Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Orphanet J Rare Dis ; 14(1): 243, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31699126

ABSTRACT

BACKGROUND: Barth syndrome (BTHS, OMIM 302060) is a rare, life-threatening, x-linked genetic disorder that occurs almost exclusively in males and is characterized by cardiomyopathy, neutropenia, skeletal muscle myopathy primarily affecting larger muscles, and shorter stature in youth. A greater number of individuals with BTHS are now surviving into adulthood due to advancements in diagnosis and disease management. Given these improvements in life expectancy, understanding the disease experience over time has become increasingly important to individuals with the condition, treatment developers, and regulatory agencies. A study was conducted to explore the experience of BTHS from the perspective of adult males at least 35 years of age with the condition via in-depth qualitative interviews. RESULTS: Findings showed that adults with BTHS experienced a variety of signs/symptoms with variable onset and severity throughout their lives, the most frequently reported being the symptoms of tiredness, muscle weakness, and a fast and/or irregular heart rate, and the sign of short stature in youth. These signs/symptoms negatively impacted individuals' emotional, physical, social, and role functioning. Tiredness and weakness impacted some individuals' physical functioning from an early age and into adulthood. These symptoms generally worsened over time, increasingly interfering with individuals' ability to fully participate in paid and unpaid labor and to partake in family and leisure activities. CONCLUSIONS: This research complements recent studies characterizing the potentially degenerative and progressive nature of BTHS and can encourage future research into the natural history and progression of BTHS in untreated individuals. Participants' interview responses revealed a range of symptoms and the potential for multiple impacts on individuals' physical, social, emotional, and role functioning as a result of BTHS symptoms, yet also revealed variability in severity of experience as well as the possibility of resilience and adaptation to the condition.


Subject(s)
Barth Syndrome/pathology , Life Change Events , Adult , Humans , Interviews as Topic , Male , Middle Aged
2.
Orphanet J Rare Dis ; 13(1): 92, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29903031

ABSTRACT

BACKGROUND: Pontocerebellar hypoplasia (PCH) describes a rare, heterogeneous group of neurodegenerative disorders mainly with a prenatal onset. Patients have severe hypoplasia or atrophy of cerebellum and pons, with variable involvement of supratentorial structures, motor and cognitive impairments. Based on distinct clinical features and genetic causes, current classification comprises 11 types of PCH. MAIN TEXT: In this review we describe the clinical, neuroradiological and genetic characteristics of the different PCH subtypes, summarize the differential diagnosis and reflect on potential disease mechanisms in PCH. Seventeen PCH-related genes are now listed in the OMIM database, most of them have a function in RNA processing or translation. It is unknown why defects in these apparently ubiquitous processes result in a brain-specific phenotype. CONCLUSIONS: Many new PCH related genes and phenotypes have been described due to the appliance of next generation sequencing techniques. By including such a broad range of phenotypes, including non-degenerative and postnatal onset disorders, the current classification gives rise to confusion. Despite the discovery of new pathways involved in PCH, treatment is still symptomatic. However, correct diagnosis of PCH is important to provide suitable care and counseling regarding prognosis, and offer appropriate (prenatal) genetic testing to families.


Subject(s)
Cerebellar Diseases/diagnosis , Cerebellar Diseases/genetics , Humans , Neurology , Phenotype
3.
J Inherit Metab Dis ; 39(1): 93-106, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26287655

ABSTRACT

INTRODUCTION: We describe the natural history of patients with a Zellweger spectrum disorder (ZSD) surviving into adulthood. METHODS: Retrospective cohort study in patients with a genetically confirmed ZSD. RESULTS: All patients (n = 19; aged 16-35 years) had a follow-up period of 1-24.4 years (mean 16 years). Seven patients had a progressive disease course, while 12 remained clinically stable during follow-up. Disease progression usually manifests in adolescence as a gait disorder, caused by central and/or peripheral nervous system involvement. Nine were capable of living a partly independent life with supported employment. Systematic MRI review revealed T2 hyperintense white matter abnormalities in the hilus of the dentate nucleus and/or peridentate region in nine out of 16 patients. Biochemical analyses in blood showed abnormal peroxisomal biomarkers in all patients in infancy and childhood, whereas in adolescence/adulthood we observed normalization of some metabolites. CONCLUSIONS: The patients described here represent a distinct subgroup within the ZSDs who survive into adulthood. Most remain stable over many years. Disease progression may occur and is mainly due to cerebral and cerebellar white matter abnormalities, and peripheral neuropathy.


Subject(s)
White Matter/pathology , Zellweger Syndrome/pathology , Adolescent , Adult , Biomarkers/metabolism , Disease Progression , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging/methods , Male , Retrospective Studies , White Matter/metabolism , Young Adult , Zellweger Syndrome/metabolism
4.
Hum Mutat ; 35(12): 1418-26, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205138

ABSTRACT

A mutation update on the nebulin gene (NEB) is necessary because of recent developments in analysis methodology, the identification of increasing numbers and novel types of variants, and a widening in the spectrum of clinical and histological phenotypes associated with this gigantic, 183 exons containing gene. Recessive pathogenic variants in NEB are the major cause of nemaline myopathy (NM), one of the most common congenital myopathies. Moreover, pathogenic NEB variants have been identified in core-rod myopathy and in distal myopathies. In this update, we present the disease-causing variants in NEB in 159 families, 143 families with NM, and 16 families with NM-related myopathies. Eighty-eight families are presented here for the first time. We summarize 86 previously published and 126 unpublished variants identified in NEB. Furthermore, we have analyzed the NEB variants deposited in the Exome Variant Server (http://evs.gs.washington.edu/EVS/), identifying that pathogenic variants are a minor fraction of all coding variants (∼7%). This indicates that nebulin tolerates substantial changes in its amino acid sequence, providing an explanation as to why variants in such a large gene result in relatively rare disorders. Lastly, we discuss the difficulties of drawing reliable genotype-phenotype correlations in NEB-associated disease.


Subject(s)
Muscle Proteins/genetics , Muscular Diseases/genetics , Mutation , Alternative Splicing , Animals , Chromosomes, Human, Pair 2 , Databases, Genetic , Exons , Genotype , Humans , Models, Animal , Muscular Diseases/classification , Phenotype
5.
Orphanet J Rare Dis ; 9: 70, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24886362

ABSTRACT

INTRODUCTION: Pontocerebellar hypoplasia Type 2 (PCH2) is a rare autosomal recessive condition, defined on MRI by a small cerebellum and ventral pons. Clinical features are severe developmental delay, microcephaly and dyskinesia.Ninety percent carry a p.A307S mutation in the TSEN54-gene. Our aim was to describe the natural course including neurological and developmental features and other aspects of care in a homogeneous group of PCH2 patients all carrying the p.A307S mutation. PATIENTS AND METHODS: Patients were recruited via the German patients' organizations. Inclusion criteria were imaging findings of PCH2 and a p.A307S mutation. Data were collected using medical reports and patient questionnaires discussed in a standardized telephone interview. RESULTS: Thirty-three patients were included. When considering survival until age 11 years, 53% of children had died Weight, length and head circumference, mostly in the normal range at birth, became abnormal, especially head circumference (-5.58 SD at age 5 yrs). Neurologic symptoms: Choreathetosis was present in 88% (62% with pyramidal signs), 12% had pure spasticity. Epileptic seizures were manifest in 82%, status epilepticus in 39%. Non-epileptic dystonic attacks occurred in 33%. General symptoms: feeding difficulties were recorded in 100%, sleep disorder in 96%, apneas in 67% and recurrent infections in 52%; gastroesophageal reflux disease was diagnosed in 73%, 67% got percutaneous endoscopic gastrostomy and 36% a Nissen-fundoplication. Neurodevelopmental data: All children made progress, but on a low level: such as fixing and following with the eyes was seen in 76%, attempting to grasp objects (76%), moderate head control (73%), social smile (70%), rolling from prone to supine (58%), and sitting without support (9%). Ten percent lost achieved abilities on follow-up. The presence of prenatal symptoms did not correlate with outcome. CONCLUSION: Phenotype of this genetically homogeneous group of PCH2 children was severe with reduced survival, but compatible with some developmental progress. Our data support the hypothesis of an early onset degeneration which thereafter stabilizes.


Subject(s)
Olivopontocerebellar Atrophies/physiopathology , Endoribonucleases/genetics , Humans , Mutation , Olivopontocerebellar Atrophies/genetics
6.
Am J Med Genet C Semin Med Genet ; 166C(2): 173-83, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24924738

ABSTRACT

Pontocerebellar hypoplasia (PCH) is a clinically and genetically heterogeneous group of autosomal recessively inherited neurodevelopmental disorders. Following the rapidly increasing number of genes identified in different subtypes, the clinical spectrum has been broadened to completely different neurological phenotypes. In this review we will address the clinical picture, neuroradiological, pathoanatomic, and genetic findings in the currently known PCH subtypes.


Subject(s)
Cerebellar Diseases/classification , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Cerebellum/physiopathology , Diagnosis, Differential , Humans , Magnetic Resonance Imaging , Neuroimaging , Phenotype
7.
Orphanet J Rare Dis ; 9: 23, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24524299

ABSTRACT

BACKGROUND: Pontocerebellar hypoplasia (PCH) represents a group of neurodegenerative disorders with prenatal onset. Eight subtypes have been described thus far (PCH1-8) based on clinical and genetic features. Common characteristics include hypoplasia and atrophy of the cerebellum, variable pontine atrophy, and severe mental and motor impairments. PCH1 is distinctly characterized by the combination with degeneration of spinal motor neurons. Recently, mutations in the exosome component 3 gene (EXOSC3) have been identified in approximately half of the patients with PCH subtype 1. METHODS: We selected a cohort of 99 PCH patients (90 families) tested negative for mutations in the TSEN genes, RARS2, VRK1 and CASK. Patients in this cohort were referred with a tentative diagnose PCH type 1, 2, 4, 7 or unclassified PCH. Genetic analysis of the EXOSC3 gene was performed using Sanger sequencing. Clinical data, MR images and autopsy reports of patients positive for EXOSC3 mutations were analyzed. RESULTS: EXOSC3 mutations were found in twelve families with PCH subtype 1, and were not found in patients with other PCH subtypes. Identified mutations included a large deletion, nonsense and missense mutations. Examination of clinical data reveals a prolonged disease course in patients with a homozygous p.D132A mutation. MRI shows variable pontine hypoplasia in EXOSC3 mediated PCH, where the pons is largely preserved in patients with a homozygous p.D132A mutation, but attenuated in patients with other mutations. Additionally, bilateral cerebellar cysts were found in patients compound heterozygous for a p.D132A mutation and a nonsense allele. CONCLUSIONS: EXOSC3 mediated PCH shows clear genotype-phenotype correlations. A homozygous p.D132A mutation leads to PCH with possible survival into early puberty, and preservation of the pons. Compound heterozygosity for a p.D132A mutation and a nonsense or p.Y109N allele, a homozygous p.G31A mutation or a p.G135E mutation causes a more rapidly progressive course leading to death in infancy and attenuation of the ventral pons.Our findings imply a clear correlation between genetic mutation and clinical outcome in EXOSC3 mediated PCH, including variable involvement of the pons.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , Olivopontocerebellar Atrophies/genetics , RNA-Binding Proteins/genetics , Brain/pathology , Female , Genetic Association Studies , Humans , Male , Mutation
8.
Eur J Paediatr Neurol ; 18(3): 434-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24485946

ABSTRACT

BACKGROUND: Pontine Tegmental Cap Dysplasia (PTCD) is a recently described hindbrain malformation presenting hypoplasia of the ventral pons, and a "pontine tegmental cap". Previous DTI studies identified ectopic transversely oriented nerve fibres in the cap, and absence of transverse fibre bundles in the ventral pons, characterizing PTCD as an embryonic axon guidance defect. A new case with relatively mild symptoms was investigated to identify fibre tracts in the tegmental cap by tracking their connections. In the process a new bilateral ectopic fibre tract was found. METHODS: Routine T1- and T2 weighted images and Diffusion Tensor Imaging (DTI) data were obtained on a 3 T MR scanner. Fractional Anisotropy maps colour coded for orientation were generated. High Angular Resolution Diffusion Imaging (HARDI) data were used for reconstructing maps denoting multiple fibre orientations (i.e. fibre crossings) per voxel through which accurate fibre tracking was performed. RESULTS INTERPRETATION: Peripontine arcuate fibres were identified, representing a second structural abnormality not previously recorded in PTCD.


Subject(s)
Axons/pathology , Malformations of Cortical Development/pathology , Pons/pathology , Child , Diffusion Magnetic Resonance Imaging/methods , Humans , Image Processing, Computer-Assisted/methods , Male , Malformations of Cortical Development/diagnosis
9.
Orphanet J Rare Dis ; 8: 174, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24172221

ABSTRACT

BACKGROUND: To describe the neurologic profiles of Rhizomelic chondrodysplasia punctata (RCDP); a peroxisomal disorder clinically characterized by skeletal abnormalities, congenital cataracts, severe growth and developmental impairments and immobility of joints. Defective plasmalogen biosynthesis is the main biochemical feature. METHODS: Observational study including review of clinical and biochemical abnormalities, genotype, presence of seizures and neurophysiological studies of a cohort of 16 patients with RCDP. RESULTS: Patients with the severe phenotype nearly failed to achieve any motor or cognitive skills, whereas patients with the milder phenotype had profound intellectual disability but were able to walk and had verbal communication skills. Eighty-eight percent of patients developed epileptic seizures. The age of onset paralleled the severity of the clinical and biochemical phenotype. Myoclonic jerks, followed by atypical absences were most frequently observed. All patients with clinical seizures had interictal encephalographic evidence of epilepsy. Visual evoked (VEP) and brain auditory potential (BAEP) studies showed initial normal latency times in 93% of patients. Deterioration of VEP occurred in a minority in both the severe and the milder phenotype. BAEP and somatosensory evoked potentials (SSEP) were more likely to become abnormal in the severe phenotype. Plasmalogens were deficient in all patients. In the milder phenotype levels of plasmalogens were significantly higher in erythrocytes than in the severe phenotype. Phytanic acid levels ranged from normal to severely increased, but had no relation with the neurological phenotype. CONCLUSION: Neurodevelopmental deficits and age-related occurrence of seizures are characteristic of RCDP and are related to the rest-activity in plasmalogen biosynthesis. Evoked potential studies are more likely to become abnormal in the severe phenotype, but are of no predictive value in single cases of RCDP.


Subject(s)
Chondrodysplasia Punctata, Rhizomelic/physiopathology , Adolescent , Adult , Child, Preschool , Evoked Potentials, Somatosensory/physiology , Evoked Potentials, Visual/physiology , Female , Humans , Male
10.
Brain ; 136(Pt 1): 282-93, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23365102

ABSTRACT

A cardioskeletal myopathy with onset and death in infancy, morphological features of muscle type I hypotrophy with myofibrillar disorganization and dilated cardiomyopathy was previously reported in three Dutch families. Here we report the genetic cause of this disorder. Multipoint parametric linkage analysis of six Dutch patients identified a homozygous region of 2.1 Mb on chromosome 12, which was shared between all Dutch patients, with a log of odds score of 10.82. Sequence analysis of the entire linkage region resulted in the identification of a homozygous mutation in the last acceptor splice site of the myosin regulatory light chain 2 gene (MYL2) as the genetic cause. MYL2 encodes a myosin regulatory light chain (MLC-2V). The myosin regulatory light chains bind, together with the essential light chains, to the flexible neck region of the myosin heavy chain in the hexameric myosin complex and have a structural and regulatory role in muscle contraction. The MYL2 mutation results in use of a cryptic splice site upstream of the last exon causing a frameshift and replacement of the last 32 codons by 20 different codons. Whole exome sequencing of an Italian patient with similar clinical features showed compound heterozygosity for two other mutations affecting the same exon of MYL2, also resulting in mutant proteins with altered C-terminal tails. As a consequence of these mutations, the second EF-hand domain is disrupted. EF-hands, assumed to function as calcium sensors, can undergo a conformational change upon binding of calcium that is critical for interactions with downstream targets. Immunohistochemical staining of skeletal muscle tissue of the Dutch patients showed a diffuse and weak expression of the mutant protein without clear fibre specificity, while normal protein was absent. Heterozygous missense mutations in MYL2 are known to cause dominant hypertrophic cardiomyopathy; however, none of the parents showed signs of cardiomyopathy. In conclusion, the mutations in the last exon of MYL2 are responsible for a novel autosomal recessive lethal myosinopathy due to defects changing the C-terminal tail of the ventricular form of the myosin regulatory light chain. We propose 'light chain myopathy' as a name for this MYL2-associated myopathy.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Myosin Light Chains/genetics , Cardiomyopathy, Hypertrophic/pathology , Female , Humans , Infant , Male , Muscular Diseases/pathology , Mutation
11.
J Inherit Metab Dis ; 36(6): 923-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23296368

ABSTRACT

Increased urinary 3-methylglutaconic acid excretion is a relatively common finding in metabolic disorders, especially in mitochondrial disorders. In most cases 3-methylglutaconic acid is only slightly elevated and accompanied by other (disease specific) metabolites. There is, however, a group of disorders with significantly and consistently increased 3-methylglutaconic acid excretion, where the 3-methylglutaconic aciduria is a hallmark of the phenotype and the key to diagnosis. Until now these disorders were labelled by roman numbers (I-V) in the order of discovery regardless of pathomechanism. Especially, the so called "unspecified" 3-methylglutaconic aciduria type IV has been ever growing, leading to biochemical and clinical diagnostic confusion. Therefore, we propose the following pathomechanism based classification and a simplified diagnostic flow chart for these "inborn errors of metabolism with 3-methylglutaconic aciduria as discriminative feature". One should distinguish between "primary 3-methylglutaconic aciduria" formerly known as type I (3-methylglutaconyl-CoA hydratase deficiency, AUH defect) due to defective leucine catabolism and the--currently known--three groups of "secondary 3-methylglutaconic aciduria". The latter should be further classified and named by their defective protein or the historical name as follows: i) defective phospholipid remodelling (TAZ defect or Barth syndrome, SERAC1 defect or MEGDEL syndrome) and ii) mitochondrial membrane associated disorders (OPA3 defect or Costeff syndrome, DNAJC19 defect or DCMA syndrome, TMEM70 defect). The remaining patients with significant and consistent 3-methylglutaconic aciduria in whom the above mentioned syndromes have been excluded, should be referred to as "not otherwise specified (NOS) 3-MGA-uria" until elucidation of the underlying pathomechanism enables proper (possibly extended) classification.


Subject(s)
Glutarates/urine , Metabolism, Inborn Errors/classification , Metabolism, Inborn Errors/diagnosis , Terminology as Topic , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/urine , Barth Syndrome/diagnosis , Barth Syndrome/genetics , Barth Syndrome/urine , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/urine , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Cerebellar Ataxia/urine , Chorea/diagnosis , Chorea/genetics , Chorea/urine , Diagnosis, Differential , Humans , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/urine , Optic Atrophy/diagnosis , Optic Atrophy/genetics , Optic Atrophy/urine , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/urine
13.
Orphanet J Rare Dis ; 6: 50, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21749694

ABSTRACT

Pontocerebellar Hypoplasia (PCH) is group of very rare, inherited progressive neurodegenerative disorders with prenatal onset. Up to now seven different subtypes have been reported (PCH1-7). The incidence of each subtype is unknown. All subtypes share common characteristics, including hypoplasia/atrophy of cerebellum and pons, progressive microcephaly, and variable cerebral involvement. Patients have severe cognitive and motor handicaps and seizures are often reported. Treatment is only symptomatic and prognosis is poor, as most patients die during infancy or childhood. The genetic basis of different subtypes has been elucidated, which makes prenatal testing possible in families with mutations. Mutations in three tRNA splicing endonuclease subunit genes were found to be responsible for PCH2, PCH4 and PCH5. Mutations in the nuclear encoded mitochondrial arginyl- tRNA synthetase gene underlie PCH6. The tRNA splicing endonuclease, the mitochondrial arginyl- tRNA synthetase and the vaccinia related kinase1 are mutated in the minority of PCH1 cases. These genes are involved in essential processes in protein synthesis in general and tRNA processing in particular. In this review we describe the neuroradiological, neuropathological, clinical and genetic features of the different PCH subtypes and we report on in vitro and in vivo studies on the tRNA splicing endonuclease and mitochondrial arginyl-tRNA synthetase and discuss their relation to pontocerebellar hypoplasia.


Subject(s)
Olivopontocerebellar Atrophies , Animals , Arginine-tRNA Ligase/genetics , Brain/pathology , Child , Child, Preschool , Disease Models, Animal , Endoribonucleases/genetics , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Mutation , Olivopontocerebellar Atrophies/classification , Olivopontocerebellar Atrophies/diagnostic imaging , Olivopontocerebellar Atrophies/genetics , Olivopontocerebellar Atrophies/pathology , Radiography
14.
Eur J Hum Genet ; 19(6): 724-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21368912

ABSTRACT

Pontocerebellar hypoplasia (PCH) is a group of autosomal recessive neurodegenerative disorders characterized by prenatal onset of stunted brain growth and progressive atrophy predominantly affecting cerebellum, pons and olivary nuclei, and to a lesser extent also the cerebral cortex. Six subtypes (PCH1-6) were described and genes for four types (PCH1, 2, 4 and 6) were identified. Mutations in the tRNA splicing endonuclease subunit (TSEN) genes 54, 2 and 34 are found in PCH2 and PCH4. One family with severe prenatal onset of PCH has been the only representative of PCH5 published so far, and the molecular genetic status of PCH5 has not been ascertained until now. We screened the previously reported PCH5 family for mutations in the TSEN54 gene. The PCH5 patient was found to be the result of compound heterozygosity for the common TSEN54 mutation (p.A307S) plus a novel splice site mutation. The mutations associated with PCH5 are similar to what has been reported in PCH4. Thus, PCH5, PCH4 and PCH2 represent a spectrum of clinical manifestations caused by different mutations in the TSEN genes. We, therefore, propose to classify PCH2, PCH4 and PCH5 as TSEN mutation spectrum disorders.


Subject(s)
Endoribonucleases/genetics , Mutation , Olivopontocerebellar Atrophies/genetics , Cerebellum/abnormalities , Female , Fetus , Heterozygote , Humans , Olivary Nucleus/abnormalities , Pedigree , Pons/abnormalities , Pregnancy
16.
Brain ; 134(Pt 1): 143-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20952379

ABSTRACT

Pontocerebellar hypoplasia is a group of autosomal recessive neurodegenerative disorders with prenatal onset. The common characteristics are cerebellar hypoplasia with variable atrophy of the cerebellum and the ventral pons. Supratentorial involvement is reflected by variable neocortical atrophy, ventriculomegaly and microcephaly. Mutations in the transfer RNA splicing endonuclease subunit genes (TSEN54, TSEN2, TSEN34) were found to be associated with pontocerebellar hypoplasia types 2 and 4. Mutations in the mitochondrial transfer RNA arginyl synthetase gene (RARS2) were associated with pontocerebellar hypoplasia type 6. We studied a cohort of 169 patients from 141 families for mutations in these genes, of whom 106 patients tested positive for mutations in one of the TSEN genes or the RARS2 gene. In order to delineate the neuroradiological and clinical phenotype of patients with mutations in these genes, we compared this group with 63 patients suspected of pontocerebellar hypoplasia who were negative on mutation analysis. We found a strong correlation (P < 0.0005) between TSEN54 mutations and a dragonfly-like cerebellar pattern on magnetic resonance imaging, in which the cerebellar hemispheres are flat and severely reduced in size and the vermis is relatively spared. Mutations in TSEN54 are clinically associated with dyskinesia and/or dystonia and variable degrees of spasticity, in some cases with pure generalized spasticity. Nonsense or splice site mutations in TSEN54 are associated with a more severe phenotype of more perinatal symptoms, ventilator dependency and early death. In addition, we present ten new mutations in TSEN54, TSEN2 and RARS2. Furthermore, we show that pontocerebellar hypoplasia type 1 together with elevated cerebrospinal fluid lactate may be caused by RARS2 mutations.


Subject(s)
Arginine-tRNA Ligase/genetics , Brain/pathology , Endoribonucleases/genetics , Adolescent , Chi-Square Distribution , Child , Child, Preschool , Female , Humans , Image Processing, Computer-Assisted , Infant , Magnetic Resonance Imaging , Male , Mutation , Olivopontocerebellar Atrophies/genetics , Olivopontocerebellar Atrophies/pathology
17.
Hum Mutat ; 30(1): 93-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18712838

ABSTRACT

Proteins destined for the peroxisomal matrix are targeted by virtue of a peroxisomal targeting sequence type 1 (PTS1) or type 2 (PTS2). In humans, targeting of either class of proteins relies on a cytosolic receptor protein encoded by the PEX5 gene. Alternative splicing of PEX5 results in two protein variants, PEX5S and PEX5L. PEX5S is exclusively involved in PTS1 protein import, whereas PEX5L mediates the import of both PTS1 and PTS2 proteins. Genetic complementation testing with over 500 different fibroblast cell lines from patients diagnosed with a peroxisome biogenesis disorder (PBD) identified 11 cell lines with a defect in PEX5. The aim of this study was to characterize these cell lines at a biochemical and genetic level. To this end, the cultured fibroblasts were analyzed for very long chain fatty acid (VLCFA) concentrations, peroxisomal beta-and alpha-oxidation, dihydroxyacetone-phosphate acyltransferase (DHAPAT) activity, peroxisomal thiolase, and catalase immunofluorescence. Mutation analysis of the PEX5 gene revealed 11 different mutations, eight of which are novel. PTS1- and PTS2-protein import capacity was assessed by transfection of the cells with green fluorescent protein (GFP) tagged with either PTS1 or PTS2. Six cell lines showed a defect in both PTS1 and PTS2 protein import, whereas four cell lines only showed a defect in PTS1 protein import. The location of the different mutations within the PEX5 amino acid sequence correlates rather well with the peroxisomal protein import defect observed in the cell lines.


Subject(s)
Genotype , Peroxisomal Disorders/genetics , Peroxisomes/metabolism , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Amino Acid Sequence , Cell Line , DNA Mutational Analysis , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Models, Biological , Molecular Sequence Data , Mutation , Peroxisomal Disorders/metabolism , Peroxisome-Targeting Signal 1 Receptor , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Transfection
19.
Nat Genet ; 40(9): 1113-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18711368

ABSTRACT

Pontocerebellar hypoplasias (PCH) represent a group of neurodegenerative autosomal recessive disorders with prenatal onset, atrophy or hypoplasia of the cerebellum, hypoplasia of the ventral pons, microcephaly, variable neocortical atrophy and severe mental and motor impairments. In two subtypes, PCH2 and PCH4, we identified mutations in three of the four different subunits of the tRNA-splicing endonuclease complex. Our findings point to RNA processing as a new basic cellular impairment in neurological disorders.


Subject(s)
Cerebellum/abnormalities , Endoribonucleases/genetics , Mutation , Pons/abnormalities , Brain/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 17 , Humans , Models, Molecular , Polymorphism, Single Nucleotide , Syndrome
20.
Neuromuscul Disord ; 18(1): 52-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17825555

ABSTRACT

Pontocerebellar hypoplasia type 2, an autosomal recessive neurodegeneration with prenatal onset, is characterised by progressive microcephaly and chorea/dystonia and has not previously been associated with muscular involvement. The gene associated with PCH-2 is unknown. An episode of rhabdomyolysis is reported in two non-related children with PCH-2, fatal in one, precipitated by intercurrent disease. Muscle biopsies in two other PCH-2 patients, and in one rhabdomyolysis patient whose biopsy antedated this complication showed areas of myofibrillar disruption or necrosis. Postmortem muscle sampled in another case without neuromuscular symptoms revealed focal necrosis, regenerating small fibres and upregulation of HLA-ABC. Random serum creatine kinase values in six other PCH-2 patients without clinical signs of neuromuscular involvement were increased in four. Collected data provide preliminary evidence of a subclinical myopathy associated with PCH-2.


Subject(s)
Cerebellum/abnormalities , Muscle, Skeletal/pathology , Olivopontocerebellar Atrophies/complications , Pons/abnormalities , Rhabdomyolysis/pathology , Adult , Child, Preschool , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosome Disorders/physiopathology , Creatine Kinase/blood , Female , Genes, Recessive/genetics , HLA Antigens/analysis , HLA Antigens/metabolism , Humans , Infant , Infant, Newborn , Male , Microscopy, Electron, Transmission , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Necrosis/genetics , Necrosis/pathology , Necrosis/physiopathology , Olivopontocerebellar Atrophies/pathology , Olivopontocerebellar Atrophies/physiopathology , Rhabdomyolysis/genetics , Rhabdomyolysis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...