45 results on '"John A. Damiano"'
Search Results
2. Recognition and epileptology of protracted <scp>CLN3</scp> disease
- Author
-
Jillian M. Cameron, John A. Damiano, Bronwyn Grinton, Patrick W. Carney, Penny McKelvie, Peter Silbert, Nicholas Lawn, Ingrid E. Scheffer, Karen L. Oliver, Michael S. Hildebrand, and Samuel F. Berkovic
- Subjects
Neurology ,Neurology (clinical) - Published
- 2023
- Full Text
- View/download PDF
3. Sporadic hypothalamic hamartoma is a ciliopathy with somatic and bi-allelic contributions
- Author
-
Timothy E Green, Joshua E Motelow, Mark F Bennett, Zimeng Ye, Caitlin A Bennett, Nicole G Griffin, John A Damiano, Richard J Leventer, Jeremy L Freeman, A Simon Harvey, Paul J Lockhart, Lynette G Sadleir, Amber Boys, Ingrid E Scheffer, Heather Major, Benjamin W Darbro, Melanie Bahlo, David B Goldstein, John F Kerrigan, Erin L Heinzen, Samuel F Berkovic, and Michael S Hildebrand
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Hamartoma ,Genetics ,Humans ,Original Article ,Hedgehog Proteins ,General Medicine ,Magnetic Resonance Imaging ,Molecular Biology ,Ciliopathies ,Hypothalamic Diseases ,Genetics (clinical) - Abstract
Hypothalamic hamartoma with gelastic seizures is a well-established cause of drug-resistant epilepsy in early life. The development of novel surgical techniques has permitted the genomic interrogation of hypothalamic hamartoma tissue. This has revealed causative mosaic variants within GLI3, OFD1 and other key regulators of the sonic-hedgehog pathway in a minority of cases. Sonic-hedgehog signalling proteins localize to the cellular organelle primary cilia. We therefore explored the hypothesis that cilia gene variants may underlie hitherto unsolved cases of sporadic hypothalamic hamartoma. We performed high-depth exome sequencing and chromosomal microarray on surgically resected hypothalamic hamartoma tissue and paired leukocyte-derived DNA from 27 patients. We searched for both germline and somatic variants under both dominant and bi-allelic genetic models. In hamartoma-derived DNA of seven patients we identified bi-allelic (one germline, one somatic) variants within one of four cilia genes—DYNC2I1, DYNC2H1, IFT140 or SMO. In eight patients, we identified single somatic variants in the previously established hypothalamic hamartoma disease genes GLI3 or OFD1. Overall, we established a plausible molecular cause for 15/27 (56%) patients. Here, we expand the genetic architecture beyond single variants within dominant disease genes that cause sporadic hypothalamic hamartoma to bi-allelic (one germline/one somatic) variants, implicate three novel cilia genes and reconceptualize the disorder as a ciliopathy.
- Published
- 2022
- Full Text
- View/download PDF
4. Genome-wide association study of febrile seizures implicates fever response and neuronal excitability genes
- Author
-
Line Skotte, João Fadista, Jonas Bybjerg-Grauholm, Vivek Appadurai, Michael S Hildebrand, Thomas F Hansen, Karina Banasik, Jakob Grove, Clara Albiñana, Frank Geller, Carmen F Bjurström, Bjarni J Vilhjálmsson, Matthew Coleman, John A Damiano, Rosemary Burgess, Ingrid E Scheffer, Ole Birger Vesterager Pedersen, Christian Erikstrup, David Westergaard, Kaspar René Nielsen, Erik Sørensen, Mie Topholm Bruun, Xueping Liu, Henrik Hjalgrim, Tune H Pers, Preben Bo Mortensen, Ole Mors, Merete Nordentoft, Julie W Dreier, Anders D Børglum, Jakob Christensen, David M Hougaard, Alfonso Buil, Anders Hviid, Mads Melbye, Henrik Ullum, Samuel F Berkovic, Thomas Werge, Bjarke Feenstra, and Institute for Molecular Medicine Finland
- Subjects
GAMMA-2-SUBUNIT ,HIPPOCAMPAL SCLEROSIS ,CHANNELS ,genome-wide association study ,Fever ,MUTATIONS ,3112 Neurosciences ,Anoctamins ,ABSENCE EPILEPSY ,PROTEIN ,VARIANTS ,Seizures, Febrile ,3124 Neurology and psychiatry ,NAV1.1 Voltage-Gated Sodium Channel ,Child, Preschool ,Humans ,epilepsy ,febrile seizures ,neuronal excitability genes ,Neurology (clinical) ,Child ,fever response genes - 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.
- Published
- 2022
- Full Text
- View/download PDF
5. Defective lipid signalling caused by mutations in PIK3C2B underlies focal epilepsy
- Author
-
Luca Gozzelino, Gaga Kochlamazashvili, Sara Baldassari, Albert Ian Mackintosh, Laura Licchetta, Emanuela Iovino, Yu Chi Liu, Caitlin A Bennett, Mark F Bennett, John A Damiano, Gábor Zsurka, Caterina Marconi, Tania Giangregorio, Pamela Magini, Marijn Kuijpers, Tanja Maritzen, Giuseppe Danilo Norata, Stéphanie Baulac, Laura Canafoglia, Marco Seri, Paolo Tinuper, Ingrid E Scheffer, Melanie Bahlo, Samuel F Berkovic, Michael S Hildebrand, Wolfram S Kunz, Lucio Giordano, Francesca Bisulli, Miriam Martini, Volker Haucke, Emilio Hirsch, Tommaso Pippucci, Gozzelino L., Kochlamazashvili G., Baldassari S., Mackintosh A.I., Licchetta L., Iovino E., Liu Y.-C., Bennett C.A., Bennett M.F., Damiano J.A., Zsurka G., Marconi C., Giangregorio T., Magini P., Kuijpers M., Maritzen T., Norata G.D., Baulac S., Canafoglia L., Seri M., Tinuper P., Scheffer I.E., Bahlo M., Berkovic S.F., Hildebrand M.S., Kunz W.S., Giordano L., Bisulli F., Martini M., Haucke V., Hirsch E., and Pippucci T.
- Subjects
PI3K-C2B ,class II PI3K ,epilepsy ,mTOR ,variants ,Animals ,Humans ,Lipids ,Mechanistic Target of Rapamycin Complex 1 ,Mice ,Mutation ,Phosphatidylinositol 3-Kinases ,Seizures ,Class II Phosphatidylinositol 3-Kinases ,Epilepsies, Partial ,Epilepsies ,Animal ,Lipid ,Seizure ,variant ,Class II Phosphatidylinositol 3-Kinase ,Settore BIO/14 - Farmacologia ,Neurology (clinical) ,Phosphatidylinositol 3-Kinase ,Human ,Partial - Abstract
Epilepsy is one of the most frequent neurological diseases, with focal epilepsy accounting for the largest number of cases. The genetic alterations involved in focal epilepsy are far from being fully elucidated.Here, we show that defective lipid signalling caused by heterozygous ultra-rare variants in PIK3C2B, encoding for the class II phosphatidylinositol 3-kinase PI3K-C2β, underlie focal epilepsy in humans. We demonstrate that patients’ variants act as loss-of-function alleles, leading to impaired synthesis of the rare signalling lipid phosphatidylinositol 3,4-bisphosphate, resulting in mTORC1 hyperactivation. In vivo, mutant Pik3c2b alleles caused dose-dependent neuronal hyperexcitability and increased seizure susceptibility, indicating haploinsufficiency as a key driver of disease. Moreover, acute mTORC1 inhibition in mutant mice prevented experimentally induced seizures, providing a potential therapeutic option for a selective group of patients with focal epilepsy.Our findings reveal an unexpected role for class II PI3K-mediated lipid signalling in regulating mTORC1-dependent neuronal excitability in mice and humans.
- Published
- 2022
6. SCN1A Variants in vaccine‐related febrile seizures: A prospective study
- Author
-
Jim Buttery, Michael Gold, Amy L Schneider, Nicholas Wood, Lucy Deng, John A. Damiano, Wenhui Li, Rosemary Burgess, Samuel F. Berkovic, Kristine Macartney, Peter Richmond, Michael S. Hildebrand, Ingrid E. Scheffer, and Nigel W Crawford
- Subjects
Male ,0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,Influenza vaccine ,Seizures, Febrile ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Dravet syndrome ,Febrile seizure ,medicine ,Humans ,Genetic Predisposition to Disease ,Prospective Studies ,Prospective cohort study ,Vaccines ,business.industry ,Case-control study ,Infant ,Odds ratio ,medicine.disease ,NAV1.1 Voltage-Gated Sodium Channel ,Vaccination ,030104 developmental biology ,Neurology ,Case-Control Studies ,Child, Preschool ,Mutation ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - 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.
- Published
- 2019
- Full Text
- View/download PDF
7. Epidemiology and etiology of infantile developmental and epileptic encephalopathies in Tasmania
- Author
-
Alexander Rosenfeld, D. L. Jones, Shannon R. Huskins, Katherine B. Howell, Michael Harvey, Simone Mandelstam, Jeremy L. Freeman, Ingrid E. Scheffer, Bronwyn E. Grinton, Samuel F. Berkovic, Melanie Bahlo, Jacinta M McMahon, Richard J. Leventer, Danae Andreopoulos-Malikotsinas, Yu Chi Liu, Michael S. Hildebrand, A. Simon Harvey, Mark F. Bennett, Mark T Mackay, John A. Damiano, and Tyson L Ware
- Subjects
Pediatrics ,medicine.medical_specialty ,Candidate gene ,Genetic counseling ,Population ,lcsh:RC346-429 ,whole exome sequencing ,03 medical and health sciences ,0302 clinical medicine ,Epidemiology ,medicine ,Short Research Article ,developmental and epileptic encephalopathy ,education ,lcsh:Neurology. Diseases of the nervous system ,Exome sequencing ,030304 developmental biology ,Genetic testing ,0303 health sciences ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Incidence (epidemiology) ,Neurology ,incidence ,Etiology ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
We sought to determine incidence, etiologies, and yield of genetic testing in infantile onset developmental and epileptic encephalopathies (DEEs) in a population isolate, with an intensive multistage approach. Infants born in Tasmania between 2011 and 2016, with seizure onset
- Published
- 2019
- Full Text
- View/download PDF
8. Genome-wide association study of febrile seizures identifies seven new loci implicating fever response and neuronal excitability genes
- Author
-
Julie Werenberg Dreier, Michael S. Hildebrand, Jakob Grove, Preben Bo Mortensen, Bjurström Cf, René Nielsen K, Ole Mors, Line Skotte, Bjarni J. Vilhjálmsson, Thomas Hansen, Matthew Coleman, Appadurai, Tune H. Pers, Samuel F. Berkovic, Mads Melbye, Vesterager Pedersen Ob, Bjarke Feenstra, Clara Albinana Climent, Jonas Bybjerg-Grauholm, Ingrid E. Scheffer, Frank Geller, Anders D. Børglum, Karina Banasik, Mie Topholm Bruun, Jakob Christensen, John A. Damiano, Erik Sørensen, David Westergaard, Alfonso Buil, Henrik Ullum, David M. Hougaard, Anders Hviid, Merete Nordentoft, Xueping Liu, Rosemary Burgess, Thomas Werge, Helle Hjalgrim, João Fadista, and Christian Erikstrup
- Subjects
0303 health sciences ,biology ,Genome-wide association study ,medicine.disease ,Synapse ,03 medical and health sciences ,Epilepsy ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,Febrile seizure ,Immunology ,biology.protein ,medicine ,Neurotransmitter ,Gene ,Pathological ,030217 neurology & neurosurgery ,030304 developmental biology ,GABRG2 - Abstract
Febrile seizures represent the most common type of pathological brain activity in young children and are influenced by genetic, environmental, and developmental factors. While usually benign, in a minority of cases, febrile seizures precede later development of epilepsy. Here, we conducted a genome-wide association study of febrile seizures with 7,635 cases and 93,966 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 harbored genes (BSN, ERC2, GABRG2, HERC1) influencing neuronal excitability by regulating neurotransmitter release and binding, vesicular transport or membrane trafficking at the synapse. GABRG2 is a well-established epilepsy gene comprising variants associated with febrile seizures, and overall we found positive genetic correlations with epilepsies (rg = 0.39, P = 1.68 × 10−4). Finally, a polygenic risk score based on all genome-wide significant loci was associated within patients with number of hospital admissions with febrile seizures and age at first admission, suggesting potential clinical utility of improved genetic understanding of febrile seizure genesis.
- Published
- 2020
- Full Text
- View/download PDF
9. Cerebrospinal fluid liquid biopsy for detecting somatic mosaicism in brain
- Author
-
Greta Gillies, AS Harvey, Amy L Schneider, Alexander Dobrovic, William Maixner, John A. Damiano, Martin Wood, Paul J. Lockhart, Hongdo Do, Samuel F. Berkovic, Wei Shern Lee, Richard J. Leventer, Michael S. Hildebrand, Zac Chatterton, Jahnvi Pflueger, Ingrid E. Scheffer, Ryan Lister, Lara McQuillan, Kate Pope, Melanie Bahlo, Michael Wilson, Stephen M. Malone, Zimeng Ye, and Bernadette Nolan
- Subjects
focal epilepsy ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Autopsy ,cerebrospinal fluid ,Ganglioglioma ,cell-free DNA ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Cerebrospinal fluid ,medicine ,Liquid biopsy ,Uncategorized ,liquid biopsy ,medicine.diagnostic_test ,business.industry ,AcademicSubjects/SCI01870 ,Brain biopsy ,General Engineering ,Cortical dysplasia ,medicine.disease ,030104 developmental biology ,Cell-free fetal DNA ,somatic mutations ,Original Article ,AcademicSubjects/MED00310 ,business ,Corrigendum ,030217 neurology & neurosurgery - Abstract
Brain somatic mutations are an increasingly recognized cause of epilepsy, brain malformations and autism spectrum disorders and may be a hidden cause of other neurodevelopmental and neurodegenerative disorders. At present, brain mosaicism can be detected only in the rare situations of autopsy or brain biopsy. Liquid biopsy using cell-free DNA derived from cerebrospinal fluid has detected somatic mutations in malignant brain tumours. Here, we asked if cerebrospinal fluid liquid biopsy can be used to detect somatic mosaicism in non-malignant brain diseases. First, we reliably quantified cerebrospinal fluid cell-free DNA in 28 patients with focal epilepsy and 28 controls using droplet digital PCR. Then, in three patients we identified somatic mutations in cerebrospinal fluid: in one patient with subcortical band heterotopia the LIS1 p. Lys64* variant at 9.4% frequency; in a second patient with focal cortical dysplasia the TSC1 p. Phe581His*6 variant at 7.8% frequency; and in a third patient with ganglioglioma the BRAF p. Val600Glu variant at 3.2% frequency. To determine if cerebrospinal fluid cell-free DNA was brain-derived, whole-genome bisulphite sequencing was performed and brain-specific DNA methylation patterns were found to be significantly enriched (P = 0.03). Our proof of principle study shows that cerebrospinal fluid liquid biopsy is valuable in investigating mosaic neurological disorders where brain tissue is unavailable., Somatic mutations can cause brain diseases. Currently, the only established route to study the brain-only somatic mutations is by sequencing brain tissues obtained from neurosurgery or autopsy. Ye et al. report a new strategy to use CSF cell-free DNA to detect brain-only somatic mutations as a surrogate for brain tissues., Graphical Abstract Graphical Abstract
- Published
- 2020
10. Progressive myoclonus epilepsies-Residual unsolved cases have marked genetic heterogeneity including dolichol-dependent protein glycosylation pathway genes
- Author
-
Leanne M. Dibbens, Anna-Elina Lehesjoki, Roberto Michelucci, Aarno Palotie, Barbara Castellotti, Jillian M. Cameron, Amos D. Korczyn, Loretta Giuliano, Davide Uccellini, Alessandro Filla, Edith Said, Karen Oliver, Zaid Afawi, William C. Sessa, Hui Bein Chew, Silvana Franceschetti, Carlo Di Bonaventura, Eva Andermann, Anna Teresa Giallonardo, Angelo Labate, Samuel F. Berkovic, Betül Baykan, Kariona A. Grabińska, Carolina Courage, Patrizia Riguzzi, Melanie Bahlo, Antonio Gambardella, John A. Damiano, Laura Canafoglia, Tarja Joensuu, Pasquale Striano, Christopher B. Jackson, Mikko Muona, Christian Brandt, Chiara Criscuolo, Sara Kivity, Eon Joo Park, Ingrid E. Scheffer, Géza Berecki, HUSLAB, Medicum, Department of Medical and Clinical Genetics, University of Helsinki, Clinicum, Research Programs Unit, STEMM - Stem Cells and Metabolism Research Program, Faculty of Medicine, Centre of Excellence in Complex Disease Genetics, Aarno Palotie / Principal Investigator, Institute for Molecular Medicine Finland, Genomics of Neurological and Neuropsychiatric Disorders, Helsinki Institute of Life Science HiLIFE, Courage, Carolina, Oliver, Karen L, Park, Eon Joo, Cameron, Jillian M, Dibbens, Leanne M, and Lehesjoki, Anna Elina
- Subjects
0301 basic medicine ,Male ,Glycosylation ,medicine.disease_cause ,Whole Exome Sequencing ,Cohort Studies ,0302 clinical medicine ,Dolichols ,whole-exome sequencing ,Age of Onset ,Child ,Genetics (clinical) ,Exome sequencing ,Genetics ,Mutation ,1184 Genetics, developmental biology, physiology ,dolichol-dependent glycosylation ,Middle Aged ,3. Good health ,Child, Preschool ,ASAH1 ,Female ,medicine.symptom ,Adult ,Adolescent ,DNA Copy Number Variations ,Progressive myoclonus epilepsy ,Biology ,progressive myoclonus epilepsy ,Article ,03 medical and health sciences ,Young Adult ,Progressive ,Myoclonic Epilepsies ,Exome Sequencing ,medicine ,Humans ,epilepsy genetics ,Preschool ,Gene ,Genetic heterogeneity ,Intron ,Introns ,Myoclonic Epilepsies, Progressive ,medicine.disease ,030104 developmental biology ,3111 Biomedicine ,Myoclonus ,030217 neurology & neurosurgery - Abstract
Progressive myoclonus epilepsies (PMEs) comprise a group of clinically and genetically heterogeneous rare diseases. Over 70% of PME cases can now be molecularly solved. Known PME genes encode a variety of proteins, many involved in lysosomal and endosomal function. We performed whole-exome sequencing (WES) in 84 (78 unrelated) unsolved PME-affected individuals, with or without additional family members, to discover novel causes. We identified likely disease-causing variants in 24 out of 78 (31%) unrelated individuals, despite previous genetic analyses. The diagnostic yield was significantly higher for individuals studied as trios or families (14/28) versus singletons (10/50) (OR = 3.9, p value = 0.01, Fisher's exact test). The 24 likely solved cases of PME involved 18 genes. First, we found and functionally validated five heterozygous variants in NUS1 and DHDDS and a homozygous variant in ALG10, with no previous disease associations. All three genes are involved in dolichol-dependent protein glycosylation, a pathway not previously implicated in PME. Second, we independently validate SEMA6B as a dominant PME gene in two unrelated individuals. Third, in five families, we identified variants in established PME genes; three with intronic or copy-number changes (CLN6, GBA, NEU1) and two very rare causes (ASAH1, CERS1). Fourth, we found a group of genes usually associated with developmental and epileptic encephalopathies, but here, remarkably, presenting as PME, with or without prior developmental delay. Our systematic analysis of these cases suggests that the small residuum of unsolved cases will most likely be a collection of very rare, genetically heterogeneous etiologies. Refereed/Peer-reviewed
- Published
- 2020
11. Evidence for a Dual-Pathway, 2-Hit Genetic Model for Focal Cortical Dysplasia and Epilepsy
- Author
-
Mark F. Bennett, Michael S. Hildebrand, Sayaka Kayumi, Mark A. Corbett, Sachin Gupta, Zimeng Ye, Michael Krivanek, Rosemary Burgess, Olivia J. Henry, John A. Damiano, Amber Boys, Jozef Gécz, Melanie Bahlo, Ingrid E. Scheffer, and Samuel F. Berkovic
- Subjects
Neurology (clinical) ,Article ,Genetics (clinical) - Abstract
Background and ObjectivesThe 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.MethodsWe 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.ResultsExome 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.DiscussionWe 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.
- Published
- 2022
- Full Text
- View/download PDF
12. Evidence of linkage to chromosome 5p13.2-q11.1 in a large inbred family with genetic generalized epilepsy
- Author
-
Samuel F. Berkovic, Canan Ulusoy, Melanie Bahlo, Erdem Tüzün, Demet Kinay, Michael S. Hildebrand, Karen Oliver, Eva Andermann, and John A. Damiano
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Adolescent ,Turkey ,Genetic Linkage ,Myoclonic Jerk ,Consanguinity ,Biology ,Polymorphism, Single Nucleotide ,Young Adult ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Genetic linkage ,Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels ,medicine ,Humans ,Child ,Exome sequencing ,Family Health ,Genetics ,Haplotype ,medicine.disease ,030104 developmental biology ,Neurology ,Chromosomes, Human, Pair 5 ,Medical genetics ,Epilepsy, Generalized ,Female ,Neurology (clinical) ,Juvenile myoclonic epilepsy ,030217 neurology & neurosurgery - Abstract
The clinical genetics of genetic generalized epilepsy suggests complex inheritance; large pedigrees, with multiple affected individuals, are rare exceptions. We studied a large consanguineous family from Turkey where extensive electroclinical phenotyping revealed a familial phenotype most closely resembling juvenile myoclonic epilepsy. For a subject to be considered affected (n = 14), a diagnostic electroencephalogram was required. Seizure onset ranged between 6 and 19 years (mean = 12 years). Thirteen of 14 experienced myoclonic jerks; in 11, this was associated with eyelid blinking, and in 10 it was interspersed with absences. Generalized tonic-clonic seizures were seen in 11. One individual had generalized tonic-clonic seizures alone. Electroencephalograms demonstrated generalized polyspike and wave discharges that were not associated with photoparoxysmal response. Intellect was normal. Nineteen family members were subsequently chosen for nonparametric multipoint linkage analyses, which identified a 39.5 Mb region on chromosome 5 (P < 0.0001). Iterative analysis, including discovery of a subtly affected individual, narrowed the critical region to 15.4 Mb and possibly to 5.5 Mb. Homozygous versus heterozygous state of the refined 5p13.2-q11.1 haplotype was not associated with phenotypic severity or onset age, suggesting that one versus two pathogenic variants may result in similar phenotypes. Whole exome sequencing (n = 3) failed to detect any rare, protein-coding variants within the highly significant linkage region that includes HCN1 as a promising candidate.
- Published
- 2018
- Full Text
- View/download PDF
13. Sensitive quantitative detection of somatic mosaic mutation in 'double cortex' syndrome
- Author
-
Rosemary Burgess, John A. Damiano, Samuel F. Berkovic, Michael S. Hildebrand, Alexander Dobrovic, Renate M Kalnins, Ezgi Ozturk, Hongdo Do, and Nigel C. Jones
- Subjects
Adult ,0301 basic medicine ,Drug Resistant Epilepsy ,Somatic cell ,Classical Lissencephalies and Subcortical Band Heterotopias ,Biology ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Intellectual Disability ,Tissue mosaicism ,Humans ,Language Development Disorders ,Digital polymerase chain reaction ,Mutation frequency ,Genetics ,Mutagenesis ,General Medicine ,Phenotype ,030104 developmental biology ,Neurology ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Mutation ,Mutation (genetic algorithm) ,Female ,Epilepsies, Partial ,Neurology (clinical) ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery - 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.
- Published
- 2017
- Full Text
- View/download PDF
14. Genetic epilepsy with febrile seizures plus
- Author
-
Samuel F. Berkovic, Andrew Bleasel, Hadassa Goldberg-Stern, Bronwyn E. Grinton, Sara Kivity, Leanne M. Dibbens, Elizabeth K. Ruzzo, John A. Damiano, Lata Vadlamudi, Zaid Afawi, Georgie C. Glubb, Jodie P. Malone, Rosemary Burgess, Padraic Grattan-Smith, Yue-Hua Zhang, Danya F. Vears, Katherine L. Helbig, Amos D. Korczyn, Ingrid E. Scheffer, Susannah T. Bellows, and Michael S. Hildebrand
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Seizures, Febrile ,Genetic epilepsy ,Young Adult ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Childhood absence epilepsy ,medicine ,Humans ,Age of Onset ,Focal Epilepsies ,Generalized epilepsy ,Child ,business.industry ,Infant ,Genetic data ,Middle Aged ,medicine.disease ,Pedigree ,Phenotype ,030104 developmental biology ,Child, Preschool ,Immunology ,Epilepsy, Generalized ,Female ,Epilepsies, Partial ,Neurology (clinical) ,Age of onset ,business ,Generalized epilepsy with febrile seizures plus ,030217 neurology & neurosurgery - 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.
- Published
- 2017
- Full Text
- View/download PDF
15. Evaluation of GLUT1 variation in non-acquired focal epilepsy
- Author
-
IE Scheffer, Alexander Peeraer, Michael S. Hildebrand, Samuel F. Berkovic, Saul A. Mullen, Susannah T. Bellows, and John A. Damiano
- Subjects
Adult ,Male ,0301 basic medicine ,Microcephaly ,Movement disorders ,Adolescent ,DNA Mutational Analysis ,Biology ,medicine.disease_cause ,Severity of Illness Index ,Temporal lobe ,Cohort Studies ,Young Adult ,03 medical and health sciences ,symbols.namesake ,Epilepsy ,0302 clinical medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Generalized epilepsy ,Aged ,Aged, 80 and over ,Sanger sequencing ,Genetics ,Glucose Transporter Type 1 ,Mutation ,Electroencephalography ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,030104 developmental biology ,Neurology ,Cohort ,symbols ,Female ,Epilepsies, Partial ,Neurology (clinical) ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Brain glucose transport is dependent on glucose transporter 1 (GLUT1), encoded by the solute carrier family 2 member 1 (SLC2A1) gene. Mutations in SLC2A1 cause GLUT1 deficiency which is characterized by a broad spectrum of neurological phenotypes including generalized epilepsy, motor disorders, developmental delay and microcephaly. Recent case reports suggest SLC2A1 mutations can contribute to non-acquired focal epilepsy (NAFE) but interrogation of a large patient cohort has not been reported. We studied 200 patients with NAFE (126 with temporal lobe epilepsy) comprising 104 females and 96 males with a mean age of onset of 18 years. Polymerase chain reaction (PCR) and Sanger sequencing was performed to detect variants in all 10 coding exons and splice site regions of the SLC2A1 gene. We did not detect any pathogenic mutations in SLC2A1 in this cohort. Our data suggests that the frequency of GLUT1 mutations in NAFE is low. Limitations of this study include the mean age of onset and cohort size. Future research should focus on subpopulations of focal epilepsy with lower age of seizure onset particularly with co-existent movement disorders in which GLUT1 mutations may play a more important role.
- Published
- 2017
- Full Text
- View/download PDF
16. Frequency ofCNKSR2mutation in the X-linked epilepsy-aphasia spectrum
- Author
-
Rosemary Burgess, John A. Damiano, Zaid Afawi, Ingrid E. Scheffer, Tally Lerman-Sagie, Samuel F. Berkovic, Michael S. Hildebrand, and Sara Kivity
- Subjects
Male ,0301 basic medicine ,Proband ,Genetic counseling ,DNA Mutational Analysis ,Nonsense mutation ,Biology ,Cohort Studies ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Aphasia ,medicine ,Humans ,Adaptor Proteins, Signal Transducing ,Family Health ,Genetics ,Point mutation ,Infant ,Electroencephalography ,medicine.disease ,Phenotype ,030104 developmental biology ,Neurology ,Mutation ,Mutation (genetic algorithm) ,Speech delay ,Female ,CNKSR2 ,Neurology (clinical) ,medicine.symptom ,Sleep ,Spasms, Infantile ,030217 neurology & neurosurgery - 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.
- Published
- 2017
- Full Text
- View/download PDF
17. Pathogenic Variants in CEP85L Cause Sporadic and Familial Posterior Predominant Lissencephaly
- Author
-
Michele G. Mehaffey, Mei-Feng Wu, Edith P. Almanza Fuerte, Laura A. Jansen, Ingrid E. Scheffer, Wen-Hung Chung, Michael S. Hildebrand, Meng-Han Tsai, Paul J. Lockhart, Richard J. Leventer, Wen-Lang Fan, Heather C Mefford, Ying-Chao Chang, Chung-Kin Chan, Nian-Hsin Chao, Alison M. Muir, William B. Dobyns, Shih-Chi Su, Kheng Seang Lim, Guillaume Sébire, Nicolas Deconinck, Won Jing Wang, Ching Ching Ng, Kate Riney, Kun-Chuan Yang, Samuel F. Berkovic, Jin Wu Tsai, John A. Damiano, Yi-Ning Kang, Brenda E. Porter, and Ghayda M. Mirzaa
- Subjects
0301 basic medicine ,Adult ,Male ,Heterozygote ,Adolescent ,Oncogene Proteins, Fusion ,Lissencephaly ,Classical Lissencephalies and Subcortical Band Heterotopias ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,PAFAH1B1 ,Mice ,Young Adult ,0302 clinical medicine ,Seizures ,medicine ,Animals ,Humans ,Age of Onset ,Child ,Gene ,Pericentriolar material ,Genetics ,Centrosome ,Chromosome Aberrations ,Mutation ,General Neuroscience ,Pachygyria ,Genetic Variation ,Infant ,Heterozygote advantage ,medicine.disease ,Magnetic Resonance Imaging ,Pedigree ,Cytoskeletal Proteins ,030104 developmental biology ,Child, Preschool ,Gene Knockdown Techniques ,Female ,030217 neurology & neurosurgery - Abstract
Lissencephaly (LIS), denoting a "smooth brain," is characterized by the absence of normal cerebral convolutions with abnormalities of cortical thickness. Pathogenic variants in over 20 genes are associated with LIS. The majority of posterior predominant LIS is caused by pathogenic variants in LIS1 (also known as PAFAH1B1), although a significant fraction remains without a known genetic etiology. We now implicate CEP85L as an important cause of posterior predominant LIS, identifying 13 individuals with rare, heterozygous CEP85L variants, including 2 families with autosomal dominant inheritance. We show that CEP85L is a centrosome protein localizing to the pericentriolar material, and knockdown of Cep85l causes a neuronal migration defect in mice. LIS1 also localizes to the centrosome, suggesting that this organelle is key to the mechanism of posterior predominant LIS.
- Published
- 2019
18. Kufs disease due to mutation of CLN6: Clinical, pathological and molecular genetic features
- Author
-
Cigdem Ozkara, Angelo Labate, Sara E. Mole, Alan McDougall, Antonio Gambardella, Michael S. Hildebrand, Sulekha Rajagopalan, Vincenzo Belcastro, Danya F. Vears, Hans Henrik M. Dahl, Loretta Giuliano, Karen Oliver, Michael Farrell, Vito Sofia, Barbara Garavaglia, Samuel F. Berkovic, Frederick Andermann, Julia Rankin, Silvana Franceschetti, Stirling Carpenter, Michela Morbin, Alessandro Simonati, Eva Andermann, Penina Krieger, Umberto Aguglia, John A. Damiano, Adam Zeman, Barbara Castellotti, Susan Brammah, Filippo M. Santorelli, and Laura Canafoglia
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Ataxia ,Adolescent ,Progressive myoclonus epilepsy ,Compound heterozygosity ,03 medical and health sciences ,Epilepsy ,Young Adult ,0302 clinical medicine ,Neuronal Ceroid-Lipofuscinoses ,Kufs disease ,medicine ,Dementia ,Humans ,Age of Onset ,Aged ,business.industry ,ataxia ,neurodegeneration ,Brain ,Membrane Proteins ,CLN6 ,Middle Aged ,medicine.disease ,Survival Rate ,030104 developmental biology ,Mutation ,neuronal ceroid lipofuscinosis ,Female ,Neuronal ceroid lipofuscinosis ,Neurology (clinical) ,Age of onset ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Kufs disease is the major adult form of neuronal ceroid lipofuscinosis, but is rare and difficult to diagnose. Diagnosis was traditionally dependent on the demonstration of characteristic storage material, but distinction from normal age-related accumulation of lipofuscin can be challenging. Mutation of CLN6 has emerged as the most important cause of recessive Kufs disease but, remarkably, is also responsible for variant late infantile ceroid lipofuscinosis. Here we provide a detailed description of Kufs disease due to CLN6 pathogenic variants. We studied 20 cases of Kufs disease with CLN6 pathogenic variants from 13 unrelated families. Mean age of onset was 28 years (range 12-51) with bimodal peaks in teenage and early adult life. The typical presentation was of progressive myoclonus epilepsy with debilitating myoclonic seizures and relatively infrequent tonic-clonic seizures. Patients became wheelchair-bound with a mean 12 years post-onset. Ataxia was the most prominent motor feature. Dementia appeared to be an invariable accompaniment, although it could take a number of years to manifest and occasionally cognitive impairment preceded myoclonic seizures. Patients were usually highly photosensitive on EEG. MRI showed progressive cerebral and cerebellar atrophy. The median survival time was 26 years from disease onset. Ultrastructural examination of the pathology revealed fingerprint profiles as the characteristic inclusions, but they were not reliably seen in tissues other than brain. Curvilinear profiles, which are seen in the late infantile form, were not a feature. Of the 13 unrelated families we observed homozygous CLN6 pathogenic variants in four and compound heterozygous variants in nine. Compared to the variant late infantile form, there was a lower proportion of variants that predicted protein truncation. Certain heterozygous missense variants in the same amino acid position were found in both variant late infantile and Kufs disease. There was a predominance of cases from Italy and surrounding regions; this was partially explained by the discovery of three founder pathogenic variants. Clinical distinction of type A (progressive myoclonus epilepsy) and type B (dementia with motor disturbance) Kufs disease was supported by molecular diagnoses. Type A is usually caused by recessive pathogenic variants in CLN6 or dominant variants in DNAJC5. Type B Kufs is usually associated with recessive CTSF pathogenic variants. The diagnosis of Kufs remains challenging but, with the availability of genetic diagnosis, this will largely supersede the use of diagnostic biopsies, particularly as biopsies of peripheral tissues has unsatisfactory sensitivity and specificity.
- Published
- 2019
- Full Text
- View/download PDF
19. Contribution of rare genetic variants to drug response in absence epilepsy
- Author
-
Amy L Schneider, Jeffrey Buchhalter, Sabrina D'Alfonso, Lynette G. Sadleir, John A. Damiano, Bronwyn E. Grinton, Samuel F. Berkovic, Melanie Bahlo, Eunice K Chan, Kenneth A. Myers, Luis Bello-Espinosa, Mark F. Bennett, Ingrid E. Scheffer, Gabriel Dabscheck, and Michael S. Hildebrand
- Subjects
0301 basic medicine ,Bioinformatics ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,medicine ,CACNA1H ,Humans ,gamma-Aminobutyric Acid ,Exome sequencing ,Valproic Acid ,biology ,business.industry ,Odds ratio ,medicine.disease ,Pathophysiology ,030104 developmental biology ,Ethosuximide ,Epilepsy, Absence ,Pharmaceutical Preparations ,Neurology ,Pharmacogenomics ,biology.protein ,Anticonvulsants ,lipids (amino acids, peptides, and proteins) ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Objective We investigated the possible significance of rare genetic variants to response to valproic acid (VPA) and ethosuximide (ETX) in patients with absence epilepsy. Our primary hypothesis was that rare CACNA1H variants are more frequent in ETX-non-responsive patients compared to ETX-responsive. Our secondary hypothesis was that rare variants in GABA-receptor genes are more frequent in VPA-non-responsive patients compared to VPA-responsive. Methods We recruited patients with absence epilepsy treated with both VPA and ETX, and performed whole exome sequencing in order to investigate the potential role of rare variants in CACNA1H, other voltage-gated calcium channel (VGCC) genes, or GABA-receptor genes in predicting response to ETX or VPA. Results Sixty-two patients were included; 12 were ETX-responsive, 14 VPA-responsive, and 36 did not have a clear positive response to either medication. We did not find significant enrichment inCACNA1H rare variants in ETX-responsive patients (odds ratio 3.43; 0.43–27.65; p = 0.20), nor was there enrichment for other VGCC genes. No significant enrichment of GABA-receptor gene rare variants was seen for VPA-non-responsive patients versus VPA-responsive. We found enrichment of rare GABA-receptor variants in our absence cohort compared to controls (odds ratio 3.82; 1.68−8.69). There was no difference in frequency of CACNA1H rs61734410 and CACNA1I rs3747178 polymorphisms between ETX-responsive and ETX-non-responsive groups; these polymorphisms have previously been reported to predict lack of response to ETX in absence epilepsy. Significance We conclude that if CACNA1H rare variants predict lack of response to ETX, a larger sample is necessary to test this with sufficient power. Increased GABA-receptor gene rare variant frequency in absence epilepsy patients who fail initial anti-seizure therapy suggests subtle GABA receptor dysfunction may contribute to the underlying pathophysiology.
- Published
- 2021
- Full Text
- View/download PDF
20. Mutations of the Sonic Hedgehog Pathway Underlie Hypothalamic Hamartoma with Gelastic Epilepsy
- Author
-
Nigel C. Jones, David Goldstein, Michael S. Hildebrand, John A. Damiano, John F. Kerrigan, Ezgi Ozturk, Benjamin W. Darbro, Erin L. Heinzen, Lynette G. Sadleir, Nicole G. Griffin, Heather J. Major, Rosemary Burgess, Jeremy L. Freeman, Richard J. Leventer, Andrew S. Allen, A. Simon Harvey, Ingrid E. Scheffer, Samuel F. Berkovic, and Elisa J. Cops
- Subjects
Male ,0301 basic medicine ,Candidate gene ,animal structures ,Hamartoma ,Kruppel-Like Transcription Factors ,Loss of Heterozygosity ,Nerve Tissue Proteins ,Zinc Finger Protein Gli2 ,03 medical and health sciences ,0302 clinical medicine ,Hypothalamic hamartoma ,Zinc Finger Protein Gli3 ,Report ,GLI2 ,GLI3 ,Genetics ,Humans ,Exome ,Hedgehog Proteins ,Genetics(clinical) ,Sonic hedgehog ,Genetics (clinical) ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,biology ,Wnt signaling pathway ,Nuclear Proteins ,CREB-Binding Protein ,Hedgehog signaling pathway ,3. Good health ,PRKACA ,030104 developmental biology ,Mutation ,embryonic structures ,Cancer research ,biology.protein ,Female ,Epilepsies, Partial ,Hypothalamic Diseases ,030217 neurology & neurosurgery ,Signal Transduction - 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.
- Published
- 2016
- Full Text
- View/download PDF
21. Somatic
- Author
-
Michael S, Hildebrand, A Simon, Harvey, Stephen, Malone, John A, Damiano, Hongdo, Do, Zimeng, Ye, Lara, McQuillan, Wirginia, Maixner, Renate, Kalnins, Bernadette, Nolan, Martin, Wood, Ezgi, Ozturk, Nigel C, Jones, Greta, Gillies, Kate, Pope, Paul J, Lockhart, Alexander, Dobrovic, Richard J, Leventer, Ingrid E, Scheffer, and Samuel F, Berkovic
- Subjects
Article - Abstract
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.
- Published
- 2018
22. Evaluation of multiple putative risk alleles within the 15q13.3 region for genetic generalized epilepsy
- Author
-
Benjamin W. Darbro, Michael S. Hildebrand, Leanne M. Dibbens, Heather C Mefford, Susannah T. Bellows, Samuel F. Berkovic, Saul A. Mullen, Ingrid E. Scheffer, Todor Arsov, Kate M. Lawrence, John A. Damiano, Heather J. Major, Hans Henrik M. Dahl, Damiano, John A, Mullen, Saul A, Hildebrand, Michael S, Bellows, Susannah T, Lawrence, Kate M, Arsov, Todor, Dibbens, Leanne, Major, Heather, Dahl, Hans-Henrik M, Mefford, Heather C, Darbro, Benjamin W, Scheffer, Ingrid E, and Berkovic, Samuel F
- Subjects
Male ,Proband ,medicine.medical_specialty ,DNA Copy Number Variations ,alpha7 Nicotinic Acetylcholine Receptor ,Epilepsy ,symbols.namesake ,Gene Frequency ,Molecular genetics ,Genetics ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Copy-number variation ,Allele ,Alleles ,Sanger sequencing ,Chromosomes, Human, Pair 15 ,Polymorphism, Genetic ,biology ,CHRNA7 ,Complex traits ,medicine.disease ,Pedigree ,Epilepsy and seizures ,Neurology ,Genetic Loci ,biology.protein ,symbols ,Epilepsy, Generalized ,Female ,Neurology (clinical) - Abstract
The chromosome 15q13.3 region has been implicated in epilepsy, intellectual disability and neuropsychiatric disorders, especially schizophrenia. Deficiency of the acetylcholine receptor gene CHRNA7 and the partial duplication, CHRFAM7A, may contribute to these phenotypes and we sought to comprehensively analyze these genes in genetic generalized epilepsy. We analyzed using DHPLC, Sanger sequencing and long range PCR, 174 probands with genetic generalized epilepsy with or without intellectual disability or psychosis, including 8 with the recurrent 15q13.3 microdeletion. We searched CHRNA7 and CHRFAM7A for single sequence variants, small copy number variants, and the common 2-bp deletion in CHRFAM7A. We identified two novel and one reported missense variants. The common 2-bp deletion was not enriched in patients compared to controls. Our data suggest that missense mutations in CHRNA7 contribute to complex inheritance in genetic generalized epilepsy in a similar fashion to the 15q13.3 microdeletion. They do not support a pathogenic role for the common 2-bp CHRFAM7A deletion. Refereed/Peer-reviewed
- Published
- 2015
- Full Text
- View/download PDF
23. Atypical multifocal Dravet syndrome lacks generalized seizures and may show later cognitive decline
- Author
-
Deepak Gill, Kent Kelley, Ingrid E. Scheffer, Xenia Iona, Susannah T. Bellows, J. Helen Cross, Leanne M. Dibbens, Young Ok Kim, Jacinta M McMahon, Samuel F. Berkovic, John A. Damiano, Kim, Young Ok, Bellows, Susannah, McMahon, Jacinta M, Iona, Xenia, Damiano, John, Dibbens, Leanne, Kelley, Kent, Gill, Deepak, Cross, J Helen, Berkovic, Samuel F, and Scheffer, Ingrid E
- Subjects
Adult ,Pediatrics ,medicine.medical_specialty ,Adolescent ,DNA Copy Number Variations ,Molecular Sequence Data ,Clinical Neurology ,Epilepsies, Myoclonic ,Status epilepticus ,Electroencephalography ,Child Development ,Developmental Neuroscience ,Dravet syndrome ,Seizures ,Intellectual Disability ,Convulsion ,Intellectual disability ,medicine ,Humans ,Age of Onset ,Cognitive decline ,Child ,medicine.diagnostic_test ,Seizure types ,business.industry ,medicine.disease ,NAV1.1 Voltage-Gated Sodium Channel ,Anesthesia ,Mutation ,Pediatrics, Perinatology and Child Health ,Female ,Neurology (clinical) ,medicine.symptom ,Age of onset ,Cognition Disorders ,business - Abstract
Aim To show that atypical multifocal Dravet syndrome is a recognizable, electroclinical syndrome associated with sodium channel gene (SCN1A) mutations that readily escapes diagnosis owing to later cognitive decline and tonic seizures. Method Eight patients underwent electroclinical characterization. SCN1A was sequenced and copy number variations sought by multiplex ligation-dependent probe amplification. Results All patients were female (age range at assessment 5–26y) with median seizure onset at 6.5 months (range 4–19mo). The initial seizure was brief in seven and status epilepticus only occurred in one; three were febrile. Focal seizures occurred in four patients and bilateral convulsion in the other four. All patients developed multiple focal seizure types and bilateral convulsions, with seizure clusters in six. The most common focal seizure semiology (six out of eight) comprised unilateral clonic activity. Five also had focal or asymmetric tonic seizures. Rare or transient myoclonic seizures occurred in six individuals, often triggered by specific antiepileptic drugs. Developmental slowing occurred in all: six between 3 years and 8 years, and two around 1 year 6 months. Cognitive outcome varied from severe to mild intellectual disability. Multifocal epileptiform discharges were seen on electroencephalography. Seven out of eight patients had SCN1A mutations. Interpretation Atypical, multifocal Dravet syndrome with SCN1A mutations may not be recognized because of later cognitive decline and frequent tonic seizures.
- Published
- 2013
- Full Text
- View/download PDF
24. Mutations inTNK2in severe autosomal recessive infantile onset epilepsy
- Author
-
Yuki Hitomi, Jacinta M McMahon, Samuel F. Berkovic, Myriam Rai, Hans-Henrik M. Dahl, Arvid Suls, Benjamin Legros, Peter De Jonghe, Ingrid E. Scheffer, Chantal Depondt, Patrick Van Bogaert, Simona Donatello, David Goldstein, Massimo Pandolfo, Sarah Weckhuysen, John A. Damiano, and Erin L. Heinzen
- Subjects
Genetics ,Mutation ,Biology ,medicine.disease_cause ,Compound heterozygosity ,medicine.disease ,Phenotype ,Epilepsy ,Neurology ,Genotype ,medicine ,Missense mutation ,Neurology (clinical) ,Gene ,Exome sequencing - Abstract
We identified a small family with autosomal recessive, infantile onset epilepsy and intellectual disability. Exome sequencing identified a homozygous missense variant in the gene TNK2, encoding a brain-expressed tyrosine kinase. Sequencing of the coding region of TNK2 in 110 patients with a similar phenotype failed to detect further homozygote or compound heterozygote mutations. Pathogenicity of the variant is supported by the results of our functional studies, which demonstrated that the variant abolishes NEDD4 binding to TNK2, preventing its degradation after epidermal growth factor stimulation. Definitive proof of pathogenicity will require confirmation in unrelated patients.
- Published
- 2013
- Full Text
- View/download PDF
25. GRIN2A mutations cause epilepsy-aphasia spectrum disorders
- Author
-
John A. Damiano, Eileen Geraghty, Gemma L. Carvill, Heather C Mefford, Samantha J. Turner, Adiba Khan, Robert A. Ouvrier, Ingrid E. Scheffer, Meng-Han Tsai, Jay Shendure, Samuel F. Berkovic, Brian J. O'Roak, Natalia Lozovaya, Nadine Bruneau, Michael S. Hildebrand, Brigid M. Regan, Richard Webster, Simone C. Yendle, Nail Burnashev, Pierre Szepetowski, Lynette G. Sadleir, and Joseph Cook
- Subjects
Male ,Proband ,Landau–Kleffner syndrome ,Biology ,Bioinformatics ,Receptors, N-Methyl-D-Aspartate ,Article ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Aphasia ,Genetics ,medicine ,Humans ,030304 developmental biology ,Landau-Kleffner Syndrome ,0303 health sciences ,Electroencephalography ,medicine.disease ,Pedigree ,3. Good health ,Rolandic epilepsy ,Phenotype ,Mutation ,Epilepsy syndromes ,Etiology ,Female ,medicine.symptom ,Developmental regression ,030217 neurology & neurosurgery - Abstract
Epilepsy-aphasia syndromes (EAS) are a group of rare, severe epileptic encephalopathies of unknown etiology with a characteristic electroencephalogram (EEG) pattern and developmental regression particularly affecting language. Rare pathogenic deletions that include GRIN2A have been implicated in neurodevelopmental disorders. We sought to delineate the pathogenic role of GRIN2A in 519 probands with epileptic encephalopathies with diverse epilepsy syndromes. We identified four probands with GRIN2A variants that segregated with the disorder in their families. Notably, all four families presented with EAS, accounting for 9% of epilepsy-aphasia cases. We did not detect pathogenic variants in GRIN2A in other epileptic encephalopathies (n = 475) nor in probands with benign childhood epilepsy with centrotemporal spikes (n = 81). We report the first monogenic cause, to our knowledge, for EAS. GRIN2A mutations are restricted to this group of cases, which has important ramifications for diagnostic testing and treatment and provides new insights into the pathogenesis of this debilitating group of conditions.
- Published
- 2013
26. SCN1A clinical spectrum includes the self-limited focal epilepsies of childhood
- Author
-
Karen Oliver, John A. Damiano, Samuel F. Berkovic, Sara Kivity, Zaid Afawi, Todor Arsov, and Melanie Bahlo
- Subjects
0301 basic medicine ,Male ,Adolescent ,Mutation, Missense ,Consanguinity ,03 medical and health sciences ,Epilepsy ,Young Adult ,0302 clinical medicine ,Dravet syndrome ,Genetic variation ,medicine ,Missense mutation ,Humans ,Generalized epilepsy ,Child ,Genetics ,Genetic Variation ,Infant ,Panayiotopoulos syndrome ,medicine.disease ,Penetrance ,Pedigree ,NAV1.1 Voltage-Gated Sodium Channel ,030104 developmental biology ,Neurology ,Child, Preschool ,Female ,Neurology (clinical) ,Epilepsies, Partial ,Psychology ,030217 neurology & neurosurgery - Abstract
Introduction Amongst autosomal dominant genetic epilepsy with febrile seizures plus (GEFS+) families, SCN1A variants are the most common genetic cause. Initially regarded as a generalized form of epilepsy, the GEFS+ spectrum is now known to include some focal epilepsies, but it is generally not conceptualized as extending to the self-limited focal epilepsies of childhood, such as Panayiotopoulos syndrome. There are, however, three reports of SCN1A variants in Panayiotopoulos syndrome. We describe the variable clinical phenotypes that include the self-limited focal epilepsies of childhood, present in a large GEFS+ family, segregating a heterozygous SCN1A missense variant. Material and methods Electro-clinical details on all putatively affected family members were sought and blood samples were taken for genetic analysis. Two individuals were chosen for SCN1A testing. All 26 exons and exon–intron junctions were amplified, sequenced and analyzed. This was followed by pedigree segregation analysis of the variant identified. Results A pathogenic heterozygous SCN1A (c.2624C>A; p.Thr875Lys) variant was identified. Sixteen of the 18 variant positive family members were affected (88% penetrance): 8 with febrile seizures, 2 febrile seizures plus, 1 unclassified seizures and 5 with self-limited focal epilepsy of childhood. Of these, one was diagnosed with atypical childhood epilepsy with centrotemporal spikes and four with Panayiotopoulos syndrome. Discussion By characterizing the heterogeneous clinical phenotypes in a large, SCN1A mutation positive GEFS+ family, we conclude that the GEFS+ spectrum can extend to the self-limited focal epilepsies of childhood, including Panayiotopoulos syndrome, and in turn highlight the complex genotype-phenotype correlations associated with SCN1A -related epilepsies.
- Published
- 2016
27. Real-world utility of whole exome sequencing with targeted gene analysis for focal epilepsy
- Author
-
Natalie P. Thorne, Sebastian Lunke, Michael S. Hildebrand, John A. Damiano, Clara Gaff, Piero Perucca, Ingrid E. Scheffer, A. Simon Harvey, Patrick Kwan, Terence J. O'Brien, Paul A. James, Brigid M. Regan, and Samuel F. Berkovic
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Pediatrics ,Adolescent ,03 medical and health sciences ,Epilepsy ,Young Adult ,0302 clinical medicine ,Intellectual disability ,Exome Sequencing ,medicine ,Humans ,Epilepsy surgery ,Genetic Testing ,Prospective Studies ,Family history ,Child ,Exome sequencing ,Genetic testing ,Aged ,medicine.diagnostic_test ,business.industry ,Genetic Variation ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,DEPDC5 ,Surgery ,Pedigree ,030104 developmental biology ,Clinical research ,Neurology ,Child, Preschool ,Female ,Neurology (clinical) ,Epilepsies, Partial ,business ,030217 neurology & neurosurgery - Abstract
Objective Driven by advances in genomic technology and reduction in costs, next-generation sequencing (NGS) is venturing into routine clinical care. The ‘real-world' clinical utility of NGS remains to be determined in focal epilepsies, which account for 60% of all epilepsies and for which the importance of genetic factors is just beginning to emerge. We investigated the diagnostic yield and management implications of whole exome sequencing (WES)-based screening of selected genes in the routine care of common focal epilepsies suspected to have a genetic basis. Methods We performed WES, followed by targeted analysis of 64 epilepsy genes, on 40 consecutive children and adults enrolled prospectively from routine clinical practice who had MRI-negative focal epilepsy and a family history of febrile seizures or any type of epilepsy in at least one first- or second-degree relative. Exclusion criteria were previous genetic testing, severe intellectual disability and benign focal epilepsies of childhood. Results 5/40 (12.5%) patients had a pathogenic or likely pathogenic variant, detected in SCN1A , DEPDC5 , PCDH19, GABRG2 or NPRL2 . Identifying a pathogenic SCN1A variant in a patient with drug-resistant epilepsy prompted to halt presurgical investigations due to concern of unfavorable post-surgical outcome. It also led in the same patient to discontinue long-standing carbamazepine therapy (a potentially aggravating drug in epilepsies due to SCN1A mutations), resulting in complete seizure control. Patients with pathogenic or likely pathogenic variants had a younger median age of seizure onset (range) compared to those without [18 months (8 months-18 years) vs 18 years (18 months-70 years), p=0.02]. Significance Our data demonstrate that WES with targeted gene analysis is an effective diagnostic tool for patients with common focal epilepsies in whom a genetic etiology is suspected. It can also influence clinical decision-making, including antiepileptic drug selection and consideration of epilepsy surgery, hence supporting its incorporation in the routine clinical care of this patient group.
- Published
- 2016
28. Evaluation of non-coding variation in GLUT1 deficiency
- Author
-
Michael S. Hildebrand, Samuel F. Berkovic, Melanie Bahlo, John A. Damiano, Jia Wei Audrey Lee, Susannah T. Bellows, Ingrid E. Scheffer, Saul A. Mullen, and Yu Chi Liu
- Subjects
0301 basic medicine ,Proband ,Adult ,Male ,CSF glucose ,Monosaccharide Transport Proteins ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,Humans ,Exome ,Copy-number variation ,Exome sequencing ,Genetics ,Glucose Transporter Type 1 ,Splice site mutation ,Epilepsy ,biology ,Infant ,Pedigree ,030104 developmental biology ,Glucose ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Hypoglycorrhachia ,biology.protein ,GLUT1 ,Female ,Neurology (clinical) ,Sequence Analysis ,030217 neurology & neurosurgery ,Carbohydrate Metabolism, Inborn Errors - Abstract
Aim Loss-of-function mutations in SLC2A1, encoding glucose transporter-1 (GLUT-1), lead to dysfunction of glucose transport across the blood–brain barrier. Ten percent of cases with hypoglycorrhachia (fasting cerebrospinal fluid [CSF] glucose
- Published
- 2016
29. Glucose transporter 1 deficiency in the idiopathic generalized epilepsies
- Author
-
Steven Petrou, A. Marie Phillips, Todor Arsov, Sara Kivity, Ingrid E. Scheffer, Chantal Trager, Zaid Afawi, Samuel F. Berkovic, John A. Damiano, Sue Rogers, Hadassa Goldberg-Stern, Saul A. Mullen, and Kate M. Lawrence
- Subjects
Adult ,Male ,Proband ,Genotype ,Monosaccharide Transport Proteins ,medicine.medical_treatment ,DNA Mutational Analysis ,Biology ,Evolution, Molecular ,Young Adult ,medicine ,Animals ,Humans ,Allele ,Aged ,Genetics ,Glucose Transporter Type 1 ,Glucose transporter ,Middle Aged ,Phenotype ,Neurology ,Mutation ,biology.protein ,Epilepsy, Generalized ,Female ,GLUT1 ,Neurology (clinical) ,Haploinsufficiency ,Carbohydrate Metabolism, Inborn Errors ,Follow-Up Studies ,Ketogenic diet - Abstract
Objective: We examined whether glucose transporter 1 (GLUT1) deficiency causes common idiopathic generalized epilepsies (IGEs). Methods: The IGEs are common, heritable epilepsies that usually follow complex inheritance; currently little is known about their genetic architecture. Previously considered rare, GLUT1 deficiency, due to mutations in SLC2A1, leads to failure of glucose transport across the blood–brain barrier and inadequate glucose for brain metabolism. GLUT1 deficiency was first associated with an encephalopathy and more recently found in rare dominant families with epilepsy and paroxysmal exertional dyskinesia (PED). Five hundred four probands with IGEs and 470 controls underwent SLC2A1 sequencing. Glucose transport was assayed following expression of SLC2A1 variants in Xenopus oocytes. All available relatives were phenotyped, and SLC2A1 was sequenced. Results: Functionally validated mutations in SLC2A1 were present in 7 of 504 (1.4%) probands and 0 of 470 controls. PED, undiagnosed prior to study, occurred in 1 proband and 3 of 13 relatives with mutations. The IGEs in probands and relatives were indistinguishable from typical IGE. Three cases (0.6%) had mutations of large functional effect and showed autosomal dominant inheritance or were de novo. Four (0.8%) cases had a subtle functional effect; 2 showed possible dominant inheritance, and 2 did not. These alleles leading to subtle functional impairment may contribute to complex, polygenic inheritance of IGE. Interpretation: SLC2A1 mutations contribute to approximately 1% of IGE both as a dominant gene and as a susceptibility allele in complex inheritance. Diagnosis of GLUT1 deficiency has important treatment (ketogenic diet) and genetic counseling implications. The mechanism of restricted glucose delivery differs from the current focus on IGEs as ion channel disorders. ANN NEUROL 2012;72:807–815
- Published
- 2012
- Full Text
- View/download PDF
30. Early onset absence epilepsy: 1 in 10 cases is caused by GLUT1 deficiency
- Author
-
Samuel F. Berkovic, Hans-Henrik M. Dahl, Ingrid E. Scheffer, Anaïs Thouin, Helen Young, Saul A. Mullen, Todor Arsov, Melinda Nolan, Douglas E. Crompton, John A. Damiano, Kate M. Lawrence, Linda L Huh, and Lynette G. Sadleir
- Subjects
Proband ,Genetics ,medicine.medical_specialty ,Genetic counseling ,Biology ,medicine.disease ,Gastroenterology ,Epilepsy ,Neurology ,Internal medicine ,Cohort ,medicine ,biology.protein ,Missense mutation ,GLUT1 ,Neurology (clinical) ,Haploinsufficiency ,Atonic seizure - Abstract
Glucose transporter 1 (GLUT1) deficiency caused by mutations of SLC2A1 is an increasingly recognized cause of genetic generalized epilepsy. We previously reported that >10% (4 of 34) of a cohort with early onset absence epilepsy (EOAE) had GLUT1 deficiency. This study uses a new cohort of 55 patients with EOAE to confirm that finding. Patients with typical absence seizures beginning before 4 years of age were screened for solute carrier family 2 (facilitated glucose transporter), member 1 (SLC2A1) mutations or deletions. All had generalized spike-waves on electroencephalography (EEG). Those with tonic and/or atonic seizures were excluded. Mutations were found in 7 (13%) of 55 cases, including five missense mutations, an in-frame deletion leading to loss of a single amino acid, and a deletion spanning two exons. Over both studies, 11 (12%) of 89 probands with EOAE have GLUT1 deficiency. Given the major treatment and genetic counseling implications, this study confirms that SLC2A1 mutational analysis should be strongly considered in EOAE.
- Published
- 2012
- Full Text
- View/download PDF
31. Kufs Disease, the Major Adult Form of Neuronal Ceroid Lipofuscinosis, Caused by Mutations in CLN6
- Author
-
Sara E. Mole, Danya F. Vears, John A. Damiano, Silvana Franceschetti, Saul A. Mullen, Sulekha Rajagopalan, Samuel F. Berkovic, Catherine J. Bromhead, Alan McDougall, Umberto Aguglia, Stefano Meletti, Vito Sofia, Patrick Cossette, Michela Morbin, Eva Andermann, Andrea Zini, Todor Arsov, Laura Canafoglia, Frederick Andermann, Katherine R. Smith, Michael A. Farrell, and Melanie Bahlo
- Subjects
Adult ,Male ,Candidate gene ,Heterozygote ,Adolescent ,Genotype ,Genetic Linkage ,Biopsy ,Progressive myoclonus epilepsy ,Biology ,Polymorphism, Single Nucleotide ,Chromosome 15 ,Neuronal Ceroid-Lipofuscinoses ,medicine ,Genetics ,Dementia ,Humans ,Genetics(clinical) ,Genetic Testing ,Allele ,Kufs disease ,Age of Onset ,Genetics (clinical) ,Membrane Proteins ,Exons ,Middle Aged ,medicine.disease ,Pedigree ,Mutation ,Neuronal ceroid lipofuscinosis ,Female ,CLN6 ,progressive myoclonus epilepsy ,Age of onset - Abstract
The molecular basis of Kufs disease is unknown, whereas a series of genes accounting for most of the childhood-onset forms of neuronal ceroid lipofuscinosis (NCL) have been identified. Diagnosis of Kufs disease is difficult because the characteristic lipopigment is largely confined to neurons and can require a brain biopsy or autopsy for final diagnosis. We mapped four families with Kufs disease for whom there was good evidence of autosomal-recessive inheritance and found two peaks on chromosome 15. Three of the families were affected by Kufs type A disease and presented with progressive myoclonus epilepsy, and one was affected by type B (presenting with dementia and motor system dysfunction). Sequencing of a candidate gene in one peak shared by all four families identified no mutations, but sequencing of CLN6, found in the second peak and shared by only the three families affected by Kufs type A disease, revealed pathogenic mutations in all three families. We subsequently sequenced CLN6 in eight other families, three of which were affected by recessive Kufs type A disease. Mutations in both CLN6 alleles were found in the three type A cases and in one family affected by unclassified Kufs disease. Mutations in CLN6 are the major cause of recessive Kufs type A disease. The phenotypic differences between variant late-infantile NCL, previously found to be caused by CLN6, and Kufs type A disease are striking; there is a much later age at onset and lack of visual involvement in the latter. Sequencing of CLN6 will provide a simple diagnostic strategy in this disorder, in which definitive identification usually requires invasive biopsy.
- Published
- 2011
- Full Text
- View/download PDF
32. Development of a rapid functional assay that predicts GLUT1 disease severity
- Author
-
John A. Damiano, Yvonne G. Weber, Snezana Maljevic, Ingrid E. Scheffer, Christopher A. Reid, Michael S. Hildebrand, Slavé Petrovski, Steven Petrou, Elena V. Gazina, Saul A. Mullen, Stéphane Auvin, Sasha M. Zaman, Holger Lerche, Gabriel Davis Jones, A. Marie Phillips, and Samuel F. Berkovic
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Glucose uptake ,Population ,Encephalopathy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Missense mutation ,education ,Genetics (clinical) ,education.field_of_study ,biology ,Glucose transporter ,medicine.disease ,Glucose binding ,030104 developmental biology ,Endocrinology ,biology.protein ,GLUT1 ,Neurology (clinical) ,030217 neurology & neurosurgery ,Glucose Transporter Type 1 - Abstract
ObjectiveTo 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.MethodsGLUT1 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).ResultsDisease 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.ConclusionsDisease 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.
- Published
- 2018
- Full Text
- View/download PDF
33. Somatic GNAQ mutation in the forme fruste of Sturge-Weber syndrome
- Author
-
Renate M Kalnins, Ezgi Ozturk, Alexander Dobrovic, Samuel F. Berkovic, Greta Gillies, Zimeng Ye, Nigel C. Jones, Paul J. Lockhart, Michael S. Hildebrand, Lara McQuillan, Bernadette Nolan, John A. Damiano, Stephen M. Malone, Kate Pope, Hongdo Do, Martin Wood, Richard J. Leventer, Simon Harvey, Ingrid E. Scheffer, and Wirginia J. Maixner
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,business.industry ,Somatic cell ,Sturge–Weber syndrome ,Forme fruste ,Brain tissue ,medicine.disease ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Mutation (genetic algorithm) ,Medicine ,Digital polymerase chain reaction ,Epilepsy surgery ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Genetics (clinical) ,GNAQ - Abstract
ObjectiveTo determine whether the GNAQ R183Q mutation is present in the forme fruste cases of Sturge-Weber syndrome (SWS) to establish a definitive molecular diagnosis.MethodsWe 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.ResultsLow 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.ConclusionsWe 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.
- Published
- 2018
- Full Text
- View/download PDF
34. Loss of synaptic Zn2+ transporter function increases risk of febrile seizures
- Author
-
Steven Petrou, Paul A. Adlard, Susannah T. Bellows, Christopher A. Reid, Arvid Suls, A. Marie Phillips, Verena C. Wimmer, Katia Hardies, Sarah Weckhuysen, Samuel F. Berkovic, John A. Damiano, Rik Hendrickx, Rosemary Burgess, Ingrid E. Scheffer, Michael S. Hildebrand, Jacinta M McMahon, Peter De Jonghe, and Saul A. Mullen
- Subjects
inorganic chemicals ,Risk ,medicine.medical_specialty ,DNA Mutational Analysis ,Molecular Sequence Data ,Inheritance Patterns ,Kaplan-Meier Estimate ,medicine.disease_cause ,Synaptic vesicle ,Seizures, Febrile ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Amino Acid Sequence ,Generalized epilepsy ,Cation Transport Proteins ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Mutation ,Multidisciplinary ,business.industry ,Sodium channel ,Glutamate receptor ,Transporter ,Sequence Analysis, DNA ,medicine.disease ,Transport protein ,Pedigree ,Rats ,Zinc ,Endocrinology ,Anesthesia ,Case-Control Studies ,business ,Engineering sciences. Technology ,Sequence Alignment ,030217 neurology & neurosurgery ,Homeostasis - Abstract
Febrile seizures (FS) are the most common seizure syndrome and are potentially a prelude to more severe epilepsy. Although zinc (Zn2+) metabolism has previously been implicated in FS, whether or not variation in proteins essential for Zn2+ homeostasis contributes to susceptibility is unknown. Synaptic Zn2+ is co-released with glutamate and modulates neuronal excitability. SLC30A3 encodes the zinc transporter 3 (ZNT3), which is primarily responsible for moving Zn2+ 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 Zn2+ increases the risk of FS and more broadly support the idea that impaired synaptic Zn2+ homeostasis can contribute to neuronal hyperexcitability.
- Published
- 2015
35. PRIMA1 mutation: a new cause of nocturnal frontal lobe epilepsy
- Author
-
Antonio Gambardella, Brigid M. Regan, Aiden Eliot Shearer, Samuel F. Berkovic, Francesca Bisulli, Sara Baldassari, Carla Marini, Renzo Guerrini, Hans Henrik M. Dahl, Tommaso Pippucci, Richard J.H. Smith, Ingrid E. Scheffer, Laura Lichetta, Rick M. Tankard, Christopher A. Reid, Steven Petrou, Angelo Labate, Kate M. Lawrence, Melanie Bahlo, Michael S. Hildebrand, John A. Damiano, Paolo Tinuper, Elena V. Gazina, Hildebrand M.S., Tankard R., Gazina E.V., Damiano J.A., Lawrence K.M., Dahl H-H. M., Regan B.M., Shearer A.E., Smith R.J.H., Marini C., Guerrini R., Labate A., Gambardella A., Tinuper P., Licchetta L., Baldassari S., Bisulli F., Pippucci T., Scheffer I.E., Reid C.A., Petrou S., Bahlo M., and Berkovic S.F.
- Subjects
Genetics ,Mutation ,Splice site mutation ,Autosomal recessive nocturnal frontal lobe epilepsy ,General Neuroscience ,Nocturnal frontal lobe epilepsy ,Biology ,medicine.disease_cause ,PRIMA1 ,NFLE ,Exon ,Genetic linkage ,intellectual disability ,Autosomal Dominant ,medicine ,Neurology (clinical) ,Exome ,Exome sequencing ,Research Articles ,Minigene - Abstract
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.
- Published
- 2015
36. Glucose metabolism transporters and epilepsy: only GLUT1 has an established role
- Author
-
Samuel F. Berkovic, Ingrid E. Scheffer, Saul A. Mullen, Karen Oliver, Michael S. Hildebrand, Hans-Henrik M. Dahl, Susannah T. Bellows, and John A. Damiano
- Subjects
Male ,medicine.medical_specialty ,Cohort Studies ,Epilepsy ,Internal medicine ,medicine ,Humans ,Generalized epilepsy ,Child ,Glucose Transporter Type 1 ,biology ,Glucose transporter ,Genetic Variation ,Infant ,medicine.disease ,Endocrinology ,Glucose ,Neurology ,Myoclonic astatic epilepsy ,Child, Preschool ,Mutation ,biology.protein ,GLUT1 ,Female ,Neurology (clinical) ,Haploinsufficiency ,Energy Metabolism ,GLUT3 - Abstract
The availability of glucose, and its glycolytic product lactate, for cerebral energy metabolism is regulated by specific brain transporters. Inadequate energy delivery leads to neurologic impairment. Haploinsufficiency of the glucose transporter GLUT1 causes a characteristic early onset encephalopathy, and has recently emerged as an important cause of a variety of childhood or later-onset generalized epilepsies and paroxysmal exercise-induced dyskinesia. We explored whether mutations in the genes encoding the other major glucose (GLUT3) or lactate (MCT1/2/3/4) transporters involved in cerebral energy metabolism also cause generalized epilepsies. A cohort of 119 cases with myoclonic astatic epilepsy or early onset absence epilepsy was screened for nucleotide variants in these five candidate genes. No epilepsy-causing mutations were identified, indicating that of the major energetic fuel transporters in the brain, only GLUT1 is clearly associated with generalized epilepsy.
- Published
- 2013
37. Recent advances in the molecular genetics of epilepsy
- Author
-
Ingrid E. Scheffer, John A. Damiano, Michael S. Hildebrand, Samuel F. Berkovic, Hans-Henrik M. Dahl, and Richard J.H. Smith
- Subjects
medicine.medical_specialty ,Multifactorial Inheritance ,DNA Copy Number Variations ,Computational biology ,Novel gene ,Epilepsy ,Molecular genetics ,Genetic variation ,Genetics ,medicine ,Clinical genetic ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Molecular Biology ,Genetics (clinical) ,Genetic testing ,medicine.diagnostic_test ,business.industry ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Mutation (genetic algorithm) ,Mutation ,Medical genetics ,business - Abstract
Recent advances in molecular genetics have translated into the increasing utilisation of genetic testing in the routine clinical practice of neurologists. There has been a steady, incremental increase in understanding the genetic variation associated with epilepsies. Genetic testing in the epilepsies is not yet widely practiced, but the advent of new screening technologies promises to exponentially expand both knowledge and clinical utility. To maximise the value of this new genetic insight we need to rapidly extrapolate genetic findings to inform patients of their diagnosis, prognosis, recurrence risk and the clinical management options available for their specific genetic condition. Comprehensive, highly specific and sensitive genetic test results improve the management of patients by neurologists and clinical geneticists. Here we discuss the latest developments in clinical genetic testing for epilepsy and describe new molecular genetics platforms that will transform both genetic screening and novel gene discovery.
- Published
- 2013
38. Cathepsin F mutations cause Type B Kufs disease, an adult-onset neuronal ceroid lipofuscinosis
- Author
-
Frederick Andermann, Paul Saftig, John F. Staropoli, Michael Schwake, Hans Henrik M. Dahl, John A. Damiano, Melanie Bahlo, Harold A. Chapman, Joachim Grötzinger, Eva Andermann, Noreen A. Alexander, Patrick Cossette, Laura Canafoglia, Silvana Franceschetti, Jonathan D. Cooper, Amalia C. Bruni, Michela Morbin, Stirling Carpenter, Katherine R. Smith, Katherine B. Sims, Sara E. Mole, Giorgio Giaccone, and Samuel F. Berkovic
- Subjects
Models, Molecular ,Secondary ,Aging ,Cathepsin F ,DNA Mutational Analysis ,Neurodegenerative ,Compound heterozygosity ,Medical and Health Sciences ,Protein Structure, Secondary ,Consanguinity ,Mice ,Models ,Anterior Horn Cells ,2.1 Biological and endogenous factors ,Missense mutation ,Exome ,Aetiology ,Kufs disease ,Genetics (clinical) ,Exome sequencing ,Genetics & Heredity ,Genetics ,Mice, Knockout ,Chromosome Mapping ,Batten Disease ,General Medicine ,Articles ,Biological Sciences ,Middle Aged ,Pedigree ,Neurological ,Female ,Sequence Analysis ,Adult ,Protein Structure ,Knockout ,Mutation, Missense ,Progressive myoclonus epilepsy ,Biology ,Rare Diseases ,Clinical Research ,Neuronal Ceroid-Lipofuscinoses ,Acquired Cognitive Impairment ,medicine ,Animals ,Humans ,Molecular Biology ,Genetic Association Studies ,Sequence Analysis, RNA ,Neurosciences ,Molecular ,medicine.disease ,Brain Disorders ,Protein Structure, Tertiary ,Case-Control Studies ,Mutation ,RNA ,Dementia ,Neuronal ceroid lipofuscinosis ,Missense ,Lod Score ,Tertiary - Abstract
Kufs disease, an adult-onset neuronal ceroid lipofuscinosis, is challenging to diagnose and genetically heterogeneous. Mutations in CLN6 were recently identified in recessive Kufs disease presenting as progressive myoclonus epilepsy (Type A), whereas the molecular basis of cases presenting with dementia and motor features (Type B) is unknown. We performed genome-wide linkage mapping of two families with recessive Type B Kufs disease and identified a single region on chromosome 11 to which both families showed linkage. Exome sequencing of five samples from the two families identified homozygous and compound heterozygous missense mutations in CTSF within this linkage region. We subsequently sequenced CTSF in 22 unrelated individuals with suspected recessive Kufs disease, and identified an additional patient with compound heterozygous mutations. CTSF encodes cathepsin F, a lysosomal cysteine protease, dysfunction of which is a highly plausible candidate mechanism for a storage disorder like ceroid lipofuscinosis. In silico modeling suggested the missense mutations would alter protein structure and function. Moreover, re-examination of a previously published mouse knockout of Ctsf shows that it recapitulates the light and electron-microscopic pathological features of Kufs disease. Although CTSF mutations account for a minority of cases of type B Kufs, CTSF screening should be considered in cases with early-onset dementia and may avoid the need for invasive biopsies.
- Published
- 2013
39. Small intragenic deletion in FOXP2 associated with childhood apraxia of speech and dysarthria
- Author
-
Samantha J. Turner, Susan Block, John A. Damiano, Sheena Reilly, Michael C Fahey, Ingrid E. Scheffer, Melanie Bahlo, Michael S. Hildebrand, and Angela T Morgan
- Subjects
Adult ,Male ,Stuttering ,Adolescent ,Genotype ,Apraxias ,Dysarthria ,Young Adult ,Motor speech disorders ,Genetics ,medicine ,Humans ,Speech ,Child ,Genetics (clinical) ,Sequence Deletion ,Base Sequence ,business.industry ,FOXP2 ,Forkhead Transcription Factors ,Middle Aged ,medicine.disease ,Uniparental disomy ,Pedigree ,Motor Skills ,Expressive language disorder ,Childhood apraxia of speech ,Speech disorder ,medicine.symptom ,business - Abstract
Relatively little is known about the neurobiological basis of speech disorders although genetic determinants are increasingly recognized. The first gene for primary speech disorder was FOXP2, identified in a large, informative family with verbal and oral dyspraxia. Subsequently, many de novo and familial cases with a severe speech disorder associated with FOXP2 mutations have been reported. These mutations include sequencing alterations, translocations, uniparental disomy, and genomic copy number variants. We studied eight probands with speech disorder and their families. Family members were phenotyped using a comprehensive assessment of speech, oral motor function, language, literacy skills, and cognition. Coding regions of FOXP2 were screened to identify novel variants. Segregation of the variant was determined in the probands' families. Variants were identified in two probands. One child with severe motor speech disorder had a small de novo intragenic FOXP2 deletion. His phenotype included features of childhood apraxia of speech and dysarthria, oral motor dyspraxia, receptive and expressive language disorder, and literacy difficulties. The other variant was found in a family in two of three family members with stuttering, and also in the mother with oral motor impairment. This variant was considered a benign polymorphism as it was predicted to be non-pathogenic with in silico tools and found in database controls. This is the first report of a small intragenic deletion of FOXP2 that is likely to be the cause of severe motor speech disorder associated with language and literacy problems.
- Published
- 2012
40. Early onset absence epilepsy: 1 in 10 cases is caused by GLUT1 deficiency
- Author
-
Todor, Arsov, Saul A, Mullen, John A, Damiano, Kate M, Lawrence, Linda L, Huh, Melinda, Nolan, Helen, Young, Anaïs, Thouin, Hans-Henrik M, Dahl, Samuel F, Berkovic, Douglas E, Crompton, Lynette G, Sadleir, and Ingrid E, Scheffer
- Subjects
Adult ,Male ,Glucose Transporter Type 1 ,Adolescent ,Monosaccharide Transport Proteins ,DNA Mutational Analysis ,Cohort Studies ,Evolution, Molecular ,Epilepsy, Absence ,Child, Preschool ,Mutation ,Animals ,Humans ,Female ,Child ,Carbohydrate Metabolism, Inborn Errors - Abstract
Glucose transporter 1 (GLUT1) deficiency caused by mutations of SLC2A1 is an increasingly recognized cause of genetic generalized epilepsy. We previously reported that10% (4 of 34) of a cohort with early onset absence epilepsy (EOAE) had GLUT1 deficiency. This study uses a new cohort of 55 patients with EOAE to confirm that finding. Patients with typical absence seizures beginning before 4 years of age were screened for solute carrier family 2 (facilitated glucose transporter), member 1 (SLC2A1) mutations or deletions. All had generalized spike-waves on electroencephalography (EEG). Those with tonic and/or atonic seizures were excluded. Mutations were found in 7 (13%) of 55 cases, including five missense mutations, an in-frame deletion leading to loss of a single amino acid, and a deletion spanning two exons. Over both studies, 11 (12%) of 89 probands with EOAE have GLUT1 deficiency. Given the major treatment and genetic counseling implications, this study confirms that SLC2A1 mutational analysis should be strongly considered in EOAE.
- Published
- 2012
41. Glucose transporter 1 deficiency as a treatable cause of myoclonic astatic epilepsy
- Author
-
Davide Mei, John A. Damiano, Saul A. Mullen, Carla Marini, Daniela Buti, Kate M. Lawrence, Peter De Jonghe, Elvio Della Giustina, Samuel F. Berkovic, Renzo Guerrini, Todor Arsov, Ingrid E. Scheffer, and Arvid Suls
- Subjects
Proband ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Epilepsies, Myoclonic ,medicine.disease_cause ,Gastroenterology ,Severity of Illness Index ,Cohort Studies ,Epilepsy ,Arts and Humanities (miscellaneous) ,De Vivo disease ,Internal medicine ,Severity of illness ,Medicine ,Humans ,Child ,Genetics ,Mutation ,Glucose Transporter Type 1 ,business.industry ,medicine.disease ,Treatment Outcome ,Dyskinesia ,Myoclonic astatic epilepsy ,Child, Preschool ,Neurology (clinical) ,Human medicine ,medicine.symptom ,business ,Diet, Ketogenic ,Ketogenic diet - Abstract
ObjectiveTo determine if a significant proportion of patients with myoclonic-astatic epilepsy (MAE) have glucose transporter 1 (GLUT1) deficiency.DesignGenetic analysis.SettingAmbulatory and hospitalized care.PatientsEighty-four unrelated probands with MAE were phenotyped and SLC2A1 was sequenced and analyzed by multiplex ligation-dependent probe amplification. Any identified mutations were then screened in controls.Main Outcome MeasureAny SLC2A1 mutations.ResultsFour of 84 probands with MAE had a mutation of SLC2A1 on sequencing. Multiplex ligation-dependent probe amplification analysis did not reveal any genomic rearrangements in 75 of the remaining cases; 5 could not be tested. Two patients with MAE with SLC2A1 mutations also developed paroxysmal exertional dyskinesia in childhood.ConclusionsFive percent of our patients with MAE had SLC2A1 mutations, suggesting that patients with MAE should be tested for GLUT1 deficiency. Diagnosis of GLUT1 deficiency is a strong indication for early use of the ketogenic diet, which may substantially improve outcome of this severe disorder.
- Published
- 2011
42. Recurrence risk of epilepsy and mental retardation in females due to parental mosaicism of PCDH19 mutations
- Author
-
Tejal A. Desai, John A. Damiano, Anne Ronan, Fiona Haslam McKenzie, Rachel Kneen, Marta A. Bayly, Sarah E. Heron, John C. Mulley, Leanne M. Dibbens, J. Gibbs, Ingrid E. Scheffer, Todor Arsov, Dibbens, LM, Kneen, R, Bayly, MA, Heron, SE, Arsov, T, Damiano, JA, Desai, T, Gibbs, J, McKenzie, F, Mulley, JC, Ronan, A, and Scheffer, IE
- Subjects
Male ,Parents ,Adolescent ,Genetic counseling ,media_common.quotation_subject ,DNA Mutational Analysis ,Germline mosaicism ,Sister ,Polymorphism, Single Nucleotide ,Young Adult ,Epilepsy ,Recurrence ,single nucleotide polymorphism ,Intellectual Disability ,medicine ,Humans ,Missense mutation ,media_common ,Family Health ,Genetics ,Daughter ,Mosaicism ,Cadherins ,medicine.disease ,Protocadherins ,Dravet syndrome ,Developmental disorder ,Mutation (genetic algorithm) ,epilepsy ,Female ,Neurology (clinical) ,Psychology - Abstract
Objective: Two unrelated families were ascertained in which sisters had infantile onset of epilepsy and developmental delay. Mutations in the protocadherin 19 ( PCDH19 ) gene cause epilepsy and mental retardation limited to females (EFMR). Despite both sister pairs having a PCDH19 mutation, neither parent in each family was a heterozygous carrier of the mutation. The possibility of parental mosaicism of PCDH19 mutations was investigated. Methods: Genomic DNA from peripheral blood was obtained and sequenced for PCDH19 mutations. Parentage was confirmed by markers. Results: Both sister pairs have a mutation in PCDH19 . Sister pair 1 has a missense mutation, c.74T>C, L25P, while sequence analysis indicates both of their parents are negative for the mutation. Diagnostic restriction enzyme analysis detected low-level mosaicism of the mutation in their mother. Sister pair 2 are half-sisters who share a mother and each has the missense PCDH19 mutation c.1019 A>G, N340S. The sequence chromatograph of their mother shows reduced signal for the same mutation. These data indicate maternal somatic and gonadal mosaicism of the PCDH19 mutation in both sister pairs. Phenotyping is suggestive of, and PCDH19 mutation detection is diagnostic for, the disorder EFMR in the affected girls. Conclusions: We show that gonadal mosaicism of a PCDH19 mutation in a parent is an important molecular mechanism associated with the inheritance of EFMR. This should be considered when providing genetic counseling for couples who have one affected daughter as they may risk recurrence of affected daughters and having sons at risk of transmitting EFMR.
- Published
- 2011
43. Does variation in NIPA2 contribute to genetic generalized epilepsy?
- Author
-
Saul A. Mullen, Samuel F. Berkovic, Susannah T. Bellows, Ingrid E. Scheffer, John A. Damiano, and Michael S. Hildebrand
- Subjects
Genetics ,Epilepsy ,Variation (linguistics) ,Membrane protein ,medicine ,Biology ,medicine.disease ,Genetic generalized epilepsy ,Molecular medicine ,Genetics (clinical) ,Human genetics - Published
- 2014
- Full Text
- View/download PDF
44. Positional cloning of a cyromazine resistance gene in Drosophila melanogaster
- Author
-
J. Lydall, Zhenzhong Chen, David G. Heckel, Philip J. Batterham, Charles Robin, A. Blasetti, John A. Damiano, Phillip J. Daborn, Christopher Lumb, John A. McKenzie, and Katherine R. Smith
- Subjects
Male ,Positional cloning ,Genes, Insect ,Biology ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Nucleic acid metabolism ,Insecticide Resistance ,chemistry.chemical_compound ,RNA interference ,Genetics ,medicine ,Animals ,Drosophila Proteins ,Cloning, Molecular ,Molecular Biology ,Gene ,Alleles ,Mutation ,Sequence Homology, Amino Acid ,Triazines ,fungi ,biology.organism_classification ,Cyromazine ,Drosophila melanogaster ,chemistry ,Insect Science ,Female ,RNA Interference ,Drosophila Protein - Abstract
Cyromazine is an effective insecticide used to control dipteran insects. Its precise mode of action is yet to be determined, although it has been suggested that it interferes with the hormone system, sclerotization of the cuticle, or nucleic acid metabolism. To understand the way in which cyromazine acts, we have positionally cloned a cyromazine resistance gene from Drosophila melanogaster. Six cyromazine resistance alleles had previously been generated by ethyl methanasulphonate treatment. Two of these failed to complement each other and here we identify them as having independent non-sense mutations in CG32743, which is an ortholog of Smg1 of worms and mammals and encodes a phosphatidylinositol kinase-like kinase (PIKK). RNAi experiments confirm that cyromazine resistance can be achieved by knocking down CG32743. These are the first cyromazine resistant mutations identified at the nucleotide level. In mammals Smg1 phosphorylates P53 in response to DNA damage. This finding supports the hypothesis that cyromazine interferes with nucleic acid metabolism.
- Published
- 2006
45. The Formation and Composition of Lithium Aluminate
- Author
-
John B. Damiano and Harold A. Horan
- Subjects
chemistry.chemical_compound ,Colloid and Surface Chemistry ,chemistry ,Chemical engineering ,Composition (visual arts) ,General Chemistry ,Lithium aluminate ,Biochemistry ,Catalysis - Published
- 1935
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.