Medicine (Austin & Northern Health) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 17
  • Item
    Thumbnail Image
    Gain-of-function HCN2 variants in genetic epilepsy
    Li, M ; Maljevic, S ; Phillips, AM ; Petrovski, S ; Hildebrand, MS ; Burgess, R ; Mount, T ; Zara, F ; Striano, P ; Schubert, J ; Thiele, H ; Nuernberg, P ; Wong, M ; Weisenberg, JL ; Thio, LL ; Lerche, H ; Scheffer, IE ; Berkovic, SF ; Petrou, S ; Reid, CA (WILEY, 2018-02)
    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.
  • Item
    Thumbnail Image
    Frequency of CNKSR2 mutation in the X-linked epilepsy-aphasia spectrum
    Damiano, JA ; Burgess, R ; Kivity, S ; Lerman-Sagie, T ; Afawi, Z ; Scheffer, IE ; Berkovic, SF ; Hildebrand, MS (WILEY, 2017-03)
    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.
  • Item
    Thumbnail Image
    De novo SCN1A pathogenic variants in the GEFS plus spectrum: Not always a familial syndrome
    Myers, KA ; Burgess, R ; Afawi, Z ; Damiano, JA ; Berkovic, SF ; Hildebrand, MS ; Scheffer, IE (WILEY, 2017-02)
    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.
  • Item
    Thumbnail Image
    The Genetic Landscape of Epilepsy of Infancy with Migrating Focal Seizures
    Burgess, R ; Wang, S ; McTague, A ; Boysen, KE ; Yang, X ; Zeng, Q ; Myers, KA ; Rochtus, A ; Trivisano, M ; Gill, D ; Sadleir, LG ; Specchio, N ; Guerrini, R ; Marini, C ; Zhang, Y-H ; Mefford, HC ; Kurian, MA ; Poduri, AH ; Scheffer, IE (WILEY, 2019-12)
    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.
  • Item
    Thumbnail Image
    Epilepsy, hippocampal sclerosis and febrile seizures linked by common genetic variation around SCN1A
    Kasperaviciute, D ; Catarino, CB ; Matarin, M ; Leu, C ; Novy, J ; Tostevin, A ; Leal, B ; Hessel, EVS ; Hallmann, K ; Hildebrand, MS ; Dahl, H-HM ; Ryten, M ; Trabzuni, D ; Ramasamy, A ; Alhusaini, S ; Doherty, CP ; Dorn, T ; Hansen, J ; Kraemer, G ; Steinhoff, BJ ; Zumsteg, D ; Duncan, S ; Kaelviaeinen, RK ; Eriksson, KJ ; Kantanen, A-M ; Pandolfo, M ; Gruber-Sedlmayr, U ; Schlachter, K ; Reinthaler, EM ; Stogmann, E ; Zimprich, F ; Theatre, E ; Smith, C ; O'Brien, TJ ; Tan, KM ; Petrovski, S ; Robbiano, A ; Paravidino, R ; Zara, F ; Striano, P ; Sperling, MR ; Buono, RJ ; Hakonarson, H ; Chaves, J ; Costa, PP ; Silva, BM ; da Silva, AM ; de Graan, PNE ; Koeleman, BPC ; Becker, A ; Schoch, S ; von Lehe, M ; Reif, PS ; Rosenow, F ; Becker, F ; Weber, Y ; Lerche, H ; Roessler, K ; Buchfelder, M ; Hamer, HM ; Kobow, K ; Coras, R ; Blumcke, I ; Scheffer, IE ; Berkovic, SF ; Weale, ME ; Delanty, N ; Depondt, C ; Cavalleri, GL ; Kunz, WS ; Sisodiya, SM (OXFORD UNIV PRESS, 2013-10)
    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.
  • Item
    Thumbnail Image
    Is a microRNA-328 binding site in PAX6 associated with Rolandic epilepsy?
    McGlade, A ; Myers, KA ; Berkovic, SF ; Scheffer, IE ; Petrovski, S ; Hildebrand, MS (WILEY, 2017-04)
  • Item
    Thumbnail Image
    PRIMA1 mutation: a new cause of nocturnal frontal lobe epilepsy
    Hildebrand, MS ; Tankard, R ; Gazina, EV ; Damiano, JA ; Lawrence, KM ; Dahl, H-HM ; Regan, BM ; Shearer, AE ; Smith, RJH ; Marini, C ; Guerrini, R ; Labate, A ; Gambardella, A ; Tinuper, P ; Lichetta, L ; Baldassari, S ; Bisulli, F ; Pippucci, T ; Scheffer, IE ; Reid, CA ; Petrou, S ; Bahlo, M ; Berkovic, SF (WILEY, 2015-08)
    OBJECTIVE: Nocturnal frontal lobe epilepsy (NFLE) can be sporadic or autosomal dominant; some families have nicotinic acetylcholine receptor subunit mutations. We report a novel autosomal recessive phenotype in a single family and identify the causative gene. METHODS: Whole exome sequencing data was used to map the family, thereby narrowing exome search space, and then to identify the mutation. RESULTS: Linkage analysis using exome sequence data from two affected and two unaffected subjects showed homozygous linkage peaks on chromosomes 7, 8, 13, and 14 with maximum LOD scores between 1.5 and 1.93. Exome variant filtering under these peaks revealed that the affected siblings were homozygous for a novel splice site mutation (c.93+2T>C) in the PRIMA1 gene on chromosome 14. No additional PRIMA1 mutations were found in 300 other NFLE cases. The c.93+2T>C mutation was shown to lead to skipping of the first coding exon of the PRIMA1 mRNA using a minigene system. INTERPRETATION: PRIMA1 is a transmembrane protein that anchors acetylcholinesterase (AChE), an enzyme hydrolyzing acetycholine, to membrane rafts of neurons. PRiMA knockout mice have reduction of AChE and accumulation of acetylcholine at the synapse; our minigene analysis suggests that the c.93+2T>C mutation leads to knockout of PRIMA1. Mutations with gain of function effects in acetylcholine receptor subunits cause autosomal dominant NFLE. Thus, enhanced cholinergic responses are the likely cause of the severe NFLE and intellectual disability segregating in this family, representing the first recessive case to be reported and the first PRIMA1 mutation implicated in disease.
  • Item
    Thumbnail Image
    KANSL1 variation is not a major contributing factor in self-limited focal epilepsy syndromes of childhood
    Myers, KA ; McGlade, A ; Neubauer, BA ; Lal, D ; Berkovic, SF ; Scheffer, IE ; Hildebrand, MS ; van Luijtelaar, G (PUBLIC LIBRARY SCIENCE, 2018-01-19)
    BACKGROUND: KANSL1 haploinsufficiency causes Koolen-de Vries syndrome (KdVS), characterized by dysmorphic features and intellectual disability; amiable personality, congenital malformations and seizures also commonly occur. The epilepsy phenotypic spectrum in KdVS is broad, but most individuals have focal seizures with some having a phenotype resembling the self-limited focal epilepsies of childhood (SFEC). We hypothesized that variants in KANSL1 contribute to pathogenesis of SFEC. MATERIALS AND METHODS: We screened KANSL1 for single nucleotide variants in 90 patients with SFEC. We then screened a cohort of 208 patients with two specific SFEC syndromes, childhood epilepsy with centrotemporal spikes (CECTS) and atypical childhood epilepsy with centrotemporal spikes (ACECTS) for KANSL1 variants. The second cohort was also used to evaluate minor allelic variants that appeared overrepresented in the initial cohort. RESULTS: One variant, p.Lys104Thr, was predicted damaging and appeared overrepresented in our 90-patient cohort compared to Genome Aggregation Database (gnomAD) allele frequency (0.217 to 0.116, with no homozygotes in gnomAD). However, there was no difference in p.Lys104Thr allele frequency in the follow-up CECTS/ACECTS cohort and controls. Four rare KANSL1 variants of uncertain significance were identified in the CECTS/ACECTS cohort. DISCUSSION: Our data do not support a major role for KANSL1 variants in pathogenesis of SFEC.
  • Item
    Thumbnail Image
    Somatic GNAQ mutation in the forme fruste of Sturge-Weber syndrome
    Hildebrand, MS ; Harvey, AS ; Malone, S ; Damiano, JA ; Do, H ; Ye, Z ; McQuillan, L ; Maixner, W ; Kalnins, R ; Nolan, B ; Wood, M ; Ozturk, E ; Jones, NC ; Gillies, G ; Pope, K ; Lockhart, PJ ; Dobrovic, A ; Leventer, RJ ; Scheffer, IE ; Berkovic, SF (LIPPINCOTT WILLIAMS & WILKINS, 2018-06)
    OBJECTIVE: To determine whether the GNAQ R183Q mutation is present in the forme fruste cases of Sturge-Weber syndrome (SWS) to establish a definitive molecular diagnosis. METHODS: We used sensitive droplet digital PCR (ddPCR) to detect and quantify the GNAQ mutation in tissues from epilepsy surgery in 4 patients with leptomeningeal angiomatosis; none had ocular or cutaneous manifestations. RESULTS: Low levels of the GNAQ mutation were detected in the brain tissue of all 4 cases-ranging from 0.42% to 7.1% frequency-but not in blood-derived DNA. Molecular evaluation confirmed the diagnosis in 1 case in which the radiologic and pathologic data were equivocal. CONCLUSIONS: We detected the mutation at low levels, consistent with mosaicism in the brain or skin (1.0%-18.1%) of classic cases. Our data confirm that the forme fruste is part of the spectrum of SWS, with the same molecular mechanism as the classic disease and that ddPCR is helpful where conventional diagnosis is uncertain.
  • Item
    Thumbnail Image
    Development of a rapid functional assay that predicts GLUT1 disease severity
    Zaman, SM ; Mullen, SA ; Petrovski, S ; Maljevic, S ; Gazina, E ; Phillips, AM ; Jones, GD ; Hildebrand, MS ; Damiano, J ; Auvin, S ; Lerche, H ; Weber, YG ; Berkovic, SF ; Scheffer, IE ; Reid, CA ; Petrou, S (LIPPINCOTT WILLIAMS & WILKINS, 2018-12)
    OBJECTIVE: To examine the genotype to phenotype connection in glucose transporter type 1 (GLUT1) deficiency and whether a simple functional assay can predict disease outcome from genetic sequence alone. METHODS: GLUT1 deficiency, due to mutations in SLC2A1, causes a wide range of epilepsies. One possible mechanism for this is variable impact of mutations on GLUT1 function. To test this, we measured glucose transport by GLUT1 variants identified in population controls and patients with mild to severe epilepsies. Controls were reference sequence from the NCBI and 4 population missense variants chosen from public reference control databases. Nine variants associated with epilepsies or movement disorders, with normal intellect in all individuals, formed the mild group. The severe group included 5 missense variants associated with classical GLUT1 encephalopathy. GLUT1 variants were expressed in Xenopus laevis oocytes, and glucose uptake was measured to determine kinetics (Vmax) and affinity (Km). RESULTS: Disease severity inversely correlated with rate of glucose transport between control (Vmax = 28 ± 5), mild (Vmax = 16 ± 3), and severe (Vmax = 3 ± 1) groups, respectively. Affinities of glucose binding in control (Km = 55 ± 18) and mild (Km = 43 ± 10) groups were not significantly different, whereas affinity was indeterminate in the severe group because of low transport rates. Simplified analysis of glucose transport at high concentration (100 mM) was equally effective at separating the groups. CONCLUSIONS: Disease severity can be partly explained by the extent of GLUT1 dysfunction. This simple Xenopus oocyte assay complements genetic and clinical assessments. In prenatal diagnosis, this simple oocyte glucose uptake assay could be useful because standard clinical assessments are not available.