Adult
19.
Mitochondrion ; 37: 55-61, 2017 11.
Article in English | MEDLINE | ID: mdl-28694194

ABSTRACT

LYRM7 is involved in the last steps of mitochondrial complex III assembly where it acts as a chaperone for the Rieske iron­sulfur (Fe-S) protein in the mitochondrial matrix. Using exome sequencing, we identified homozygosity for a splice site destroying 4 base pair deletion in LYRM7 in a child with recurrent lactic acidotic crises and distinct early-onset leukencephalopathy. Sanger sequencing showed variant segregation in similarly affected family members. Functional analyses revealed a reduced amount of the Rieske Fe-S protein, which was restored after re-expression of LYRM7. Our data provide further evidence for the importance of LYRM7 for mitochondrial function and emphasize the importance of whole exome sequencing in the diagnosis of rare mitochondrial diseases.


Subject(s)
Electron Transport Complex III/deficiency , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Acidosis, Lactic/complications , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Child, Preschool , Electron Transport Complex III/analysis , Female , Humans , Infant , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Sequence Deletion
20.
Am J Hum Genet ; 97(6): 894-903, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637979

ABSTRACT

SLC39A8 is a membrane transporter responsible for manganese uptake into the cell. Via whole-exome sequencing, we studied a child that presented with cranial asymmetry, severe infantile spasms with hypsarrhythmia, and dysproportionate dwarfism. Analysis of transferrin glycosylation revealed severe dysglycosylation corresponding to a type II congenital disorder of glycosylation (CDG) and the blood manganese levels were below the detection limit. The variants c.112G>C (p.Gly38Arg) and c.1019T>A (p.Ile340Asn) were identified in SLC39A8. A second individual with the variants c.97G>A (p.Val33Met) and c.1004G>C (p.Ser335Thr) on the paternal allele and c.610G>T (p.Gly204Cys) on the maternal allele was identified among a group of unresolved case subjects with CDG. These data demonstrate that variants in SLC39A8 impair the function of manganese-dependent enzymes, most notably ß-1,4-galactosyltransferase, a Golgi enzyme essential for biosynthesis of the carbohydrate part of glycoproteins. Impaired galactosylation leads to a severe disorder with deformed skull, severe seizures, short limbs, profound psychomotor retardation, and hearing loss. Oral galactose supplementation is a treatment option and results in complete normalization of glycosylation. SLC39A8 deficiency links a trace element deficiency with inherited glycosylation disorders.


Subject(s)
Cation Transport Proteins/genetics , Congenital Disorders of Glycosylation/genetics , Dwarfism/genetics , Manganese/blood , Spasms, Infantile/genetics , Amino Acid Sequence , Carbohydrate Sequence , Cation Transport Proteins/deficiency , Cations, Divalent , Congenital Disorders of Glycosylation/blood , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/diet therapy , Dwarfism/blood , Dwarfism/complications , Dwarfism/diet therapy , Female , Galactose/therapeutic use , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant , Ion Transport , Manganese/deficiency , Molecular Sequence Data , Mutation , Pedigree , Sequence Alignment , Spasms, Infantile/blood , Spasms, Infantile/complications , Spasms, Infantile/diet therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...