Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
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
2.
Am J Hum Genet ; 109(9): 1692-1712, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36055214

ABSTRACT

Leucine zipper-EF-hand containing transmembrane protein 1 (LETM1) encodes an inner mitochondrial membrane protein with an osmoregulatory function controlling mitochondrial volume and ion homeostasis. The putative association of LETM1 with a human disease was initially suggested in Wolf-Hirschhorn syndrome, a disorder that results from de novo monoallelic deletion of chromosome 4p16.3, a region encompassing LETM1. Utilizing exome sequencing and international gene-matching efforts, we have identified 18 affected individuals from 11 unrelated families harboring ultra-rare bi-allelic missense and loss-of-function LETM1 variants and clinical presentations highly suggestive of mitochondrial disease. These manifested as a spectrum of predominantly infantile-onset (14/18, 78%) and variably progressive neurological, metabolic, and dysmorphic symptoms, plus multiple organ dysfunction associated with neurodegeneration. The common features included respiratory chain complex deficiencies (100%), global developmental delay (94%), optic atrophy (83%), sensorineural hearing loss (78%), and cerebellar ataxia (78%) followed by epilepsy (67%), spasticity (53%), and myopathy (50%). Other features included bilateral cataracts (42%), cardiomyopathy (36%), and diabetes (27%). To better understand the pathogenic mechanism of the identified LETM1 variants, we performed biochemical and morphological studies on mitochondrial K+/H+ exchange activity, proteins, and shape in proband-derived fibroblasts and muscles and in Saccharomyces cerevisiae, which is an important model organism for mitochondrial osmotic regulation. Our results demonstrate that bi-allelic LETM1 variants are associated with defective mitochondrial K+ efflux, swollen mitochondrial matrix structures, and loss of important mitochondrial oxidative phosphorylation protein components, thus highlighting the implication of perturbed mitochondrial osmoregulation caused by LETM1 variants in neurological and mitochondrial pathologies.


Subject(s)
Calcium-Binding Proteins , Mitochondrial Diseases , Calcium-Binding Proteins/genetics , Homeostasis/genetics , Humans , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nervous System/metabolism , Saccharomyces cerevisiae/metabolism
3.
J Med Genet ; 61(7): 689-698, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38458752

ABSTRACT

BACKGROUND: Plexins are large transmembrane receptors for the semaphorin family of signalling proteins. Semaphorin-plexin signalling controls cellular interactions that are critical during development as well as in adult life stages. Nine plexin genes have been identified in humans, but despite the apparent importance of plexins in development, only biallelic PLXND1 and PLXNA1 variants have so far been associated with Mendelian genetic disease. METHODS: Eight individuals from six families presented with a recessively inherited variable clinical condition, with core features of amelogenesis imperfecta (AI) and sensorineural hearing loss (SNHL), with variable intellectual disability. Probands were investigated by exome or genome sequencing. Common variants and those unlikely to affect function were excluded. Variants consistent with autosomal recessive inheritance were prioritised. Variant segregation analysis was performed by Sanger sequencing. RNA expression analysis was conducted in C57Bl6 mice. RESULTS: Rare biallelic pathogenic variants in plexin B2 (PLXNB2), a large transmembrane semaphorin receptor protein, were found to segregate with disease in all six families. The variants identified include missense, nonsense, splicing changes and a multiexon deletion. Plxnb2 expression was detected in differentiating ameloblasts. CONCLUSION: We identify rare biallelic pathogenic variants in PLXNB2 as a cause of a new autosomal recessive, phenotypically diverse syndrome with AI and SNHL as core features. Intellectual disability, ocular disease, ear developmental abnormalities and lymphoedema were also present in multiple cases. The variable syndromic human phenotype overlaps with that seen in Plxnb2 knockout mice, and, together with the rarity of human PLXNB2 variants, may explain why pathogenic variants in PLXNB2 have not been reported previously.


Subject(s)
Amelogenesis Imperfecta , Intellectual Disability , Pedigree , Humans , Animals , Male , Female , Mice , Intellectual Disability/genetics , Intellectual Disability/pathology , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/pathology , Receptors, Cell Surface/genetics , Nerve Tissue Proteins/genetics , Alleles , Child , Hearing Loss/genetics , Hearing Loss/pathology , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Adult , Mutation/genetics , Adolescent , Child, Preschool , Phenotype
4.
Genet Med ; 26(2): 101013, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37924258

ABSTRACT

PURPOSE: RNF213, encoding a giant E3 ubiquitin ligase, has been recognized for its role as a key susceptibility gene for moyamoya disease. Case reports have also implicated specific variants in RNF213 with an early-onset form of moyamoya disease with full penetrance. We aimed to expand the phenotypic spectrum of monogenic RNF213-related disease and to evaluate genotype-phenotype correlations. METHODS: Patients were identified through reanalysis of exome sequencing data of an unselected cohort of unsolved pediatric cases and through GeneMatcher or ClinVar. Functional characterization was done by proteomics analysis and oxidative phosphorylation enzyme activities using patient-derived fibroblasts. RESULTS: We identified 14 individuals from 13 unrelated families with (de novo) missense variants in RNF213 clustering within or around the Really Interesting New Gene (RING) domain. Individuals presented either with early-onset stroke (n = 11) or with Leigh syndrome (n = 3). No genotype-phenotype correlation could be established. Proteomics using patient-derived fibroblasts revealed no significant differences between clinical subgroups. 3D modeling revealed a clustering of missense variants in the tertiary structure of RNF213 potentially affecting zinc-binding suggesting a gain-of-function or dominant negative effect. CONCLUSION: De novo missense variants in RNF213 clustering in the E3 RING or other regions affecting zinc-binding lead to an early-onset syndrome characterized by stroke or Leigh syndrome.


Subject(s)
Leigh Disease , Moyamoya Disease , Stroke , Humans , Child , Moyamoya Disease/genetics , Leigh Disease/complications , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Zinc , Genetic Predisposition to Disease , Adenosine Triphosphatases/genetics
5.
J Inherit Metab Dis ; 47(2): 270-279, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38084664

ABSTRACT

In this study, we investigated the metabolic signatures of different mitochondrial defects (two different complex I and complex V, and the one MDH2 defect) in human skin fibroblasts (HSF). We hypothesized that using a selective culture medium would cause defect specific adaptation of the metabolome and further our understanding of the biochemical implications for the studied defects. All cells were cultivated under galactose stress condition and compared to glucose-based cell culture condition. We investigated the bioenergetic profile using Seahorse XFe96 cell analyzer and assessed the extracellular metabolic footprints and the intracellular metabolic fingerprints using NMR. The galactose-based culture condition forced a bioenergetic switch from a glycolytic to an oxidative state in all cell lines which improved overall separation of controls from the different defect groups. The extracellular metabolome was discriminative for separating controls from defects but not the specific defects, whereas the intracellular metabolome suggests CI and CV changes and revealed clear MDH2 defect-specific changes in metabolites associated with the TCA cycle, malate aspartate shuttle, and the choline metabolism, which are pronounced under galactose condition.


Subject(s)
Energy Metabolism , Galactose , Humans , Galactose/metabolism , Glycolysis , Magnetic Resonance Spectroscopy , Electron Transport Complex I/metabolism , Fibroblasts/metabolism , Malate Dehydrogenase
6.
Am J Hum Genet ; 106(2): 256-263, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004446

ABSTRACT

We report an inborn error of metabolism caused by TKFC deficiency in two unrelated families. Rapid trio genome sequencing in family 1 and exome sequencing in family 2 excluded known genetic etiologies, and further variant analysis identified rare homozygous variants in TKFC. TKFC encodes a bifunctional enzyme involved in fructose metabolism through its glyceraldehyde kinase activity and in the generation of riboflavin cyclic 4',5'-phosphate (cyclic FMN) through an FMN lyase domain. The TKFC homozygous variants reported here are located within the FMN lyase domain. Functional assays in yeast support the deleterious effect of these variants on protein function. Shared phenotypes between affected individuals with TKFC deficiency include cataracts and developmental delay, associated with cerebellar hypoplasia in one case. Further complications observed in two affected individuals included liver dysfunction and microcytic anemia, while one had fatal cardiomyopathy with lactic acidosis following a febrile illness. We postulate that deficiency of TKFC causes disruption of endogenous fructose metabolism leading to generation of by-products that can cause cataract. In line with this, an affected individual had mildly elevated urinary galactitol, which has been linked to cataract development in the galactosemias. Further, in light of a previously reported role of TKFC in regulating innate antiviral immunity through suppression of MDA5, we speculate that deficiency of TKFC leads to impaired innate immunity in response to viral illness, which may explain the fatal illness observed in the most severely affected individual.


Subject(s)
Cataract/etiology , Cerebellum/abnormalities , Developmental Disabilities/etiology , Mutation , Nervous System Malformations/etiology , Phosphorus-Oxygen Lyases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Alleles , Amino Acid Sequence , Cataract/pathology , Cerebellum/pathology , Child, Preschool , Developmental Disabilities/pathology , Female , Homozygote , Humans , Infant , Male , Nervous System Malformations/pathology , Pedigree , Phenotype , Phosphorylation , Sequence Homology , Exome Sequencing
7.
Am J Hum Genet ; 106(1): 102-111, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31883641

ABSTRACT

Isolated complex III (CIII) deficiencies are among the least frequently diagnosed mitochondrial disorders. Clinical symptoms range from isolated myopathy to severe multi-systemic disorders with early death and disability. To date, we know of pathogenic variants in genes encoding five out of 10 subunits and five out of 13 assembly factors of CIII. Here we describe rare bi-allelic variants in the gene of a catalytic subunit of CIII, UQCRFS1, which encodes the Rieske iron-sulfur protein, in two unrelated individuals. Affected children presented with low CIII activity in fibroblasts, lactic acidosis, fetal bradycardia, hypertrophic cardiomyopathy, and alopecia totalis. Studies in proband-derived fibroblasts showed a deleterious effect of the variants on UQCRFS1 protein abundance, mitochondrial import, CIII assembly, and cellular respiration. Complementation studies via lentiviral transduction and overexpression of wild-type UQCRFS1 restored mitochondrial function and rescued the cellular phenotype, confirming UQCRFS1 variants as causative for CIII deficiency. We demonstrate that mutations in UQCRFS1 can cause mitochondrial disease, and our results thereby expand the clinical and mutational spectrum of CIII deficiencies.


Subject(s)
Alopecia/pathology , Cardiomyopathies/pathology , Electron Transport Complex III/deficiency , Iron-Sulfur Proteins/genetics , Mitochondrial Diseases/pathology , Mutation , Alleles , Alopecia/genetics , Cardiomyopathies/genetics , Child , Electron Transport Complex III/genetics , Humans , Infant , Male , Mitochondrial Diseases/genetics , Pedigree
8.
Am J Hum Genet ; 107(2): 364-373, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32707086

ABSTRACT

We report bi-allelic pathogenic HPDL variants as a cause of a progressive, pediatric-onset spastic movement disorder with variable clinical presentation. The single-exon gene HPDL encodes a protein of unknown function with sequence similarity to 4-hydroxyphenylpyruvate dioxygenase. Exome sequencing studies in 13 families revealed bi-allelic HPDL variants in each of the 17 individuals affected with this clinically heterogeneous autosomal-recessive neurological disorder. HPDL levels were significantly reduced in fibroblast cell lines derived from more severely affected individuals, indicating the identified HPDL variants resulted in the loss of HPDL protein. Clinical presentation ranged from severe, neonatal-onset neurodevelopmental delay with neuroimaging findings resembling mitochondrial encephalopathy to milder manifestation of adolescent-onset, isolated hereditary spastic paraplegia. All affected individuals developed spasticity predominantly of the lower limbs over the course of the disease. We demonstrated through bioinformatic and cellular studies that HPDL has a mitochondrial localization signal and consequently localizes to mitochondria suggesting a putative role in mitochondrial metabolism. Taken together, these genetic, bioinformatic, and functional studies demonstrate HPDL is a mitochondrial protein, the loss of which causes a clinically variable form of pediatric-onset spastic movement disorder.


Subject(s)
Brain Diseases/genetics , Mitochondrial Proteins/genetics , Neurodegenerative Diseases/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Adult , Alleles , Amino Acid Sequence , Child , Female , Humans , Male , Mitochondria/genetics , Pedigree , Phenotype , Young Adult
9.
Genet Med ; 25(6): 100314, 2023 06.
Article in English | MEDLINE | ID: mdl-36305855

ABSTRACT

PURPOSE: This study aimed to define the genotypic and phenotypic spectrum of reversible acute liver failure (ALF) of infancy resulting from biallelic pathogenic TRMU variants and determine the role of cysteine supplementation in its treatment. METHODS: Individuals with biallelic (likely) pathogenic variants in TRMU were studied within an international retrospective collection of de-identified patient data. RESULTS: In 62 individuals, including 30 previously unreported cases, we described 47 (likely) pathogenic TRMU variants, of which 17 were novel, and 1 intragenic deletion. Of these 62 individuals, 42 were alive at a median age of 6.8 (0.6-22) years after a median follow-up of 3.6 (0.1-22) years. The most frequent finding, occurring in all but 2 individuals, was liver involvement. ALF occurred only in the first year of life and was reported in 43 of 62 individuals; 11 of whom received liver transplantation. Loss-of-function TRMU variants were associated with poor survival. Supplementation with at least 1 cysteine source, typically N-acetylcysteine, improved survival significantly. Neurodevelopmental delay was observed in 11 individuals and persisted in 4 of the survivors, but we were unable to determine whether this was a primary or a secondary consequence of TRMU deficiency. CONCLUSION: In most patients, TRMU-associated ALF was a transient, reversible disease and cysteine supplementation improved survival.


Subject(s)
Liver Failure, Acute , Liver Failure , Adolescent , Child , Child, Preschool , Humans , Infant , Young Adult , Acetylcysteine/therapeutic use , Liver Failure/drug therapy , Liver Failure/genetics , Liver Failure, Acute/drug therapy , Liver Failure, Acute/genetics , Mitochondrial Proteins/genetics , Mutation , Retrospective Studies , tRNA Methyltransferases/genetics
10.
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
11.
Ann Neurol ; 91(2): 225-237, 2022 02.
Article in English | MEDLINE | ID: mdl-34954817

ABSTRACT

OBJECTIVE: ATP synthase (ATPase) is responsible for the majority of ATP production. Nevertheless, disease phenotypes associated with mutations in ATPase subunits are extremely rare. We aimed at expanding the spectrum of ATPase-related diseases. METHODS: Whole-exome sequencing in cohorts with 2,962 patients diagnosed with mitochondrial disease and/or dystonia and international collaboration were used to identify deleterious variants in ATPase-encoding genes. Findings were complemented by transcriptional and proteomic profiling of patient fibroblasts. ATPase integrity and activity were assayed using cells and tissues from 5 patients. RESULTS: We present 10 total individuals with biallelic or de novo monoallelic variants in nuclear ATPase subunit genes. Three unrelated patients showed the same homozygous missense ATP5F1E mutation (including one published case). An intronic splice-disrupting alteration in compound heterozygosity with a nonsense variant in ATP5PO was found in one patient. Three patients had de novo heterozygous missense variants in ATP5F1A, whereas another 3 were heterozygous for ATP5MC3 de novo missense changes. Bioinformatics methods and populational data supported the variants' pathogenicity. Immunohistochemistry, proteomics, and/or immunoblotting revealed significantly reduced ATPase amounts in association to ATP5F1E and ATP5PO mutations. Diminished activity and/or defective assembly of ATPase was demonstrated by enzymatic assays and/or immunoblotting in patient samples bearing ATP5F1A-p.Arg207His, ATP5MC3-p.Gly79Val, and ATP5MC3-p.Asn106Lys. The associated clinical profiles were heterogeneous, ranging from hypotonia with spontaneous resolution (1/10) to epilepsy with early death (1/10) or variable persistent abnormalities, including movement disorders, developmental delay, intellectual disability, hyperlactatemia, and other neurologic and systemic features. Although potentially reflecting an ascertainment bias, dystonia was common (7/10). INTERPRETATION: Our results establish evidence for a previously unrecognized role of ATPase nuclear-gene defects in phenotypes characterized by neurodevelopmental and neurodegenerative features. ANN NEUROL 2022;91:225-237.


Subject(s)
Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/genetics , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Neurodevelopmental Disorders/enzymology , Neurodevelopmental Disorders/genetics , Dystonia/enzymology , Dystonia/genetics , Epilepsy/genetics , Genetic Variation , Humans , Mitochondria/genetics , Mitochondrial ADP, ATP Translocases/genetics , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/genetics , Models, Molecular , Mutation , Mutation, Missense , Pedigree , Phenotype , Proteomics , Exome Sequencing
12.
Neuropediatrics ; 54(5): 351-355, 2023 10.
Article in English | MEDLINE | ID: mdl-36603837

ABSTRACT

OBJECTIVE: By loading transfer RNAs with their cognate amino acids, aminoacyl-tRNA synthetases (ARS) are essential for protein translation. Both cytosolic ARS1-deficiencies and mitochondrial ARS2 deficiencies can cause severe diseases. Amino acid supplementation has shown to positively influence the clinical course of four individuals with cytosolic ARS1 deficiencies. We hypothesize that this intervention could also benefit individuals with mitochondrial ARS2 deficiencies. METHODS: This study was designed as a N-of-1 trial. Daily oral L-phenylalanine supplementation was used in a 3-year-old girl with FARS2 deficiency. A period without supplementation was implemented to discriminate the effects of treatment from age-related developments and continuing physiotherapy. Treatment effects were measured through a physiotherapeutic testing battery, including movement assessment battery for children, dynamic gait index, gross motor function measure 66, and quality of life questionnaires. RESULTS: The individual showed clear improvement in all areas tested, especially in gross motor skills, movement abilities, and postural stability. In the period without supplementation, she lost newly acquired motor skills but regained these upon restarting supplementation. No adverse effects and good tolerance of treatment were observed. INTERPRETATION AND CONCLUSION: Our positive results encourage further studies both on L-phenylalanine for other individuals with FARS2 deficiency and the exploration of this treatment rationale for other ARS2 deficiencies. Additionally, treatment costs were relatively low at 1.10 €/day.


Subject(s)
Phenylalanine-tRNA Ligase , Child , Female , Humans , Child, Preschool , Phenylalanine-tRNA Ligase/genetics , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/metabolism , Phenylalanine/metabolism , Quality of Life , Mitochondria/genetics , Mitochondria/metabolism , RNA, Transfer/metabolism , Mitochondrial Proteins/genetics
13.
Arch Orthop Trauma Surg ; 143(5): 2363-2372, 2023 May.
Article in English | MEDLINE | ID: mdl-35438332

ABSTRACT

BACKGROUND: When determining the dimensions of an anterior cruciate ligament (ACL) graft in children, bone age should be considered in addition to the patient's age, gender, and body height. OBJECTIVES: We aimed to determine the relationship between age, gender, and ACL dimensions as well as thickness of growth plate cartilage of the distal femur during puberty. METHODS: We retrospectively analyzed MRI scans of the knee in 131 children (82 girls, 49 boys) aged 6-18 years (mean age: 14.9 ± 2.6 years). ACL length and width as well as thickness of the growth plate cartilage at the distal femoral epiphysis were measured. RESULTS: Mean ACL length increased linearly up to the age of 12 years in females and 14 years in males; thereafter, mean ACL length remained constant. Mean ACL length was largest at the age of 12 to < 13 years (38.18 mm) in females and at 15 to < 16 years (39.38 mm) in males. Mean ACL width increased up to the age of 12 years in girls and 13 years in boys. After the age of 12 years, both the ACL length and width were significantly larger in boys than girls (p = 0.002 and p = 0.045, respectively). Mean thickness of the growth plate cartilage of the distal femur remained stable up to the age of 12 years in girls and 14 years in boys. Thickness of the growth plate cartilage changed most markedly between the age intervals of 11 to < 12 years and 12 to < 13 years in girls and between the age intervals of 13 to < 14 years and 14 to < 15 years in boys. CONCLUSIONS: ACL dimensions depended on both age and gender during the growth period. Measurement of cartilage thickness of the femoral distal growth plate proved to be an objective parameter to assess the maturation stage of local bone. This may be useful for the planning and timing of orthopedic ACL procedures. LEVEL OF EVIDENCE: III-retrospective cohort study.


Subject(s)
Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament , Male , Female , Humans , Child , Anterior Cruciate Ligament/diagnostic imaging , Retrospective Studies , Growth Plate/diagnostic imaging , Tibia , Femur/diagnostic imaging , Knee Joint/diagnostic imaging , Epiphyses/diagnostic imaging , Magnetic Resonance Imaging/methods
14.
Genet Med ; 24(1): 179-191, 2022 01.
Article in English | MEDLINE | ID: mdl-34906456

ABSTRACT

PURPOSE: Haploinsufficiency of PSMD12 has been reported in individuals with neurodevelopmental phenotypes, including developmental delay/intellectual disability (DD/ID), facial dysmorphism, and congenital malformations, defined as Stankiewicz-Isidor syndrome (STISS). Investigations showed that pathogenic variants in PSMD12 perturb intracellular protein homeostasis. Our objective was to further explore the clinical and molecular phenotypic spectrum of STISS. METHODS: We report 24 additional unrelated patients with STISS with various truncating single nucleotide variants or copy-number variant deletions involving PSMD12. We explore disease etiology by assessing patient cells and CRISPR/Cas9-engineered cell clones for various cellular pathways and inflammatory status. RESULTS: The expressivity of most clinical features in STISS is highly variable. In addition to previously reported DD/ID, speech delay, cardiac and renal anomalies, we also confirmed preaxial hand abnormalities as a feature of this syndrome. Of note, 2 patients also showed chilblains resembling signs observed in interferonopathy. Remarkably, our data show that STISS patient cells exhibit a profound remodeling of the mTORC1 and mitophagy pathways with an induction of type I interferon-stimulated genes. CONCLUSION: We refine the phenotype of STISS and show that it can be clinically recognizable and biochemically diagnosed by a type I interferon gene signature.


Subject(s)
Intellectual Disability , Language Development Disorders , Musculoskeletal Abnormalities , Haploinsufficiency , Humans , Intellectual Disability/diagnosis , Language Development Disorders/genetics , Musculoskeletal Abnormalities/genetics , Phenotype
15.
Ann Neurol ; 90(1): 143-158, 2021 07.
Article in English | MEDLINE | ID: mdl-33999436

ABSTRACT

OBJECTIVE: Precursors of peptide hormones undergo posttranslational modifications within the trans-Golgi network (TGN). Dysfunction of proteins involved at different steps of this process cause several complex syndromes affecting the central nervous system (CNS). We aimed to clarify the genetic cause in a group of patients characterized by hypopituitarism in combination with brain atrophy, thin corpus callosum, severe developmental delay, visual impairment, and epilepsy. METHODS: Whole exome sequencing was performed in seven individuals of six unrelated families with these features. Postmortem histopathological and HID1 expression analysis of brain tissue and pituitary gland were conducted in one patient. Functional consequences of the homozygous HID1 variant p.R433W were investigated by Seahorse XF Assay in fibroblasts of two patients. RESULTS: Bi-allelic variants in the gene HID1 domain-containing protein 1 (HID1) were identified in all patients. Postmortem examination confirmed cerebral atrophy with enlarged lateral ventricles. Markedly reduced expression of pituitary hormones was found in pituitary gland tissue. Colocalization of HID1 protein with the TGN was not altered in fibroblasts of patients compared to controls, while the extracellular acidification rate upon stimulation with potassium chloride was significantly reduced in patient fibroblasts compared to controls. INTERPRETATION: Our findings indicate that mutations in HID1 cause an early infantile encephalopathy with hypopituitarism as the leading presentation, and expand the list of syndromic CNS diseases caused by interference of TGN function. ANN NEUROL 2021;90:149-164.


Subject(s)
Brain Diseases/genetics , Epilepsy/genetics , Hypopituitarism/genetics , Alleles , Brain Diseases/pathology , Child, Preschool , Epilepsy/pathology , Female , Humans , Hypopituitarism/pathology , Infant , Male , Pituitary Gland/pathology , Exome Sequencing , Young Adult
16.
Blood ; 136(9): 1033-1043, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32294159

ABSTRACT

Neutropenia and neutrophil dysfunction cause serious infections and inflammatory bowel disease in glycogen storage disease type Ib (GSD-Ib). Our discovery that accumulating 1,5-anhydroglucitol-6-phosphate (1,5AG6P) caused neutropenia in a glucose-6-phosphatase 3 (G6PC3)-deficient mouse model and in 2 rare diseases (GSD-Ib and G6PC3 deficiency) led us to repurpose the widely used antidiabetic drug empagliflozin, an inhibitor of the renal glucose cotransporter sodium glucose cotransporter 2 (SGLT2). Off-label use of empagliflozin in 4 GSD-Ib patients with incomplete response to granulocyte colony-stimulating factor (GCSF) treatment decreased serum 1,5AG and neutrophil 1,5AG6P levels within 1 month. Clinically, symptoms of frequent infections, mucosal lesions, and inflammatory bowel disease resolved, and no symptomatic hypoglycemia was observed. GCSF could be discontinued in 2 patients and tapered by 57% and 81%, respectively, in the other 2. The fluctuating neutrophil numbers in all patients were increased and stabilized. We further demonstrated improved neutrophil function: normal oxidative burst (in 3 of 3 patients tested), corrected protein glycosylation (2 of 2), and normal neutrophil chemotaxis (1 of 1), and bactericidal activity (1 of 1) under treatment. In summary, the glucose-lowering SGLT2 inhibitor empagliflozin, used for type 2 diabetes, was successfully repurposed for treating neutropenia and neutrophil dysfunction in the rare inherited metabolic disorder GSD-Ib without causing symptomatic hypoglycemia. We ascribe this to an improvement in neutrophil function resulting from the reduction of the intracellular concentration of 1,5AG6P.


Subject(s)
Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Glycogen Storage Disease Type I/complications , Hexosephosphates/blood , Neutropenia/drug therapy , Neutrophils/pathology , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Benzhydryl Compounds/adverse effects , Blood Glucose/analysis , Chemotaxis, Leukocyte/drug effects , Child, Preschool , Drug Repositioning , Drug Resistance , Female , Glucosides/adverse effects , Glycogen Storage Disease Type I/blood , Glycogen Storage Disease Type I/immunology , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocytes/chemistry , Humans , Infant, Newborn , Lysosomal-Associated Membrane Protein 2/blood , Male , Neutropenia/blood , Off-Label Use , Respiratory Burst/drug effects , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Young Adult
17.
Mov Disord ; 37(10): 2147-2153, 2022 10.
Article in English | MEDLINE | ID: mdl-36047608

ABSTRACT

BACKGROUND: COQ4 codes for a mitochondrial protein required for coenzyme Q10 (CoQ10 ) biosynthesis. Autosomal recessive COQ4-associated CoQ10 deficiency leads to an early-onset mitochondrial multi-organ disorder. METHODS: In-house exome and genome datasets (n = 14,303) were screened for patients with bi-allelic variants in COQ4. Work-up included clinical characterization and functional studies in patient-derived cell lines. RESULTS: Six different COQ4 variants, three of them novel, were identified in six adult patients from four different families. Three patients had a phenotype of hereditary spastic paraparesis, two sisters showed a predominant cerebellar ataxia, and one patient had mild signs of both. Studies in patient-derived fibroblast lines revealed significantly reduced amounts of COQ4 protein, decreased CoQ10 concentrations, and elevated levels of the metabolic intermediate 6-demethoxyubiquinone. CONCLUSION: We report bi-allelic variants in COQ4 causing an adult-onset ataxia-spasticity spectrum phenotype and a disease course much milder than previously reported. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Cerebellar Ataxia , Mitochondrial Proteins , Ubiquinone , Ataxia/genetics , Cerebellar Ataxia/genetics , Humans , Mitochondrial Diseases , Mitochondrial Proteins/genetics , Muscle Spasticity , Muscle Weakness , Mutation/genetics , Ubiquinone/deficiency , Ubiquinone/genetics , Ubiquinone/metabolism
18.
Amino Acids ; 54(3): 365-383, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34477981

ABSTRACT

BACKGROUND: Accumulating evidence indicates that free amino acids (FAA) might be bioactive compounds with potential immunomodulatory capabilities. However, the FAA composition in human milk is still poorly characterized with respect to its correlation to maternal serum levels and its physiological significance for the infant. Studies addressing the relation of human milk FAA to the infants' intestinal microbiota are still missing. METHODS: As part of a pilot study, maternal serum and breast milk FAA concentrations as well as infant intestinal microbiota (16S rRNA) were determined 2 months after birth. The study cohort consisted of 41 healthy mothers and their term delivered, healthy infants with normal birthweight. The relationship between maternal serum and milk FAA was determined by correlation analyses. Associations between (highly correlated) milk FAA and infant intestinal beta diversity were tested using PERMANOVA, LefSe and multivariate regression models adjusted for common confounders. RESULTS: Seven breast milk FAA correlated significantly with serum concentrations. One of these, threonine showed a negative association with abundance of members of the class Gammaproteobacteria (R2adj = 17.1%, p = 0.006; ß= - 0.441). In addition, on the level of families and genera, threonine explained 23.2% of variation of the relative abundance of Enterobacteriaceae (R2adj; p = 0.001; ß = - 0.504) and 11.1% of variability in the abundance of Escherichia/Shigella (R2adj, p = 0.025; ß = - 0.368), when adjusted for confounders. CONCLUSION: Our study is the first to suggest potential interactions between breast milk FAA and infant gut microbiota composition during early lactation. The results might be indicative of a potential protective role of threonine against members of the Enterobacteriaceae family in breast-fed infants. Still, results are based on correlation analyses and larger cohorts are needed to support the findings and elucidate possible underlying mechanisms to assess the complex interplay between breast milk FAA and infant intestinal microbiota in detail.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Female , Gastrointestinal Microbiome/genetics , Humans , Infant , Milk, Human/chemistry , Milk, Human/metabolism , Milk, Human/microbiology , Pilot Projects , RNA, Ribosomal, 16S/genetics , Threonine/metabolism
19.
J Inherit Metab Dis ; 45(4): 663-681, 2022 07.
Article in English | MEDLINE | ID: mdl-35506430

ABSTRACT

Exome sequencing (ES) in the clinical setting of inborn metabolic diseases (IMDs) has created tremendous improvement in achieving an accurate and timely molecular diagnosis for a greater number of patients, but it still leaves the majority of patients without a diagnosis. In parallel, (personalized) treatment strategies are increasingly available, but this requires the availability of a molecular diagnosis. IMDs comprise an expanding field with the ongoing identification of novel disease genes and the recognition of multiple inheritance patterns, mosaicism, variable penetrance, and expressivity for known disease genes. The analysis of trio ES is preferred over singleton ES as information on the allelic origin (paternal, maternal, "de novo") reduces the number of variants that require interpretation. All ES data and interpretation strategies should be exploited including CNV and mitochondrial DNA analysis. The constant advancements in available techniques and knowledge necessitate the close exchange of clinicians and molecular geneticists about genotypes and phenotypes, as well as knowledge of the challenges and pitfalls of ES to initiate proper further diagnostic steps. Functional analyses (transcriptomics, proteomics, and metabolomics) can be applied to characterize and validate the impact of identified variants, or to guide the genomic search for a diagnosis in unsolved cases. Future diagnostic techniques (genome sequencing [GS], optical genome mapping, long-read sequencing, and epigenetic profiling) will further enhance the diagnostic yield. We provide an overview of the challenges and limitations inherent to ES followed by an outline of solutions and a clinical checklist, focused on establishing a diagnosis to eventually achieve (personalized) treatment.


Subject(s)
Exome , Genomics , DNA, Mitochondrial , Exome/genetics , Genetic Testing/methods , Genomics/methods , Phenotype , Exome Sequencing/methods
20.
Arch Orthop Trauma Surg ; 142(4): 615-624, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33236185

ABSTRACT

PURPOSE: Benign lesions of the proximal femur region, such as simple bone cysts, aneurysmal bone cysts, and fibrous dysplasia, are common in children. Benign lesions may cause pathologic fractures, limb length inequities, and growth disturbances. Differential diagnoses, e.g., malignant bone tumors and osteomyelitis, are sometimes difficult to rule out. OBJECTIVE: We aimed to evaluate outcomes in children with benign lesions of the proximal femur treated with curettage, bone grafting, and plate fixation. METHODS: In this retrospective study, we included 30 children (median age 10.5 years; range 1.1-17.8 years) suffering from bone cysts and tumor-like lesions of the proximal femur region treated between 2002 and 2018. We analyzed plain X-ray images and CT scans in all children and obtained MRI scans in a selected group of children (63.3%). We examined histopathologic biopsy results for all bone lesions before initiating treatment. Surgical management comprised tumor curettage with adjuvant high-speed drilling and allogenic bone grafting supplemented by bone graft substitutes before plate fixation. Median follow-up interval was 87 months (range 24-156 months). We evaluated the healing of lesions according to Capanna's classification and rated functional outcomes according to Merle d'Aubigné and Postel score. RESULTS: Overall, 25 of 30 (83.3%) patients were admitted to hospital because of a pathologic fracture. We diagnosed simple bone cysts in 15 (50.0%) patients, aneurysmal bone cysts in 7 (23.5%) patients, and fibrous dysplasia in 8 (26.5%) patients. Bone consolidation was achieved in 22 of 30 (73.3%) patients after a mean of 5 months (range 3-7 months). The main complication was recurrence of the lesion in 4 of 30 (13.3%) patients. With respect to the Merle d'Aubigné and Postel scores, 17 of 30 (56.7%) patients obtained an excellent result (18 points), while 12 (40.0%) patients had a good result (15-17 points) and only 1 (3.3%) patient had a fair result (14 points). CONCLUSION: Surgical treatment of bone cysts and tumor-like lesions of the proximal femur by local resection or destruction of the lesion, followed by filling the defect with bone graft material and internal stabilization represents a safe and effective treatment option in children. LEVEL OF EVIDENCE: Therapeutic, retrospective comparative study-Level III.


Subject(s)
Bone Cysts , Bone Neoplasms , Fractures, Spontaneous , Adolescent , Bone Cysts/diagnostic imaging , Bone Cysts/surgery , Bone Neoplasms/complications , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/surgery , Bone Transplantation/methods , Child , Child, Preschool , Femur/surgery , Fractures, Spontaneous/diagnostic imaging , Fractures, Spontaneous/etiology , Fractures, Spontaneous/surgery , Humans , Infant , Retrospective Studies , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL