1
|
Surdi P, Trivisano M, De Dominicis A, Mercier M, Piscitello LM, Pavia GC, Calabrese C, Cappelletti S, Correale C, Mazzone L, Vigevano F, Specchio N. Unveiling the disease progression in developmental and epileptic encephalopathies: Insights from EEG and neuropsychology. Epilepsia 2024; 65:3279-3292. [PMID: 39287605 DOI: 10.1111/epi.18127] [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: 03/03/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Developmental and epileptic encephalopathies (DEEs) are neurological disorders characterized by developmental impairment and epilepsy. Our study aims to assess disease progression by comparing clinical findings, electroencephalography (EEG), and neuropsychological data from seizure onset to the last follow-up evaluation. METHODS We retrospectively reviewed patients with genetic DEEs who were followed-up at the epilepsy unit of Bambino Gesù Children's Hospital, Rome. We collected information regarding gender, family history, genetic variant, age at onset and at last follow-up, neurological examination, type of seizure, drug resistance, occurrence of status epilepticus, and movement and cognitive and behavioral disorders. We compared EEG background activity, epileptiform abnormalities, and cognitive functions between seizure onset and the last follow-up evaluation using the McNemar-Bowker test (α = 5%). RESULTS A total of 160 patients (94 female) were included. Genetic analysis revealed a spectrum of pathogenic variants, with SCN1A being the most prevalent (25%). The median age at seizure onset and at the last follow-up was 0.37 (interquartile range [IQR]: 0.09-0.75) and 8.54 years (IQR: 4.32-14.55), respectively. We documented a statistically significant difference in EEG background activity (p = .017) and cognitive impairment (p = .01) from seizure onset to the last follow-up evaluation. No significant differences were detected for epileptiform abnormalities (p = .2). In addition, high prevalence rates were observed for drug resistance (81.9%), movement disorders (60.6%), behavioral and autism spectrum disorders (45%), neurological deficits (31.3%), and occurrence of status epilepticus (23.1%). SIGNIFICANCE Our study provides evidence that a clinical progression may appear in genetic DEEs, manifesting as development or worsening of cognitive impairment and disruption of EEG background activity. These results highlight the challenging clinical course and the importance of early intervention and personalized care in the management of patients with DEEs.
Collapse
Affiliation(s)
- Paolo Surdi
- Child Neurology and Psychiatry Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Marina Trivisano
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Angela De Dominicis
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Mattia Mercier
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Ludovica Maria Piscitello
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Giusy Carfì Pavia
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Costanza Calabrese
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Simona Cappelletti
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Cinzia Correale
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| | - Luigi Mazzone
- Child Neurology and Psychiatry Unit, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Federico Vigevano
- Pediatric Neurorehabilitation Department, IRCCS San Raffaele, Rome, Italy
| | - Nicola Specchio
- Neurology, Epilepsy and Movement Disorders Unit, Bambino Gesù Children's Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Rome, Italy
| |
Collapse
|
2
|
Papatheodorou I, Stavrakaki S, Koukoulioti V, Spilioti M, Kimiskidis V. How Encephalopathy Impacts Language Ability: A Scoping Review of the Linguistic Abilities of Adults with Developmental and Epileptic Encephalopathy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1635. [PMID: 39459422 PMCID: PMC11509453 DOI: 10.3390/medicina60101635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Developmental and epileptic encephalopathy refers to a group of conditions where patients experience abnormal development due to various causes as well as frequent epileptiform discharges that ultimately contribute, in an independent and additive fashion, to cognitive and linguistic impairments. The language and cognition outcome of these patients in adulthood has been understudied. This paper aims to present a scoping review of linguistic abilities in adults with developmental and epileptic encephalopathy to determine the extent to which language outcomes in adulthood and their relation to cognitive outcomes have been studied. Design: Two online databases were searched and the methodological framework by Arksey & O'Malley (2005) was adopted. Results: Out of the 27 selected studies, only 13 exclusively examined adults, 15 were group studies, 5 were case studies and 7 were case series. A total of 9 out of the 15 group studies provided individual results for adults. Twenty-two studies included a follow-up examination. Twenty-three studies addressed the relationship between language and cognition. The selected studies indicate the presence of language impairments, which are nevertheless differentially manifested in the syndromes under investigation, whereas individual variability is also reported. Aspects of cognition seem to correlate with linguistic abilities. Conclusions: In sum, despite variability in linguistic abilities, language deficits constitute a significant aspect of the clinical profile of many adults with developmental and epileptic encephalopathy, a finding that should be taken into account for the treatment protocols of these individuals.
Collapse
Affiliation(s)
- Ioanna Papatheodorou
- School of Medicine, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (I.P.); (M.S.)
| | - Stavroula Stavrakaki
- Department of Italian Language and Literature, School of Philosophy, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece;
| | - Vasiliki Koukoulioti
- Department of German Language and Literature, School of Philosophy, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece;
| | - Martha Spilioti
- School of Medicine, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (I.P.); (M.S.)
| | - Vasileios Kimiskidis
- School of Medicine, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (I.P.); (M.S.)
| |
Collapse
|
3
|
Jaramillo-Jiménez E, Sandoval-Barrios J, Walsh FJ, Jaramillo-Jiménez MC, Echeverri-Sánchez JD, Rodríguez-Márquez IA, Barrientos-Montoya HD, Ascencio-Lancheros JL, Giraldo-Palacio JF, Sierra-Arrieta IM, Gómez-Duque DI, Pérez-López S, Bustamante MT. Epileptic encephalopathies secondary to hypothalamic hamartomas treated with radiosurgery: A case series. Epileptic Disord 2024; 26:581-590. [PMID: 38804823 DOI: 10.1002/epd2.20246] [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: 05/05/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE Hypothalamic hamartomas are congenital lesions that typically present with gelastic seizures, refractory epilepsy, neurodevelopmental delay, and severe cognitive impairment. Surgical procedures have been reported to be effective in removing the hamartomas, however, they are associated with significant morbidity. Therefore, it is not considered a safe therapeutic modality. Image-guided robotic radiosurgery (CyberKnife® Radiosurgery System) has been shown to provide good outcomes without lasting complications. METHODS This series of cases describes the clinical, radiological, radiotherapeutic, and postsurgical outcomes of five patients with epileptic encephalopathies secondary to hypothalamic hamartomas who were treated with CyberKnife®. RESULTS All patients exhibited refractory epilepsy with gelastic seizures and were unsuitable candidates for surgical resection The prescribed dose ranged between 16 and 25 Gy, delivered in a single fraction for four patients and five fractions for one patient while adhering strictly to visual pathway constraints. After radiosurgery, four patients maintained seizure control (one with an Engel class Ia, three with an Engel class 1d), and another presented sporadic, nondisabling gelastic seizures (with an Engel class IIa). After 24-26 months of follow-up, in three patients, their intelligence quotient scores increased. No complications were reported. SIGNIFICANCE This report suggests that Cyberknife may be a good option for treating hypothalamic hamartoma, particularly in cases where other noninvasive alternatives are unavailable. Nevertheless, additional studies are essential in order to evaluate the effectiveness of the technique in these cases.
Collapse
Affiliation(s)
| | | | - Fergus John Walsh
- School of Medicine, College of Health & Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Li PP, Zhou YY, Gao L, Lv JN, Xu SS, Zhao YW, Xu D, Huang R, Zhang X, Li P, Fu X, He Z. The de novo missense mutation F224S in GABRB2, identified in epileptic encephalopathy and developmental delay, impairs GABA AR function. Neuroscience 2024; 553:172-184. [PMID: 38964454 DOI: 10.1016/j.neuroscience.2024.06.029] [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: 03/01/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Genetic variants in genes encoding subunits of the γ-aminobutyric acid-A receptor (GABAAR) have been found to cause neurodevelopmental disorders and epileptic encephalopathy. In a patient with epilepsy and developmental delay, a de novo heterozygous missense mutation c.671 T > C (p.F224S) was discovered in the GABRB2 gene, which encodes the β2 subunit of GABAAR. Based on previous studies on GABRB2 variants, this new GABRB2 variant (F224S) would be pathogenic. To confirm and investigate the effects of this GABRB2 mutation on GABAAR channel function, we conducted transient expression experiments using GABAAR subunits in HEK293T cells. The GABAARs containing mutant β2 (F224S) subunit showed poor trafficking to the cell membrane, while the expression and distribution of the normal α1 and γ2 subunits were unaffected. Furthermore, the peak current amplitude of the GABAAR containing the β2 (F224S) subunit was significantly smaller compared to the wild type GABAAR. We propose that GABRB2 variant F224S is pathogenic and GABAARs containing this β2 mutant reduce response to GABA under physiological conditions, which could potentially disrupt the excitation/inhibition balance in the brain, leading to epilepsy.
Collapse
Affiliation(s)
- Ping-Ping Li
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yue-Yuan Zhou
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Li Gao
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jia-Nan Lv
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shi-Shi Xu
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yan-Wen Zhao
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Di Xu
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Ruoke Huang
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiong Zhang
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peijun Li
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Xiaoqin Fu
- Department of Geriatrics and Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zhiyong He
- Department of Pediatric Rehabilitation, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
5
|
Xu Y, Chen D, Liu L. Optimal dose of fenfluramine in adjuvant treatment of drug-resistant epilepsy: evidence from randomized controlled trials. Front Neurol 2024; 15:1371704. [PMID: 38590719 PMCID: PMC10999678 DOI: 10.3389/fneur.2024.1371704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024] Open
Abstract
Objective Several clinical trials have suggested that fenfluramine (FFA) is effective for the treatment of epilepsy in Dravet syndrome (DS) and Lennox-Gastaut syndrome (LGS). However, the exploration of its optimal target dose is ongoing. This study aimed to summarize the best evidence to inform this clinical issue. Materials and methods We searched PubMed, Embase (via Ovid), and Web of Science for relevant literature published before December 1st, 2023. Randomized, double-blind, placebo-controlled studies that evaluated the efficacy, safety, and tolerability of FFA in DS and LGS were identified and meta-analysis was performed according to doses. The study was registered with PROSPERO (CRD42023392454). Results Six hundred and twelve patients from four randomized controlled trials were enrolled. The results demonstrated that FFA at 0.2, 0.4, or 0.7 mg/kg/d showed significantly greater efficacy compared to placebo in terms of at least 50% reduction (p < 0.001, p < 0.001, p < 0.001) and at least 75% reduction (p < 0.001, p = 0.007, p < 0.001) in monthly seizure frequency from baseline. Moreover, significantly more patients receiving FFA than placebo were rated as much improved or very much improved in CGI-I by both caregivers/parents and investigators (p < 0.001). The most common treatment-emergent adverse events were decreased appetite, diarrhea, fatigue, and weight loss, with no valvular heart disease or pulmonary hypertension observed in any participant. For dose comparison, 0.7 mg/kg/d group presented higher efficacy on at least 75% reduction in seizure (p = 0.006) but not on at least 50% reduction. Weight loss (p = 0.002), decreased appetite (p = 0.04), and all-cause withdrawal (p = 0.036) were more common in 0.7 mg/kg/d group than 0.2 mg/kg/d. There was no statistical difference in other safety parameters between these two groups. Conclusion The higher range of the licensed dose achieves the optimal balance between efficacy, safety, and tolerability in patients with DS and LGS. Clinical trial registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023392454.
Collapse
Affiliation(s)
| | | | - Ling Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Çapan ÖY, Yapıcı Z, Özbil M, Çağlayan HS. Exome data of developmental and epileptic encephalopathy patients reveals de novo and inherited pathologic variants in epilepsy-associated genes. Seizure 2024; 116:51-64. [PMID: 37353388 DOI: 10.1016/j.seizure.2023.06.009] [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: 02/24/2023] [Revised: 06/04/2023] [Accepted: 06/10/2023] [Indexed: 06/25/2023] Open
Abstract
PURPOSE In Developmental and Epileptic Encephalopathies (DEEs), identifying the precise genetic factors guides the clinicians to apply the most appropriate treatment for the patient. Due to high locus heterogeneity, WES analysis is a promising approach for the genetic diagnosis of DEE. Therefore, the aim of the present study is to evaluate the utility of WES in the diagnosis and treatment of DEE patients. METHODS The exome data of 29 DEE patients were filtrated for destructive and missense mutations in 1896 epilepsy-related genes to detect the causative variants and examine the genotype-phenotype correlations. We performed Sanger sequencing with the available DNA samples to follow the co-segregation of the variants with the disease phenotype in the families. Also, the structural effects of p.Asn1053Ser, p.Pro120Ser and p.Glu1868Gly mutations on KCNMA1, NPC2, and SCN2A proteins, respectively, were evaluated by molecular dynamics (MD) and molecular docking simulations. RESULTS Out of 29, nine patients (31%) harbor pathological (P) or likely pathological (LP) mutations in SCN2A, KCNQ2, ATP1A2, KCNMA1, and MECP2 genes, and three patients have VUS variants (10%) in SCN1A and SCN2A genes. Sanger sequencing results indicated that three of the patients have de novo mutations while eight of them carry paternally and/or maternally inherited causative variants. MD and molecular docking simulations supported the destructive effects of the mutations on KCNMA1, NPC2, and SCN2A protein structures. CONCLUSION Herein we demonstrated the effectiveness of WES for DEE with high locus heterogeneity. Identification of the genetic etiology guided the clinicians to adjust the proper treatment for the patients.
Collapse
Affiliation(s)
- Özlem Yalçın Çapan
- Department of Medical Biology, Faculty of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey; Department of Molecular Biology and Genetics, İstanbul Arel University, İstanbul, Turkey.
| | - Zuhal Yapıcı
- Division of Child Neurology, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mehmet Özbil
- Institute of Biotechnology, Gebze Technical University, Kocaeli, Turkiye
| | - Hande S Çağlayan
- Department of Molecular Biology and Genetics, Boğaziçi University, İstanbul, Turkey (formerly)
| |
Collapse
|
7
|
Zhao X, He Z, Li Y, Yang X, Li B. Atypical absence seizures and gene variants: A gene-based review of etiology, electro-clinical features, and associated epilepsy syndrome. Epilepsy Behav 2024; 151:109636. [PMID: 38232560 DOI: 10.1016/j.yebeh.2024.109636] [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: 11/08/2023] [Revised: 01/01/2024] [Accepted: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Atypical absence seizures are generalized non-convulsive seizures that often occur in children with cognitive impairment. They are common in refractory epilepsy and have been recognized as one of the hallmarks of developmental epileptic encephalopathies. Notably, pathogenic variants associated with AAS, such as GABRG2, GABRG3, SLC6A1, CACNB4, SCN8A, and SYNGAP1, are also linked to developmental epileptic encephalopathies. Atypical absences differ from typical absences in that they are frequently drug-resistant and the prognosis is dependent on the etiology or related epileptic syndromes. To improve clinicians' understanding of atypical absences and provide novel perspectives for clinical treatment, we have reviewed the electro-clinical characteristics, etiologies, treatment, and prognosis of atypical absences, with a focus on the etiology of advancements in gene variants, shedding light on potential avenues for improved clinical management.
Collapse
Affiliation(s)
| | - Zimeng He
- Shandong University, Jinan, Shandong, China
| | - Yumei Li
- Shandong University, Jinan, Shandong, China
| | - Xiaofan Yang
- Shandong University, Jinan, Shandong, China; Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Baomin Li
- Shandong University, Jinan, Shandong, China; Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
8
|
Battaglia L, Scorrano G, Spiaggia R, Basile A, Palmucci S, Foti PV, Spatola C, Iacomino M, Marinangeli F, Francia E, Comisi F, Corsello A, Salpietro V, Vittori A, David E. Neuroimaging features of WOREE syndrome: a mini-review of the literature. Front Pediatr 2023; 11:1301166. [PMID: 38161429 PMCID: PMC10757851 DOI: 10.3389/fped.2023.1301166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
The WWOX gene encodes a 414-amino-acid protein composed of two N-terminal WW domains and a C-terminal short-chain dehydrogenase/reductase (SDR) domain. WWOX protein is highly conserved among species and mainly expressed in the cerebellum, cerebral cortex, brain stem, thyroid, hypophysis, and reproductive organs. It plays a crucial role in the biology of the central nervous system, and it is involved in neuronal development, migration, and proliferation. Biallelic pathogenic variants in WWOX have been associated with an early infantile epileptic encephalopathy known as WOREE syndrome. Both missense and null variants have been described in affected patients, leading to a reduction in protein function and stability. The most severe WOREE phenotypes have been related to biallelic null/null variants, associated with the complete loss of function of the protein. All affected patients showed brain anomalies on magnetic resonance imaging (MRI), suggesting the pivotal role of WWOX protein in brain homeostasis and developmental processes. We provided a literature review, exploring both the clinical and radiological spectrum related to WWOX pathogenic variants, described to date. We focused on neuroradiological findings to better delineate the WOREE phenotype with diagnostic and prognostic implications.
Collapse
Affiliation(s)
- Laura Battaglia
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Giovanna Scorrano
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rossana Spiaggia
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Antonio Basile
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Stefano Palmucci
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Pietro Valerio Foti
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Corrado Spatola
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| | - Michele Iacomino
- Unit of Medical Genetics, IRCCS Instituto Giannina Gaslini, Genoa, Italy
| | - Franco Marinangeli
- Department of Anesthesia, Critical Care and Pain Therapy, University of L’aquila, L’aquila, Italy
| | - Elisa Francia
- Department of Anesthesia and Critical Care, ARCO ROMA, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | | | | | - Vincenzo Salpietro
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Alessandro Vittori
- Department of Anesthesia and Critical Care, ARCO ROMA, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Emanuele David
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinic “G. Rodolico-San Marco”, Catania, Italy
| |
Collapse
|
9
|
Vidal E, Castro-Gutierrez E, Arisaca R, Paz-Valderrama A, Albiol-Pérez S. Serious Game for Fine Motor Control Rehabilitation for Children With Epileptic Encephalopathy: Development and Usability Study. JMIR Form Res 2023; 7:e50492. [PMID: 37788071 PMCID: PMC10582812 DOI: 10.2196/50492] [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: 07/03/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Epileptic encephalopathy (EE) is defined as the presence of frequent epileptiform activity that adversely impacts development, typically causing the slowing or regression of developmental skills, and is usually associated with frequent seizures. One of the main disturbances in EE is in the coordination of the upper extremities and hands. Traditional rehabilitation for this type of pathology focuses on the alleviation of gross or fine motor disability. In the last few years, the use of low-cost devices together with customized serious games has shown improvements in motor disorders and enrichments in activities of daily living. OBJECTIVE This study aims to explore the feasibility of a new serious game for improving fine motor control in children with EE. METHODS The participants were 4 children with EE (male: n=2, 50%; female: n=2, 50%) who were classified as belonging to level 1 in the Gross Motor Classification System. The children were tested over 10 sessions during the intervention period (before and after treatment). The clinical tests performed were the Bruininks-Oseretsky Test of Motor Proficiency, 2nd edition and Pittsburgh Rehabilitation Participation Scale. The subscales of the Bruininks-Oseretsky Test of Motor Proficiency, 2nd edition were fine motor precision, fine motor integration, manual dexterity, and upper-limb coordination. At the end of the first session, we used the User Satisfaction Evaluation Questionnaire to analyze user satisfaction. RESULTS The significance outcomes for a Student t test (1-tailed) were as follows: P=.009 for fine motor precision, P=.002 for fine motor integration, P=.56 for manual dexterity, and P=.99 for upper-limb coordination. The participation rate as measured using the Pittsburgh Rehabilitation Participation Scale was between good and very good, which means that, based on the therapist's evaluation, interest, independence, and motivation were achieved by each participant. The mean User Satisfaction Evaluation Questionnaire score was close to 30, which is the maximum value. CONCLUSIONS The results support the use of the proposed serious game as a complement in therapeutic sessions during the rehabilitation processes for children with EE. Significant improvements in fine motor control and activities of daily living revealed that the proposed serious game is beneficial for fine motor disorders of this pathology.
Collapse
Affiliation(s)
- Elizabeth Vidal
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
| | | | - Robert Arisaca
- Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru
| | | | - Sergio Albiol-Pérez
- Aragón Health Research Institute (IIS Aragón), Universidad de Zaragoza, Teruel, Spain
| |
Collapse
|
10
|
Muthaffar OY, Jan MMS, Alyazidi AS, Alotibi TK, Alsulami EA. Insight into Genetic Mutations of SZT2: Is It a Syndrome? Biomedicines 2023; 11:2402. [PMID: 37760843 PMCID: PMC10525120 DOI: 10.3390/biomedicines11092402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The seizure threshold 2 (SZT2) gene encodes a protein of unknown function, which is widely expressed, confers a low seizure threshold, and enhances epileptogenesis. It also comprises the KICSTOR protein complex, which inhibits the mTORC1 pathway. A pathogenic variant in the SZT2 gene could result in hyperactive mTORC1 signaling, which can lead to several neurological disorders. AIM OF THE STUDY To review every reported case and present two novel cases to expand the current knowledge and understanding of the mutation. METHODS Whole exome sequencing (WES) was used to identify the novel cases and present their clinical and radiological findings. A detailed revision of the literature was conducted to illustrate and compare findings. The clinical, genetical, neuroimaging, and electrophysiological data were extracted. RESULTS The study included 16 female patients and 13 male patients in addition to the 2 novel male cases. Eighteen patients had heterozygous mutations; others were homozygous. The majority presented with facial dysmorphism (n = 22). Seizures were noted as the predominant hallmark (n = 26). Developmental delay and hypotonia were reported in 27 and 15 patients, respectively. The majority of patients had multifocal epileptiform discharges on the electroencephalogram (EEG) and short and thick corpus callosum on the magnetic resonance imaging (MRI). CONCLUSION Several promising features are becoming strongly linked to patients with SZT2 mutations. High variability among the cases was observed. Developmental delay and facial dysmorphism can be investigated as potential hallmarks; aiding clinicians in diagnosing the condition and optimizing management plans.
Collapse
Affiliation(s)
- Osama Y. Muthaffar
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (O.Y.M.); (M.M.S.J.)
| | - Mohammed M. S. Jan
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (O.Y.M.); (M.M.S.J.)
| | - Anas S. Alyazidi
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.K.A.); (E.A.A.)
| | - Taif K. Alotibi
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.K.A.); (E.A.A.)
| | - Eman A. Alsulami
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.K.A.); (E.A.A.)
| |
Collapse
|
11
|
Ko YJ, Kim SY, Lee S, Yoon JG, Kim MJ, Jun H, Kim H, Chae JH, Kim KJ, Kim K, Lim BC. Epilepsy phenotype and gene ontology analysis of the 129 genes in a large neurodevelopmental disorders cohort. Front Neurol 2023; 14:1218706. [PMID: 37645600 PMCID: PMC10461058 DOI: 10.3389/fneur.2023.1218706] [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: 05/08/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023] Open
Abstract
Objective Although pediatric epilepsy is an independent disease entity, it is often observed in pediatric neurodevelopmental disorders (NDDs) as a major or minor clinical feature, which might provide diagnostic clues. This study aimed to identify the clinical and genetic characteristics of patients with epilepsy in an NDD cohort and demonstrate the importance of genetic testing. Methods We retrospectively analyzed the detailed clinical differences of pediatric NDD patients with epilepsy according to their genetic etiology. Among 1,213 patients with NDDs, 477 were genetically diagnosed by exome sequencing, and 168 had epilepsy and causative variants in 129 genes. Causative genes were classified into two groups: (i) the "epilepsy-genes" group resulting in epilepsy as the main phenotype listed in OMIM, Epi25, and ClinGen (67 patients) and (ii) the "NDD-genes" group not included in the "epilepsy-genes" group (101 patients). Results Patients in the "epilepsy-genes" group started having seizures, often characterized by epilepsy syndrome, at a younger age. However, overall clinical features, including treatment responses and all neurologic manifestations, showed no significant differences between the two groups. Gene ontology analysis revealed the close interactions of epilepsy genes associated with ion channels and neurotransmitters. Conclusion We demonstrated a similar clinical presentation of different gene groups regarding biological/molecular processes in a large NDDs cohort with epilepsy. Phenotype-driven genetic analysis should cover a broad scope, and further studies are required to elucidate integrated pathomechanisms.
Collapse
Affiliation(s)
- Young Jun Ko
- Department of Pediatrics, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Republic of Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Pediatric Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seungbok Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jihoon G. Yoon
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Man Jin Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Jun
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hunmin Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jong-Hee Chae
- Department of Pediatrics, Pediatric Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ki Joong Kim
- Department of Pediatrics, Pediatric Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Byung Chan Lim
- Department of Pediatrics, Pediatric Neuroscience Center, Seoul National University Children's Hospital, Seoul, Republic of Korea
| |
Collapse
|
12
|
Garg RK, Pandey S, Rizvi I, Uniyal R, Sharma PK, Kumar N, Paliwal V. Seizures as Presenting Feature of Subacute Sclerosing Panencephalitis: a Systematic Review of Case Reports and Case Series. CURRENT TROPICAL MEDICINE REPORTS 2023; 10:166-185. [DOI: 10.1007/s40475-023-00291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 01/05/2025]
|
13
|
Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs 2023; 37:399-440. [PMID: 37166702 PMCID: PMC10173947 DOI: 10.1007/s40263-023-01007-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
The sigma-1 receptor is a 223 amino acid-long protein with a recently identified structure. The sigma-2 receptor is a genetically unrelated protein with a similarly shaped binding pocket and acts to influence cellular activities similar to the sigma-1 receptor. Both proteins are highly expressed in neuronal tissues. As such, they have become targets for treating neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), Rett syndrome (RS), developmental and epileptic encephalopathies (DEE), and motor neuron disease/amyotrophic lateral sclerosis (MND/ALS). In recent years, there have been many pre-clinical and clinical studies of sigma receptor (1 and 2) ligands for treating neurological disease. Drugs such as blarcamesine, dextromethorphan and pridopidine, which have sigma-1 receptor activity as part of their pharmacological profile, are effective in treating multiple aspects of several neurological diseases. Furthermore, several sigma-2 receptor ligands are under investigation, including CT1812, rivastigmine and SAS0132. This review aims to provide a current and up-to-date analysis of the current clinical and pre-clinical data of drugs with sigma receptor activities for treating neurological disease.
Collapse
Affiliation(s)
- Dicson S Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokphorn S Ruckvongacheep
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - James M Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Room 409, ChulaPat-1 Building, 154 Rama 1 Road, Bangkok, 10330, Thailand.
| |
Collapse
|
14
|
Oliver KL, Trivisano M, Mandelstam SA, De Dominicis A, Francis DI, Green TE, Muir AM, Chowdhary A, Hertzberg C, Goldhahn K, Metreau J, Prager C, Pinner J, Cardamone M, Myers KA, Leventer RJ, Lesca G, Bahlo M, Hildebrand MS, Mefford HC, Kaindl AM, Specchio N, Scheffer IE. WWOX developmental and epileptic encephalopathy: Understanding the epileptology and the mortality risk. Epilepsia 2023; 64:1351-1367. [PMID: 36779245 PMCID: PMC10952634 DOI: 10.1111/epi.17542] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/14/2023]
Abstract
OBJECTIVE WWOX is an autosomal recessive cause of early infantile developmental and epileptic encephalopathy (WWOX-DEE), also known as WOREE (WWOX-related epileptic encephalopathy). We analyzed the epileptology and imaging features of WWOX-DEE, and investigated genotype-phenotype correlations, particularly with regard to survival. METHODS We studied 13 patients from 12 families with WWOX-DEE. Information regarding seizure semiology, comorbidities, facial dysmorphisms, and disease outcome were collected. Electroencephalographic (EEG) and brain magnetic resonance imaging (MRI) data were analyzed. Pathogenic WWOX variants from our cohort and the literature were coded as either null or missense, allowing individuals to be classified into one of three genotype classes: (1) null/null, (2) null/missense, (3) missense/missense. Differences in survival outcome were estimated using the Kaplan-Meier method. RESULTS All patients experienced multiple seizure types (median onset = 5 weeks, range = 1 day-10 months), the most frequent being focal (85%), epileptic spasms (77%), and tonic seizures (69%). Ictal EEG recordings in six of 13 patients showed tonic (n = 5), myoclonic (n = 2), epileptic spasms (n = 2), focal (n = 1), and migrating focal (n = 1) seizures. Interictal EEGs demonstrated slow background activity with multifocal discharges, predominantly over frontal or temporo-occipital regions. Eleven of 13 patients had a movement disorder, most frequently dystonia. Brain MRIs revealed severe frontotemporal, hippocampal, and optic atrophy, thin corpus callosum, and white matter signal abnormalities. Pathogenic variants were located throughout WWOX and comprised both missense and null changes including five copy number variants (four deletions, one duplication). Survival analyses showed that patients with two null variants are at higher mortality risk (p-value = .0085, log-rank test). SIGNIFICANCE Biallelic WWOX pathogenic variants cause an early infantile developmental and epileptic encephalopathy syndrome. The most common seizure types are focal seizures and epileptic spasms. Mortality risk is associated with mutation type; patients with biallelic null WWOX pathogenic variants have significantly lower survival probability compared to those carrying at least one presumed hypomorphic missense pathogenic variant.
Collapse
Affiliation(s)
- Karen L. Oliver
- Epilepsy Research Centre, Department of MedicineUniversity of Melbourne, Austin HealthHeidelbergVictoriaAustralia
- Population Health and Immunity DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of NeuroscienceBambino Gesù Children's Hospital IRCCS, full member of European Reference Network EpiCARERomeItaly
| | - Simone A. Mandelstam
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
- Department of Radiology, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Angela De Dominicis
- Rare and Complex Epilepsy Unit, Department of NeuroscienceBambino Gesù Children's Hospital IRCCS, full member of European Reference Network EpiCARERomeItaly
- Department of Biomedicine and PreventionUniversity of Rome “Tor Vergata”RomeItaly
| | - David I. Francis
- Victorian Clinical Genetics ServicesMurdoch Children's Research Institute, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Timothy E. Green
- Epilepsy Research Centre, Department of MedicineUniversity of Melbourne, Austin HealthHeidelbergVictoriaAustralia
| | - Alison M. Muir
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Apoorva Chowdhary
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
| | - Christoph Hertzberg
- Zentrum für Sozialpädiatrie und Neuropädiatrie (DBZ)Vivantes Hospital NeukoellnBerlinGermany
| | - Klaus Goldhahn
- Department of Pediatrics and Neuropediatrics, DRK Klinikum WestendBerlinGermany
| | - Julia Metreau
- Department of Pediatric NeurologyHôpital Bicêtre, Assistance Publique Hopitaux de ParisLe Kremlin‐BicêtreFrance
| | - Christine Prager
- Center for Chronically Sick Children (SPZ)Charité‐Universitätsmedizin BerlinBerlinGermany
- Department of Pediatric NeurologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Jason Pinner
- Sydney Children's HospitalRandwickNew South WalesAustralia
- School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Michael Cardamone
- Sydney Children's HospitalRandwickNew South WalesAustralia
- School of Women's and Children's HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Kenneth A. Myers
- Division of Child Neurology, Department of PediatricsMcGill UniversityMontrealQuebecCanada
- Research Institute of the McGill University Health CentreMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMontreal Children's Hospital, McGill UniversityMontrealQuebecCanada
| | - Richard J. Leventer
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of NeurologyRoyal Children's HospitalMelbourneVictoriaAustralia
| | - Gaetan Lesca
- Department of Medical Genetics, Lyon University HospitalUniversité Claude Bernard Lyon 1, member of the European Reference Network EpiCARELyonFrance
| | - Melanie Bahlo
- Population Health and Immunity DivisionWalter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Michael S. Hildebrand
- Epilepsy Research Centre, Department of MedicineUniversity of Melbourne, Austin HealthHeidelbergVictoriaAustralia
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
| | - Heather C. Mefford
- Department of PediatricsUniversity of WashingtonSeattleWashingtonUSA
- Center for Pediatric Neurological Disease ResearchSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Angela M. Kaindl
- Center for Chronically Sick Children (SPZ)Charité‐Universitätsmedizin BerlinBerlinGermany
- Department of Pediatric NeurologyCharité–Universitätsmedizin BerlinBerlinGermany
- Institute of Cell Biology and NeurobiologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of NeuroscienceBambino Gesù Children's Hospital IRCCS, full member of European Reference Network EpiCARERomeItaly
| | - Ingrid E. Scheffer
- Epilepsy Research Centre, Department of MedicineUniversity of Melbourne, Austin HealthHeidelbergVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
| |
Collapse
|
15
|
Raga SV, Essajee F, Solomons R, Van Toorn R, Wilmshurst JM. Epileptic spasms: A South African overview of aetiologies, interventions, and outcomes. Dev Med Child Neurol 2023; 65:526-533. [PMID: 36229895 DOI: 10.1111/dmcn.15433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/28/2022]
Abstract
AIM To better understand the aetiologies of epileptic spasms in infants, as well as the safety and efficacy of high dose corticosteroids in tuberculosis and human immunodeficiency virus (HIV) endemic resource-limited settings. METHOD This was a retrospective analysis of infants with epileptic spasms managed at the tertiary referral centres in the Western Cape, South Africa. RESULTS Of 175 children with epileptic spasms, the median age at onset was 6 months (interquartile range 4-8 months). Structural aetiologies were most common (115 out of 175 [66%]), with two-thirds related to perinatal insults. A lead time to treatment (LTTT) of less than 1 month was more likely in the epileptic encephalopathy/developmental and epileptic encephalopathy (DEE) group: 58 out of 92 (63%), compared to 28 out of 76 (37%) of those with developmental encephalopathy (p = 0.001). Failure to recognize preceding developmental delay was common. Ninety-nine children (57%) received first line hormonal therapy such as adrenocorticotropic hormone. A total of 111 out of 172 children (65%) from the developmental encephalopathy and epileptic encephalopathy/DEE groups had clinical and/or electroencephalogram resolution of spasms within 14 days. In our population, children in whom an aetiology could not be identified were statistically more likely to have moderate to profound developmental delay at 1 year of age: 33 out of 44 (p = 0.001). Based on reported incidence of epileptic spasms, 23 to 58 cases per annum would be expected but a far smaller proportion presented to our centres. INTERPRETATION Whilst this is the largest cohort of infants with epileptic spasms from sub-Saharan Africa, the study size is less than expected; this may reflect misdiagnosis and failure of referral pathways. Despite a reported shorter LTTT, infants with DEE had worse developmental outcomes compared to international studies. Hormonal therapy was safe and effective in our setting, despite exposure to high levels of tuberculosis and HIV. WHAT THIS PAPER ADDS The number of unreferred cases of epileptic spasms in South Africa remains high. Caregivers and health care workers in primary care facilities often fail to recognize developmental delay. The burden of disease from hypoxic-ischaemic encephalopathy remains high in our resource-limited setting. Hormonal treatment (e.g. adrenocorticotropic hormone) was safe and effective despite the high prevalence of human immunodeficiency virus and tuberculosis.
Collapse
Affiliation(s)
- Sharika V Raga
- Paediatric Neurology Division, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Farida Essajee
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Regan Solomons
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ronald Van Toorn
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jo M Wilmshurst
- Paediatric Neurology Division, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
16
|
Song Z, Mao H, Liu J, Sun W, Wu S, Lu X, Jin C, Yang J. Lanthanum Chloride Induces Axon Abnormality Through LKB1-MARK2 and LKB1-STK25-GM130 Signaling Pathways. Cell Mol Neurobiol 2023; 43:1181-1196. [PMID: 35661286 PMCID: PMC11414431 DOI: 10.1007/s10571-022-01237-0] [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: 03/08/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022]
Abstract
Lanthanum (La) is a natural rare-earth element that can damage the central nervous system and impair learning and memory. However, its neurotoxic mechanism remains unclear. In this study, adult female rats were divided into 4 groups and given distilled water solution containing 0%, 0.125%, 0.25%, and 0.5% LaCl3, respectively, and this was done from conception to the end of the location. Their offspring rats were used to establish animal models to investigate LaCl3 neurotoxicity. Primary neurons cultured in vitro were treated with LaCl3 and infected with LKB1 overexpression lentivirus. The results showed that LaCl3 exposure resulted in abnormal axons in the hippocampus and primary cultured neurons. LaCl3 reduced the expression of LKB1, p-LKB1, STRAD and MO25 proteins, and directly or indirectly affected the expression of LKB1, leading to decreased activity of LKB1-MARK2 and LKB1-STK25-GM130 pathways. This study indicated that LaCl3 exposure could interfere with the normal effects of LKB1 in the brain and downregulate LKB1-MARK2 and LKB1-STK25-GM130 signaling pathways, resulting in abnormal axon in offspring rats.
Collapse
Affiliation(s)
- Zeli Song
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Haoyue Mao
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Jinxuan Liu
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Wenchang Sun
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Cuihong Jin
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China
| | - Jinghua Yang
- Department of Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
17
|
Chang YT, Hong SY, Lin WD, Lin CH, Lin SS, Tsai FJ, Chou IC. Genetic Testing in Children with Developmental and Epileptic Encephalopathies: A Review of Advances in Epilepsy Genomics. CHILDREN 2023; 10:children10030556. [PMID: 36980114 PMCID: PMC10047509 DOI: 10.3390/children10030556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Advances in disease-related gene discovery have led to tremendous innovations in the field of epilepsy genetics. Identification of genetic mutations that cause epileptic encephalopathies has opened new avenues for the development of targeted therapies. Clinical testing using extensive gene panels, exomes, and genomes is currently accessible and has resulted in higher rates of diagnosis and better comprehension of the disease mechanisms underlying the condition. Children with developmental disabilities have a higher risk of developing epilepsy. As our understanding of the mechanisms underlying encephalopathies and epilepsies improves, there may be greater potential to develop innovative therapies tailored to an individual’s genotype. This article provides an overview of the significant progress in epilepsy genomics in recent years, with a focus on developmental and epileptic encephalopathies in children. The aim of this review is to enhance comprehension of the clinical utilization of genetic testing in this particular patient population. The development of effective and precise therapeutic strategies for epileptic encephalopathies may be facilitated by a comprehensive understanding of their molecular pathogenesis.
Collapse
Affiliation(s)
- Yu-Tzu Chang
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung 40447, Taiwan; (Y.-T.C.)
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
| | - Syuan-Yu Hong
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
- Department of Medicine, School of Medicine, China Medical University, Taichung 40447, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40447, Taiwan
| | - Wei-De Lin
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung 40447, Taiwan; (Y.-T.C.)
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chien-Heng Lin
- Division of Pediatric Pulmonology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
- Department of Biomedical Imaging and Radiological Science, College of Medicine, China Medial University, Taichung 40447, Taiwan
| | - Sheng-Shing Lin
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung 40447, Taiwan; (Y.-T.C.)
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
- Division of Genetics and Metabolism, China Medical University Children’s Hospital, Taichung 40447, Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung 40447, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 40447, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 40447, Taiwan
| | - I-Ching Chou
- Division of Pediatric Neurology, China Medical University Children’s Hospital, Taichung 40447, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40447, Taiwan
- Correspondence: ; Tel.: +886-4-22052121
| |
Collapse
|
18
|
Marcinkowska AB, Jóźwiak S, Tarasewicz A, Dębska-Ślizień A, Szurowska E. Tuberous Sclerosis Complex Patients' Needs and Difficulties-Results of TAND Questionnaire Analysis in Polish Adult Population. J Clin Med 2022; 11:6536. [PMID: 36362764 PMCID: PMC9658121 DOI: 10.3390/jcm11216536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 09/08/2024] Open
Abstract
INTRODUCTION Tuberous Sclerosis Complex (TSC) is a rare genetic disease. Around 90% of individuals with TSC present some neuropsychiatric manifestations (TSC-associated neuropsychiatric disorders, TAND). To date, none of the studies have focused on the TAND profile of the adult population. Thus, the aim of the study was to describe their potential specific needs and difficulties, including differences in cohorts with or without epilepsy and/or intellectual disability. METHOD The Polish version of the TAND Checklist was used for assessment of individuals with TSC. Participants had to meet the criteria for diagnosis of TSC. One hundred adult participants (forty-eight males/ fifty-two females; mean age 32.33 ± 11.29) were enrolled in the study. Epilepsy was present in 71% of patients; intellectual disability occurred in a total of 37%. RESULTS Only 11% of individuals received complete TAND features examination in the past. Moreover, 91.5 of the subjects had four and more TAND symptoms. Intellectually disabled patients and those with epilepsy had more neuropsychiatric problems than epilepsy-free subjects. CONCLUSIONS Findings reveal that TANDs are common in adults with TSC and are underdiagnosed. Most individuals present several behavioural and cognitive problems. Among psychiatric disorders, the most common are ASD, depression, and anxiety disorder. TAND screening should be widely disseminated and applied in clinical practice for early identification, prevention, and rehabilitation of their difficulties. TAND is one of the most significant issues affecting the quality of life of TSC patients and their carers.
Collapse
Affiliation(s)
- Anna B. Marcinkowska
- Applied Cognitive Neuroscience Lab, Department of Human Physiology, Medical University of Gdansk, Tuwima Str. 15, 80-210 Gdansk, Poland
- 2nd Department of Radiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Agnieszka Tarasewicz
- Department of Nephrology, Transplantology and Internal Diseases, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Diseases, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Edyta Szurowska
- 2nd Department of Radiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
19
|
Brun L, Viemari J, Villard L. Mouse models of Kcnq2 dysfunction. Epilepsia 2022; 63:2813-2826. [PMID: 36047730 PMCID: PMC9828481 DOI: 10.1111/epi.17405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/12/2023]
Abstract
Variants in the Kv7.2 channel subunit encoded by the KCNQ2 gene cause epileptic disorders ranging from a benign form with self-limited epileptic seizures and normal development to severe forms with intractable epileptic seizures and encephalopathy. The biological mechanisms involved in these neurological diseases are still unclear. The disease remains intractable in patients affected by the severe form. Over the past 20 years, KCNQ2 models have been developed to elucidate pathological mechanisms and to identify new therapeutic targets. The diversity of Kcnq2 mouse models has proven invaluable to access neuronal networks and evaluate the associated cognitive deficits. This review summarizes the available models and their contribution to our current understanding of KCNQ2 epileptic disorders.
Collapse
Affiliation(s)
- Lucile Brun
- Aix Marseille Univ, Inserm, MMGMarseilleFrance
| | | | - Laurent Villard
- Aix Marseille Univ, Inserm, MMGMarseilleFrance,Service de Génétique Médicale, AP‐HM, Hôpital de La TimoneMarseilleFrance
| |
Collapse
|
20
|
Concise Review: Stem Cell Models of SCN1A-Related Encephalopathies—Current Perspective and Future Therapies. Cells 2022; 11:cells11193119. [PMID: 36231081 PMCID: PMC9561991 DOI: 10.3390/cells11193119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the SCN1A gene can cause a variety of phenotypes, ranging from mild forms, such as febrile seizures and generalized epilepsy with febrile seizures plus, to severe, such as Dravet and non-Dravet developmental epileptic encephalopathies. Until now, more than two thousand pathogenic variants of the SCN1A gene have been identified and different pathogenic mechanisms (loss vs. gain of function) described, but the precise molecular mechanisms responsible for the deficits exhibited by patients are not fully elucidated. Additionally, the phenotypic variability proves the involvement of other genetic factors in its final expression. This is the reason why animal models and cell line models used to explore the molecular pathology of SCN1A-related disorders are only of limited use. The results of studies based on such models cannot be directly translated to affected individuals because they do not address each patient’s unique genetic background. The generation of functional neurons and glia for patient-derived iPSCs, together with the generation of isogenic controls using CRISPR/Cas technology, and finally, the 3D brain organoid models, seem to be a good way to solve this problem. Here, we review SCN1A-related encephalopathies, as well as the stem cell models used to explore their molecular basis.
Collapse
|
21
|
Gataullina S, Galvani G, Touchet S, Nous C, Lemaire E, Laschet J, Chiron C, Dulac O, Dossi E, Brion JD, Messaoudi S, Alami M, Huberfeld G. GluN2C
selective inhibition is a target to develop new antiepileptic compounds. Epilepsia 2022; 63:2911-2924. [PMID: 36054371 DOI: 10.1111/epi.17396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Many early-onset epilepsies present as developmental and epileptic encephalopathy associated with refractory seizures, altered psychomotor development, and disorganized interictal cortical activity. Abnormal upregulation of specific N-methyl-d-aspartate receptor (NMDA-R) subunits is being disentangled as one of the mechanisms of severe early-onset epilepsies. In tuberous sclerosis complex (TSC), upregulation of the GluN2C subunit of the NMDA-R with slow deactivation kinetic results in increased neuronal excitation and synchronization. METHODS Starting from an available GluN2C/D antagonist, NMDA-R-modulating compounds were developed and screened using a patch clamp on neuronal culture to select those with the strongest inhibitory effect on glutamatergic NMDA currents. For these selected compounds, blood pharmacokinetics and passage through the blood-brain barrier were studied. We tested the effect of the most promising compounds on epileptic activity in Tsc1+/- mice brain slices with multielectrode array, and then in vivo at postnatal ages P14-P17, comparable with the usual age at epilepsy onset in human TSC. RESULTS Using a double-electrode voltage clamp on isolated NMDA currents, we identified the most prominent antagonists of the GluN2C subunit with no effect on GluN2A as a means of preventing side effects. The best compound passing through the blood-brain barrier was selected. Applied in vivo in six Tsc1+/- mice at P14-P17, this compound reduced or completely stopped spontaneous seizures in four of them, and decreased the background activity disorganization. Furthermore, ictal-like discharges stopped on a human brain sample from an infant with epilepsy due to TSC. INTERPRETATION Subunit-selective inhibition is a valuable target for developing drugs for severe epilepsies resulting from an upregulation of NMDA-R subunit-mediated transmission.
Collapse
Affiliation(s)
- S. Gataullina
- Service d’explorations fonctionnelles multidisciplinaires Centre de médecine du sommeil, Antoine Béclère Hospital, APHP, Université Paris Saclay Clamart France
| | - G. Galvani
- AdPueriVitam (APV), Antony France
- Université de Lorraine CNRS, L2CM Nancy France
| | - S. Touchet
- AdPueriVitam (APV), Antony France
- Université de Lorraine CNRS, L2CM Nancy France
| | - C. Nous
- Institut de la Vision, UFR Sciences et Technologies Paris France
| | | | | | - C. Chiron
- Inserm U1141, Paris & APHP, Neuropediatrics, Necker Hospital Paris France
| | - O. Dulac
- AdPueriVitam (APV), Antony France
| | - E. Dossi
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050 Université PSL Paris France
| | - J. D. Brion
- Université Paris‐Saclay CNRS UMR 8076, BioCIS Châtenay‐Malabry France
| | - S. Messaoudi
- Université Paris‐Saclay CNRS UMR 8076, BioCIS Châtenay‐Malabry France
| | - M. Alami
- Université Paris‐Saclay CNRS UMR 8076, BioCIS Châtenay‐Malabry France
| | - G. Huberfeld
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050 Université PSL Paris France
- Neurology Department, Hôpital Fondation Adolphe de Rothschild Paris France
| |
Collapse
|
22
|
Schirmer C, Abboud MA, Lee SC, Bass JS, Mazumder AG, Kamen JL, Krishnan V. Home-cage behavior in the Stargazer mutant mouse. Sci Rep 2022; 12:12801. [PMID: 35896608 PMCID: PMC9329369 DOI: 10.1038/s41598-022-17015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
In many childhood-onset genetic epilepsies, seizures are accompanied by neurobehavioral impairments and motor disability. In the Stargazer mutant mouse, genetic disruptions of Cacng2 result in absence-like spike-wave seizures, cerebellar gait ataxia and vestibular dysfunction, which limit traditional approaches to behavioral phenotyping. Here, we combine videotracking and instrumented home-cage monitoring to resolve the neurobehavioral facets of the murine Stargazer syndrome. We find that despite their gait ataxia, stargazer mutants display horizontal hyperactivity and variable rates of repetitive circling behavior. While feeding rhythms, circadian or ultradian oscillations in activity are unchanged, mutants exhibit fragmented bouts of behaviorally defined "sleep", atypical licking dynamics and lowered sucrose preference. Mutants also display an attenuated response to visual and auditory home-cage perturbations, together with profound reductions in voluntary wheel-running. Our results reveal that the seizures and ataxia of Stargazer mutants occur in the context of a more pervasive behavioral syndrome with elements of encephalopathy, repetitive behavior and anhedonia. These findings expand our understanding of the function of Cacng2.
Collapse
Affiliation(s)
- Catharina Schirmer
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - Mark A Abboud
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - Samuel C Lee
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - John S Bass
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - Arindam G Mazumder
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - Jessica L Kamen
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA
| | - Vaishnav Krishnan
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza St, Neurosensory BCM: MS NB302, Houston, TX, 77030, USA.
| |
Collapse
|
23
|
Lin JJ, Meletti S, Vaudano AE, Lin KL. Developmental and epileptic encephalopathies: Is prognosis related to different epileptic network dysfunctions? Epilepsy Behav 2022; 131:107654. [PMID: 33349540 DOI: 10.1016/j.yebeh.2020.107654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/19/2022]
Abstract
Developmental and epileptic encephalopathies are a group of rare, severe epilepsies, which are characterized by refractory seizures starting in infancy or childhood and developmental delay or regression. Developmental changes might be independent of epilepsy. However, interictal epileptic activity and seizures can further deteriorate cognition and behavior. Recently, the concept of developmental and epileptic encephalopathies has moved from the lesions associated with epileptic encephalopathies toward the epileptic network dysfunctions on the functioning of the brain. Early recognition and differentiation of patients with developmental and epileptic encephalopathies is important, as precision therapies need to be holistic to address the often devastating symptoms. In this review, we discuss the evolution of the concept of developmental and epileptic encephalopathies in recent years, as well as the current understanding of the genetic basis of developmental and epileptic encephalopathies. Finally, we will discuss the role of epileptic network dysfunctions on prognosis for these severe conditions.
Collapse
Affiliation(s)
- Jainn-Jim Lin
- Division of Pediatric Critical Care and Pediatric Neurocritical Care Center, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Taoyuan, Taiwan; Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Study Group for Intensive and Integrated Care of Pediatric Central Nervous System (iCNS Group), Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Stefano Meletti
- Division of Neurology, University Hospital of Modena, Modena, Italy; Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Elisabetta Vaudano
- Division of Neurology, University Hospital of Modena, Modena, Italy; Department of Biomedical, Metabolic and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Kuang-Lin Lin
- Division of Pediatric Neurology, Chang Gung Children's Hospital and Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Study Group for Intensive and Integrated Care of Pediatric Central Nervous System (iCNS Group), Chang Gung Children's Hospital, Taoyuan, Taiwan.
| |
Collapse
|
24
|
Clinical whole exome sequencing revealed de novo Heterozygous Stop-Gain and Missense variants in the STXBP1 gene associated with Epilepsy in Saudi Families. Saudi J Biol Sci 2022; 29:103309. [PMID: 35663845 PMCID: PMC9160351 DOI: 10.1016/j.sjbs.2022.103309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/06/2022] [Accepted: 05/15/2022] [Indexed: 11/21/2022] Open
Abstract
Intellectual disability and developmental encephalopathies are mostly linked with infant epilepsy. Epileptic encephalopathy is a term that is used to define association between developmental delay and epilepsy. Mutations in the STXBP1 (Syntaxin-binding protein 1) gene have been previously reported in association with multiple severe early epileptic encephalopathies along with many neurodevelopmental disorders. Among the disorders produced due to any mutations in the STXBP1 gene is developmental and epileptic encephalopathy 4 (OMIM: 612164), is an autosomal dominant neurologic disorder categorized by the onset of tonic seizures in early infancy (usually in the first months of life). In this article, we report two Saudi families one with de novo heterozygous stop-gain mutation c.364C > T and a novel missense c. 305C > A p.Ala102Glu in exon 5 of the STXBP1 gene (OMIM: 602926) lead to development of epileptic encephalopathy 4. The variants identified in the current study broadened the genetic spectrum of STXBP1 gene related with diseases, which will help to add in the literature and benefit to the studies addressing this disease in the future.
Collapse
|
25
|
Hieu NLT, Thu NTM, Ngan LTA, Van LTK, Huy DP, Linh PTT, Mai NTQ, Hien HTD, Hang DTT. Genetic analysis using targeted exome sequencing of 53 Vietnamese children with developmental and epileptic encephalopathies. Am J Med Genet A 2022; 188:2048-2060. [PMID: 35365919 DOI: 10.1002/ajmg.a.62741] [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: 08/15/2021] [Revised: 01/14/2022] [Accepted: 03/13/2022] [Indexed: 11/07/2022]
Abstract
Developmental and epileptic encephalopathies (DEE) refers to a group of rare and severe neurodevelopmental disorders where genetic etiologies can play a major role. This study aimed to elucidate the genetic etiologies of a cohort of 53 Vietnamese patients with DEE. All patients were classified into known electroclinical syndromes where possible. Exome sequencing (ES) followed by a targeted analysis on 294 DEE-related genes was then performed. Patients with identified causative variants were followed for 6 months to determine the impact of genetic testing on their treatment. The diagnostic yield was 38.0% (20/53), which was significantly higher in the earlier onset group (<12 months) than in the later onset group (≥12 months). The 19 identified variants belonged to 11 genes with various cellular functions. Genes encoding ion channels especially sodium voltage-gated channel were the most frequently involved. Most variants were missense variants and located in key protein functional domains. Four variants were novel and four had been reported previously but in different phenotypes. Within 6 months of further follow-up, treatment changes were applied for six patients based on the identified disease-causing variants, with five patients showing a positive impact. This is the first study in Vietnam to analyze the genetics of DEE. This study confirms the strong involvement of genetic etiologies in DEE, especially early onset DEE. The study also contributes to clarify the genotype-phenotype correlations of DEE and highlights the efficacy of targeted ES in the diagnosis and treatment of DEE.
Collapse
Affiliation(s)
- Nguyen Le Trung Hieu
- Neurology Department, Children Hospital 2, Ho Chi Minh City, Vietnam.,University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | | | - Le Tran Anh Ngan
- Neurology Department, Children Hospital 2, Ho Chi Minh City, Vietnam
| | - Le Thi Khanh Van
- Neurology Department, Children Hospital 2, Ho Chi Minh City, Vietnam
| | - Do Phuoc Huy
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | - Pham Thi Truc Linh
- Functional Genomic Unit, DNA Medical Technology Company, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Quynh Mai
- Research Center for Genetics and Reproductive Health, School of Medicine, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Huynh Thi Dieu Hien
- Research Center for Genetics and Reproductive Health, School of Medicine, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Do Thi Thu Hang
- Research Center for Genetics and Reproductive Health, School of Medicine, Vietnam National University, Ho Chi Minh City, Vietnam
| |
Collapse
|
26
|
Zanus C, Carrozzi M, Costa P, Musante L, Faletra F. Encefalopatie epilettiche e dello sviluppo: dalla pratica alla genetica, andata e ritorno. MEDICO E BAMBINO 2022; 41:33-40. [DOI: 10.53126/meb41033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
In the last decades the research on the genetics of epilepsy has greatly expanded, supported by the development of effective next-generation sequencing (NGS) methods. In particular, the studies in Developmental and Epileptic Encephalopathies (DEEs) discover an everyday increasing number of new epilepsy-associated genes and provide information with positive effects on genetic counselling and treatment choice. Thanks to NGS, genetics is becoming a first line diagnostic test in DEEs but a consensus on who should receive genetic testing, on the best testing strategy and on the utility of genetic testing is still lacking. Many patients are being studied with research sequencing and analysis. The participation in a study on the genetic causes of DEE led to share some considerations about this topic and the most relevant aspects of the described experience.
Collapse
Affiliation(s)
- Caterina Zanus
- SC di Neuropsichiatria Infantile, RCCS Materno-Infantile “Burlo Garofolo”, Trieste
| | - Marco Carrozzi
- SC di Neuropsichiatria Infantile, RCCS Materno-Infantile “Burlo Garofolo”, Trieste
| | - Paola Costa
- SC di Neuropsichiatria Infantile, RCCS Materno-Infantile “Burlo Garofolo”, Trieste
| | - Luciana Musante
- SC di Genetica Medica, IRCCS Materno-Infantile “Burlo Garofolo”, Trieste
| | - Flavio Faletra
- SC di Genetica Medica, IRCCS Materno-Infantile “Burlo Garofolo”, Trieste
| |
Collapse
|
27
|
Geng H, Chen X. Development and validation of a nomogram for the early prediction of drug resistance in children with epilepsy. Front Pediatr 2022; 10:905177. [PMID: 36110106 PMCID: PMC9468368 DOI: 10.3389/fped.2022.905177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/28/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND PURPOSE This study aimed to effectively identify children with drug-resistant epilepsy (DRE) in the early stage of epilepsy, and take personalized interventions, to improve patients' prognosis, reduce serious comorbidity, and save social resources. Herein, we developed and validated a nomogram prediction model for children with DRE. METHODS The training set was patients with epilepsy who visited the Children's Hospital of Soochow University (Suzhou Industrial Park, Jiangsu Province, China) between January 2015 and December 2017. The independent risk factors for DRE were screened by univariate and multivariate logistic regression analyses using SPSS21 software. The nomogram was designed according to the regression coefficient. The nomogram was validated in the training and validation sets. Internal validation was conducted using bootstrapping analyses. We also externally validated this instrument in patients with epilepsy from the Children's Hospital of Soochow University (Gusu District, Jiangsu Province, China) and Yancheng Maternal and Child Health Hospital between January 2018 and December 2018. The nomogram's performance was assessed by concordance (C-index), calibration curves, as well as GiViTI calibration belts. RESULTS Multivariate logistic regression analysis of 679 children with epilepsy from the Children's Hospital of Soochow University (Suzhou Industrial Park, Jiangsu Province, China) showed that onset age<1, status epilepticus (SE), focal seizure, > 20 pre-treatment seizures, clear etiology (caused by genetic, structural, metabolic, or infectious), development and epileptic encephalopathy (DEE), and neurological abnormalities were all independent risk factors for DRE. The AUC of 0.92 for the training set compared to that of 0.91 for the validation set suggested a good discrimination ability of the prediction model. The C-index was 0.92 and 0.91 in the training and validation sets. Additionally, both good calibration curves and GiViTI calibration belts (P-value: 0.849 and 0.291, respectively) demonstrated that the predicted risks had strong consistency with the observed outcomes, suggesting that the prediction model in both groups was perfectly calibrated. CONCLUSION A nomogram prediction model for DRE was developed, with good discrimination and calibration in the training set and the validation set. Furthermore, the model demonstrated great accuracy, consistency, and prediction ability. Therefore, the nomogram prediction model can aid in the timely identification of DRE in children.
Collapse
Affiliation(s)
- Hua Geng
- Neurology Department, Children's Hospital of Soochow University, Suzhou, China
| | - Xuqin Chen
- Neurology Department, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
28
|
Specchio N, Di Micco V, Trivisano M, Ferretti A, Curatolo P. The epilepsy-autism spectrum disorder phenotype in the era of molecular genetics and precision therapy. Epilepsia 2021; 63:6-21. [PMID: 34741464 DOI: 10.1111/epi.17115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 11/30/2022]
Abstract
Autism spectrum disorder (ASD) is frequently associated with infants with epileptic encephalopathy, and early interventions targeting social and cognitive deficits can have positive effects on developmental outcome. However, early diagnosis of ASD among infants with epilepsy is complicated by variability in clinical phenotypes. Commonality in both biological and molecular mechanisms have been suggested between ASD and epilepsy, such as occurs with tuberous sclerosis complex. This review summarizes the current understanding of causal mechanisms between epilepsy and ASD, with a particularly genetic focus. Hypothetical explanations to support the conjugation of the two conditions include abnormalities in synaptic growth, imbalance in neuronal excitation/inhibition, and abnormal synaptic plasticity. Investigation of the probable genetic basis has implemented many genes, although the main risk supports existing hypotheses in that these cluster to abnormalities in ion channels, synaptic function and structure, and transcription regulators, with the mammalian target of rapamycin (mTOR) pathway and "mTORpathies" having been a notable research focus. Experimental models not only have a crucial role in determining gene functions but are also useful instruments for tracing disease trajectory. Precision medicine from gene therapy remains a theoretical possibility, but more contemporary developments continue in molecular tests to aid earlier diagnoses and better therapeutic targeting.
Collapse
Affiliation(s)
- Nicola Specchio
- Rare and Complex Epilepsy Unit, Division of Neurology, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome, Italy
| | - Valentina Di Micco
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| | - Marina Trivisano
- Rare and Complex Epilepsy Unit, Division of Neurology, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome, Italy
| | - Alessandro Ferretti
- Rare and Complex Epilepsy Unit, Division of Neurology, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy
| |
Collapse
|
29
|
Specchio N, Curatolo P. Developmental and epileptic encephalopathies: what we do and do not know. Brain 2021; 144:32-43. [PMID: 33279965 DOI: 10.1093/brain/awaa371] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/16/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Developmental encephalopathies, including intellectual disability and autistic spectrum disorder, are frequently associated with infant epilepsy. Epileptic encephalopathy is used to describe an assumed causal relationship between epilepsy and developmental delay. Developmental encephalopathies pathogenesis more independent from epilepsy is supported by the identification of several gene variants associated with both developmental encephalopathies and epilepsy, the possibility for gene-associated developmental encephalopathies without epilepsy, and the continued development of developmental encephalopathies even when seizures are controlled. Hence, 'developmental and epileptic encephalopathy' may be a more appropriate term than epileptic encephalopathy. This update considers the best studied 'developmental and epileptic encephalopathy' gene variants for illustrative support for 'developmental and epileptic encephalopathy' over epileptic encephalopathy. Moreover, the interaction between epilepsy and developmental encephalopathies is considered with respect to influence on treatment decisions. Continued research in genetic testing will increase access to clinical tests, earlier diagnosis, better application of current treatments, and potentially provide new molecular-investigated treatments.
Collapse
Affiliation(s)
- Nicola Specchio
- Department of Neuroscience, Bambino Gesu Children's Hospital, IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies EpiCARE, Piazza S, 00165 Rome, Italy
| | - Paolo Curatolo
- Systems Medicine Department, Child Neurology and Psychiatry Unit, Tor Vergata University Hospital of Rome, 00133 Rome, Italy
| |
Collapse
|
30
|
Trivisano M, Rivera M, Terracciano A, Ciolfi A, Napolitano A, Pepi C, Calabrese C, Digilio MC, Tartaglia M, Curatolo P, Vigevano F, Specchio N. Developmental and epileptic encephalopathy due to SZT2 genomic variants: Emerging features of a syndromic condition. Epilepsy Behav 2020; 108:107097. [PMID: 32402703 DOI: 10.1016/j.yebeh.2020.107097] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/04/2020] [Accepted: 04/04/2020] [Indexed: 10/24/2022]
Abstract
Seizure threshold 2 (SZT2) gene mutations have been associated with developmental and epileptic encephalopathies (DEEs). Following a literature review, we collected 22 patients and identified the main clinical features related to SZT2 variants that are epilepsy with onset within the first years of life, intellectual disability (ID), macrocephaly with dysmorphic facial features, corpus callosum (CC) shape abnormalities, and cortical migration disorders. Moreover, we identified the c.7825T>G homozygous missense variant in SZT2 in two female siblings presenting with focal seizures, mild-moderate ID, behavioral disturbances, and facial dysmorphisms. Interictal Electroencephalogram (EEG) and ictal EEG were both informative and revealed, respectively, temporal bilateral asynchronous slow and epileptiform abnormalities and a focal onset in both of them. Neuroimaging study revealed a thick and abnormally shaped CC. Seizure threshold 2 has been identified as a component of the KICSTOR complex, a newly recognized protein complex involved in the mammalian target of rapamycin (mTOR) pathway. mTOR signaling dysregulation represents common pathogenetic mechanisms that can explain the presence of both epileptogenesis and ID. Even if few cases had been reported, a new clinical phenotype is emerging, and recent hypothesis of hyperactivation of mTORC1 signaling might also open to targeted treatments, challenging an early diagnosis as of paramount importance.
Collapse
Affiliation(s)
- Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manuel Rivera
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Departamento de Neuropediatria, Fleni, Montañeses 2325, C1428AQK Ciudad de Buenos Aires, Argentina
| | | | - Andrea Ciolfi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Antonio Napolitano
- Neuroradiology Unit, Department of Imaging, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chiara Pepi
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy
| | - Costanza Calabrese
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Cristina Digilio
- Medical Genetics, Department of Pediatrics, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Via Montpellier 1, 00133 Rome, Italy
| | - Federico Vigevano
- Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome; Member of European Reference Network EpiCARE
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Member of European Reference Network EpiCARE.
| |
Collapse
|
31
|
Trivisano M, Ferretti A, Bebin E, Huh L, Lesca G, Siekierska A, Takeguchi R, Carneiro M, De Palma L, Guella I, Haginoya K, Shi RM, Kikuchi A, Kobayashi T, Jung J, Lagae L, Milh M, Mathieu ML, Minassian BA, Novelli A, Pietrafusa N, Takeshita E, Tartaglia M, Terracciano A, Thompson ML, Cooper GM, Vigevano F, Villard L, Villeneuve N, Buyse GM, Demos M, Scheffer IE, Specchio N. Defining the phenotype of FHF1 developmental and epileptic encephalopathy. Epilepsia 2020; 61:e71-e78. [PMID: 32645220 PMCID: PMC8168379 DOI: 10.1111/epi.16582] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/25/2023]
Abstract
Fibroblast growth-factor homologous factor (FHF1) gene variants have recently been associated with developmental and epileptic encephalopathy (DEE). FHF1 encodes a cytosolic protein that modulates neuronal sodium channel gating. We aim to refine the electroclinical phenotypic spectrum of patients with pathogenic FHF1 variants. We retrospectively collected clinical, genetic, neurophysiologic, and neuroimaging data of 17 patients with FHF1-DEE. Sixteen patients had recurrent heterozygous FHF1 missense variants: 14 had the recurrent p.Arg114His variant and two had a novel likely pathogenic variant p.Gly112Ser. The p.Arg114His variant is associated with an earlier onset and more severe phenotype. One patient carried a chromosomal microduplication involving FHF1. Twelve patients carried a de novo variant, five (29.5%) inherited from parents with gonadic or somatic mosaicism. Seizure onset was between 1 day and 41 months; in 76.5% it was within 30 days. Tonic seizures were the most frequent seizure type. Twelve patients (70.6%) had drug-resistant epilepsy, 14 (82.3%) intellectual disability, and 11 (64.7%) behavioral disturbances. Brain magnetic resonance imaging (MRI) showed mild cerebral and/or cerebellar atrophy in nine patients (52.9%). Overall, our findings expand and refine the clinical, EEG, and imaging phenotype of patients with FHF1-DEE, which is characterized by early onset epilepsy with tonic seizures, associated with moderate to severe ID and psychiatric features.
Collapse
Affiliation(s)
- Marina Trivisano
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Alessandro Ferretti
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Elizabeth Bebin
- University of Alabama at Birmingham, Department of Pediatric Neurology, Birmingham, AL, USA
| | - Linda Huh
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, BC, Canada
| | - Gaetan Lesca
- Service de Génétique, Hospices Civils de Lyon - Lyon – France and Institut Neuromyogène, Equipe Métabolisme énergétique et développement neuronal, CNRS UMR 5310, INSERM U1217, Université Lyon 1 - Lyon - France
| | | | - Ryo Takeguchi
- Department of Pediatrics, Asahikawa Medical University, Asahikawa, Japan
| | - Maryline Carneiro
- Department of Pediatric Neurology, Femme Mère Enfant Hospital, Hospices Civils de Lyon, France
| | - Luca De Palma
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Ilaria Guella
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, BC, Canada
| | - Kazuhiro Haginoya
- Department of Pediatric Neurology, Miyagi Children’s Hopital, Sendai 989-3126, Japan
| | - Ruo Ming Shi
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan and Department of Pediatrics, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan
| | - Tomoko Kobayashi
- Department of Pediatrics, Tohoku University Hospital, Sendai, Japan and Division of Child Development, Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Julien Jung
- Service de Génétique, Hospices Civils de Lyon - Lyon – France and Institut Neuromyogène, Equipe Métabolisme énergétique et développement neuronal, CNRS UMR 5310, INSERM U1217, Université Lyon 1 - Lyon - France
| | - Lieven Lagae
- Department of Development and Regeneration, University Hospitals KU Leuven, Leuven, Belgium
| | - Mathieu Milh
- Department of Pediatric Neurology, Femme Mère Enfant Hospital, Hospices Civils de Lyon, France
| | - Marie L Mathieu
- Department of Pediatric Neurology, Femme Mère Enfant Hospital, Hospices Civils de Lyon, France
| | - Berge A Minassian
- Department of Pediatrics, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonio Novelli
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Nicola Pietrafusa
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Eri Takeshita
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Alessandra Terracciano
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | | | - Federico Vigevano
- Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | | | - Nathalie Villeneuve
- APHM, Department of Pediatric Neurology, Hopital de la Timone, Marseille, France
| | - Gunnar M Buyse
- Pediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Michelle Demos
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, BC, Canada
| | - Ingrid E Scheffer
- University of Melbourne, Austin Health, and Royal Children’s Hospital, Florey and Murdoch Institutes, Melbourne, Australia
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| |
Collapse
|