Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

From molecules to medicines: the dawn of targeted therapies for genetic epilepsies

Abstract

Precision medicine is the treatment of patients with therapy targeted to their specific pathophysiology. This lofty ideal currently has limited application in clinical practice. However, new technological advances in epilepsy models and genomics suggest that the precision medicine revolution is closer than ever before. We are gaining an improved understanding of the true complexity underlying the pathophysiology of genetic epilepsies and the sources of phenotypic variation that continue to frustrate efforts at genotype–phenotype correlation. Conventional experimental models of epilepsy, such as mouse models and heterologous expression systems, have provided many of the advances in our understanding of genetic epilepsies, but fail to account for some of these complexities. Novel high-throughput models of epilepsy such as zebrafish and induced pluripotent stems cells can be combined with CRISPR–Cas9 gene editing techniques to explore the pathogenesis of a specific gene change and rapidly screen drug libraries for potential therapeutics. The knowledge gained from these models must be combined with thorough natural history studies to determine appropriate patient populations for pragmatic clinical trials. Advances in the ‘omics’, genetic epilepsy models and deep-phenotyping techniques have revolutionary translational research potential that can bring precision medicine to the forefront of clinical practice in the coming decade.

Key points

  • Efforts to develop precision medicine for epilepsy have been limited by the complexity of the pathophysiology underlying genotype–phenotype correlations.

  • A complete picture of genetic epilepsy pathogenesis recognizes the contribution of intragenic, cellular and network-level variability that is influenced by genomic and environmental differences between patients.

  • Conventional models of epilepsy (heterologous expression systems and mouse models) can address some of these layers of complexity but fail to address others.

  • New models of epilepsy (such as zebrafish and induced pluripotent stem cells) offer insights that were previously not possible.

  • Complementary preclinical approaches that include both conventional and novel models of epilepsy have the power to advance research in precision medicine like never before.

  • The combination of these robust preclinical developments with collaborative clinical research, including deep phenotyping and pragmatic clinical trials, has the potential to improve precision treatment of the genetic epilepsies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The monogenic–polygenic spectrum.
Fig. 2: Sources of phenotypic variation in genetic epilepsies.
Fig. 3: Example of a national precision medicine model.

Similar content being viewed by others

References

  1. Online Mendelian Inheritance in Man. OMIM Entry Search–epilepsy. OMIM https://www.omim.org/search/?index=entry&sort=score+desc%2C+prefix_sort+desc&start=1&limit=10&search=epilepsy (2017).

  2. Hutchison, C. A. III. DNA sequencing: bench to bedside and beyond. Nucleic Acids Res. 35, 6227–6237 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Greenberg, D. A. & Subaran, R. Blinders, phenotype, and fashionable genetic analysis: a critical examination of the current state of epilepsy genetic studies. Epilepsia 52, 1–9 (2011).

    PubMed  Google Scholar 

  4. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    CAS  PubMed  Google Scholar 

  5. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    CAS  PubMed  Google Scholar 

  6. Chamberlain, S. J., Li, X. J. & Lalande, M. Induced pluripotent stem (iPS) cells as in vitro models of human neurogenetic disorders. Neurogenetics 9, 227–235 (2008).

    PubMed  Google Scholar 

  7. Smith, L. A. et al. A model program for translational medicine in epilepsy genetics. J. Child Neurol. 32, 429–436 (2017).

    PubMed  PubMed Central  Google Scholar 

  8. Gussow, A. B., Petrovski, S., Wang, Q., Allen, A. S. & Goldstein, D. B. The intolerance to functional genetic variation of protein domains predicts the localization of pathogenic mutations within genes. Genome Biol. 17, 9 (2016).

    PubMed  PubMed Central  Google Scholar 

  9. Petrovski, S., Wang, Q., Heinzen, E. L., Allen, A. S. & Goldstein, D. B. Genic intolerance to functional variation and the interpretation of personal genomes. PLOS Genet. 9, e1003709 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Millichap, J. J. et al. KCNQ2 encephalopathy: features, mutational hot spots, and ezogabine treatment of 11 patients. Neurol. Genet. 2, e96 (2016).

    PubMed  PubMed Central  Google Scholar 

  11. Miceli, F. et al. Genotype-phenotype correlations in neonatal epilepsies caused by mutations in the voltage sensor of K(v)7.2 potassium channel subunits. Proc. Natl Acad. Sci. USA 110, 4386–4391 (2013).

    CAS  PubMed  Google Scholar 

  12. Soldovieri, M. V. et al. Novel KCNQ2 and KCNQ3 mutations in a large cohort of families with benign neonatal epilepsy: first evidence for an altered channel regulation by syntaxin-1A. Hum. Mutat. 35, 356–367 (2014).

    CAS  PubMed  Google Scholar 

  13. Miceli, F. et al. Early-onset epileptic encephalopathy caused by gain-of-function mutations in the voltage sensor of Kv7.2 and Kv7.3 potassium channel subunits. J. Neurosci. 35, 3782–3793 (2015).

    CAS  PubMed  Google Scholar 

  14. Mulkey, S. B. et al. Neonatal nonepileptic myoclonus is a prominent clinical feature of KCNQ2 gain-of-function variants R201C and R201H. Epilepsia 58, 436–445 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Millichap, J. J. et al. Infantile spasms and encephalopathy without preceding neonatal seizures caused by KCNQ2 R198Q, a gain-of-function variant. Epilepsia 58, e10–e15 (2017).

    CAS  PubMed  Google Scholar 

  16. Kole, M. H. & Cooper, E. C. Axonal Kv7.2/7.3 channels: caught in the act. Channels (Austin) 8, 288–289 (2014).

    Google Scholar 

  17. Kanaumi, T. et al. Developmental changes in KCNQ2 and KCNQ3 expression in human brain: possible contribution to the age-dependent etiology of benign familial neonatal convulsions. Brain Dev. 30, 362–369 (2008).

    PubMed  Google Scholar 

  18. Safiulina, V. F., Zacchi, P., Taglialatela, M., Yaari, Y. & Cherubini, E. Low expression of Kv7/M channels facilitates intrinsic and network bursting in the developing rat hippocampus. J. Physiol. 586, 5437–5453 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Rubinstein, M. et al. Genetic background modulates impaired excitability of inhibitory neurons in a mouse model of Dravet syndrome. Neurobiol. Dis. 73, 106–117 (2015).

    PubMed  Google Scholar 

  20. Casillas-Espinosa, P. M. et al. Evaluating whole genome sequence data from the genetic absence epilepsy rat from Strasbourg and its related non-epileptic strain. PLOS ONE 12, e0179924 (2017).

    PubMed  PubMed Central  Google Scholar 

  21. Weaving, L. S. et al. Mutations of CDKL5 cause a severe neurodevelopmental disorder with infantile spasms and mental retardation. Am. J. Hum. Genet. 75, 1079–1093 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Valli, E. et al. CDKL5, a novel MYCN-repressed gene, blocks cell cycle and promotes differentiation of neuronal cells. Biochim. Biophys. Acta 1819, 1173–1185 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhu, Y. C. et al. Palmitoylation-dependent CDKL5-PSD-95 interaction regulates synaptic targeting of CDKL5 and dendritic spine development. Proc. Natl Acad. Sci. USA 110, 9118–9123 (2013).

    CAS  PubMed  Google Scholar 

  24. Fuchs, C. et al. Loss of CDKL5 impairs survival and dendritic growth of newborn neurons by altering AKT/GSK-3beta signaling. Neurobiol. Dis. 70, 53–68 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Livide, G. et al. GluD1 is a common altered player in neuronal differentiation from both MECP2-mutated and CDKL5-mutated iPS cells. Eur. J. Hum. Genet. 23, 195–201 (2015).

    CAS  PubMed  Google Scholar 

  26. Della Sala, G. et al. Dendritic spine instability in a mouse model of CDKL5 disorder is rescued by insulin-like growth factor 1. Biol. Psychiatry 80, 302–311 (2016).

    PubMed  Google Scholar 

  27. Hector, R. D. et al. Characterisation of CDKL5 transcript isoforms in human and mouse. PLOS ONE 11, e0157758 (2016).

    PubMed  PubMed Central  Google Scholar 

  28. Nawaz, M. S. et al. CDKL5 and Shootin1 interact and concur in regulating neuronal polarization. PLOS ONE 11, e0148634 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Sivilia, S. et al. CDKL5 knockout leads to altered inhibitory transmission in the cerebellum of adult mice. Genes Brain Behav. 15, 491–502 (2016).

    CAS  PubMed  Google Scholar 

  30. Bahi-Buisson, N. & Bienvenu, T. CDKL5-related disorders: from clinical description to molecular genetics. Mol. Syndromol. 2, 137–152 (2011).

    PubMed  PubMed Central  Google Scholar 

  31. Bahi-Buisson, N. et al. Recurrent mutations in the CDKL5 gene: genotype-phenotype relationships. Am. J. Med. Genet. A 158A, 1612–1619 (2012).

    PubMed  Google Scholar 

  32. Moseley, B. D., Dhamija, R., Wirrell, E. C. & Nickels, K. C. Historic, clinical, and prognostic features of epileptic encephalopathies caused by CDKL5 mutations. Pediatr. Neurol. 46, 101–105 (2012).

    PubMed  Google Scholar 

  33. Fehr, S. et al. There is variability in the attainment of developmental milestones in the CDKL5 disorder. J. Neurodev. Disord. 7, 2 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Fehr, S. et al. Seizure variables and their relationship to genotype and functional abilities in the CDKL5 disorder. Neurology 87, 2206–2213 (2016).

    CAS  PubMed  Google Scholar 

  35. Ishii, A., Watkins, J. C., Chen, D., Hirose, S. & Hammer, M. F. Clinical implications of SCN1A missense and truncation variants in a large Japanese cohort with Dravet syndrome. Epilepsia 58, 282–290 (2017).

    CAS  PubMed  Google Scholar 

  36. Dilena, R. et al. Efficacy of sodium channel blockers in SCN2A early infantile epileptic encephalopathy. Brain Dev. 39, 345–348 (2017).

    PubMed  Google Scholar 

  37. Wolff, M. et al. Genetic and phenotypic heterogeneity suggest therapeutic implications in SCN2A-related disorders. Brain 140, 1316–1336 (2017).

    PubMed  Google Scholar 

  38. Yamagata, T., Ogiwara, I., Mazaki, E., Yanagawa, Y. & Yamakawa, K. Nav1.2 is expressed in caudal ganglionic eminence-derived disinhibitory interneurons: mutually exclusive distributions of Nav1.1 and Nav1.2. Biochem. Biophys. Res. Commun. 491, 1070–1076 (2017).

    CAS  PubMed  Google Scholar 

  39. Tai, C., Abe, Y., Westenbroek, R. E., Scheuer, T. & Catterall, W. A. Impaired excitability of somatostatin- and parvalbumin-expressing cortical interneurons in a mouse model of Dravet syndrome. Proc. Natl Acad. Sci. USA 111, E3139–E3148 (2014).

    CAS  PubMed  Google Scholar 

  40. Kalume, F. et al. Sleep impairment and reduced interneuron excitability in a mouse model of Dravet Syndrome. Neurobiol. Dis. 77, 141–154 (2015).

    PubMed  PubMed Central  Google Scholar 

  41. Kurbatova, P. et al. Dynamic changes of depolarizing GABA in a computational model of epileptogenic brain: insight for Dravet syndrome. Exp. Neurol. 283, 57–72 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Milh, M. et al. Variable clinical expression in patients with mosaicism for KCNQ2 mutations. Am. J. Med. Genet. A 167, 2314–2318 (2015).

    CAS  Google Scholar 

  43. McElroy, P. B., Liang, L. P., Day, B. J. & Patel, M. Scavenging reactive oxygen species inhibits status epilepticus-induced neuroinflammation. Exp. Neurol. 298, 13–22 (2017).

    CAS  PubMed  Google Scholar 

  44. Pauletti, A. et al. Targeting oxidative stress improves disease outcomes in a rat model of acquired epilepsy. Brain 140, 1885–1899 (2017).

    PubMed  Google Scholar 

  45. Pearson-Smith, J. N., Liang, L. P., Rowley, S. D., Day, B. J. & Patel, M. Oxidative stress contributes to status epilepticus associated mortality. Neurochem. Res. 42, 2024–2032 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Pearson-Smith, J. N. & Patel, M. Metabolic dysfunction and oxidative stress in epilepsy. Int. J. Mol. Sci. 18, E2365 (2017).

    PubMed  Google Scholar 

  47. Kumar, M. G. et al. Altered glycolysis and mitochondrial respiration in a zebrafish model of Dravet syndrome. eNeuro https://doi.org/10.1523/eneuro.0008-16.2016 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Veeramah, K. R. 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. 90, 502–510 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Vaher, U. et al. De novo SCN8A mutation identified by whole-exome sequencing in a boy with neonatal epileptic encephalopathy, multiple congenital anomalies, and movement disorders. J. Child Neurol. 29, 202–206 (2014).

    Google Scholar 

  50. Wagnon, J. L. & Meisler, M. H. Recurrent and non-recurrent mutations of SCN8A in epileptic encephalopathy. Front. Neurol. 6, 104 (2015).

    PubMed  PubMed Central  Google Scholar 

  51. Wagnon, J. L. et al. Convulsive seizures and SUDEP in a mouse model of SCN8A epileptic encephalopathy. Hum. Mol. Genet. 24, 506–515 (2015).

    CAS  PubMed  Google Scholar 

  52. Lopez-Santiago, L. F. et al. Neuronal hyperexcitability in a mouse model of SCN8A epileptic encephalopathy. Proc. Natl Acad. Sci. USA 114, 2383–2388 (2017).

    CAS  PubMed  Google Scholar 

  53. Barcia, G. et al. De novo gain-of-function KCNT1 channel mutations cause malignant migrating partial seizures of infancy. Nat. Genet. 44, 1255–1259 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lim, C. X., Ricos, M. G., Dibbens, L. M. & Heron, S. E. KCNT1 mutations in seizure disorders: the phenotypic spectrum and functional effects. J. Med. Genet. 53, 217–225 (2016).

    CAS  PubMed  Google Scholar 

  55. Vanderver, A. et al. Identification of a novel de novo p. Phe932Ile KCNT1 mutation in a patient with leukoencephalopathy and severe epilepsy. Pediatr. Neurol. 50, 112–114 (2014).

    PubMed  Google Scholar 

  56. Evely, K. M., Pryce, K. D. & Bhattacharjee, A. The Phe932Ile mutation in KCNT1 channels associated with severe epilepsy, delayed myelination and leukoencephalopathy produces a loss-of-function channel phenotype. Neuroscience 351, 65–70 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Bearden, D. et al. Targeted treatment of migrating partial seizures of infancy with quinidine. Ann. Neurol. 76, 457–461 (2014).

    CAS  PubMed  Google Scholar 

  58. Milligan, C. J. et al. KCNT1 gain of function in 2 epilepsy phenotypes is reversed by quinidine. Ann. Neurol. 75, 581–590 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Mikati, M. A. et al. Quinidine in the treatment of KCNT1-positive epilepsies. Ann. Neurol. 78, 995–999 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Fukuoka, M. et al. Quinidine therapy for West syndrome with KCNTI mutation: a case report. Brain Dev. 39, 80–83 (2017).

    PubMed  Google Scholar 

  61. McTague, A. et al. Clinical and molecular characterization of KCNT1-related severe early-onset epilepsy. Neurology 90, e55–e66 (2017).

    PubMed  Google Scholar 

  62. Mullen, S. A. et al. Precision therapy for epilepsy due to KCNT1 mutations: a randomized trial of oral quinidine. Neurology 90, e67–e72 (2017).

    PubMed  Google Scholar 

  63. Chong, P. F., Nakamura, R., Saitsu, H., Matsumoto, N. & Kira, R. Ineffective quinidine therapy in early onset epileptic encephalopathy with KCNT1 mutation. Ann. Neurol. 79, 502–503 (2016).

    CAS  PubMed  Google Scholar 

  64. Griffin, A., Krasniak, C. & Baraban, S. C. Advancing epilepsy treatment through personalized genetic zebrafish models. Prog. Brain Res. 226, 195–207 (2016).

    CAS  PubMed  Google Scholar 

  65. Hong, S., Lee, P., Baraban, S. C. & Lee, L. P. A novel long-term, multi-channel and non-invasive electrophysiology platform for zebrafish. Sci. Rep. 6, 28248 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Meyer, M. et al. Microarray noninvasive neuronal seizure recordings from intact larval zebrafish. PLOS ONE 11, e0156498 (2016).

    PubMed  PubMed Central  Google Scholar 

  67. Hunyadi, B., Siekierska, A., Sourbron, J., Copmans, D. & de Witte, P. A. M. Automated analysis of brain activity for seizure detection in zebrafish models of epilepsy. J. Neurosci. Methods 287, 13–24 (2017).

    PubMed  Google Scholar 

  68. Turrini, L. et al. Optical mapping of neuronal activity during seizures in zebrafish. Sci. Rep. 7, 3025 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Baraban, S. C., Dinday, M. T. & Hortopan, G. A. Drug screening in Scn1a zebrafish mutant identifies clemizole as a potential Dravet syndrome treatment. Nat. Commun. 4, 2410 (2013).

    PubMed  PubMed Central  Google Scholar 

  70. Mahmood, F. et al. A zebrafish model of CLN2 disease is deficient in tripeptidyl peptidase 1 and displays progressive neurodegeneration accompanied by a reduction in proliferation. Brain 136, 1488–1507 (2013).

    PubMed  Google Scholar 

  71. Mahmood, F. et al. Generation and validation of a zebrafish model of EAST (epilepsy, ataxia, sensorineural deafness and tubulopathy) syndrome. Dis. Model. Mech. 6, 652–660 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Colin, E. et al. Biallelic variants in UBA5 reveal that disruption of the UFM1 cascade can result in early-onset encephalopathy. Am. J. Hum. Genet. 99, 695–703 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Cooper, S. R., Jontes, J. D. & Sotomayor, M. Structural determinants of adhesion by Protocadherin-19 and implications for its role in epilepsy. eLife 5, e18529 (2016).

    PubMed  PubMed Central  Google Scholar 

  74. Sicca, F. et al. Gain-of-function defects of astrocytic Kir4.1 channels in children with autism spectrum disorders and epilepsy. Sci. Rep. 6, 34325 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Wager, K. et al. Neurodegeneration and epilepsy in a zebrafish model of CLN3 disease (Batten disease). PLOS ONE 11, e0157365 (2016).

    PubMed  PubMed Central  Google Scholar 

  76. Anttonen, A. K. et al. ZNHIT3 is defective in PEHO syndrome, a severe encephalopathy with cerebellar granule neuron loss. Brain 140, 1267–1279 (2017).

    PubMed  Google Scholar 

  77. Cao, S. et al. Homozygous EEF1A2 mutation causes dilated cardiomyopathy, failure to thrive, global developmental delay, epilepsy and early death. Hum. Mol. Genet. 26, 3545–3552 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Pena, I. A. et al. Pyridoxine-dependent epilepsy in zebrafish caused by Aldh7a1 deficiency. Genetics 207, 1501–1518 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Scheldeman, C. et al. mTOR-related neuropathology in mutant tsc2 zebrafish: phenotypic, transcriptomic and pharmacological analysis. Neurobiol. Dis. 108, 225–237 (2017).

    CAS  PubMed  Google Scholar 

  80. Zabinyakov, N. et al. Characterization of the first knock-out aldh7a1 zebrafish model for pyridoxine-dependent epilepsy using CRISPR-Cas9 technology. PLOS ONE 12, e0186645 (2017).

    PubMed  PubMed Central  Google Scholar 

  81. Griffin, A. et al. Clemizole and modulators of serotonin signalling suppress seizures in Dravet syndrome. Brain 140, 669–683 (2017).

    PubMed  PubMed Central  Google Scholar 

  82. Schoonjans, A. et al. Low-dose fenfluramine significantly reduces seizure frequency in Dravet syndrome: a prospective study of a new cohort of patients. Eur. J. Neurol. 24, 309–314 (2017).

    CAS  PubMed  Google Scholar 

  83. Son, D. et al. Generation of two induced pluripotent stem cell (iPSC) lines from X-linked adrenoleukodystrophy (X-ALD) patients with adrenomyeloneuropathy (AMN). Stem Cell Res. 25, 46–49 (2017).

    CAS  PubMed  Google Scholar 

  84. Bershteyn, M. et al. Human iPSC-derived cerebral organoids model cellular features of lissencephaly and reveal prolonged mitosis of outer radial glia. Cell Stem Cell 20, 435–449.e4 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Marinowic, D. R. et al. Induced pluripotent stem cells from patients with focal cortical dysplasia and refractory epilepsy. Mol. Med. Rep. 15, 2049–2056 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Chen, W. et al. Generation of the SCN1A epilepsy mutation in hiPS cells using the TALEN technique. Sci. Rep. 4, 5404 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Maeda, H. et al. Establishment of isogenic iPSCs from an individual with SCN1A mutation mosaicism as a model for investigating neurocognitive impairment in Dravet syndrome. J. Hum. Genet. 61, 565–569 (2016).

    CAS  PubMed  Google Scholar 

  88. Sun, Y. et al. A deleterious Nav1.1 mutation selectively impairs telencephalic inhibitory neurons derived from Dravet Syndrome patients. eLife 5, e13073 (2016).

    PubMed  PubMed Central  Google Scholar 

  89. Yamashita, S. et al. Mislocalization of syntaxin-1 and impaired neurite growth observed in a human iPSC model for STXBP1-related epileptic encephalopathy. Epilepsia 57, e81–e86 (2016).

    CAS  PubMed  Google Scholar 

  90. Ricciardi, S. et al. CDKL5 ensures excitatory synapse stability by reinforcing NGL-1-PSD95 interaction in the postsynaptic compartment and is impaired in patient iPSC-derived neurons. Nat. Cell Biol. 14, 911–923 (2012).

    CAS  PubMed  Google Scholar 

  91. Spampanato, J., Escayg, A., Meisler, M. H. & Goldin, A. L. Functional effects of two voltage-gated sodium channel mutations that cause generalized epilepsy with febrile seizures plus type 2. J. Neurosci. 21, 7481–7490 (2001).

    CAS  PubMed  Google Scholar 

  92. Lossin, C. et al. Epilepsy-associated dysfunction in the voltage-gated neuronal sodium channel SCN1A. J. Neurosci. 23, 11289–11295 (2003).

    CAS  PubMed  Google Scholar 

  93. Liu, Y. et al. Dravet syndrome patient-derived neurons suggest a novel epilepsy mechanism. Ann. Neurol. 74, 128–139 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Tidball, A. M. et al. Rapid generation of human genetic loss-of-function iPSC lines by simultaneous reprogramming and gene editing. Stem Cell Rep. 9, 725–731 (2017).

    CAS  Google Scholar 

  95. Hadoux, J. et al. Transcriptional landscape of a RET(C634Y)-mutated iPSC and its CRISPR-corrected isogenic control reveals the putative role of EGR1 transcriptional program in the development of multiple endocrine neoplasia type 2A-associated cancers. Stem Cell Res. 26, 8–16 (2017).

    PubMed  Google Scholar 

  96. Tidball, A. M. & Parent, J. M. Concise review: exciting cells: modeling genetic epilepsies with patient-derived induced pluripotent stem cells. Stem Cells 34, 27–33 (2016).

    PubMed  Google Scholar 

  97. Lee, G. et al. Large-scale screening using familial dysautonomia induced pluripotent stem cells identifies compounds that rescue IKBKAP expression. Nat. Biotechnol. 30, 1244–1248 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Kaufmann, M. et al. High-throughput screening using iPSC-derived neuronal progenitors to identify compounds counteracting epigenetic gene silencing in Fragile X syndrome. J. Biomol. Screen. 20, 1101–1111 (2015).

    CAS  PubMed  Google Scholar 

  99. Darville, H. et al. Human pluripotent stem cell-derived cortical neurons for high throughput medication screening in autism: a proof of concept study in SHANK3 haploinsufficiency Syndrome. EBioMedicine 9, 293–305 (2016).

    PubMed  PubMed Central  Google Scholar 

  100. Li, R. et al. Recapitulating cortical development with organoid culture in vitro and modeling abnormal spindle-like (ASPM related primary) microcephaly disease. Protein Cell 8, 823–833 (2017).

    PubMed  PubMed Central  Google Scholar 

  101. Quadrato, G. & Arlotta, P. Present and future of modeling human brain development in 3D organoids. Curr. Opin. Cell Biol. 49, 47–52 (2017).

    CAS  PubMed  Google Scholar 

  102. Tomaskovic-Crook, E. & Crook, J. M. Clinically amendable, defined, and rapid induction of human brain organoids from induced pluripotent stem cells. Methods Mol. Biol. https://doi.org/10.1007/7651_2017_95 (2017).

    Article  PubMed  Google Scholar 

  103. Zhang, S. C., Wernig, M., Duncan, I. D., Brustle, O. & Thomson, J. A. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat. Biotechnol. 19, 1129–1133 (2001).

    CAS  PubMed  Google Scholar 

  104. Maroof, A. M. et al. Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells. Cell Stem Cell 12, 559–572 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. DeRosa, B. A. et al. hVGAT-mCherry: a novel molecular tool for analysis of GABAergic neurons derived from human pluripotent stem cells. Mol. Cell. Neurosci. 68, 244–257 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Chen, Z., Brodie, M. J., Liew, D. & Kwan, P. Treatment outcomes in patients with newly diagnosed epilepsy treated with established and new antiepileptic drugs: a 30-year longitudinal cohort study. JAMA Neurol. 75, 279–286 (2018).

    PubMed  Google Scholar 

  107. Schulz, A. et al. Study of intraventricular cerliponase Alfa for CLN2 disease. N. Engl. J. Med. 378, 1898–1907 (2018).

    CAS  PubMed  Google Scholar 

  108. Rosenow, F. et al. Personalized translational epilepsy research - novel approaches and future perspectives: part I: clinical and network analysis approaches. Epilepsy Behav. 76, 13–18 (2017).

    PubMed  Google Scholar 

  109. Kohler, S. et al. The human phenotype ontology in 2017. Nucleic Acids Res. 45, D865–D876 (2017).

    PubMed  Google Scholar 

  110. Robinson, P. N., Mungall, C. J. & Haendel, M. Capturing phenotypes for precision medicine. Cold Spring Harb. Mol. Case Stud. 1, a000372 (2015).

    PubMed  PubMed Central  Google Scholar 

  111. Robinson, P. N. Deep phenotyping for precision medicine. Hum. Mutat. 33, 777–780 (2012).

    PubMed  Google Scholar 

  112. Haring, R. & Wallaschofski, H. Diving through the “-omics”: the case for deep phenotyping and systems epidemiology. OMICS 16, 231–234 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Tracy, R. P. ‘Deep phenotyping’: characterizing populations in the era of genomics and systems biology. Curr. Opin. Lipidol. 19, 151–157 (2008).

    CAS  PubMed  Google Scholar 

  114. Moeller, F. et al. EEG-fMRI in atypical benign partial epilepsy. Epilepsia 54, e103–e108 (2013).

    PubMed  Google Scholar 

  115. Warren, A. E. L., Abbott, D. F., Jackson, G. D. & Archer, J. S. Thalamocortical functional connectivity in Lennox-Gastaut syndrome is abnormally enhanced in executive-control and default-mode networks. Epilepsia 58, 2085–2097 (2017).

    PubMed  Google Scholar 

  116. Siniatchkin, M. & Capovilla, G. Functional neuroimaging in epileptic encephalopathies. Epilepsia 54 (Suppl. 8), 27–33 (2013).

    PubMed  Google Scholar 

  117. Frey, L. J., Lenert, L. & Lopez-Campos, G. EHR big data deep phenotyping. Contribution of the IMIA Genomic Medicine Working Group. Yearb. Med. Inform. 9, 206–211 (2014).

    Google Scholar 

  118. Underhill, M. L. et al. A state-wide initiative to promote genetic testing in an underserved population. Cancer Med. 6, 1837–1844 (2017).

    PubMed  PubMed Central  Google Scholar 

  119. Childers, K. K., Maggard-Gibbons, M., Macinko, J. & Childers, C. P. National distribution of cancer genetic testing in the united states: evidence for a gender disparity in hereditary breast and ovarian cancer. JAMA Oncol. 4, 876–879 (2018).

    PubMed  Google Scholar 

  120. Gerhard, G. S., Fisher, S. G. & Feldman, A. M. Genetic testing for inherited cardiac diseases in underserved populations of non-european ancestry: double disparity. JAMA Cardiol. 3, 273–274 (2018).

    PubMed  Google Scholar 

  121. Manrriquez, E., Chapman, J. S., Mak, J., Blanco, A. M. & Chen, L. M. Disparities in genetics assessment for women with ovarian cancer: can we do better? Gynecol. Oncol. 149, 84–88 (2018).

    PubMed  Google Scholar 

  122. Sayani, A. Inequities in genetic testing for hereditary breast cancer: implications for public health practice. J. Community Genet. https://doi.org/10.1007/s12687-018-0370-8 (2018).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Reviewer information

Nature Reviews Neurology thanks A. Poduri, X. Wang and E. Russo for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Both authors contributed to the discussion of content, writing and editing of the manuscript before submission.

Corresponding author

Correspondence to Scott T. Demarest.

Ethics declarations

Competing interests

S.D. has consulted for Upsher-Smith on an unrelated subject matter. A.B.K. has no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Demarest, S.T., Brooks-Kayal, A. From molecules to medicines: the dawn of targeted therapies for genetic epilepsies. Nat Rev Neurol 14, 735–745 (2018). https://doi.org/10.1038/s41582-018-0099-3

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41582-018-0099-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing