You are on page 1of 7

REVIEW

CURRENT
OPINION Advances in epilepsy gene discovery and
implications for epilepsy diagnosis and treatment
Joseph D. Symonds a,b, Sameer M. Zuberi a,b, and Michael R. Johnson c

Purpose of review
Epilepsy genetics is shifting from the academic pursuit of gene discovery to a clinical discipline based on
molecular diagnosis and stratified medicine. We consider the latest developments in epilepsy genetics and
review how gene discovery in epilepsy is influencing the clinical classification of epilepsy and informing
new therapeutic approaches and drug discovery.
Recent findings
Recent studies highlighting the importance of mutation in GABA receptors, NMDA receptors, potassium
channels, G-protein coupled receptors, mammalian target of rapamycin pathway and chromatin
remodeling are discussed. Examples of precision medicine in epilepsy targeting gain-of-function mutations
in KCNT1, GRIN2A, GRIN2D and SCN8A are presented. Potential reasons for the paucity of examples of
precision medicine for loss-of-function mutations or in non-ion channel epilepsy genes are explored. We
highlight how systems genetics and gene network analyses have suggested that pathways disrupted in
epilepsy overlap with those of other neurodevelopmental traits including human cognition. We review how
network-based computational approaches are now being applied to epilepsy drug discovery.
Summary
We are living in an unparalleled era of epilepsy gene discovery. Advances in clinical care from this
progress are already materializing through improved clinical diagnosis and stratified medicine. The
application of targeted drug repurposing based on single gene defects has shown promise for epilepsy
arising from gain-of-function mutations in ion-channel subunit genes, but important barriers remain to
translating these approaches to non-ion channel epilepsy genes and loss-of-function mutations. Gene
network analysis offers opportunities to discover new pathways for epilepsy, to decipher epilepsy’s
relationship to other neurodevelopmental traits and to frame a new approach to epilepsy drug discovery.
Keywords
drug discovery, epilepsy, genetics, networks, next-generation sequencing, precision medicine, systems
genetics

INTRODUCTION &
encephalopathy [3,4,5 ], and neurodevelopmental
Epilepsy genetics can be conceptualized under two disease more generally [6], represents a fundamental
broad headings: monogenic epilepsy in which a scientific advance. NGS studies have revealed how
single variant of large effect is considered causative mutations in the same gene can give rise to a spec-
and complex genetic epilepsy in which a presumed trum of epilepsy phenotypes (or even different
combinatorial effect of multiple susceptibility var-
iants is thought to underlie the disease. Although
a
advances in next-generation sequencing (NGS) have Paediatric Neurosciences Research Group, Fraser of Allander Neuro-
led to substantial progress in the discovery of genes sciences Unit, Royal Hospital for Children, bSchool of Medicine, Univer-
sity of Glasgow, Glasgow and cDivision of Brain Sciences, Imperial
for monogenic epilepsy, attempts to identify var-
College London, London, United Kingdom
iants that confer susceptibility to complex epilepsy
Correspondence to Prof Michael R. Johnson, MD, PhD, Division of Brain
using genome-wide association study (GWAS) have Sciences, Faculty of Medicine, Imperial College London, Room E419,
identified few contributory variants [1], most likely Burlington Danes Building, Hammersmith Hospital Campus, 160 Du
because of the small sample sizes of the epilepsy Cane Road, London W12 0NN, United Kingdom.
GWAS to date [2]. In contrast to the current state of Tel: +44 0203 311 7508; e-mail: m.johnson@imperial.ac.uk
the art for epilepsy GWAS, NGS-enabled discovery of Curr Opin Neurol 2017, 30:193–199
the importance of de novo mutation in epileptic DOI:10.1097/WCO.0000000000000433

1350-7540 Copyright ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Seizure disorders

aphasia spectrum disorders and idiopathic focal


KEY POINTS epilepsy with rolandic spikes [14–16]. GRIN1
 Next-generation sequencing has enabled rapid mutations have been found to cause epilepsy with
progress in gene discovery for epilepsy, and this has profound developmental delay associated with a
already led to material advances in clinical diagnosis hyperkinetic movement disorder and infantile
and care. &
hypotonia [17 ], and mutations in GRIN2D are
present in some patients with severe infantile-onset
 Epilepsy genetics has implications for the classification &

of epilepsy, which will evolve with time and requires an epilepsy [18 ].
agreed framework of terminology. This includes a need One of the most discussed epilepsy genes of the
to understand the relationship between the genetic past 12 months is KCNA2. De novo mutations in this
underpinnings of epilepsy and other gene have been found in severe childhood-onset
neurodevelopmental diseases. & &
epilepsy [19 ,20 ], but also in an autosomal-domi-
 There is optimism that drug therapy for epilepsy can be nant family with the predominant phenotype of
targeted to the underlying genetic cause, but to date infantile-onset pharmacoresponsive epilepsy in
examples of precision medicine are mostly in gain-of- the setting of normal intellect and episodic ataxia
function mutations in ion-channel genes. This highlights [21]. Along with SCN1A and SCN2A-related epilepsy,
a need to develop new approaches to drug discovery this observation highlights how mutations in the
in epilepsy that address non-ion channel epilepsy genes same gene can give rise to a spectrum of epilepsy
and loss-of-function mutations.
severity, from severe childhood epilepsy with devel-
opmental delay to pharmacoresponsive self-limit-
ing epilepsy. The mechanistic explanations for such
phenotypic diversity are unknown, although in the
forms of neurodevelopmental disease) and have case of KCNA2 it has been observed that gain-of-
highlighted the genetic heterogeneity underlying function and loss-of-functions appear to cause dis-
some relatively well defined epilepsy phenotypes tinct epilepsy phenotypes [20 ].
&

&
[7]. As analysis of epilepsy gene panels [8 ] and The phenotypic spectrum of another potassium
clinical whole exome sequencing becomes main- channel subunit gene KCNT1 has also recently
stream, the diagnostic approach in epilepsy genetics expanded, from epilepsy of infancy with migrating
is increasingly one that moves from genotype to focal seizures (EIMFS) to include West syndrome and
phenotype. In this review, we consider the recent
&
early-onset epileptic encephalopathy [22 ].
advances in epilepsy genetics and in particular how Mutations in GNAO1, encoding the Gao
these discoveries are changing the conceptual G-protein subunit were first described in four patients
boundaries between epilepsy phenotypes and the with early-onset epileptic encephalopathy [23]. The
prospects for precision medicine and new drug phenotypic spectrum of this disorder has also broad-
discovery based on epilepsy gene discovery. ened to now include cases with clear developmental
&
delay prior to childhood onset of epilepsy [24 ]. The
majority of these cases demonstrate a dyskinetic
RECENT ADVANCES IN MONOGENIC movement disorder that may have onset at any age
EPILEPSY between infancy and late childhood [25 ]. A second
&

Below, we consider the latest advances in gene G-protein receptor subunit gene GNB1 has also been
discovery for monogenic epilepsy. implicated in epilepsy, with patients manifesting a
The extent of phenotypic variability associated wide variety of seizure types [26].
with GABA receptor mutations is becoming appa- The gap activity toward rags (GATOR) complex
rent, with GABRA1 mutations originally reported is involved in the inhibition of the mammalian
in a family with dominantly inherited Juvenile target of rapamycin (mTOR) complex, which plays
Myoclonic Epilepsy now also described in severe an essential role in regulating cell growth and pro-
& &
infantile onset epileptic encephalopathies [9 ,10 ]. liferation. Three GATOR complex genes (DEPDC5,
GABRB3 mutations, first observed in families with NPRL2 and NPRL3) have been associated with fam-
Childhood Absence Epilepsy, are now also found in ilial focal epilepsy with or without focal cortical
& & &
epileptic encephalopathy [5 ,10 –12 ], and a new
&& &&
dysplasia [27 –30 ], and mutations in a fourth
entity, GABRB1 encephalopathy, presenting with mTOR regulator gene, NEDD4L, have been found
epileptic seizures and developmental regression in to cause epilepsy and periventricular nodular heter-
& &
infancy has been described [11 –13 ].
&
otopia [31 ]. The importance of somatic mTOR path-
Mutations in the NMDA receptor subunit genes way mutation in focal cortical dysplasia has also
GRIN2A and GRIN2B are an important cause of become increasingly apparent with deep sequencing
neurodevelopmental epilepsy including epilepsy- of paired blood/brain DNA [32 ,33].
&

194 www.co-neurology.com Volume 30  Number 2  April 2017

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Advances in epilepsy gene discovery and implications Symonds et al.

Another fascinating development has been the Terminology alongside commentary articles and
&
association between epilepsy and genes involved online feedback [47 ]. The process will be complete
in transcriptional regulation through chromatin with the publication of two companion articles, one
remodeling. Examples include CHD2, associated on the classification of seizure types and the other
with myoclonic encephalopathy and photosensi- on the overall classification of the epilepsies in the
& &
tivity [34 ,35 ], SMARCA2 associated with Nico- journal Epilepsia in 2017.
laides– Baraitser Syndrome and Myoclonic Astatic One of the principal drivers for a new classifi-
&
Epilepsy [36 ] and SMC1A, associated with a severe cation was the clinical imperative to consider cause
epilepsy with clusters of seizures in women at each of three levels of epilepsy classification.
& &
[37 ,38,39 ]. It is perhaps surprising that genes These levels are seizure type (generalized onset, focal
involved in chromatin remodeling have been associ- onset and unknown onset), epilepsy type (general-
ated with such apparently well delineated clinical ized, focal, combined generalized and focal and
syndromes as intuitively, one might expect a large unknown type) and epilepsy syndrome. The frame-
number of downstream genes to be disrupted and work for the classification of epilepsies divides cause
therefore the phenotypes to be broad and variable. into six groups chosen for their treatment implica-
In fact, chromatin remodeling genes appear to be tions – structural, genetic, infectious, metabolic,
&
highly selective in the genes that they regulate [40 ]. immune and unknown. The concept of a genetic
As well as facilitating the discovery of new epi- epilepsy is that it results directly from a known or
lepsy genes, the ability of NGS to screen for genetic presumed genetic mutation in which seizures are a
variants in multiple genes in parallel has revealed core symptom of the disorder. In the new classifi-
the potential for so-called blended phenotypes – cation scheme, there are three ways in which an
patients whose disorder might be explained by more epilepsy may be classified as genetic. First, evidence
than one large-effect genetic variant. Recently pub- for a genetic cause may be based solely on a family
lished cases of blended phenotypes in epilepsy history whether or not the disorder has a known
include dominantly coinherited SLC20A2 and gene. Second, clinical research in populations with
CHRNB2 mutations associated with familial gener- the same syndrome may suggest that a disorder is
alized epilepsy with basal ganglia calcifications primarily genetic, for example twin studies in Juven-
&
[41 ]; a de novo GNAO1 mutation combined with ile Myoclonic Epilepsy [48]. Third, a pathogenic
a de novo HESX1 mutation associated with progress- gene variant may have been reproducibly associated
ive encephalopathy with edema, hypsarrhythmia with an epilepsy phenotype. In the new ILAE classi-
&
and optic atrophy syndrome [42 ]; de-novo deletion fication of the epilepsies, ‘genetic’ will not equate to
of MEF2C with an inherited SCN1A variant associ- ‘inherited’ as de novo pathogenic gene variants are
ated with drug-resistant childhood-onset epilepsy important particularly in severe infantile-onset
&
[43 ]; and a patient with 7q11.23 deletion (Williams disorders.
syndrome) plus a de novo GABRA1 variant present- As new genes are associated with epilepsy, it is
&
ing with a severe drug-resistant epilepsy [44 ]. likely that the genetic disorders will be named after
Whether some patients with sudden unexpected the gene. For example ‘SCN8A encephalopathy’
& &
death in epilepsy (SUDEP), who have been found [49 ] or ‘GRIN2 encephalopathy’ [17 ]. Epilepsy
to harbor an increased burden of de novo mutations genes are typically expressed widely in the central
at a population level, represent a multiallelic severe nervous system, and (as detailed above) pathogenic
&
epilepsy phenotype remains to be determined [45 ]. variants in these genes can produce diverse pheno-
types including movement disorders, learning
disability and autistic features, which may be more
GENE DISCOVERY AND THE prominent and clinically relevant than the epilepsy
INTERNATIONAL LEAGUE AGAINST &
[50 ]. Currently, epilepsy genetics appears to be on
EPILEPSY CLASSIFICATION the cusp of an epistemological crossroads – should
The last ratified International League Against Epi- these conditions be considered genetic epilepsies, or
lepsy (ILAE) Classification of the Epilepsies was in genetic developmental disorders in which epilepsy
1989 [46]. Development of an updated classification may (or may not) be a component of the symptom
for primarily clinical purposes, which acknowledges complex?
the scientific advances in our understanding of the
causes of the epilepsies, has involved a wide engage-
ment with the epilepsy community over a period of PRECISION MEDICINE IN EPILEPSY: HOPE
7 years. This long iterative process has included VERSUS DATA
publication of proposals for classification from The importance of highly penetrant de novo
the ILAE Commission for Classification and mutation in epilepsy along with advances in our

1350-7540 Copyright ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 195

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Seizure disorders

ability to quantitatively assess the pathogenicity of memantine inhibited GluN2A-Leu812Met-contain-


gene variants [51] has led to optimism that antiepi- ing NMDARs and off-label treatment with oral
leptic treatments could be targeted to a person’s memantine led to a reduction in the child’s seizure
&
specific genetic diagnosis (precision medicine) burden. Li et al. [18 ] subsequently reported two
&
[52 ]. In this section, we discuss some of the studies unrelated children with epileptic encephalopathy
that have established a proof-of-principle for pre- associated with gain-of-function mutations in
cision medicine in epilepsy, but we also consider GRIN2D. Here again, FDA-approved drugs that act
potential barriers to its wider implementation. In as NMDAR blockers were evaluated in vitro, and
the discussion below, we narrowly define precision subsequently both children were treated off-label
medicine as a specific drug therapy targeting an with oral memantine with a reported ‘mild to
underlying genetic defect, rather than the use of moderate’ improvement in seizure burden. Boerma
&
genetics to inform a stratified treatment approach et al. [58 ] reported four patients with severe
such as the use of the ketogenic diet in GLUT1 epilepsy associated with putative gain-of-function
&
deficiency syndrome [53 ], or prescription of stiri- SCN8A missense mutations in which seizure control
pentol and avoidance of sodium channel blockers in was obtained with the sodium channel blocker
&
SCN1A disease [54 ]. phenytoin.
One of the first reports of a targeted precision Although these cases illustrate the principles of
therapy in epilepsy was the off-label use of quinidine precision medicine in epilepsy, in each of the
in the treatment of a child with EIMFS secondary to examples described above the epilepsy resulted from
a gain-of-function missense mutation in KCNT1 a putative gain-of-function mutation. However,
(Arg428Gln) [55]. Prior in-vitro studies had estab- substantial evidence implicates protein loss-of-func-
lished that quinidine is a blocker of KCNT1 channels, tion and haploinsufficiency as a key mechanism of
suggesting that the drug might be a precision therapy both epileptic encephalopathy and monogenic neu-
in epilepsy associated with gain-of-function KCNT1 rodevelopmental disease more broadly. Examples of
mutation. Subsequent treatment with quinidine precision medicine in epilepsy in which the
resulted in a marked reduction in seizure frequency approach has been to increase the activity of the
and improved psychomotor development. wild-type allele to compensate for protein loss-of-
Seizure outcomes following treatment with qui- function have been relatively few. To date, the only
nidine have since been reported for two additional example of precision medicine targeting a loss-of-
&
epilepsy patients with KCNT1 mutation [56 ]. The function mutation is a report of improvement of
epilepsy phenotypes were severe nocturnal seizures seizure frequency with the KCNQ2/KCNQ3 agonist
with onset in childhood, and a second case of retigabine in a number of patients with neonatal-
EIMFS, due to Tyr796His and Lys629Asn missense onset epileptic encephalopathy associated with
&
mutations, respectively. Both mutations resulted in KCNQ2 mutation [59 ]. With the field still in its
a gain-of-function, and in both cases quinidine infancy, it is probably too early to tell if the paucity
restored mutant function toward wild-type in vitro. of reports of precision medicine in loss-of-function
However, although the patient with EIMFS had an mutation reflects a more challenging pharmacologi-
80% reduction in seizure frequency following treat- cal environment related to developing protein acti-
ment with quinidine, the other child with nocturnal vators compared with blockers.
seizures did not improve. The reason for the discor- A further observation from the published
dant clinical response to quinidine in these two examples of precision medicine in epilepsy is that
cases despite similar in-vitro evidence for efficacy so far all the cases resulted from mutation in an ion-
is not known and highlights the need to consider channel subunit gene. To date, with the exception
&&
the functional impact of mutations within a more of the use of everolimus in tuberous sclerosis [60 ],
systems-wide context. there are no reported examples of precision medi-
Other recent cases exemplifying the potential of cine targeting a monogenic epilepsy arising from
precision medicine in epilepsy have been described mutation in the growing list of non-ion channel
for patients with de novo mutations in GRIN2A [57] epilepsy genes such as LGI1, STXBP1, PCDH19,
&
and GRIN2D [18 ] treated with NMDA receptor TCF4, CDKL5 and others. Given that many of these
blockers, and in SCN8A-associated epilepsy treated proteins have not been traditional targets for drug
&
with phenytoin [58 ]. Thus, Pierson et al. reported a development, and their functional role in epilepsy is
child with epileptic encephalopathy and severe cog- poorly understood, the development of precision
nitive impairment associated with a gain-of-func- therapies for epilepsy resulting from mutation in
tion Leu812Met missense mutation in GRIN2A [57]. non-ion channel genes may advance slowly. This
In-vitro analysis revealed the US Food and Drug highlights the need for new approaches to drug
Administration (FDA) approved NMDAR blocker discovery for monogenic epilepsy. One proposal

196 www.co-neurology.com Volume 30  Number 2  April 2017

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Advances in epilepsy gene discovery and implications Symonds et al.

has been to develop systems for the experimental discoveries highlight a plethora of potential new
high-throughput screening of compounds such as mechanisms for epilepsy. Whether these mechan-
the use of stem cell models of epilepsy (‘epilepsy in a isms will converge around common pathways or
&
dish’) [52 ]. However, with potentially many thou- each require the development of a specific targeted
sands of causal variants for epilepsy in hundreds of therapy is currently unclear, but there is genuine
different genes [2], and thousands of known drugs optimism that the era of NGS-enabled gene discov-
(www.drugbank.ca/stats), there are potentially tens ery will lead to advances in the care of people
of millions of drug–disease pairings, making the with epilepsy.
exhaustive experimental screening of all known
drugs for all epilepsy variants unfeasible. This com- Acknowledgements
binatorial problem will require, at least in part, a None.
computational solution.
One approach to developing a computational Financial support and sponsorship
framework for new drug discovery and drug screen- J.D.S. is funded for a PhD in epilepsy genetics by the
ing makes use of a systems perspective of disease. Glasgow Children’s Hospital Charity (Scottish Charity
Here, a disease is viewed in terms of its molecular Number SCO 007856). S.M.Z. receives research funding
drivers arising from the interaction of sets of genes from Epilepsy Research UK, Dravet Syndrome UK, UCB
in regulatory networks [61]. In epilepsy, network Pharma and Glasgow Childrens Hospital Charity. He is
analyses have revealed that many different genes Editor-in-Chief of the European Journal of Paediatric
for epileptic encephalopathy interact in gene regu- Neurology for which he receives an honorarium. M.J.R.
&& &
latory networks [62 ,63 ], and that these networks receives funding from Imperial College NIHR Biomedical
&&
overlap with those for both human cognition [62 ] Research Centre (BRC) Scheme, UCB Pharma, and the
and non-Mendelian common forms of epilepsy Medical Research Council.
&
[63 ]. These studies suggest the substantial and
heterogeneous genetic contributions to epilepsy Conflicts of interest
may (at least in part) converge on common There are no conflicts of interest.
pathways that are currently poorly understood.
Importantly, gene regulatory networks represent REFERENCES AND RECOMMENDED
measurable molecular entities that can be targeted READING
& &
for both drug target [64 ] and drug [63 ] discovery in Papers of particular interest, published within the annual period of review, have
been highlighted as:
epilepsy. As proof-of-concept for network-based & of special interest
drug discovery in epilepsy, a recent study predicted && of outstanding interest

sodium valproate as a treatment for epilepsy from an 1. International League Against Epilepsy Consortium on Complex Epilepsies.
unsupervised computational screen of 1300 small Genetic determinants of common epilepsies: a meta-analysis of genome-wide
& association studies. Lancet Neurol 2014; 13:893–903.
molecules [63 ]. Critically, the computational 2. Speed D, O’Brien TJ, Palotie A, et al. Describing the genetic architecture of
screen was conducted entirely independently of epilepsy through heritability analysis. Brain 2014; 137:2680–2689.
3. Allen AS, Berkovic SF, Cossette P, et al. De novo mutations in epileptic
valproate’s known mechanism of action or role in encephalopathies. Nature 2013; 501:217–221.
epilepsy, and was based solely on an unsupervised 4. Consortium E, Phenome E, Project G, Consortium E. De novo mutations in
synaptic transmission genes including DNM1 cause epileptic encephalopa-
analysis of disease-related and drug-related gene thies. Am J Hum Genet 2014; 100:179.
expression profiles. Such studies highlight the 5. Consortium E. De novo mutations in SLC1A2 and CACNA1A are important
causes of epileptic encephalopathies. Am J Hum Genet 2016; 99:287–298.
potential of network-based drug discovery as a novel &

Study identifies important new genes for monogenic epilepsy using next-genera-
and efficient strategy for new drug discovery in tion sequencing (NGS).
6. Iossifov I, O’Roak BJ, Sanders SJ, et al. The contribution of de novo coding
epilepsy. Moreover, since network approaches target mutations to autism spectrum disorder. Nature 2014; 515:216–221.
sets of coregulated genes, they may also offer an 7. Zhu X, Need AC, Petrovski S, Goldstein DB. One gene, many neuropsychia-
tric disorders: lessons from Mendelian diseases. Nat Neurosci 2014;
approach to developing therapies for epilepsies aris- 17:773–781.
ing from the coordinated dysregulation of sets of 8. Trump N, McTague A, Brittain H, et al. Improving diagnosis and broadening
the phenotypes in early-onset seizure and severe developmental delay dis-
genes due to mutation in chromatin remodeling &

orders through gene panel analysis. J Med Genet 2016; 53:310–317.


genes or other gene regulatory sequences, or arising Study reports diagnostic yields from clinical epilepsy gene panel tests.
9. Johannesen K, Marini C, Pfeffer S, et al. Phenotypic spectrum of GABRA1:
from aberrant homeostatic neuronal responses & from generalized epilepsies to severe epileptic encephalopathies. Neurology
to mutation. 2016; 87:1140–1151.
Study reports the phenotypic spectrum from GABBRA1 mutations.
10. Kodera H, Ohba C, Kato M, et al. De novo GABRA1 mutations in Ohtahara
& and West syndromes. Epilepsia 2016; 57:566–573.
CONCLUSION Study reports de novo GABRA1 mutations in patients with infantile-onset epilepsy.
11. Janve VS, Hernandez CC, Verdier KM, et al. Epileptic encephalopathy de novo
We live in an unprecedented era of epilepsy gene & GABRB mutations impair GABAA receptor function. Ann Neurol 2016;
79:806–825.
discovery. Epilepsy genetics is moving rapidly from Study reports de novo GABAA and GABAB receptor mutations in epileptic
gene discovery to clinical practice, and these encephalopathy.

1350-7540 Copyright ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 197

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Seizure disorders

12. Papandreou A, Mctague A, Trump N, et al. GABRB3 mutations: a new and 32. Nakashima M, Saitsu H, Takei N, et al. Somatic mutations in the MTOR gene
& emerging cause of early infantile epileptic encephalopathy. Dev Med Child & cause focal cortical dysplasia type IIb. Ann Neurol 2015; 78:375–386.
Neurol 2016; 58:416–420. Discovery of importance of somatic mutation in focal cortical dysplasia.
GABRB3 mutations first observed in families with CAE are found in epileptic 33. Lim JS, Kim W, Kang H-C, et al. Brain somatic mutations in MTOR cause focal
encephalopathy. cortical dysplasia type II leading to intractable epilepsy. Nat Med 2015;
13. Lien E, Våtevik AK, Østern R, et al. A second patient with a de novo GABRB1 21:395–400.
& mutation and epileptic encephalopathy. Ann Neurol 2016; 80:311–312. 34. Thomas RH, Zhang LM, Carvill GL, et al. CHD2 myoclonic encephalopathy is
Study reports de novo GABRB1 mutation in epileptic encephalopathy. & frequently associated with self-induced seizures. Neurology 2015; 84:951–
14. Endele S, Rosenberger G, Geider K, et al. Mutations in GRIN2A and GRIN2B 958.
encoding regulatory subunits of NMDA receptors cause variable neurodeve- Discovery of CHD2 mutations in myoclonic encephalopathy.
lopmental phenotypes. Nat Genet 2010; 42:1021–1026. 35. Galizia EC, Myers CT, Leu C, et al. CHD2 variants are a risk factor for
15. Carvill GL, Regan BM, Yendle SC, et al. GRIN2A mutations cause epilepsy- & photosensitivity in epilepsy. Brain 2015; 138:1198–1207.
aphasia spectrum disorders. Nat Genet 2013; 45:1073–1076. CHD2 implicated as a risk factor for common forms of photosensitive epilepsy.
16. Lesca G, Rudolf G, Bruneau N, et al. GRIN2A mutations in acquired 36. Tang S, Hughes E, Lascelles K, et al. New SMARCA2 mutation in a patient
epileptic aphasia and related childhood focal epilepsies and encephalopa- & with Nicolaides–Baraitser syndrome and myoclonic astatic epilepsy. Am J
thies with speech and language dysfunction. Nat Genet 2013; 45:1061– Med Genet A 2016; 173:195–199.
1066. Report of a de novo SMARCA2 missense mutation discovered by exome sequen-
17. Lemke JR, Geider K, Helbig KL, et al. Delineating the GRIN1 phenotypic cing.
& spectrum: a distinct genetic NMDA receptor encephalopathy. Neurology 37. Goldstein JHR, Tim-aroon T, Shieh J, et al. Novel SMC1A frameshift mutations
2016; 86:2171–2178. & in children with developmental delay and epilepsy. Eur J Med Genet 2015;
Discovery of GRIN1 mutations as a cause of epilepsy with profound developmental 58:562-568.
delay associated with movement disorder and infantile hypotonia. Report of de novo mutations in SMC1A in epilepsy and expand the phenotypic
18. Li D, Yuan H, Ortiz-Gonzalez XR, et al. GRIN2D recurrent de novo dominant spectrum.
& mutation causes a severe epileptic encephalopathy treatable with NMDA 38. Lebrun N, Lebon S, Jeannet P-Y, et al. Early-onset encephalopathy with
receptor channel blockers. Am J Hum Genet 2016; 99:802–816. epilepsy associated with a novel splice site mutation in SMC1A. Am J Med
Report of de novo GRIN2D mutation and example of precision medicine with off- Genet A 2015; 167A:3076-3081.
label use of an FDA-approved NMDAR blocker. 39. Jansen S, Kleefstra T, Willemsen MH, et al. De novo loss-of-function mutations
19. Syrbe S, Hedrich UBS, Riesch E, et al. De novo loss- or gain-of-function & in X-linked SMC1A cause severe ID and therapy-resistant epilepsy in females:
& mutations in KCNA2 cause epileptic encephalopathy. Nat Genet 2015; expanding the phenotypic spectrum. Clin Genet 2016; 90:413–419.
47:393–399. Report of de novo mutations in SMC1A in epilepsy and expand the phenotypic
New report of both gain and loss-of-function mutations in KCNA2 as a cause of spectrum.
epileptic encephalopathy. 40. Liu JC, Ferreira CG, Yusufzai T. Human CHD2 is a chromatin assembly
20. Allou L, Julia S, Amsallem D, et al. Rett-like phenotypes: expanding the genetic & ATPase regulated by its chromo- And DNA-binding domains. J Biol Chem
& heterogeneity to the KCNA2 gene and first familial case of CDKL5-related 2015; 290:25–34.
disease. Clin Genet 2016; doi:10.1111/cge.12784. [Epub ahead of print] Shows that the accessory domains of CHD2 play roles in regulating the ATPase
Reports novel KCNA2-related epilepsy phenotypes. domain and conferring selectivity to chromatin substrates.
21. Corbett MA, Bellows ST, Li M, et al. Dominant KCNA2 mutation causes 41. Fjaer R, Brodtkorb E, Øye AM, et al. Generalized epilepsy in a family with basal
episodic ataxia and pharmacoresponsive epilepsy. 2016; 87:1975– & ganglia calcifications and mutations in SLC20A2 and CHRNB2. Eur J Med
1984. Genet 2015; 58:624–628.
22. Ohba C, Kato M, Takahashi N, et al. De novo KCNT1 mutations in early-onset Example of a ‘blended phenotype’.
& epileptic encephalopathy. Epilepsia 2015; 56:e121–e128. 42. Gawlinski P, Posmyk R, Gambin T, et al. PEHO syndrome may represent
Expansion of the phenotypic spectrum of KCNT1 epilepsy. & phenotypic expansion at the severe end of the early-onset encephalopathies.
23. Nakamura K, Kodera H, Akita T, et al. De novo mutations in GNAO1, encoding Pediatr Neurol 2016; 60:83–87.
a Gao subunit of heterotrimeric G proteins, cause epileptic encephalopathy. Example of a ‘blended phenotype.
Am J Hum Genet 2013; 93:496–505. 43. Rocha H, Sampaio M, Rocha R, et al. MEF2C haploinsufficiency syndrome:
24. Saitsu H, Fukai R, Ben-Zeev B, et al. Phenotypic spectrum of GNAO1 & report of a new MEF2C mutation and review. Eur J Med Genet 2015;
& variants: epileptic encephalopathy to involuntary movements with severe 59:478–482.
developmental delay. Eur J Hum Genet 2016; 24:129–134. Example of a ‘blended phenotype.
Important expansion of the phenotype of GNAO1-related epilepsy. 44. Popp B, Trollmann R, Büttner C, et al. Do the exome: a case of Williams–
25. Ananth AL, Robichaux-Viehoever A, Kim YM, et al. Clinical course of six & Beuren syndrome with severe epilepsy due to a truncating de novo variant in
& children with GNAO1 mutations causing a severe and distinctive movement GABRA1. Eur J Med Genet 2016; 59:549–553.
disorder. Pediatr Neurol 2016; 59:81–84. Example of a ‘blended phenotype.
Important expansion of the phenotype including disease course of GNAO1 45. Leu C, Balestrini S, Maher B, et al. Genome-wide polygenic burden of rare
epilepsy. & deleterious variants in sudden unexpected death in epilepsy. EBioMedicine
26. Petrovski S, Küry S, Myers CT, et al. Germline de novo mutations in GNB1 2015; 2:1063–1070.
cause severe neurodevelopmental disability, hypotonia, and seizures. Am J First study examining the role of rare variation in SUDEP using NGS.
Hum Genet 2016; 98:1001–1010. 46. Berg AT, Berkovic SF, Brodie MJ, et al. Revised terminology and concepts for
27. Baldassari S, Licchetta L, Tinuper P, et al. GATOR1 complex: the common organization of seizures and epilepsies: report of the ILAE commission on
&& genetic actor in focal epilepsies. J Med Genet 2016; 53:503–510. classification and terminology, 2005–2009. Epilepsia 2010; 51:676–685.
This study reports germline mutations in gap activity toward rags (GATOR) 47. Scheffer IE, French J, Hirsch E, et al. Classification of the epilepsies: new
complex genesin association with familial focal epilepsy with or without focal & concepts for discussion and debate. Special Report of the ILAE Classification
cortical dysplasia. Task Force of the Commission for Classification and Terminology. Epilepsia
28. Ricos MG, Hodgson BL, Pippucci T, et al. Mutations in the mammalian target Open 2016.
&& of rapamycin pathway regulators NPRL2 and NPRL3 cause focal epilepsy. Discussion document on epilepsy and seizure classification produced by the
Ann Neurol 2016; 79:120–131. International League Against Epilepsy Task Force.
This study reports germline mutations in gap activity toward rags (GATOR) 48. Vadlamudi L, Milne RL, Lawrence K, et al. Genetics of epilepsy: the testimony
complex genesin association with familial focal epilepsy with or without focal of twins in the molecular era. Neurology 2014; 83:1042–1048.
cortical dysplasia. 49. Larsen J, Carvill GL, Gardella E, et al. The phenotypic spectrum of SCN8A
29. Weckhuysen S, Marsan E, Lambrecq V, et al. Involvement of GATOR complex & encephalopathy. Neurology 2015; 84:480–489.
&& genes in familial focal epilepsies and focal cortical dysplasia. Epilepsia 2016; Reports and expands and the phenotypic spectrum of SCN8A encephalopathy.
57:994–1003. 50. Korff CM, Brunklaus A, Zuberi SM. Epileptic activity is a surrogate for an
This study reports germline mutations in gap activity toward rags (GATOR) & underlying etiology and stopping the activity has a limited impact on devel-
complex genesin association with familial focal epilepsy with or without focal opmental outcome. Epilepsia 2015; 56:1477–1481.
cortical dysplasia. Re-examines the concept of ‘epileptic encephalopathy’.
30. Sim JC, Scerri T, Fanjul-Fernández M, et al. Familial cortical dysplasia caused 51. Lek M, Tewksbury J, Services H. Analysis of protein-coding genetic variation in
&& by mutation in the mTOR regulator NPRL3. Ann Neurol 2016; 79:132–137. 60,706 humans. Nature 2016; 536:285–291.
This study reports germline mutations in GATOR complex genesin association 52. Consortium E. A roadmap for precision medicine in the epilepsies. Lancet
with familial focal epilepsy with or without focal cortical dysplasia & Neurol 2015; 4422:1–10.
31. Broix L, Jagline HL, Ivanova E, et al. Mutations in the HECT domain of NEDD4L Review discussing prospects and roadmap for precision medicine in epilepsy.
& lead to AKT-mTOR pathway deregulation and cause periventricular nodular 53. Kass HR, Winesett SP, Bessone SK, et al. Use of dietary therapies amongst
heterotopia. Nat Genet 2016; 48:1349–1358. & patients with GLUT1 deficiency syndrome. Seizure 2016; 35:83–87.
Reports mutation in NEDD4L, a regulator of mammalian target of rapamycin in Review of dietary therapies including ketogenic diet in GLUT1 deficiency syn-
association with epilepsy and periventricular nodular heterotopia. drome.

198 www.co-neurology.com Volume 30  Number 2  April 2017

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.


Advances in epilepsy gene discovery and implications Symonds et al.

54. Wilmshurst JM, Gaillard WD, Vinayan KP, et al. Summary of recommendations 60. French JA, Lawson JA, Yapici Z, et al. Adjunctive everolimus therapy for
& for the management of infantile seizures: task force report for the ILAE && treatment-resistant focal-onset seizures associated with tuberous sclerosis
commission of pediatrics. Epilepsia 2015; 56:1185–1197. (EXIST-3): a phase 3, randomised, double-blind, placebo-controlled study.
Review of treatment and management recommendations in infantile seizures. Lancet 2016; 388:2153–2163.
55. Bearden D, Strong A, Ehnot J, et al. Targeted treatment of migrating partial Landmark phase 3 randomised controlled trial.
seizures of infancy with quinidine. Ann Neurol 2014; 76:457–461. 61. Civelek M, Lusis AJ. Systems genetics approaches to understand complex
56. Mikati Ma, Jiang Y, Carboni M, et al. Quinidine in the treatment of KCNT1 traits. Nat Rev Genet 2014; 15:34–48.
& positive epilepsies. Ann Neurol 2015; 78:995–959. 62. Johnson MR, Shkura K, Langley SR, et al. Systems genetics identifies a
Two patients with gain of function KCNT1-related epilepsy with discordant clinical && convergent gene network for cognition and neurodevelopmental disease. Nat
response to quinidine as a precision therapy. Neurosci 2016; 19:223–232.
57. Pierson TM, Yuan H, Marsh ED, et al. GRIN2A mutation and early-onset Reports a convergent gene network for neurodevelopmental disease (including
epileptic encephalopathy: personalized therapy with memantine. Ann Clin epilepsy) and healthy human cognition.
Transl Neurol 2014; 1:190–198. 63. Delahaye-Duriez A, Srivastava P, Shkura K, et al. Rare and common epilepsies
58. Boerma RS, Braun KP, van de Broek MPH, et al. Remarkable phenytoin & converge on a shared gene regulatory network providing opportunities for
& sensitivity in 4 children with SCN8A-related epilepsy: a molecular neuro- novel antiepileptic drug discovery. Genome Biol 2016; 17:245.
pharmacological approach. Neurotherapeutics 2016; 13:192–197. Reports a convergent gene network for monogenic and complex epilepsy and
Exemplers of precision medicine in SCN8A-related epilepsy. outlines a network-based framework for new drug discovery in epilepsy.
59. Millichap JJ, Park KL, Tsuchida T, et al. KCNQ2 encephalopathy: features, 64. Johnson MR, Behmoaras J, Bottolo L, et al. Systems genetics identifies
& mutational hot spots, and ezogabine treatment of 11 patients. Neurol Genet & Sestrin 3 as a regulator of a proconvulsant gene network in human epileptic
2016; 2:e96. hippocampus. Nat Commun 2015; 6:6031.
To date, the only report of precision medicine in epilepsy occurring as a result of a Describes the network-based discovery of a new drug target for epilepsy based on
loss-of-function mutation. mapping the master regulator of an epilepsy gene network.

1350-7540 Copyright ß 2017 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 199

Copyright © 2017 Wolters Kluwer Health, Inc. All rights reserved.

You might also like