Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Sep 14;145(9):2991-3009.
doi: 10.1093/brain/awab321.

Genotype-phenotype correlations in SCN8A-related disorders reveal prognostic and therapeutic implications

Katrine M Johannesen  1   2 Yuanyuan Liu  3 Mahmoud Koko  3 Cathrine E Gjerulfsen  1 Lukas Sonnenberg  3   4 Julian Schubert  3 Christina D Fenger  1 Ahmed Eltokhi  3 Maert Rannap  3 Nils A Koch  4 Stephan Lauxmann  3   4 Johanna Krüger  3 Josua Kegele  3 Laura Canafoglia  5 Silvana Franceschetti  5 Thomas Mayer  6 Johannes Rebstock  6 Pia Zacher  6 Susanne Ruf  7 Michael Alber  7 Katalin Sterbova  8 Petra Lassuthová  8 Marketa Vlckova  8 Johannes R Lemke  9 Konrad Platzer  9 Ilona Krey  9 Constanze Heine  9 Dagmar Wieczorek  10 Judith Kroell-Seger  11 Caroline Lund  12 Karl Martin Klein  13 P Y Billie Au  14 Jong M Rho  15 Alice W Ho  15 Silvia Masnada  16 Pierangelo Veggiotti  16   17 Lucio Giordano  18 Patrizia Accorsi  18 Christina E Hoei-Hansen  19   20 Pasquale Striano  21   22 Federico Zara  22 Helene Verhelst  23 Judith S Verhoeven  24 Hilde M H Braakman  25 Bert van der Zwaag  26 Aster V E Harder  26 Eva Brilstra  26 Manuela Pendziwiat  27 Sebastian Lebon  28   29 Maria Vaccarezza  30 Ngoc Minh Le  31 Jakob Christensen  32 Sabine Grønborg  33 Stephen W Scherer  34   35 Jennifer Howe  36 Walid Fazeli  37   38 Katherine B Howell  38   39   40 Richard Leventer  38   39   40 Chloe Stutterd  39   40 Sonja Walsh  41 Marion Gerard  42 Bénédicte Gerard  43 Sara Matricardi  44 Claudia M Bonardi  45 Stefano Sartori  46 Andrea Berger  47 Dorota Hoffman-Zacharska  48 Massimo Mastrangelo  49 Francesca Darra  50 Arve Vøllo  51 M Mahdi Motazacker  52 Phillis Lakeman  53 Mathilde Nizon  54 Cornelia Betzler  55   56 Cecilia Altuzarra  57 Roseline Caume  58 Agathe Roubertie  59 Philippe Gélisse  59 Carla Marini  60 Renzo Guerrini  61 Frederic Bilan  62 Daniel Tibussek  63 Margarete Koch-Hogrebe  64 M Scott Perry  65 Shoji Ichikawa  66 Elena Dadali  67   68 Artem Sharkov  68   69 Irina Mishina  67 Mikhail Abramov  68 Ilya Kanivets  70   71 Sergey Korostelev  70   72 Sergey Kutsev  67 Karen E Wain  73 Nancy Eisenhauer  73 Monisa Wagner  73 Juliann M Savatt  73 Karen Müller-Schlüter  74 Haim Bassan  75   76 Artem Borovikov  67 Marie Cecile Nassogne  77 Anne Destrée  78 An Sofie Schoonjans  79 Marije Meuwissen  80 Marga Buzatu  80 Anna Jansen  81 Emmanuel Scalais  82 Siddharth Srivastava  83 Wen Hann Tan  84 Heather E Olson  83   85 Tobias Loddenkemper  83 Annapurna Poduri  83   85 Katherine L Helbig  86   87 Ingo Helbig  86   87   88   89   90   91 Mark P Fitzgerald  86   87   88   90 Ethan M Goldberg  86   87 Timo Roser  92 Ingo Borggraefe  92   93 Tobias Brünger  94 Patrick May  95 Dennis Lal  94   96   97   98 Damien Lederer  78 Guido Rubboli  1   99 Henrike O Heyne  9   100   101   102 Gaetan Lesca  103   104 Ulrike B S Hedrich  3 Jan Benda  4 Elena Gardella  1   2 Holger Lerche  3 Rikke S Møller  1   2
Affiliations

Genotype-phenotype correlations in SCN8A-related disorders reveal prognostic and therapeutic implications

Katrine M Johannesen et al. Brain. .

Abstract

We report detailed functional analyses and genotype-phenotype correlations in 392 individuals carrying disease-causing variants in SCN8A, encoding the voltage-gated Na+ channel Nav1.6, with the aim of describing clinical phenotypes related to functional effects. Six different clinical subgroups were identified: Group 1, benign familial infantile epilepsy (n = 15, normal cognition, treatable seizures); Group 2, intermediate epilepsy (n = 33, mild intellectual disability, partially pharmaco-responsive); Group 3, developmental and epileptic encephalopathy (n = 177, severe intellectual disability, majority pharmaco-resistant); Group 4, generalized epilepsy (n = 20, mild to moderate intellectual disability, frequently with absence seizures); Group 5, unclassifiable epilepsy (n = 127); and Group 6, neurodevelopmental disorder without epilepsy (n = 20, mild to moderate intellectual disability). Those in Groups 1-3 presented with focal or multifocal seizures (median age of onset: 4 months) and focal epileptiform discharges, whereas the onset of seizures in patients with generalized epilepsy was later (median: 42 months) with generalized epileptiform discharges. We performed functional studies expressing missense variants in ND7/23 neuroblastoma cells and primary neuronal cultures using recombinant tetrodotoxin-insensitive human Nav1.6 channels and whole-cell patch-clamping. Two variants causing developmental and epileptic encephalopathy showed a strong gain-of-function (hyperpolarizing shift of steady-state activation, strongly increased neuronal firing rate) and one variant causing benign familial infantile epilepsy or intermediate epilepsy showed a mild gain-of-function (defective fast inactivation, less increased firing). In contrast, all three variants causing generalized epilepsy induced a loss-of-function (reduced current amplitudes, depolarizing shift of steady-state activation, reduced neuronal firing). Functional effects were known for 170 individuals. All 136 individuals carrying a functionally tested gain-of-function variant had either focal (n = 97, Groups 1-3) or unclassifiable (n = 39) epilepsy, whereas 34 individuals with a loss-of-function variant had either generalized (n = 14), no (n = 11) or unclassifiable (n = 6) epilepsy; only three had developmental and epileptic encephalopathy. Computational modelling in the gain-of-function group revealed a significant correlation between the severity of the electrophysiological and clinical phenotypes. Gain-of-function variant carriers responded significantly better to sodium channel blockers than to other anti-seizure medications, and the same applied for all individuals in Groups 1-3. In conclusion, our data reveal clear genotype-phenotype correlations between age at seizure onset, type of epilepsy and gain- or loss-of-function effects of SCN8A variants. Generalized epilepsy with absence seizures is the main epilepsy phenotype of loss-of-function variant carriers and the extent of the electrophysiological dysfunction of the gain-of-function variants is a main determinant of the severity of the clinical phenotype in focal epilepsies. Our pharmacological data indicate that sodium channel blockers present a treatment option in SCN8A-related focal epilepsy with onset in the first year of life.

Keywords: SCN8A; epilepsy; genetics; personalized medicine.

PubMed Disclaimer

Conflict of interest statement

The authors report no competing interests.

Figures

Figure 1
Figure 1
Functional characterizations of SCN8A variants in the neuroblastoma cell line ND7/23. Wild-type or mutant Nav1.6 channels were transfected into ND7/23 cells and recorded in the presence of tetrodotoxin to block endogenous Na+ channels. (A) Representative Na+ current traces for transfected Nav1.6 wild-type (WT, black) or mutant channels (colour code in the bottom right). (B) Peak Na+ currents normalized by cell capacitance were plotted versus voltage. Both the p.(Phe846Ser) and p.(Leu840Pro) variants caused a hyperpolarizing shift of the current-voltage relationship, whereas the p.(Val1758Ala) and p.(Thr1787Pro) variants caused a depolarizing shift compared with the wild-type channels. The p.(Ile1654Asn), p.(Val1758Ala) and p.(Thr1787Pro) variants significantly decreased the current density in comparison with the wild-type. Wild-type: n = 30; mutants: n = 14–19. (C) Voltage-dependent steady-state activation curves. Lines represent Boltzmann functions fit to the data-points. (D) Voltage-dependent steady-state inactivation curves. Lines represent Boltzmann functions fit to the data-points. (E) Time course of recovery from fast inactivation at −100 mV. The p.(Val1758Ala), p.(Thr1787Pro) and p.(Asn374Lys) variants accelerated the recovery from fast inactivation compared with the wild-type. (F) Voltage-dependence of the major time constant of fast inactivation τh. Shown are means ± SEM (BF). Numbers of recorded cells and statistical analysis for all experiments are provided in Supplementary Table 2.
Figure 2
Figure 2
Effects of SCN8A variants in primary cultured hippocampal mouse neurons in the absence of tetrodotoxin. Neurons were transfected with wild-type or mutant Nav1.6 channels and recorded in the absence of tetrodotoxin. (A) Representative voltage traces of evoked action potentials (APs) in the absence of tetrodotoxin from neurons transfected with wild-type (WT, black) or mutant neurons (colour code indicated in B). (B) Numbers of evoked action potentials plotted versus injected current in the absence of tetrodotoxin. Shown are means ± SEM. (C) Area under the curve for the input-output relationships. The p.(Thr1787Pro) variant shows a significantly decreased area under the curve compared with the wild-type channels. Rheobase (D) and threshold (E) of action potentials were decreased for neurons transfected with the p.(Phe846Ser) variant but increased for neurons transfected with the p.(Thr1787Pro) variant compared with the wild-type channels. Box-and-whisker plots (CE) show means (plus sign), the 25th, 50th and 75th percentiles, minima and maxima; *P < 0.05; **P < 0.01; ***P < 0.001; one-way ANOVA with Dunnett’s post hoc test or ANOVA on ranks with Dunn’s post hoc test were performed. Numbers of recorded cells and statistical analysis are provided in Supplementary Table 3.
Figure 3
Figure 3
Neuronal properties carried only by transfected wild-type or mutant Nav1.6 channels. Hippocampal neurons transfected with wild-type or mutant Nav1.6 channels were recorded in the presence of tetrodotoxin to block endogenous Na+ channels. (A) Representative voltage traces of evoked action potentials (APs) from neurons transfected with wild-type (black) or mutant channels (colour code in the bottom left). (B) Numbers of evoked action potentials plotted versus injected current in the presence of tetrodotoxin. Shown are means ± SEM. (C) Area under the curve for the input-output relationships. (D) Peak Na+ current amplitudes of neurons transfected with wild-type or mutant Nav1.6 channels in the presence of tetrodotoxin. Rheobase (E) and threshold (F) were significantly decreased in neurons transfected with p.(Phe846Ser) or p.(Leu840Pro) variants compared with the wild-type channels. The p.(Asn1877Ser) variant also significantly decreased the rheobase. The rheobase or threshold could not be obtained in neurons transfected with p.(Asn374Lys), p.(Val1758Ala), p.(Thr1787Pro) and p.(Ile 1654Asn) mutant channels due to very few evoked action potentials. Box-and-whisker plots (CF) show means (plus symbol), the 25th, 50th and 75th percentiles, minima and maxima; *P < 0.05; **P < 0.01; ***P < 0.001; one-way ANOVA with Dunnett’s post hoc test or ANOVA on ranks with Dunn’s post hoc test were performed. The numbers of recorded cells and statistical analysis are provided in Supplementary Table 4.
Figure 4
Figure 4
Distribution of affected individuals carrying SCN8A variants according to phenotype and age of seizure onset. Phenotypic subgroups of the total cohort (A) and individuals carrying LOF (B) or GOF (C) variants. (A) In the total cohort, 225 affected individuals (57.4%) had BFIE, intermediate epilepsy or DEE; 20 individuals (5.1%) had generalized epilepsy; and 20 individuals (5.1%) had a NDDwoE. (B) Twenty-five affected individuals had generalized epilepsy or an NDDwoE, accounting for 73.5% of LOF variant carriers, whereas 71.6% of GOF variant carriers had BFIE, intermediate epilepsy or DEE (C). Histogram of the age at seizure onset in affected individuals with BFIE (D), intermediate epilepsy (E), DEE (F) and generalized epilepsy (G). (H) Stacked histogram of age at seizure onset in affected individuals with focal epilepsy (FE) carrying GOF (FE+GOF), LOF (FE+LOF) or SCN8A variants which are not functionally characterized. (I) Stacked histogram of age at seizure onset in affected individuals with generalized epilepsy (GE) carrying LOF (GE+LOF) or SCN8A variants which are not functionally characterized. (J) Stacked histogram of age at seizure onset in affected individuals carrying GOF variants with FE (FE+GOF) or unclassifiable epilepsy (UE+GOF). (K) Stacked histogram of age at seizure onset in affected individuals carrying LOF variants with generalized epilepsy (GE+LOF), FE (FE+LOF) or unclassifiable epilepsy (UE+GOF). Affected individuals with BNIE, intermediate epilepsy or DEE exhibited a significant earlier age at seizure onset than individuals with generalized epilepsy (Kruskal-Wallis test, P < 0.001), which is also observed for GOF versus LOF variant carriers (Mann-Whitney test, P < 0.001). Histogram bin size = 1 month. Seizure type distributions of affected individuals with FE versus generalized epilepsy (L) and GOF versus LOF variant carriers (M).
Figure 5
Figure 5
Correlation of a computed electrophysiological score with the clinical severity of SCN8A GOF variants. Electrophysiological scores of SCN8A GOF variants were obtained according to the effect of variants on action potential firing simulated by a single-compartment conductance-based model. (AF) The contrast of the simulated area under the input-output curve (AUC, Equation 1) as a function of the changes in single Na+ current gating parameters, such as: (A) the V1/2 of the activation curve; (B) the slope factor k of the activation curve; (C) the V1/2 of the fast inactivation curve; (D) the slope factor k of the fast inactivation curve; (E) the persistent Na+ current; and (F) the Na+ conductivity (or current density). Changes in the V1/2 and the slope of the activation curve had a much stronger effect on the AUC than other parameters. (G) Correlation (dashed grey line) of the simulation-based score (Equation 9) with the severity of each SCN8A GOF variant averaged over affected individuals (blue dots with the respective one-amino acid code). (H) Distributions of simulation-based scores of each patient (blue dots) for each of the four categories of clinical severities. *P < 0.05; ***P < 0.001 (ANOVA on ranks with Dunn’s post hoc test).
Figure 6
Figure 6
Location of SCN8A variants associated with neurodevelopmental disorders. Schematic 2D representation of the Nav1.6 channel displaying the location of pathogenic variants. A comparison of the location of missense variants with proven GOF or a BFIE/intermediate epilepsy/DEE phenotype (without functional analysis) on one hand, and variants with proven LOF or generalized epilepsy/NDDwoE phenotypes (without functional analysis) on the other, revealed a significant difference in the distribution of variants (see main text and Supplementary material). Recurring variants are indicated with larger symbol size relative to the number of patients and in frame indels or deletions are indicated showing the whole affected regions [p.(Ile888_Val892delinsMet), p.(Glu1774_Ala1777del) and p.(Pro1428_Lys1473del)].
Figure 7
Figure 7
Treatment responses to anti-seizure medications in affected individuals carrying SCN8A variants. Treatment effects of ASMs on seizures in affected individuals with BFIE, intermediate epilepsy or DEE versus those with generalized epilepsy (A) and those carrying SCN8A GOF versus LOF variants (B). Phenytoin, carbamazepine, lamotrigine, oxcarbazepine, zonisamide and lacosamide are SCBs. (C and D) Individuals with BFIE, intermediate epilepsy or DEE and those carrying SCN8A GOF variants responded significantly better to SCBs than non-SCBs, whereas treatment with SCBs or non-SCBs did not cause a different effect in individuals with generalized epilepsy and those carrying SCN8A LOF variants (please consider the very small numbers in these latter categories). P values derived from Fisher’s exact test are provided in the figure. Responders were defined as those who became seizure-free or experienced a seizure reduction while staying on the drug; non-responders were defined as those experiencing no effect or seizure worsening. CBZ = carbamazepine; CLB = clobazam; CLZ = clonazepam; ESM = ethosuximide; KD = ketogenic diet; LCM = lacosamide; LEV = levetiracetam; LTG = lamotrigine; OXC= oxcarbazepine; PB = phenobarbital; PHT = phenytoin; TPM = topiramate; VGB = vigabatrin; VPA = valproate; ZNS = zonisamide.

Similar articles

  • Biological concepts in human sodium channel epilepsies and their relevance in clinical practice.
    Brunklaus A, Du J, Steckler F, Ghanty II, Johannesen KM, Fenger CD, Schorge S, Baez-Nieto D, Wang HR, Allen A, Pan JQ, Lerche H, Heyne H, Symonds JD, Zuberi SM, Sanders S, Sheidley BR, Craiu D, Olson HE, Weckhuysen S, DeJonge P, Helbig I, Van Esch H, Busa T, Milh M, Isidor B, Depienne C, Poduri A, Campbell AJ, Dimidschstein J, Møller RS, Lal D. Brunklaus A, et al. Epilepsia. 2020 Mar;61(3):387-399. doi: 10.1111/epi.16438. Epub 2020 Feb 23. Epilepsia. 2020. PMID: 32090326
  • Phenotypic and genetic spectrum of SCN8A-related disorders, treatment options, and outcomes.
    Gardella E, Møller RS. Gardella E, et al. Epilepsia. 2019 Dec;60 Suppl 3:S77-S85. doi: 10.1111/epi.16319. Epilepsia. 2019. PMID: 31904124
  • Genetic and phenotypic heterogeneity suggest therapeutic implications in SCN2A-related disorders.
    Wolff M, Johannesen KM, Hedrich UBS, Masnada S, Rubboli G, Gardella E, Lesca G, Ville D, Milh M, Villard L, Afenjar A, Chantot-Bastaraud S, Mignot C, Lardennois C, Nava C, Schwarz N, Gérard M, Perrin L, Doummar D, Auvin S, Miranda MJ, Hempel M, Brilstra E, Knoers N, Verbeek N, van Kempen M, Braun KP, Mancini G, Biskup S, Hörtnagel K, Döcker M, Bast T, Loddenkemper T, Wong-Kisiel L, Baumeister FM, Fazeli W, Striano P, Dilena R, Fontana E, Zara F, Kurlemann G, Klepper J, Thoene JG, Arndt DH, Deconinck N, Schmitt-Mechelke T, Maier O, Muhle H, Wical B, Finetti C, Brückner R, Pietz J, Golla G, Jillella D, Linnet KM, Charles P, Moog U, Õiglane-Shlik E, Mantovani JF, Park K, Deprez M, Lederer D, Mary S, Scalais E, Selim L, Van Coster R, Lagae L, Nikanorova M, Hjalgrim H, Korenke GC, Trivisano M, Specchio N, Ceulemans B, Dorn T, Helbig KL, Hardies K, Stamberger H, de Jonghe P, Weckhuysen S, Lemke JR, Krägeloh-Mann I, Helbig I, Kluger G, Lerche H, Møller RS. Wolff M, et al. Brain. 2017 May 1;140(5):1316-1336. doi: 10.1093/brain/awx054. Brain. 2017. PMID: 28379373
  • SCN8A-Related Epilepsy and/or Neurodevelopmental Disorders.
    Hammer MF, Xia M, Schreiber JM. Hammer MF, et al. 2016 Aug 25 [updated 2023 Apr 6]. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2024. 2016 Aug 25 [updated 2023 Apr 6]. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2024. PMID: 27559564 Free Books & Documents. Review.
  • Gain of function SCN1A disease-causing variants: Expanding the phenotypic spectrum and functional studies guiding the choice of effective antiseizure medication.
    Matricardi S, Cestèle S, Trivisano M, Kassabian B, Leroudier N, Vittorini R, Nosadini M, Cesaroni E, Siliquini S, Marinaccio C, Longaretti F, Podestà B, Operto FF, Luisi C, Sartori S, Boniver C, Specchio N, Vigevano F, Marini C, Mantegazza M. Matricardi S, et al. Epilepsia. 2023 May;64(5):1331-1347. doi: 10.1111/epi.17509. Epub 2023 Jan 26. Epilepsia. 2023. PMID: 36636894 Review.

Cited by

  • Sex differences in physiological response to increased neuronal excitability in a knockin mouse model of pediatric epilepsy.
    Hammer MF, Krzyzaniak CT, Bahramnejad E, Smelser KJ, Hack JB, Watkins JC, Ronaldson PT. Hammer MF, et al. Clin Sci (Lond). 2024 Feb 21;138(4):205-223. doi: 10.1042/CS20231572. Clin Sci (Lond). 2024. PMID: 38348743 Free PMC article.
  • Clinical and electrophysiological features of SCN8A variants causing episodic or chronic ataxia.
    Lyu H, Boßelmann CM, Johannesen KM, Koko M, Ortigoza-Escobar JD, Aguilera-Albesa S, Garcia-Navas Núñez D, Linnankivi T, Gaily E, van Ruiten HJA, Richardson R, Betzler C, Horvath G, Brilstra E, Geerdink N, Orsucci D, Tessa A, Gardella E, Fleszar Z, Schöls L, Lerche H, Møller RS, Liu Y. Lyu H, et al. EBioMedicine. 2023 Dec;98:104855. doi: 10.1016/j.ebiom.2023.104855. Epub 2023 Oct 28. EBioMedicine. 2023. PMID: 38251463 Free PMC article.
  • Expanding the genotype-phenotype spectrum in SCN8A-related disorders.
    Hebbar M, Al-Taweel N, Gill I, Boelman C, Dean RA, Goodchild SJ, Mezeyova J, Shuart NG, Johnson JP Jr, Lee J, Michoulas A, Huh LL, Armstrong L, Connolly MB, Demos MK. Hebbar M, et al. BMC Neurol. 2024 Jan 17;24(1):31. doi: 10.1186/s12883-023-03478-y. BMC Neurol. 2024. PMID: 38233770 Free PMC article.
  • Follow the allosteric transitions to predict variant pathogenicity: a channel-specific approach.
    Absalom NL, El-Kamand S, Chua HC, Ahring PK. Absalom NL, et al. Brain. 2024 May 3;147(5):e37-e40. doi: 10.1093/brain/awae008. Brain. 2024. PMID: 38198781 Free PMC article. No abstract available.
  • Gain-of-function and loss-of-function variants in GRIA3 lead to distinct neurodevelopmental phenotypes.
    Rinaldi B, Bayat A, Zachariassen LG, Sun JH, Ge YH, Zhao D, Bonde K, Madsen LH, Awad IAA, Bagiran D, Sbeih A, Shah SM, El-Sayed S, Lyngby SM, Pedersen MG, Stenum-Berg C, Walker LC, Krey I, Delahaye-Duriez A, Emrick LT, Sully K, Murali CN, Burrage LC, Plaud Gonzalez JA, Parnes M, Friedman J, Isidor B, Lefranc J, Redon S, Heron D, Mignot C, Keren B, Fradin M, Dubourg C, Mercier S, Besnard T, Cogne B, Deb W, Rivier C, Milani D, Bedeschi MF, Di Napoli C, Grilli F, Marchisio P, Koudijs S, Veenma D, Argilli E, Lynch SA, Au PYB, Ayala Valenzuela FE, Brown C, Masser-Frye D, Jones M, Patron Romero L, Li WL, Thorpe E, Hecher L, Johannsen J, Denecke J, McNiven V, Szuto A, Wakeling E, Cruz V, Sency V, Wang H, Piard J, Kortüm F, Herget T, Bierhals T, Condell A, Ben-Zeev B, Kaur S, Christodoulou J, Piton A, Zweier C, Kraus C, Micalizzi A, Trivisano M, Specchio N, Lesca G, Møller RS, Tümer Z, Musgaard M, Gerard B, Lemke JR, Shi YS, Kristensen AS. Rinaldi B, et al. Brain. 2024 May 3;147(5):1837-1855. doi: 10.1093/brain/awad403. Brain. 2024. PMID: 38038360

References

    1. Veeramah KR, O'Brien JE, Meisler MH, et al. De novo pathogenic SCN8A mutation identified by whole-genome sequencing of a family quartet affected by infantile epileptic encephalopathy and SUDEP. Am J Hum Genet. 2012;90(3):502–510. - PMC - PubMed
    1. Anand G, Collett-White F, Orsini A, et al. Autosomal dominant SCN8A mutation with an unusually mild phenotype. Eur J Paediatr Neurol. 2016;20(5):761–765. - PubMed
    1. Gardella E, Becker F, Moller RS, et al. Benign infantile seizures and paroxysmal dyskinesia caused by an SCN8A mutation. Ann Neurol. 2016;79(3):428–436. - PubMed
    1. Han JY, Jang JH, Lee IG, Shin S, Park J. A novel inherited mutation of SCN8A in a Korean family with benign familial infantile epilepsy using diagnostic exome sequencing. Ann Clin Lab Sci. 2017;47:747–753. - PubMed
    1. Johannesen KM, Gardella E, Encinas AC, et al. The spectrum of intermediate SCN8A-related epilepsy. Epilepsia. 2019;60(5):830–844. - PubMed

Publication types

Substances