Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Article in English | MEDLINE | ID: mdl-36229191

ABSTRACT

BACKGROUND AND OBJECTIVE: The spectrum of myelin oligodendrocyte glycoprotein (MOG) antibody-associated disorder (MOGAD) comprises monophasic diseases such as acute disseminated encephalomyelitis (ADEM), optic neuritis (ON), and transverse myelitis and relapsing courses of these presentations. Persistently high MOG antibodies (MOG immunoglobulin G [IgG]) are found in patients with a relapsing disease course. Prognostic factors to determine the clinical course of children with a first MOGAD are still lacking. The objective of the study is to assess the clinical and laboratory prognostic parameters for a risk of relapse and the temporal dynamics of MOG-IgG titers in children with MOGAD in correlation with clinical presentation and disease course. METHODS: In this prospective multicenter hospital-based study, children with a first demyelinating attack and complete data set comprising clinical and radiologic findings, MOG-IgG titer at onset, and clinical and serologic follow-up data were included. Serum samples were analyzed by live cell-based assay, and a titer level of ≥1:160 was classified as MOG-IgG-positive. RESULTS: One hundred sixteen children (f:m = 57:59) with MOGAD were included and initially diagnosed with ADEM (n = 59), unilateral ON (n = 12), bilateral ON (n = 16), myelitis (n = 6), neuromyelitis optica spectrum disorder (n = 8) or encephalitis (n = 6). The median follow-up time was 3 years in monophasic and 5 years in relapsing patients. There was no significant association between disease course and MOG-IgG titers at onset, sex, age at presentation, or clinical phenotype. Seroconversion to MOG-IgG-negative within 2 years of the initial event showed a significant risk reduction for a relapsing disease course. Forty-two/one hundred sixteen patients (monophasic n = 26, relapsing n = 16) had serial MOG-IgG testing in years 1 and 2 after the initial event. In contrast to relapsing patients, monophasic patients showed a significant decrease of MOG-IgG titers during the first and second years, often with seroconversion to negative titers. During the follow-up, MOG-IgG titers were persistently higher in relapsing than in monophasic patients. Decrease in MOG-IgG of ≥3 dilution steps after the first and second years was shown to be associated with a decreased risk of relapses. In our cohort, no patient experienced a relapse after seroconversion to MOG-IgG-negative. DISCUSSION: In this study, patients with declining MOG-IgG titers, particularly those with seroconversion to MOG-IgG-negative, are shown to have a significantly reduced relapse risk.


Subject(s)
Encephalomyelitis, Acute Disseminated , Neuromyelitis Optica , Optic Neuritis , Humans , Immunoglobulin G , Myelin-Oligodendrocyte Glycoprotein , Neoplasm Recurrence, Local , Prospective Studies , Syndrome
2.
Am J Hum Genet ; 108(5): 857-873, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33961779

ABSTRACT

The ALF transcription factor paralogs, AFF1, AFF2, AFF3, and AFF4, are components of the transcriptional super elongation complex that regulates expression of genes involved in neurogenesis and development. We describe an autosomal dominant disorder associated with de novo missense variants in the degron of AFF3, a nine amino acid sequence important for its binding to ubiquitin ligase, or with de novo deletions of this region. The sixteen affected individuals we identified, along with two previously reported individuals, present with a recognizable pattern of anomalies, which we named KINSSHIP syndrome (KI for horseshoe kidney, NS for Nievergelt/Savarirayan type of mesomelic dysplasia, S for seizures, H for hypertrichosis, I for intellectual disability, and P for pulmonary involvement), partially overlapping the AFF4-associated CHOPS syndrome. Whereas homozygous Aff3 knockout mice display skeletal anomalies, kidney defects, brain malformations, and neurological anomalies, knockin animals modeling one of the microdeletions and the most common of the missense variants identified in affected individuals presented with lower mesomelic limb deformities like KINSSHIP-affected individuals and early lethality, respectively. Overexpression of AFF3 in zebrafish resulted in body axis anomalies, providing some support for the pathological effect of increased amount of AFF3. The only partial phenotypic overlap of AFF3- and AFF4-associated syndromes and the previously published transcriptome analyses of ALF transcription factors suggest that these factors are not redundant and each contributes uniquely to proper development.


Subject(s)
Brain Diseases/genetics , Epilepsy/genetics , Fused Kidney/genetics , Intellectual Disability/genetics , Mutation, Missense , Nuclear Proteins/genetics , Osteochondrodysplasias/genetics , Adolescent , Amino Acid Sequence , Animals , Brain Diseases/etiology , Child , Child, Preschool , Epilepsy/complications , Evolution, Molecular , Female , Gene Frequency , Humans , Infant , Male , Mice , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Phenotype , Protein Stability , Syndrome , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/genetics , Young Adult , Zebrafish/genetics
3.
J Neuromuscul Dis ; 7(1): 41-46, 2020.
Article in English | MEDLINE | ID: mdl-31744015

ABSTRACT

The natural history of patients with spinal muscular atrophy (SMA) has changed due to advances in standard care and development of targeted treatments. Nusinersen was the first drug approved for the treatment of all SMA patients. The transfer of clinical trial data into a real-life environment is challenging, especially regarding the advice of patients and families to what extent they can expect a benefit from the novel treatment. We report the results of a modified Delphi consensus process among child neurologists from Germany, Austria and Switzerland about the indication or continuation of nusinersen treatment in children with SMA type 1 based on different clinical case scenarios.


Subject(s)
Consensus , Neurologists , Oligonucleotides/therapeutic use , Pediatricians , Spinal Muscular Atrophies of Childhood/drug therapy , Austria , Child , Delphi Technique , Germany , Humans , Switzerland
4.
Eur J Hum Genet ; 26(2): 258-264, 2018 02.
Article in English | MEDLINE | ID: mdl-29358611

ABSTRACT

Rolandic epilepsy (RE) is the most common focal epilepsy in childhood. To date no hypothesis-free exome-wide mutational screen has been conducted for RE and atypical RE (ARE). Here we report on whole-exome sequencing of 194 unrelated patients with RE/ARE and 567 ethnically matched population controls. We identified an exome-wide significantly enriched burden for deleterious and loss-of-function variants only for the established RE/ARE gene GRIN2A. The statistical significance of the enrichment disappeared after removing ARE patients. For several disease-related gene-sets, an odds ratio >1 was detected for loss-of-function variants.


Subject(s)
Epilepsy, Rolandic/genetics , Loss of Function Mutation , Receptors, N-Methyl-D-Aspartate/genetics , Adolescent , Child , Epilepsy, Rolandic/pathology , Exome , Female , Humans , Male
5.
Neurology ; 89(9): 900-908, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28768844

ABSTRACT

OBJECTIVE: To assess the prognostic value of MOG antibodies (abs) in the differential diagnosis of acquired demyelinating syndromes (ADS). METHODS: Clinical course, MRI, MOG-abs, AQP4-abs, and CSF cells and oligoclonal bands (OCB) in children with ADS and 24 months of follow-up were reviewed in this observational prospective multicenter hospital-based study. RESULTS: Two hundred ten children with ADS were included and diagnosed with acute disseminated encephalomyelitis (ADEM) (n = 60), neuromyelitis optica spectrum disorder (NMOSD) (n = 12), clinically isolated syndrome (CIS) (n = 101), and multiple sclerosis (MS) (n = 37) after the first episode. MOG-abs were predominantly found in ADEM (57%) and less frequently in NMOSD (25%), CIS (25%), or MS (8%). Increased MOG-ab titers were associated with younger age (p = 0.0001), diagnosis of ADEM (p = 0.005), increased CSF cell counts (p = 0.011), and negative OCB (p = 0.012). At 24-month follow-up, 96 children had no further relapses. Thirty-five children developed recurrent non-MS episodes (63% MOG-, 17% AQP4-abs at onset). Seventy-nine children developed MS (4% MOG-abs at onset). Recurrent non-MS episodes were associated with high MOG-ab titers (p = 0.0003) and older age at onset (p = 0.024). MS was predicted by MS-like MRI (p < 0.0001) and OCB (p = 0.007). An MOG-ab cutoff titer ≥1:1,280 predicted a non-MS course with a sensitivity of 47% and a specificity of 100% and a recurrent non-MS course with a sensitivity of 46% and a specificity of 86%. CONCLUSIONS: Our results show that the presence of MOG-abs strongly depends on the age at disease onset and that high MOG-ab titers were associated with a recurrent non-MS disease course.


Subject(s)
Encephalomyelitis, Acute Disseminated/immunology , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/blood , Myelin-Oligodendrocyte Glycoprotein/immunology , Neuromyelitis Optica/immunology , Adolescent , Autoantibodies , Biomarkers/metabolism , Child , Child, Preschool , Diagnosis, Differential , Disease Progression , Encephalomyelitis, Acute Disseminated/blood , Encephalomyelitis, Acute Disseminated/cerebrospinal fluid , Encephalomyelitis, Acute Disseminated/diagnostic imaging , Female , Follow-Up Studies , Humans , Infant , Magnetic Resonance Imaging , Male , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/diagnostic imaging , Neuromyelitis Optica/blood , Neuromyelitis Optica/cerebrospinal fluid , Neuromyelitis Optica/diagnostic imaging , Oligoclonal Bands , Prognosis , Prospective Studies
6.
Pediatr Rheumatol Online J ; 15(1): 67, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28830446

ABSTRACT

BACKGROUND: Loss-of-function CECR1 mutations cause polyarteritis nodosa (PAN) with childhood onset, an autoinflammatory disorder without significant signs of autoimmunity. Herein we describe the unusual presentation of an autoimmune phenotype with constitutive type I interferon activation in siblings with adenosine deaminase 2 (ADA2) deficiency. CASE PRESENTATION: We describe two siblings with early-onset recurrent strokes, arthritis, oral ulcers, discoid rash, peripheral vascular occlusive disease and high antinuclear antibody titers. Assessment of interferon signatures in blood revealed constitutive type I interferon activation. Aicardi-Goutières syndrome (AGS) was suspected, but no mutation in the known AGS genes were detected. Whole exome sequencing identified compound heterozygosity for a known and a novel mutation in the CECR1 gene. Functional consequences of the mutations were demonstrated by marked reduction in ADA2 catalytic activity. CONCLUSIONS: Our findings demonstrate that ADA2 deficiency can cause an unusual autoimmune phenotype extending the phenotypic spectrum of PAN. Constitutive interferon I activation in patient blood suggests a possible role of type I interferon in disease pathogenesis which may have therapeutic implications.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/genetics , Intercellular Signaling Peptides and Proteins/genetics , Interferon Type I/metabolism , Polyarteritis Nodosa/genetics , Severe Combined Immunodeficiency/genetics , Adenosine Deaminase/genetics , Agammaglobulinemia/complications , Agammaglobulinemia/diagnosis , Child , Child, Preschool , Humans , Infant , Interferon Type I/genetics , Male , Mutation , Pedigree , Phenotype , Polyarteritis Nodosa/complications , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/diagnosis
7.
PLoS One ; 11(3): e0150426, 2016.
Article in English | MEDLINE | ID: mdl-26990884

ABSTRACT

OBJECTIVE: The SCN1A gene, coding for the voltage-gated Na+ channel alpha subunit NaV1.1, is the clinically most relevant epilepsy gene. With the advent of high-throughput next-generation sequencing, clinical laboratories are generating an ever-increasing catalogue of SCN1A variants. Variants are more likely to be classified as pathogenic if they have already been identified previously in a patient with epilepsy. Here, we critically re-evaluate the pathogenicity of this class of variants in a cohort of patients with common epilepsy syndromes and subsequently ask whether a significant fraction of benign variants have been misclassified as pathogenic. METHODS: We screened a discovery cohort of 448 patients with a broad range of common genetic epilepsies and 734 controls for previously reported SCN1A mutations that were assumed to be disease causing. We re-evaluated the evidence for pathogenicity of the identified variants using in silico predictions, segregation, original reports, available functional data and assessment of allele frequencies in healthy individuals as well as in a follow up cohort of 777 patients. RESULTS AND INTERPRETATION: We identified 8 known missense mutations, previously reported as pathogenic, in a total of 17 unrelated epilepsy patients (17/448; 3.80%). Our re-evaluation indicates that 7 out of these 8 variants (p.R27T; p.R28C; p.R542Q; p.R604H; p.T1250M; p.E1308D; p.R1928G; NP_001159435.1) are not pathogenic. Only the p.T1174S mutation may be considered as a genetic risk factor for epilepsy of small effect size based on the enrichment in patients (P = 6.60 x 10-4; OR = 0.32, fishers exact test), previous functional studies but incomplete penetrance. Thus, incorporation of previous studies in genetic counseling of SCN1A sequencing results is challenging and may produce incorrect conclusions.


Subject(s)
Epilepsy/genetics , Mutation, Missense , NAV1.1 Voltage-Gated Sodium Channel/genetics , Amino Acid Substitution , Case-Control Studies , Epilepsy/epidemiology , Female , Humans , Male , Risk Factors , Syndrome
8.
J Neurol Neurosurg Psychiatry ; 87(8): 897-905, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26645082

ABSTRACT

OBJECTIVE: To determine the frequency and clinical-radiological associations of antibodies to myelin oligodendrocyte glycoprotein (MOG) and aquaporin-4 (AQP4) in children presenting with neuromyelitis optica (NMO) and limited forms. METHODS: Children with a first event of NMO, recurrent (RON), bilateral ON (BON), longitudinally extensive transverse myelitis (LETM) or brainstem syndrome (BS) with a clinical follow-up of more than 12 months were enrolled. Serum samples were tested for MOG- and AQP4-antibodies using live cell-based assays. RESULTS: 45 children with NMO (n=12), LETM (n=14), BON (n=6), RON (n=12) and BS (n=1) were included. 25/45 (56%) children had MOG-antibodies at initial presentation (7 NMO, 4 BON, 8 ON, 6 LETM). 5/45 (11%) children showed AQP4-antibodies (3 NMO, 1 LETM, 1 BS) and 15/45 (33%) were seronegative for both antibodies (2 NMO, 2 BON, 4 RON, 7 LETM). No differences were found in the age at presentation, sex ratio, frequency of oligoclonal bands or median EDSS at last follow-up between the three groups. Children with MOG-antibodies more frequently (1) had a monophasic course (p=0.018) after one year, (2) presented with simultaneous ON and LETM (p=0.004) and (3) were less likely to receive immunosuppressive therapies (p=0.0002). MRI in MOG-antibody positive patients (4) less frequently demonstrated periependymal lesions (p=0.001), (5) more often were unspecific (p=0.004) and (6) resolved more frequently (p=0.016). CONCLUSIONS: 67% of all children presenting with NMO or limited forms tested positive for MOG- or AQP4-antibodies. MOG-antibody positivity was associated with distinct features. We therefore recommend to measure both antibodies in children with demyelinating syndromes.


Subject(s)
Aquaporin 4/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelitis, Transverse/immunology , Neuromyelitis Optica/immunology , Adolescent , Aquaporin 4/blood , Autoantibodies/blood , Autoantibodies/immunology , Brain Stem/diagnostic imaging , Brain Stem/pathology , Child , Child, Preschool , Female , Humans , Infant , Leukocytosis/cerebrospinal fluid , Magnetic Resonance Imaging , Male , Myelin-Oligodendrocyte Glycoprotein/blood , Myelitis, Transverse/blood , Myelitis, Transverse/cerebrospinal fluid , Myelitis, Transverse/diagnostic imaging , Neuroimaging , Neuromyelitis Optica/blood , Neuromyelitis Optica/cerebrospinal fluid , Neuromyelitis Optica/diagnostic imaging , Oligoclonal Bands/cerebrospinal fluid , Risk Factors , Syndrome
9.
J Neurol ; 262(9): 2124-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26100331

ABSTRACT

Dynein, cytoplasmic 1, heavy chain 1 (DYNC1H1) encodes a necessary subunit of the cytoplasmic dynein complex, which traffics cargo along microtubules. Dominant DYNC1H1 mutations are implicated in neural diseases, including spinal muscular atrophy with lower extremity dominance (SMA-LED), intellectual disability with neuronal migration defects, malformations of cortical development, and Charcot-Marie-Tooth disease, type 2O. We hypothesized that additional variants could be found in these and novel motoneuron and related diseases. Therefore, we analyzed our database of 1024 whole exome sequencing samples of motoneuron and related diseases for novel single nucleotide variations. We filtered these results for significant variants, which were further screened using segregation analysis in available family members. Analysis revealed six novel, rare, and highly conserved variants. Three of these are likely pathogenic and encompass a broad phenotypic spectrum with distinct disease clusters. Our findings suggest that DYNC1H1 variants can cause not only lower, but also upper motor neuron disease. It thus adds DYNC1H1 to the growing list of spastic paraplegia related genes in microtubule-dependent motor protein pathways.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Cytoplasmic Dyneins/genetics , Motor Neuron Disease/genetics , Mutation , Phenotype , Charcot-Marie-Tooth Disease/pathology , DNA Mutational Analysis , Humans , Motor Neuron Disease/pathology , Motor Neurons/pathology , Muscle, Skeletal/pathology
10.
Ann Neurol ; 77(6): 972-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25726841

ABSTRACT

OBJECTIVE: To test whether mutations in γ-aminobutyric acid type A receptor (GABAA -R) subunit genes contribute to the etiology of rolandic epilepsy (RE) or its atypical variants (ARE). METHODS: We performed exome sequencing to compare the frequency of variants in 18 GABAA -R genes in 204 European patients with RE/ARE versus 728 platform-matched controls. Identified GABRG2 variants were functionally assessed for protein stability, trafficking, postsynaptic clustering, and receptor function. RESULTS: Of 18 screened GABAA -R genes, we detected an enrichment of rare variants in the GABRG2 gene in RE/ARE patients (5 of 204, 2.45%) in comparison to controls (1 of 723, 0.14%; odds ratio = 18.07, 95% confidence interval = 2.01-855.07, p = 0.0024, pcorr = 0.043). We identified a GABRG2 splice variant (c.549-3T>G) in 2 unrelated patients as well as 3 nonsynonymous variations in this gene (p.G257R, p.R323Q, p.I389V). Functional assessment showed reduced surface expression of p.G257R and decreased GABA-evoked currents for p.R323Q. The p.G257R mutation displayed diminished levels of palmitoylation, a post-translational modification crucial for trafficking of proteins to the cell membrane. Enzymatically raised palmitoylation levels restored the surface expression of the p.G257R variant γ2 subunit. INTERPRETATION: The statistical association and the functional evidence suggest that mutations of the GABRG2 gene may increase the risk of RE/ARE. Restoring the impaired membrane trafficking of some GABRG2 mutations by enhancing palmitoylation might be an interesting therapeutic approach to reverse the pathogenic effect of such mutants.


Subject(s)
Epilepsy, Rolandic/genetics , Lipoylation/genetics , Mutation/genetics , Receptors, GABA-A/genetics , Exome , Female , HEK293 Cells , Humans , Landau-Kleffner Syndrome/genetics , Male , Pedigree , Syndrome , White People/genetics
11.
Epilepsia ; 55(8): e89-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24995671

ABSTRACT

Rolandic epilepsy (RE) and its atypical variants (atypical rolandic epilepsy, ARE) along the spectrum of epilepsy-aphasia disorders are characterized by a strong but largely unknown genetic basis. Two genes with a putative (ELP4) or a proven (SRPX2) function in neuronal migration were postulated to confer susceptibility to parts of the disease spectrum: the ELP4 gene to centrotemporal spikes and SRPX2 to ARE. To reexamine these findings, we investigated a cohort of 280 patients of European ancestry with RE/ARE for the etiological contribution of these genes and their close interaction partners. We performed next-generation sequencing and single-nucleotide polymorphism (SNP)-array based genotyping to screen for sequence and structural variants. In comparison to European controls we could not detect an enrichment of rare deleterious variants of ELP4, SRPX2, or their interaction partners in affected individuals. The previously described functional p.N327S variant in the X chromosomal SRPX2 gene was detected in two affected individuals (0.81%) and also in controls (0.26%), with some preponderance of male patients. We did not detect an association of SNPs in the ELP4 gene with centrotemporal spikes as previously reported. In conclusion our data do not support a major role of ELP4 and SRPX2 in the etiology of RE/ARE.


Subject(s)
Epilepsy, Rolandic/diagnosis , Epilepsy, Rolandic/genetics , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Austria/epidemiology , Canada/epidemiology , Child , Epilepsy, Rolandic/epidemiology , Female , Genetic Variation/genetics , Germany/epidemiology , Humans , Male , Membrane Proteins , Neoplasm Proteins
12.
Hum Mol Genet ; 23(22): 6069-80, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24939913

ABSTRACT

Rolandic epilepsy (RE) is the most common idiopathic focal childhood epilepsy. Its molecular basis is largely unknown and a complex genetic etiology is assumed in the majority of affected individuals. The present study tested whether six large recurrent copy number variants at 1q21, 15q11.2, 15q13.3, 16p11.2, 16p13.11 and 22q11.2 previously associated with neurodevelopmental disorders also increase risk of RE. Our association analyses revealed a significant excess of the 600 kb genomic duplication at the 16p11.2 locus (chr16: 29.5-30.1 Mb) in 393 unrelated patients with typical (n = 339) and atypical (ARE; n = 54) RE compared with the prevalence in 65,046 European population controls (5/393 cases versus 32/65,046 controls; Fisher's exact test P = 2.83 × 10(-6), odds ratio = 26.2, 95% confidence interval: 7.9-68.2). In contrast, the 16p11.2 duplication was not detected in 1738 European epilepsy patients with either temporal lobe epilepsy (n = 330) and genetic generalized epilepsies (n = 1408), suggesting a selective enrichment of the 16p11.2 duplication in idiopathic focal childhood epilepsies (Fisher's exact test P = 2.1 × 10(-4)). In a subsequent screen among children carrying the 16p11.2 600 kb rearrangement we identified three patients with RE-spectrum epilepsies in 117 duplication carriers (2.6%) but none in 202 carriers of the reciprocal deletion. Our results suggest that the 16p11.2 duplication represents a significant genetic risk factor for typical and atypical RE.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 16/genetics , Epilepsy, Rolandic/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 22/genetics , DNA Copy Number Variations , Female , Humans , Infant , Male , Polymorphism, Single Nucleotide
13.
Brain ; 136(Pt 10): 3140-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24014518

ABSTRACT

Epilepsy comprises several syndromes, amongst the most common being mesial temporal lobe epilepsy with hippocampal sclerosis. Seizures in mesial temporal lobe epilepsy with hippocampal sclerosis are typically drug-resistant, and mesial temporal lobe epilepsy with hippocampal sclerosis is frequently associated with important co-morbidities, mandating the search for better understanding and treatment. The cause of mesial temporal lobe epilepsy with hippocampal sclerosis is unknown, but there is an association with childhood febrile seizures. Several rarer epilepsies featuring febrile seizures are caused by mutations in SCN1A, which encodes a brain-expressed sodium channel subunit targeted by many anti-epileptic drugs. We undertook a genome-wide association study in 1018 people with mesial temporal lobe epilepsy with hippocampal sclerosis and 7552 control subjects, with validation in an independent sample set comprising 959 people with mesial temporal lobe epilepsy with hippocampal sclerosis and 3591 control subjects. To dissect out variants related to a history of febrile seizures, we tested cases with mesial temporal lobe epilepsy with hippocampal sclerosis with (overall n = 757) and without (overall n = 803) a history of febrile seizures. Meta-analysis revealed a genome-wide significant association for mesial temporal lobe epilepsy with hippocampal sclerosis with febrile seizures at the sodium channel gene cluster on chromosome 2q24.3 [rs7587026, within an intron of the SCN1A gene, P = 3.36 × 10(-9), odds ratio (A) = 1.42, 95% confidence interval: 1.26-1.59]. In a cohort of 172 individuals with febrile seizures, who did not develop epilepsy during prospective follow-up to age 13 years, and 6456 controls, no association was found for rs7587026 and febrile seizures. These findings suggest SCN1A involvement in a common epilepsy syndrome, give new direction to biological understanding of mesial temporal lobe epilepsy with hippocampal sclerosis with febrile seizures, and open avenues for investigation of prognostic factors and possible prevention of epilepsy in some children with febrile seizures.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Sclerosis/genetics , Seizures, Febrile/genetics , Epilepsy, Temporal Lobe/etiology , Genome-Wide Association Study/methods , Hippocampus/pathology , Humans , Prospective Studies , Seizures, Febrile/diagnosis , Temporal Lobe/pathology
14.
BMC Med Genet ; 13: 60, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22830313

ABSTRACT

BACKGROUND: Left ventricular hypertrabeculation/noncompaction (LVHT) is a cardiac abnormality of unknown etiology which has been described in children as well as in adults with and without chromosomal aberrations. LVHT has been reported in association with various cardiac and extracardiac abnormalities like epilepsy and facial dysmorphism. CASE PRESENTATION: A unique combination of LVHT, atrial septal defect, pulmonary valve stenosis, aortic stenosis, epilepsy and minor facial anomalies is presented in a 5.5 years old girl. Microarray-based genomic hybridization (array-CGH) detected six previously not described copy number variants (CNVs) inherited from a clinically unaffected father and minimally affected mother, thus, most likely, not clinically significant but rare benign variants. CONCLUSIONS: Despite this complex phenotype de novo microdeletions or microduplications were not detected by array CGH. Further investigations, such as whole exome sequencing, could reveal point mutations and small indels as the possible cause.


Subject(s)
Aortic Valve Stenosis/diagnosis , Epilepsy/diagnosis , Face/abnormalities , Isolated Noncompaction of the Ventricular Myocardium/diagnosis , Pulmonary Valve Stenosis/diagnosis , Aortic Valve Stenosis/genetics , Child, Preschool , DNA Copy Number Variations , Epilepsy/genetics , Female , Humans , Isolated Noncompaction of the Ventricular Myocardium/genetics , Oligonucleotide Array Sequence Analysis , Pulmonary Valve Stenosis/genetics
15.
Am J Hum Genet ; 90(1): 61-8, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22243965

ABSTRACT

Low copper and ceruloplasmin in serum are the diagnostic hallmarks for Menkes disease, Wilson disease, and aceruloplasminemia. We report on five patients from four unrelated families with these biochemical findings who presented with a lethal autosomal-recessive syndrome of congenital cataracts, hearing loss, and severe developmental delay. Cerebral MRI showed pronounced cerebellar hypoplasia and hypomyelination. Homozygosity mapping was performed and displayed a region of commonality among three families at chromosome 3q25. Deep sequencing and conventional sequencing disclosed homozygous or compound heterozygous mutations for all affected subjects in SLC33A1 encoding a highly conserved acetylCoA transporter (AT-1) required for acetylation of multiple gangliosides and glycoproteins. The mutations were found to cause reduced or absent AT-1 expression and abnormal intracellular localization of the protein. We also showed that AT-1 knockdown in HepG2 cells leads to reduced ceruloplasmin secretion, indicating that the low copper in serum is due to reduced ceruloplasmin levels and is not a sign of copper deficiency. The severity of the phenotype implies an essential role of AT-1 in proper posttranslational modification of numerous proteins, without which normal lens and brain development is interrupted. Furthermore, AT-1 defects are a new and important differential diagnosis in patients with low copper and ceruloplasmin in serum.


Subject(s)
Cataract/genetics , Ceruloplasmin/metabolism , Copper/blood , Hearing Loss/genetics , Membrane Transport Proteins/genetics , Mutation/genetics , Base Sequence , Cataract/congenital , Cerebellum/abnormalities , Cerebellum/growth & development , Ceruloplasmin/analysis , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 3/genetics , Female , Hearing Loss/congenital , Hep G2 Cells , Humans , Infant , Male , Membrane Transport Proteins/biosynthesis , Molecular Sequence Data , Severity of Illness Index
16.
Neurogenetics ; 12(4): 273-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21965147

ABSTRACT

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by cerebellar degeneration, immunodeficiency, oculocutaneous telangiectasias, chromosomal instability, radiosensitivity, and cancer predisposition. The gene mutated in the patients, ATM, encodes a member of the phosphatidylinositol 3-kinase family proteins. The ATM protein has a key role in the cellular response to DNA damage. Truncating and splice site mutations in ATM have been found in most patients with the classical AT phenotype. Here we report of our extensive ATM mutation screening on 25 AT patients from 19 families of different ethnic origin. Previously unknown mutations were identified in six patients including a new homozygous missense mutation, c.8110T>C (p.Cys2704Arg), in a severely affected patient. Comprehensive clinical data are presented for all patients described here along with data on ATM function generated by analysis of cell lines established from a subset of the patients.


Subject(s)
Ataxia Telangiectasia/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Child , Child, Preschool , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Female , Haplotypes , Humans , Male , Phenotype , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Tumor Suppressor Proteins/metabolism
17.
PLoS One ; 5(9)2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20862287

ABSTRACT

BACKGROUND: Human mesial temporal lobe epilepsies (MTLE) represent the most frequent form of partial epilepsies and are frequently preceded by febrile seizures (FS) in infancy and early childhood. Genetic associations of several complement genes including its central component C3 with disorders of the central nervous system, and the existence of C3 dysregulation in the epilepsies and in the MTLE particularly, make it the C3 gene a good candidate for human MTLE. METHODOLOGY/PRINCIPAL FINDINGS: A case-control association study of the C3 gene was performed in a first series of 122 patients with MTLE and 196 controls. Four haplotypes (HAP1 to 4) comprising GF100472, a newly discovered dinucleotide repeat polymorphism [(CA)8 to (CA)15] in the C3 promoter region showed significant association after Bonferroni correction, in the subgroup of MTLE patients having a personal history of FS (MTLE-FS+). Replication analysis in independent patients and controls confirmed that the rare HAP4 haplotype comprising the minimal length allele of GF100472 [(CA)8], protected against MTLE-FS+. A fifth haplotype (HAP5) with medium-size (CA)11 allele of GF100472 displayed four times higher frequency in controls than in the first cohort of MTLE-FS+ and showed a protective effect against FS through a high statistical significance in an independent population of 97 pure FS. Consistently, (CA)11 allele by its own protected against pure FS in a second group of 148 FS patients. Reporter gene assays showed that GF100472 significantly influenced C3 promoter activity (the higher the number of repeats, the lower the transcriptional activity). Taken together, the consistent genetic data and the functional analysis presented here indicate that a newly-identified and functional polymorphism in the promoter of the complement C3 gene might participate in the genetic susceptibility to human MTLE with a history of FS, and to pure FS. CONCLUSIONS/SIGNIFICANCE: The present study provides important data suggesting for the first time the involvement of the complement system in the genetic susceptibility to epileptic seizures and to epilepsy.


Subject(s)
Complement C3/genetics , Epilepsy, Temporal Lobe/genetics , Genetic Predisposition to Disease , Promoter Regions, Genetic , Seizures, Febrile/genetics , Adolescent , Case-Control Studies , Child , Child, Preschool , Female , Genetic Variation , Humans , Male , Microsatellite Repeats , Polymorphism, Single Nucleotide , Young Adult
18.
Acta Paediatr ; 97(5): 535-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18394095

ABSTRACT

BACKGROUND: Tick-borne encephalitis in children appears to be more benign than in adults and shows also a more favourable outcome. Only some authors report of sequelae like paralysis, paresis or seizures and behavioural abnormalities. The aim was to describe the clinical features of tick-borne encephalitis in children with special attention to sequelae and to review the literature. METHODS: Retrospective review of all charts of children with serologically confirmed tick-borne encephalitis hospitalised in Styria between 1981 and 2005. RESULTS: One hundred sixteen children were diagnosed with tick-borne encephalitis. Ninety-two children (79.3%) developed meningitis and 24 (20.7%) meningoencephalitis. Eleven patients with meningoencephalitis showed somnolence, 5 confusion, 5 tremors, 2 facial palsy, 1 ataxia, 1 epileptic seizure and 1 hemi paresis. Seven patients had to be admitted to the intensive care unit. Two children (1.7%) developed long time neurological sequelae: one epileptic seizure with requirement of antiepileptic therapy and one left-sided hemi paresis. One hundred twelve children had been not and 3 incompletely vaccinated against tick-borne encephalitis. Only one child had been fully vaccinated according to the Austrian vaccination schedule. CONCLUSION: Our study provides further evidence that tick-borne encephalitis in children has a substantial morbidity and in single cases severe long-time neurological sequelae are observed.


Subject(s)
Encephalitis, Tick-Borne/epidemiology , Meningoencephalitis/epidemiology , Adolescent , Austria/epidemiology , Child , Child, Preschool , Encephalitis, Tick-Borne/complications , Encephalitis, Tick-Borne/prevention & control , Humans , Incidence , Infant , Medical Records , Meningitis/classification , Meningitis/epidemiology , Meningitis/physiopathology , Meningoencephalitis/etiology , Meningoencephalitis/physiopathology , Retrospective Studies , Viral Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL
...