1
|
Checri R, Baldassari S, Baulac S. Detection of brain somatic mutations from stereo-EEG electrodes in focal epilepsy: Current advances and future perspectives. Rev Neurol (Paris) 2025; 181:425-431. [PMID: 40187987 DOI: 10.1016/j.neurol.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/02/2025] [Indexed: 04/07/2025]
Abstract
Brain somatic mutations are increasingly recognized as major drivers of focal epilepsy particularly in malformations of cortical development. While traditionally relying on surgically resected tissue for genetic analysis, recent advances in molecular techniques now enable the recovery and analysis of DNA from stereo-electroencephalography (SEEG) electrodes. This minimally invasive approach provides unprecedented opportunities to identify somatic mutations in patients who may not undergo resective surgery. Here, we review the current state of molecular analyses from SEEG electrodes, including recent developments in DNA sequencing, transcriptomics, and epigenetic profiling. We discuss how genetic testing may be integrated into presurgical evaluations, providing new opportunities for comprehensive molecular phenotyping of focal epilepsies. These innovations hold promises in enhancing surgical outcome prediction and guiding toward targeted therapies.
Collapse
Affiliation(s)
- R Checri
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France; Pediatric Neurosurgery Department, CCMR Epilepsies Rares, European Reference Network EpiCare Member, Rothschild Hospital Foundation, 75019 Paris, France
| | - S Baldassari
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - S Baulac
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France.
| |
Collapse
|
2
|
Galvão IC, Lemoine M, Kandratavicius L, Yasuda CL, Alvim MKM, Ghizoni E, Blümcke I, Cendes F, Rogerio F, Lopes-Cendes I, Veiga DFT. Cell type mapping of mild malformations of cortical development with oligodendroglial hyperplasia in epilepsy using single-nucleus multiomics. Epilepsia 2025. [PMID: 40293058 DOI: 10.1111/epi.18413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE Mild malformations of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE) are brain lesions associated with focal epilepsy and characterized by increased oligodendroglial density, heterotopic neurons, and hypomyelination in the white matter. Although previous studies have implicated somatic mutations in the SLC35A2 gene, the cellular and molecular mechanisms underlying MOGHE pathogenesis remain elusive. To address this gap, this study aimed to systematically characterize the cell type composition and molecular alterations of MOGHE lesions at cellular resolution using single-nucleus multiomic profiling. METHODS We performed single-nucleus multiomic sequencing to obtain paired gene expression and chromatin accessibility profiles of >31 000 nuclei from gray matter and white matter regions of MOGHE lesions and compared the results with publicly available neurotypical control datasets. RESULTS The analysis of gray and white matter regions from two MOGHE patients revealed significant cellular composition alterations, including the presence of heterotopic neurons and disease-specific oligodendrocyte populations within the subcortical white matter. MOGHE-specific oligodendrocytes were characterized by the upregulation of synaptic functions and enhanced neuron communication, denoting a possible role in synaptic support and the mediation of glia-neuron interactions in the disease. On the other hand, MOGHE heterotopic neurons were characterized by the upregulation of genes associated with neuronal migration and the Wnt signaling pathway, suggesting a mechanism underlying their atypical localization. SIGNIFICANCE This high-resolution cell type mapping of MOGHE lesions in clinical samples unveils neuronal and glial populations affected by the disease and provides novel insights into the pathophysiological mechanisms of MOGHE.
Collapse
Affiliation(s)
- Isabella C Galvão
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
| | - Manuela Lemoine
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
| | - Ludmyla Kandratavicius
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Pathology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Clarissa L Yasuda
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Marina K M Alvim
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Enrico Ghizoni
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Ingmar Blümcke
- Department of Neuropathology, partner of the European Reference Network EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fernando Cendes
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Neurology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Fabio Rogerio
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
- Department of Pathology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
| | - Diogo F T Veiga
- Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Campinas, Brazil
| |
Collapse
|
3
|
Ono R, Miki S, Koiso T, Sugii N, Sakamoto N, Masuda Y, Matsuda M, Shibuya M, Miyata H, Ishikawa E. Asymptomatic extensive right cerebral hemispheric lesion in an adult with histopathological findings consistent with mild malformation of cortical development with oligodendroglial hyperplasia and epilepsy: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2025; 9:CASE24827. [PMID: 40228416 PMCID: PMC12001064 DOI: 10.3171/case24827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND A mild malformation of cortical development (MCD) with oligodendroglial hyperplasia and epilepsy (MOGHE) is characterized by increased Olig2-positive atypical cells in the white matter and is associated with drug-resistant intractable epilepsy, typically the frontal and temporal lobes. MOGHE is a newly classified subtype of focal cortical dysplasia associated with epilepsy, according to the latest International League Against Epilepsy guidelines. There have been no reports of MOGHE without epilepsy and associated with extensive lesions. OBSERVATIONS A 31-year-old man was incidentally diagnosed with an extensive right cerebral hemispheric lesion on MRI. Pathological examination of the biopsy sample showed no evidence of an invasive tumor but revealed an increase in Olig2-positive cells, consistent with MOGHE. Sanger sequencing showed no IDH1/2, histone H3 TERTpromoter, or SLC35A2 mutations. Electroencephalography showed no evidence of epilepsy. LESSONS This case suggests the existence of a condition with similar histological findings to MOGHE but without epilepsy. The authors propose naming this condition "mild malformation of cortical development with oligodendroglial hyperplasia and no epilepsy." https://thejns.org/doi/10.3171/CASE24827.
Collapse
Affiliation(s)
- Ryohei Ono
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| | - Shunichiro Miki
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| | - Takao Koiso
- Department of Neurosurgery, Mito Saiseikai General Hospital, Mito, Japan
| | - Narushi Sugii
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| | - Noriaki Sakamoto
- Department of Diagnostic Pathology, University of Tsukuba, Japan
| | - Yosuke Masuda
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| | - Masahide Matsuda
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| | - Makoto Shibuya
- Central Laboratory, Hachioji Medical Center, Tokyo Medical University, Hachioji, Japan
| | - Hajime Miyata
- Department of Neuropathology, Research Institute for Brain and Blood Vessels, Akita Cerebrospinal and Cardiovascular Center, Akita, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Japan
| |
Collapse
|
4
|
Luo H, Ruan X, Liu X, Liu Q, Sun Y, Wang Y, Zhang J, Cai L, Jiang Y, Wu Y. Deep sequencing reveals SLC35A2 somatic variants in MOGHE: molecular and clinical insights. J Genet Genomics 2025:S1673-8527(25)00098-0. [PMID: 40220859 DOI: 10.1016/j.jgg.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Affiliation(s)
- Huaxia Luo
- Children's Medical Center, Peking University First Hospital, Beijing 102627, China
| | - Xiaoqin Ruan
- Children's Medical Center, Peking University First Hospital, Beijing 102627, China
| | - Xianyu Liu
- Children's Medical Center, Peking University First Hospital, Beijing 102627, China
| | - Qingzhu Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China
| | - Yu Sun
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China
| | - Yao Wang
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Beijing 102627, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China
| | - Yuwu Jiang
- Children's Medical Center, Peking University First Hospital, Beijing 102627, China; Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China
| | - Ye Wu
- Children's Medical Center, Peking University First Hospital, Beijing 102627, China; Pediatric Epilepsy Center, Peking University First Hospital, Beijing 102627, China.
| |
Collapse
|
5
|
D’Gama AM, Phillips HW, Wang Y, Chiu MY, Chahine Y, Swanson AC, Smith RS, Pearl PL, Tsuboyama M, Madsen JR, Lidov H, Lee EA, Prabhu SP, Huang AY, Stone SSD, Walsh CA, Poduri A. Analysis of DNA from brain tissue on stereo-EEG electrodes reveals mosaic epilepsy-related variants. Brain Commun 2025; 7:fcaf113. [PMID: 40177531 PMCID: PMC11961356 DOI: 10.1093/braincomms/fcaf113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/26/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Somatic mosaic variants contribute to focal epilepsy, with variants often present only in brain tissue and not in blood or other samples typically assayed for genetic testing. Thus, genetic analysis for mosaic variants in focal epilepsy has been limited to patients with drug-resistant epilepsy who undergo surgical resection and have resected brain tissue samples available. Stereo-EEG (sEEG) has become part of the evaluation for many patients with focal drug-resistant epilepsy, and sEEG electrodes provide a potential source of small amounts of brain-derived DNA. We aimed to identify, validate, and assess the distribution of deleterious mosaic variants in epilepsy-associated genes in DNA extracted from trace brain tissue on individual sEEG electrodes. We enrolled a prospective cohort of 10 paediatric patients with drug-resistant epilepsy who had sEEG electrodes implanted for invasive monitoring. We extracted unamplified DNA and in parallel performed whole-genome amplification from trace brain tissue on each sEEG electrode. We also extracted DNA from resected brain tissue and blood/saliva samples where available. We performed deep sequencing (panel and exome) and analysis for candidate germline and mosaic variants. We validated candidate mosaic variants and assessed the variant allele fraction in amplified and unamplified electrode-derived DNA and across electrodes. We extracted unamplified DNA and performed whole-genome amplification from >150 individual electrodes from 10 individuals. Immunohistochemistry confirmed the presence of neurons in the brain tissue on electrodes. Deep sequencing and analysis demonstrated similar depth of coverage between amplified and unamplified DNA samples but significantly more potential mosaic variants in amplified samples. We validated four deleterious mosaic variants in epilepsy-associated genes in electrode-derived DNA in three patients who underwent laser ablation and did not have resected brain tissue samples available. Three of the four variants were detected in both amplified and unamplified electrode-derived DNA, with higher variant allele fraction observed in DNA from electrodes in closest proximity to the electrical seizure focus in one case. We demonstrate that mosaic variants can be identified and validated from DNA extracted from trace brain tissue on individual sEEG electrodes in patients with drug-resistant focal epilepsy, from both unamplified and amplified electrode-derived DNA. Our findings support a relationship between the extent of regional genetic abnormality and electrophysiology and suggest that with further optimization, this minimally invasive diagnostic approach holds promise for advancing precision medicine for patients with drug-resistant epilepsy as part of the surgical evaluation.
Collapse
Affiliation(s)
- Alissa M D’Gama
- Epilepsy Genetics Program, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - H Westley Phillips
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Yilan Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle Y Chiu
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Yasmine Chahine
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Amanda C Swanson
- Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Richard S Smith
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Phillip L Pearl
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Melissa Tsuboyama
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hart Lidov
- Division of Neuropathology, Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eunjung Alice Lee
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Sanjay P Prabhu
- Department of Radiology, Division of Neuroradiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - August Yue Huang
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Scellig S D Stone
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher A Walsh
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Annapurna Poduri
- Epilepsy Genetics Program, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
- National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Cecchini E, Geffers S, Coras R, Schultheis D, Holtzhausen C, Karandasheva K, Herrmann H, Paulsen F, Stadelmann C, Kobow K, Hartlieb T, Bien CG, Lal D, Blumcke I, Hoffmann L. Human brain tissue with MOGHE carrying somatic SLC35A2 variants reveal aberrant protein expression and protein loss in the white matter. Acta Neuropathol 2025; 149:23. [PMID: 40042641 PMCID: PMC11882685 DOI: 10.1007/s00401-025-02858-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/09/2025]
Abstract
Mild Malformation of Cortical Development with Oligodendroglial Hyperplasia in Epilepsy (MOGHE) is a recently described disease entity primarily affecting young children with drug-resistant epilepsy, mainly affecting the frontal lobe. The condition is histopathologically defined by focal lesions with patchy areas of increased oligodendroglial cell density at the grey-white matter boundary and heterotopic neurons in the white matter. Approximately half of the individuals with MOGHE carry brain somatic variants in the SLC35A2 gene, which affects the UDP-galactose transporter and thus sphingolipid glycosylation. To investigate the impact of SLC35A2 variants on protein expression, we analysed MOGHE brain tissue with and without SLC35A2 mosaicism, distinguishing missense from nonsense variants. We developed an antibody targeting the N-terminus of the SLC35A2 galactose transporter and applied it for immunofluorescence (IF) analyses in a MOGHE cohort comprising 59 genetically tested individuals selected from three centres in Germany. The cohort included 13 individuals with SLC35A2 missense variants and 15 with SLC35A2 nonsense variants. Our findings confirm the localisation of the SLC35A2 protein in the Golgi apparatus of all neuroepithelial cell types as well as within Golgi outposts along oligodendroglial processes. The protein distribution was altered in MOGHE samples dependent on the SLC35A2 variant and its allelic frequency. Western blot and IF analyses revealed a significant SLC35A2 reduction in MOGHE tissues carrying nonsense variants. Ultrastructural analyses from three MOGHE samples demonstrated hypomyelination in regions with increased oligodendroglial cell densities, regardless of the harbouring of SLC35A2 variants. Notably, this hypomyelination pattern decreased with age. These results suggested a role for the SLC35A2 protein in the pathogenesis of MOGHE and indicated the presence of additional myelin-associated pathomechanisms in those individuals who do not carry a pathogenic SLC35A2 variant.
Collapse
Affiliation(s)
- Erica Cecchini
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Geffers
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Coras
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Dorothea Schultheis
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Holtzhausen
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Karandasheva
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Herrmann
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Katja Kobow
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Till Hartlieb
- Center for Pediatric Neurology, Neurorehabilitation, and Epileptology, Schoen-Clinic, Vogtareuth, Germany
- Research Institute for Rehabilitation, Transition, and Palliation, Paracelsus Medical University, Salzburg, Austria
| | - Christian G Bien
- Department of Epileptology, Krankenhaus Mara, Bethel Epilepsy Center, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Dennis Lal
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Neurogenetics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ingmar Blumcke
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Lucas Hoffmann
- Department of Neuropathology, Partner of the European Reference Network (ERN) EpiCARE, Universitätsklinikum Erlangen, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
7
|
Blümcke I, Vorndran J. Neuropathology of focal epilepsy: the promise of artificial intelligence and digital Neuropathology 3.0. Pathology 2025; 57:171-177. [PMID: 39827065 DOI: 10.1016/j.pathol.2024.12.386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
Focal lesions of the human neocortex often cause drug-resistant epilepsy, yet surgical resection of the epileptogenic region has been proven as a successful strategy to control seizures in a carefully selected patient cohort. Continuous efforts to study neurosurgically resected brain samples at the microscopic level, i.e., Neuropathology 1.0, unravelled a comprehensive description of the spectrum of underlying aetiologies, e.g., hippocampal sclerosis, congenital brain tumours or cortical malformations as the three most common aetiologies representing almost 80% of the entire lesional landscape. Human brain tissue was also instrumental to discover underlying molecular pathways and common somatic variants, e.g., MTOR, DEPDC5, SLC35A2, BRAF or PTPN11, that helped us to define specific phenotype-genotype associations, thereby promoting novel targets for medical treatment, i.e., Neuropathology 2.0. The increasing gap in accessing necessary resources to perform such studies around the world could be bridged, however, when introducing artificial intelligence (AI)-based algorithms to classify epileptogenic brain lesions on digital slide scans obtained from routine haematoxylin and eosin-stained, formalin-fixed paraffin-embedded tissue sections. This may also provide an advanced prediction of the lesion's phenotype-genotype association in the near future. Thus, digital Neuropathology 3.0 may be the promising next level of laboratory advancement in the realm of neuropathology in focal epilepsy.
Collapse
Affiliation(s)
- Ingmar Blümcke
- Neuropathologisches Institut, Universitätsklinikum Erlangen, Friedrich-Alexander Universität, Germany; Partner of the European Reference Network (ERN) EpiCARE, Germany.
| | - Jörg Vorndran
- Partner of the European Reference Network (ERN) EpiCARE, Germany
| |
Collapse
|
8
|
Liu X, Tang Q, Xia X, Liu Q, Liu J, Jin Y, Wu P, Luo H, Gao K, Ruan X, Sun Y, Ji T, Wang S, Liu X, Cai L, Jiang Y, Dai P, Chen X, Wu Y. Somatic variants in SLC35A2 leading to defects in N-glycosylation in mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE). Acta Neuropathol 2025; 149:13. [PMID: 39900685 DOI: 10.1007/s00401-025-02850-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/05/2025]
Abstract
Mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE) is a new histopathological entity identified in the surgically resected brain tissue of patients with drug-resistant epilepsy. Somatic variants in SLC35A2 have been increasingly identified in MOGHE brain resections. SLC35A2 protein transports uridine 5'-diphosphogalactose (UDP-Gal) into the Golgi lumen, playing a crucial role in the process of N-glycosylation. Currently, research on the pathogenic mechanism of SLC35A2 variants in MOGHE is limited. Here we conducted genetic testing on brain samples and paired blood samples from 28 pediatric patients pathologically diagnosed with MOGHE. We performed an in-depth functional analysis of somatic variants identified in SLC35A2, integrating glycan labeling and intact glycopeptide profiling to assess N-glycosylation defects. With whole-exome sequencing and validation with ultra-deep amplicon sequencing, we identified 101 potentially pathogenic somatic variants (PPSVs) across 87 genes. Nine PPSVs in SLC35A2 were found in 10 samples. The 9 identified variants of SLC35A2, characterized by various mutation types (4 frameshift, 3 missense and 2 nonsense variants), were all confirmed to be loss-of-function via altered glycan chains. Intact glycopeptide analysis at the cellular level indicated an increase in truncated N-glycan glycoforms. Analysis of brain tissue revealed N-glycosylated proteins and glycosites modified with agalactosylated glycoforms, and glycoproteins bearing agalactosylated N-glycans were significantly enriched in cell adhesion and axon guidance-related pathways. Additionally, chemoenzymatic glycan labeling in lesions demonstrated N-glycan damage of heterotopic neurons, suggesting a potential diagnostic approach for MOGHE. Our findings provide a comprehensive somatic landscape of MOGHE and a rich resource of somatic SLC35A2 variant-related glycoform and glycoprotein abnormalities, thereby unveiling valuable insights into compromised N-glycosylation and MOGHE formation.
Collapse
Affiliation(s)
- Xianyu Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Qi Tang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Xiaoqian Xia
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Qingzhu Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Jialin Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yangyanan Jin
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Pengxia Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Huaxia Luo
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xiaoqin Ruan
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yu Sun
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Taoyun Ji
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Shuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Xiaoyan Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Peng Dai
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China.
- Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Peking University, Beijing, China.
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China.
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Synthetic and Functional Biomolecules Center, and Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Beijing National Laboratory for Molecular Sciences, Synthetic and Functional Biomolecules Center, Peking University, Beijing, China.
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China.
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China.
| |
Collapse
|
9
|
Macdonald‐Laurs E, Leventer RJ. ILAE genetic literacy series: Focal cortical dysplasia. Epileptic Disord 2025; 27:1-8. [PMID: 39641771 PMCID: PMC11829622 DOI: 10.1002/epd2.20308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 12/07/2024]
Abstract
Focal cortical dysplasia (FCD) is a common cause of drug-resistant focal epilepsy in children and young adults and is often surgically remediable. The genetics of FCD are increasingly understood due to the ability to perform genomic testing including deep sequencing of resected FCD tissue specimens. There is clear evidence that FCD type II occurs secondary to both germline and somatic mTOR pathway variants, while emerging literature supports the role of SLC35A2, a glycosylation gene, in mild malformation of cortical development with oligodendroglial hyperplasia and epilepsy (MOGHE). Herein, we provide a review of FCDs focusing on their clinical phenotypes, genetic basis, and management considerations when performing genetic testing in this patient group.
Collapse
Affiliation(s)
- Emma Macdonald‐Laurs
- Department of NeurologyThe Royal Children's HospitalParkvilleVictoriaAustralia
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
| | - Richard J. Leventer
- Department of NeurologyThe Royal Children's HospitalParkvilleVictoriaAustralia
- Murdoch Children's Research InstituteParkvilleVictoriaAustralia
- Department of PaediatricsThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
10
|
Zhan Y, Chen S, Jin Z, Zhou J, Zhang YX, Hou Q, Wang Y, Zheng G, Zheng Y. Mild Malformation of Cortical Development With Oligodendroglial Hyperplasia and Epilepsy: A Systematic Review. Neurol Genet 2025; 11:e200240. [PMID: 39902276 PMCID: PMC11790282 DOI: 10.1212/nxg.0000000000200240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/02/2024] [Indexed: 02/05/2025]
Abstract
Background and Objectives Mild malformation of cortical development with oligodendroglial hyperplasia and epilepsy (MOGHE) is a newly described rare entity of drug-resistant epilepsy, with a wide spectrum of presentations. We aim to describe the diagnostic features and prognosis of MOGHE in a large cohort. Methods We performed a systematic review preregistered on PROSPERO (CRD42023472978), in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement. We searched PubMed, Embase, Scopus, and ScienceDirect between database inception and November 30, 2023, for all published studies on MOGHE. Inclusion criteria were a histopathologic diagnosis of MOGHE. The risk of bias was analyzed with a standardized tool specifically for case reports and case series. The demographic, clinical, EEG, neuroimaging, genetic, and neuropathologic features; treatments; and prognosis were extracted and analyzed. Subgroup analysis was performed with the age at onset and SLC35A2 variant status. Results A total of 163 patients with MOGHE from 18 studies were included in the analysis. The median age at seizure onset was 1.2 years, and 103 were male. Ninety-five patients presented with unilobed lesions. Ninety-nine had lesions in the frontal lobe. A total of 101 patients achieved a favorable surgical outcome. Patients with an onset before 10 years were more likely to present with epileptic spasms, the West syndrome, a circumscribed pattern of interictal EEG, intellectual disabilities, and a better seizure outcome, compared with those with an onset age 10 years and older. Forty-five patients (72.6%) were SLC35A2-positive. Patients harboring the SLC35A2 variants were more likely to present as Lennox-Gastaut syndrome, when compared with those who were SLC35A2-negative. Discussion MOGHE is a distinct entity of drug-resistant epilepsy associated with SLC35A2 variants, characterized by age-dependent phenotypes. The study emphasizes the clinical pearls indicative of the rare disease, which may facilitate early recognition and appropriate selection of treatments. The included studies were case reports or series, which were mainly limited by selection and reporting biases.
Collapse
Affiliation(s)
- Yixin Zhan
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shijia Chen
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenghan Jin
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiping Zhou
- Department of Neurology, Wayne State University/Detroit Medical Center, Detroit, MI; and
| | - Yin-Xi Zhang
- Department of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Hou
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Wang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guoqing Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Kim SH, Kang H, Roh YH, Hahn J, Min KL, Lee S, Yang D, Choi HS, Park S, Lee JH, Lee S, Kim SH, Chang MJ, Kim HD. Efficacy and safety of everolimus for patients with focal cortical dysplasia type 2. Epilepsia Open 2025; 10:243-257. [PMID: 39607729 PMCID: PMC11803298 DOI: 10.1002/epi4.13104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE This study aimed to evaluate the effectiveness and safety of everolimus in treating seizures associated with focal cortical dysplasia type 2 (FCD 2). METHODS A prospective, crossover, placebo-controlled clinical trial (ClinicalTrials.gov: NCT03198949) enrolled patients aged 4-40 years with pathologically confirmed FCD 2 and a history of ≥3 seizures per month for two out of the 3 months prior to screening. The trial included a 4-week baseline phase, two 12-week core phases, and a 29-week extension phase. Patients received everolimus or placebo in a blinded manner during core phase I, with crossover to the alternate treatment in core phase II. Everolimus dosage started at 4.5 mg/m2/day, targeting a serum level of 5-15 ng/mL. The primary outcome was the proportion of patients achieving ≥50% seizure reduction from baseline in the last month of each core phase. Safety profiles were compared between groups. RESULTS Between May 11, 2017, and June 19, 2020, 21 patients completed the core phases. There was no significant difference in the primary outcome between everolimus and placebo groups (24% vs. 19%, p = 0.66). The patients showed varied responses. Three patients with a pathogenic variant in the MTOR gene or no genetic abnormalities achieved seizure freedom with everolimus in the last month of the core phase, while none of the patients with variants in other genes did. Adverse events, such as mucositis or skin ulceration, were more common with everolimus (19/21 vs. 7/21, p < 0.001). All adverse events resolved without study drug withdrawal. SIGNIFICANCE Everolimus treatment for 12 weeks did not show overall superiority in reducing seizures compared to placebo. However, it showed promise, mostly in patients with a pathogenic variant in the MTOR gene, highlighting the need for further research into patient-specific factors influencing treatment response. The everolimus treatment was generally safe and manageable. PLAIN LANGUAGE SUMMARY This study tested everolimus for reducing seizures in patients with focal cortical dysplasia type 2 (FCD 2). While the drug was not more effective than a placebo for most, few patients showed better results, with some becoming seizure-free. Side effects were common but manageable. More research is needed to understand why certain patients respond better to treatment.
Collapse
Affiliation(s)
- Se Hee Kim
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
| | - Hoon‐Chul Kang
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
| | - Yun Ho Roh
- Biostatistics Collaboration Unit, Department of Biomedical Systems InformaticsYonsei University College of MedicineSeoulRepublic of Korea
| | - Jongsung Hahn
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of PharmacyYonsei UniversityIncheonRepublic of Korea
- School of PharmacyJeonbuk National UniversityJeonjuRepublic of Korea
| | - Kyung Lok Min
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of PharmacyYonsei UniversityIncheonRepublic of Korea
- Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and PharmacyYonsei UniversityIncheonRepublic of Korea
| | - Seok‐Jin Lee
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
| | - Donghwa Yang
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
- Division of Pediatric Neurology, Department of PediatricsNational Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Han Som Choi
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
- Department of PediatricsEwha Womans University Seoul Hospital, Ewha Womans University School of MedicineSeoulRepublic of Korea
| | - Soyoung Park
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
- Department of PediatricsSoonchunhyang University Bucheon Hospital, Soonchunhyang University College of MedicineBucheonRepublic of Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST)SoVarGen, Inc.DaejeonRepublic of Korea
| | - Sang‐Guk Lee
- Department of Laboratory MedicineSeverance Hospital, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Se Hoon Kim
- Department of PathologyYonsei University College of MedicineSeoulRepublic of Korea
| | - Min Jung Chang
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of PharmacyYonsei UniversityIncheonRepublic of Korea
- Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and PharmacyYonsei UniversityIncheonRepublic of Korea
- Graduate Program of Industrial Pharmaceutical ScienceYonsei UniversityIncheonRepublic of Korea
| | - Heung Dong Kim
- Pediatric Neurology, Department of PediatricsYonsei University College of Medicine, Severance Children's Hospital, Epilepsy Research InstituteSeoulRepublic of Korea
- Department of PediatricsKangbuk Samsung Hospital, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| |
Collapse
|
12
|
Coleman M, Wang M, Snell P, Lee WS, D'Arcy C, Mignone C, Pope K, Gillies G, Maixner W, Wray A, Harvey AS, Simons C, Leventer RJ, Stephenson SEM, Lockhart PJ, Howell KB. The genetic landscape and classification of infantile epileptic spasms syndrome requiring surgery due to suspected focal brain malformations. Brain Commun 2025; 7:fcaf034. [PMID: 39926610 PMCID: PMC11806418 DOI: 10.1093/braincomms/fcaf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/17/2024] [Accepted: 01/24/2025] [Indexed: 02/11/2025] Open
Abstract
Infantile epileptic spasms syndrome is a severe epilepsy of infancy that is often associated with focal malformations of cortical development. This study aimed to elucidate the genetic landscape and histopathologic aetiologies of infantile epileptic spasms syndrome due to focal malformations of cortical development requiring surgery. Fifty-nine children with a history of infantile epileptic spasms syndrome and focal malformations of cortical development on MRI were studied. Genetic testing of resected brain tissue was performed by high-coverage targeted panel sequencing or exome sequencing. Histopathology and MRI were reviewed, and integrated clinico-pathological diagnoses were established. A genetic diagnosis was achieved in 47 children (80% of cohort). Germline pathogenic variants were identified in 27/59 (46%) children, in TSC2 (x19), DEPDC5 (x2), CDKL5 (x2), NPRL3 (x1), FGFR1 (x1), TSC1 (x1), and one child with both a TUBB2A/TUBB2B deletion and a pathogenic variant in COL4A1 (x1). Pathogenic brain somatic variants were identified in 21/59 (36%) children, in SLC35A2 (x9), PIK3CA (x3), AKT3 (x2), TSC2 (x2), MTOR (x2), OFD1 (x1), TSC1 (x1) and DEPDC5 (x1). One child had 'two-hit' diagnosis, with both germline and somatic pathogenic DEPDC5 variants in trans. Multimodal data integration resulted in clinical diagnostic reclassifications in 24% of children, emphasizing the importance of combining genetic, histopathologic and imaging findings. Mammalian target of rapamycin pathway variants were identified in most children with tuberous sclerosis or focal cortical dysplasia type II. All nine children with somatic SLC35A2 variants in brain were reclassified to mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy. Somatic mosaicism was a major cause of focal cortical dysplasia type II/hemimegalencephaly (81%) and mild malformation of cortical development with oligodendroglial hyperplasia (100%). The genetic landscape of infantile epileptic spasms syndrome due to focal malformations comprises germline and somatic variants in a range of genes, with mTORopathies and SLC35A2-related mild malformation of cortical development with oligodendroglial hyperplasia being the major causes. Multimodal data integration incorporating genetic data aids in optimizing diagnostic pathways and can guide surgical decision-making and inform future research and therapeutic interventions.
Collapse
Affiliation(s)
- Matthew Coleman
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Min Wang
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
| | - Penny Snell
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
| | - Wei Shern Lee
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Colleen D'Arcy
- Department of Anatomical Pathology, The Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Cristina Mignone
- Department of Medical Imaging, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Kate Pope
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
| | - Greta Gillies
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
| | - Wirginia Maixner
- Department of Neurosurgery, The Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Alison Wray
- Department of Neurosurgery, The Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - A Simon Harvey
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria 3052, Australia
- Department of Neurology, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Cas Simons
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Centre for Population Genomics, Garvin Institute of Medical Research, and UNSW Sydney, Sydney, New South Wales 2010, Australia
| | - Richard J Leventer
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Neurology, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Sarah E M Stephenson
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Paul J Lockhart
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Katherine B Howell
- Murdoch Children’s Research Institute, Parkville, Victoria 3052, Australia
- Department of Neurology, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
| |
Collapse
|
13
|
Corrigan RR, Mashburn-Warren LM, Yoon H, Bedrosian TA. Somatic Mosaicism in Brain Disorders. ANNUAL REVIEW OF PATHOLOGY 2025; 20:13-32. [PMID: 39227323 DOI: 10.1146/annurev-pathmechdis-111523-023528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Research efforts over the past decade have defined the genetic landscape of somatic variation in the brain. Neurons accumulate somatic mutations from development through aging with potentially profound functional consequences. Recent studies have revealed the contribution of somatic mosaicism to various brain disorders including focal epilepsy, neuropsychiatric disease, and neurodegeneration. One notable finding is that the effect of somatic mosaicism on clinical outcomes can vary depending on contextual factors, such as the developmental origin of a variant or the number and type of cells affected. In this review, we highlight current knowledge regarding the role of somatic mosaicism in brain disorders and how biological context can mediate phenotypes. First, we identify the origins of brain somatic variation throughout the lifespan of an individual. Second, we explore recent discoveries that suggest somatic mosaicism contributes to various brain disorders. Finally, we discuss neuropathological associations of brain mosaicism in different biological contexts and potential clinical utility.
Collapse
Affiliation(s)
- Rachel R Corrigan
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA;
| | | | - Hyojung Yoon
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA;
| | - Tracy A Bedrosian
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA;
| |
Collapse
|
14
|
Lai D, Sosicka P, Williams DJ, Bowyer ME, Ressler AK, Kohrt SE, Muron SJ, Crino PB, Freeze HH, Boland MJ, Heinzen EL. SLC35A2 loss of function variants affect glycomic signatures, neuronal fate, and network dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630524. [PMID: 39763953 PMCID: PMC11703275 DOI: 10.1101/2024.12.27.630524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
SLC35A2 encodes a UDP-galactose transporter essential for glycosylation of proteins and galactosylation of lipids and glycosaminoglycans. Germline genetic SLC35A2 variants have been identified in congenital disorders of glycosylation and somatic SLC35A2 variants have been linked to intractable epilepsy associated with malformations of cortical development. However, the functional consequences of these pathogenic variants on brain development and network integrity remain elusive. In this study, we use an isogenic human induced pluripotent stem cell-derived neuron model to comprehensively interrogate the functional impact of loss of function variants in SLC35A2 through the integration of cellular and molecular biology, protein glycosylation analysis, neural network dynamics, and single cell electrophysiology. We show that loss of function variants in SLC35A2 result in disrupted glycomic signatures and precocious neurodevelopment, yielding hypoactive, asynchronous neural networks. This aberrant network activity is attributed to an inhibitory/excitatory imbalance as characterization of neural composition revealed preferential differentiation of SLC35A2 loss of function variants towards the GABAergic fate. Additionally, electrophysiological recordings of synaptic activity reveal a shift in excitatory/inhibitory balance towards increased inhibitory drive, indicating changes occurring specifically at the pre-synaptic terminal. Our study is the first to provide mechanistic insight regarding the early development and functional connectivity of SLC35A2 loss of function variant harboring human neurons, providing important groundwork for future exploration of potential therapeutic interventions.
Collapse
Affiliation(s)
- Dulcie Lai
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Paulina Sosicka
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Damian J Williams
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - MaryAnn E Bowyer
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Andrew K Ressler
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sarah E Kohrt
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Savannah J Muron
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Michael J Boland
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Epilepsy and Neurodevelopmental Disorders, Perelman School of Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Erin L Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
15
|
Sanders MWCB, Koeleman BPC, Brilstra EH, Jansen FE, Baldassari S, Chipaux M, Sim NS, Ko A, Kang HC, Blümcke I, Lal D, Baulac S, Lee JH, Aronica E, Braun KPJ. Somatic variant analysis of resected brain tissue in epilepsy surgery patients. Epilepsia 2024; 65:e209-e215. [PMID: 39460693 DOI: 10.1111/epi.18148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
We studied the distribution of germline and somatic variants in epilepsy surgery patients with (suspected) malformations of cortical development (MCD) who underwent surgery between 2015 and 2020 at University Medical Center Utrecht (the Netherlands) and pooled our data with four previously published cohort studies. Tissue analysis yielded a pathogenic variant in 203 of 663 (31%) combined cases. In 126 of 379 (33%) focal cortical dysplasia (FCD) type II cases and 23 of 37 (62%) hemimegalencephaly cases, a pathogenic variant was identified, mostly involving the mTOR signaling pathway. Pathogenic variants in 10 focal epilepsy genes were found in 48 of 178 (27%) FCDI/mild MCD/mMCD with oligodendroglial hyperplasia and epilepsy cases; 36 of these (75%) were SLC35A2 variants. Six of 69 (9%) patients without a histopathological lesion had a pathogenic variant in SLC35A2 (n = 5) or DEPDC5 (n = 1). A germline variant in blood DNA was confirmed in all cases with a pathogenic variant in tissue, with a variant allele frequency (VAF) of ~50%. In seven of 114 patients (6%) with a somatic variant in tissue, mosaicism in blood was detected. More than half of pathogenic somatic variants had a VAF < 5%. Further analysis of the correlation between genetic variants and surgical outcomes will improve patient counseling and may guide postoperative treatment decisions.
Collapse
Affiliation(s)
- Maurits W C B Sanders
- Department of Child Neurology, University Medical Center Utrecht Brain Center, Utrecht, the Netherlands
| | - Bobby P C Koeleman
- Department of Genetics, Center for Molecular Medicine, University Medical Center, Utrecht, the Netherlands
| | - Eva H Brilstra
- Department of Genetics, Center for Molecular Medicine, University Medical Center, Utrecht, the Netherlands
| | - Floor E Jansen
- Department of Child Neurology, University Medical Center Utrecht Brain Center, Utrecht, the Netherlands
| | - Sara Baldassari
- Hôpital de la Pitié Salpêtrière, Institut du Cerveau-Paris Brain Institute, Sorbonne Université, Paris, France
| | - Mathilde Chipaux
- Department of Pediatric Neurosurgery, Rothschild Foundation Hospital, Paris, France
| | - Nam Suk Sim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ara Ko
- Division of Pediatric Neurology, Department of Pediatrics, Pediatric Epilepsy Clinics, Severance Children's Hospital, Seoul, Republic of Korea
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Pediatric Epilepsy Clinics, Severance Children's Hospital, Seoul, Republic of Korea
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States
- Cologne Center for Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Stéphanie Baulac
- Hôpital de la Pitié Salpêtrière, Institut du Cerveau-Paris Brain Institute, Sorbonne Université, Paris, France
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
- Stichting Epilepsie Instellingen Nederland, Heemstede, the Netherlands
| | - Kees P J Braun
- Department of Child Neurology, University Medical Center Utrecht Brain Center, Utrecht, the Netherlands
| |
Collapse
|
16
|
Yoon H, Ringland A, Anderson JJ, Sran S, Elziny S, Huynh C, Shinagawa N, Badertscher S, Corrigan RR, Mashburn-Warren L, Amari F, Chen M, Coppola V, Crino PB, Bedrosian TA. Mouse models of Slc35a2 brain mosaicism reveal mechanisms of mild malformations of cortical development with oligodendroglial hyperplasia in epilepsy. Epilepsia 2024; 65:3717-3731. [PMID: 39460689 DOI: 10.1111/epi.18166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVE Brain somatic variants in SLC35A2 were recently identified as a genetic marker for mild malformations of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE). The role of SLC35A2 in cortical development and the contributions of abnormal neurons and oligodendrocytes to seizure activity in MOGHE remain largely unexplored. METHODS Here, we generated a novel Slc35a2 floxed allele, which we used to develop two Slc35a2 conditional knockout mouse lines targeting (1) the Emx1 dorsal telencephalic lineage (excitatory neurons and glia) and (2) the Olig2 lineage (oligodendrocytes). We examined brain structure, behavior, and seizure activity. RESULTS Knockout of Slc35a2 from the Emx1 lineage, which targets both cortical neurons and oligodendrocytes, resulted in early lethality and caused abnormal cortical development, increased oligodendroglial cell density, early onset seizures, and developmental delays akin to what is observed in patients with MOGHE. By tracing neuronal development with 5-Ethynyl-2'-deoxyuridine (EdU) birthdating experiments, we found that Slc35a2 deficiency disrupts corticogenesis by delaying radial migration of neurons from the subventricular zone. To discern the contributions of oligodendrocytes to these phenotypes, we knocked out Slc35a2 from the Olig2 lineage. This recapitulated the increased oligodendroglial cell density and resulted in abnormal electroencephalographic activity, but without a clear seizure phenotype, suggesting Slc35a2 deficiency in neurons is required for epileptogenesis. SIGNIFICANCE This study presents two novel Slc35a2 conditional knockout mouse models and characterizes the effects on brain development, behavior, and epileptogenesis. Together, these results demonstrate a direct causal role for SLC35A2 in MOGHE-like phenotypes, including a critical role in neuronal migration during brain development, and identify neurons as key contributors to SLC35A2-related epileptogenesis.
Collapse
Affiliation(s)
- Hyojung Yoon
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Amanda Ringland
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - James J Anderson
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sahibjot Sran
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Soad Elziny
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cindy Huynh
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Noriyuki Shinagawa
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Samantha Badertscher
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Rachel R Corrigan
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | - Foued Amari
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio, USA
| | - Min Chen
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, Ohio, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tracy A Bedrosian
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
17
|
Cases‐Cunillera S, Quatraccioni A, Rossini L, Ruffolo G, Ono T, Baulac S, Auvin S, O'Brien TJ, Henshall DC, Akman Ö, Sankar R, Galanopoulou AS. WONOEP appraisal: The role of glial cells in focal malformations associated with early onset epilepsies. Epilepsia 2024; 65:3457-3468. [PMID: 39401070 PMCID: PMC11647439 DOI: 10.1111/epi.18126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024]
Abstract
Epilepsy represents a common neurological disorder in patients with developmental brain lesions, particularly in association with malformations of cortical development and low-grade glioneuronal tumors. In these diseases, genetic and molecular alterations in neurons are increasingly discovered that can trigger abnormalities in the neuronal network, leading to higher neuronal excitability levels. However, the mechanisms underlying epilepsy cannot rely solely on assessing the neuronal component. Growing evidence has revealed the high degree of complexity underlying epileptogenic processes, in which glial cells emerge as potential modulators of neuronal activity. Understanding the role of glial cells in developmental brain lesions such as malformations of cortical development and low-grade glioneuronal tumors is crucial due to the high degree of pharmacoresistance characteristic of these lesions. This has prompted research to investigate the role of glial and immune cells in epileptiform activity to find new therapeutic targets that could be used as combinatorial drug therapy. In a special session of the XVI Workshop of the Neurobiology of Epilepsy (WONOEP, Talloires, France, July 2022) organized by the Neurobiology Commission of the International League Against Epilepsy, we discussed the evidence exploring the genetic and molecular mechanisms of glial cells and immune response and their implications in the pathogenesis of neurodevelopmental pathologies associated with early life epilepsies.
Collapse
Affiliation(s)
- Silvia Cases‐Cunillera
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and GliomaParisFrance
| | - Anne Quatraccioni
- Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of BonnBonnGermany
| | - Laura Rossini
- Epilepsy UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Gabriele Ruffolo
- Department of Physiology and PharmacologyIstituto Pasteur–Fondazione Cenci Bolognetti, University of Rome SapienzaRomeItaly
- IRCCS San Raffaele RomaRomeItaly
| | - Tomonori Ono
- Epilepsy Center, National Hospital Organization Nagasaki Medical CenterŌmuraJapan
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, AP‐HP, Hôpital de la Pitié SalpêtrièreParisFrance
| | - Stéphane Auvin
- Pediatric Neurology Department, AP‐HP, Robert Debré University HospitalCRMR épilepsies Rares, EpiCARE memberParisFrance
- Université Paris Cité, INSERM NeuroDiderotParisFrance
- Institut Universitaire de FranceParisFrance
| | - Terence J. O'Brien
- Department of Neuroscience, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of NeurologyRoyal Melbourne HospitalMelbourneVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
- Department of Medicine (Royal Melbourne Hospital)University of MelbourneMelbourneVictoriaAustralia
| | - David C. Henshall
- Department of Physiology and Medical Physics, RCSIUniversity of Medicine and Health SciencesDublinIreland
| | - Özlem Akman
- Department of PhysiologyFaculty of Medicine, Demiroglu Bilim UniversityIstanbulTurkey
| | - Raman Sankar
- Department of Pediatrics and NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominique P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
18
|
Vorndran J, Blümcke I. A deep-learning-based histopathology classifier for focal cortical dysplasia (FCD) unravels a complex scenario of comorbid FCD subtypes. Epilepsia 2024; 65:3501-3512. [PMID: 39440630 DOI: 10.1111/epi.18161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Recently, we developed a first artificial intelligence (AI)-based digital pathology classifier for focal cortical dysplasia (FCD) as defined by the ILAE classification. Herein, we tested the usefulness of the classifier in a retrospective histopathology workup scenario. METHODS Eighty-six new cases with histopathologically confirmed FCD ILAE type Ia (FCDIa), FCDIIa, FCDIIb, mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE), or mild malformations of cortical development were selected, 20 of which had confirmed gene mosaicism. RESULTS The classifier always recognized the correct histopathology diagnosis in four or more 1000 × 1000-μm digital tiles in all cases. Furthermore, the final diagnosis overlapped with the largest batch of tiles assigned by the algorithm to one diagnostic entity in 80.2% of all cases. However, 86.2% of all cases revealed more than one diagnostic category. As an example, FCDIIb was identified in all of the 23 patients with histopathologically assigned FCDIIb, whereas the classifier correctly recognized FCDIIa tiles in 19 of these cases (83%), that is, dysmorphic neurons but no balloon cells. In contrast, the classifier misdiagnosed FCDIIb tiles in seven of 23 cases histopathologically assigned to FCDIIa (33%). This mandates a second look by the signing histopathologist to either confirm balloon cells or differentiate from reactive astrocytes. The algorithm also recognized coexisting architectural dysplasia, for example, vertically oriented microcolumns as in FCDIa, in 22% of cases classified as FCDII and in 62% of cases with MOGHE. Microscopic review confirmed microcolumns in the majority of tiles, suggesting that vertically oriented architectural abnormalities are more common than previously anticipated. SIGNIFICANCE An AI-based diagnostic classifier will become a helpful tool in our future histopathology laboratory, in particular when large anatomical resections from epilepsy surgery require extensive resources. We also provide an open access web application allowing the histopathologist to virtually review digital tiles obtained from epilepsy surgery to corroborate their final diagnosis.
Collapse
Affiliation(s)
- Jörg Vorndran
- Department of Neuropathology, University Hospital Erlangen, member of EpiCare European Reference Network, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, member of EpiCare European Reference Network, Erlangen, Germany
| |
Collapse
|
19
|
Quatraccioni A, Cases-Cunillera S, Balagura G, Coleman M, Rossini L, Mills JD, Casillas-Espinosa PM, Moshé SL, Sankar R, Baulac S, Noebels JL, Auvin S, O'Brien TJ, Henshall DC, Akman Ö, Galanopoulou AS. WONOEP appraisal: Genetic insights into early onset epilepsies. Epilepsia 2024; 65:3138-3154. [PMID: 39302576 DOI: 10.1111/epi.18124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
Early onset epilepsies occur in newborns and infants, and to date, genetic aberrations and variants have been identified in approximately one quarter of all patients. With technological sequencing advances and ongoing research, the genetic diagnostic yield for specific seizure disorders and epilepsies is expected to increase. Genetic variants associated with epilepsy include chromosomal abnormalities and rearrangements of various sizes as well as single gene variants. Among these variants, a distinction can be made between germline and somatic, with the latter being increasingly identified in epilepsies with focal cortical malformations in recent years. The identification of the underlying genetic mechanisms of epilepsy syndromes not only revolutionizes the diagnostic schemes but also leads to a better understanding of the diseases and their interrelationships, ultimately providing new opportunities for therapeutic targeting. At the XVI Workshop on Neurobiology of Epilepsy (WONOEP 2022, Talloires, France, July 2022), various etiologies, research models, and mechanisms of genetic early onset epilepsies were presented and discussed.
Collapse
Affiliation(s)
- Anne Quatraccioni
- Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Silvia Cases-Cunillera
- Neuronal Signaling in Epilepsy and Glioma, Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Paris, France
| | - Ganna Balagura
- Department of Neuroscience, Ophthalmology, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Matthew Coleman
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Laura Rossini
- Epilepsy Unit, IRCCS Foundation, C. Besta Neurological Institute, Milan, Italy
| | - James D Mills
- Department of Clinical and Experimental Epilepsy, University College London Queen Square Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Chalfont St. Peter, UK
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Solomon L Moshé
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, and Department of Neuroscience and Pediatrics, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | - Raman Sankar
- Department of Neurology and Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jeffrey L Noebels
- Departments of Neurology, Neuroscience, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Stéphane Auvin
- AP-HP, Hôpital Robert-Debré, INSERM NeuroDiderot, DMU Innov-RDB, Neurologie Pédiatrique, member of European Reference Network EpiCARE, Université Paris Cité and Institut Universitaire de France, Paris, France
| | - Terence J O'Brien
- Department of Neuroscience, Alfred Hospital, Monash University, Melbourne, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - David C Henshall
- Department of Physiology and Medical Physics and FutureNeuro SFI Research Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Özlem Akman
- Department of Physiology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
20
|
Spyrou J, Aung KP, Vanyai H, Leventer RJ, Maljevic S, Lockhart PJ, Howell KB, Reid CA. Slc35a2 mosaic knockout impacts cortical development, dendritic arborisation, and neuronal firing. Neurobiol Dis 2024; 201:106657. [PMID: 39236911 DOI: 10.1016/j.nbd.2024.106657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE) is an important cause of drug-resistant epilepsy. A significant subset of individuals diagnosed with MOGHE display somatic mosaicism for loss-of-function variants in SLC35A2, which encodes the UDP-galactose transporter. We developed a mouse model to investigate how disruption of this transporter leads to a malformation of cortical development. We used in utero electroporation and CRISPR/Cas9 to knockout Slc35a2 in a subset of layer 2/3 cortical neuronal progenitors in the developing brains of male and female fetal mice to model mosaic expression. Mosaic Slc35a2 knockout was verified through next-generation sequencing and immunohistochemistry of GFP-labelled transfected cells. Histology of brain tissue in mosaic Slc35a2 knockout mice revealed the presence of upper layer-derived cortical neurons in the white matter. Reconstruction of single filled neurons identified altered dendritic arborisation with Slc35a2 knockout neurons having increased complexity. Whole-cell electrophysiological recordings revealed that Slc35a2 knockout neurons display reduced action potential firing, increased afterhyperpolarisation duration and reduced burst-firing when compared with control neurons. Mosaic Slc35a2 knockout mice also exhibited significantly increased epileptiform spiking and increased locomotor activity. We successfully generated a mouse model of mosaic Slc35a2 deficiency, which recapitulates features of the human phenotype, including impaired neuronal migration. We show that knockout in layer 2/3 cortical neuron progenitors is sufficient to disrupt neuronal excitability, increase epileptiform activity and cause hyperactivity in mosaic mice. Our mouse model provides an opportunity to further investigate the disease mechanisms that contribute to MOGHE and facilitate the development of precision therapies.
Collapse
Affiliation(s)
- James Spyrou
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Khaing Phyu Aung
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Hannah Vanyai
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Richard J Leventer
- Department of Neurology, Royal Children's Hospital, Parkville, VIC 3052, Australia; Murdoch Children's Research Institute, Parkville, VIC 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Snezana Maljevic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia
| | - Paul J Lockhart
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Katherine B Howell
- Department of Neurology, Royal Children's Hospital, Parkville, VIC 3052, Australia; Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3052, Australia; Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia.
| |
Collapse
|
21
|
Liu J, Binding L, Puntambekar I, Patodia S, Lim YM, Mryzyglod A, Xiao F, Pan S, Mito R, de Tisi J, Duncan JS, Baxendale S, Koepp M, Thom M. Microangiopathy in temporal lobe epilepsy with diffusion MRI alterations and cognitive decline. Acta Neuropathol 2024; 148:49. [PMID: 39377933 PMCID: PMC11461556 DOI: 10.1007/s00401-024-02809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024]
Abstract
White matter microvascular alterations in temporal lobe epilepsy (TLE) may be relevant to acquired neurodegenerative processes and cognitive impairments associated with this condition. We quantified microvascular changes, myelin, axonal, glial and extracellular-matrix labelling in the gyral core and deep temporal lobe white matter regions in surgical resections from 44 TLE patients with or without hippocampal sclerosis. We compared this pathology data with in vivo pre-operative MRI diffusion measurements in co-registered regions and neuropsychological measures of cognitive impairment and decline. In resections, increased arteriolosclerosis was observed in TLE compared to non-epilepsy controls (greater sclerotic index, p < 0.001), independent of age. Microvascular changes included increased vascular densities in some regions but uniformly reduced mean vascular size (quantified with collagen-4, p < 0.05-0.0001), and increased pericyte coverage of small vessels and capillaries particularly in deep white matter (quantified with platelet-derived growth factor receptorβ and smooth muscle actin, p < 0.01) which was more marked the longer the duration of epilepsy (p < 0.05). We noted increased glial numbers (Olig2, Iba1) but reduced myelin (MAG, PLP) in TLE compared to controls, particularly prominent in deep white matter. Gene expression analysis showed a greater reduction of myelination genes in HS than non-HS cases and with age and correlation with diffusion MRI alterations. Glial densities and vascular size were increased with increased MRI diffusivity and vascular density with white matter abnormality quantified using fixel-based analysis. Increased perivascular space was associated with reduced fractional anisotropy as well as age-accelerated cognitive decline prior to surgery (p < 0.05). In summary, likely acquired microangiopathic changes in TLE, including vascular sclerosis, increased pericyte coverage and reduced small vessel size, may indicate a functional alteration in contractility of small vessels and haemodynamics that could impact on tissue perfusion. These morphological features correlate with white matter diffusion MRI alterations and might explain cognitive decline in TLE.
Collapse
Affiliation(s)
- Joan Liu
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Neuroscience, University of Westminster, London, UK
| | - Lawrence Binding
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Centre for Medical Image Computing, Department of Computer Science, UCL, London, UK
| | - Isha Puntambekar
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Smriti Patodia
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Yau Mun Lim
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alicja Mryzyglod
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Fenglai Xiao
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Shengning Pan
- Department of Statistical Science, University College London, Gower St., London, UK
| | - Remika Mito
- Department of Neuroscience and Mental Health, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Jane de Tisi
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - John S Duncan
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Sallie Baxendale
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
22
|
Liu Y, Li Y, Zhang Y, Fang Y, Lei L, Yu J, Tan H, Sui L, Guo Q, Zhou L. Excitatory neurons and oligodendrocyte precursor cells are vulnerable to focal cortical dysplasia type IIIa as suggested by single-nucleus multiomics. Clin Transl Med 2024; 14:e70072. [PMID: 39440467 PMCID: PMC11497056 DOI: 10.1002/ctm2.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Focal cortical dysplasia (FCD) is a heterogeneous group of cortical developmental malformations that constitute a common cause of medically intractable epilepsy. FCD type IIIa (FCD IIIa) refers to temporal neocortex alterations in architectural organisation or cytoarchitectural composition in the immediate vicinity of hippocampal sclerosis. Slight alterations in the temporal neocortex of FCD IIIa patients pose a challenge for the preoperative diagnosis and definition of the resection range. METHODS We have performed multimodal integration of single-nucleus RNA sequencing and single-nucleus assay for transposase-accessible chromatin sequencing in the epileptogenic cortex of four patients with FCD IIIa, and three relatively normal temporal neocortex were chosen as controls. RESULTS Our study revealed that the most significant dysregulation occurred in excitatory neurons (ENs) and oligodendrocyte precursor cells (OPCs) in the epileptogenic cortex of FCD IIIa patients. In ENs, we constructed a transcription factor (TF)-hub gene regulatory network and found DAB1high ENs subpopulation mediates neuronal immunity characteristically in FCD IIIa. Western blotting and immunofluorescence were used to validate the changes in protein expression levels caused by some of the key genes. The OPCs were activated and exhibited aberrant phenotypes in FCD IIIa, and TFs regulating reconstructed pseudotime trajectory were identified. Finally, our results revealed aberrant intercellular communication between ENs and OPCs in FCD IIIa patients. CONCLUSIONS Our study revealed significant and intricate alterations in the transcriptomes and epigenomes in ENs and OPCs of FCD IIIa patients, shedding light on their cell type-specific regulation and potential pathogenic involvement in this disorder. This work will help evaluate the pathogenesis of cortical dysplasia and epilepsy and explore potential therapeutic targets. KEY POINTS Paired snRNA-seq and snATAC-seq data were intergrated and analysed to identify crucial subpopulations of ENs and OPCs in the epileptogenic cortex of FCD IIIa patients and explore their possible pathogenic role in the disease. A TF-hub gene regulatory network was constructed in ENs, and the DAB1high Ex-1 mediated neuronal immunity was characterstically in FCD IIIa patients. The OPCs were activated and exhibited aberrant phenotypes in FCD IIIa patients, and TFs regulating reconstructed pseudotime traectory were identified. Aberrant intercelluar communications between ENs and OPCs in FCD IIIa patients were identified.
Collapse
Affiliation(s)
- Yingying Liu
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Department of NeurologyThird Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yinchao Li
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yaqian Zhang
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yubao Fang
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Lei Lei
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Jiabin Yu
- Department of Epilepsy CenterThe Second Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Hongping Tan
- Epilepsy CenterGuangdong Sanjiu Brain HospitalGuangzhouGuangdongChina
| | - Lisen Sui
- Department of Epilepsy CenterThe Second Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Qiang Guo
- Epilepsy CenterGuangdong Sanjiu Brain HospitalGuangzhouGuangdongChina
| | - Liemin Zhou
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
23
|
Kim J, Park SM, Koh HY, Ko A, Kang HC, Chang WS, Kim DS, Lee JH. Threshold of somatic mosaicism leading to brain dysfunction with focal epilepsy. Brain 2024; 147:2983-2990. [PMID: 38916065 DOI: 10.1093/brain/awae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 06/26/2024] Open
Abstract
Somatic mosaicism in a fraction of brain cells causes neurodevelopmental disorders, including childhood intractable epilepsy. However, the threshold for somatic mosaicism leading to brain dysfunction is unknown. In this study, we induced various mosaic burdens in focal cortical dysplasia type II (FCD II) mice, featuring mTOR somatic mosaicism and spontaneous behavioural seizures. The mosaic burdens ranged from approximately 1000 to 40 000 neurons expressing the mTOR mutant in the somatosensory or medial prefrontal cortex. Surprisingly, approximately 8000-9000 neurons expressing the MTOR mutant, extrapolated to constitute 0.08%-0.09% of total cells or roughly 0.04% of variant allele frequency in the mouse hemicortex, were sufficient to trigger epileptic seizures. The mutational burden was correlated with seizure frequency and onset, with a higher tendency for electrographic inter-ictal spikes and beta- and gamma-frequency oscillations in FCD II mice exceeding the threshold. Moreover, mutation-negative FCD II patients in deep sequencing of their bulky brain tissues revealed somatic mosaicism of the mTOR pathway genes as low as 0.07% in resected brain tissues through ultra-deep targeted sequencing (up to 20 million reads). Thus, our study suggests that extremely low levels of somatic mosaicism can contribute to brain dysfunction.
Collapse
Affiliation(s)
- Jintae Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Sang Min Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- SoVarGen Co., Ltd., Daejeon 34051, Republic of Korea
| | - Hyun Yong Koh
- Department of Pediatrics and Neurology, Baylor College of Medicine, Houston 77030, USA
| | - Ara Ko
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hoon-Chul Kang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Won Seok Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Seok Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- SoVarGen Co., Ltd., Daejeon 34051, Republic of Korea
| |
Collapse
|
24
|
Sran S, Ringland A, Bedrosian TA. Building the brain mosaic: an expanded view. Trends Genet 2024; 40:747-756. [PMID: 38853120 PMCID: PMC11387136 DOI: 10.1016/j.tig.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024]
Abstract
The complexity of the brain is closely tied to its nature as a genetic mosaic, wherein each cell is distinguished by a unique constellation of somatic variants that contribute to functional and phenotypic diversity. Postzygotic variation arising during neurogenesis is recognized as a key contributor to brain mosaicism; however, recent advances have broadened our understanding to include sources of neural genomic diversity that develop throughout the entire lifespan, from embryogenesis through aging. Moving beyond the traditional confines of neurodevelopment, in this review, we delve into the complex mechanisms that enable various origins of brain mosaicism.
Collapse
Affiliation(s)
- Sahibjot Sran
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Amanda Ringland
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Tracy A Bedrosian
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
25
|
Cui TY, Wu H, Tang CY, Wang XF, Li TF, Zhou J. Surgical outcomes of patients with genetically refractory epilepsy: A systematic review and meta-analysis. Seizure 2024; 120:124-134. [PMID: 38959583 DOI: 10.1016/j.seizure.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE To summarize the surgical outcomes of genetically refractory epilepsy and identify prognostic factors for these outcomes. METHODS A literature search of the PubMed, Web of Science, and Embase databases for relevant studies, published between January 1, 2002 and December 31, 2023, was performed using specific search terms. All studies addressing surgical outcomes and follow-up of genetically refractory epilepsy were included. All statistical analyses were performed using STATA software (StataCorp LLC, College Station, TX, USA). This review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses, 2020 (i.e., "PRISMA") reporting guidelines. RESULTS Of the 3833 studies retrieved, 55 fulfilled the inclusion criteria. Eight studies were eligible for meta-analysis at the study level. Pooled outcomes revealed that 74 % of patients who underwent resective surgery (95 % confidence interval [CI] 0.55-0.89; z = 9.47, p < 0.05) achieved Engel I status at the last follow-up. In the study level analysis, pooled outcomes revealed that 9 % of patients who underwent vagus nerve stimulation achieved seizure-free status (95 % CI 0.00-0.31; z = 1.74, p < 0.05), and 61 % (95 % CI 0.55-0.89; z = 11.96, p < 0.05) achieved a 50 % reduction in seizure frequency at the last follow-up. Fifty-three studies comprising 249 patients were included in an individual-level analysis. Among patients who underwent lesion resection or lobectomy/multilobar resection, 65 % (100/153) achieved Engel I status at the last follow-up. Univariate analysis indicated that female sex, somatic mutations, and presenting with focal seizure symptoms were associated with better prognosis (p < 0.05). Additionally, 75 % (21/28) of patients who underwent hemispherectomy/hemispherotomy achieved Engel I status at the last follow-up. In the individual-level analysis, among patients treated with vagus nerve stimulation, 21 % (10/47) were seizure-free and 64 % (30/47) experienced >50 % reduction in seizure frequency compared with baseline. CONCLUSION Meticulous presurgical evaluation and selection of appropriate surgical procedures can, to a certain extent, effectively control seizures. Therefore, various surgical procedures should be considered when treating patients with genetically refractory epilepsy.
Collapse
Affiliation(s)
- Tian-Yi Cui
- Functional Neurosurgery Department, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Han Wu
- Functional Neurosurgery Department, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chong-Yang Tang
- Functional Neurosurgery Department, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Xiong-Fei Wang
- Functional Neurosurgery Department, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tian-Fu Li
- Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Clinical Research on Epilepsy, Beijing, China; Centre of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Jian Zhou
- Functional Neurosurgery Department, Sanbo Brain Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Clinical Research on Epilepsy, Beijing, China; Centre of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
26
|
Chowdhury SR, Whitney R, RamachandranNair R, Bijarnia Mahay S, Sharma S. Genetic Testing in Pediatric Epilepsy: Tools, Tips, and Navigating the Traps. Pediatr Neurol 2024; 157:42-49. [PMID: 38865949 DOI: 10.1016/j.pediatrneurol.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/17/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
With the advent of high-throughput sequencing and computational methods, genetic testing has become an integral part of contemporary clinical practice, particularly in epilepsy. The toolbox for genetic testing has evolved from conventional chromosomal microarray and epilepsy gene panels to state-of-the-art sequencing techniques in the modern genomic era. Beyond its potential for therapeutic benefits through precision medicine, optimizing the choice of antiseizure medications, or exploring nonpharmacological therapeutic modalities, genetic testing carries substantial diagnostic, prognostic, and personal implications. Developmental and epileptic encephalopathies, the coexistence of neurodevelopmental comorbidities, early age of epilepsy onset, unexplained drug-refractory epilepsy, and positive family history have demonstrated the highest likelihood of yielding positive genetic test results. Given the diagnostic efficacy across different testing modalities, reducing costs of next-generation sequencing tests, and genetic diversity of epilepsies, exome sequencing or genome sequencing, where feasible and available, have been recommended as the first-tier test. Comprehensive clinical phenotyping at the outset, corroborative evidence from radiology and electrophysiology-based investigations, reverse phenotyping, and periodic reanalysis are some of the valuable strategies when faced with inconclusive test results. In this narrative review, the authors aim to simplify the approach to genetic testing in epilepsy by guiding on the selection of appropriate testing tools in the indicated clinical scenarios, addressing crucial aspects during pre- and post-test counseling sessions, adeptly navigating the traps posed by uncertain or negative genetic variants, and paving the way forward to the emerging testing modalities beyond DNA sequencing.
Collapse
Affiliation(s)
- Sayoni Roy Chowdhury
- Department of Paediatrics, Lady Hardinge Medical College and Associated Kalawati Saran Children's Hospital, New Delhi, India
| | - Robyn Whitney
- Comprehensive Paediatric Epilepsy Program, Division of Neurology, Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Rajesh RamachandranNair
- Comprehensive Paediatric Epilepsy Program, Division of Neurology, Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Sunita Bijarnia Mahay
- Sr. Consultant, Clinical & Metabolic Geneticist, Institute of Medical Genetics & Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Suvasini Sharma
- Department of Paediatrics, Lady Hardinge Medical College and Associated Kalawati Saran Children's Hospital, New Delhi, India.
| |
Collapse
|
27
|
Elziny S, Sran S, Yoon H, Corrigan RR, Page J, Ringland A, Lanier A, Lapidus S, Foreman J, Heinzen EL, Iffland P, Crino PB, Bedrosian TA. Loss of Slc35a2 alters development of the mouse cerebral cortex. Neurosci Lett 2024; 836:137881. [PMID: 38909838 DOI: 10.1016/j.neulet.2024.137881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/25/2024]
Abstract
Brain somatic variants in SLC35A2, an intracellular UDP-galactose transporter, are commonly identified mutations associated with drug-resistant neocortical epilepsy and developmental brain malformations, including focal cortical dysplasia type I and mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE). However, the causal effects of altered SLC35A2 function on cortical development remain untested. We hypothesized that focal Slc35a2 knockout (KO) or knockdown (KD) in the developing mouse cortex would disrupt cortical development and change network excitability. Through two independent studies, we used in utero electroporation (IUE) to introduce CRISPR/Cas9/targeted guide RNAs or short-hairpin RNAs into the embryonic mouse brain at day 14.5-15.5 to achieve Slc35a2 KO or KD, respectively, from neural precursor cells. Slc35a2 KO or KD caused disrupted radial migration of electroporated neurons evidenced by heterotopic cells located in lower cortical layers and in the sub-cortical white matter. Slc35a2 KO in neurons did not induce changes in oligodendrocyte number, importantly suggesting that the oligodendroglial hyperplasia observed in MOGHE originates from distinct cell autonomous effects of Slc35a2 mutations. Adult KO mice were implanted with EEG electrodes for 72-hour continuous recording. Spontaneous seizures were not observed in focal Slc35a2 KO mice, but there was reduced seizure threshold following pentylenetetrazol injection. Here we demonstrate that focal Slc35a2 KO or KD in vivo disrupts corticogenesis through altered neuronal migration and that KO leads to reduced seizure threshold. Together these results demonstrate a direct causal role for SLC35A2 in cortical development.
Collapse
Affiliation(s)
- Soad Elziny
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sahibjot Sran
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Hyojung Yoon
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Rachel R Corrigan
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - John Page
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Amanda Ringland
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Anna Lanier
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Sara Lapidus
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - James Foreman
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Erin L Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy and Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Philip Iffland
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Peter B Crino
- Dept. of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tracy A Bedrosian
- Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, United States; Dept. of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States.
| |
Collapse
|
28
|
D'Gama AM, Phillips HW, Wang Y, Chiu MY, Chahine Y, Swanson AC, Smith RS, Pearl PL, Tsuboyama M, Madsen JR, Lidov H, Lee EA, Prabhu SP, Huang AY, Stone SSD, Walsh CA, Poduri A. Analysis of DNA from brain tissue on stereo-EEG electrodes reveals mosaic epilepsy-related variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.21.24310779. [PMID: 39108522 PMCID: PMC11302611 DOI: 10.1101/2024.07.21.24310779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Somatic mosaic variants contribute to focal epilepsy, but genetic analysis has been limited to patients with drug-resistant epilepsy (DRE) who undergo surgical resection, as the variants are mainly brain-limited. Stereoelectroencephalography (sEEG) has become part of the evaluation for many patients with focal DRE, and sEEG electrodes provide a potential source of small amounts of brain-derived DNA. We aimed to identify, validate, and assess the distribution of potentially clinically relevant mosaic variants in DNA extracted from trace brain tissue on individual sEEG electrodes. We enrolled a prospective cohort of eleven pediatric patients with DRE who had sEEG electrodes implanted for invasive monitoring, one of whom was previously reported. We extracted unamplified DNA from the trace brain tissue on each sEEG electrode and also performed whole-genome amplification for each sample. We extracted DNA from resected brain tissue and blood/saliva samples where available. We performed deep panel and exome sequencing on a subset of samples from each case and analysis for potentially clinically relevant candidate germline and mosaic variants. We validated candidate mosaic variants using amplicon sequencing and assessed the variant allele fraction (VAF) in amplified and unamplified electrode-derived DNA and across electrodes. We extracted DNA from >150 individual electrodes from 11 individuals and obtained higher concentrations of whole-genome amplified vs unamplified DNA. Immunohistochemistry confirmed the presence of neurons in the brain tissue on electrodes. Deep sequencing and analysis demonstrated similar depth of coverage between amplified and unamplified samples but significantly more called mosaic variants in amplified samples. In addition to the mosaic PIK3CA variant detected in a previously reported case from our group, we identified and validated four potentially clinically relevant mosaic variants in electrode-derived DNA in three patients who underwent laser ablation and did not have resected brain tissue samples available. The variants were detected in both amplified and unamplified electrode-derived DNA, with higher VAFs observed in DNA from electrodes in closest proximity to the electrical seizure focus in some cases. This study demonstrates that mosaic variants can be identified and validated from DNA extracted from trace brain tissue on individual sEEG electrodes in patients with drug-resistant focal epilepsy and in both amplified and unamplified electrode-derived DNA samples. Our findings support a relationship between the extent of regional genetic abnormality and electrophysiology, and suggest that with further optimization, this minimally invasive diagnostic approach holds promise for advancing precision medicine for patients with DRE as part of the surgical evaluation.
Collapse
|
29
|
Macdonald-Laurs E, Dzau W, Warren AEL, Coleman M, Mignone C, Stephenson SEM, Howell KB. Identification and treatment of surgically-remediable causes of infantile epileptic spasms syndrome. Expert Rev Neurother 2024; 24:661-680. [PMID: 38814860 DOI: 10.1080/14737175.2024.2360117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Infantile epileptic spasms syndrome (IESS) is a common developmental and epileptic encephalopathy with poor long-term outcomes. A substantial proportion of patients with IESS have a potentially surgically remediable etiology. Despite this, epilepsy surgery is underutilized in this patient group. Some surgically remediable etiologies, such as focal cortical dysplasia and malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE), are under-diagnosed in infants and young children. Even when a surgically remediable etiology is recognised, for example, tuberous sclerosis or focal encephalomalacia, epilepsy surgery may be delayed or not considered due to diffuse EEG changes, unclear surgical boundaries, or concerns about operating in this age group. AREAS COVERED In this review, the authors discuss the common surgically remediable etiologies of IESS, their clinical and EEG features, and the imaging techniques that can aid in their diagnosis. They then describe the surgical approaches used in this patient group, and the beneficial impact that early epilepsy surgery can have on developing brain networks. EXPERT OPINION Epilepsy surgery remains underutilized even when a potentially surgically remediable cause is recognized. Overcoming the barriers that result in under-recognition of surgical candidates and underutilization of epilepsy surgery in IESS will improve long-term seizure and developmental outcomes.
Collapse
Affiliation(s)
- Emma Macdonald-Laurs
- Department of Neurology, The Royal Children's Hospital, Parkville, VIC, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Winston Dzau
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Aaron E L Warren
- Department of Medicine (Austin Health), The University of Melbourne, Melbourne, VIC, Australia
- Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Matthew Coleman
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Cristina Mignone
- Department of Medical Imaging, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Sarah E M Stephenson
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Katherine B Howell
- Department of Neurology, The Royal Children's Hospital, Parkville, VIC, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| |
Collapse
|
30
|
Klein KM, Mascarenhas R, Merrikh D, Khanbabaei M, Maroilley T, Kaur N, Liu Y, Soule T, Manalo M, Tamura G, Jacobs J, Hader W, Pfeffer G, Tarailo-Graovac M. Identification of a mosaic MTOR variant in purified neuronal DNA in a patient with focal cortical dysplasia using a novel depth electrode harvesting technique. Epilepsia 2024; 65:1768-1776. [PMID: 38587282 DOI: 10.1111/epi.17980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
OBJECTIVE Recent studies have identified brain somatic variants as a cause of focal epilepsy. These studies relied on resected tissue from epilepsy surgery, which is not available in most patients. The use of trace tissue adherent to depth electrodes used for stereo electroencephalography (EEG) has been proposed as an alternative but is hampered by the low cell quality and contamination by nonbrain cells. Here, we use our improved depth electrode harvesting technique that purifies neuronal nuclei to achieve molecular diagnosis in a patient with focal cortical dysplasia (FCD). METHODS Depth electrode tips were collected, pooled by brain region and seizure onset zone, and nuclei were isolated and sorted using fluorescence-activated nuclei sorting (FANS). Somatic DNA was amplified from neuronal and astrocyte nuclei using primary template amplification followed by exome sequencing of neuronal DNA from the affected pool, unaffected pool, and saliva. The identified variant was validated using droplet digital polymerase chain reaction (PCR). RESULTS An 11-year-old male with drug-resistant genetic-structural epilepsy due to left anterior insula FCD had seizures from age 3 years. Stereo EEG confirmed seizure onset in the left anterior insula. The two anterior insula electrodes were combined as the affected pool and three frontal electrodes as the unaffected pool. FANS isolated 140 neuronal nuclei from the affected and 245 neuronal nuclei from the unaffected pool. A novel somatic missense MTOR variant (p.Leu489Met, CADD score 23.7) was identified in the affected neuronal sample. Droplet digital PCR confirmed a mosaic gradient (variant allele frequency = .78% in affected neuronal sample; variant was absent in all other samples). SIGNIFICANCE Our findings confirm that harvesting neuronal DNA from depth electrodes followed by molecular analysis to identify brain somatic variants is feasible. Our novel method represents a significant improvement compared to the previous method by focusing the analysis on high-quality cells of the cell type of interest.
Collapse
Affiliation(s)
- Karl Martin Klein
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rumika Mascarenhas
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Daria Merrikh
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Maryam Khanbabaei
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Tatiana Maroilley
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Navprabhjot Kaur
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Yiping Liu
- Flow Cytometry Core Facility, University of Calgary, Calgary, Alberta, Canada
| | - Tyler Soule
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Minette Manalo
- Department of Pediatrics, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Goichiro Tamura
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Julia Jacobs
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Pediatrics, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada
| | - Walter Hader
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Gerald Pfeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Maja Tarailo-Graovac
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Arizono E, Tanei ZI, Iijima K, Kimura Y, Shigemoto Y, Maki H, Kusama M, Murayama K, Iwasaki M, Saito T, Saito Y, Saito K, Sato N. MRI detection of mild malformation of cortical development with oligodendroglial hyperplasia (MOGHE) on T1WI-CHESS. Epilepsy Behav Rep 2024; 26:100674. [PMID: 38764719 PMCID: PMC11101927 DOI: 10.1016/j.ebr.2024.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024] Open
Abstract
Mild malformation of cortical development with oligodendroglial hyperplasia (MOGHE) is a recently proposed epileptogenic entity that is difficult to detect on MRI. We present a case of MOGHE that was successfully detected on T1WI-chemical shift-selective saturation (CHESS) MRI. The clinical presentation, MRI including T1WI-CHESS, functional images, and pathology findings of a 14-year-old Japanese girl diagnosed with MOGHE are described. T1WI-CHESS revealed an abnormal high signal along the affected lesion, whereas the findings shown by the other MR sequences were less obvious; interictal fluorodeoxyglucose-positron emission tomography indicated slightly decreased accumulation in the lesion, and subtraction ictal single photon emission computed tomography co-registered to MRI showed an increased blood flow. Together these observations suggest that T1WI-CHESS may be a useful MR sequence for detecting the lesions in patients with MOGHE.
Collapse
Affiliation(s)
- Elly Arizono
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Radiology, Tokyo Medical University, School of Medicine, Tokyo, Japan
| | - Zen-ichi Tanei
- Department of Cancer Pathology, Hokkaido University, Faculty of Medicine, Hokkaido, Japan
| | - Keiya Iijima
- Department of Neurosurgery, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yukio Kimura
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoko Shigemoto
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroyuki Maki
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Midori Kusama
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kumiko Murayama
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masaki Iwasaki
- Department of Neurosurgery, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takashi Saito
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuko Saito
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kazuhiro Saito
- Department of Radiology, Tokyo Medical University, School of Medicine, Tokyo, Japan
| | - Noriko Sato
- Department of Radiology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
32
|
Wang Y, Peng X, Wu M, Wang B, Chen T, Zhan X. SLC35A2 expression is associated with HER2 expression in breast cancer. Discov Oncol 2024; 15:124. [PMID: 38639872 PMCID: PMC11031507 DOI: 10.1007/s12672-024-00978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
The role of SLC35A2 in breast cancer remains poorly understood, with limited available information on its significance. This study aimed to investigate the expression of SLC35A2 and clinicopathological variables in breast cancer patients. Immunohistochemical analysis of SLC35A2 protein was conductedon 40 adjacent non-neoplastic tissues and 320 breast cancer tissues. The study also assesed the association between SLC35A2 expression and breast cancer clinicopathological features of breast cancer, as well as its impact on overall survival. In comparison to adjacent non-neoplastic tissues, a significantly higher expression of SLC35A2 was observed in breast cancer tissues (P = 0.020), and this expression was found to be independently correlated with HER2 positivity (P = 0.001). Survival analysis indicated that patients with low SLC35A2 expression had a more favorable prognosis in HER2-positive subtype breast cancer (P = 0.017). These results suggest that SLC35A2 is overexpressed in breast cancer tissues compared to adjacent non-neoplastic tissues and may serve as a potential prognostic marker for HER2-positive subtype breast cancer. Furthermore, breast cancer patients with the HER2 positive subtype who exhibited decreased levels of SLC35A2 expression demonstrated improved long-term prognostic outcomes.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xiaobo Peng
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meihong Wu
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Bin Wang
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Tianran Chen
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xianbao Zhan
- Department of Oncology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
33
|
Gooley S, Perucca P, Tubb C, Hildebrand MS, Berkovic SF. Somatic mosaicism in focal epilepsies. Curr Opin Neurol 2024; 37:105-114. [PMID: 38235675 DOI: 10.1097/wco.0000000000001244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
PURPOSE OF REVIEW Over the past decade, it has become clear that brain somatic mosaicism is an important contributor to many focal epilepsies. The number of cases and the range of underlying pathologies with somatic mosaicism are rapidly increasing. This growth in somatic variant discovery is revealing dysfunction in distinct molecular pathways in different focal epilepsies. RECENT FINDINGS We briefly summarize the current diagnostic yield of pathogenic somatic variants across all types of focal epilepsy where somatic mosaicism has been implicated and outline the specific molecular pathways affected by these variants. We will highlight the recent findings that have increased diagnostic yields such as the discovery of pathogenic somatic variants in novel genes, and new techniques that allow the discovery of somatic variants at much lower variant allele fractions. SUMMARY A major focus will be on the emerging evidence that somatic mosaicism may contribute to some of the more common focal epilepsies such as temporal lobe epilepsy with hippocampal sclerosis, which could lead to it being re-conceptualized as a genetic disorder.
Collapse
Affiliation(s)
- Samuel Gooley
- Epilepsy Research Centre, Department of Medicine, University of Melbourne
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Heidelberg
| | - Piero Perucca
- Epilepsy Research Centre, Department of Medicine, University of Melbourne
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Heidelberg
- Department of Neuroscience, Central Clinical School, Monash University
- Department of Neurology, Alfred Health, Melbourne
- Department of Neurology, The Royal Melbourne Hospital
| | - Caitlin Tubb
- Epilepsy Research Centre, Department of Medicine, University of Melbourne
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, University of Melbourne
- Neuroscience Group, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Heidelberg
| |
Collapse
|
34
|
Blümcke I. Neuropathology and epilepsy surgery - 2024 update. FREE NEUROPATHOLOGY 2024; 5:8. [PMID: 38532826 PMCID: PMC10964794 DOI: 10.17879/freeneuropathology-2024-5347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
Neuropathology-based studies in neurosurgically resected brain tissue obtained from carefully examined patients with focal epilepsies remain a treasure box for excellent insights into human neuroscience, including avenues to better understand the neurobiology of human brain organization and neuronal hyperexcitability at the cellular level including glio-neuronal interaction. It also allows to translate results from animal models in order to develop personalized treatment strategies in the near future. A nice example of this is the discovery of a new disease entity in 2017, termed mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy or MOGHE, in the frontal lobe of young children with intractable seizures. In 2021, a brain somatic missense mutation of the galactose transporter SLC35A2 leading to altered glycosylation of lipoproteins in the Golgi apparatus was detected in 50 % of MOGHE samples. In 2023, the first clinical trial evaluated galactose supplementation in patients with histopathologically confirmed MOGHE carrying brain somatic SLC35A2 mutations that were not seizure free after surgery. The promising results of this pilot trial are an example of personalized medicine in the arena of epileptology. Besides this, neuropathological studies of epilepsy samples have revealed many other fascinating results for the main disease categories in focal epilepsies, such as the first deep-learning based classifier for Focal Cortical Dysplasia, or the genomic landscape of cortical malformations showing new candidate genes such as PTPN11, which is associated with ganglioglioma and adverse clinical outcome. This update will also ask why common pathogenic variants accumulate in certain brain regions, e.g., MTOR in the frontal lobe, and BRAF in the temporal lobe. Finally, I will highlight the ongoing discussion addressing commonalities between temporal lobe epilepsy and Alzheimer's disease, the impact of adult neurogenesis and gliogenesis for the initiation and progression of temporal lobe seizures in the human brain as well as the immunopathogenesis of glutamic acid decarboxylase antibody associated temporal lobe epilepsy as a meaningful disease entity. This review will update the reader on some of these fascinating publications from 2022 and 2023 which were selected carefully, yet subjectively, by the author.
Collapse
Affiliation(s)
- Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Germany
| |
Collapse
|
35
|
Johannesen KM, Tümer Z, Weckhuysen S, Barakat TS, Bayat A. Solving the unsolved genetic epilepsies: Current and future perspectives. Epilepsia 2023; 64:3143-3154. [PMID: 37750451 DOI: 10.1111/epi.17780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
Many patients with epilepsy undergo exome or genome sequencing as part of a diagnostic workup; however, many remain genetically unsolved. There are various factors that account for negative results in exome/genome sequencing for patients with epilepsy: (1) the underlying cause is not genetic; (2) there is a complex polygenic explanation; (3) the illness is monogenic but the causative gene remains to be linked to a human disorder; (4) family segregation with reduced penetrance; (5) somatic mosaicism or the complexity of, for example, a structural rearrangement; or (6) limited knowledge or diagnostic tools that hinder the proper classification of a variant, resulting in its designation as a variant of unknown significance. The objective of this review is to outline some of the diagnostic options that lie beyond the exome/genome, and that might become clinically relevant within the foreseeable future. These options include: (1) re-analysis of older exome/genome data as knowledge increases or symptoms change; (2) looking for somatic mosaicism or long-read sequencing to detect low-complexity repeat variants or specific structural variants missed by traditional exome/genome sequencing; (3) exploration of the non-coding genome including disruption of topologically associated domains, long range non-coding RNA, or other regulatory elements; and finally (4) transcriptomics, DNA methylation signatures, and metabolomics as complementary diagnostic methods that may be used in the assessment of variants of unknown significance. Some of these tools are currently not integrated into standard diagnostic workup. However, it is reasonable to expect that they will become increasingly available and improve current diagnostic capabilities, thereby enabling precision diagnosis in patients who are currently undiagnosed.
Collapse
Affiliation(s)
- Katrine M Johannesen
- Department of Genetics, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Center, Dianalund, Denmark
| | - Zeynep Tümer
- Department of Genetics, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Weckhuysen
- Applied and Translational Neurogenomics Group, VIB Centre for Molecular Neurology, Antwerp, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
- Department of Neurology, University Hospital Antwerp, Antwerp, Belgium
- μNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Allan Bayat
- Department of Epilepsy Genetics and Personalized Medicine, The Danish Epilepsy Center, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Elziny S, Sran S, Yoon H, Corrigan RR, Page J, Ringland A, Lanier A, Lapidus S, Foreman J, Heinzen EL, Iffland P, Crino PB, Bedrosian TA. Loss of Slc35a2 alters development of the mouse cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569243. [PMID: 38077069 PMCID: PMC10705455 DOI: 10.1101/2023.11.29.569243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Brain somatic variants in SLC35A2 are associated with clinically drug-resistant epilepsy and developmental brain malformations, including mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE). SLC35A2 encodes a uridine diphosphate galactose translocator that is essential for protein glycosylation; however, the neurodevelopmental mechanisms by which SLC35A2 disruption leads to clinical and histopathological features remain unspecified. We hypothesized that focal knockout (KO) or knockdown (KD) of Slc35a2 in the developing mouse cortex would disrupt cerebral cortical development through altered neuronal migration and cause changes in network excitability. We used in utero electroporation (IUE) to introduce CRISPR/Cas9 and targeted guide RNAs or short-hairpin RNAs to achieve Slc35a2 KO or KD, respectively, during early corticogenesis. Following Slc35a2 KO or KD, we observed disrupted radial migration of transfected neurons evidenced by heterotopic cells located in lower cortical layers and in the sub-cortical white matter. Slc35a2 KO in neurons did not induce changes in oligodendrocyte number, suggesting that the oligodendroglial hyperplasia observed in MOGHE originates from distinct cell autonomous effects. Spontaneous seizures were not observed, but intracranial EEG recordings after focal KO showed a reduced seizure threshold following pentylenetetrazol injection. These results demonstrate that Slc35a2 KO or KD in vivo disrupts corticogenesis through altered neuronal migration.
Collapse
Affiliation(s)
- Soad Elziny
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sahibjot Sran
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Hyojung Yoon
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Rachel R. Corrigan
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - John Page
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amanda Ringland
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Anna Lanier
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Sara Lapidus
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James Foreman
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erin L. Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy and Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Philip Iffland
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter B. Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tracy A. Bedrosian
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
37
|
Straka B, Splitkova B, Vlckova M, Tesner P, Rezacova H, Krskova L, Koblizek M, Kyncl M, Maulisova A, Bukacova K, Uhrova-Meszarosova A, Musilova A, Kudr M, Ebel M, Belohlavkova A, Jahodova A, Liby P, Tichy M, Jezdik P, Zamecnik J, Aronica E, Krsek P. Genetic testing in children enrolled in epilepsy surgery program. A real-life study. Eur J Paediatr Neurol 2023; 47:80-87. [PMID: 37812946 DOI: 10.1016/j.ejpn.2023.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVE Although genetic causes of drug-resistant focal epilepsy and selected focal malformations of cortical development (MCD) have been described, a limited number of studies comprehensively analysed genetic diagnoses in patients undergoing pre-surgical evaluation, their outcomes and the effect of genetic diagnosis on surgical strategy. METHODS We analysed a prospective cohort of children enrolled in epilepsy surgery program over January 2018-July 2022. The majority of patients underwent germline and/or somatic genetic testing. We searched for predictors of surgical outcome and positive result of germline genetic testing. RESULTS Ninety-five patients were enrolled in epilepsy surgery program and 64 underwent resective epilepsy surgery. We ascertained germline genetic diagnosis in 13/74 patients having underwent germline gene testing (pathogenic or likely pathogenic variants in CHRNA4, NPRL3, DEPDC5, FGF12, GRIA2, SZT2, STXBP1) and identified three copy number variants. Thirty-five patients underwent somatic gene testing; we detected 10 pathogenic or likely pathogenic variants in genes SLC35A2, PTEN, MTOR, DEPDC5, NPRL3. Germline genetic diagnosis was significantly associated with the diagnosis of focal epilepsy with unknown seizure onset. SIGNIFICANCE Germline and somatic gene testing can ascertain a definite genetic diagnosis in a significant subgroup of patients in epilepsy surgery programs. Diagnosis of focal genetic epilepsy may tip the scales against the decision to proceed with invasive EEG study or surgical resection; however, selected patients with genetic focal epilepsies associated with MCD may benefit from resective epilepsy surgery and therefore, a genetic diagnosis does not disqualify patients from presurgical evaluation and epilepsy surgery.
Collapse
Affiliation(s)
- Barbora Straka
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Barbora Splitkova
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Marketa Vlckova
- Department of Biology and Medical Genetics, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Pavel Tesner
- Department of Biology and Medical Genetics, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Hana Rezacova
- Department of Biology and Medical Genetics, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Lenka Krskova
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Miroslav Koblizek
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Martin Kyncl
- Department of Radiology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Alice Maulisova
- Department of Clinical Psychology, Motol University Hospital, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Katerina Bukacova
- Department of Clinical Psychology, Motol University Hospital, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Anna Uhrova-Meszarosova
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Alena Musilova
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Martin Kudr
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Matyas Ebel
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Anezka Belohlavkova
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Alena Jahodova
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Petr Liby
- Department of Neurosurgery, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Michal Tichy
- Department of Neurosurgery, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Petr Jezdik
- Faculty of Electrical Engineering, Department of Circuit Theory, Czech Technical University in Prague, Technicka 2, Praha 6, 166 27, Czech Republic.
| | - Josef Zamecnik
- Department of Pathology and Molecular Medicine, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| | - Eleonora Aronica
- Amsterdam UMC Location University of Amsterdam, Department of Neuropathology, Amsterdam Neuroscience, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands.
| | - Pavel Krsek
- Department of Paediatric Neurology, Motol Epilepsy Center, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Full Member of the ERN EpiCARE, V Uvalu 84, 15006, Prague, Czech Republic.
| |
Collapse
|
38
|
Cao H, Wei P, Huang Y, Wang N, Guo LA, Fan X, Wang Z, Ren L, Piao Y, Lu J, Shan Y, He X, Zhao G. The alteration of cortical microstructure similarity in drug-resistant epilepsy correlated with mTOR pathway genes. EBioMedicine 2023; 97:104847. [PMID: 39492369 PMCID: PMC10628344 DOI: 10.1016/j.ebiom.2023.104847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Drug-resistant epilepsy (DRE) is associated with distributed laminar disruptions due to cytoarchitectonic pathologies, which may be characterized by multimodal MRI approaches such as morphometric similarity networks (MSNs). However, the genetic and histological underpinning of MSN alterations in DRE remains poorly understood, hampering its clinical application. METHODS We enrolled 60 patients with DRE and 23 controls, acquiring T1 and diffusion spectrum imaging data with a 3.0T GE SIGNA Premier scanner. Morphometric similarity networks (MSNs) were constructed and analyzed to identify microstructure similarity differences between patients and controls. Subsequently, patient-specific MSN alteration patterns were associated with gene expression using the GAMBA tool, and layer-specific neuronal signature mapping were also applied. During these analyses, sex and age were adjusted as covariates and multiple comparisons corrections were applied when appropriate. FINDINGS We observed widespread MSN changes in patients with DRE and identified five distinct MSN alteration patterns. Major patterns presented pattern-specific associations with expressions of epilepsy-related genes, particularly involving the mTOR pathway. Histological analysis confirmed the presence of cortical microstructure changes in areas with MSN alterations and revealed cellular abnormalities matching the aforementioned genetic risks. INTERPRETATION Our findings highlight the potential of quantifying laminar-related microstructure integrity using MSN to uncover the cytoarchitectonic changes in the pathophysiology of DRE. This approach may facilitate the identification of genetic and histological underpinnings of MSN alterations in DRE, ultimately aiding in the development of targeted therapeutic strategies. FUNDINGS The National Natural Science Foundation of China, the Ministry of Science and Technology of the People's Republic of China, and the Beijing Municipal Health Commission.
Collapse
Affiliation(s)
- Hang Cao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Penghu Wei
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Clinical Research Center for Epilepsy, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Beijing Municipal Geriatric Medical Research Center, 45 Changchun St, Beijing, 100053, China
| | - Yuda Huang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Ningrui Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Lin-Ai Guo
- Department of Pathology, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Xiaotong Fan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Zhenming Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Liankun Ren
- Department of Neurology, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China
| | - Yueshan Piao
- Department of Pathology, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Clinical Research Center for Epilepsy, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; National Medical Center for Neurological Diseases, 45 Changchun St, Beijing, 100053, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, 45 Changchun St, Beijing, 100053, China
| | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China.
| | - Xiaosong He
- Department of Psychology, University of Science and Technology of China, No 96 Jinzhai Rd, Hefei, 230026, China.
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Clinical Research Center for Epilepsy, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China; Beijing Municipal Geriatric Medical Research Center, 45 Changchun St, Beijing, 100053, China.
| |
Collapse
|
39
|
Elziny S, Crino PB, Winawer M. SLC35A2 somatic variants in drug resistant epilepsy: FCD and MOGHE. Neurobiol Dis 2023; 187:106299. [PMID: 37739137 PMCID: PMC10994450 DOI: 10.1016/j.nbd.2023.106299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
De novo somatic (post-zygotic) gene mutations affecting neuroglial progenitor cell types in embryonic cerebral cortex are increasingly identified in patients with drug resistant epilepsy (DRE) associated with malformations of cortical development, in particular, focal cortical dysplasias (FCD). Somatic variants in at least 16 genes have been linked to FCD type II, all encoding components of the mechanistic target of rapamycin (mTOR) pathway. FCD type II is characterized histopathologically by cytomegalic dysmorphic neurons and balloon cells. In contrast, the molecular pathogenesis of FCD I subtypes is less well understood, and histological features are characterized by alterations in columnar or laminar organization without cytomegalic dysmorphic neurons or balloon cells. In 2018, we reported somatic mutations in Solute Carrier Family 35 member A2 (SLC35A2) linked to DRE underlying FCD type I and subsequently to a new histopathological phenotype: excess oligodendrocytes and heterotopic neurons in subcortical white matter known as MOGHE (mild malformation of cortical development with oligodendroglial hyperplasia). These discoveries opened the door to studies linking somatic mutations to FCD. In this review, we discuss the biology of SLC35A2 somatic mutations in epilepsy in FCD and MOGHE, and insights into SLC35A2 epilepsy pathogenesis, describing progress to date and critical areas for investigation.
Collapse
Affiliation(s)
- Soad Elziny
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Melodie Winawer
- Department of Neurology, Columbia University, New York, NY, United States of America.
| |
Collapse
|
40
|
Nishioka M, Takayama J, Sakai N, Kazuno AA, Ishiwata M, Ueda J, Hayama T, Fujii K, Someya T, Kuriyama S, Tamiya G, Takata A, Kato T. Deep exome sequencing identifies enrichment of deleterious mosaic variants in neurodevelopmental disorder genes and mitochondrial tRNA regions in bipolar disorder. Mol Psychiatry 2023; 28:4294-4306. [PMID: 37248276 PMCID: PMC10827672 DOI: 10.1038/s41380-023-02096-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Bipolar disorder (BD) is a global medical issue, afflicting around 1% of the population with manic and depressive episodes. Despite various genetic studies, the genetic architecture and pathogenesis of BD have not been fully resolved. Besides germline variants, postzygotic mosaic variants are proposed as new candidate mechanisms contributing to BD. Here, we performed extensive deep exome sequencing (DES, ~300×) and validation experiments to investigate the roles of mosaic variants in BD with 235 BD cases (194 probands of trios and 41 single cases) and 39 controls. We found an enrichment of developmental disorder (DD) genes in the genes hit by deleterious mosaic variants in BD (P = 0.000552), including a ClinVar-registered pathogenic variant in ARID2. An enrichment of deleterious mosaic variants was also observed for autism spectrum disorder (ASD) genes (P = 0.000428). The proteins coded by the DD/ASD genes with non-synonymous mosaic variants in BD form more protein-protein interaction than expected, suggesting molecular mechanisms shared with DD/ASD but restricted to a subset of cells in BD. We also found significant enrichment of mitochondrial heteroplasmic variants, another class of mosaic variants, in mitochondrial tRNA genes in BD (P = 0.0102). Among them, recurrent m.3243 A > G variants known as causal for mitochondrial diseases were found in two unrelated BD probands with allele fractions of 5-12%, lower than in mitochondrial diseases. Despite the limitation of using peripheral tissues, our DES investigation supports the possible contribution of deleterious mosaic variants in the nuclear genome responsible for severer phenotypes, such as DD/ASD, to the risk of BD and further demonstrates that the same paradigm can be applied to the mitochondrial genome. These results, as well as the enrichment of heteroplasmic mitochondrial tRNA variants in BD, add a new piece to the understanding of the genetic architecture of BD and provide general insights into the pathological roles of mosaic variants in human diseases.
Collapse
Affiliation(s)
- Masaki Nishioka
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Department of Molecular Pathology of Mood Disorders, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Jun Takayama
- Department of AI and Innovative Medicine, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8573, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Naomi Sakai
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - An-A Kazuno
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Mizuho Ishiwata
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Junko Ueda
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takashi Hayama
- Yokohama Mental Clinic Totsuka, 494-8 Kamikurata-cho, Totsuka-ku, Yokohama, 244-0816, Japan
| | - Kumiko Fujii
- Department of Psychiatry, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, 757 Asahimachidori-ichibancho, Chuo-ku, Niigata, 951-8510, Japan
| | - Shinichi Kuriyama
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8573, Japan
- Department of Molecular Epidemiology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Gen Tamiya
- Department of AI and Innovative Medicine, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-Ku, Sendai, Miyagi, 980-8573, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Atsushi Takata
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
- Laboratory for Molecular Pathology of Psychiatric Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
| | - Tadafumi Kato
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Department of Molecular Pathology of Mood Disorders, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-Ku, Tokyo, 113-8421, Japan.
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
41
|
Liu X, Zhu Y, Liu Q, Zhang S, Wu P, Sun Y, Zhang J, Wang R, Ji T, Wang S, Liu X, Jiang Y, Cai L, Wu Y. Clinical characteristics and surgical outcomes in children with mild malformation of cortical development and oligodendroglial hyperplasia in epilepsy. Epilepsia Open 2023; 8:898-911. [PMID: 37144544 PMCID: PMC10472402 DOI: 10.1002/epi4.12755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023] Open
Abstract
OBJECTIVE Mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE) is a new and rare histopathological entity of cortical developmental malformations. The clinical characteristics of MOGHE remain challenging. METHODS Children with histologically confirmed MOGHE were retrospectively studied. The clinical findings, electroclinical and imaging features, and postoperative outcomes were analyzed, and previously published studies were reviewed up to June 2022. RESULTS Thirty-seven children were included in our cohort. Clinical characteristics included early onset in infancy (94.6% before 3 years), multiple seizure types, and moderate or severe delay. Epileptic spasm is the most common seizure type and initial manifestation. The lesions were mainly multilobar (59.5% multiple lobes and 8.1% hemispheres), and predominance in the frontal lobe was observed. The interictal EEG pattern was circumscribed or widespread. The prominent MRI characteristics were cortical thickening, cortical/subcortical hyperintense T2/FLAIR signal, and blurring at the GM and WM transition. Among the 21 children followed up for more than 1 year after surgery, 76.2% were seizure-free. Preoperative interictal circumscribed discharges and larger resections were significantly associated with a good postoperative outcome. The clinical features of 113 patients in the reviewed studies were similar to those we reported, but the lesions were mainly unilobar (73.5%) and Engel I was achieved in only 54.2% after surgery. SIGNIFICANCE Distinct clinical characteristics in MOGHE, especially age at onset, epileptic spasm, and age-related MRI characteristics, can help in early diagnosis. Preoperative interictal discharge and surgical strategy may be predictors of postoperative outcomes.
Collapse
Affiliation(s)
- Xianyu Liu
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Ying Zhu
- Department of RadiologyPeking University First HospitalBeijingChina
| | - Qingzhu Liu
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Shuang Zhang
- Department of PathologyPeking University First HospitalBeijingChina
| | - Pengxia Wu
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Yu Sun
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Jie Zhang
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Ruofan Wang
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Taoyun Ji
- Department of PediatricsPeking University First HospitalBeijingChina
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Shuang Wang
- Department of PediatricsPeking University First HospitalBeijingChina
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Xiaoyan Liu
- Department of PediatricsPeking University First HospitalBeijingChina
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Yuwu Jiang
- Department of PediatricsPeking University First HospitalBeijingChina
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Lixin Cai
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| | - Ye Wu
- Department of PediatricsPeking University First HospitalBeijingChina
- Pediatric Epilepsy CenterPeking University First HospitalBeijingChina
| |
Collapse
|
42
|
Sun X, Yuan Z, Zhang L, Ren M, Yang J, Xu Y, Hao J. Comprehensive Analysis of SLC35A2 in Pan-Cancer and Validation of Its Role in Breast Cancer. J Inflamm Res 2023; 16:3381-3398. [PMID: 37593196 PMCID: PMC10427759 DOI: 10.2147/jir.s419994] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023] Open
Abstract
Purpose Elucidation of the oncogenic role of SLC35A2 in human tumors and the potential function and clinical significance in breast cancer. Methods Pan-cancer analysis was performed via various bioinformatics tools to explain the pathogenic role of SLC35A2. A prognostic nomogram was also developed based on the SLC35A2 expression and clinicopathological characteristics in breast cancer patients. In addition, the role of SLC35A2 was validated in breast cancer by in vivo and in vitro experiments. Results SLC35A2 expression is increased in 27 tumor types, and its high expression is substantially correlated with poor prognosis in patients with a variety of cancers. Receiver operating characteristic (ROC) curves showed that SLC35A2 expression levels could accurately distinguish most tumor tissues from normal tissues. High SLC35A2 expression was linked to increased immune infiltration in myeloid-derived suppressor cells (MDSC), as well as immune checkpoints, ferroptosis-related genes, tumor mutational burden (TMB), and microsatellite instability (MSI). SLC35A2 may be involved in tumorigenesis by regulating the glycosylation process. Furthermore, multivariate Cox analysis showed that SLC35A2 was an independent prognostic factor for breast cancer. And the nomogram model had good predictive accuracy for the prognosis of breast cancer patients. Meanwhile, cellular experiments demonstrated that knockdown of SLC35A2 could significantly inhibit the proliferation, migration and invasion of breast cancer cells, while increasing the protein level of E-cadherin and decreasing N-cadherin. A nude mouse xenograft model showed that inhibition of SLA35A2 expression could significantly inhibit tumor growth. Conclusion SLC35A2 has good diagnostic and prognostic values in multiple cancers and is closely related to tumor immune infiltration. In addition, SLA35A2 as an oncogene in breast cancer may be involved in the progression of epithelial mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Xiaonan Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Zhichao Yuan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Lu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Min Ren
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Jing Yang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Yidan Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Jiqing Hao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| |
Collapse
|
43
|
Wang DD, Katoch M, Jabari S, Blumcke I, Blumenthal DB, Lu DH, Coras R, Wang YJ, Shi J, Zhou WJ, Kobow K, Piao YS. The specific DNA methylation landscape in focal cortical dysplasia ILAE type 3D. Acta Neuropathol Commun 2023; 11:129. [PMID: 37559109 PMCID: PMC10410964 DOI: 10.1186/s40478-023-01618-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/09/2023] [Indexed: 08/11/2023] Open
Abstract
Focal Cortical Dysplasia (FCD) is a frequent cause of drug-resistant focal epilepsy in children and young adults. The international FCD classifications of 2011 and 2022 have identified several clinico-pathological subtypes, either occurring isolated, i.e., FCD ILAE Type 1 or 2, or in association with a principal cortical lesion, i.e., FCD Type 3. Here, we addressed the DNA methylation signature of a previously described new subtype of FCD 3D occurring in the occipital lobe of very young children and microscopically defined by neuronal cell loss in cortical layer 4. We studied the DNA methylation profile using 850 K BeadChip arrays in a retrospective cohort of 104 patients with FCD 1 A, 2 A, 2B, 3D, TLE without FCD, and 16 postmortem specimens without neurological disorders as controls, operated in China or Germany. DNA was extracted from formalin-fixed paraffin-embedded tissue blocks with microscopically confirmed lesions, and DNA methylation profiles were bioinformatically analyzed with a recently developed deep learning algorithm. Our results revealed a distinct position of FCD 3D in the DNA methylation map of common FCD subtypes, also different from non-FCD epilepsy surgery controls or non-epileptic postmortem controls. Within the FCD 3D cohort, the DNA methylation signature separated three histopathology subtypes, i.e., glial scarring around porencephalic cysts, loss of layer 4, and Rasmussen encephalitis. Differential methylation in FCD 3D with loss of layer 4 mapped explicitly to biological pathways related to neurodegeneration, biogenesis of the extracellular matrix (ECM) components, axon guidance, and regulation of the actin cytoskeleton. Our data suggest that DNA methylation signatures in cortical malformations are not only of diagnostic value but also phenotypically relevant, providing the molecular underpinnings of structural and histopathological features associated with epilepsy. Further studies will be necessary to confirm these results and clarify their functional relevance and epileptogenic potential in these difficult-to-treat children.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Xicheng District, Beijing, 100053, China
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, 100053, China
- National Center for Neurological Disorders, Beijing, 100053, China
| | - Mitali Katoch
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Samir Jabari
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ingmar Blumcke
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - David B Blumenthal
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - De-Hong Lu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Xicheng District, Beijing, 100053, China
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, 100053, China
- National Center for Neurological Disorders, Beijing, 100053, China
| | - Roland Coras
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yu-Jiao Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Xicheng District, Beijing, 100053, China
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, 100053, China
- National Center for Neurological Disorders, Beijing, 100053, China
| | - Jie Shi
- Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, 100049, China
| | - Wen-Jing Zhou
- Department of Neurosurgery, Tsinghua University Yuquan Hospital, Beijing, 100049, China
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Yue-Shan Piao
- Department of Pathology, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Xicheng District, Beijing, 100053, China.
- Clinical Research Center for Epilepsy, Capital Medical University, Beijing, 100053, China.
- National Center for Neurological Disorders, Beijing, 100053, China.
| |
Collapse
|
44
|
Boßelmann CM, Leu C, Lal D. Technological and computational approaches to detect somatic mosaicism in epilepsy. Neurobiol Dis 2023:106208. [PMID: 37343892 DOI: 10.1016/j.nbd.2023.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023] Open
Abstract
Lesional epilepsy is a common and severe disease commonly associated with malformations of cortical development, including focal cortical dysplasia and hemimegalencephaly. Recent advances in sequencing and variant calling technologies have identified several genetic causes, including both short/single nucleotide and structural somatic variation. In this review, we aim to provide a comprehensive overview of the methodological advancements in this field while highlighting the unresolved technological and computational challenges that persist, including ultra-low variant allele fractions in bulk tissue, low availability of paired control samples, spatial variability of mutational burden within the lesion, and the issue of false-positive calls and validation procedures. Information from genetic testing in focal epilepsy may be integrated into clinical care to inform histopathological diagnosis, postoperative prognosis, and candidate precision therapies.
Collapse
Affiliation(s)
- Christian M Boßelmann
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Costin Leu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London, UK.
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T., Cambridge, MA, USA; Cologne Center for Genomics (CCG), University of Cologne, Cologne, DE, USA
| |
Collapse
|
45
|
D'Gama AM, Poduri A. Brain somatic mosaicism in epilepsy: Bringing results back to the clinic. Neurobiol Dis 2023; 181:106104. [PMID: 36972791 DOI: 10.1016/j.nbd.2023.106104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/28/2023] Open
Abstract
Over the past decade, there has been tremendous progress in understanding brain somatic mosaicism in epilepsy in the research setting. Access to resected brain tissue samples from patients with medically refractory epilepsy undergoing epilepsy surgery has been key to making these discoveries. In this review, we discuss the gap between making discoveries in the research setting and bringing results back to the clinical setting. Current clinical genetic testing mainly uses clinically accessible tissue samples, like blood and saliva, and can detect inherited and de novo germline variants and potentially non-brain-limited mosaic variants that have resulted from post-zygotic mutation (also called "somatic mutations"). Methods developed in the research setting to detect brain-limited mosaic variants using brain tissue samples need to be further translated and validated in the clinical setting, which will allow post-resection brain tissue genetic diagnoses. However, obtaining a genetic diagnosis after surgery for refractory focal epilepsy, when brain tissue samples are available, is arguably "too late" to guide precision management. Emerging methods using cerebrospinal fluid (CSF) and stereoelectroencephalography (SEEG) electrodes hold promise for establishing genetic diagnoses pre-resection without the need for actual brain tissue. In parallel, development of curation rules for interpreting the pathogenicity of mosaic variants, which have unique considerations compared to germline variants, will assist clinically accredited laboratories and epilepsy geneticists in making genetic diagnoses. Returning results of brain-limited mosaic variants to patients and their families will end their diagnostic odyssey and advance epilepsy precision management.
Collapse
Affiliation(s)
- Alissa M D'Gama
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA 02115, United States of America; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States of America; Epilepsy Genetics Program, Boston Children's Hospital, Boston, MA 02115, United States of America
| | - Annapurna Poduri
- Epilepsy Genetics Program, Boston Children's Hospital, Boston, MA 02115, United States of America; Department of Neurology, Boston Children's Hospital, Boston, MA 02115, United States of America; Department of Neurology, Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
46
|
Bizzotto S. The human brain through the lens of somatic mosaicism. Front Neurosci 2023; 17:1172469. [PMID: 37250426 PMCID: PMC10213359 DOI: 10.3389/fnins.2023.1172469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Every cell in the human brain possesses a unique genome that is the product of the accumulation of somatic mutations starting from the first postzygotic cell division and continuing throughout life. Somatic mosaicism in the human brain has been the focus of several recent efforts that took advantage of key technological innovations to start elucidating brain development, aging and disease directly in human tissue. On one side, somatic mutation occurring in progenitor cells has been used as a natural barcoding system to address cell phylogenies of clone formation and cell segregation in the brain lineage. On the other side, analyses of mutation rates and patterns in the genome of brain cells have revealed mechanisms of brain aging and disorder predisposition. In addition to the study of somatic mosaicism in the normal human brain, the contribution of somatic mutation has been investigated in both developmental neuropsychiatric and neurodegenerative disorders. This review starts with a methodological perspective on the study of somatic mosaicism to then cover the most recent findings in brain development and aging, and ends with the role of somatic mutations in brain disease. Thus, this review underlies what we have learned and what is still possible to discover by looking at somatic mosaicism in the brain genome.
Collapse
|
47
|
Metodiev D, Minkin K, Ruseva M, Ganeva R, Parvanov D, Nachev S. Pathomorphological Diagnostic Criteria for Focal Cortical Dysplasias and Other Common Epileptogenic Lesions—Review of the Literature. Diagnostics (Basel) 2023; 13:diagnostics13071311. [PMID: 37046529 PMCID: PMC10092959 DOI: 10.3390/diagnostics13071311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Focal cortical dysplasia (FCD) represents a heterogeneous group of morphological changes in the brain tissue that can predispose the development of pharmacoresistant epilepsy (recurring, unprovoked seizures which cannot be managed with medications). This group of neurological disorders affects not only the cerebral cortex but also the subjacent white matter. This work reviews the literature describing the morphological substrate of pharmacoresistant epilepsy. All illustrations presented in this study are obtained from brain biopsies from refractory epilepsy patients investigated by the authors. Regarding classification, there are three main FCD types, all of which involve cortical dyslamination. The 2022 revision of the International League Against Epilepsy (ILAE) FCD classification includes new histologically defined pathological entities: mild malformation of cortical development (mMCD), mild malformation of cortical development with oligodendroglial hyperplasia in frontal lobe epilepsy (MOGHE), and “no FCD on histopathology”. Although the pathomorphological characteristics of the various forms of focal cortical dysplasias are well known, their aetiologic and pathogenetic features remain elusive. The identification of genetic variants in FCD opens an avenue for novel treatment strategies, which are of particular utility in cases where total resection of the epileptogenic area is impossible.
Collapse
|
48
|
Sahly AN, Whitney R, Costain G, Chau V, Otsubo H, Ochi A, Donner EJ, Cunningham J, Jones KC, Widjaja E, Ibrahim GM, Jain P. Epilepsy surgery outcomes in patients with GATOR1 gene complex variants: Report of new cases and review of literature. Seizure 2023; 107:13-20. [PMID: 36931189 DOI: 10.1016/j.seizure.2023.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/19/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023] Open
Abstract
AIM To report seizure outcomes in children with GATOR1 gene complex disorders who underwent epilepsy surgery and perform a systematic literature search to study the available evidence. METHODS The records of children with pathogenic/likely pathogenic variants in GATOR1 gene complex who underwent epilepsy surgery were reviewed. Clinical, radiological, neurophysiological, and histological data were extracted/summarized. The systematic review included all case series/reports and observational studies reporting on children or adults with genetic (germline or somatic) variants in the GATOR1 complex genes (DEPDC5, NPRL2, NPRL3) with focal epilepsy with/without focal cortical dysplasia who underwent epilepsy surgery; seizure outcomes were analyzed. RESULTS Eight children with pathogenic/likely pathogenic variants in GATOR1 complex genes were included. All had drug-resistant epilepsy. Six children had significant neurodevelopmental delay. Epilepsy surgery was performed in all; clinical seizure freedom was noted in 4 children (50%). Systematic literature search identified 17 eligible articles; additional 30 cases with patient-level data were studied. Lesional MRI brain was seen in 80% cases. The pooled rate of seizure freedom following surgery was 60%; FCD IIa was the most encountered pathology. INTERPRETATION Epilepsy surgery may be effective in some children with GATOR1 complex gene variants. Seizure outcomes may be compromised by extensive epileptogenic zones.
Collapse
Affiliation(s)
- Ahmed N Sahly
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, Quebec, Canada; Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Gregory Costain
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Vann Chau
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hiroshi Otsubo
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ayako Ochi
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth J Donner
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jessie Cunningham
- Hospital Library and Archives, Learning Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kevin C Jones
- Division of Neurology, Department of Paediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Elysa Widjaja
- Department of Diagnostic Imaging, Hospital for Sick Children, Toronto, Ontario, Canada
| | - George M Ibrahim
- Division of Neurosurgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Puneet Jain
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Balestrini S, Barba C, Thom M, Guerrini R. Focal cortical dysplasia: a practical guide for neurologists. Pract Neurol 2023:pn-2022-003404. [PMID: 36823117 DOI: 10.1136/pn-2022-003404] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2023] [Indexed: 02/25/2023]
Abstract
Focal cortical dysplasia (FCD) is a malformation of cortical development characterised by disruption of cortical cytoarchitecture. Classification of FCDs subtypes has initially been based on correlation of the histopathology with relevant clinical, electroencephalographic and neuroimaging features. A recently proposed classification update recommends a multilayered, genotype-phenotype approach, integrating findings from histopathology, genetic analysis of resected tissue and presurgical MRI. FCDs are caused either by single somatic activating mutations in MTOR pathway genes or by double-hit inactivating mutations with a constitutional and a somatic loss-of-function mutation in repressors of the signalling pathway. Mild malformation with oligodendroglial hyperplasia in epilepsy is caused by somatic pathogenic SLC35A2 mutations. FCDs most often present with drug-resistant focal epilepsy or epileptic encephalopathy. Most patients respond to surgical treatment. The use of mechanistic target of rapamycin inhibitors may complement the surgical approach. Treatment approaches and outcomes have improved with advances in neuroimaging, neurophysiology and genetics, although predictors of treatment response have only been determined in part.
Collapse
Affiliation(s)
- Simona Balestrini
- Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital IRCCS, Florence, Italy .,University of Florence, Florence, Italy.,Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Carmen Barba
- Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital IRCCS, Florence, Italy.,University of Florence, Florence, Italy
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK
| | - Renzo Guerrini
- Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital IRCCS, Florence, Italy.,University of Florence, Florence, Italy
| |
Collapse
|
50
|
Barba C, Blumcke I, Winawer MR, Hartlieb T, Kang HC, Grisotto L, Chipaux M, Bien CG, Heřmanovská B, Porter BE, Lidov HGW, Cetica V, Woermann FG, Lopez-Rivera JA, Canoll PD, Mader I, D'Incerti L, Baldassari S, Yang E, Gaballa A, Vogel H, Straka B, Macconi L, Polster T, Grant GA, Krsková L, Shin HJ, Ko A, Crino PB, Krsek P, Lee JH, Lal D, Baulac S, Poduri A, Guerrini R. Clinical Features, Neuropathology, and Surgical Outcome in Patients With Refractory Epilepsy and Brain Somatic Variants in the SLC35A2 Gene. Neurology 2023; 100:e528-e542. [PMID: 36307217 PMCID: PMC9931085 DOI: 10.1212/wnl.0000000000201471] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/09/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The SLC35A2 gene, located at chromosome Xp11.23, encodes for a uridine diphosphate-galactose transporter. We describe clinical, genetic, neuroimaging, EEG, and histopathologic findings and assess possible predictors of postoperative seizure and cognitive outcome in 47 patients with refractory epilepsy and brain somatic SLC35A2 gene variants. METHODS This is a retrospective multicenter study where we performed a descriptive analysis and classical hypothesis testing. We included the variables of interest significantly associated with the outcomes in the generalized linear models. RESULTS Two main phenotypes were associated with brain somatic SLC35A2 variants: (1) early epileptic encephalopathy (EE, 39 patients) with epileptic spasms as the predominant seizure type and moderate to severe intellectual disability and (2) drug-resistant focal epilepsy (DR-FE, 8 patients) associated with normal/borderline cognitive function and specific neuropsychological deficits. Brain MRI was abnormal in all patients with EE and in 50% of those with DR-FE. Histopathology review identified mild malformation of cortical development with oligodendroglial hyperplasia in epilepsy in 44/47 patients and was inconclusive in 3. The 47 patients harbored 42 distinct mosaic SLC35A2 variants, including 14 (33.3%) missense, 13 (30.9%) frameshift, 10 (23.8%) nonsense, 4 (9.5%) in-frame deletions/duplications, and 1 (2.4%) splicing variant. Variant allele frequencies (VAFs) ranged from 1.4% to 52.6% (mean VAF: 17.3 ± 13.5). At last follow-up (35.5 ± 21.5 months), 30 patients (63.8%) were in Engel Class I, of which 26 (55.3%) were in Class IA. Cognitive performances remained unchanged in most patients after surgery. Regression analyses showed that the probability of achieving both Engel Class IA and Class I outcomes, adjusted by age at seizure onset, was lower when the duration of epilepsy increased and higher when postoperative EEG was normal or improved. Lower brain VAF was associated with improved postoperative cognitive outcome in the analysis of associations, but this finding was not confirmed in regression analyses. DISCUSSION Brain somatic SLC35A2 gene variants are associated with 2 main clinical phenotypes, EE and DR-FE, and a histopathologic diagnosis of MOGHE. Additional studies will be needed to delineate any possible correlation between specific genetic variants, mutational load in the epileptogenic tissue, and surgical outcomes.
Collapse
Affiliation(s)
- Carmen Barba
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Ingmar Blumcke
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Melodie R Winawer
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Till Hartlieb
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Hoon-Chul Kang
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Laura Grisotto
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Mathilde Chipaux
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Christian G Bien
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Barbora Heřmanovská
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Brenda E Porter
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Hart G W Lidov
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Valentina Cetica
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Friedrich G Woermann
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Javier A Lopez-Rivera
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Peter D Canoll
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Irina Mader
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Ludovico D'Incerti
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Sara Baldassari
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Edward Yang
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Ahmed Gaballa
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Hannes Vogel
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Barbora Straka
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Letizia Macconi
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Tilman Polster
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Gerald A Grant
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Lenka Krsková
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Hui Jin Shin
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Ara Ko
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Peter B Crino
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Pavel Krsek
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Jeong Ho Lee
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Dennis Lal
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Stéphanie Baulac
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Annapurna Poduri
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| | - Renzo Guerrini
- From the IRCCS Meyer Children's Hospital (C.B., V.C., L.D.I., L.M., R.G.), Florence, Italy; University of Florence (C.B., L.G., R.G.), Florence, Italy; University Hospital Erlangen (I.B.), Germany; Columbia University (M.R.W., P.D.C.), New York, NY; Neurorehabilitation and Epileptology (T.H., I.M.), Vogtareuth, Germany; PMU Salzburg (T.H.), Austria; Yonsei University College of Medicine (H.-C.K., H.J.S.), Seoul, Republic of Korea; Rothschild Foundation Hospital (M.C.), Paris, France; Krankenhaus Mara (C.G.B., F.G.W., A.G., T.P.), Bielefeld University, Medical School, Germany; Charles University (B.H., B.S., L.K., P.K.), 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic; Stanford University (B.E.P., H.V.), School of Medicine Stanford, CA; Boston Children's Hospital and Harvard Medical School (H.G.W.L., E.Y., A.P.), MA; Society of Epilepsy Research (F.G.W.), Bielefeld, Germany; Case Western Reserve University (J.A.L.-R.), OH; Cleveland Clinic (J.A.L.-R., D.L.), Cleveland, OH; Sorbonne University (Sara Baldassari, Stéphanie Baulac), Paris Brain Institute (ICM), INSERM, CNRS, AP-HP, Pitié-Salpêtrière Hospital, France; Lucile Packard Children's Hospital at Stanford University (G.A.G.), School of Medicine Stanford, CA; Korea Advanced Institute of Science and Technology (A.K., J.H.L.), Daejeon, South Korea; University of Maryland School of Medicine (P.B.C.), Baltimore, MD; and Broad Institute of Harvard and M.I.T (D.L.), Cambridge, MA
| |
Collapse
|