Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Genet ; 60(1): 65-73, 2023 01.
Article in English | MEDLINE | ID: mdl-34872991

ABSTRACT

BACKGROUND: Large-scale mitochondrial DNA deletions (LMD) are a common genetic cause of mitochondrial disease and give rise to a wide range of clinical features. Lack of longitudinal data means the natural history remains unclear. This study was undertaken to describe the clinical spectrum in a large cohort of patients with paediatric disease onset. METHODS: A retrospective multicentre study was performed in patients with clinical onset <16 years of age, diagnosed and followed in seven European mitochondrial disease centres. RESULTS: A total of 80 patients were included. The average age at disease onset and at last examination was 10 and 31 years, respectively. The median time from disease onset to death was 11.5 years. Pearson syndrome was present in 21%, Kearns-Sayre syndrome spectrum disorder in 50% and progressive external ophthalmoplegia in 29% of patients. Haematological abnormalities were the hallmark of the disease in preschool children, while the most common presentations in older patients were ptosis and external ophthalmoplegia. Skeletal muscle involvement was found in 65% and exercise intolerance in 25% of the patients. Central nervous system involvement was frequent, with variable presence of ataxia (40%), cognitive involvement (36%) and stroke-like episodes (9%). Other common features were pigmentary retinopathy (46%), short stature (42%), hearing impairment (39%), cardiac disease (39%), diabetes mellitus (25%) and renal disease (19%). CONCLUSION: Our study provides new insights into the phenotypic spectrum of childhood-onset, LMD-associated syndromes. We found a wider spectrum of more prevalent multisystem involvement compared with previous studies, most likely related to a longer time of follow-up.


Subject(s)
Kearns-Sayre Syndrome , Muscular Diseases , Ophthalmoplegia, Chronic Progressive External , Child, Preschool , Humans , Child , Aged , DNA, Mitochondrial/genetics , Kearns-Sayre Syndrome/epidemiology , Kearns-Sayre Syndrome/genetics , Ophthalmoplegia, Chronic Progressive External/epidemiology , Ophthalmoplegia, Chronic Progressive External/genetics , Muscular Diseases/genetics , Disease Progression
2.
Exp Eye Res ; 232: 109500, 2023 07.
Article in English | MEDLINE | ID: mdl-37178956

ABSTRACT

Primary open-angle glaucoma (POAG) is characterized by optic nerve degeneration and irreversible loss of retinal ganglion cells (RGCs). The pathophysiology is not fully understood. Since RGCs have a high energy demand, suboptimal mitochondrial function may put the survival of these neurons at risk. In the present study, we explored whether mtDNA copy number or mtDNA deletions could reveal a mitochondrial component in POAG pathophysiology. Buffy coat DNA was isolated from EDTA blood of age- and sex-matched study groups, namely POAG patients with high intraocular pressure (IOP) at diagnosis (high tension glaucoma: HTG; n = 97), normal tension glaucoma patients (NTG, n = 37), ocular hypertensive controls (n = 9), and cataract controls (without glaucoma; n = 32), all without remarkable comorbidities. The number of mtDNA copies was assessed through qPCR quantification of the mitochondrial D-loop and nuclear B2M gene. Presence of the common 4977 base pair mtDNA deletion was assessed by a highly sensitive breakpoint PCR. Analysis showed that HTG patients had a lower number of mtDNA copies per nuclear DNA than NTG patients (p-value <0.01, Dunn test) and controls (p-value <0.001, Dunn test). The common 4977 base pair mtDNA deletion was not detected in any of the participants. A lower mtDNA copy number in blood of HTG patients suggests a role for a genetically defined, deficient mtDNA replication in the pathology of HTG. This may cause a low number of mtDNA copies in RGCs, which together with aging and high IOP, may lead to mitochondrial dysfunction, and contribute to glaucoma pathology.


Subject(s)
Glaucoma, Open-Angle , Glaucoma , Low Tension Glaucoma , Humans , Glaucoma, Open-Angle/diagnosis , DNA, Mitochondrial/genetics , DNA Copy Number Variations , Intraocular Pressure , Low Tension Glaucoma/genetics , Mitochondria/genetics
3.
J Inherit Metab Dis ; 44(2): 469-480, 2021 03.
Article in English | MEDLINE | ID: mdl-32857451

ABSTRACT

The aim of this study was to compare the value of serum biomarkers, fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15), with histological analysis of muscle in the diagnosis of mitochondrial disease. We collected 194 serum samples from patients with a suspected or known mitochondrial disease. Biomarkers were analyzed blinded using enzyme-labeled immunosorbent assay. Clinical data were collected using a structured questionnaire. Only 39% of patients with genetically verified mitochondrial disease had mitochondrial pathology in their muscle histology. In contrast, biomarkers were elevated in 62% of patients with genetically verified mitochondrial disease. Those with both biomarkers elevated had a muscle manifesting disorder and a defect affecting mitochondrial DNA expression. If at least one of the biomarkers was induced and the patient had a myopathic disease, a mitochondrial DNA expression disease was the cause with 94% probability. Among patients with biomarker analysis and muscle biopsy taken <12 months apart, a mitochondrial disorder would have been identified in 70% with analysis of FGF21 and GDF15 compared to 50% of patients whom could have been identified with muscle biopsy alone. Muscle findings were nondiagnostic in 72% (children) and 45% (adults). Induction of FGF21 and GDF15 suggest a mitochondrial etiology as an underlying cause of a muscle manifesting disease. Normal biomarker values do not, however, rule out a mitochondrial disorder, especially if the disease does not manifest in muscle. We suggest that FGF21 and GDF15 together should be first-line diagnostic investigations in mitochondrial disease complementing muscle biopsy.


Subject(s)
DNA, Mitochondrial/genetics , Fibroblast Growth Factors/genetics , Growth Differentiation Factor 15/genetics , Mitochondrial Diseases/genetics , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mitochondrial Diseases/blood , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Young Adult
5.
PLoS Genet ; 13(5): e1006809, 2017 May.
Article in English | MEDLINE | ID: mdl-28542170

ABSTRACT

Integrator is an RNA polymerase II (RNAPII)-associated complex that was recently identified to have a broad role in both RNA processing and transcription regulation. Importantly, its role in human development and disease is so far largely unexplored. Here, we provide evidence that biallelic Integrator Complex Subunit 1 (INTS1) and Subunit 8 (INTS8) gene mutations are associated with rare recessive human neurodevelopmental syndromes. Three unrelated individuals of Dutch ancestry showed the same homozygous truncating INTS1 mutation. Three siblings harboured compound heterozygous INTS8 mutations. Shared features by these six individuals are severe neurodevelopmental delay and a distinctive appearance. The INTS8 family in addition presented with neuronal migration defects (periventricular nodular heterotopia). We show that the first INTS8 mutation, a nine base-pair deletion, leads to a protein that disrupts INT complex stability, while the second missense mutation introduces an alternative splice site leading to an unstable messenger. Cells from patients with INTS8 mutations show increased levels of unprocessed UsnRNA, compatible with the INT function in the 3'-end maturation of UsnRNA, and display significant disruptions in gene expression and RNA processing. Finally, the introduction of the INTS8 deletion mutation in P19 cells using genome editing alters gene expression throughout the course of retinoic acid-induced neural differentiation. Altogether, our results confirm the essential role of Integrator to transcriptome integrity and point to the requirement of the Integrator complex in human brain development.


Subject(s)
Developmental Disabilities/genetics , Gene Deletion , Mutation, Missense , Protein Subunits/genetics , RNA, Messenger/metabolism , Adult , Alternative Splicing , Brain/growth & development , Brain/metabolism , Brain/pathology , Cells, Cultured , Child , Developmental Disabilities/diagnosis , Female , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Male , Mutation , Pedigree , Protein Subunits/metabolism , RNA, Messenger/genetics , Syndrome , Transcriptome , Wnt1 Protein
6.
J Med Genet ; 55(1): 21-27, 2018 01.
Article in English | MEDLINE | ID: mdl-29101127

ABSTRACT

BACKGROUND: Leigh syndrome is a phenotypically and genetically heterogeneous mitochondrial disorder. While some genetic defects are associated with well-described phenotypes, phenotype-genotype correlations in Leigh syndrome are not fully explored. OBJECTIVE: We aimed to identify phenotype-genotype correlations in Leigh syndrome in a large cohort of systematically evaluated patients. METHODS: We studied 96 patients with genetically confirmed Leigh syndrome diagnosed and followed in eight European centres specialising in mitochondrial diseases. RESULTS: We found that ataxia, ophthalmoplegia and cardiomyopathy were more prevalent among patients with mitochondrial DNA defects. Patients with mutations in MT-ND and NDUF genes with complex I deficiency shared common phenotypic features, such as early development of central nervous system disease, followed by high occurrence of cardiac and ocular manifestations. The cerebral cortex was affected in patients with NDUF mutations significantly more often than the rest of the cohort. Patients with the m.8993T>G mutation in MT-ATP6 gene had more severe clinical and radiological manifestations and poorer disease outcome compared with patients with the m.8993T>C mutation. CONCLUSION: Our study provides new insights into phenotype-genotype correlations in Leigh syndrome and particularly in patients with complex I deficiency and with defects in the mitochondrial ATP synthase.


Subject(s)
Genetic Association Studies , Leigh Disease/genetics , Cell Nucleus/metabolism , DNA/genetics , DNA, Mitochondrial/genetics , Female , Follow-Up Studies , Humans , Infant , Male , Mutation/genetics , Phenotype
7.
Ann Neurol ; 82(3): 317-330, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28856750

ABSTRACT

Primary and secondary conditions leading to thiamine deficiency have overlapping features in children, presenting with acute episodes of encephalopathy, bilateral symmetric brain lesions, and high excretion of organic acids that are specific of thiamine-dependent mitochondrial enzymes, mainly lactate, alpha-ketoglutarate, and branched chain keto-acids. Undiagnosed and untreated thiamine deficiencies are often fatal or lead to severe sequelae. Herein, we describe the clinical and genetic characterization of 79 patients with inherited thiamine defects causing encephalopathy in childhood, identifying outcome predictors in patients with pathogenic SLC19A3 variants, the most common genetic etiology. We propose diagnostic criteria that will aid clinicians to establish a faster and accurate diagnosis so that early vitamin supplementation is considered. Ann Neurol 2017;82:317-330.


Subject(s)
Thiamine Deficiency/genetics , Adolescent , Age of Onset , Child , Child, Preschool , Female , Humans , Infant , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins , Mutation , Prognosis , Survival Rate , Thiamine Deficiency/mortality , Young Adult
8.
BMC Gastroenterol ; 18(1): 149, 2018 Oct 19.
Article in English | MEDLINE | ID: mdl-30340467

ABSTRACT

BACKGROUND: Gastrointestinal complications are the main cause of death in patients with mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). Available treatments often restore biochemical homeostasis, but fail to cure gastrointestinal symptoms. METHODS: We evaluated the small intestine neuromuscular pathology of an untreated MNGIE patient and two recipients of hematopoietic stem cells, focusing on enteric neurons and glia. Additionally, we evaluated the intestinal neuromuscular pathology in a mouse model of MNGIE treated with hematopoietic stem cell gene therapy. Quantification of muscle wall thickness and ganglion cell density was performed blind to the genotype with ImageJ. Significance of differences between groups was determined by two-tailed Mann-Whitney U test (P < 0.05). RESULTS: Our data confirm that MNGIE presents with muscle atrophy and loss of Cajal cells and CD117/c-kit immunoreactivity in the small intestine. We also show that hematopoietic stem cell transplantation does not benefit human intestinal pathology at least on short-term. CONCLUSIONS: We suggest that hematopoietic stem cell transplantation may be insufficient to restore intestinal neuropathology, especially at later stages of MNGIE. As interstitial Cajal cells and their networks play a key role in development of gastrointestinal dysmotility, alternative therapeutic approaches taking absence of these cells into account could be required.


Subject(s)
Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Intestine, Small/pathology , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Encephalomyopathies/therapy , Adolescent , Animals , Child , Disease Models, Animal , Humans , Interstitial Cells of Cajal/pathology , Mice , Muscular Atrophy/pathology , Young Adult
9.
J Med Genet ; 54(2): 73-83, 2017 02.
Article in English | MEDLINE | ID: mdl-27450679

ABSTRACT

BACKGROUND: Severe, disease-causing germline mitochondrial (mt)DNA mutations are maternally inherited or arise de novo. Strategies to prevent transmission are generally available, but depend on recurrence risks, ranging from high/unpredictable for many familial mtDNA point mutations to very low for sporadic, large-scale single mtDNA deletions. Comprehensive data are lacking for de novo mtDNA point mutations, often leading to misconceptions and incorrect counselling regarding recurrence risk and reproductive options. We aim to study the relevance and recurrence risk of apparently de novo mtDNA point mutations. METHODS: Systematic study of prenatal diagnosis (PND) and recurrence of mtDNA point mutations in families with de novo cases, including new and published data. 'De novo' based on the absence of the mutation in multiple (postmitotic) maternal tissues is preferred, but mutations absent in maternal blood only were also included. RESULTS: In our series of 105 index patients (33 children and 72 adults) with (likely) pathogenic mtDNA point mutations, the de novo frequency was 24.6%, the majority being paediatric. PND was performed in subsequent pregnancies of mothers of four de novo cases. A fifth mother opted for preimplantation genetic diagnosis because of a coexisting Mendelian genetic disorder. The mtDNA mutation was absent in all four prenatal samples and all 11 oocytes/embryos tested. A literature survey revealed 137 de novo cases, but PND was only performed for 9 (including 1 unpublished) mothers. In one, recurrence occurred in two subsequent pregnancies, presumably due to germline mosaicism. CONCLUSIONS: De novo mtDNA point mutations are a common cause of mtDNA disease. Recurrence risk is low. This is relevant for genetic counselling, particularly for reproductive options. PND can be offered for reassurance.


Subject(s)
DNA, Mitochondrial/genetics , Genetic Diseases, Inborn/diagnosis , Maternal Inheritance/genetics , Prenatal Diagnosis , Adult , Child , Female , Genetic Counseling , Humans , Male , Oocytes/metabolism , Point Mutation/genetics , Pregnancy , Preimplantation Diagnosis
10.
J Pediatr ; 182: 371-374.e2, 2017 03.
Article in English | MEDLINE | ID: mdl-28081892

ABSTRACT

Whole-exome sequencing identified multiple genetic causes in 2 infants with heterogeneous disease. Three gene defects in the first patient explained all symptoms, but manifestations were overlapping (blended phenotype). Two gene defects in the second patient explained nonoverlapping symptoms (composite phenotype). Whole-exome sequencing rapidly and comprehensively resolves heterogeneous genetic disease.


Subject(s)
Congenital Abnormalities/genetics , Genetic Diseases, Inborn/diagnosis , Mutation , Sequence Analysis, DNA/methods , Amidohydrolases/genetics , Carboxylic Ester Hydrolases/genetics , Congenital Abnormalities/diagnosis , Exome/genetics , Genetic Testing/methods , Genomics , Genotype , Humans , Infant , Membrane Proteins/genetics , Microtubule-Associated Proteins , Mutagenicity Tests , Phenotype , Receptors, Peptide/genetics , Sensitivity and Specificity , Severity of Illness Index
11.
Am J Physiol Regul Integr Comp Physiol ; 312(5): R689-R701, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28179228

ABSTRACT

Muscle weakness and exercise intolerance negatively affect the quality of life of patients with mitochondrial myopathy. Short-term dietary nitrate supplementation has been shown to improve exercise performance and reduce oxygen cost of exercise in healthy humans and trained athletes. We investigated whether 1 wk of dietary inorganic nitrate supplementation decreases the oxygen cost of exercise and improves mitochondrial function in patients with mitochondrial myopathy. Ten patients with mitochondrial myopathy (40 ± 5 yr, maximal whole body oxygen uptake = 21.2 ± 3.2 ml·min-1·kg body wt-1, maximal work load = 122 ± 26 W) received 8.5 mg·kg body wt-1·day-1 inorganic nitrate (~7 mmol) for 8 days. Whole body oxygen consumption at 50% of the maximal work load, in vivo skeletal muscle oxidative capacity (evaluated from postexercise phosphocreatine recovery using 31P-magnetic resonance spectroscopy), and ex vivo mitochondrial oxidative capacity in permeabilized skinned muscle fibers (measured with high-resolution respirometry) were determined before and after nitrate supplementation. Despite a sixfold increase in plasma nitrate levels, nitrate supplementation did not affect whole body oxygen cost during submaximal exercise. Additionally, no beneficial effects of nitrate were found on in vivo or ex vivo muscle mitochondrial oxidative capacity. This is the first time that the therapeutic potential of dietary nitrate for patients with mitochondrial myopathy was evaluated. We conclude that 1 wk of dietary nitrate supplementation does not reduce oxygen cost of exercise or improve mitochondrial function in the group of patients tested.


Subject(s)
Exercise , Mitochondria, Muscle/metabolism , Mitochondrial Myopathies/drug therapy , Mitochondrial Myopathies/physiopathology , Nitrates/administration & dosage , Oxygen Consumption/drug effects , Administration, Oral , Adult , Aged , Exercise Tolerance/drug effects , Female , Humans , Male , Middle Aged , Mitochondria, Muscle/drug effects , Muscle Strength/drug effects , Psychomotor Performance/drug effects , Treatment Outcome , Young Adult
12.
J Neuroophthalmol ; 37(4): 371-381, 2017 12.
Article in English | MEDLINE | ID: mdl-28991104

ABSTRACT

Leber hereditary optic neuropathy (LHON) is currently estimated as the most frequent mitochondrial disease (1 in 27,000-45,000). Its molecular pathogenesis and natural history is now fairly well understood. LHON also is the first mitochondrial disease for which a treatment has been approved (idebenone-Raxone, Santhera Pharmaceuticals) by the European Medicine Agency, under exceptional circumstances because of the rarity and severity of the disease. However, what remains unclear includes the optimal target population, timing, dose, and frequency of administration of idebenone in LHON due to lack of accepted definitions, criteria, and general guidelines for the clinical management of LHON. To address these issues, a consensus conference with a panel of experts from Europe and North America was held in Milan, Italy, in 2016. The intent was to provide expert consensus statements for the clinical and therapeutic management of LHON based on the currently available evidence. We report the conclusions of this conference, providing the guidelines for clinical and therapeutic management of LHON.


Subject(s)
Consensus , Disease Management , Ophthalmology , Optic Atrophy, Hereditary, Leber/drug therapy , Societies, Medical , Ubiquinone/analogs & derivatives , Antioxidants/therapeutic use , Congresses as Topic , Humans , International Cooperation , Ubiquinone/therapeutic use
13.
Am J Hum Genet ; 92(5): 774-80, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23643384

ABSTRACT

Inherited white-matter disorders are a broad class of diseases for which treatment and classification are both challenging. Indeed, nearly half of the children presenting with a leukoencephalopathy remain without a specific diagnosis. Here, we report on the application of high-throughput genome and exome sequencing to a cohort of ten individuals with a leukoencephalopathy of unknown etiology and clinically characterized by hypomyelination with brain stem and spinal cord involvement and leg spasticity (HBSL), as well as the identification of compound-heterozygous and homozygous mutations in cytoplasmic aspartyl-tRNA synthetase (DARS). These mutations cause nonsynonymous changes to seven highly conserved amino acids, five of which are unchanged between yeast and man, in the DARS C-terminal lobe adjacent to, or within, the active-site pocket. Intriguingly, HBSL bears a striking resemblance to leukoencephalopathy with brain stem and spinal cord involvement and elevated lactate (LBSL), which is caused by mutations in the mitochondria-specific DARS2, suggesting that these two diseases might share a common underlying molecular pathology. These findings add to the growing body of evidence that mutations in tRNA synthetases can cause a broad range of neurologic disorders.


Subject(s)
Aspartate-tRNA Ligase/genetics , Leukoencephalopathies/genetics , Models, Molecular , Muscle Spasticity/genetics , Protein Conformation , Aspartate-tRNA Ligase/chemistry , Brain Stem/pathology , Crystallography, X-Ray , Humans , Leg/pathology , Leukoencephalopathies/pathology , Mutation/genetics , Spinal Cord/pathology
14.
Brain ; 138(Pt 10): 2847-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26264513

ABSTRACT

Haematopoietic stem cell transplantation has been proposed as treatment for mitochondrial neurogastrointestinal encephalomyopathy, a rare fatal autosomal recessive disease due to TYMP mutations that result in thymidine phosphorylase deficiency. We conducted a retrospective analysis of all known patients suffering from mitochondrial neurogastrointestinal encephalomyopathy who underwent allogeneic haematopoietic stem cell transplantation between 2005 and 2011. Twenty-four patients, 11 males and 13 females, median age 25 years (range 10-41 years) treated with haematopoietic stem cell transplantation from related (n = 9) or unrelated donors (n = 15) in 15 institutions worldwide were analysed for outcome and its associated factors. Overall, 9 of 24 patients (37.5%) were alive at last follow-up with a median follow-up of these surviving patients of 1430 days. Deaths were attributed to transplant in nine (including two after a second transplant due to graft failure), and to mitochondrial neurogastrointestinal encephalomyopathy in six patients. Thymidine phosphorylase activity rose from undetectable to normal levels (median 697 nmol/h/mg protein, range 262-1285) in all survivors. Seven patients (29%) who were engrafted and living more than 2 years after transplantation, showed improvement of body mass index, gastrointestinal manifestations, and peripheral neuropathy. Univariate statistical analysis demonstrated that survival was associated with two defined pre-transplant characteristics: human leukocyte antigen match (10/10 versus <10/10) and disease characteristics (liver disease, history of gastrointestinal pseudo-obstruction or both). Allogeneic haematopoietic stem cell transplantation can restore thymidine phosphorylase enzyme function in patients with mitochondrial neurogastrointestinal encephalomyopathy and improve clinical manifestations of mitochondrial neurogastrointestinal encephalomyopathy in the long term. Allogeneic haematopoietic stem cell transplantation should be considered for selected patients with an optimal donor.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Intestinal Pseudo-Obstruction/surgery , Mitochondrial Encephalomyopathies/surgery , Treatment Outcome , Adolescent , Adult , Body Weight , Brain/pathology , Child , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Muscular Dystrophy, Oculopharyngeal , Neural Conduction/physiology , Neurologic Examination , Neutrophils , Ophthalmoplegia/congenital , Retrospective Studies , Survival Analysis , Thymidine Phosphorylase/metabolism , Transplantation, Homologous/methods , Young Adult
15.
Am J Hum Genet ; 91(3): 533-40, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22939636

ABSTRACT

Polymicrogyria is a malformation of the developing cerebral cortex caused by abnormal organization and characterized by many small gyri and fusion of the outer molecular layer. We have identified autosomal-recessive mutations in RTTN, encoding Rotatin, in individuals with bilateral diffuse polymicrogyria from two separate families. Rotatin determines early embryonic axial rotation, as well as anteroposterior and dorsoventral patterning in the mouse. Human Rotatin has recently been identified as a centrosome-associated protein. The Drosophila melanogaster homolog of Rotatin, Ana3, is needed for structural integrity of centrioles and basal bodies and maintenance of sensory neurons. We show that Rotatin colocalizes with the basal bodies at the primary cilium. Cultured fibroblasts from affected individuals have structural abnormalities of the cilia and exhibit downregulation of BMP4, WNT5A, and WNT2B, which are key regulators of cortical patterning and are expressed at the cortical hem, the cortex-organizing center that gives rise to Cajal-Retzius (CR) neurons. Interestingly, we have shown that in mouse embryos, Rotatin colocalizes with CR neurons at the subpial marginal zone. Knockdown experiments in human fibroblasts and neural stem cells confirm a role for RTTN in cilia structure and function. RTTN mutations therefore link aberrant ciliary function to abnormal development and organization of the cortex in human individuals.


Subject(s)
Carrier Proteins/genetics , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Cilia/physiology , Malformations of Cortical Development/genetics , Adolescent , Cell Cycle Proteins , Cell Line , Child , Female , Gene Knockout Techniques , Genes, Recessive , Humans , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/diagnosis , Mutation
16.
Am J Hum Genet ; 88(5): 657-63, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21549339

ABSTRACT

The progressive myoclonus epilepsies (PMEs) are a group of predominantly recessive disorders that present with action myoclonus, tonic-clonic seizures, and progressive neurological decline. Many PMEs have similar clinical presentations yet are genetically heterogeneous, making accurate diagnosis difficult. A locus for PME was mapped in a consanguineous family with a single affected individual to chromosome 17q21. An identical-by-descent, homozygous mutation in GOSR2 (c.430G>T, p.Gly144Trp), a Golgi vesicle transport gene, was identified in this patient and in four apparently unrelated individuals. A comparison of the phenotypes in these patients defined a clinically distinct PME syndrome characterized by early-onset ataxia, action myoclonus by age 6, scoliosis, and mildly elevated serum creatine kinase. This p.Gly144Trp mutation is equivalent to a loss of function and results in failure of GOSR2 protein to localize to the cis-Golgi.


Subject(s)
Mutation , Myoclonic Epilepsies, Progressive/genetics , Qb-SNARE Proteins/genetics , Spinocerebellar Degenerations/genetics , Amino Acid Sequence , Child , Consanguinity , Female , Genes, Recessive , Genetic Markers , Golgi Apparatus/genetics , Homozygote , Humans , Lod Score , Male , Molecular Sequence Data , Myoclonic Epilepsies, Progressive/pathology , Pedigree , Phenotype , SNARE Proteins/genetics , Spinocerebellar Degenerations/pathology
17.
Brain ; 136(Pt 4): 1146-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23449775

ABSTRACT

We previously identified a homozygous mutation in the Golgi SNAP receptor complex 2 gene (GOSR2) in six patients with progressive myoclonus epilepsy. To define the syndrome better we analysed the clinical and electrophysiological phenotype in 12 patients with GOSR2 mutations, including six new unrelated subjects. Clinical presentation was remarkably similar with early onset ataxia (average 2 years of age), followed by myoclonic seizures at the average age of 6.5 years. Patients developed multiple seizure types, including generalized tonic clonic seizures, absence seizures and drop attacks. All patients developed scoliosis by adolescence, making this an important diagnostic clue. Additional skeletal deformities were present, including pes cavus in four patients and syndactyly in two patients. All patients had elevated serum creatine kinase levels (median 734 IU) in the context of normal muscle biopsies. Electroencephalography revealed pronounced generalized spike and wave discharges with a posterior predominance and photosensitivity in all patients, with focal EEG features seen in seven patients. The disease course showed a relentless decline; patients uniformly became wheelchair bound (mean age 13 years) and four had died during their third or early fourth decade. All 12 cases had the same variant (c.430G>T, G144W) and haplotype analyses confirmed a founder effect. The cases all came from countries bounding the North Sea, extending to the coastal region of Northern Norway. 'North Sea' progressive myoclonus epilepsy has a homogeneous clinical presentation and relentless disease course allowing ready identification from the other progressive myoclonus epilepsies.


Subject(s)
Mutation , Myoclonic Epilepsies, Progressive/genetics , Myoclonic Epilepsies, Progressive/physiopathology , Phenotype , Qb-SNARE Proteins/genetics , Adolescent , Adult , Ataxia/genetics , Ataxia/physiopathology , Child , Electroencephalography , Europe , Female , Humans , Male , Mutation/genetics , Myoclonic Epilepsies, Progressive/mortality , North Sea , Young Adult
18.
Am J Med Genet A ; 161A(9): 2376-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23873601

ABSTRACT

Mutations in FLNA (Filamin A, OMIM 300017) cause X-linked periventricular nodular heterotopia (XL-PNH). XL-PNH-associated mutations are considered lethal in hemizygous males. However, a few males with unusual mutations (including distal truncating and hypomorphic missense mutations), and somatic mosaicism have been reported to survive past infancy. Two brothers had an atypical presentation with failure to thrive and distinct facial appearance including hypertelorism. Evaluations of these brothers and their affected cousin showed systemic involvement including severe intestinal malfunction, malrotation, congenital short bowel, PNH, pyloric stenosis, wandering spleen, patent ductus arteriosus, atrial septal defect, inguinal hernia, and vesicoureteral reflux. The unanticipated finding of PNH led to FLNA testing and subsequent identification of a novel no-stop FLNA mutation (c.7941_7942delCT, p.(*2648Serext*100)). Western blotting and qRT-PCR of patients' fibroblasts showed diminished levels of protein and mRNA. This FLNA mutation, the most distal reported so far, causes in females classical XL-PNH, but in males an unusual, multi-organ phenotype, providing a unique insight into the FLNA-associated phenotypes.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Filamins/genetics , Mutation, Missense , Base Sequence , Brain/pathology , Facies , Female , Genotype , Humans , Infant , Magnetic Resonance Imaging , Male , Pedigree , Periventricular Nodular Heterotopia/diagnosis , Periventricular Nodular Heterotopia/genetics , Phenotype , Radiography , Spleen/diagnostic imaging , Spleen/pathology
19.
Eur J Med Genet ; 66(6): 104746, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36967043

ABSTRACT

Phelan-McDermid syndrome (PMS) is a 22q13.3 deletion syndrome that presents with a disturbed development, neurological and psychiatric characteristics, and sometimes other comorbidities like seizures. The epilepsy manifests itself in a variety of seizure semiologies. Further diagnostics using electroencephalogram (EEG) and brain magnetic resonance imaging (MRI) are important in conjunction with the clinical picture of the seizures to decide whether anticonvulsant therapy is necessary. As part of the development of European consensus guidelines we focussed on the prevalence and semiology of epileptic seizures in PMS associated with a pathogenic variant in the SHANK3 gene or the 22q13 deletion involving SHANK3, in order to then be able to make recommendations regarding diagnosis and therapy.


Subject(s)
Chromosome Disorders , Epilepsy , Humans , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosome Deletion , Epilepsy/diagnosis , Epilepsy/genetics , Seizures/genetics , Chromosomes, Human, Pair 22/genetics
20.
Front Ophthalmol (Lausanne) ; 3: 1309836, 2023.
Article in English | MEDLINE | ID: mdl-38983060

ABSTRACT

Introduction: Primary open-angle glaucoma (POAG) is a characteristic optic neuropathy, caused by degeneration of the optic nerve-forming neurons, the retinal ganglion cells (RGCs). High intraocular pressure (IOP) and aging have been identified as major risk factors; yet the POAG pathophysiology is not fully understood. Since RGCs have high energy requirements, mitochondrial dysfunction may put the survivability of RGCs at risk. We explored in buffy coat DNA whether mtDNA variants and their distribution throughout the mtDNA could be risk factors for POAG. Methods: The mtDNA was sequenced from age- and sex-matched study groups, being high tension glaucoma (HTG, n=71), normal tension glaucoma patients (NTG, n=33), ocular hypertensive subjects (OH, n=7), and cataract controls (without glaucoma; n=30), all without remarkable comorbidities. Results: No association was found between the number of mtDNA variants in genes encoding proteins, tRNAs, rRNAs, and in non-coding regions in the different study groups. Next, variants that controls shared with the other groups were discarded. A significantly higher number of exclusive variants was observed in the D-loop region for the HTG group (~1.23 variants/subject), in contrast to controls (~0.35 variants/subject). In the D-loop, specifically in the 7S DNA sub-region within the Hypervariable region 1 (HV1), we found that 42% of the HTG and 27% of the NTG subjects presented variants, while this was only 14% for the controls and OH subjects. As we have previously reported a reduction in mtDNA copy number in HTG, we analysed if specific D-loop variants could explain this. While the majority of glaucoma patients with the exclusive D-loop variants m.72T>C, m.16163 A>G, m.16186C>T, m.16298T>C, and m.16390G>A presented a mtDNA copy number below controls median, no significant association between these variants and low copy number was found and their possible negative role in mtDNA replication remains uncertain. Approximately 38% of the HTG patients with reduced copy number did not carry any exclusive D-loop or other mtDNA variants, which indicates that variants in nuclear-encoded mitochondrial genes, environmental factors, or aging might be involved in those cases. Conclusion: In conclusion, we found that variants in the D-loop region may be a risk factor in a subgroup of POAG, possibly by affecting mtDNA replication.

SELECTION OF CITATIONS
SEARCH DETAIL