Skip to main content
Erschienen in: Molecular Brain 1/2018

Open Access 01.12.2018 | Research

Novel and de novo mutations in pediatric refractory epilepsy

verfasst von: Jing Liu, Lili Tong, Shuangshuang Song, Yue Niu, Jun Li, Xiu Wu, Jie Zhang, Clement C. Zai, Fang Luo, Jian Wu, Haiyin Li, Albert H. C. Wong, Ruopeng Sun, Fang Liu, Baomin Li

Erschienen in: Molecular Brain | Ausgabe 1/2018

Abstract

Pediatric refractory epilepsy is a broad phenotypic spectrum with great genetic heterogeneity. Next-generation sequencing (NGS) combined with Sanger sequencing could help to understand the genetic diversity and underlying disease mechanisms in pediatric epilepsy. Here, we report sequencing results from a cohort of 172 refractory epilepsy patients aged 0–14 years. The pathogenicity of identified variants was evaluated in accordance with the American College of Medical Genetics and Genomics (ACMG) criteria. We identified 43 pathogenic or likely pathogenic variants in 40 patients (23.3%). Among these variants, 74.4% mutations (32/43) were de novo and 60.5% mutations (26/43) were novel. Patients with onset age of seizures ≤12 months had higher yields of deleterious variants compared to those with onset age of seizures > 12 months (P = 0.006). Variants in ion channel genes accounted for the greatest functional gene category (55.8%), with SCN1A coming first (16/43). 81.25% (13/16) of SCN1A mutations were de novo and 68.8% (11/16) were novel in Dravet syndrome. Pathogenic or likely pathogenic variants were found in the KCNQ2, STXBP1, SCN2A genes in Ohtahara syndrome. Novel deleterious variants were also found in West syndrome, Doose syndrome and glucose transporter type 1 deficiency syndrome patients. One de novo MECP2 mutation were found in a Rett syndrome patient. TSC1/TSC2 variants were found in 60% patients with tuberous sclerosis complex patients. Other novel mutations detected in unclassified epilepsy patients involve the SCN8A, CACNA1A, GABRB3, GABRA1, IQSEC2, TSC1, VRK2, ATP1A2, PCDH19, SLC9A6 and CHD2 genes. Our study provides novel insights into the genetic origins of pediatric epilepsy and represents a starting-point for further investigations into the molecular pathophysiology of pediatric epilepsy that could eventually lead to better treatments.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13041-018-0392-5) contains supplementary material, which is available to authorized users.
The original version of this article was revised: the name of one of the authors had been spelled incorrectly. It should be Clement C. Zai, not Celement C. Zai.
A correction to this article is available online at https://​doi.​org/​10.​1186/​s13041-018-0399-y.
Abkürzungen
ACMG
American College of Medical Genetics and Genomics
AEDs
antiepileptic drugs
EEOC
childhood-onset epileptic encephalopathy
EIEE
early-infantile epileptic encephalopathies
MT
Mutation Taster
NGS
next-generation sequencing
OMIM
Online Mendelian Inheritance in Man
PP2
PolyPhen-2
SCN1A
subunit alpha of sodium channel type 1
SIFT
Sorting Intolerant From Tolerant

Introduction

Epilepsy is a complex group of chronic brain disorders that are characterized by recurrent spontaneous seizures, and these can often begin in childhood. Repeated and refractory seizures can cause long-term cognitive impairment, decreased social participation and significantly lower quality of life [1, 2]. Epilepsy is one of the most common neurological disorders with 50 to 100 million affected worldwide, and 2 to 4 million new cases diagnosed each year [3].
Epilepsy is a heterogeneous disease with diverse clinical manifestations and causes, including altered ion channel expression, neurotransmitter signaling, synaptic structure, gliosis, and inflammation [1]. Estimates of heritability from twin studies range from 25% to 70% [4, 5]. Although the range of heritability estimates is quite large, disparate studies using varied methods and studying divergent populations are all consistent in concluding that there is a substantial inherited component to epilepsy [6]. Because of this, we sought to investigate the genome in a heterogeneous set of patients with epilepsy and their parents, with the hope that we would identify novel mutations and confirm existing reports of genetic associations with epilepsy. This type of genetic information can provide an entry point into the biology of epilepsy that could eventually lead to new molecular treatment targets.
With the rapid progress of next-generation sequencing (NGS) techniques, our knowledge of the genetic etiology in many brain disorders such as epilepsy, autism and intellectual disability has expanded greatly [7, 8]. NGS is now capable of efficient and accurate sequencing of entire genomes with small amounts of tissue at ever decreasing costs and has required new approaches to analysing the very large amount of data obtained. For this study, our priority was to separate common and benign genetic variants from those that are likely to be related to the cause of epilepsy, and we chose to apply the American College of Medical Genetics and Genomics (ACMG) guidelines [911]. The ACMG guidelines classify variants into pathogenic, likely pathogenic, uncertain significance, likely benign, and benign categories based on genetic information that includes population, functional, computational and segregation data. In this study, we investigated 153 epilepsy candidate genes in a cohort of 172 refractory epilepsy pediatric patients. We aimed to provide genetic diagnoses of this patient cohort and explore the genetic etiology of pediatric refractory epilepsy.

Method

Participants

We retrospectively collected and analyzed 172 cases of pediatric refractory epilepsy patients between the ages of 1 day to 14 years old in the Department of Pediatrics of Qilu Hospital, China. The program adhered to guidelines of patients’ consent for participation and research was supported by the Ethics Committee of Qilu hospital, Shandong University (No. 2016(027)).
All patients were examined and diagnosed at the Pediatric Department in Qilu Hospital using a combination of patients’ illness history, previous history, family history, physical examinations, developmental evaluation, hematological examination, ambulatory or video electroencephalography (AEEG/VEEG) monitoring, magnetic resonance imaging (MRI) or computed tomography (CT), and genetic sequencing. Developmental evaluation included gross motor, fine motor, language, and personal-social skills. The above information was reviewed by two qualified pediatric epileptologists. Seizure types and epilepsy syndromes were diagnosed and classified according to the guidelines of International League Against Epilepsy (2014, 2017) [12, 13].

Next-generation sequencing

Targeted gene capture and sequencing

Blood samples of the patients and their biological parents were collected to test if the mutations were de novo or inherited. Genomic DNA was extracted from peripheral blood using the QIAamp DNA Mini Kit (Qiagen, China).
One hundred fifty-three genes (Table 1) associated with epilepsy were selected by a gene capture strategy, using the GenCap custom enrichment kit (MyGenostics, China) following the manufacturer’s protocol. The biotinylated capture probes were designed to tile all of the exons without repeated regions. The captured DNAs were eluted, amplified and then their polymerase chain reaction (PCR) products were purified with SPRI beads (Beckman, USA). The enriched libraries were sequenced for paired-end reads of 150 bp by Illumina HiSeq X Ten.
Table 1
One hundred fifty-three epilepsy genes tested in this study by NGS
ADSL
CHD2
DHFR
GLB1
MAGI2
PNPO
SLC9A6
ALDH7A1
CHRNA2
DIAPH3
GLRA1
MAPK10
POLG
SPTAN1
ALG13
CHRNA4
DNAJC6
GPR56
MBD5
PPT1
SRPX2
ARG1
CHRNA7
DNM1
GPR98
MDGA2
PROC
ST3GAL2
ARHGEF15
CHRNB2
DOCK7
GRIN1
ME2
PRRT2
ST3GAL5
ARHGEF9
CLCN2
EEF1A2
GRIN2A
MECP2
RBFOX1
STRADA
ARX
CLCN4
EFHC1
GRIN2B
MEF2C
RBFOX2
STXBP1
ASAH1
CLN3
ELP4
HAX1
MFSD8
RBFOX3
SYNGAP1
ATP13A4
CLN5
EPHB2
HDAC4
MTHFR
RELN
SYNJ1
ATP1A2
CLN6
ERBB4
HEXA
MTOR
RYR3
SZT2
ATP1A3
CLN8
FASN
HEXB
NDE1
SCN1A
TBC1D24
ATP6AP2
CNTN5
FLNA
HNRNPH1
NEDD4L
SCN1B
TCF4
ATP7A
CNTNAP2
FOLR1
HNRNPU
NID2
SCN2A
TNK2
BRAF
COX6B1
FOXG1
IQSEC2
NRXN1
SCN8A
TPP1
BSN
CSTB
FOXP2
KCNB1
PAFAH1B1
SHANK3
TSC1
CACNA1A
CTNNA3
GABBR2
KCNH5
PCDH19
SLC13A5
TSC2
CACNA1H
CTSD
GABRA1
KCNMA1
PDHA1
SLC19A3
TUBA1A
CACNB4
CYB5R3
GABRA6
KCNQ2
PIGA
SLC1A3
UBE3A
CASR
DBH
GABRB2
KCNQ3
PIGV
SLC25A22
VRK2
CDH13
DCX
GABRB3
KCNT1
PLCB1
SLC2A1
WDR45
CDH9
DEPDC5
GABRD
LGI1
PNKD
SLC35A2
ZEB2
CDKL5
DGKD
GABRG2
LIAS
PNKP
SLC46A1
 

Data analysis and pathogenicity of candidate variants

After sequencing, raw data were saved in FASTQ format. Illumina sequencing adapters and low quality reads (< 80 bp) were filtered by Cutadapt [14]. Clean reads were aligned to UCSC hg19 human reference genome using the Burrows-Wheeler Alignment [15] tool. Duplicated reads were removed using Picard (http://​broadinstitute.​github.​io/​picard). Insertions, deletions and SNP variants were detected and filtered using the Genome Analysis Toolkit [16]. Then the identified variants were annotated using ANNOVAR [17] and associated with the following databases: 1000 genomes, Exome Aggregation Consortium, The Human Gene Mutation Database, and predicted by Mutation Taster (MT) [18], Sorting Intolerant From Tolerant (SIFT) [19], PolyPhen-2 (PP2) [20] and Genomic Evolutionary Rate Profiling (GERP++) [21, 22]. Splice-site were predicted by Human Splicing Finder [23]. All variants identified by the Illumina HiSeq X Ten sequencer were confirmed by Sanger sequencing. The pathogenicity of mutations was assessed in accordance with American College of Medical Genetics and Genomics guideline (ACMG) [911].

Statistical analysis

Statistical analysis was performed using SPSS19. The yields of deleterious variants in patients with different onset age or family history were compared using the chi-squared test.

Results

In the current study, we recruited 172 epilepsy pediatric patients, including 23 with Dravet syndrome, ten with Ohtahara syndrome, two with Ohtahara syndrome evolving to West syndrome, ten with West syndrome, two with West syndrome evolving to Lennox-Gastaut syndrome, five with Lennox-Gastaut syndrome, four with Doose syndrome, two with epilepsy of infancy with migrating focal seizures, two with epileptic encephalopathy with continuous spike and wave during sleep, and one each with temporal lobe epilepsy, early myoclonic encephalopathy, Landau-Kleffner syndrome, and glucose transporter type 1 deficiency syndrome. Three patients had Rett syndrome, five had tuberous sclerosis complex, and one had Sturge-Weber syndrome. Forty-two patients were diagnosed as unclassified epileptic encephalopathy and 57 patients were diagnosed as unclassified refractory epilepsy due to nonspecific manifestations (Table 2).
Table 2
Clinical diagnosis in 172 refractory epilepsy and their pathogenic or likely pathogenic mutations
Clinical diagnosis
Cases
P/LP mutations
P/LP gene(recurrent no.)
DS
23
16
SCN1A (16)
OS
10
2
KCNQ2 (1), SCN2A (1)
OS-WS
2
1
STXBP1 (1)
WS
10
4
STXBP1 (1), KCNT1 (1), CDKL5 (1), ADSL (1)
WS-LGS
2
LGS
5
EIMFS
2
ECSWS
2
EME
1
LKS
1
UEE
42
8
CACNA1A (1), GABRA1 (1), GABRB3 (1), SCN8A (2), IQSEC2 (1), PCDH19 (1), CHD2 (1)
Doose
4
1
SYNGAP1 (1)
TLE
1
GLUT1-DS
1
1
SLC2A1 (1)
Rett
3
1
MECP2 (1)
TSC
5
5
TSC2 (5)
SWS
1
UE
57
4
VRK2 (1), ATP1A2 (1), TSC (1), SLC9A6 (1)
Total
172
43
P pathogenic, LP likely pathogenic, DS Dravet syndrome, OS Ohtahara syndrome, OS-WS Ohtahara syndrome evolves to West syndrome, WS West syndrome, WS-LGS West syndrome evolves to Lennox-Gastaut syndrome, LGS Lennox-Gastaut syndrome, Doose Doose syndrome, ECSWS epileptic encephalopathy with continuous spike and wave during sleep, EIMFS epilepsy of infancy with migrating focal seizures, TLE temporal lobe epilepsy, EME early myoclonic encephalopathy, LKS Landau-Kleffner syndromes, UEE unclassified epileptic encephalopathy, GLUT1-DS glucose transporter type 1 deficiency syndrome. Rett Rett syndrome, TSC tuberous sclerosis complex, SWS Sturge-Weber syndrome, UE unclassified refractory epilepsy
One hundred fifty-three epilepsy-related genes were selected for sequencing in all patients. The expression pattern of the targeted 153 genes across tissues were analyzed and classified according to the National Center for Biotechnology Information (NCBI, https://​www.​ncbi.​nlm.​nih.​gov) and The Human Protein Atlas (https://​www.​proteinatlas.​org) database (Additional file 1: Table S1). In our 153-gene panel, 51 genes show elevated expression, 14 genes have low expression, and 88 of them exhibit medium levels of expression in brain. The 14 low-expression genes have been associated with epilepsy, including: ARG1 [2427], ARHGEF15 [28], CASR [29, 30], CHRNA2 [31], DBH [3234], DIAPH3 [35], FOLR1 [36, 37], GABRA6 [38, 39], GLRA1 [40, 41], NID2 [42, 43], PROC [44], SLC13A5 [45, 46], SLC19A3 [47], SRPX2 [48]. Specifically, among 51 elevated genes in brain, 4 genes (GABRG2, GABBR2, GABRA1, GRIN1) show restricted brain expression.
The DNA samples of patients were analyzed by using NGS and the variants were validated by Sanger Sequencing. For the samples subjected to targeted sequencing, the quality assurance (QA) /quality control (QC) file are provided in Additional file 1: Table S2.
After sequencing the 153 epilepsy genes, we identified 43 deleterious variants in 23.3% patients (40 of 172), with three children harbouring more than one deleterious variant. Our results were similar to previous reports, with diagnostic yields ranging between 10% and 48.5% [4956]. There were 60.5% (26/43) novel deleterious variants found in our study. A total of 43 variants in 22 genes were scored as pathogenic or likely pathogenic, including SCN1A (16), TSC2 (5), STXBP1 (2), SCN8A (2), TSC1(1), MECP2 (1), CHD2 (1), PCDH19 (1), GABRA1 (1), GABRB3 (1), SLC2A1 (1), SLC9A6 (1), IQSEC2 (1), KCNQ2 (1), SCN2A (1), CACNA1A (1), KCNT1 (1), SYNGAP1 (1), ATP1A2 (1), CDKL5 (1), ADSL (1), VRK2 (1) (Fig. 1a). Among these 43 pathogenic or likely pathogenic variants, there were 18 (41.9%) missense mutations, 3 (7%) splice site mutations, 11 (25.6%) nonsense mutations, 10 (23.3%) frame-shifts, and 1 (2.3%) deletion mutations (Fig. 1a, Table 3).
Table 3
Pathogenic and likely pathogenic mutations adhered to ACMG guidelines in 172 refractory epilepsy children
Case code
Gene
Gene location
Transcript
cDNA change
Protein change
SIFT
PP2
MT
HSF
GERP++
MAF-ExAC
MAF-KG
Parental Origin
ACMG scoring
ACMG pathogenicity
Diagosis
13
SCN1A
chr2–166,901,702
NM_006920
c.1513A > T
p.K505X
A
6.17 (C)
De novo
PVS1 + PS2 + PM2
LP
DS
23
SCN1A
chr2–166,854,657 166,854,660 a [101]
NM_006920
c.4331_4334del
p.E1444fs
De novo
PVS1 + PS1 + PS2 + PM2
P
DS
26
SCN1A
chr2–166,870,270
NM_001165963
c.3689T>C
p.L1230P
D
D
D
5.28 (C)
De novo
PS2 + PM1 + PM2 + PP3
LP
DS
35
SCN1A
chr2–166,900,287 166,900,288
NM_001165963
c.1934_1935del
p.V645fs
De novo
PVS1 + PS2 + PM2
P
DS
38
SCN1A
chr2–166,859,121
NM_006920
c.G4112T
p.G1371V
D
P
D
5.54 (C)
De novo
PS2 + PM2
LP
DS
53
SCN1A
chr2–166,894,306 166,894,337
NM_001165963
c.2895_2926del
p.Q965fs
Unknown
PVS1 + PM2
LP
DS
56
SCN1A
chr2–166,908,355 a [102]
NM_006920
c.838T > C
p.W280R
D
D
D
5.41 (C)
De novo
PS1 + PS2 + PM2 + PP3
P
DS
65
SCN1A
chr2–166,850,927
NM_006920
c.4549-1G > C
splicing
D
+
5.76 (C)
De novo
PVS1 + PS2 + PM2
P
DS
115
SCN1A
chr2–166,848,614
NM_006920
c.5138C > A
p.A1713D
D
D
D
5.8 (C)
De novo
PS2 + PM2 + PP3
LP
DS
124
SCN1A
chr2–166,848,438 a [103]
NM_006920
c.5314G > A
p.A1772T
D
D
D
5.69 (C)
De novo
PS1 + PS2 + PM2 + PP3
P
DS
130
SCN1A
chr2–166,854,634 166,854,639 a [101]
NM_006920
c.4352_4356del
p.Y1451Cfs*22
De novo
PVS1 + PS1 + PS2 + PM2
P
DS
140
SCN1A
chr2–166,911,210 166,911,211
NM_006920
c.539delT
p.L180X
De novo
PVS1 + PS2 + PM2
P
DS
148
SCN1A
chr2–166,901,579
NM_001165963
c.1636G > T
p.E546X
A
6.17 (C)
Unknown
PVS1 + PM2
LP
DS
149
SCN1A
chr2–166,894,430 a [104]
NM_006920
c.2769G > A
p.M923I
D
D
D
5.18 (C)
Paternal
PS1 + PM2 + PP3
LP
DS
162
SCN1A
chr2–166,848,043 166,848,045
NM_001165963
c.5740_5742del
p.1914_1914del
De novo
PS2 + PM2 + PM4
LP
DS
172
SCN1A
chr2–166,903,330
NM_006920
c.1327G > T
p.E443X
A
5.31 (C)
De novo
PVS1 + PS2 + PM2
P
DS
93
SCN2A
chr2–166,243,416
NM_001040142
c.4712T > C
p.I1571T
D
D
D
5.17 (C)
De novo
PS2 + PM1 + PM2 + PP3
LP
OS
55
KCNQ2
chr20–62,073,781 a [105]
NM_172107
c.794C > T
p.A265V
D
P
D
3.38 (C)
De novo
PS1 + PS2 + PM2
P
OS
90
STXBP1
chr9–130,423,419 a [53]
NM_003165
c.364C > T
p.R122X
A
4.92 (C)
Unknown
PVS1 + PS1 + PM2
P
OS-WS
52
ADSL
chr22–40,745,935
NM_000026
c.253C > T
p.R85X
A
5.59 (C)
Maternal
PVS1 + PM2
LP
WS
chr22–40,742,633 [58]
NM_000026
c.71C > T
p.P24L
T
B
D
0.153 (N)
Paternal
PM2
UC
89
KCNT1
chr9–138,651,532 a [106]
NM_020822
c.862G > A
p.G288S
T
D
D
5.05 (C)
De novo
PS1 + PS2 + PM1 + PM2
P
WS
104
CDKL5
chrX-18,593,592 18,593,593
NM_003159
c.265delT
p.F89Lfs*24
De novo
PVS1 + PS2 + PM2
P
WS
151
STXBP1
chr9–130,428,529
NM_003165
c.748C > T
p.Q250X
A
5.72 (C)
De novo
PVS1 + PS2 + PM2
P
WS
29
SYNGAP1
chr6–33,393,659 33,393,662
NM_006772
c.274_277del
p.G92fs
De novo
PVS1 + PS2 + PM2
P
Doose
164
SLC2A1
chr1–43,396,517
NM_006516
c.296T > G
p.M99R
D
B
D
5.51 (C)
De novo
PS2 + PM2
LP
GLUT1-DS
30
MECP2
chrX-153,296,516 a [63]
NM_001110792
c.799C > T
p.R267X
A
3.55 (C)
De novo
PVS1 + PS1 + PS2 + PM2
P
Rett
32
TSC2
chr16–2,126,095 a [91]
NM_000548
c.2666C > T
p.A889V
D
D
D
5.09 (C)
Paternal
PS1 + PM2 + PP3
LP
TSC
94
TSC2
chr16–2,130,180 a [107]
NM_000548
c.3412C > T
p.R1138X
A
4.74 (C)
De novo
PVS1 + PS1 + PS2 + PM2
P
TSC
TSC2
chr16–2,130,366 a [66]
NM_000548
c.3598C > T
p.R1200W
D
D
D
4.74 (C)
De novo
PS1 + PS2 + PM2 + PP3
P
98
TSC2
chr16–2,138,467
NM_001077183
c.5079C > G
p.Y1693X
D
0.137 (N)
Paternal
PVS1 + PM2
LP
TSC
TSC2
chr16–2,138,465 2,138,466
NM_001077183
c.5077delT
p.Y1693fs
Paternal
PVS1 + PM2
LP
7
SCN8A
chr12–52,184,209 a [108]
NM_001177984
c.4324G > A
p.E1442K
D
D
D
4.68 (C)
Paternal
PS1 + PM2 + PP3
LP
UEE
IQSEC2
chrX-53,263,621 53,263,622
NM_001111125
c.4246_4247insG
p.S1416fs
De novo
PVS1 + PS2 + PM2
P
63
CACNA1A
chr19–13,566,019 a [109]
NM_001127221
c.301G > C
p.E101Q
D
D
D
5.01 (C)
De novo
PS1 + PS2 + PM1 + PM2 + PP3
P
UEE
66
SCN8A
chr12–52,200,885 a [110]
NM_001177984
c.5492G > A
p.R1831Q
D
D
D
4.91 (C)
De novo
PS1 + PS2 + PM2 + PP3
P
UEE
69
PCDH19
chrX-99,551,873 99,551,874
NM_001184880
c.2849-1G > −
splicing
+
Unknown
PVS1 + PM2
LP
UEE
157
GABRB3
chr15–26,812,802 a [111]
NM_021912
c.761C > T
p.S254F
D
D
D
6.06 (C)
De novo
PS1 + PS2 + PM1 + PM2 + PP3
P
UEE
160
GABRA1
chr5–161,309,645 a [112]
NM_001127648
c.641G > A
p.R214H
D
D
D
5.34 (C)
De novo
PS1 + PS2 + PM1 + PM2 + PP3
P
UEE
54
CHD2
chr15–93,540,231
NM_001271
c.3640G > T
p.G1214X
A
5.64 (C)
De novo
PVS1 + PS2 + PM2
P
UEE
40
VRK2
chr2–58,312,086
NM_001130483
c.C256 + 1G > A
splicing
D
+
5.86 (C)
Unknown
PVS1 + PM2
LP
UE
44
ATP1A2
chr1–160,098,521
NM_000702
c.1097G > T
p.G366V
D
D
D
4.77 (C)
De novo
PS2 + PM1 + PM2 + PP3
LP
UE
68
TSC1
chr9–135,772,854
NM_000368
c.2768_2769insC
p.L924Ffs*26
De novo
PVS1 + PS2 + PM2
P
UE
79
SLC9A6
chrX-135,080,322 135,080,336
NM_001042537
c.582_595del
p.Y194fs
De novo
PVS1 + PS2 + PM2
P
UE
Abbreviations: M male, F female, m month, y year, SIFT Sorts intolerant from tolerant (D, damaging; T, tolerant), PP2, polymorphism phenotyping v2 (D, damaging; P, possible damaging; B, benign), MT mutation taster (D, disease causing; A, disease causing automatic), HSF human splicing finder (+, altering splicing), GERP++ genomic evolutionary rate profiling (C, conserved; N, nonconserved), KG 1000 Genomes project, LP likely pathogenic, P pathogenic, DS Dravet syndrome, OS Ohtahara syndrome, OS-WS OS syndrome evolves to West syndrome, WS West syndrome, Doose Doose syndrome, GLUT1-DS glucose transporter type 1 deficiency syndrome, Rett Rett syndrome, TSC tuberous sclerosis complex, UEE unclassified epileptic encephalopathy, UE unclassified refractory epilepsy
a Mutations have been reported in HGMD database
More recent studies suggest that many severe epilepsy types begin in infancy or childhood, especially those with psychomotor retardation and epileptic encephalopathies are often due to de novo mutations [30, 31]. In our study, 32/43 (74.4%) pathogenic or likely pathogenic variants were de novo, five (11.6%) were paternal, one (2.3%) was maternal, and five (11.6%) were unknown due to blood samples from parents were unavailable (Table 3).
To further explore the genetic pathogenesis of epilepsy, we subdivided the mutated genes into nine groups according to the molecular and biological function of the gene produce. These functional groups included voltage-gated ion channels, enzyme/enzyme modulators, membrane trafficking, ligand-gated ion channels, DNA/RNA binding, cell-adhesion proteins, glucose transporter, proton antiporter, and GTP/GDP exchanges. Variants in ion channel genes (SCN1A, SCN2A, SCN8A, CACNA1A, KCNT1, KCNQ2) accounted for 51.2% (22/43) of the pathogenic or likely pathogenic variants. Variants in enzyme/enzyme modulator genes (TSC1, TSC2, SYNGAP1, ATP1A2, CDKL5, ADSL, VRK2) accounted for 25.6% (11/43) of pathogenic or likely pathogenic variants. Variants in genes encoded membrane trafficking (STXBP1), ligand-gated ion channels (GABRA1, GABRB3), DNA/RNA binding proteins (MECP2, CHD2) each accounted for 4.7% (2/43) (Fig. 1b). Ion channels (voltage-gated and ligand-gated) accounted for 55.8% in total, suggesting that dysfunction of ion channels plays critical roles in the pathogenesis of epilepsy.
We then analyzed the yield of the epilepsy gene panel testing based on electroclinical syndrome (Fig. 1c). The yield of deleterious variants in Dravet syndrome (69.6%, 16/23) and glucose transporter type 1 deficiency syndrome (100%, 1/1) patients was higher than that in others. Patients with onset age of seizures ≤12 months had higher yields of deleterious variants compared to those with onset age of seizures > 12 months (31/101 vs 9/71; χ2 = 7.583, df = 1, P = 0.006). The family history did not affect whether or not a deleterious genetic variant was identified (7/27 vs 33/145; χ2 = 0.128, df = 1, P = 0.804).
There were 16 mutations in SCN1A gene, of which six (37.5%) were missense mutations, one (6.25%) was a splice site mutation, four (25%) were nonsense mutations, four (25%) were frame-shifts, and one (6.25%) was deletion mutation. Thirteen of the 16 (81.3%) SCN1A mutations were de novo and 11 (68.8%) were novel. We further analysed the positions of the mutations in the affected proteins corresponding to gene mutations and found that 43.8% (7/16) of protein changes are in the intracellular loop of sodium channel protein type 1 subunit alpha, 31.3% (5/16) are in the extracellular loop, 18.8% (3/16) are in the transmembrane region, and 6.25% (1/16) are in the pore forming area (Fig. 2).
There has been a marked increase in genetic diagnoses of a number of key childhood-onset epilepsy syndromes, such as Dravet syndrome, which has been mainly linked to SCN1A [17]. In our 16 patients diagnosed as Dravet syndrome with pathogenic or likely pathogenic variants, all identified mutations were in the SCN1A gene. These 16 Dravet syndrome patients had typical manifestations: onset between 3 to 8 months of age, fever-sensitive, multiple seizure types, and developmental delay after seizure onset. 81.25% (13/16) SCN1A mutations were de novo in Dravet syndrome patients and one was inherited from the father who had a history of febrile seizures (FS). 12.5% (2/16) SCN1A mutations were unknown. Pathogenic and likely pathogenic mutations each accounted for 50% (Table 4). 50% (8/16) of the Dravet syndrome variants cause nonsense or frameshift mutations that result in truncated proteins, which was consistent with a previous study [57]. We evaluated whether different seizure types, family history, abnormal brain MRI, or developmental delay were associated with specific SCN1A mutation types or locations within the gene. We did not detect any bias towards particular regions of the gene or in the type of mutation, although our small sample size did not provide substantial power (Additional file 1: Tables S3 and S4).
Table 4
Clinical features in DS patients
Case code
Gender/age
Diagosis
Age of onset
Seizure types
EEG
Brain MRI/CT
Developmental delay
Gene
cDNA change
Protein change
Parental Origin
ACMG pathogenicity
13
F/2y6m
DS
3m
FS, FoS, Myo
FSW
Normal
Yes
SCN1A
c.1513A > T
p.K505X
De novo
LP
23
F/3y
DS
7m
FS, FoS (A), Myo, FBTC
Multi. FD
Underdeveloped myelin
Yes
SCN1A
c.4331_4334del
p.E1444fs
De novo
P
26
F/5y11m
DS
5m
FS, SE, FoS (A), Myo, FBTC
FSW
Normal
Yes
SCN1A
c.3689T>C
p.L1230P
De novo
LP
35
M/4y
DS
3m
FS, SE, GTCS, aAb
Multi. FD
Normal
Yes
SCN1A
c.1934_1935del
p.V645fs
De novo
P
38
F/1y6m
DS
4m
FS, SE, Myo
FSW
Nonspecific
Yes
SCN1A
c.G4112T
p.G1371V
De novo
LP
53
M/5y
DS
7m
FS, aAb, Myo, Fos (I)
Multi. FD
Normal
Yes
SCN1A
c.2895_2926del
p.Q965fs
Unknown
LP
56
F/3y6m
DS
5m
FS, Myo, GTCS, SE, FoS (A), aAb
Multi. FD
Nonspecific
Yes
SCN1A
c.838T > C
p.W280R
De novo
P
65
M/2y4m
DS
5m
FS, SE, FoS (A)
FSW
Normal
Yes
SCN1A
c.4549-1G > C
splicing
De novo
P
115
M/2y1m
DS
8m
FS, FoS (I), FoS (hemi clonic), GTCS
FSW
Enlargement of the subarachnoid space in front of left temporal lobe
Yes
SCN1A
c.5138C > A
p.A1713D
De novo
LP
124
M/3y
DS
5m
FS, FoS (A), FBTC
FSW
Nonspecific
Yes
SCN1A
c.5314G > A
p.A1772T
De novo
P
130
F/11y
DS
6m
FS, FoS (A), aAb, Myo, GTCS
Multi. FD
Normal
Yes
SCN1A
c.4352_4356del
p.Y1451Cfs*22
De novo
P
140
F/1y9m
DS
3m
FS, GTCS, C, FoS (I)
FSW
Normal
Yes
SCN1A
c.539delT
p.L180X
De novo
P
148
F/6y8m
DS
4m
FS, GTCS, FoS, aAb
Multi. FD
Normal
Yes
SCN1A
c.1636G > T
p.E546X
Unknown
LP
149
M/3y6m
DS
4m
FS, FoS (A), Myo, GTCS
Multi. FD, GSW
Normal
Yes
SCN1A
c.2769G > A
p.M923I
Paternal (FS)
LP
162
M/4y
DS
5m
FS, FoS (A), Myo, FBTC
Multi. FD
Normal
Yes
SCN1A
c.5740_5742del
p.1914_1914del
De novo
LP
172
F/8y
DS
5m
FS, aAb, Myo, FBTC
Multi. FD, GSW, GPSW
Normal
Yes
SCN1A
c.1327G > T
p.E443X
De novo
P
Abbreviations: M male, F female, m month, y year, P pathogenic, LP likely pathogenic, UC uncertain, DS Dravet syndrome, FS febrile seizures, SE status epilepticus, FoS focal seizures, FoS (I) focal seizures (impaired awareness), FoS (A) focal seizures (aware), FBTC focal to bilateral tonic-clonic, Myo myoclonic, aAb atypical absence, GTCS generalized tonic-clonic seizures, FSW focal spike wave, Multi. FD multifocal discharges, GSW generalized spike-wave, GPSW generalized polyspike-wave
Twelve patients presented typical manifestation of Ohtahara syndrome: onset age within postnatal 30 days, tonic spasms, burst suppression EEG and developmental delay. Pathogenic or likely pathogenic variants in Ohtahara syndrome were in the KCNQ2 (1), STXBP1 (1), SCN2A (1) genes. The nonsense mutation in STXBP1 (c.364C > T, p.R122X) was detected in one Ohtahara syndrome patients that evolved to West syndrome. This patient had an onset age of postnatal 17 day, spasms, and burst-suppression EEG at postnatal 22 day and hypsarrhythmia EEG at 4 months (Table 5).
Table 5
Clinical features in OS, WS, LGS, Doose, GLUT1-DS, Rett, TSC, UEE and UE patients
Case code
Gender/age
Diagosis
Age of onset
Seizure types
EEG
Brain MRI/CT
Developmental delay
Gene
cDNA change
Protein change
Parental Origin
ACMG pathogenicity
55
M/54d
OS
1d
FoS, Tonic spasms
BS, FSW
Normal
Yes
KCNQ2
c.794C > T
p.A265V
De novo
P
93
M/40d
OS
3d
Tonic spasms
BS
Normal
Yes
SCN2A
c.4712T > C
p.I1571T
De novo
LP
90
M/2y11m
OS-WS
17d
Tonic spasms, Spa.
BS, Hypsarrhy.
Normal
Yes
STXBP1
c.364C > T
p.R122X
Unknown
P
52
F/1y8m
WS
2m
Spa.
Multi. FD, Hypsarrhy.
Cerebral dysplasia
Yes
ADSL
c.253C > T
p.R85X
Maternal
LP
ADSL
c.71C > T
p.P24L
Paternal
UC
89
F/1y11m
WS
19d
FoS, Spa.
Multi. FD, Hypsarrhy.
Subdural hemorrhage
Yes
KCNT1
c.862G > A
p.G288S
De novo
P
104
F/2y10m
WS
3m7d
Spa.
Hypsarrhy., Multi.FD
Normal
Yes
CDKL5
c.265delT
p.F89Lfs*24
De novo
P
151
F/9m
WS
3m
Spa.
Hypsarrhy., Multi. FD
Enlargement of the subarachnoid space
Yes
STXBP1
c.748C > T
p.Q250X
De novo
P
29
M/5y6m
Doose
1y3m
Myo-At., Myo, aAb
Abnormal background theta, GSW, GPSW
Normal
No
SYNGAP1
c.274_277del
p.G92fs
De novo
P
164
F/6y
GLUT1-DS
2y4m
GTCS
FSW, Multi. FD
Nonspecific (Hair loss leads to bald)
No
SLC2A1
c.296T > G
p.M99R
De novo
LP
30
F/4y4m
Rett
3y2m
Fos (I), FBTC
Multi. FD
Normal
Yes
MECP2
c.799C > T
p.R267X
De novo
P
32
M/8y
TSC
1y6m
FoS (I), FBTC
Multi. FD
Multi nodules
No
TSC2
c.2666C > T
p.A889V
Paternal
LP
94
F/9m
TSC (WS)
3m
Spa.
Multi. FD, Hypsarrhy.
Multi nodules
Yes
TSC2
c.3412C > T
p.R1138X
De novo
P
TSC2
c.3598C > T
p.R1200W
De novo
P
98
M/3y
TSC (WS)
4m
Spa., aAb
Multi. FD, Hypsarrhy.
Nonspecific
Yes
TSC2
c.5079C > G
p.Y1693X
Paternal
LP
TSC2
c.5077delT
p.Y1693fs
Paternal
LP
7
M/2y
UEE (EIEE13)
6m
FoS (I), FBTC
Multi. FD
Enlargement of the subarachnoid space
Yes
SCN8A
c.4324G > A
p.E1442K
Paternal
LP
IQSEC2
c.4246_4247insG
p.S1416fs
De novo
P
63
M/4y
UEE (EIEE42)
5m
FoS, GTCS
Multi. FD
Normal
Yes
CACNA1A
c.301G > C
p.E101Q
De novo
P
66
M/1y9m
UEE (EIEE13)
4m
FBTC, FoS
Multi. FD
Enlargement of the subarachnoid space
Yes
SCN8A
c.5492G > A
p.R1831Q
De novo
P
69
F/2y1m
UEE (EIEE9)
1y3m
FBTC, C, T
Multi. FD
Normal
Yes
PCDH19
c.2849-1G > −
splicing
Unknown
LP
157
F/2y
UEE (EIEE43)
2m
C, FoS (I)
FSW
Normal
Yes
GABRB3
c.761C > T
p.S254F
De novo
P
160
M/6y
UEE (EIEE19)
6m
FoS (I), GTCS
FSW
Normal
Yes
GABRA1
c.641G > A
p.R214H
De novo
P
54
F/7y
UEE (EEOC)
4y2m
SE, GTCS, FoS (I)
Mult. FD
Normal
Yes
CHD2
c.3640G > T
p.G1214X
De novo
P
40
F/2y11m
UE
4m
FoS
FSW
Normal
No
VRK2
c.C256 + 1G > A
splicing
Unknown
LP
44
F/5y6m
UE
4y
FoS (automatisms, emotional)
Multi. FD
Nodules in internal side of left anterior limb of internal capsule; caput of caudate nucleus or heterotopic gray matter
Yes
ATP1A2
c.1097G > T
p.G366V
De novo
LP
68
M/6y
UE
4y
FoS (A)
FSW
Normal
No
TSC1
c.2768_2769insC
p.L924Ffs*26
De novo
P
79
M/3y
UE
1y2m
FoS (I), FBTC
Multi. FD
Normal
Yes
SLC9A6
c.582_595del
p.Y194fs
De novo
P
Abbreviations: M male, F female, m month, y year, P pathogenic, LP likely pathogenic, UC uncertain, OS Ohtahara syndrome, OS-WS Ohtahara syndrome evolves to West syndrome, WS West syndrome, Doose Doose syndrome, GLUT1-DS glucose transporter type 1 deficiency syndrome, Rett Rett syndrome, TSC tuberous sclerosis complex, UEE unclassified epileptic encephalopathy, UE unclassified refractory epilepsy, EEIE early-infantile epileptic encephalopathies, EEOC childhood-onset epileptic encephalopathy, Spa. Spasms, FoS focal seizures, FoS (I) focal seizures (impaired awareness), FoS (A) focal seizures (aware), FBTC focal to bilateral tonic-clonic, T tonic, C clonic, Myo myoclonic, aAb atypical absence, At. atonic, GTCS generalized tonic-clonic seizures, SE status epilepticus, BS burst suppression, Hypsarrhy. hypsarrhythmia, Multi. FD multifocal discharges, FSW focal spike-wave, GSW generalized spike-wave, GPSW generalized polyspike-wave
West syndrome patients in our study had onset ages of seizures ranging from postnatal 19 days to 6 months. Typical clinical manifestations were all observed, including spasms, hypsarrhythmia EEG, and developmental delay. 16.7% (2/12) of the West syndrome children evolves to Lennox-Gastaut syndrome. After sequencing, we identified 4 pathogenic or likely pathogenic mutations in the following genes: STXBP1 (1), KCNT1 (1), CDKL5 (1), ADSL (1). 75% (3/4) of these variants were de novo.
One of the West syndrome patients were found to carry two mutations: a nonsense ADSL (c.253C > T, p.R85X) mutation was scored as likely pathogenic and was inherited from her unaffected mother. Another reported missense ADSL (c.71C > T, p.P24L) [58] mutation which was inherited from her unaffected father were scored as uncertain pathogenicity. ADSL has been reported to be related to adenylosuccinate lyase deficiency, which is an autosomal recessive defect of purine metabolism [59, 60]. The patient presented with spasms 2 months after birth. Brain MRI showed cerebral dysplasia and EEG showed hypsarrhythmia and multifocal discharges. The patient also had developmental delay and lack of eye contact. A definitive diagnosis can be made with high performance liquid chromatography examination of the urine to detect the ratio of succinyladenosine and succinyl-aminoimidazole carboximide riboside, but this was not available for the patient in question. Thus, this patient was diagnosed clinically as having West syndrome.
A novel frame-shift mutation in SYNGAP5 (c.274_277del, p.G92fs) was detected in a patient with Doose syndrome. This patient presented with myoclonic and myoclonic-astatic seizures, as well as having atypical absence seizures. SYNGAP5 had been reported to be associated with Doose syndrome and mental retardation, autosomal dominant 5 (MRD5) [51, 61, 62]. This mutation, which is very rare, was de novo, and caused frameshift changes in Ras/Rap GTPase-activating protein SynGAP, was therefore scored as pathogenic (Table 5).
One glucose transporter type 1 deficiency syndrome patient presented with seizures at age 28 months. The patient has alopecia and was almost bald at 4 years old. The child did not have other abnormalities in blood tests, brain MRI, or neurological exam. Her cerebrospinal fluid glucose value was 2.04 mmol/L (blood glucose value was 7.2 mmol/L before lumbar puncture; fasting blood glucose value was 5.2 mmol/L). NGS identified a missense mutation in SLC2A1 (c.296T > G, p.M99R). The mutation was de novo and novel. The patient’s parents and sister were normal, which is consistent with the sequencing results. Symptoms improved with a ketogenic diet, with seizures controlled for more than 6 months.
One MECP2 mutation (c.799C > T, p.R267X) was detected in a girl diagnosed as Rett syndrome. The girl developed normally for the first 18 months, gradually lost speech ability while developing repetitive hand-wringing. Seizures began at age 3 years. The MECP2 gene is located on the X-chromosome, and Rett syndrome is inherited through this gene in a dominant fashion [63]. This patient had a de novo MECP2 nonsense mutation, consistent with her parents being unaffected.
40% (2/5) of tuberous sclerosis complex patients were diagnosed with West syndrome associated with tuberous sclerosis complex in our study. Tuberous sclerosis complex is closely related to the TSC1/TSC2 genes [6467].
In our study, all of the tuberous sclerosis complex patients’ initial presentations were seizures, of which 80% (4/5) presented in the first year of life. 60% (3/5) had hypomelanotic macules and 40% (2/5) had multi nodules. One patient’s only clinical manifestation was seizures and three (60%) patients with seizures had only one major feature of tuberous sclerosis complex. After sequencing, 60% (3/5) patients were found to have deleterious TSC1 or TSC2 mutations.
We identified more than one TSC1/2 mutations in 2 patients. One patient has two TSC2 mutations inherited from his affected father. Facial angiofibromas appeared by age 3–4 years in 60% (3/5) patients in the follow-up period. Gilboa et al. [68] reported four patients with the same TSC1 genomic deletion (9q34.13q34.2) in a family and none of them fulfilled the clinical criteria for tuberous sclerosis complex. In our study, one patient with pathogenic TSC1 (c.2768_2769insC, p.L924Ffs*26) mutation presented with focal seizures beginning at age four. There were two hypopigmented macules on the patient’s abdomen. The brain MRI results were normal and there are no other features of tuberous sclerosis complex. This de novo mutation causes a frame-shift in hamartin and has not been reported previously. Thus, this patient was considered to have unclassified refractory epilepsy.
One unclassified epileptic encephalopathy patient had two deleterious mutations: SCN8A inherited from his affected father (c.4324G > A, p.E1442K) and IQSEC2 (c.4246_4247insG, p.S1416fs). Early-infantile epileptic encephalopathies (EIEE) caused by SCN8A mutations are designated as EIEE13 (OMIM #614558) [69]. The missense mutation in SCN8A is very rare in the general population, and had been previously predicted to be damaging by SIFT, MT and PP2. IQSEC2 is an X-linked gene that has been reported to be related to intellectual disability and epilepsy, and it encodes the IQ motif and SEC7 domain-containing protein 2 [70]. The identified novel IQSEC2 mutation was de novo and was scored as being pathogenic.
Other pathogenic or likely pathogenic mutations found in patients with unclassified epileptic encephalopathy included CACNA1A, GABRA1, GABRB3, PCDH19, and CHD2. Epileptic encephalopathies with the above mutations had been designated as EIEE42, EIEE19, EIEE43, EIEE9 and EEOC (childhood-onset epileptic encephalopathy) according to Online Mendelian Inheritance in Man (OMIM). Other deleterious variants found in patients with unclassified refractory epilepsy were in VRK2, ATP1A2, and SLC9A6. Taking these unclassified epileptic encephalopathies and unclassified refractory epilepsy patients’ clinical manifestations into consideration, we found that all patients with deleterious mutations in genes encoding ion channels (SCN8A, CACNA1A, GABRB3, GABRA1) had similar clinical symptoms: onset age of seizures within the first year, epileptic encephalopathy and developmental delay. In contrast, patients with mutations in VRK2, ATP1A2, and SLC9A6, had relatively later onset age of seizures.
We then assessed the clinical benefit of genetic testing in those patients with identified deleterious variants. NGS helped with the diagnosis (n = 8), medication selection (n = 18), reproductive planning (n = 4), and treatment planning (n = 1). The finding of the SLC2A1 variant in Case 164 prompted other tests such as cerebrospinal fluid (CSF) glucose that were clinically useful. Identification of deleterious SCN1A mutations in five young infants with clinically suspected Dravet syndrome helped early diagnosis (Case 13, 38, 65, 115, 140) and led to the discontinuation of oxcarbazepine (Case 13) that exacerbated seizures. Identification of SCN1A mutations in other Dravet syndrome patients helped to avoid sodium channel blockers such as oxcarbazepine, carbamazepine and lamotrigine. Among the four Dravet syndrome patients who responded to anticonvulsants (Case 13, 26, 149, 172), 75% (3/4) of them were prescribed sodium valproate or clonazepam suggesting that these medications may be effective in Dravet syndrome. The finding of the TSC2 variants in Cases 94 and 98 helped early diagnosis and Case 32 experienced remission with administration of rapamune. Identification of TSC1 prompted clinical surveillance for tuberous sclerosis complex in Case 68. The findings of patients with deleterious variants in TSC2 (Case 32, 98), SCN8A (Case 7), SCN1A (Case 149), ADSL (Case 52) which were inherited, helped in prenatal counselling (Table 6).
Table 6
Clinical benefits after molecular diagnosis
Clinical benefits
Effects (Case details)
Diagnosis
SLC2A1 (GLUT1-DS)
Definitive diagnosis (Case 164)
SCN1A (DS)
Definitive diagnosis (Case 13, 38, 65, 115, 140)
TSC2 (TSC)
Definitive diagnosis (Case 94, 98)
Management implications
SLC2A1, using KD
Controlled (Case 164, KD)
SCN1A, stopping OXC
Remitted (Case 13, VPA, TPM,10–20 / month)
SCN1A, avoiding OXC, CBZ, and LTG
Remitted (Case 23, VPA, TPM, seizure-free for 5 months; Case 26, LEV, TPM, CZP, seizure-free for 6 months; Case 149, VPA, TPM, LEV, CZP, seizure-free for 4 months; Case 172, VPA, TPM, CZP, seizure-free for 1 year)
Uncontrolled (Case 35, 38, 53, 56, 65, 115, 124, 130, 140, 148, 162)
TSC2, using rapamune
Remitted (Case 32, seizure-free for 7 months)
Long-term follow up
TSC1 (risk of TSC)
Case 68
Reproductive planning
Suggesting the family conduct genetic counseling
TSC2 (Case 32, 98), SCN8A (Case 7), SCN1A (Case 149), ADSL (Case 52)
Abbreviations: DS Dravet syndrome, GLUT1-DS glucose transporter type 1 deficiency syndrome, Rett Rett syndrome, TSC tuberous sclerosis complex, KD ketogenic diet, OXC oxcarbazepine, CBZ carbamazepine, LTG lamotrigine, VPA sodium valproate, TPM topiramate, LEV levetiracetam, CZP clonazepam

Discussion

Epilepsy is highly heterogeneous and can be primarily genetic in origin, or be secondary to structural or metabolic disorders of the central nervous system [71, 72]. To date, over 500 genes have been implicated in epilepsy [7376]. However, the overlapping clinical features of different epilepsy syndromes and non-specific phenotypes can hamper clinical and genetic diagnosis [53]. The correct genetic diagnosis can help to guide treatment and prognosis. In addition to genetic origins, pediatric epilepsy may also arise from epigenetic mechanisms mediating gene-environment interactions during neurodevelopment. In this study, we used NGS to investigate 153 epilepsy related genes in a cohort of 172 refractory epilepsy children.
Approximately one quarter of genes identified in epilepsy encode ion channel proteins, including voltage-gated channels (Na+, K+, Ca2+ channels and hyperpolarization-activated cyclic nucleotide-gated channels) and ligand-gated ion channels (N-Methyl-D-Aspartate receptors, Gamma-aminobutyric acid receptors and Nicotinic Acetylcholine receptors) [77]. The genes that encode ion channels and are relevant to epilepsy include SCN1A, SCN1B, SCN2A, SCN8A, KCNA1, KCNA2, KCNB1, KCNC1, KCNMA1, KCNQ2, KCNQ3, KCNT1, KCTD7, HCN1, CACAN1A, CACNA1H, GRIN1, GRIN2A, GRIN2B, GRIN2D, GABRA1, GABRB3, GABRG2, CHRNA2, CHRNA4, CHRNB2. In our study, 51.2% pathogenic or likely pathogenic variants were found in voltage-gated ion channels and 4.7% were found in ligand-gated ion channels. Thus, we further confirmed that ion channels play an important role in the pathogenesis of epilepsy.
An SCN1A mutation was first discovered in epilepsy in 2000 [72], and now hundreds of new SCN1A mutations have been described in epilepsy patients, making it the most common epilepsy-related gene [78]. In our study, we found SCN1A mutations in 16/44 deleterious variants, making it the most common gene to show variation in our study. SCN1A encodes the Nav1.1 pore-forming α-subunit, expressed mainly in inhibitory GABAergic neurons. The α-subunit comprises four homologous domains (I–IV), forming a tetrameric structure. Each domain is composed of six transmembrane segments (S1–S6) [77]. The S4, voltage-sensing segment has multiple positively charged amino acids. The intracellular loop between III and IV domain functions as the inactivation gate. The α-subunit is usually associated with two β-subunits that influence α-subunit localization and function [77]. Among α-subunit of sodium channel genetic variants in our study, 43.8% (7/16) are within the intracellular loop, 31.3% (5/16) in the extracellular loop, 18.8% (3/16) in the transmembrane area, and 6.25% (1/16) in the pore forming area. All the extracellular mutations are between S5 and S6, which is very close to the pore forming area. These variants may influence the initiation and propagation of action potentials, making these inhibitory GABAergic neurons less excitable. Some antiepileptic drugs (AEDs) bind to the inner cavity of the pore of the sodium channel (IS6, IIIS6 and IVS6) [77, 79]. The pore forming area or internal/external loop could be promising targets for new seizure prophylaxis medications.
Patients harboring SCN1A mutations can have with Dravet syndrome or generalized epilepsy with febrile seizures plus. One Dravet syndrome patient inherited the SCN1A mutations from his father only had febrile seizures. This could be due to somatic mosaicism [72, 80, 81]. A Dravet syndrome mouse model (Nav1.1 knockout-based) responded well to stiripentol and clobazam, which are commonly used to treat Dravet syndrome [8285]. One of the patients in our study was treated with oxcarbazepine, which blocks sodium channels and worsened seizures, before the diagnosis of Dravet syndrome was made. This case illustrates the importance of correct molecular diagnosis in selecting the best anticonvulsant.
Approximately half of Ohtahara syndrome patients with STXBP1 mutations evolve to West syndrome [86]. In our study, there was one such patient with a nonsense mutation in STXBP1, suggesting that this gene could play a role in the etiology of West syndrome. Our findings also suggest that STXBP1 is related to both Ohtahara syndrome and West syndrome.
KCNT1 is associated with epilepsy of infancy with migrating focal seizures, autosomal dominant nocturnal frontal lobe epilepsy, and other types of early onset epileptic encephalopathies [8789]. Ohba et al. [88] found 11 KCNT1 mutations in a total of 362 epilepsy patients: 9/18 epilepsy of infancy with migrating focal seizures cases (50%), 1/180 West syndrome cases (0.56%), and 1/66 unclassified early onset epileptic encephalopathy cases (1.52%), suggesting that KCNT1 may be a causal gene for West syndrome. In our study, one KCNT1 (c.862G > A, p.G288S) mutation was found in a patient diagnosed as West syndrome.
Genetic studies of neuropsychiatric disease have led to the discovery of molecular etiology and pathophysiology. For example, most cases of Rett syndrome are now known to arise from mutations in the MECP2 gene, which codes for a methyl-CpG-binding protein 2 [90]. Another example is glucose transporter type 1 deficiency syndrome, which has been attributed to variants in SLC2A1, SLC2A2, and GLUT1. In our study, the glucose transporter type 1 deficiency syndrome patient did not have cerebrospinal fluid analysis as part of their diagnostic work-up until the genetic data suggested the diagnosis. This example illustrates the utility of NGS in clinical scenarios, and in time this may become an important part of the evaluation of pediatric patients with epilepsy. In some epilepsy syndromes, crucial interventions such as diet modification can have dramatic beneficial effects, so early diagnosis is vital [91, 92].
In our study, SCN1A was the main deleterious variant in Dravet syndrome and KCNQ2, STXBP1, SCN2A were found in Ohtahara syndrome. Deleterious variants in STXBP1, KCNT1, CDKL5, ADSL genes were found in West syndrome. Novel mutations in SYNGAP1 were found in Doose syndrome, a SLC2A1 mutation was found in GLUT1-DS and a de novo MECP2 mutation were found in Rett syndrome. TSC1/TSC2 variants were found in 60% of patients with tuberous sclerosis complex. Mutations found in unclassified epileptic encephalopathy were mainly in ion-channel genes. Thus, our study reinforces previous observations that the clinical syndrome and genetic etiology do not always match.
We tested 153 epilepsy genes and found 43 pathogenic and likely pathogenic variants in this study. Considering that over 500 epilepsy genes have been reported [7376], our work was not comprehensive, which is a limitation of this study. With the decreasing cost of whole genome sequencing, the interrogation of the entire genome is now feasible for larger samples of epilepsy patients, and this approach has already been fruitful in other neuropsychiatric disorders such as autism, Kabuki syndrome, Bohring-Opitz syndrome and others [93, 94].
For genetic testing, it is proposed to conduct the strong candidate gene sequencing first (SCN1A for Dravet syndrome, MECP2 for Rett syndrome and TSC1/2 for tuberous sclerosis complex) before a NGS multi-gene panel testing [9597]. In our study, we conducted targeted panel sequencing on Dravet syndrome and Rett syndrome patients before screening the strongest candidate gene for the following reasons. First, the correct clinical diagnosis of these syndromes can be difficult, especially in some of the younger patients in our sample, and often requires longitudinal assessment, which delays the correct diagnosis. Thus, we elected to perform NGS on our subjects before knowing the clinical diagnosis in some cases, such as these syndromes. Since our NGS panel that contains 153 epilepsy genes, our approach could facilitate the correct diagnosis in some cases. Second, it is now apparent that while 70–80% Dravet syndrome patients have SCN1A mutations, mutations in other genes such as SCN1B, SCN2A, SCN8A, PCDH19, GABRA1, GABRG2, STXBP1, CHD2 genes can cause Dravet syndrome like phenotypes [98], which would be missed if only SCN1A was sequenced. Similarly, CDKL5 and FOXG1 have been associated with atypical Rett syndrome [99], in addition to MECP2.
In tuberous sclerosis complex patients, we have a similar clinical scenario in which most features of tuberous sclerosis complex become evident only after 3 years of age, limiting their usefulness for early diagnosis [100]. In our study, all of the tuberous sclerosis complex patients’ initial presentations were seizures, of which 80% presented in the first year of life. 60% had hypomelanotic macules and 40% had multi nodules. 20% patient’s only clinical manifestation was seizures and 60% patients with seizures had only one major feature of tuberous sclerosis complex. 60% patients were found to have deleterious TSC1 or TSC2 mutations by NGS sequencing. Facial angiofibromas appeared by age 3–4 years in 60% patients in the follow-up period.
In summary, we identified 43 pathogenic or likely pathogenic variants, of which 26 mutations were novel and 32 were de novo. Variants in ion channel genes accounted for the largest category of gene in children with refractory epilepsy. Dravet syndrome is closely related to the SCN1A gene, which was the most frequently-appearing gene showing variants in our study. Novel and de novo mutations were found in Ohtahara syndrome, West syndrome, Doose syndrome and tuberous sclerosis complex pediatric patients. We also found a novel mutation in glucose transporter type 1 deficiency. Our results reinforce the importance and feasibility of precise genetic diagnosis for epilepsy, with the hope that in future, this will both aid in understanding the molecular pathophysiology and lead to new treatment targets.

Acknowledgements

All authors greatly appreciate to the families that take part in this research.

Funding

The project was funded by National Key Research and Development Program of China (NO. 2016YFC1306202), Key Research and Development Plan in Shandong Province (NO. 2016GSF201073), and General program of Qilu hospital, Shandong University (NO.2015QLMS08).

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
The program adhered to guidelines of patients’ consent for participation and research and was supported by the Ethics Committee of Qilu hospital, Shandong University (No. 2016(027)).
All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Henshall DC, Hamer HM, Pasterkamp RJ, Goldstein DB, Kjems J, Prehn JHM, et al. MicroRNAs in epilepsy: pathophysiology and clinical utility. Lancet Neurol. 2016;15(13):1368–76.PubMedCrossRef Henshall DC, Hamer HM, Pasterkamp RJ, Goldstein DB, Kjems J, Prehn JHM, et al. MicroRNAs in epilepsy: pathophysiology and clinical utility. Lancet Neurol. 2016;15(13):1368–76.PubMedCrossRef
2.
Zurück zum Zitat Nickels KC, Zaccariello MJ, Hamiwka LD, Wirrell EC. Cognitive and neurodevelopmental comorbidities in paediatric epilepsy. Nat Rev Neurol. 2016;12(8):465–76.PubMedCrossRef Nickels KC, Zaccariello MJ, Hamiwka LD, Wirrell EC. Cognitive and neurodevelopmental comorbidities in paediatric epilepsy. Nat Rev Neurol. 2016;12(8):465–76.PubMedCrossRef
3.
Zurück zum Zitat Pitkanen A, Loscher W, Vezzani A, Becker AJ, Simonato M, Lukasiuk K, et al. Advances in the development of biomarkers for epilepsy. Lancet Neurol. 2016;15(8):843–56.PubMedCrossRef Pitkanen A, Loscher W, Vezzani A, Becker AJ, Simonato M, Lukasiuk K, et al. Advances in the development of biomarkers for epilepsy. Lancet Neurol. 2016;15(8):843–56.PubMedCrossRef
4.
Zurück zum Zitat Miller LL, Pellock JM, DeLorenzo RJ, Meyer JM, Corey LA. Univariate genetic analyses of epilepsy and seizures in a population-based twin study: the Virginia Twin Registry. Genet Epidemiol. 1998;15(1):33–49.PubMedCrossRef Miller LL, Pellock JM, DeLorenzo RJ, Meyer JM, Corey LA. Univariate genetic analyses of epilepsy and seizures in a population-based twin study: the Virginia Twin Registry. Genet Epidemiol. 1998;15(1):33–49.PubMedCrossRef
5.
Zurück zum Zitat Kjeldsen MJ, Kyvik KO, Christensen K, Friis ML. Genetic and environmental factors in epilepsy: a population-based study of 11900 Danish twin pairs. Epilepsy Res. 2001;44(2–3):167–78.PubMedCrossRef Kjeldsen MJ, Kyvik KO, Christensen K, Friis ML. Genetic and environmental factors in epilepsy: a population-based study of 11900 Danish twin pairs. Epilepsy Res. 2001;44(2–3):167–78.PubMedCrossRef
6.
Zurück zum Zitat Speed D, O'Brien TJ, Palotie A, Shkura K, Marson AG, Balding DJ, et al. Describing the genetic architecture of epilepsy through heritability analysis. Brain. 2014;137:2680–9.PubMedPubMedCentralCrossRef Speed D, O'Brien TJ, Palotie A, Shkura K, Marson AG, Balding DJ, et al. Describing the genetic architecture of epilepsy through heritability analysis. Brain. 2014;137:2680–9.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Brandler WM, Sebat J. From de novo mutations to personalized therapeutic interventions in autism. Annu Rev Med. 2015;66:487–507.PubMedCrossRef Brandler WM, Sebat J. From de novo mutations to personalized therapeutic interventions in autism. Annu Rev Med. 2015;66:487–507.PubMedCrossRef
8.
Zurück zum Zitat Hoischen A, Krumm N, Eichler EE. Prioritization of neurodevelopmental disease genes by discovery of new mutations. Nat Neurosci. 2014;17(6):764–72.PubMedPubMedCentralCrossRef Hoischen A, Krumm N, Eichler EE. Prioritization of neurodevelopmental disease genes by discovery of new mutations. Nat Neurosci. 2014;17(6):764–72.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Directors ABo. ACMG policy statement: updated recommendations regarding analysis and reporting of secondary findings in clinical genome-scale sequencing. Genet Med. 2015;17(1):68–9.CrossRef Directors ABo. ACMG policy statement: updated recommendations regarding analysis and reporting of secondary findings in clinical genome-scale sequencing. Genet Med. 2015;17(1):68–9.CrossRef
10.
Zurück zum Zitat Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.PubMedPubMedCentralCrossRef Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Green RC, Berg JS, Grody WW, Kalia SS, Korf BR, Martin CL, et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet Med. 2013;15(7):565–74.PubMedPubMedCentralCrossRef Green RC, Berg JS, Grody WW, Kalia SS, Korf BR, Martin CL, et al. ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet Med. 2013;15(7):565–74.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–82.CrossRefPubMed Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–82.CrossRefPubMed
13.
Zurück zum Zitat Fisher RS, Cross JH, D'Souza C, French JA, Haut SR, Higurashi N, et al. Instruction manual for the ILAE 2017 operational classification of seizure types. Epilepsia. 2017;58(4):531–42.PubMedCrossRef Fisher RS, Cross JH, D'Souza C, French JA, Haut SR, Higurashi N, et al. Instruction manual for the ILAE 2017 operational classification of seizure types. Epilepsia. 2017;58(4):531–42.PubMedCrossRef
16.
Zurück zum Zitat Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11.10.1–33. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11.10.1–33.
17.
Zurück zum Zitat Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164.PubMedPubMedCentralCrossRef Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38(16):e164.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Schwarz JM, Cooper DN, Schuelke M, Seelow D. MutationTaster2: mutation prediction for the deep-sequencing age. Nat Methods. 2014;11(4):361–2.CrossRefPubMed Schwarz JM, Cooper DN, Schuelke M, Seelow D. MutationTaster2: mutation prediction for the deep-sequencing age. Nat Methods. 2014;11(4):361–2.CrossRefPubMed
19.
Zurück zum Zitat Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4(7):1073–81.PubMedCrossRef Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4(7):1073–81.PubMedCrossRef
20.
Zurück zum Zitat Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248–9.PubMedPubMedCentralCrossRef Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, et al. A method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248–9.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Cooper GM, Stone EA, Asimenos G, Program NCS, Green ED, Batzoglou S, et al. Distribution and intensity of constraint in mammalian genomic sequence. Genome Res. 2005;15(7):901–13.PubMedPubMedCentralCrossRef Cooper GM, Stone EA, Asimenos G, Program NCS, Green ED, Batzoglou S, et al. Distribution and intensity of constraint in mammalian genomic sequence. Genome Res. 2005;15(7):901–13.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Davydov EV, Goode DL, Sirota M, Cooper GM, Sidow A, Batzoglou S. Identifying a high fraction of the human genome to be under selective constraint using GERP++. PLoS Comput Biol. 2010;6(12):e1001025.PubMedPubMedCentralCrossRef Davydov EV, Goode DL, Sirota M, Cooper GM, Sidow A, Batzoglou S. Identifying a high fraction of the human genome to be under selective constraint using GERP++. PLoS Comput Biol. 2010;6(12):e1001025.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Desmet FO, Hamroun D, Lalande M, Collod-Beroud G, Claustres M, Beroud C. Human splicing finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 2009;37(9):e67.PubMedPubMedCentralCrossRef Desmet FO, Hamroun D, Lalande M, Collod-Beroud G, Claustres M, Beroud C. Human splicing finder: an online bioinformatics tool to predict splicing signals. Nucleic Acids Res. 2009;37(9):e67.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Uchino T, Haraguchi Y, Aparicio JM, Mizutani N, Higashikawa M, Naitoh H, et al. Three novel mutations in the liver-type arginase gene in three unrelated Japanese patients with argininemia. Am J Hum Genet. 1992;51(6):1406–12.PubMedPubMedCentral Uchino T, Haraguchi Y, Aparicio JM, Mizutani N, Higashikawa M, Naitoh H, et al. Three novel mutations in the liver-type arginase gene in three unrelated Japanese patients with argininemia. Am J Hum Genet. 1992;51(6):1406–12.PubMedPubMedCentral
25.
Zurück zum Zitat Carvalho DR, Brand GD, Brum JM, Takata RI, Speck-Martins CE, Pratesi R. Analysis of novel ARG1 mutations causing hyperargininemia and correlation with arginase I activity in erythrocytes. Gene. 2012;509(1):124–30.PubMedCrossRef Carvalho DR, Brand GD, Brum JM, Takata RI, Speck-Martins CE, Pratesi R. Analysis of novel ARG1 mutations causing hyperargininemia and correlation with arginase I activity in erythrocytes. Gene. 2012;509(1):124–30.PubMedCrossRef
26.
Zurück zum Zitat Uchino T, Snyderman SE, Lambert M, Qureshi IA, Shapira SK, Sansaricq C, et al. Molecular basis of phenotypic variation in patients with argininemia. Hum Genet. 1995;96(3):255–60.PubMedCrossRef Uchino T, Snyderman SE, Lambert M, Qureshi IA, Shapira SK, Sansaricq C, et al. Molecular basis of phenotypic variation in patients with argininemia. Hum Genet. 1995;96(3):255–60.PubMedCrossRef
27.
Zurück zum Zitat Wu TF, Liu YP, Li XY, Wang Q, Ding Y, Ma YY, et al. Five novel mutations in ARG1 gene in Chinese patients of argininemia. Pediatr Neurol. 2013;49(2):119–23.PubMedCrossRef Wu TF, Liu YP, Li XY, Wang Q, Ding Y, Ma YY, et al. Five novel mutations in ARG1 gene in Chinese patients of argininemia. Pediatr Neurol. 2013;49(2):119–23.PubMedCrossRef
28.
Zurück zum Zitat Veeramah KR, Johnstone L, Karafet TM, Wolf D, Sprissler R, Salogiannis J, et al. Exome sequencing reveals new causal mutations in children with epileptic encephalopathies. Epilepsia. 2013;54(7):1270–81.PubMedPubMedCentralCrossRef Veeramah KR, Johnstone L, Karafet TM, Wolf D, Sprissler R, Salogiannis J, et al. Exome sequencing reveals new causal mutations in children with epileptic encephalopathies. Epilepsia. 2013;54(7):1270–81.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Kapoor A, Satishchandra P, Ratnapriya R, Reddy R, Kadandale J, Shankar SK, et al. An idiopathic epilepsy syndrome linked to 3q13.3-q21 and missense mutations in the extracellular calcium sensing receptor gene. Ann Neurol. 2008;64(2):158–67.PubMedCrossRef Kapoor A, Satishchandra P, Ratnapriya R, Reddy R, Kadandale J, Shankar SK, et al. An idiopathic epilepsy syndrome linked to 3q13.3-q21 and missense mutations in the extracellular calcium sensing receptor gene. Ann Neurol. 2008;64(2):158–67.PubMedCrossRef
30.
Zurück zum Zitat Sato K, Hasegawa Y, Nakae J, Nanao K, Takahashi I, Tajima T, et al. Hydrochlorothiazide effectively reduces urinary calcium excretion in two Japanese patients with gain-of-function mutations of the calcium-sensing receptor gene. J Clin Endocrinol Metab. 2002;87(7):3068–73.PubMedCrossRef Sato K, Hasegawa Y, Nakae J, Nanao K, Takahashi I, Tajima T, et al. Hydrochlorothiazide effectively reduces urinary calcium excretion in two Japanese patients with gain-of-function mutations of the calcium-sensing receptor gene. J Clin Endocrinol Metab. 2002;87(7):3068–73.PubMedCrossRef
31.
Zurück zum Zitat Kurahashi H, Hirose S. Autosomal Dominant Nocturnal Frontal Lobe Epilepsy. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, et al., editors. GeneReviews((R)). Seattle (WA): University of Washington, Seattle; 1993–2018. Kurahashi H, Hirose S. Autosomal Dominant Nocturnal Frontal Lobe Epilepsy. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, et al., editors. GeneReviews((R)). Seattle (WA): University of Washington, Seattle; 1993–2018.
32.
Zurück zum Zitat Lauterborn JC, Ribak CE. Differences in dopamine beta-hydroxylase immunoreactivity between the brains of genetically epilepsy-prone and Sprague-Dawley rats. Epilepsy Res. 1989;4(3):161–76.PubMedCrossRef Lauterborn JC, Ribak CE. Differences in dopamine beta-hydroxylase immunoreactivity between the brains of genetically epilepsy-prone and Sprague-Dawley rats. Epilepsy Res. 1989;4(3):161–76.PubMedCrossRef
33.
Zurück zum Zitat Schank JR, Liles LC, Weinshenker D. Reduced anticonvulsant efficacy of valproic acid in dopamine beta-hydroxylase knockout mice. Epilepsy Res. 2005;65(1–2):23–31.PubMedCrossRef Schank JR, Liles LC, Weinshenker D. Reduced anticonvulsant efficacy of valproic acid in dopamine beta-hydroxylase knockout mice. Epilepsy Res. 2005;65(1–2):23–31.PubMedCrossRef
34.
Zurück zum Zitat Warter JM, Coquillat G, Kurtz D. Human circulating dopamine-beta-hydroxylase and epilepsy. Psychopharmacologia. 1975;41(1):75–9.PubMedCrossRef Warter JM, Coquillat G, Kurtz D. Human circulating dopamine-beta-hydroxylase and epilepsy. Psychopharmacologia. 1975;41(1):75–9.PubMedCrossRef
35.
Zurück zum Zitat Lesca G, Rudolf G, Labalme A, Hirsch E, Arzimanoglou A, Genton P, et al. Epileptic encephalopathies of the Landau-Kleffner and continuous spike and waves during slow-wave sleep types: genomic dissection makes the link with autism. Epilepsia. 2012;53(9):1526–38.PubMedCrossRef Lesca G, Rudolf G, Labalme A, Hirsch E, Arzimanoglou A, Genton P, et al. Epileptic encephalopathies of the Landau-Kleffner and continuous spike and waves during slow-wave sleep types: genomic dissection makes the link with autism. Epilepsia. 2012;53(9):1526–38.PubMedCrossRef
36.
Zurück zum Zitat Steele SU, Cheah SM, Veerapandiyan A, Gallentine W, Smith EC, Mikati MA. Electroencephalographic and seizure manifestations in two patients with folate receptor autoimmune antibody-mediated primary cerebral folate deficiency. Epilepsy Behav. 2012;24(4):507–12.PubMedCrossRef Steele SU, Cheah SM, Veerapandiyan A, Gallentine W, Smith EC, Mikati MA. Electroencephalographic and seizure manifestations in two patients with folate receptor autoimmune antibody-mediated primary cerebral folate deficiency. Epilepsy Behav. 2012;24(4):507–12.PubMedCrossRef
37.
Zurück zum Zitat Perez-Duenas B, Toma C, Ormazabal A, Muchart J, Sanmarti F, Bombau G, et al. Progressive ataxia and myoclonic epilepsy in a patient with a homozygous mutation in the FOLR1 gene. J Inherit Metab Dis. 2010;33(6):795–802.PubMedCrossRef Perez-Duenas B, Toma C, Ormazabal A, Muchart J, Sanmarti F, Bombau G, et al. Progressive ataxia and myoclonic epilepsy in a patient with a homozygous mutation in the FOLR1 gene. J Inherit Metab Dis. 2010;33(6):795–802.PubMedCrossRef
38.
Zurück zum Zitat Kumari R, Lakhan R, Kalita J, Garg RK, Misra UK, Mittal B. Potential role of GABAA receptor subunit; GABRA6, GABRB2 and GABRR2 gene polymorphisms in epilepsy susceptibility and pharmacotherapy in North Indian population. Clin Chim Acta. 2011;412(13–14):1244–8.PubMedCrossRef Kumari R, Lakhan R, Kalita J, Garg RK, Misra UK, Mittal B. Potential role of GABAA receptor subunit; GABRA6, GABRB2 and GABRR2 gene polymorphisms in epilepsy susceptibility and pharmacotherapy in North Indian population. Clin Chim Acta. 2011;412(13–14):1244–8.PubMedCrossRef
39.
Zurück zum Zitat Hernandez CC, Gurba KN, Hu N, Macdonald RL. The GABRA6 mutation, R46W, associated with childhood absence epilepsy, alters 6beta22 and 6beta2 GABA(A) receptor channel gating and expression. J Physiol. 2011;589(Pt 23):5857–78.PubMedPubMedCentralCrossRef Hernandez CC, Gurba KN, Hu N, Macdonald RL. The GABRA6 mutation, R46W, associated with childhood absence epilepsy, alters 6beta22 and 6beta2 GABA(A) receptor channel gating and expression. J Physiol. 2011;589(Pt 23):5857–78.PubMedPubMedCentralCrossRef
40.
41.
Zurück zum Zitat Bakker MJ, van Dijk JG, van den Maagdenberg AMJM, Tijssen MAJ. Startle syndromes. Lancet Neurol. 2006;5(6):513–24.PubMedCrossRef Bakker MJ, van Dijk JG, van den Maagdenberg AMJM, Tijssen MAJ. Startle syndromes. Lancet Neurol. 2006;5(6):513–24.PubMedCrossRef
42.
Zurück zum Zitat Conroy J, McGettigan PA, McCreary D, Shah N, Collins K, Parry-Fielder B, et al. Towards the identification of a genetic basis for Landau-Kleffner syndrome. Epilepsia. 2014;55(6):858–65.PubMedCrossRef Conroy J, McGettigan PA, McCreary D, Shah N, Collins K, Parry-Fielder B, et al. Towards the identification of a genetic basis for Landau-Kleffner syndrome. Epilepsia. 2014;55(6):858–65.PubMedCrossRef
43.
Zurück zum Zitat Dong L, Chen Y, Lewis M, Hsieh JC, Reing J, Chaillet JR, et al. Neurologic defects and selective disruption of basement membranes in mice lacking entactin-1/nidogen-1. Lab Investig. 2002;82(12):1617–30.PubMedCrossRef Dong L, Chen Y, Lewis M, Hsieh JC, Reing J, Chaillet JR, et al. Neurologic defects and selective disruption of basement membranes in mice lacking entactin-1/nidogen-1. Lab Investig. 2002;82(12):1617–30.PubMedCrossRef
44.
Zurück zum Zitat Fong CY, Mumford AD, Likeman MJ, Jardine PE. Cerebral palsy in siblings caused by compound heterozygous mutations in the gene encoding protein C. Dev Med Child Neurol. 2010;52(5):489–93.PubMedCrossRef Fong CY, Mumford AD, Likeman MJ, Jardine PE. Cerebral palsy in siblings caused by compound heterozygous mutations in the gene encoding protein C. Dev Med Child Neurol. 2010;52(5):489–93.PubMedCrossRef
45.
Zurück zum Zitat Thevenon J, Milh M, Feillet F, St-Onge J, Duffourd Y, Juge C, et al. Mutations in SLC13A5 cause autosomal-recessive epileptic encephalopathy with seizure onset in the first days of life. Am J Hum Genet. 2014;95(1):113–20.PubMedPubMedCentralCrossRef Thevenon J, Milh M, Feillet F, St-Onge J, Duffourd Y, Juge C, et al. Mutations in SLC13A5 cause autosomal-recessive epileptic encephalopathy with seizure onset in the first days of life. Am J Hum Genet. 2014;95(1):113–20.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Hardies K, de Kovel CG, Weckhuysen S, Asselbergh B, Geuens T, Deconinck T, et al. Recessive mutations in SLC13A5 result in a loss of citrate transport and cause neonatal epilepsy, developmental delay and teeth hypoplasia. Brain. 2015;138(Pt 11):3238–50.PubMedCrossRef Hardies K, de Kovel CG, Weckhuysen S, Asselbergh B, Geuens T, Deconinck T, et al. Recessive mutations in SLC13A5 result in a loss of citrate transport and cause neonatal epilepsy, developmental delay and teeth hypoplasia. Brain. 2015;138(Pt 11):3238–50.PubMedCrossRef
47.
Zurück zum Zitat Zeng WQ, Al-Yamani E, Acierno JS Jr, Slaugenhaupt S, Gillis T, MacDonald ME, et al. Biotin-responsive basal ganglia disease maps to 2q36.3 and is due to mutations in SLC19A3. Am J Hum Genet. 2005;77(1):16–26.PubMedPubMedCentralCrossRef Zeng WQ, Al-Yamani E, Acierno JS Jr, Slaugenhaupt S, Gillis T, MacDonald ME, et al. Biotin-responsive basal ganglia disease maps to 2q36.3 and is due to mutations in SLC19A3. Am J Hum Genet. 2005;77(1):16–26.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Roll P, Rudolf G, Pereira S, Royer B, Scheffer IE, Massacrier A, et al. SRPX2 mutations in disorders of language cortex and cognition. Hum Mol Genet. 2006;15(7):1195–207.PubMedCrossRef Roll P, Rudolf G, Pereira S, Royer B, Scheffer IE, Massacrier A, et al. SRPX2 mutations in disorders of language cortex and cognition. Hum Mol Genet. 2006;15(7):1195–207.PubMedCrossRef
49.
Zurück zum Zitat Hildebrand MS, Myers CT, Carvill GL, Regan BM, Damiano JA, Mullen SA, et al. A targeted resequencing gene panel for focal epilepsy. Nerology. 2016;86(17):1605–12.CrossRef Hildebrand MS, Myers CT, Carvill GL, Regan BM, Damiano JA, Mullen SA, et al. A targeted resequencing gene panel for focal epilepsy. Nerology. 2016;86(17):1605–12.CrossRef
50.
Zurück zum Zitat Parrini E, Marini C, Mei D, Galuppi A, Cellini E, Pucatti D, et al. Diagnostic targeted resequencing in 349 patients with drug-resistant pediatric epilepsies identifies causative mutations in 30 different genes. Hum Mutat. 2017;38(2):216–25.PubMedCrossRef Parrini E, Marini C, Mei D, Galuppi A, Cellini E, Pucatti D, et al. Diagnostic targeted resequencing in 349 patients with drug-resistant pediatric epilepsies identifies causative mutations in 30 different genes. Hum Mutat. 2017;38(2):216–25.PubMedCrossRef
51.
Zurück zum Zitat Carvill GL, Heavin SB, Yendle SC, McMahon JM, O'Roak BJ, Cook J, et al. Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1. Nat Genet. 2013;45(7):825–30.PubMedPubMedCentralCrossRef Carvill GL, Heavin SB, Yendle SC, McMahon JM, O'Roak BJ, Cook J, et al. Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1. Nat Genet. 2013;45(7):825–30.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Trump N, McTague A, Brittain H, Papandreou A, Meyer E, Ngoh A, et al. Improving diagnosis and broadening the phenotypes in early-onset seizure and severe developmental delay disorders through gene panel analysis. J Med Genet. 2016;53(5):310–7.PubMedCrossRef Trump N, McTague A, Brittain H, Papandreou A, Meyer E, Ngoh A, et al. Improving diagnosis and broadening the phenotypes in early-onset seizure and severe developmental delay disorders through gene panel analysis. J Med Genet. 2016;53(5):310–7.PubMedCrossRef
53.
Zurück zum Zitat Lemke JR, Riesch E, Scheurenbrand T, Schubach M, Wilhelm C, Steiner I, et al. Targeted next generation sequencing as a diagnostic tool in epileptic disorders. Epilepsia. 2012;53(8):1387–98.PubMedCrossRef Lemke JR, Riesch E, Scheurenbrand T, Schubach M, Wilhelm C, Steiner I, et al. Targeted next generation sequencing as a diagnostic tool in epileptic disorders. Epilepsia. 2012;53(8):1387–98.PubMedCrossRef
54.
Zurück zum Zitat Møller RS, Larsen LH, Johannesen KM, Talvik I, Talvik T, Vaher U, et al. Gene panel testing in epileptic encephalopathies and familial epilepsies. Mol Syndromol. 2016;7(4):210–9.PubMedPubMedCentralCrossRef Møller RS, Larsen LH, Johannesen KM, Talvik I, Talvik T, Vaher U, et al. Gene panel testing in epileptic encephalopathies and familial epilepsies. Mol Syndromol. 2016;7(4):210–9.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Kodera H, Kato M, Nord AS, Walsh T, Lee M, Yamanaka G, et al. Targeted capture and sequencing for detection of mutations causing early onset epileptic encephalopathy. Epilepsia. 2013;54(7):1262–9.PubMedCrossRef Kodera H, Kato M, Nord AS, Walsh T, Lee M, Yamanaka G, et al. Targeted capture and sequencing for detection of mutations causing early onset epileptic encephalopathy. Epilepsia. 2013;54(7):1262–9.PubMedCrossRef
58.
Zurück zum Zitat Mao X, Li K, Tang B, Luo Y, Ding D, Zhao Y, et al. Novel mutations in ADSL for Adenylosuccinate Lyase deficiency identified by the combination of trio-WES and constantly updated guidelines. Sci Rep. 2017;7(1):1625.PubMedPubMedCentralCrossRef Mao X, Li K, Tang B, Luo Y, Ding D, Zhao Y, et al. Novel mutations in ADSL for Adenylosuccinate Lyase deficiency identified by the combination of trio-WES and constantly updated guidelines. Sci Rep. 2017;7(1):1625.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Jurecka A, Zikanova M, Kmoch S, Tylki-Szymańska A. Adenylosuccinate lyase deficiency. J Inherit Metab Dis. 2015;38(2):231–42.PubMedCrossRef Jurecka A, Zikanova M, Kmoch S, Tylki-Szymańska A. Adenylosuccinate lyase deficiency. J Inherit Metab Dis. 2015;38(2):231–42.PubMedCrossRef
60.
Zurück zum Zitat Chen BC, McGown IN, Thong MK, Pitt J, Yunus ZM, Khoo TB, et al. Adenylosuccinate lyase deficiency in a Malaysian patient, with novel adenylosuccinate lyase gene mutations. J Inherit Metab Dis. 2010;33(Suppl 3):S159–62.PubMedCrossRef Chen BC, McGown IN, Thong MK, Pitt J, Yunus ZM, Khoo TB, et al. Adenylosuccinate lyase deficiency in a Malaysian patient, with novel adenylosuccinate lyase gene mutations. J Inherit Metab Dis. 2010;33(Suppl 3):S159–62.PubMedCrossRef
61.
Zurück zum Zitat Hamdan FF, Gauthier J, Spiegelman D, Noreau A, Yang Y, Pellerin S, et al. Mutations in SYNGAP1 in autosomal nonsyndromic mental retardation. N Engl J Med. 2009;360(6):599–605.PubMedPubMedCentralCrossRef Hamdan FF, Gauthier J, Spiegelman D, Noreau A, Yang Y, Pellerin S, et al. Mutations in SYNGAP1 in autosomal nonsyndromic mental retardation. N Engl J Med. 2009;360(6):599–605.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Berryer MH, Hamdan FF, Klitten LL, Møller RS, Carmant L, Schwartzentruber J, et al. Mutations in SYNGAP1 cause intellectual disability, autism, and a specific form of epilepsy by inducing haploinsufficiency. Hum Mutat. 2013;34(2):385–94.PubMedCrossRef Berryer MH, Hamdan FF, Klitten LL, Møller RS, Carmant L, Schwartzentruber J, et al. Mutations in SYNGAP1 cause intellectual disability, autism, and a specific form of epilepsy by inducing haploinsufficiency. Hum Mutat. 2013;34(2):385–94.PubMedCrossRef
63.
Zurück zum Zitat Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23:185–8.CrossRefPubMed Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23:185–8.CrossRefPubMed
64.
65.
Zurück zum Zitat Zhang H, Nanba E, Yamamoto T, Ninomiya H, Ohno K, Mizuguchi M, et al. Mutational analysis of TSC1 and TSC2 genes in Japanese patients with tuberous sclerosis complex. J Hum Genet. 1999;44(6):391–6.PubMedCrossRef Zhang H, Nanba E, Yamamoto T, Ninomiya H, Ohno K, Mizuguchi M, et al. Mutational analysis of TSC1 and TSC2 genes in Japanese patients with tuberous sclerosis complex. J Hum Genet. 1999;44(6):391–6.PubMedCrossRef
66.
Zurück zum Zitat Wilson PJ, Ramesh V, Kristiansen A, Bove C, Jozwiak S, Kwiatkowski DJ, et al. Novel mutations detected in the TSC2 gene from both sporadic and familial TSC patients. Hum Mol Genet. 1996;5(2):249–56.PubMedCrossRef Wilson PJ, Ramesh V, Kristiansen A, Bove C, Jozwiak S, Kwiatkowski DJ, et al. Novel mutations detected in the TSC2 gene from both sporadic and familial TSC patients. Hum Mol Genet. 1996;5(2):249–56.PubMedCrossRef
67.
Zurück zum Zitat Hoogeveen-Westerveld M, Wentink M, van den Heuvel D, Mozaffari M, Ekong R, Povey S, et al. Functional assessment of variants in the TSC1 and TSC2 genes identified in individuals with tuberous sclerosis complex. Hum Mutat. 2011;32(4):424–35.PubMedCrossRef Hoogeveen-Westerveld M, Wentink M, van den Heuvel D, Mozaffari M, Ekong R, Povey S, et al. Functional assessment of variants in the TSC1 and TSC2 genes identified in individuals with tuberous sclerosis complex. Hum Mutat. 2011;32(4):424–35.PubMedCrossRef
68.
Zurück zum Zitat Gilboa T, Segel R, Zeligson S, Alterescu G, Ben-Pazi H. Ganglioglioma, epilepsy, and intellectual impairment due to familial TSC1 deletion. J Child Neurol. 2018;33(7):482–6.PubMedCrossRef Gilboa T, Segel R, Zeligson S, Alterescu G, Ben-Pazi H. Ganglioglioma, epilepsy, and intellectual impairment due to familial TSC1 deletion. J Child Neurol. 2018;33(7):482–6.PubMedCrossRef
70.
Zurück zum Zitat Zerem A, Haginoya K, Lev D, Blumkin L, Kivity S, Linder I, et al. The molecular and phenotypic spectrum of IQSEC2-related epilepsy. Epilepsia. 2016;57(11):1858–69.PubMedCrossRef Zerem A, Haginoya K, Lev D, Blumkin L, Kivity S, Linder I, et al. The molecular and phenotypic spectrum of IQSEC2-related epilepsy. Epilepsia. 2016;57(11):1858–69.PubMedCrossRef
71.
Zurück zum Zitat Wilmshurst JM, Berg AT, Lagae L, Newton CR, Cross JH. The challenges and innovations for therapy in children with epilepsy. Nat Rev Neurol. 2014;10(5):249–60.PubMedPubMedCentralCrossRef Wilmshurst JM, Berg AT, Lagae L, Newton CR, Cross JH. The challenges and innovations for therapy in children with epilepsy. Nat Rev Neurol. 2014;10(5):249–60.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Oyrer J, Maljevic S, Scheffer IE, Berkovic SF, Petrou S, Reid CA. Ion channels in genetic epilepsy: from genes and mechanisms to disease-targeted therapies. Pharmacol Rev. 2018;70(1):142–73.PubMedPubMedCentralCrossRef Oyrer J, Maljevic S, Scheffer IE, Berkovic SF, Petrou S, Reid CA. Ion channels in genetic epilepsy: from genes and mechanisms to disease-targeted therapies. Pharmacol Rev. 2018;70(1):142–73.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Orsini A, Zara F, Striano P. Recent advances in epilepsy genetics. Neurosci Lett. 2018;667:4–9.PubMedCrossRef Orsini A, Zara F, Striano P. Recent advances in epilepsy genetics. Neurosci Lett. 2018;667:4–9.PubMedCrossRef
74.
Zurück zum Zitat Weber YG, Biskup S, Helbig KL, Von Spiczak S, Lerche H. The role of genetic testing in epilepsy diagnosis and management. Expert Rev Mol Diagn. 2017;17(8):739–50.PubMedCrossRef Weber YG, Biskup S, Helbig KL, Von Spiczak S, Lerche H. The role of genetic testing in epilepsy diagnosis and management. Expert Rev Mol Diagn. 2017;17(8):739–50.PubMedCrossRef
75.
Zurück zum Zitat Wang J, Lin ZJ, Liu L, Xu HQ, Shi YW, Yi YH, et al. Epilepsy-associated genes. Seizure. 2017;44:11–20.PubMedCrossRef Wang J, Lin ZJ, Liu L, Xu HQ, Shi YW, Yi YH, et al. Epilepsy-associated genes. Seizure. 2017;44:11–20.PubMedCrossRef
76.
Zurück zum Zitat Samarasinghe TD, Sands SA, Skuza EM, Joshi MS, Nold-Petry CA, Berger PJ. The effect of prenatal maternal infection on respiratory function in mouse offspring: evidence for enhanced chemosensitivity. J Appl Physiol (1985). 2015;119(3):299–307.CrossRef Samarasinghe TD, Sands SA, Skuza EM, Joshi MS, Nold-Petry CA, Berger PJ. The effect of prenatal maternal infection on respiratory function in mouse offspring: evidence for enhanced chemosensitivity. J Appl Physiol (1985). 2015;119(3):299–307.CrossRef
77.
Zurück zum Zitat Catterall WA, Goldin AL, Waxman SG. International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels. Pharmacol Rev. 2005;57(4):397–409.PubMedCrossRef Catterall WA, Goldin AL, Waxman SG. International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels. Pharmacol Rev. 2005;57(4):397–409.PubMedCrossRef
78.
Zurück zum Zitat Oliva M, Berkovic SF, Petrou S. Sodium channels and the neurobiology of epilepsy. Epilepsia. 2012;53(11):1849–59.PubMedCrossRef Oliva M, Berkovic SF, Petrou S. Sodium channels and the neurobiology of epilepsy. Epilepsia. 2012;53(11):1849–59.PubMedCrossRef
79.
Zurück zum Zitat McDonald CL, Saneto RP, Carmant L, Sotero de Menezes MA. Focal seizures in patients with SCN1A mutations. J Child Neurol. 2017;32(2):170–6.PubMedCrossRef McDonald CL, Saneto RP, Carmant L, Sotero de Menezes MA. Focal seizures in patients with SCN1A mutations. J Child Neurol. 2017;32(2):170–6.PubMedCrossRef
80.
Zurück zum Zitat Depienne C, Trouillard O, Gourfinkel-An I, Saint-Martin C, Bouteiller D, Graber D, et al. Mechanisms for variable expressivity of inherited SCN1A mutations causing Dravet syndrome. J Med Genet. 2010;47(6):404–10.PubMedCrossRef Depienne C, Trouillard O, Gourfinkel-An I, Saint-Martin C, Bouteiller D, Graber D, et al. Mechanisms for variable expressivity of inherited SCN1A mutations causing Dravet syndrome. J Med Genet. 2010;47(6):404–10.PubMedCrossRef
81.
Zurück zum Zitat Poduri A, Evrony GD, Cai X, Elhosary PC, Beroukhim R, Lehtinen MK, et al. Somatic activation of AKT3 causes hemispheric developmental brain malformations. Neuron. 2012;74(1):41–8.PubMedPubMedCentralCrossRef Poduri A, Evrony GD, Cai X, Elhosary PC, Beroukhim R, Lehtinen MK, et al. Somatic activation of AKT3 causes hemispheric developmental brain malformations. Neuron. 2012;74(1):41–8.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Hawkins NA, Anderson LL, Gertler TS, Laux L, George AL Jr, Kearney JA. Screening of conventional anticonvulsants in a genetic mouse model of epilepsy. Ann Clin Transl Neurol. 2017;4(5):326–39.PubMedPubMedCentralCrossRef Hawkins NA, Anderson LL, Gertler TS, Laux L, George AL Jr, Kearney JA. Screening of conventional anticonvulsants in a genetic mouse model of epilepsy. Ann Clin Transl Neurol. 2017;4(5):326–39.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Chiron C, Dulac O. The pharmacologic treatment of Dravet syndrome. Epilepsia. 2011;52(Suppl 2):72–5.PubMedCrossRef Chiron C, Dulac O. The pharmacologic treatment of Dravet syndrome. Epilepsia. 2011;52(Suppl 2):72–5.PubMedCrossRef
84.
Zurück zum Zitat Cao D, Ohtani H, Ogiwara I, Ohtani S, Takahashi Y, Yamakawa K, et al. Efficacy of stiripentol in hyperthermia-induced seizures in a mouse model of Dravet syndrome. Epilepsia. 2012;53(7):1140–5.PubMedCrossRef Cao D, Ohtani H, Ogiwara I, Ohtani S, Takahashi Y, Yamakawa K, et al. Efficacy of stiripentol in hyperthermia-induced seizures in a mouse model of Dravet syndrome. Epilepsia. 2012;53(7):1140–5.PubMedCrossRef
85.
Zurück zum Zitat Oakley JC, Cho AR, Cheah CS, Scheuer T, Catterall WA. Synergistic GABA-enhancing therapy against seizures in a mouse model of Dravet syndrome. J Pharmacol Exp Ther. 2013;345(2):215–24.PubMedPubMedCentralCrossRef Oakley JC, Cho AR, Cheah CS, Scheuer T, Catterall WA. Synergistic GABA-enhancing therapy against seizures in a mouse model of Dravet syndrome. J Pharmacol Exp Ther. 2013;345(2):215–24.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Di Meglio C, Lesca G, Villeneuve N, Lacoste C, Abidi A, Cacciagli P, et al. Epileptic patients with de novo STXBP1 mutations: key clinical features based on 24 cases. Epilepsia. 2015;56(12):1931–40.PubMedCrossRef Di Meglio C, Lesca G, Villeneuve N, Lacoste C, Abidi A, Cacciagli P, et al. Epileptic patients with de novo STXBP1 mutations: key clinical features based on 24 cases. Epilepsia. 2015;56(12):1931–40.PubMedCrossRef
87.
Zurück zum Zitat Ko A, Youn SE, Kim SH, Lee JS, Kim S, Choi JR, et al. Targeted gene panel and genotype-phenotype correlation in children with developmental and epileptic encephalopathy. Epilepsy Res. 2018;141:48–55.PubMedCrossRef Ko A, Youn SE, Kim SH, Lee JS, Kim S, Choi JR, et al. Targeted gene panel and genotype-phenotype correlation in children with developmental and epileptic encephalopathy. Epilepsy Res. 2018;141:48–55.PubMedCrossRef
88.
Zurück zum Zitat Ohba C, Kato M, Takahashi N, Osaka H, Shiihara T, Tohyama J, et al. De novo KCNT1 mutations in early-onset epileptic encephalopathy. Epilepsia. 2015;56(9):e121–8.CrossRefPubMed Ohba C, Kato M, Takahashi N, Osaka H, Shiihara T, Tohyama J, et al. De novo KCNT1 mutations in early-onset epileptic encephalopathy. Epilepsia. 2015;56(9):e121–8.CrossRefPubMed
89.
Zurück zum Zitat Fukuoka M, Kuki I, Kawawaki H, Okazaki S, Kim K, Hattori Y, et al. Quinidine therapy for West syndrome with KCNTI mutation: a case report. Brain Dev. 2017;39(1):80–3.CrossRefPubMed Fukuoka M, Kuki I, Kawawaki H, Okazaki S, Kim K, Hattori Y, et al. Quinidine therapy for West syndrome with KCNTI mutation: a case report. Brain Dev. 2017;39(1):80–3.CrossRefPubMed
90.
Zurück zum Zitat Leonard H, Cobb S, Downs J. Clinical and biological progress over 50 years in Rett syndrome. Nat Rev Neurol. 2017;13(1):37–51.PubMedCrossRef Leonard H, Cobb S, Downs J. Clinical and biological progress over 50 years in Rett syndrome. Nat Rev Neurol. 2017;13(1):37–51.PubMedCrossRef
92.
Zurück zum Zitat Daci A, Bozalija A, Jashari F, Krasniqi S. Individualizing treatment approaches for epileptic patients with glucose transporter type1 (GLUT-1) deficiency. Int J Mol Sci. 2018;19(1):122.CrossRefPubMedCentral Daci A, Bozalija A, Jashari F, Krasniqi S. Individualizing treatment approaches for epileptic patients with glucose transporter type1 (GLUT-1) deficiency. Int J Mol Sci. 2018;19(1):122.CrossRefPubMedCentral
93.
Zurück zum Zitat Ku CS, Polychronakos C, Tan EK, Naidoo N, Pawitan Y, Roukos DH, et al. A new paradigm emerges from the study of de novo mutations in the context of neurodevelopmental disease. Mol Psychiatry. 2013;18(2):141–53.PubMedCrossRef Ku CS, Polychronakos C, Tan EK, Naidoo N, Pawitan Y, Roukos DH, et al. A new paradigm emerges from the study of de novo mutations in the context of neurodevelopmental disease. Mol Psychiatry. 2013;18(2):141–53.PubMedCrossRef
94.
Zurück zum Zitat Veltman JA, Brunner HG. De novo mutations in human genetic disease. Nat Rev Genet. 2012;13(8):565–75.PubMedCrossRef Veltman JA, Brunner HG. De novo mutations in human genetic disease. Nat Rev Genet. 2012;13(8):565–75.PubMedCrossRef
95.
Zurück zum Zitat Hirose S, Scheffer IE, Marini C, De Jonghe P, Andermann E, Goldman AM, et al. SCN1A testing for epilepsy: application in clinical practice. Epilepsia. 2013;54(5):946–52.PubMedCrossRef Hirose S, Scheffer IE, Marini C, De Jonghe P, Andermann E, Goldman AM, et al. SCN1A testing for epilepsy: application in clinical practice. Epilepsia. 2013;54(5):946–52.PubMedCrossRef
96.
Zurück zum Zitat Kammoun F. Screening of MECP2 coding sequence in patients with phenotypes of decreasing likelihood for Rett syndrome: a cohort of 171 cases. J Med Genet. 2004;41(6):e85.PubMedPubMedCentralCrossRef Kammoun F. Screening of MECP2 coding sequence in patients with phenotypes of decreasing likelihood for Rett syndrome: a cohort of 171 cases. J Med Genet. 2004;41(6):e85.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Avgeris S, Fostira F, Vagena A, Ninios Y, Delimitsou A, Vodicka R, et al. Mutational analysis of TSC1 and TSC2 genes in tuberous sclerosis complex patients from Greece. Sci Rep. 2017;7(1):16697.PubMedPubMedCentralCrossRef Avgeris S, Fostira F, Vagena A, Ninios Y, Delimitsou A, Vodicka R, et al. Mutational analysis of TSC1 and TSC2 genes in tuberous sclerosis complex patients from Greece. Sci Rep. 2017;7(1):16697.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Steel D, Symonds JD, Zuberi SM, Brunklaus A. Dravet syndrome and its mimics: beyond SCN1A. Epilepsia. 2017;58(11):1807–16.PubMedCrossRef Steel D, Symonds JD, Zuberi SM, Brunklaus A. Dravet syndrome and its mimics: beyond SCN1A. Epilepsia. 2017;58(11):1807–16.PubMedCrossRef
99.
Zurück zum Zitat Guerrini R, Parrini E. Epilepsy in Rett syndrome, and CDKL5- and FOXG1-gene-related encephalopathies. Epilepsia. 2012;53(12):2067–78.PubMedCrossRef Guerrini R, Parrini E. Epilepsy in Rett syndrome, and CDKL5- and FOXG1-gene-related encephalopathies. Epilepsia. 2012;53(12):2067–78.PubMedCrossRef
100.
101.
Zurück zum Zitat Depienne C, Trouillard O, Saint-Martin C, Gourfinkel-An I, Bouteiller D, Carpentier W, et al. Spectrum of SCN1A gene mutations associated with Dravet syndrome: analysis of 333 patients. J Med Genet. 2009;46(3):183–91.PubMedCrossRef Depienne C, Trouillard O, Saint-Martin C, Gourfinkel-An I, Bouteiller D, Carpentier W, et al. Spectrum of SCN1A gene mutations associated with Dravet syndrome: analysis of 333 patients. J Med Genet. 2009;46(3):183–91.PubMedCrossRef
102.
Zurück zum Zitat Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini E, et al. Spectrum of SCN1A mutations in severe myoclonic epilepsy of infancy. Neurology. 2003;60(12):1961–7.PubMedCrossRef Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini E, et al. Spectrum of SCN1A mutations in severe myoclonic epilepsy of infancy. Neurology. 2003;60(12):1961–7.PubMedCrossRef
103.
Zurück zum Zitat Harkin LA, McMahon JM, Iona X, Dibbens L, Pelekanos JT, Zuberi SM, et al. The spectrum of SCN1A-related infantile epileptic encephalopathies. Brain. 2007;130(Pt 3):843–52.PubMedCrossRef Harkin LA, McMahon JM, Iona X, Dibbens L, Pelekanos JT, Zuberi SM, et al. The spectrum of SCN1A-related infantile epileptic encephalopathies. Brain. 2007;130(Pt 3):843–52.PubMedCrossRef
104.
Zurück zum Zitat Fukuma G, Oguni H, Shirasaka Y, Watanabe K, Miyajima T, Yasumoto S, et al. Mutations of neuronal voltage-gated Na+ channel alpha 1 subunit gene SCN1A in core severe myoclonic epilepsy in infancy (SMEI) and in borderline SMEI (SMEB). Epilepsia. 2004;45(2):140–8.PubMedCrossRef Fukuma G, Oguni H, Shirasaka Y, Watanabe K, Miyajima T, Yasumoto S, et al. Mutations of neuronal voltage-gated Na+ channel alpha 1 subunit gene SCN1A in core severe myoclonic epilepsy in infancy (SMEI) and in borderline SMEI (SMEB). Epilepsia. 2004;45(2):140–8.PubMedCrossRef
105.
Zurück zum Zitat Saitsu H, Kato M, Koide A, Goto T, Fujita T, Nishiyama K, et al. Whole exome sequencing identifies KCNQ2 mutations in Ohtahara syndrome. Ann Neurol. 2012;72(2):298–300.PubMedCrossRef Saitsu H, Kato M, Koide A, Goto T, Fujita T, Nishiyama K, et al. Whole exome sequencing identifies KCNQ2 mutations in Ohtahara syndrome. Ann Neurol. 2012;72(2):298–300.PubMedCrossRef
106.
Zurück zum Zitat Ishii A, Shioda M, Okumura A, Kidokoro H, Sakauchi M, Shimada S, et al. A recurrent KCNT1 mutation in two sporadic cases with malignant migrating partial seizures in infancy. Gene. 2013;531(2):467–71.PubMedCrossRef Ishii A, Shioda M, Okumura A, Kidokoro H, Sakauchi M, Shimada S, et al. A recurrent KCNT1 mutation in two sporadic cases with malignant migrating partial seizures in infancy. Gene. 2013;531(2):467–71.PubMedCrossRef
107.
Zurück zum Zitat Mayer K, Ballhausen W, Rott HD. Mutation screening of the entire coding regions of the TSC1 and the TSC2 gene with the protein truncation test (PTT) identifies frequent splicing defects. Hum Mutat. 1999;14:401–11.PubMedCrossRef Mayer K, Ballhausen W, Rott HD. Mutation screening of the entire coding regions of the TSC1 and the TSC2 gene with the protein truncation test (PTT) identifies frequent splicing defects. Hum Mutat. 1999;14:401–11.PubMedCrossRef
108.
Zurück zum Zitat Gardella E, Becker F, Møller RS, Schubert J, Lemke JR, Larsen LH, et al. Benign infantile seizures and paroxysmal dyskinesia caused by an SCN8A mutation. Ann Neurol. 2016;79(3):428–36.PubMedCrossRef Gardella E, Becker F, Møller RS, Schubert J, Lemke JR, Larsen LH, et al. Benign infantile seizures and paroxysmal dyskinesia caused by an SCN8A mutation. Ann Neurol. 2016;79(3):428–36.PubMedCrossRef
109.
Zurück zum Zitat Epi4K Consortium. De novo mutations in SLC1A2 and CACNA1A are important causes of epileptic encephalopathies. Am J Hum Genet. 2016;99(2):287–98.CrossRef Epi4K Consortium. De novo mutations in SLC1A2 and CACNA1A are important causes of epileptic encephalopathies. Am J Hum Genet. 2016;99(2):287–98.CrossRef
110.
Zurück zum Zitat Larsen J, Carvill GL, Gardella E, Kluger G, Schmiedel G, Barisic N, et al. The phenotypic spectrum of SCN8A encephalopathy. Neurology. 2015;84(5):480–9.PubMedPubMedCentralCrossRef Larsen J, Carvill GL, Gardella E, Kluger G, Schmiedel G, Barisic N, et al. The phenotypic spectrum of SCN8A encephalopathy. Neurology. 2015;84(5):480–9.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Hirouchi M, Suzuki H, Itoda M, Ozawa S, Sawada J-i, Ieiri I, et al. Characterization of the cellular localization, expression level, and function of SNP variants of MRP2/ABCC2. Pharm Res. 2004;21(5):742–8.PubMedCrossRef Hirouchi M, Suzuki H, Itoda M, Ozawa S, Sawada J-i, Ieiri I, et al. Characterization of the cellular localization, expression level, and function of SNP variants of MRP2/ABCC2. Pharm Res. 2004;21(5):742–8.PubMedCrossRef
112.
Zurück zum Zitat Fokstuen S, Makrythanasis P, Hammar E, Guipponi M, Ranza E, Varvagiannis K, et al. Experience of a multidisciplinary task force with exome sequencing for Mendelian disorders. Hum Genomics. 2016;10(1):24.PubMedPubMedCentralCrossRef Fokstuen S, Makrythanasis P, Hammar E, Guipponi M, Ranza E, Varvagiannis K, et al. Experience of a multidisciplinary task force with exome sequencing for Mendelian disorders. Hum Genomics. 2016;10(1):24.PubMedPubMedCentralCrossRef
Metadaten
Titel
Novel and de novo mutations in pediatric refractory epilepsy
verfasst von
Jing Liu
Lili Tong
Shuangshuang Song
Yue Niu
Jun Li
Xiu Wu
Jie Zhang
Clement C. Zai
Fang Luo
Jian Wu
Haiyin Li
Albert H. C. Wong
Ruopeng Sun
Fang Liu
Baomin Li
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Brain / Ausgabe 1/2018
Elektronische ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-018-0392-5

Weitere Artikel der Ausgabe 1/2018

Molecular Brain 1/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.