Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Brain ; 145(2): 555-568, 2022 04 18.
Article in English | MEDLINE | ID: mdl-35022648

ABSTRACT

Febrile seizures represent the most common type of pathological brain activity in young children and are influenced by genetic, environmental and developmental factors. In a minority of cases, febrile seizures precede later development of epilepsy. We conducted a genome-wide association study of febrile seizures in 7635 cases and 83 966 controls identifying and replicating seven new loci, all with P < 5 × 10-10. Variants at two loci were functionally related to altered expression of the fever response genes PTGER3 and IL10, and four other loci harboured genes (BSN, ERC2, GABRG2, HERC1) influencing neuronal excitability by regulating neurotransmitter release and binding, vesicular transport or membrane trafficking at the synapse. Four previously reported loci (SCN1A, SCN2A, ANO3 and 12q21.33) were all confirmed. Collectively, the seven novel and four previously reported loci explained 2.8% of the variance in liability to febrile seizures, and the single nucleotide polymorphism heritability based on all common autosomal single nucleotide polymorphisms was 10.8%. GABRG2, SCN1A and SCN2A are well-established epilepsy genes and, overall, we found positive genetic correlations with epilepsies (rg = 0.39, P = 1.68 × 10-4). Further, we found that higher polygenic risk scores for febrile seizures were associated with epilepsy and with history of hospital admission for febrile seizures. Finally, we found that polygenic risk of febrile seizures was lower in febrile seizure patients with neuropsychiatric disease compared to febrile seizure patients in a general population sample. In conclusion, this largest genetic investigation of febrile seizures to date implicates central fever response genes as well as genes affecting neuronal excitability, including several known epilepsy genes. Further functional and genetic studies based on these findings will provide important insights into the complex pathophysiological processes of seizures with and without fever.


Subject(s)
Epilepsy , Seizures, Febrile , Anoctamins/genetics , Child , Child, Preschool , Epilepsy/genetics , Fever/complications , Fever/genetics , Genome-Wide Association Study , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures, Febrile/genetics
2.
Ann Neurol ; 87(2): 281-288, 2020 02.
Article in English | MEDLINE | ID: mdl-31755124

ABSTRACT

OBJECTIVE: Febrile seizures may follow vaccination. Common variants in the sodium channel gene, SCN1A, are associated with febrile seizures, and rare pathogenic variants in SCN1A cause the severe developmental and epileptic encephalopathy Dravet syndrome. Following vaccination, febrile seizures may raise the specter of poor outcome and inappropriately implicate vaccination as the cause. We aimed to determine the prevalence of SCN1A variants in children having their first febrile seizure either proximal to vaccination or unrelated to vaccination compared to controls. METHODS: We performed SCN1A sequencing, blind to clinical category, in a prospective cohort of children presenting with their first febrile seizure as vaccine proximate (n = 69) or as non-vaccine proximate (n = 75), and children with no history of seizures (n = 90) recruited in Australian pediatric hospitals. RESULTS: We detected 2 pathogenic variants in vaccine-proximate cases (p.R568X and p.W932R), both of whom developed Dravet syndrome, and 1 in a non-vaccine-proximate case (p.V947L) who had febrile seizures plus from 9 months. All had generalized tonic-clonic seizures lasting >15 minutes. We also found enrichment of a reported risk allele, rs6432860-T, in children with febrile seizures compared to controls (odds ratio = 1.91, 95% confidence interval = 1.31-2.81). INTERPRETATION: Pathogenic SCN1A variants may be identified in infants with vaccine-proximate febrile seizures. As early diagnosis of Dravet syndrome is essential for optimal management and outcome, SCN1A sequencing in infants with prolonged febrile seizures, proximate to vaccination, should become routine. ANN NEUROL 2020;87:281-288.


Subject(s)
Genetic Predisposition to Disease/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures, Febrile/genetics , Vaccines/adverse effects , Case-Control Studies , Child, Preschool , Female , Humans , Infant , Male , Mutation , Prospective Studies
3.
Ann Neurol ; 86(6): 821-831, 2019 12.
Article in English | MEDLINE | ID: mdl-31618474

ABSTRACT

OBJECTIVE: Epilepsy of infancy with migrating focal seizures (EIMFS) is one of the most severe developmental and epileptic encephalopathies. We delineate the genetic causes and genotype-phenotype correlations of a large EIMFS cohort. METHODS: Phenotypic and molecular data were analyzed on patients recruited through an international collaborative study. RESULTS: We ascertained 135 patients from 128 unrelated families. Ninety-three of 135 (69%) had causative variants (42/55 previously reported) across 23 genes, including 9 novel EIMFS genes: de novo dominant GABRA1, GABRB1, ATP1A3; X-linked CDKL5, PIGA; and recessive ITPA, AIMP1, KARS, WWOX. The most frequently implicated genes were KCNT1 (36/135, 27%) and SCN2A (10/135, 7%). Mosaicism occurred in 2 probands (SCN2A, GABRB3) and 3 unaffected mothers (KCNT1). Median age at seizure onset was 4 weeks, with earlier onset in the SCN2A, KCNQ2, and BRAT1 groups. Epileptic spasms occurred in 22% patients. A total of 127 patients had severe to profound developmental impairment. All but 7 patients had ongoing seizures. Additional features included microcephaly, movement disorders, spasticity, and scoliosis. Mortality occurred in 33% at median age 2 years 7 months. INTERPRETATION: We identified a genetic cause in 69% of patients with EIMFS. We highlight the genetic heterogeneity of EIMFS with 9 newly implicated genes, bringing the total number to 33. Mosaicism was observed in probands and parents, carrying critical implications for recurrence risk. EIMFS pathophysiology involves diverse molecular processes from gene and protein regulation to ion channel function and solute trafficking. ANN NEUROL 2019;86:821-831.


Subject(s)
Genetic Predisposition to Disease/genetics , Seizures/diagnosis , Seizures/genetics , Spasms, Infantile/diagnosis , Spasms, Infantile/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Seizures/physiopathology , Spasms, Infantile/physiopathology
4.
Am J Hum Genet ; 99(2): 423-9, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27453577

ABSTRACT

Hypothalamic hamartoma (HH) with gelastic epilepsy is a well-recognized drug-resistant epilepsy syndrome of early life.(1) Surgical resection allows limited access to the small deep-seated lesions that cause the disease. Here, we report the results of a search for somatic mutations in paired hamartoma- and leukocyte-derived DNA samples from 38 individuals which we conducted by using whole-exome sequencing (WES), chromosomal microarray (CMA), and targeted resequencing (TRS) of candidate genes. Somatic mutations were identified in genes involving regulation of the sonic hedgehog (Shh) pathway in 14/38 individuals (37%). Three individuals had somatic mutations in PRKACA, which encodes a cAMP-dependent protein kinase that acts as a repressor protein in the Shh pathway, and four subjects had somatic mutations in GLI3, an Shh pathway gene associated with HH. In seven other individuals, we identified two recurrent and three single brain-tissue-specific, large copy-number or loss-of-heterozygosity (LOH) variants involving multiple Shh genes, as well as other genes without an obvious biological link to the Shh pathway. The Shh pathway genes in these large somatic lesions include the ligand itself (SHH and IHH), the receptor SMO, and several other Shh downstream pathway members, including CREBBP and GLI2. Taken together, our data implicate perturbation of the Shh pathway in at least 37% of individuals with the HH epilepsy syndrome, consistent with the concept of a developmental pathway brain disease.


Subject(s)
Epilepsies, Partial/genetics , Hamartoma/genetics , Hedgehog Proteins/metabolism , Hypothalamic Diseases/genetics , Mutation/genetics , Signal Transduction/genetics , CREB-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Exome/genetics , Female , Humans , Kruppel-Like Transcription Factors/genetics , Loss of Heterozygosity , Male , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
5.
Hum Mutat ; 39(2): 202-209, 2018 02.
Article in English | MEDLINE | ID: mdl-29064616

ABSTRACT

Genetic generalized epilepsy (GGE) is a common epilepsy syndrome that encompasses seizure disorders characterized by spike-and-wave discharges (SWDs). Pacemaker hyperpolarization-activated cyclic nucleotide-gated channels (HCN) are considered integral to SWD genesis, making them an ideal gene candidate for GGE. We identified HCN2 missense variants from a large cohort of 585 GGE patients, recruited by the Epilepsy Phenome-Genome Project (EPGP), and performed functional analysis using two-electrode voltage clamp recordings from Xenopus oocytes. The p.S632W variant was identified in a patient with idiopathic photosensitive occipital epilepsy and segregated in the family. This variant was also independently identified in an unrelated patient with childhood absence seizures from a European cohort of 238 familial GGE cases. The p.V246M variant was identified in a patient with photo-sensitive GGE and his father diagnosed with juvenile myoclonic epilepsy. Functional studies revealed that both p.S632W and p.V246M had an identical functional impact including a depolarizing shift in the voltage dependence of activation that is consistent with a gain-of-function. In contrast, no biophysical changes resulted from the introduction of common population variants, p.E280K and p.A705T, and the p.R756C variant from EPGP that did not segregate with disease. Our data suggest that HCN2 variants can confer susceptibility to GGE via a gain-of-function mechanism.


Subject(s)
DNA, Complementary/genetics , Epilepsy, Generalized/genetics , Epilepsy/genetics , Gain of Function Mutation/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Electrophysiology , Female , Humans , Male , Models, Biological , Pedigree
6.
Epilepsia ; 58(2): e26-e30, 2017 02.
Article in English | MEDLINE | ID: mdl-28084635

ABSTRACT

Genetic epilepsy with febrile seizures plus (GEFS+) is a familial epilepsy syndrome characterized by heterogeneous phenotypes ranging from mild disorders such as febrile seizures to epileptic encephalopathies (EEs) such as Dravet syndrome (DS). Although DS often occurs with de novo SCN1A pathogenic variants, milder GEFS+ spectrum phenotypes are associated with inherited pathogenic variants. We identified seven cases with non-EE GEFS+ phenotypes and de novo SCN1A pathogenic variants, including a monozygotic twin pair. Febrile seizures plus (FS+) occurred in six patients, five of whom had additional seizure types. The remaining case had childhood-onset temporal lobe epilepsy without known febrile seizures. Although early development was normal in all individuals, three later had learning difficulties, and the twin girls had language impairment and working memory deficits. All cases had SCN1A missense pathogenic variants that were not found in either parent. One pathogenic variant had been reported previously in a case of DS, and the remainder were novel. Our finding of de novo pathogenic variants in mild phenotypes within the GEFS+ spectrum shows that mild GEFS+ is not always inherited. SCN1A screening should be considered in patients with GEFS+ phenotypes because identification of pathogenic variants will influence antiepileptic therapy, and prognostic and genetic counseling.


Subject(s)
Epilepsies, Myoclonic/genetics , Family Health , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Seizures, Febrile/genetics , Adolescent , Adult , Child , Electroencephalography , Female , Humans , Male , Middle Aged , Models, Molecular , Young Adult
7.
Epilepsia ; 58(3): e40-e43, 2017 03.
Article in English | MEDLINE | ID: mdl-28098945

ABSTRACT

Synaptic proteins are critical to neuronal function in the brain, and their deficiency can lead to seizures and cognitive impairments. CNKSR2 (connector enhancer of KSR2) is a synaptic protein involved in Ras signaling-mediated neuronal proliferation, migration and differentiation. Mutations in the X-linked gene CNKSR2 have been described in patients with seizures and neurodevelopmental deficits, especially those affecting language. In this study, we sequenced 112 patients with phenotypes within the epilepsy-aphasia spectrum (EAS) to determine the frequency of CNKSR2 mutation within this complex set of disorders. We detected a novel nonsense mutation (c.2314 C>T; p.Arg712*) in one Ashkenazi Jewish family, the male proband of which had a severe epileptic encephalopathy with continuous spike-waves in sleep (ECSWS). His affected brother also had ECSWS with better outcome, whereas the sister had childhood epilepsy with centrotemporal spikes. This mutation segregated in the three affected siblings in an X-linked manner, inherited from their mother who had febrile seizures. Although the frequency of point mutation is low, CNKSR2 sequencing should be considered in families with suspected X-linked EAS because of the specific genetic counseling implications.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Aphasia/genetics , Mutation/genetics , Spasms, Infantile/genetics , Aphasia/physiopathology , Cohort Studies , DNA Mutational Analysis , Electroencephalography , Family Health , Female , Humans , Infant , Male , Phenotype , Sleep/physiology , Spasms, Infantile/physiopathology
8.
Radiology ; 281(3): 896-906, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27355897

ABSTRACT

Purpose To investigate whether it is possible in patients with periventricular nodular heterotopia (PVNH) to detect abnormal fiber projections that have only previously been reported in the histopathology literature. Materials and Methods Whole-brain diffusion-weighted (DW) imaging data from 14 patients with bilateral PVNH and 14 age- and sex-matched healthy control subjects were prospectively acquired by using 3.0-T magnetic resonance (MR) imaging between August 1, 2008, and December 5, 2012. All participants provided written informed consent. The DW imaging data were processed to generate whole-brain constrained spherical deconvolution (CSD)-based tractography data and super-resolution track-density imaging (TDI) maps. The tractography data were overlaid on coregistered three-dimensional T1-weighted images to visually assess regions of heterotopia. A panel of MR imaging researchers independently assessed each case and indicated numerically (no = 1, yes = 2) as to the presence of abnormal fiber tracks in nodular tissue. The Fleiss κ statistical measure was applied to assess the reader agreement. Results Abnormal fiber tracks emanating from one or more regions of heterotopia were reported by all four readers in all 14 patients with PVNH (Fleiss κ = 1). These abnormal structures were not visible on the tractography data from any of the control subjects and were not discernable on the conventional T1-weighted images of the patients with PVNH. Conclusion Whole-brain CSD-based fiber tractography and super-resolution TDI mapping reveals abnormal fiber projections in nodular tissue suggestive of abnormal organization of white matter (with abnormal fibers both within nodules and projecting to the surrounding white matter) in patients with bilateral PVNH. © RSNA, 2016.


Subject(s)
Epilepsy/pathology , Periventricular Nodular Heterotopia/pathology , Adolescent , Adult , Aged , Diffusion Magnetic Resonance Imaging , Diffusion Tensor Imaging , Epilepsy/genetics , Female , Filamins/genetics , Humans , Infant , Male , Middle Aged , Mutation/genetics , Periventricular Nodular Heterotopia/genetics , Prospective Studies , Young Adult
9.
J Immunol ; 191(1): 424-33, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23729444

ABSTRACT

Glucocorticoid-induced leucine zipper (GILZ) is an anti-inflammatory protein first identified in T lymphocytes. We recently observed that GILZ is highly expressed in synovial endothelial cells in rheumatoid arthritis. However, the function of GILZ in endothelial cells is unknown. To investigate the actions of GILZ in this cell type, we induced GILZ expression in HUVECs via transient transfection. GILZ overexpression significantly reduced the capacity of TNF-stimulated HUVECs to support leukocyte rolling, adhesion, and transmigration. These effects were associated with decreased expression of E-selectin, ICAM-1, CCL2, CXCL8, and IL-6. Experiments in a human microvascular endothelial cell line demonstrated that TNF-inducible NF-κB activity was significantly inhibited by overexpression of GILZ. Exogenous GILZ inhibited TNF-induced NF-κB p65 DNA binding, although this occurred in the absence of an effect on p65 nuclear translocation, indicating that the mechanism of action of exogenous GILZ in endothelial cells differs from that reported in other cell types. GILZ overexpression also inhibited TNF-induced activation of p38, ERK, and JNK MAPKs, as well as increased expression of the MAPK inhibitory phosphatase, MKP-1. In contrast, silencing endogenous GILZ in glucocorticoid-treated HUVECs did not alter their capacity to support leukocyte interactions. These data demonstrate that exogenous GILZ exerts inhibitory effects on endothelial cell adhesive function via a novel pathway involving modulation of NF-κB p65 DNA binding and MAPK activity. Induction of GILZ expression in endothelial cells may represent a novel therapeutic modality with the potential to inhibit inflammatory leukocyte recruitment.


Subject(s)
Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , MAP Kinase Signaling System/immunology , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transendothelial and Transepithelial Migration/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Communication/immunology , Cell Line , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes/immunology , Leukocytes/metabolism , MAP Kinase Signaling System/genetics , Microcirculation/genetics , Microcirculation/immunology , Primary Cell Culture , Random Allocation , Transcription Factors/biosynthesis , Transcription Factors/physiology , Transendothelial and Transepithelial Migration/genetics
10.
Genes (Basel) ; 14(8)2023 07 31.
Article in English | MEDLINE | ID: mdl-37628618

ABSTRACT

Aicardi Syndrome (AIC) is a rare neurodevelopmental disorder recognized by the classical triad of agenesis of the corpus callosum, chorioretinal lacunae and infantile epileptic spasms syndrome. The diagnostic criteria of AIC were revised in 2005 to include additional phenotypes that are frequently observed in this patient group. AIC has been traditionally considered as X-linked and male lethal because it almost exclusively affects females. Despite numerous genetic and genomic investigations on AIC, a unifying X-linked cause has not been identified. Here, we performed exome and genome sequencing of 10 females with AIC or suspected AIC based on current criteria. We identified a unique de novo variant, each in different genes: KMT2B, SLF1, SMARCB1, SZT2 and WNT8B, in five of these females. Notably, genomic analyses of coding and non-coding single nucleotide variants, short tandem repeats and structural variation highlighted a distinct lack of X-linked candidate genes. We assessed the likely pathogenicity of our candidate autosomal variants using the TOPflash assay for WNT8B and morpholino knockdown in zebrafish (Danio rerio) embryos for other candidates. We show expression of Wnt8b and Slf1 are restricted to clinically relevant cortical tissues during mouse development. Our findings suggest that AIC is genetically heterogeneous with implicated genes converging on molecular pathways central to cortical development.


Subject(s)
Aicardi Syndrome , Male , Female , Animals , Mice , Aicardi Syndrome/genetics , Zebrafish/genetics , Chromosome Mapping , Genes, X-Linked/genetics , Biological Assay
11.
Neurol Genet ; 8(1): e652, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35097204

ABSTRACT

BACKGROUND AND OBJECTIVES: The 2-hit model of genetic disease is well established in cancer, yet has only recently been reported to cause brain malformations associated with epilepsy. Pathogenic germline and somatic variants in genes in the mechanistic target of rapamycin (mTOR) pathway have been implicated in several malformations of cortical development. We investigated the 2-hit model by performing genetic analysis and searching for germline and somatic variants in genes in the mTOR and related pathways. METHODS: We searched for germline and somatic pathogenic variants in 2 brothers with drug-resistant focal epilepsy and surgically resected focal cortical dysplasia (FCD) type IIA. Exome sequencing was performed on blood- and brain-derived DNA to identify pathogenic variants, which were validated by droplet digital PCR. In vitro functional assays of a somatic variant were performed. RESULTS: Exome analysis revealed a novel, maternally inherited, germline pathogenic truncation variant (c.48delG; p.Ser17Alafs*70) in NPRL3 in both brothers. NPRL3 is a known FCD gene that encodes a negative regulator of the mTOR pathway. Somatic variant calling in brain-derived DNA from both brothers revealed a low allele fraction somatic variant (c.338C>T; p.Ala113Val) in the WNT2 gene in 1 brother, confirmed by droplet digital PCR. In vitro functional studies suggested a loss of WNT2 function as a consequence of this variant. A second somatic variant has not yet been found in the other brother. DISCUSSION: We identify a pathogenic germline mTOR pathway variant (NPRL3) and a somatic variant (WNT2) in the intersecting WNT signaling pathway, potentially implicating the WNT2 gene in FCD and supporting a dual-pathway 2-hit model. If confirmed in other cases, this would extend the 2-hit model to pathogenic variants in different genes in critical, intersecting pathways in a malformation of cortical development. Detection of low allele fraction somatic second hits is challenging but promises to unravel the molecular architecture of FCDs.

12.
Am J Hum Genet ; 82(1): 19-31, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18179881

ABSTRACT

We describe a distinct retinal disorder, autosomal-recessive bestrophinopathy (ARB), that is consequent upon biallelic mutation in BEST1 and is associated with central visual loss, a characteristic retinopathy, an absent electro-oculogram light rise, and a reduced electroretinogram. Heterozygous mutations in BEST1 have previously been found to cause the two dominantly inherited disorders, Best macular dystrophy and autosomal-dominant vitreoretinochoroidopathy. The transmembrane protein bestrophin-1, encoded by BEST1, is located at the basolateral membrane of the retinal pigment epithelium in which it probably functions as a Cl(-) channel. We sequenced BEST1 in five families, identifying DNA variants in each of ten alleles. These encoded six different missense variants and one nonsense variant. The alleles segregated appropriately for a recessive disorder in each family. No clinical or electrophysiological abnormalities were identified in any heterozygotes. We conducted whole-cell patch-clamping of HEK293 cells transfected with bestrophin-1 to measure the Cl(-) current. Two ARB missense isoforms severely reduced channel activity. However, unlike two other alleles previously associated with Best disease, cotransfection with wild-type bestrophin-1 did not impair the formation of active wild-type bestrophin-1 channels, consistent with the recessive nature of the condition. We propose that ARB is the null phenotype of bestrophin-1 in humans.


Subject(s)
Chloride Channels/genetics , Eye Proteins/genetics , Retinal Diseases/genetics , Adolescent , Adult , Amino Acid Sequence , Bestrophins , Cell Line , Child , Child, Preschool , Chloride Channels/chemistry , Codon, Nonsense , Eye Proteins/chemistry , Female , Genes, Recessive , Humans , Male , Molecular Sequence Data , Mutation, Missense , Transfection
13.
Neurology ; 92(2): e96-e107, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30541864

ABSTRACT

OBJECTIVE: To delineate the epileptology, a key part of the SYNGAP1 phenotypic spectrum, in a large patient cohort. METHODS: Patients were recruited via investigators' practices or social media. We included patients with (likely) pathogenic SYNGAP1 variants or chromosome 6p21.32 microdeletions incorporating SYNGAP1. We analyzed patients' phenotypes using a standardized epilepsy questionnaire, medical records, EEG, MRI, and seizure videos. RESULTS: We included 57 patients (53% male, median age 8 years) with SYNGAP1 mutations (n = 53) or microdeletions (n = 4). Of the 57 patients, 56 had epilepsy: generalized in 55, with focal seizures in 7 and infantile spasms in 1. Median seizure onset age was 2 years. A novel type of drop attack was identified comprising eyelid myoclonia evolving to a myoclonic-atonic (n = 5) or atonic (n = 8) seizure. Seizure types included eyelid myoclonia with absences (65%), myoclonic seizures (34%), atypical (20%) and typical (18%) absences, and atonic seizures (14%), triggered by eating in 25%. Developmental delay preceded seizure onset in 54 of 56 (96%) patients for whom early developmental history was available. Developmental plateauing or regression occurred with seizures in 56 in the context of a developmental and epileptic encephalopathy (DEE). Fifty-five of 57 patients had intellectual disability, which was moderate to severe in 50. Other common features included behavioral problems (73%); high pain threshold (72%); eating problems, including oral aversion (68%); hypotonia (67%); sleeping problems (62%); autism spectrum disorder (54%); and ataxia or gait abnormalities (51%). CONCLUSIONS: SYNGAP1 mutations cause a generalized DEE with a distinctive syndrome combining epilepsy with eyelid myoclonia with absences and myoclonic-atonic seizures, as well as a predilection to seizures triggered by eating.


Subject(s)
Developmental Disabilities/genetics , Mutation/genetics , Spasms, Infantile/genetics , ras GTPase-Activating Proteins/genetics , Adolescent , Adult , Anticonvulsants/therapeutic use , Brain/diagnostic imaging , Brain Diseases/complications , Brain Diseases/diagnostic imaging , Brain Diseases/genetics , Child , Child, Preschool , Cohort Studies , Developmental Disabilities/complications , Developmental Disabilities/diagnostic imaging , Electroencephalography , Female , Genetic Association Studies , Humans , Infant , Male , Spasms, Infantile/complications , Spasms, Infantile/diagnostic imaging , Spasms, Infantile/drug therapy , Young Adult
14.
Genome Med ; 9(1): 55, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28595657

ABSTRACT

BACKGROUND: The pathogenesis of neurological and mental health disorders often involves multiple genes, complex interactions, as well as brain- and development-specific biological mechanisms. These characteristics make identification of disease genes for such disorders challenging, as conventional prioritisation tools are not specifically tailored to deal with the complexity of the human brain. Thus, we developed a novel web-application-brain-coX-that offers gene prioritisation with accompanying visualisations based on seven gene expression datasets in the post-mortem human brain, the largest such resource ever assembled. RESULTS: We tested whether our tool can correctly prioritise known genes from 37 brain-specific KEGG pathways and 17 psychiatric conditions. We achieved average sensitivity of nearly 50%, at the same time reaching a specificity of approximately 75%. We also compared brain-coX's performance to that of its main competitors, Endeavour and ToppGene, focusing on the ability to discover novel associations. Using a subset of the curated SFARI autism gene collection we show that brain-coX's prioritisations are most similar to SFARI's own curated gene classifications. CONCLUSIONS: brain-coX is the first prioritisation and visualisation web-tool targeted to the human brain and can be freely accessed via http://shiny.bioinf.wehi.edu.au/freytag.s/ .


Subject(s)
Brain/metabolism , Computational Biology/methods , Software , Transcriptome , Databases, Genetic , Gene Expression Profiling , Humans , Sensitivity and Specificity
15.
Epileptic Disord ; 19(4): 450-455, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29258966

ABSTRACT

Somatic mutation of the lissencephaly-1 gene is a cause of subcortical band heterotopia ("double cortex"). The severity of the phenotype depends on the level of mutation in brain tissue. Detecting and quantifying low-level somatic mosaic mutations is challenging. Here, we utilized droplet digital PCR, a sensitive method to detect low-level mutation. Droplet digital PCR was used in concert with classic genotyping techniques (SNaPshot assays and pyrosequencing) to detect and characterize the tissue mosaicism of a somatic mutation (LIS1 c.190A>T; p.K64X) in a patient with posterior bilateral SBH and refractory epilepsy. The high sensitivity of droplet digital PCR and the ability to target individual DNA molecules allowed us to detect the mutation at low level in the brain, despite the low quality of the DNA sample derived from formalin-fixed paraffin-embedded tissue. This low mutation frequency in the brain was consistent with the relatively subtle malformation resolved by magnetic resonance imaging. The presence of the mutation in other tissues from the patient permitted us to predict the timing of mutagenesis. This sensitive methodology will have utility for a variety of other brain malformation syndromes associated with epilepsy for which historical pathological specimens are available and specific somatic mosaic mutations are predicted.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Drug Resistant Epilepsy/genetics , Epilepsies, Partial/genetics , Microtubule-Associated Proteins/genetics , Mutation , Adult , Female , Humans , Intellectual Disability/genetics , Language Development Disorders/genetics , Real-Time Polymerase Chain Reaction
16.
Neurology ; 89(12): 1210-1219, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28842445

ABSTRACT

OBJECTIVE: Following our original description of generalized epilepsy with febrile seizures plus (GEFS+) in 1997, we analyze the phenotypic spectrum in 409 affected individuals in 60 families (31 new families) and expand the GEFS+ spectrum. METHODS: We performed detailed electroclinical phenotyping on all available affected family members. Genetic analysis of known GEFS+ genes was carried out where possible. We compared our phenotypic and genetic data to those published in the literature over the last 19 years. RESULTS: We identified new phenotypes within the GEFS+ spectrum: focal seizures without preceding febrile seizures (16/409 [4%]), classic genetic generalized epilepsies (22/409 [5%]), and afebrile generalized tonic-clonic seizures (9/409 [2%]). Febrile seizures remains the most frequent phenotype in GEFS+ (178/409 [44%]), followed by febrile seizures plus (111/409 [27%]). One third (50/163 [31%]) of GEFS+ families tested have a pathogenic variant in a known GEFS+ gene. CONCLUSION: As 37/409 (9%) affected individuals have focal epilepsies, we suggest that GEFS+ be renamed genetic epilepsy with febrile seizures plus rather than generalized epilepsy with febrile seizures plus. The phenotypic overlap between GEFS+ and the classic generalized epilepsies is considerably greater than first thought. The clinical and molecular data suggest that the 2 major groups of generalized epilepsies share genetic determinants.


Subject(s)
Epilepsies, Partial/physiopathology , Epilepsy, Generalized/physiopathology , Seizures, Febrile/physiopathology , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Epilepsies, Partial/genetics , Epilepsy, Generalized/genetics , Female , Humans , Infant , Male , Middle Aged , Pedigree , Phenotype , Seizures, Febrile/genetics , Young Adult
17.
FEBS Lett ; 580(1): 175-8, 2006 Jan 09.
Article in English | MEDLINE | ID: mdl-16364304

ABSTRACT

Protein-protein interactions between SHEP and Cas proteins influence cellular signaling through tyrosine kinases, as well as integrin-mediated signaling, and may be linked to antiestrogen resistance. Data from past studies suggests that association between SHEP and Cas proteins is critical for these cellular effects. In this study, the interacting domains of each protein were co-expressed in bacteria and a soluble stable complex was purified. Deuterium exchange mass spectrometry was used to define regions that are buried when SHEP1 is in complex with Cas. The results reveal four segments in SHEP1 that are highly protected, including a region (residues 619-640) that contains a key residue, tyrosine 635, required for association with Cas. This region is predominately hydrophilic, yet remains protected from solvent in the complex.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Crk-Associated Substrate Protein/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Animals , Crk-Associated Substrate Protein/metabolism , Deuterium , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Solvents/chemistry
18.
Genetics ; 169(4): 2035-45, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15654087

ABSTRACT

Much of our understanding of the genetic mechanisms that control planar cell polarity (PCP) in epithelia has derived from studies of the formation of polarized cell hairs during Drosophila wing development. The correct localization of an F-actin prehair to the distal vertex of the pupal wing cell has been shown to be dependent upon the polarized subcellular localization of Frizzled and other core PCP proteins. However, the core PCP proteins do not organize actin cytoskeletal polarity directly but require PCP effector proteins such as Fuzzy and Inturned to mediate this process. Here we describe the characterization of a new PCP effector gene, fritz, that encodes a novel but evolutionarily conserved coiled-coil WD40 protein. We show that the fritz gene product functions cell-autonomously downstream of the core PCP proteins to regulate both the location and the number of wing cell prehair initiation sites.


Subject(s)
Cytoskeleton/metabolism , Drosophila Proteins/biosynthesis , Drosophila melanogaster/metabolism , Epidermis/metabolism , Glycoproteins/physiology , Membrane Proteins/biosynthesis , Actins/metabolism , Alleles , Amino Acid Sequence , Animals , Body Patterning , Cell Lineage , Chromosome Mapping , Cold Temperature , Conserved Sequence , Evolution, Molecular , Frizzled Receptors , Gene Expression Regulation , Glycoproteins/chemistry , Humans , Intracellular Signaling Peptides and Proteins , Models, Genetic , Molecular Sequence Data , Mutation , Phenotype , Receptors, G-Protein-Coupled , Sequence Homology, Amino Acid , Temperature , Wings, Animal/metabolism
19.
Epilepsy Res ; 128: 48-51, 2016 12.
Article in English | MEDLINE | ID: mdl-27810516

ABSTRACT

Mutation of fibroblast growth factor 13 (FGF13) has recently been implicated in genetic epilepsy with febrile seizures plus (GEFS+) in a single family segregating a balanced translocation with a breakpoint in this X chromosome gene, predicting a partial knockout involving 3 of 5 known FGF13 isoforms. Investigation of a mouse model of complete Fgf13 knock-out revealed increased susceptibility to hyperthermia-induced seizures and epilepsy. Here we investigated whether mutation of FGF13 would explain other cases of GEFS+ compatible with X-linked inheritance. We screened the coding and splice site regions of the FGF13 gene in a sample of 45 unrelated probands where GEFS+ segregated in an X-linked pattern. We subsequently identified a de novo FGF13 missense variant in an additional patient with febrile seizures and facial edema. Our data suggests FGF13 is not a common cause of GEFS+.


Subject(s)
Epilepsy, Generalized/genetics , Fibroblast Growth Factors/genetics , Seizures, Febrile/genetics , Child, Preschool , Cohort Studies , Edema/genetics , Face , Female , Genes, X-Linked , Genetic Testing , Humans , Male , Mutation, Missense , Phenotype , SOXF Transcription Factors/genetics
20.
Sci Rep ; 5: 17816, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26647834

ABSTRACT

Febrile seizures (FS) are the most common seizure syndrome and are potentially a prelude to more severe epilepsy. Although zinc (Zn(2+)) metabolism has previously been implicated in FS, whether or not variation in proteins essential for Zn(2+) homeostasis contributes to susceptibility is unknown. Synaptic Zn(2+) is co-released with glutamate and modulates neuronal excitability. SLC30A3 encodes the zinc transporter 3 (ZNT3), which is primarily responsible for moving Zn(2+) into synaptic vesicles. Here we sequenced SLC30A3 and discovered a rare variant (c.892C > T; p.R298C) enriched in FS populations but absent in population-matched controls. Functional analysis revealed a significant loss-of-function of the mutated protein resulting from a trafficking deficit. Furthermore, mice null for ZnT3 were more sensitive than wild-type to hyperthermia-induced seizures that model FS. Together our data suggest that reduced synaptic Zn(2+) increases the risk of FS and more broadly support the idea that impaired synaptic Zn(2+) homeostasis can contribute to neuronal hyperexcitability.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Seizures, Febrile/genetics , Seizures, Febrile/metabolism , Zinc/metabolism , Amino Acid Sequence , Animals , Case-Control Studies , Cation Transport Proteins/chemistry , Cell Line , DNA Mutational Analysis , Genetic Predisposition to Disease , Humans , Inheritance Patterns , Kaplan-Meier Estimate , Mice, Knockout , Molecular Sequence Data , Mutation , Pedigree , Rats , Risk , Seizures, Febrile/mortality , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL