Journal of Pediatric Neurology 2023; 21(03): 173-185
DOI: 10.1055/s-0041-1727097
Review Article

SCN2A and Its Related Epileptic Phenotypes

Andrea D. Praticò
1   Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
,
Alessandro Giallongo
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Marta Arrabito
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Silvia D'Amico
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Maria Cristina Gauci
1   Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
,
Giulia Lombardo
2   Pediatrics Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
,
Agata Polizzi
3   Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
,
Raffaele Falsaperla
4   Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
5   Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
,
Martino Ruggieri
1   Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, Catania, Italy
› Author Affiliations

Abstract

Epilepsies due to SCN2A mutations can present with a broad range of phenotypes that are still not fully understood. Clinical characteristics of SNC2A-related epilepsy may vary from neonatal benign epilepsy to early-onset epileptic encephalopathy, including Ohtahara syndrome and West syndrome, and epileptic encephalopathies occurring at later ages (usually within the first 10 years of life). Some patient may present with intellectual disability and/or autism or movement disorders and without epilepsy. The heterogeneity of the phenotypes associated to such genetic mutations does not always allow the clinician to address his suspect on this gene. For this reason, diagnosis is usually made after a multiple gene panel examination through next generation sequencing (NGS) or after whole exome sequencing (WES) or whole genome sequencing (WGS). Subsequently, confirmation by Sanger sequencing can be obtained. Mutations in SCN2A are inherited as an autosomal dominant trait. Most individuals diagnosed with SCN2A–benign familial neonatal-infantile seizures (BFNIS) have an affected parent; however, hypothetically, a child may present SCN2A-BNFNIS as the result of a de novo pathogenic variant. Almost all individuals with SCN2A and severe epileptic encephalopathies have a de novo pathogenic variant. SNC2A-related epilepsies have not shown a clear genotype–phenotype correlation; in some cases, a same variant may lead to different presentations even within the same family and this could be due to other genetic factors or to environmental causes. There is no “standardized” treatment for SCN2A-related epilepsy, as it varies in relation to the clinical presentation and the phenotype of the patient, according to its own gene mutation. Treatment is based mainly on antiepileptic drugs, which include classic wide-spectrum drugs, such as valproic acid, levetiracetam, and lamotrigine. However, specific agents, which act directly modulating the sodium channels activity (phenytoin, carbamazepine, oxcarbamazepine, lamotrigine, and zonisamide), have shown positive result, as other sodium channel blockers (lidocaine and mexiletine) or even other drugs with different targets (phenobarbital).



Publication History

Received: 02 September 2020

Accepted: 23 January 2021

Article published online:
27 March 2021

© 2021. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Orsini A, Zara F, Striano P. Recent advances in epilepsy genetics. Neurosci Lett 2018; 667: 4-9
  • 2 McTague A, Howell KB, Cross JH, Kurian MA, Scheffer IE. The genetic landscape of the epileptic encephalopathies of infancy and childhood. Lancet Neurol 2016; 15 (03) 304-316
  • 3 Ruggieri M, Rizzo R, Pavone P, Baieli S, Sorge G, Happle R. Temporal triangular alopecia in association with mental retardation and epilepsy in a mother and daughter. Arch Dermatol 2000; 136 (03) 426-427
  • 4 Ruggieri M, Iannetti P, Clementi M. et al. Neurofibromatosis type 1 and infantile spasms. Childs Nerv Syst 2009; 25 (02) 211-216
  • 5 Sugawara T, Tsurubuchi Y, Agarwala KL. et al. A missense mutation of the Na+ channel alpha II subunit gene Na(v)1.2 in a patient with febrile and afebrile seizures causes channel dysfunction. Proc Natl Acad Sci U S A 2001; 98 (11) 6384-6389
  • 6 Litt M, Luty J, Kwak M, Allen L, Magenis RE, Mandel G. Localization of a human brain sodium channel gene (SCN2A) to chromosome 2. Genomics 1989; 5 (02) 204-208
  • 7 Liao WP, Shi YW, Long YS. et al. Partial epilepsy with antecedent febrile seizures and seizure aggravation by antiepileptic drugs: associated with loss of function of Na(v) 1.1. Epilepsia 2010; 51 (09) 1669-1678
  • 8 Ben-Shalom R, Keeshen CM, Berrios KN, An JY, Sanders SJ, Bender KJ. Opposing effects on NaV1.2 function underlie differences between SCN2A variants observed in individuals with autism spectrum disorder or infantile seizures. Biol Psychiatry 2017; 82 (03) 224-232
  • 9 Ruggieri M, Milone P, Pavone P. et al. Nevus vascularis mixtus (cutaneous vascular twin nevi) associated with intracranial vascular malformation of the Dyke-Davidoff-Masson type in two patients. Am J Med Genet A 2012; 158A (11) 2870-2880
  • 10 Sorge G, Ruggieri M, Polizzi A, Scuderi A, Di Pietro M. SHORT syndrome: a new case with probable autosomal dominant inheritance. Am J Med Genet 1996; 61 (02) 178-181
  • 11 Spratt PWE, Ben-Shalom R, Keeshen CM. et al. The autism-associated gene Scn2a contributes to dendritic excitability and synaptic function in the prefrontal cortex. Neuron 2019; 103 (04) 673-685.e5
  • 12 Jiang P, Chen C, Liu XB. et al. Generation and characterization of spiking and nonspiking oligodendroglial progenitor cells from embryonic stem cells. Stem Cells 2013; 31 (12) 2620-2631
  • 13 Vlachou V, Larsen L, Pavlidou E. et al. SCN2A mutation in an infant with Ohtahara syndrome and neuroimaging findings: expanding the phenotype of neuronal migration disorders. J Genet 2019; 98 (02) 54
  • 14 Ruggieri M, Iannetti P, Pavone L. Delineation of a newly recognized neurocutaneous malformation syndrome with “cutis tricolor”. Am J Med Genet A 2003; 120A (01) 110-116
  • 15 Pavone P, Praticò AD, Vitaliti G. et al. Hydranencephaly: cerebral spinal fluid instead of cerebral mantles. Ital J Pediatr 2014; 40: 79
  • 16 Sanders SJ, Campbell AJ, Cottrell JR. et al. Progress in understanding and treating SCN2A-mediated disorders. Trends Neurosci 2018; 41 (07) 442-456
  • 17 Brunklaus A, Du J, Steckler F. et al. Biological concepts in human sodium channel epilepsies and their relevance in clinical practice. Epilepsia 2020; 61 (03) 387-399
  • 18 Hedrich UBS, Lauxmann S, Lerche H. SCN2A channelopathies: mechanisms and models. Epilepsia 2019; 60 (Suppl. 03) S68-S76
  • 19 Wolff M, Johannesen KM, Hedrich UBS. et al. Genetic and phenotypic heterogeneity suggest therapeutic implications in SCN2A-related disorders. Brain 2017; 140 (05) 1316-1336
  • 20 Wolff M, Brunklaus A, Zuberi SM. Phenotypic spectrum and genetics of SCN2A-related disorders, treatment options, and outcomes in epilepsy and beyond. Epilepsia 2019; 60 (Suppl. 03) S59-S67
  • 21 Ito M, Shirasaka Y, Hirose S, Sugawara T, Yamakawa K. Seizure phenotypes of a family with missense mutations in SCN2A . Pediatr Neurol 2004; 31 (02) 150-152
  • 22 Kamiya K, Kaneda M, Sugawara T. et al. A nonsense mutation of the sodium channel gene SCN2A in a patient with intractable epilepsy and mental decline. J Neurosci 2004; 24 (11) 2690-2698
  • 23 Striano P, Bordo L, Lispi ML. et al. A novel SCN2A mutation in family with benign familial infantile seizures. Epilepsia 2006; 47 (01) 218-220
  • 24 Herlenius E, Heron SE, Grinton BE. et al. SCN2A mutations and benign familial neonatal-infantile seizures: the phenotypic spectrum. Epilepsia 2007; 48 (06) 1138-1142
  • 25 Ogiwara I, Ito K, Sawaishi Y. et al. De novo mutations of voltage-gated sodium channel alphaII gene SCN2A in intractable epilepsies. Neurology 2009; 73 (13) 1046-1053
  • 26 Kobayashi K, Ohzono H, Shinohara M. et al. Acute encephalopathy with a novel point mutation in the SCN2A gene. Epilepsy Res 2012; 102 (1,2): 109-112
  • 27 Dhamija R, Wirrell E, Falcao G, Kirmani S, Wong-Kisiel LC. Novel de novo SCN2A mutation in a child with migrating focal seizures of infancy. Pediatr Neurol 2013; 49 (06) 486-488
  • 28 Lauxmann S, Boutry-Kryza N, Rivier C. et al. An SCN2A mutation in a family with infantile seizures from Madagascar reveals an increased subthreshold Na(+) current. Epilepsia 2013; 54 (09) e117-e121
  • 29 Sundaram SK, Chugani HT, Tiwari VN, Huq AH. SCN2A mutation is associated with infantile spasms and bitemporal glucose hypometabolism. Pediatr Neurol 2013; 49 (01) 46-49
  • 30 Touma M, Joshi M, Connolly MC. et al. Whole genome sequencing identifies SCN2A mutation in monozygotic twins with Ohtahara syndrome and unique neuropathologic findings. Epilepsia 2013; 54 (05) e81-e85
  • 31 Hackenberg A, Baumer A, Sticht H. et al. Infantile epileptic encephalopathy, transient choreoathetotic movements, and hypersomnia due to a de novo missense mutation in the SCN2A gene. Neuropediatrics 2014; 45 (04) 261-264
  • 32 Matalon D, Goldberg E, Medne L, Marsh ED. Confirming an expanded spectrum of SCN2A mutations: a case series. Epileptic Disord 2014; 16 (01) 13-18
  • 33 Zerem A, Lev D, Blumkin L. et al. Paternal germline mosaicism of a SCN2A mutation results in Ohtahara syndrome in half siblings. Eur J Paediatr Neurol 2014; 18 (05) 567-571
  • 34 Fukasawa T, Kubota T, Negoro T. et al. A case of recurrent encephalopathy with SCN2A missense mutation. Brain Dev 2015; 37 (06) 631-634
  • 35 Saitoh M, Ishii A, Ihara Y. et al. Missense mutations in sodium channel SCN1A and SCN2A predispose children to encephalopathy with severe febrile seizures. Epilepsy Res 2015; 117: 1-6
  • 36 Samanta D, Ramakrishnaiah R. De novo R853Q mutation of SCN2A gene and West syndrome. Acta Neurol Belg 2015; 115 (04) 773-776
  • 37 Schwarz N, Hahn A, Bast T. et al. Mutations in the sodium channel gene SCN2A cause neonatal epilepsy with late-onset episodic ataxia. J Neurol 2016; 263 (02) 334-343
  • 38 Johannesen KM, Miranda MJ, Lerche H, Moller RS. Letter to the editor: confirming neonatal seizure and late onset ataxia in SCN2A Ala263Val. J Neurol 2016; 263 (07) 1459-1460
  • 39 Leach EL, van Karnebeek CD, Townsend KN, Tarailo-Graovac M, Hukin J, Gibson WT. Episodic ataxia associated with a de novo SCN2A mutation. Eur J Paediatr Neurol 2016; 20 (05) 772-776
  • 40 Syrbe S, Zhorov BS, Bertsche A. et al. Phenotypic variability from benign infantile epilepsy to Ohtahara syndrome associated with a novel mutation in SCN2A . Mol Syndromol 2016; 7 (04) 182-188
  • 41 Singh D, Lau M, Ayers T. et al. De novo heterogeneous mutations in SCN2A and GRIN2A genes and seizures with ictal vocalizations. Clin Pediatr (Phila) 2016; 55 (09) 867-870
  • 42 Bernardo S, Marchionni E, Prudente S. et al. Unusual association of SCN2A epileptic encephalopathy with severe cortical dysplasia detected by prenatal MRI. Eur J Paediatr Neurol 2017; 21 (03) 587-590
  • 43 Cheng Y, Zhang L, Huang X. et al. De novo SCN2A mutation in a Chinese infant with severe early-onset epileptic encephalopathy, bronchopulmonary dysplasia, and adrenal hypofunction. Int J Clin Exp Pathol 2017; 10 (10) 10358-10362
  • 44 Dilena R, Striano P, Gennaro E. et al. Efficacy of sodium channel blockers in SCN2A early infantile epileptic encephalopathy. Brain Dev 2017; 39 (04) 345-348
  • 45 Foster LA, Johnson MR, MacDonald JT. et al. Infantile epileptic encephalopathy associated with SCN2A mutation responsive to oral mexiletine. Pediatr Neurol 2017; 66: 108-111
  • 46 Gorman KM, King MD. SCN2A p.Ala263Val variant a phenotype of neonatal seizures followed by paroxysmal ataxia in toddlers. Pediatr Neurol 2017; 67: 111-112
  • 47 Liang JS, Lin LJ, Yang MT, Wang JS, Lu JF. The therapeutic implication of a novel SCN2A mutation associated early-onset epileptic encephalopathy with Rett-like features. Brain Dev 2017; 39 (10) 877-881
  • 48 Sauvestre F, Moutton S, Badens C. et al. In utero seizures revealing dentato-olivary dysplasia caused by SCN2A mutation. Neuropathol Appl Neurobiol 2017; 43 (07) 631-635
  • 49 Hadar PN, Kini LG, Coto C. et al. Clinical validation of automated hippocampal segmentation in temporal lobe epilepsy. Neuroimage Clin 2018; 20: 1139-1147
  • 50 Lee HH, Lau NK, Yeung CW, Ng SG, Yau KE, Mak CM. Successful adaptation of targeted gene panel next-generation sequencing in regional hospital in Hong Kong: genomic diagnosis of SCN2A-related seizure disorder. Chin Med J (Engl) 2018; 131 (18) 2262-2264
  • 51 Leroy A, Corfiotti C, Nguyen The Tich S. et al. Catatonia associated with a SCN2A-related disorder in a 4-year-old child. Pediatrics 2018; 142 (05) e20181231
  • 52 Liu XW, Li W, Han T. et al. The finding of a new heterozygous mutation site of the SCN2A gene in a monozygotic twin family carrying and exhibiting genetic epilepsy with febrile seizures plus (GEFS+) using targeted next-generation sequencing. Clin Neurol Neurosurg 2018; 169: 86-91
  • 53 Su DJ, Lu JF, Lin LJ, Liang JS, Hung KL. SCN2A mutation in an infant presenting with migrating focal seizures and infantile spasm responsive to a ketogenic diet. Brain Dev 2018; 40 (08) 724-727
  • 54 Türkdoğan D, Matthews E, Usluer S. et al. Possible role of SCN4A skeletal muscle mutation in apnea during seizure. Epilepsia Open 2019; 4 (03) 498-503
  • 55 AlSaif S, Umair M, Alfadhel M. Biallelic SCN2A gene mutation causing early infantile epileptic encephalopathy: case report and review. J Cent Nerv Syst Dis 2019; 11: 1179573519849938
  • 56 Huang Q, Yu L, Ma M, Qi H, Wu Y. Novel SCN2A mutation in a family associated with juvenile-onset myoclonus: case report. Medicine (Baltimore) 2019; 98 (08) e14698
  • 57 Welzel T, Ziesenitz VC, Waldvogel S. et al. Use of a personalized phenytoin dosing approach to manage difficult to control seizures in an infant with a SCN2A mutation. Eur J Clin Pharmacol 2019; 75 (05) 737-739
  • 58 Kong Y, Yan K, Hu L. et al. Data on mutations and clinical features in SCN1A or SCN2A gene. Data Brief 2018; 22: 492-501
  • 59 Zara F, Specchio N, Striano P. et al. Genetic testing in benign familial epilepsies of the first year of life: clinical and diagnostic significance. Epilepsia 2013; 54 (03) 425-436
  • 60 Reynolds C, King MD, Gorman KM. The phenotypic spectrum of SCN2A-related epilepsy. Eur J Paediatr Neurol 2020; 24: 117-122
  • 61 Nakamura K, Kato M, Osaka H. et al. Clinical spectrum of SCN2A mutations expanding to Ohtahara syndrome. Neurology 2013; 81 (11) 992-998
  • 62 Barcia G, Chemaly N, Kuchenbuch M. et al. Epilepsy with migrating focal seizures: KCNT1 mutation hotspots and phenotype variability. Neurol Genet 2019; 5 (06) e363
  • 63 Barcia G, Fleming MR, Deligniere A. et al. De novo gain-of-function KCNT1 channel mutations cause malignant migrating partial seizures of infancy. Nat Genet 2012; 44 (11) 1255-1259
  • 64 Parrini E, Marini C, Mei D. et al; Clinical Study Group. Diagnostic targeted resequencing in 349 patients with drug-resistant pediatric epilepsies identifies causative mutations in 30 different genes. Hum Mutat 2017; 38 (02) 216-225
  • 65 Berg AT, Palac H, Wilkening G, Zelko F, Schust Meyer L. SCN2A-developmental and epileptic encephalopathies: challenges to trial-readiness for non-seizure outcomes. Epilepsia 2021; 62 (01) 258-268
  • 66 Howell KB, McMahon JM, Carvill GL. et al. SCN2A encephalopathy: a major cause of epilepsy of infancy with migrating focal seizures. Neurology 2015; 85 (11) 958-966
  • 67 Kruth KA, Grisolano TM, Ahern CA, Williams AJ. SCN2A channelopathies in the autism spectrum of neuropsychiatric disorders: a role for pluripotent stem cells?. Mol Autism 2020; 11 (01) 23
  • 68 Lionetti E, Leonardi S, Franzonello C, Mancardi M, Ruggieri M, Catassi C. Gluten psychosis: confirmation of a new clinical entity. Nutrients 2015; 7 (07) 5532-5539
  • 69 Ruggieri M, Praticò AD, Serra A. et al. Childhood neurofibromatosis type 2 (NF2) and related disorders: from bench to bedside and biologically targeted therapies. Acta Otorhinolaryngol Ital 2016; 36 (05) 345-367
  • 70 Wang T, Guo H, Xiong B. et al. De novo genic mutations among a Chinese autism spectrum disorder cohort. Nat Commun 2016; 7: 13316
  • 71 Schwarz N, Bast T, Gaily E. et al. Clinical and genetic spectrum of SCN2A-associated episodic ataxia. Eur J Paediatr Neurol 2019; 23 (03) 438-447
  • 72 Vidal S, Brandi N, Pacheco P. et al; Rett Working Group. The most recurrent monogenic disorders that overlap with the phenotype of Rett syndrome. Eur J Paediatr Neurol 2019; 23 (04) 609-620
  • 73 Trump N, McTague A, Brittain H. et al. Improving diagnosis and broadening the phenotypes in early-onset seizure and severe developmental delay disorders through gene panel analysis. J Med Genet 2016; 53 (05) 310-317
  • 74 Pratico AD, Falsaperla R, Ruggieri M, Corsello G, Pavone P. Prognostic challenges of SCN1A genetic mutations: report on two children with mild features. J Pediatr Neurol 2016; 14: 82-88
  • 75 Oates S, Tang S, Rosch R. et al. Incorporating epilepsy genetics into clinical practice: a 360°evaluation. NPJ Genom Med 2018; 3: 13
  • 76 Lindy AS, Stosser MB, Butler E. et al. Diagnostic outcomes for genetic testing of 70 genes in 8565 patients with epilepsy and neurodevelopmental disorders. Epilepsia 2018; 59 (05) 1062-1071
  • 77 Sahli M, Zrhidri A, Elaloui SC. et al. Clinical exome sequencing identifies two novel mutations of the SCN1A and SCN2A genes in Moroccan patients with epilepsy: a case series. J Med Case Reports 2019; 13 (01) 266
  • 78 Calhoun JD, Hawkins NA, Zachwieja NJ, Kearney JA. Cacna1g is a genetic modifier of epilepsy caused by mutation of voltage-gated sodium channel Scn2a. Epilepsia 2016; 57 (06) e103-e107
  • 79 Hawkins NA, Kearney JA. Hlf is a genetic modifier of epilepsy caused by voltage-gated sodium channel mutations. Epilepsy Res 2016; 119: 20-23
  • 80 Thuresson AC, Van Buggenhout G, Sheth F. et al. Whole gene duplication of SCN2A and SCN3A is associated with neonatal seizures and a normal intellectual development. Clin Genet 2017; 91 (01) 106-110
  • 81 Pratico AD, Ruggieri M, Falsaperla R, Pavone P. A probable topiramate-induced limbs paraesthesia and rigid fingers flexion. Curr Drug Saf 2018; 13 (02) 131-136
  • 82 Pratico AD, Longo L, Mansueto S. et al. Off-label use of drugs and adverse drug reactions in pediatric units: a prospective, multicenter study. Curr Drug Saf 2018; 13 (03) 200-207
  • 83 Flor-Hirsch H, Heyman E, Livneh A. et al. Lacosamide for SCN2A-related intractable neonatal and infantile seizures. Epileptic Disord 2018; 20 (05) 440-446
  • 84 Baumer FM, Peters JM, El Achkar CM, Pearl PL. SCN2A-related early-onset epileptic encephalopathy responsive to phenobarbital. J Pediatr Epilepsy 2016; 5 (01) 42-46
  • 85 Wong VC, Fung CW, Kwong AK. SCN2A mutation in a Chinese boy with infantile spasm - response to modified Atkins Diet. Brain Dev 2015; 37 (07) 729-732
  • 86 Praticò AD, Pavone P, Scuderi MG. et al. Symptomatic hypocalcemia in an epileptic child treated with valproic acid plus lamotrigine: a case report. Cases J 2009; 2: 7394
  • 87 Ko A, Jung DE, Kim SH. et al. The efficacy of ketogenic diet for specific genetic mutation in developmental and epileptic encephalopathy. Front Neurol 2018; 9: 530
  • 88 Falsaperla R, D'Angelo G, Praticò AD. et al. Ketogenic diet for infants with epilepsy: a literature review. Epilepsy Behav 2020; 112: 107361
  • 89 Harivenkatesh N, Haribalaji N, David DC, Kumar CM. Therapeutic drug monitoring of antiepileptic drugs in a tertiary care hospital in India. Clin Neuropharmacol 2015; 38 (01) 1-5