Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Am J Hum Genet ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38744284

ABSTRACT

Anoctamins are a family of Ca2+-activated proteins that may act as ion channels and/or phospholipid scramblases with limited understanding of function and disease association. Here, we identified five de novo and two inherited missense variants in ANO4 (alias TMEM16D) as a cause of fever-sensitive developmental and epileptic or epileptic encephalopathy (DEE/EE) and generalized epilepsy with febrile seizures plus (GEFS+) or temporal lobe epilepsy. In silico modeling of the ANO4 structure predicted that all identified variants lead to destabilization of the ANO4 structure. Four variants are localized close to the Ca2+ binding sites of ANO4, suggesting impaired protein function. Variant mapping to the protein topology suggests a preliminary genotype-phenotype correlation. Moreover, the observation of a heterozygous ANO4 deletion in a healthy individual suggests a dysfunctional protein as disease mechanism rather than haploinsufficiency. To test this hypothesis, we examined mutant ANO4 functional properties in a heterologous expression system by patch-clamp recordings, immunocytochemistry, and surface expression of annexin A5 as a measure of phosphatidylserine scramblase activity. All ANO4 variants showed severe loss of ion channel function and DEE/EE associated variants presented mild loss of surface expression due to impaired plasma membrane trafficking. Increased levels of Ca2+-independent annexin A5 at the cell surface suggested an increased apoptosis rate in DEE-mutant expressing cells, but no changes in Ca2+-dependent scramblase activity were observed. Co-transfection with ANO4 wild-type suggested a dominant-negative effect. In summary, we expand the genetic base for both encephalopathic sporadic and inherited fever-sensitive epilepsies and link germline variants in ANO4 to a hereditary disease.

3.
Front Neurosci ; 17: 1219262, 2023.
Article in English | MEDLINE | ID: mdl-37502687

ABSTRACT

Introduction: Phenotypic spectrum of SLC6A1-related neurodevelopmental disorders (SLC6A1-NDD) includes intellectual disability (ID), autistic spectrum disorders (ASD), epilepsy, developmental delay, beginning from early infancy or after seizure onset, and other neurological features such as hypotonia and movement disorders. Data on familial phenotypic heterogeneity have been rarely reported, thus in our study we aimed to investigate intrafamilial phenotypic variability in families with SLC6A1 variants. Methods: We collected clinical, laboratory and genetic data on 39 individuals, including 17 probands, belonging to 13 families harboring inherited variants of SLC6A1. Data were collected through an international network of Epilepsy and Genetic Centers. Results: Main clinical findings in the whole cohort of 39 subjects were: (a) epilepsy, mainly presenting with generalized seizures, reported in 71% of probands and 36% of siblings or first/second-degree relatives. Within a family, the same epilepsy type (generalized or focal) was observed; (b) ID reported in 100% and in 13% of probands and siblings or first/second-degree relatives, respectively; (c) learning disabilities detected in 28% of the SLC6A1 carriers, all of them were relatives of a proband; (d) around 51% of the whole cohort presented with psychiatric symptoms or behavioral disorders, including 82% of the probands. Out of the 19 patients with psychiatric symptoms, ASD were diagnosed in 40% of them; (e) neurological findings (primarily tremor and speech difficulties) were observed 38.5% of the whole cohort, including 10 probands. Our families harbored 12 different SLC6A1 variants, one was a frameshift, two stop-gain, while the remaining were missense. No genotype-phenotype associations were identified. Discussion: Our study showed that first-or second-degree relatives presented with a less severe phenotype, featuring mainly mild intellectual and/or learning disabilities, at variance with the probands who suffered from moderate to severe ID, generalized, sometimes intractable, epileptic seizures, behavioral and psychiatric disorders. These findings may suggest that a proportion of individuals with mild SLC6A1-NDD might be missed, in particular those with an older age where genetic testing is not performed. Further studies on intrafamilial phenotypic variability are needed to confirm our results and possibly to expand the phenotypic spectrum of these disorders and benefit genetic counseling.

4.
EBioMedicine ; 98: 104855, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38251463

ABSTRACT

BACKGROUND: Variants in SCN8A are associated with a spectrum of epilepsies and neurodevelopmental disorders. Ataxia as a predominant symptom of SCN8A variation has not been well studied. We set out to investigate disease mechanisms and genotype-phenotype correlations of SCN8A-related ataxia. METHODS: We collected genetic and electro-clinical data of ten individuals from nine unrelated families carrying novel SCN8A variants associated with chronic progressive or episodic ataxia. Electrophysiological characterizations of these variants were performed in ND7/23 cells and cultured neurons. FINDINGS: Variants associated with chronic progressive ataxia either decreased Na+ current densities and shifted activation curves towards more depolarized potentials (p.Asn995Asp, p.Lys1498Glu and p.Trp1266Cys) or resulted in a premature stop codon (p.Trp937Ter). Three variants (p.Arg847Gln and biallelic p.Arg191Trp/p.Asp1525Tyr) were associated with episodic ataxia causing loss-of-function by decreasing Na+ current densities or a hyperpolarizing shift of the inactivation curve. Two additional episodic ataxia-associated variants caused mixed gain- and loss-of function effects in ND7/23 cells and were further examined in primary murine hippocampal neuronal cultures. Neuronal firing in excitatory neurons was increased by p.Arg1629His, but decreased by p.Glu1201Lys. Neuronal firing in inhibitory neurons was decreased for both variants. No functional effect was observed for p.Arg1913Trp. In four individuals, treatment with sodium channel blockers exacerbated symptoms. INTERPRETATION: We identified episodic or chronic ataxia as predominant phenotypes caused by variants in SCN8A. Genotype-phenotype correlations revealed a more pronounced loss-of-function effect for variants causing chronic ataxia. Sodium channel blockers should be avoided under these conditions. FUNDING: BMBF, DFG, the Italian Ministry of Health, University of Tuebingen.


Subject(s)
Ataxia , Neurons , Humans , Animals , Mice , Ataxia/diagnosis , Ataxia/genetics , Codon, Nonsense , Sodium Channel Blockers , NAV1.6 Voltage-Gated Sodium Channel/genetics
5.
Brain Commun ; 4(6): fcac295, 2022.
Article in English | MEDLINE | ID: mdl-36447560

ABSTRACT

Epilepsy in infancy links to a significant risk of neurodevelopmental delay, calling for a better understanding of its underlying mechanisms. Here, we studied cortical activity networks in infants with early-onset epilepsy to identify network properties that could pre-empt infants' neurodevelopmental course. We studied high-density (64 channel) electroencephalogram during non-rapid eye movement (N2) sleep in n = 49 infants at 1 year of age after being diagnosed with epilepsy during their first year of life. We computed frequency-specific networks in the cortical source space for two intrinsic brain modes: amplitude-amplitude and phase-phase correlations. Cortical activity networks of all frequency bands and connectivity modes were compared between the syndrome groups as well as between the three categories of neurocognitive development. The group differences were studied at three spatial levels: global, regional, and individual connections. Cortical mechanisms related to infant epilepsy were further compared with physiological networks using an automatic spindle detection algorithm. Our results show that global connectivity does not significantly differ between epilepsy syndromes; however, it co-varies with neurocognitive development. The largest network differences were observed at the lowest (<1 Hz) and mid-range (10-15 Hz) frequency bands. An algorithmic removal of sleep spindles from the data partially reduced the mid-range frequency network's strength. The centrocentral and frontocentral networks at the spindle frequencies were found to be strongest in infants with a persistent age-typical neurocognitive performance, while their low-frequency (< 1 Hz) networks were weaker for both amplitude-amplitude [P = 0.008, effect size = 0.61] and phase-phase correlations (P = 0.02, effect size = 0.54) at low (< 1 Hz). However, subjects with persistent mild neurocognitive delay from 1 to 2 years of age had higher amplitude-amplitude (P = 0.02, effect size = 0.73) and phase-phase (P = 0.06, effect size = 0.59) at low frequencies than those that deteriorated from mild to severely delayed from 1 to 2 years of age. Our findings suggest that cortical activity networks reflect the underlying clinical course of infants' epilepsy, and measures of spectrally and spatially resolved networks might become useful in better understanding infantile epilepsy as a network disease.

6.
Dev Med Child Neurol ; 64(5): 641-648, 2022 05.
Article in English | MEDLINE | ID: mdl-34716587

ABSTRACT

AIM: To investigate the prevalence of vigabatrin-attributed visual field defect (VAVFD) in infantile spasms and the utility of optical coherence tomography (OCT) in detecting vigabatrin-related damage. METHOD: We examined visual fields by Goldmann or Octopus perimetry and the thickness of peripapillary retinal nerve fiber layer (RNFL) with spectral-domain OCT at school age or adolescence. RESULTS: Out of 88 patients (38 females, mean age at study 15y, SD 4y 3mo, range 6y 4mo-23y 3mo [n=65] or deceased [n=21] or moved abroad [n=2]) exposed to vigabatrin in infancy, 28 were able to perform formal visual field testing. Two had visual field defect from structural causes. We found mild VAVFD in four patients and severe VAVFD in one patient. Median vigabatrin treatment duration for those with normal visual field was 11 months compared to 19 months for those with VAVFD (p=0.04). OCT showed concomitant attenuated RNFL in three children with VAVFD, and was normal in one. The temporal half of the peripapillary RNFL was significantly thinner in the VAVFD group compared to the normal visual field group. INTERPRETATION: The overall prevalence of VAVFD is lower after exposure in infancy compared to 52% which has been reported after exposure in adulthood. The risk increases with longer treatment duration. Further studies should identify infants particularly susceptible to VAVFD and clarify the role of OCT.


Subject(s)
Vigabatrin , Visual Fields , Adolescent , Adult , Anticonvulsants , Child , Female , Humans , Infant , Nerve Fibers , Retrospective Studies , Vigabatrin/adverse effects , Visual Field Tests
7.
Hum Genet ; 140(7): 1011-1029, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33710394

ABSTRACT

The genetics of autosomal recessive intellectual disability (ARID) has mainly been studied in consanguineous families, however, founder populations may also be of interest to study intellectual disability (ID) and the contribution of ARID. Here, we used a genotype-driven approach to study the genetic landscape of ID in the founder population of Finland. A total of 39 families with syndromic and non-syndromic ID were analyzed using exome sequencing, which revealed a variant in a known ID gene in 27 families. Notably, 75% of these variants in known ID genes were de novo or suspected de novo (64% autosomal dominant; 11% X-linked) and 25% were inherited (14% autosomal recessive; 7% X-linked; and 4% autosomal dominant). A dual molecular diagnosis was suggested in two families (5%). Via additional analysis and molecular testing, we identified three cases with an abnormal molecular karyotype, including chr21q22.12q22.2 uniparental disomy with a mosaic interstitial 2.7 Mb deletion covering DYRK1A and KCNJ6. Overall, a pathogenic or likely pathogenic variant was identified in 64% (25/39) of the families. Last, we report an alternate inheritance model for 3 known ID genes (UBA7, DDX47, DHX58) and discuss potential candidate genes for ID, including SYPL1 and ERGIC3 with homozygous founder variants and de novo variants in POLR2F and DNAH3. In summary, similar to other European populations, de novo variants were the most common variants underlying ID in the studied Finnish population, with limited contribution of ARID to ID etiology, though mainly driven by founder and potential founder variation in the latter case.


Subject(s)
Exome/genetics , Intellectual Disability/genetics , Family , Female , Finland , Genes, Recessive/genetics , Genetic Predisposition to Disease/genetics , Genotype , Homozygote , Humans , Male , Pedigree , Exome Sequencing/methods
8.
Pediatr Neurol ; 109: 56-62, 2020 08.
Article in English | MEDLINE | ID: mdl-32387008

ABSTRACT

BACKGROUND: Mutations in the X-linked gene WDR45 cause neurodegeneration with brain iron accumulation type 5. Global developmental delay occurs at an early age with slow progression to dystonia, parkinsonism, and dementia due to progressive iron accumulation in the brain. METHODS: We present 17 new cases and reviewed 106 reported cases of neurodegeneration with brain iron accumulation type 5. Detailed information related to developmental history and key time to event measures was collected. RESULTS: Within this cohort, there were 19 males. Most individuals were molecularly diagnosed by whole-exome testing. Overall 10 novel variants were identified across 11 subjects. All individuals were affected by developmental delay, most prominently in verbal skills. Most individuals experienced a decline in motor and cognitive skills. Although most individuals were affected by seizures, the spectrum ranged from provoked seizures to intractable epilepsy. The imaging findings varied as well, often evolving over time. The classic iron accumulation in the globus pallidus and substantia nigra was noted in half of our cohort and was associated with older age of image acquisition, whereas myelination abnormalities were associated with younger age. CONCLUSIONS: WDR45 mutations lead to a progressive and evolving disorder whose diagnosis is often delayed. Developmental delay and seizures predominate in early childhood, followed by a progressive decline of neurological function. There is variable expressivity in the clinical phenotypes of individuals with WDR45 mutations, suggesting that this gene should be considered in the diagnostic evaluation of children with myelination abnormalities, iron deposition, developmental delay, and epilepsy depending on the age at evaluation.


Subject(s)
Carrier Proteins/genetics , Demyelinating Diseases , Developmental Disabilities , Epilepsy , Iron Metabolism Disorders , Neuroaxonal Dystrophies , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Demyelinating Diseases/diagnosis , Demyelinating Diseases/etiology , Demyelinating Diseases/genetics , Demyelinating Diseases/physiopathology , Developmental Disabilities/diagnosis , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Epilepsy/diagnosis , Epilepsy/etiology , Epilepsy/genetics , Epilepsy/physiopathology , Female , Humans , Infant , Iron Metabolism Disorders/complications , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/physiopathology , Male , Middle Aged , Neuroaxonal Dystrophies/complications , Neuroaxonal Dystrophies/diagnosis , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/physiopathology , Phenotype , Exome Sequencing , Young Adult
9.
Epilepsia ; 60(5): 830-844, 2019 05.
Article in English | MEDLINE | ID: mdl-30968951

ABSTRACT

OBJECTIVE: Pathogenic variants in SCN8A have been associated with a wide spectrum of epilepsy phenotypes, ranging from benign familial infantile seizures (BFIS) to epileptic encephalopathies with variable severity. Furthermore, a few patients with intellectual disability (ID) or movement disorders without epilepsy have been reported. The vast majority of the published SCN8A patients suffer from severe developmental and epileptic encephalopathy (DEE). In this study, we aimed to provide further insight on the spectrum of milder SCN8A-related epilepsies. METHODS: A cohort of 1095 patients were screened using a next generation sequencing panel. Further patients were ascertained from a network of epilepsy genetics clinics. Patients with severe DEE and BFIS were excluded from the study. RESULTS: We found 36 probands who presented with an SCN8A-related epilepsy and normal intellect (33%) or mild (61%) to moderate ID (6%). All patients presented with epilepsy between age 1.5 months and 7 years (mean = 13.6 months), and 58% of these became seizure-free, two-thirds on monotherapy. Neurological disturbances included ataxia (28%) and hypotonia (19%) as the most prominent features. Interictal electroencephalogram was normal in 41%. Several recurrent variants were observed, including Ile763Val, Val891Met, Gly1475Arg, Gly1483Lys, Phe1588Leu, Arg1617Gln, Ala1650Val/Thr, Arg1872Gln, and Asn1877Ser. SIGNIFICANCE: With this study, we explore the electroclinical features of an intermediate SCN8A-related epilepsy with mild cognitive impairment, which is for the majority a treatable epilepsy.


Subject(s)
Epilepsy/genetics , Mutation, Missense , NAV1.6 Voltage-Gated Sodium Channel/genetics , Anticonvulsants/therapeutic use , Ataxia/genetics , Child , Child, Preschool , Cognitive Dysfunction/genetics , Electroencephalography , Epilepsy/drug therapy , Epilepsy/physiopathology , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Infant , Intellectual Disability/genetics , Language Development Disorders/genetics , Movement Disorders/genetics , Muscle Hypotonia/genetics , Pedigree , Severity of Illness Index
10.
Nat Genet ; 50(7): 1048-1053, 2018 07.
Article in English | MEDLINE | ID: mdl-29942082

ABSTRACT

Epilepsy is a frequent feature of neurodevelopmental disorders (NDDs), but little is known about genetic differences between NDDs with and without epilepsy. We analyzed de novo variants (DNVs) in 6,753 parent-offspring trios ascertained to have different NDDs. In the subset of 1,942 individuals with NDDs with epilepsy, we identified 33 genes with a significant excess of DNVs, of which SNAP25 and GABRB2 had previously only limited evidence of disease association. Joint analysis of all individuals with NDDs also implicated CACNA1E as a novel disease-associated gene. Comparing NDDs with and without epilepsy, we found missense DNVs, DNVs in specific genes, age of recruitment, and severity of intellectual disability to be associated with epilepsy. We further demonstrate the extent to which our results affect current genetic testing as well as treatment, emphasizing the benefit of accurate genetic diagnosis in NDDs with epilepsy.


Subject(s)
Epilepsy/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Neurodevelopmental Disorders/genetics , Exome/genetics , Female , Genetic Testing/methods , Humans , Intellectual Disability/genetics , Male
11.
Epilepsia ; 59(2): 389-402, 2018 02.
Article in English | MEDLINE | ID: mdl-29315614

ABSTRACT

OBJECTIVE: Pathogenic SLC6A1 variants were recently described in patients with myoclonic atonic epilepsy (MAE) and intellectual disability (ID). We set out to define the phenotypic spectrum in a larger cohort of SCL6A1-mutated patients. METHODS: We collected 24 SLC6A1 probands and 6 affected family members. Four previously published cases were included for further electroclinical description. In total, we reviewed the electroclinical data of 34 subjects. RESULTS: Cognitive development was impaired in 33/34 (97%) subjects; 28/34 had mild to moderate ID, with language impairment being the most common feature. Epilepsy was diagnosed in 31/34 cases with mean onset at 3.7 years. Cognitive assessment before epilepsy onset was available in 24/31 subjects and was normal in 25% (6/24), and consistent with mild ID in 46% (11/24) or moderate ID in 17% (4/24). Two patients had speech delay only, and 1 had severe ID. After epilepsy onset, cognition deteriorated in 46% (11/24) of cases. The most common seizure types were absence, myoclonic, and atonic seizures. Sixteen cases fulfilled the diagnostic criteria for MAE. Seven further patients had different forms of generalized epilepsy and 2 had focal epilepsy. Twenty of 31 patients became seizure-free, with valproic acid being the most effective drug. There was no clear-cut correlation between seizure control and cognitive outcome. Electroencephalography (EEG) findings were available in 27/31 patients showing irregular bursts of diffuse 2.5-3.5 Hz spikes/polyspikes-and-slow waves in 25/31. Two patients developed an EEG pattern resembling electrical status epilepticus during sleep. Ataxia was observed in 7/34 cases. We describe 7 truncating and 18 missense variants, including 4 recurrent variants (Gly232Val, Ala288Val, Val342Met, and Gly362Arg). SIGNIFICANCE: Most patients carrying pathogenic SLC6A1 variants have an MAE phenotype with language delay and mild/moderate ID before epilepsy onset. However, ID alone or associated with focal epilepsy can also be observed.


Subject(s)
Epilepsies, Myoclonic/physiopathology , GABA Plasma Membrane Transport Proteins/genetics , Intellectual Disability/physiopathology , Language Development Disorders/physiopathology , Adolescent , Adult , Anticonvulsants/therapeutic use , Ataxia/complications , Ataxia/genetics , Ataxia/physiopathology , Child , Child, Preschool , Cohort Studies , Electroencephalography , Epilepsies, Myoclonic/complications , Epilepsies, Myoclonic/drug therapy , Epilepsies, Myoclonic/genetics , Epilepsies, Partial/complications , Epilepsies, Partial/drug therapy , Epilepsies, Partial/genetics , Epilepsies, Partial/physiopathology , Epilepsy, Generalized/complications , Epilepsy, Generalized/drug therapy , Epilepsy, Generalized/genetics , Epilepsy, Generalized/physiopathology , Female , Genetic Association Studies , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Language Development Disorders/complications , Language Development Disorders/genetics , Male , Mutation , Mutation, Missense , Neurodevelopmental Disorders/complications , Neurodevelopmental Disorders/genetics , Phenotype , Treatment Outcome , Valproic Acid/therapeutic use , Young Adult
12.
Am J Hum Genet ; 99(3): 683-694, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545674

ABSTRACT

The ubiquitin fold modifier 1 (UFM1) cascade is a recently identified evolutionarily conserved ubiquitin-like modification system whose function and link to human disease have remained largely uncharacterized. By using exome sequencing in Finnish individuals with severe epileptic syndromes, we identified pathogenic compound heterozygous variants in UBA5, encoding an activating enzyme for UFM1, in two unrelated families. Two additional individuals with biallelic UBA5 variants were identified from the UK-based Deciphering Developmental Disorders study and one from the Northern Finland Intellectual Disability cohort. The affected individuals (n = 9) presented in early infancy with severe irritability, followed by dystonia and stagnation of development. Furthermore, the majority of individuals display postnatal microcephaly and epilepsy and develop spasticity. The affected individuals were compound heterozygous for a missense substitution, c.1111G>A (p.Ala371Thr; allele frequency of 0.28% in Europeans), and a nonsense variant or c.164G>A that encodes an amino acid substitution p.Arg55His, but also affects splicing by facilitating exon 2 skipping, thus also being in effect a loss-of-function allele. Using an in vitro thioester formation assay and cellular analyses, we show that the p.Ala371Thr variant is hypomorphic with attenuated ability to transfer the activated UFM1 to UFC1. Finally, we show that the CNS-specific knockout of Ufm1 in mice causes neonatal death accompanied by microcephaly and apoptosis in specific neurons, further suggesting that the UFM1 system is essential for CNS development and function. Taken together, our data imply that the combination of a hypomorphic p.Ala371Thr variant in trans with a loss-of-function allele in UBA5 underlies a severe infantile-onset encephalopathy.


Subject(s)
Alleles , Brain Diseases/genetics , Brain Diseases/metabolism , Mutation/genetics , Proteins/genetics , Ubiquitin-Activating Enzymes/genetics , Ubiquitin/metabolism , Animals , Animals, Newborn , Apoptosis , Brain Diseases/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Cohort Studies , Epilepsy/genetics , Exome/genetics , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Finland , Gene Frequency , Heterozygote , Humans , Infant , Intellectual Disability/genetics , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/pathology , Neurons/metabolism , Neurons/pathology , Proteins/metabolism , Spasms, Infantile/genetics , Spasms, Infantile/metabolism
13.
Hum Mutat ; 37(9): 884-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27279129

ABSTRACT

Mitochondrial aminoacyl-tRNA synthetases are an important group of disease genes typically underlying either a disorder affecting an isolated tissue or a distinct syndrome. Missense mutations in the mitochondrial seryl-tRNA synthetase gene, SARS2, have been identified in HUPRA syndrome (hyperuricemia, pulmonary hypertension, renal failure in infancy, and alkalosis). We report here a homozygous splicing mutation in SARS2 in a patient with progressive spastic paresis. We show that the mutation leads to diminished levels of the synthetase in patient's fibroblasts. This has a destabilizing effect on the tRNASer(AGY) isoacceptor, but to a lesser degree than in HUPRA syndrome patients. tRNASer(UCN) is largely unaffected in both phenotypes. In conclusion, the level of tRNASer(AGY) instability may be a factor in determining tissue manifestation in patients with SARS2 mutations. This finding exemplifies the sensitivity of the nervous system to partially reduced aminoacylation, which is sufficient in other tissues to maintain respiratory chain function.


Subject(s)
Mutation , Paraparesis, Spastic/genetics , RNA Splicing , Serine-tRNA Ligase/genetics , Child , Homozygote , Humans , Male , RNA, Messenger/metabolism
14.
Neurology ; 85(4): 306-15, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26115735

ABSTRACT

OBJECTIVE: We aimed to decipher the molecular genetic basis of disease in a cohort of children with a uniform clinical presentation of neonatal irritability, spastic or dystonic quadriplegia, virtually absent psychomotor development, axonal neuropathy, and elevated blood/CSF lactate. METHODS: We performed whole-exome sequencing of blood DNA from the index patients. Detected compound heterozygous mutations were confirmed by Sanger sequencing. Structural predictions and a bacterial activity assay were performed to evaluate the functional consequences of the mutations. Mass spectrometry, Western blotting, and protein oxidation detection were used to analyze the effects of selenoprotein deficiency. RESULTS: Neuropathology indicated laminar necrosis and severe loss of myelin, with neuron loss and astrogliosis. In 3 families, we identified a missense (p.Thr325Ser) and a nonsense (p.Tyr429*) mutation in SEPSECS, encoding the O-phosphoseryl-tRNA:selenocysteinyl-tRNA synthase, which was previously associated with progressive cerebellocerebral atrophy. We show that the mutations do not completely abolish the activity of SEPSECS, but lead to decreased selenoprotein levels, with demonstrated increase in oxidative protein damage in the patient brain. CONCLUSIONS: These results extend the phenotypes caused by defective selenocysteine biosynthesis, and suggest SEPSECS as a candidate gene for progressive encephalopathies with lactate elevation.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/metabolism , Lactic Acid/blood , Lactic Acid/cerebrospinal fluid , Selenoproteins/deficiency , Adolescent , Brain/metabolism , Brain/pathology , Brain Diseases, Metabolic, Inborn/blood , Brain Diseases, Metabolic, Inborn/cerebrospinal fluid , Child , Child, Preschool , Female , Humans , Male , Mutation , Oxidative Stress/genetics , Selenoproteins/biosynthesis
15.
Nat Genet ; 47(4): 393-399, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751627

ABSTRACT

Epileptic encephalopathies are a phenotypically and genetically heterogeneous group of severe epilepsies accompanied by intellectual disability and other neurodevelopmental features. Using next-generation sequencing, we identified four different de novo mutations in KCNA2, encoding the potassium channel KV1.2, in six isolated patients with epileptic encephalopathy (one mutation recurred three times independently). Four individuals presented with febrile and multiple afebrile, often focal seizure types, multifocal epileptiform discharges strongly activated by sleep, mild to moderate intellectual disability, delayed speech development and sometimes ataxia. Functional studies of the two mutations associated with this phenotype showed almost complete loss of function with a dominant-negative effect. Two further individuals presented with a different and more severe epileptic encephalopathy phenotype. They carried mutations inducing a drastic gain-of-function effect leading to permanently open channels. These results establish KCNA2 as a new gene involved in human neurodevelopmental disorders through two different mechanisms, predicting either hyperexcitability or electrical silencing of KV1.2-expressing neurons.


Subject(s)
Epilepsy/genetics , Kv1.2 Potassium Channel/genetics , Mutation , Spasms, Infantile/genetics , Adult , Amino Acid Sequence , Child , Child, Preschool , Cohort Studies , Female , Genetic Predisposition to Disease , Humans , Infant , Male , Pedigree , Young Adult
16.
Brain ; 137(Pt 4): 1019-29, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24566671

ABSTRACT

Leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation is a disorder caused by recessive mutations in the gene DARS2, which encodes mitochondrial aspartyl-tRNA synthetase. Recent observations indicate that the phenotypic range of the disease is much wider than initially thought. Currently, no treatment is available. The aims of our study were (i) to explore a possible genotype-phenotype correlation; and (ii) to identify potential therapeutic agents that modulate the splice site mutations in intron 2 of DARS2, present in almost all patients. A cross-sectional observational study was performed in 78 patients with two DARS2 mutations in the Amsterdam and Helsinki databases up to December 2012. Clinical information was collected via questionnaires. An inventory was made of the DARS2 mutations in these patients and those previously published. An assay was developed to assess mitochondrial aspartyl-tRNA synthetase enzyme activity in cells. Using a fluorescence reporter system we screened for drugs that modulate DARS2 splicing. Clinical information of 66 patients was obtained. The clinical severity varied from infantile onset, rapidly fatal disease to adult onset, slow and mild disease. The most common phenotype was characterized by childhood onset and slow neurological deterioration. Full wheelchair dependency was rare and usually began in adulthood. In total, 60 different DARS2 mutations were identified, 13 of which have not been reported before. Except for 4 of 42 cases published by others, all patients were compound heterozygous. Ninety-four per cent of the patients had a splice site mutation in intron 2. The groups of patients sharing the same two mutations were too small for formal assessment of genotype-phenotype correlation. However, some combinations of mutations were consistently associated with a mild phenotype. The mitochondrial aspartyl-tRNA synthetase activity was strongly reduced in patient cells. Among the compounds screened, cantharidin was identified as the most potent modulator of DARS2 splicing. In conclusion, the phenotypic spectrum of leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation is wide, but most often the disease has a relatively slow and mild course. The available evidence suggests that the genotype influences the phenotype, but because of the high number of private mutations, larger numbers of patients are necessary to confirm this. The activity of mitochondrial aspartyl-tRNA synthetase is significantly reduced in patient cells. A compound screen established a 'proof of principle' that the splice site mutation can be influenced. This finding is promising for future therapeutic strategies.


Subject(s)
Alternative Splicing/drug effects , Aspartate-tRNA Ligase/deficiency , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Mitochondrial Diseases/complications , Mitochondrial Diseases/genetics , Adolescent , Adult , Age of Onset , Aspartate-tRNA Ligase/genetics , Aspartate-tRNA Ligase/metabolism , Cantharidin/pharmacology , Child , Child, Preschool , Cross-Sectional Studies , DNA Mutational Analysis , Disease Progression , Enzyme Inhibitors/pharmacology , Female , Genetic Association Studies , Humans , Infant , Leukoencephalopathies/drug therapy , Leukoencephalopathies/enzymology , Male , Middle Aged , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/enzymology , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
18.
Am J Hum Genet ; 93(5): 967-75, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24207121

ABSTRACT

Dravet syndrome is a severe epilepsy syndrome characterized by infantile onset of therapy-resistant, fever-sensitive seizures followed by cognitive decline. Mutations in SCN1A explain about 75% of cases with Dravet syndrome; 90% of these mutations arise de novo. We studied a cohort of nine Dravet-syndrome-affected individuals without an SCN1A mutation (these included some atypical cases with onset at up to 2 years of age) by using whole-exome sequencing in proband-parent trios. In two individuals, we identified a de novo loss-of-function mutation in CHD2 (encoding chromodomain helicase DNA binding protein 2). A third CHD2 mutation was identified in an epileptic proband of a second (stage 2) cohort. All three individuals with a CHD2 mutation had intellectual disability and fever-sensitive generalized seizures, as well as prominent myoclonic seizures starting in the second year of life or later. To explore the functional relevance of CHD2 haploinsufficiency in an in vivo model system, we knocked down chd2 in zebrafish by using targeted morpholino antisense oligomers. chd2-knockdown larvae exhibited altered locomotor activity, and the epileptic nature of this seizure-like behavior was confirmed by field-potential recordings that revealed epileptiform discharges similar to seizures in affected persons. Both altered locomotor activity and epileptiform discharges were absent in appropriate control larvae. Our study provides evidence that de novo loss-of-function mutations in CHD2 are a cause of epileptic encephalopathy with generalized seizures.


Subject(s)
DNA-Binding Proteins/genetics , Epilepsies, Myoclonic/genetics , Animals , Child , Cognition Disorders/genetics , Cognition Disorders/pathology , Cohort Studies , Epilepsies, Myoclonic/pathology , Exome , Female , Gene Knockdown Techniques , Haploinsufficiency , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Larva/genetics , Male , NAV1.1 Voltage-Gated Sodium Channel/genetics , Phenotype , Seizures, Febrile/genetics , Seizures, Febrile/pathology , Young Adult , Zebrafish
19.
Nat Genet ; 45(9): 1067-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933819

ABSTRACT

Idiopathic focal epilepsy (IFE) with rolandic spikes is the most common childhood epilepsy, comprising a phenotypic spectrum from rolandic epilepsy (also benign epilepsy with centrotemporal spikes, BECTS) to atypical benign partial epilepsy (ABPE), Landau-Kleffner syndrome (LKS) and epileptic encephalopathy with continuous spike and waves during slow-wave sleep (CSWS). The genetic basis is largely unknown. We detected new heterozygous mutations in GRIN2A in 27 of 359 affected individuals from 2 independent cohorts with IFE (7.5%; P = 4.83 × 10(-18), Fisher's exact test). Mutations occurred significantly more frequently in the more severe phenotypes, with mutation detection rates ranging from 12/245 (4.9%) in individuals with BECTS to 9/51 (17.6%) in individuals with CSWS (P = 0.009, Cochran-Armitage test for trend). In addition, exon-disrupting microdeletions were found in 3 of 286 individuals (1.0%; P = 0.004, Fisher's exact test). These results establish alterations of the gene encoding the NMDA receptor NR2A subunit as a major genetic risk factor for IFE.


Subject(s)
Epilepsies, Partial/genetics , Mutation , Receptors, N-Methyl-D-Aspartate/genetics , Amino Acid Substitution , Epilepsies, Partial/diagnosis , Female , Humans , Male , Models, Molecular , Mutation, Missense , Pedigree , Protein Conformation , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
20.
Am J Med Genet A ; 158A(12): 3119-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23165795

ABSTRACT

Hypochondroplasia (HCH), an autosomal dominant skeletal dysplasia caused by mutations in the FGFR3 gene, has not been commonly associated with neurological problems. Temporal lobe dysgenesis associated with epilepsy was recently described in single patients. In this retrospective study, we assessed neurological and neuroimaging aspects of 13 FGFR3 (N540K) mutation verified HCH patients in Finland. Eight patients had neurocognitive difficulties, ranging from specific learning disorder (2/13) to mild intellectual disability (5/13) or global developmental delay (1/13). Six of 13 patients had a history of seizures or epilepsy. Eight patients had undergone MRI. They all had structural abnormalities consistent with temporal lobe dysgenesis. Six patients had peritrigonal white matter reduction, and 4 had abnormally shaped lateral ventricles. We recommend a close follow-up of development in patients with HCH and a low threshold for neuroimaging.


Subject(s)
Dwarfism/genetics , Dwarfism/pathology , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/pathology , Lordosis/genetics , Lordosis/pathology , Mutation , Receptor, Fibroblast Growth Factor, Type 3/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adolescent , Bone and Bones/abnormalities , Bone and Bones/pathology , Child , Child, Preschool , Dwarfism/diagnosis , Female , Finland , Humans , Infant , Limb Deformities, Congenital/diagnosis , Lordosis/diagnosis , Male , Neuroimaging/methods , Psychomotor Disorders/diagnosis , Psychomotor Disorders/genetics , Psychomotor Disorders/pathology , Retrospective Studies , Temporal Lobe/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...