• No results found

University of Groningen Genotyping and phenotyping epilepsies of childhood Vlaskamp, Danique

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Genotyping and phenotyping epilepsies of childhood Vlaskamp, Danique"

Copied!
214
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Genotyping and phenotyping epilepsies of childhood

Vlaskamp, Danique

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Vlaskamp, D. (2018). Genotyping and phenotyping epilepsies of childhood. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 1PDF page: 1PDF page: 1PDF page: 1

Genotyping and phenotyping

epilepsies of childhood

(3)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 2PDF page: 2PDF page: 2PDF page: 2

Printed by Ipskamp Printing, Enschede, the Netherlands

ISBN 978-94-034-1144-6 (printed version)

978-94-034-1143-9 (e-book)

© Copyright 2018 D.R.M. Vlaskamp, Groningen, the Netherlands

All rights reserved. No part of this publication may be reproduced or transmitted in any forms or by any means, without prior written permission of the author.

This research was funded by the Junior Scientifi c Masterclass.

Unrestricted travel grants were received from the Dutch Society of Child Neurology, Jo Kolk Foundation (VVAO), the Research School of Behavioural and Cognitive Neurosciences (BCN). Conference attendances were fi nancially supported by the BCN.

(4)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 3PDF page: 3PDF page: 3PDF page: 3

Genotyping and phenotyping

epilepsies of childhood

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de

rector magnificus prof. dr. E. Sterken en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op woensdag 05 december 2018 om 14.30 uur

door

Danique Rienke Maria Vlaskamp

geboren op 15 mei 1991

(5)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 4PDF page: 4PDF page: 4PDF page: 4

Prof. dr. O.F. Brouwer Copromotores Dr. P.M.C. Callenbach Dr. P. Rump

Beoordelingscommissie Prof. dr. V.V.A.M. Knoers Prof. dr. H.P.H. Kremer Prof. dr. K.P.J. Braun

(6)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 5PDF page: 5PDF page: 5PDF page: 5

COVER

The tree on the cover of this thesis symbolizes the relationship between the genotype and the phenotype

of epilepsies of childhood.

The beautiful tree has deeply anchored, widely spreading roots (genotype) that grow a new flowering tree with a unique pattern of branches,

leaves and blossoms (phenotype).

(7)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 6PDF page: 6PDF page: 6PDF page: 6

Paranimfen

Nienke te Grootenhuis Wieke Eggink

(8)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 7PDF page: 7PDF page: 7PDF page: 7

7

CONTENTS

Chapter 1 Introduction 9

PART I - THE YIELD OF GENETIC TESTING

Chapter 2 Copy number variation in a hospital-based cohort of children with epilepsy 27

Published as: Epilepsia Open. 2017; 2: 244–254.

Chapter 3 Empowerment and anxiety of patients and parents 49

during genetic counseling for epilepsy Manuscript ready to submit

PART II - GENOTYPE-PHENOTYPE STUDIES

Chapter 4 SYNGAP1 encephalopathy: a distinctive generalized 71

developmental and epileptic encephalopathy Accepted for publication in Neurology

Chapter 5 Genotype-phenotype correlation of 248 individuals with 95

GRIN2A-related disorders identifies two distinct phenotypic subgroups associated with different classes of variants, protein domains and functional consequences

Accepted for publication in Brain

Chapter 6 Schizophrenia is a later-onset feature of PCDH19 Girls Clustering Epilepsy 123

Revised version under review

Chapter 7 Haploinsufficiency of the STX1B gene is associated with 145

myoclonic astatic epilepsy

Published as: European Journal of Paediatric Neurology. 2016; 20: 489-492

Chapter 8 PRRT2-related phenotypes in patients with a 16p11.2 deletion 157

Published as: European Journal of Medical Genetics. 2018, in press.

Chapter 9 Positive effect of sodium channel blocking anti-epileptic 177

drugs on neonatal infantile epilepsy due to a SCN2A mutation Translated from the Dutch paper published as: Epilepsie. 2017; Periodiek voor Professionals 15, nr.3

Chapter 10 General discussion and a proposal for a diagnostic algorithm 185

for genetic testing for epilepsy

Chapter 11 Nederlandse samenvatting 201

Dankwoord 208

(9)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

(10)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 9PDF page: 9PDF page: 9PDF page: 9

9

9

Chapter 1

Introduction

(11)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 10PDF page: 10PDF page: 10PDF page: 10 I am about to discuss the disease called “sacred.” It is not, in my opinion, any more divine or more sacred

than any other diseases, but has a natural cause…

Its origin, like that of other diseases, lies in heredity... The fact is that the cause of this affection is... the brain...

My own view is that those who first attributed a sacred character to this malady were like the magicians, purifiers, charlatans, and quacks of our own day... So that there is no need to put the disease in a special class and to consider it more divine than the others...

Each has a nature and a power of its own; none is hopeless or incapable of treatment.1

Hippocrates, 460–370 BC, on epilepsy

Hippocrates was revolutionary for his time in arguing that the origin of epilepsy lies in ‘heredity’. Although hereditary does not always equal genetic, we now have robust evidence that genetic variants can cause or contribute to epilepsy. However, we still have a long way to go to fully understand the intriguing role of genetics in epilepsy.

To comprehend the current challenges in epilepsy genetics addressed in this thesis, it is helpful to learn more about epilepsy phenotyping and genotyping. Whereas epilepsy phenotyping concentrates on describing the clinical presentation, epilepsy genotyping focuses on identifying the underlying genetic cause. Integration of information on epilepsy phenotype and genotype is important in clinical practice and research settings in order to find new genes and risk factors for epilepsy, to better understand the disease and its presentation and to improve its management and outcome.

(12)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 11PDF page: 11PDF page: 11PDF page: 11

11

EPILEPSY PHENOTYPING

An epileptic seizure is defined as a transient occurrence of signs and/or symptoms due to abnormal

excessive or synchronous neuronal activity in the brain.2 Seizures can be provoked by acute

disturbances in the body, such as hypoglycemia or acute infection, or can occur unprovoked. Patients are diagnosed with epilepsy if they present with any of the following three conditions: 1. at least two unprovoked seizures occurring >24 hours apart, 2. one unprovoked (or reflex) seizure and a probability of further seizures in the next 10 years similar to the general recurrence risk after two

unprovoked seizures (at least 60%), or 3. a diagnosis of an epilepsy syndrome.3 Epilepsy is diagnosed

in 41-87 children per 100,000 children each year.4

Epilepsy is not a single disease entity, but should be considered as a group of disorders. Its presentation, etiology, management and prognosis can vary significantly for the different epilepsies. Epilepsy therefore warrants further classification for clinical and research purposes. The International

League Against Epilepsy (ILAE) has made several classification guidelines.5–11 In the most recent

guideline from 2017, they recommended classifying epilepsy at three consecutive levels: 1. seizure

type, 2. epilepsy type, and 3. epilepsy syndrome.12 The flowchart in Figure 1 demonstrates how to

classify epilepsies using this three-step method. Depending on the availability of clinical information, epilepsy can be classified up to level 1, 2 or 3.

1. Seizure type. Epileptic seizures should be classified based on their onset as focal, generalized

or unknown.12,13 Epilepsy is a disorder involving neuronal networks and all seizures are

hypothesized to occur somewhere within a network.14,15 Focal seizures are limited to networks

within one hemisphere, while generalized seizures rapidly engage bilaterally distributed

networks.16 Both clinical and electroencephalogram (EEG) data can help to distinguish focal

and generalized seizures. If more detailed clinical information on the seizures is available, they can be further classified based on the level of awareness (for focal seizures only) and

by describing their first feature.12,13 Focal seizures can evolve to bilateral tonic-clonic seizures

(previously called ‘secondary generalized seizures’).12,13,17

2. Epilepsy type. Since individuals can have both focal and generalized seizures, a second level of classification has been introduced: epilepsy type. Epilepsy type can be focal, generalized or

combined generalized and focal.12

3. Epilepsy syndrome. To diagnose the epilepsy syndrome, one should also consider the setting in which seizures occur. This setting includes the patient’s gender, age at seizure onset, developmental course, family history, and results from additional investigations such as EEG and imaging. Recognition of a distinctive clinical and EEG pattern allows a syndrome

diagnosis.12

INTRODUCTION

(13)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 12PDF page: 12PDF page: 12PDF page: 12

 >t z ^/  E d/ &z  d,  W /> W ^z  d /K >K ' z ^d Z h  dh Z  > ' E d / /E &  d/ K h ^ D d   K >/  /D D h E  h E <E K t E  ϭ ͘ ^ / h Z d zW  Ϯ ͘  W /> W ^z d zW  &K   > KŶůLJ ĨŽĐĂů ƐĞŝnj Ƶ ƌĞƐ  h E <E K t E  ' E Z  >/   KŶůLJ ŐĞŶĞƌĂůŝnj ĞĚ ƐĞŝnj ƵƌĞƐ ' E Z  >/    E  & K   > ŽƚŚ ĨŽĐĂů ĂŶĚ ŐĞŶĞƌĂůŝnjĞĚ ƐĞŝnjƵƌĞƐ ϯ ͘  W /> W ^z ^ zE  Z K D   K E d yd 'ĞŶĚĞƌ ͕Ă ŐĞ Ăƚ ƐĞŝnjƵƌ Ğ ŽŶƐ Ğƚ͕  ĚĞǀĞůŽƉŵĞŶƚ͕ ŝŶƚĞůůĞĐƚ͕ ĐŽŵŽƌ ďŝĚŝƚŝĞƐ͕ ĨĂŵŝůLJ ŚŝƐƚŽƌ LJ K d, Z & /E  /E ' ^ ZĞƐƵůƚƐ Ĩƌ Žŵ ŝŶǀĞƐƚŝŐĂƚŝŽŶƐ ;ŝŵĂŐŝŶŐ͕ĞůĞĐƚƌ ŽĞŶĐĞƉŚĂůŽŐƌ ĂŵͿ W /> W ^z  ^z E  Z K D   / ' E K ^/ ^ &K   >^ / h Z  ^ĞŝnjƵƌ Ğ ǁ ŝƚŚ ŝŶǀŽůǀĞŵĞŶƚ Ž Ĩ ŶĞƚǁŽƌ ŬƐ ŝŶ Ž ŶĞ ŚĞŵŝƐƉŚĞƌ Ğ ' E Z  >/   ^ / h Z  ^ĞŝnjƵƌ Ğ ǁ ŝƚŚ ŝŶǀŽůǀĞŵĞŶƚ ŽĨ ŶĞƚǁŽƌ ŬƐ ŝŶ ďŽƚŚ ŚĞŵŝƐƉŚĞƌ ĞƐ h E <E K t E hŶŬŶŽǁŶ Ž ƌ ƵŶĐůĂƐƐŝĨŝĂďůĞ ƐĞŝnjƵƌ Ğ K Ŷ ƐĞ ƚ  t  Z  E K d t  Z  D K dK Z  ƵƚŽŵ ĂƚŝƐŵ ͬ ĂƚŽŶŝĐ ͬ ĐůŽ Ŷ ŝĐ ͬ ƐƉ ĂƐ ŵƐ ͬ ŚLJƉĞ ƌŬŝŶĞ ƚŝĐ ͬ ŵ LJŽĐ ůŽ Ŷŝ Đ ͬ ƚŽŶŝ Đ E K E ͲD K dK Z  ƵƚŽŶŽŵ ŝĐ ͬ ďĞ ŚĂǀ ŝŽƌ ĂƌƌĞ Ɛƚ ͬ ĐŽ ŐŶŝƚŝǀĞ ͬĞ ŵŽ ƚŝŽ Ŷ Ăů ͬ ƐĞ ŶƐŽƌLJ  ǁ Ăƌ ĞŶ ĞƐ Ɛ &ŝ ƌƐ ƚĨ ĞĂ ƚƵ ƌĞ D K dK Z  dŽ Ŷ ŝĐ ͬ ĐůŽ Ŷ ŝĐ ͬ ƚŽ Ŷ ŝĐͲĐůŽ Ŷ ŝĐ ͬ ŵ LJŽĐ ůŽ Ŷŝ Đ ͬŵ LJŽ Đů ŽŶŝ ĐͲ ƚŽŶŝ ĐͲ Đů ŽŶŝ Đ ͬŵ LJŽĐ ůŽ Ŷŝ ĐͲ ĂƚŽ Ŷ ŝĐ ͬ ĂƚŽ Ŷ ŝĐ ͬ Ğ Ɖ ŝůĞ Ɖ ƚŝĐ ƐƉĂƐŵ E K E ͲD K dK Z  dLJƉ ŝĐĂů Ž ƌ Ăƚ LJƉ ŝĐĂů ĂďƐĞ ŶĐĞ ͬ ŵLJŽ ĐůŽ Ŷ ŝĐ ͬ ĞLJĞ ůŝĚ ŵLJŽ ĐůŽ Ŷ ŝĂ D K dK Z  dŽ Ŷ ŝĐͲĐůŽ Ŷ ŝĐ ͬ ĞƉ ŝůĞƉ ƚŝ Đ ƐƉĂƐŵ Ɛ E K E ͲD K dK Z Ğ ŚĂ ǀŝ ŽƌĂ ů ƌƌĞ Ɛƚ >t z^ / E d/& z KD KZ / /dz &Ž ĐĂ ůƚ Ž ď ŝůĂ ƚĞ ƌĂ ůƚ Ž Ŷ ŝĐ ͲĐ ůŽ Ŷ ŝĐ ƐĞ ŝnj Ƶ ƌĞ  Ž Ƶ ƌƐ Ğ W /> W ^z  dz W  F igur e 1: Flow char t f

or the classification of epilepsies f

ollowing a thr ee st ep method . M odified af ter F isher et al . 2017 & Scheff er et al . 2017. 12,13

(14)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 13PDF page: 13PDF page: 13PDF page: 13

13

Comorbidities

In parallel with classifying the epilepsy from seizure type to epilepsy syndrome, it is important to identify comorbidities. Examples of comorbidities include intellectual disability, movement disorders, autism spectrum disorders and other psychiatric and behavioral disorders. Pro-active screening for the presence of comorbidities is vital because it not only helps in epilepsy syndrome and etiology diagnosis, it also enables appropriate management of comorbidities that can improve quality of life of patients and their families.

Etiology

During the whole process of phenotyping epilepsies, it is also essential to try to identify the etiology. Understanding etiology can prevent unnecessary further diagnostic investigations and may lead to better counseling, management and, possibly, outcome. Six epilepsy etiology groups have been

proposed: genetic, structural, infectious, metabolic, immune and unknown.12 For some epilepsies,

etiology can be classified into more than one etiology category. For example, pyridoxine-dependent epilepsy due to a pathogenic ALDH7A1 variant has a metabolic and genetic etiology while DEPDC5-gene-related epilepsy with a cortical malformation has a genetic and structural etiology. For the genetic etiology, epilepsies can be considered genetic based on the presenting phenotype or

family history even when the variant itself has not yet been identified.12

Developmental and epileptic encephalopathy

In some epilepsies, “the epileptic activity itself may contribute to severe cognitive and behavioral

impairments that is beyond what might be expected from the underlying pathology alone”11,

defined as an epileptic encephalopathy (EE). There are many different EEs that can occur in infancy

and childhood, and these are often genetic.18 However, developmental slowing and regression

often precede seizure onset and cognitive deficits can persist after seizure remission. Therefore, the term developmental and epileptic encephalopathy (DEE) was introduced to describe the encephalopathies where the underlying genetic condition and the seizure activity together lead

to developmental plateauing or regression.12 An example of a DEE is SYNGAP1-encephalopathy,

which is studied in this thesis.

EPILEPSY GENOTYPING

Although the role of genetics in epilepsies has been appreciated since antiquity, research on this role only started the last century and the growth in knowledge since then has been revolutionary. What follows is a historical overview of the different consecutive study types that have led to our current understanding of the complex role of genetics in epilepsies.

INTRODUCTION

(15)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 14PDF page: 14PDF page: 14PDF page: 14

Family studies

The first evidence for epilepsy as a hereditary disease came from clinical family studies performed in the end of the last century. Family aggregation studies showed a 3-7 times higher incidence of

epilepsy in siblings and offspring of patients with epilepsy than expected by chance.19–21 Of patients

with epilepsy, 5-21% have at least one first-degree relative sharing a diagnosis of epilepsy.22–25

Furthermore, twin studies revealed a higher concordance for epilepsy among monozygotic versus

dizygotic twins.26–28 These studies together indicated that epilepsy has a genetic basis, but its

nature and extent are unknown. Nowadays, family studies are mainly performed to evaluate the phenotypic spectrum of familial epilepsy syndromes, e.g. genetic epilepsy with febrile seizures

plus29, or to identify new familial syndromes, e.g. familial posterior quadrant epilepsies.30

Searching for epilepsy candidate genes

Following these family studies, different approaches were used to identify the epilepsy candidate genes and their position within the human genome (called locus). Two important study types, linkage analysis and association studies, have been performed since the eighties and nineties of the last century, respectively.

Linkage studies.

Linkage analysis can be used in families to evaluate the segregation of epilepsy with genetic markers with a known position in the genome. This technique is based on the principle that genes that reside closely together are linked during meiosis. By identifying the genetic marker that is carried by affected relatives but not by non-affected relatives, the linked gene or locus for epilepsy can be identified. The first locus for epilepsy (juvenile myoclonus epilepsy on chromosome 6) was identified in 1989, and this was followed by the discovery of many other loci for this and other

epilepsy syndromes.31,32 Linkage analysis is still used to identify new loci for epilepsy.32

Association studies.

Association studies aim at identifying alleles that increase the risk for developing epilepsy, called ‘susceptibility alleles’. In these studies, the statistical association between a specific allele and an epilepsy syndrome is calculated by comparing the presence of this allele in large numbers of patients versus controls. Alleles found significantly more often in patients are considered susceptibility alleles. The many association studies performed to date have consistently identified a few susceptibility loci for different generalized epilepsies (1q43, 2p16.1, 2q22.3 and 17q21.32), focal and generalized epilepsy and febrile seizures (2q24.3 including SCN1A), febrile seizures (2q24.3, 11p14.2 and 12q21.33)

or focal and generalized epilepsy (4p15.1).33 Focusing on epilepsy treatment, an HLA-B*1502 allele

has been associated with the occurrence of carbamazepine-induced Stevens-Johnson syndrome.34

Unfortunately, many other findings from association studies could not be replicated, mainly due to low statistical power. Future collaboration between epilepsy research groups is necessary to increase the number of participants and thereby increase the statistical power to identify new loci

(16)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 15PDF page: 15PDF page: 15PDF page: 15

15

Molecular genetic studies

Advances in molecular genetic techniques that disentangle the human genome in greater detail together with more adequate epilepsy phenotyping have resulted in the identification of many new genes for epilepsy. Table 1 gives an overview of the different molecular techniques that are currently available to detect genetic variants at all levels of the human genome. Here, we will discuss the three most important techniques in terms of identifying new genes and loci for epilepsy.

Sanger sequencing.

In 1995, the first disease-causing gene variant for epilepsy (previously called a mutation) was identified using Sanger sequencing in a locus identified by linkage analysis. A missense mutation in the CHolinergic Receptor Nicotinic Alpha 4 Subunit gene (CHRNA4) was found in a large Australian

family with autosomal dominant nocturnal frontal lobe epilepsy.37 Sanger sequencing, invented

in 1977, has dominated genetic diagnostics for decades.38 Many disease-causing variants in

other genes (SCN1B39, KCNQ240, KCNQ341, SLC2A142, SCN1A43, GABRA144, and GABRG245) for other dominantly inherited epilepsies have been found following identification of their loci using linkage analysis. Currently, Sanger sequencing is mainly used to sequence a single gene selected based on a family history with variants in this gene or on an urgent need for specific treatment (such as SCL2A1 for glut1 deficiency requiring treatment with a ketogenic diet).

Microarray.

Microarray analysis has been a widely available technique since the early 2000’s. Microarray analysis

detects losses (deletions) or gains (duplications) of relatively small parts of the chromosomes.46

These deletions and duplications are called copy number variants (CNVs). There are two types of CNVs that can underlie epilepsy: recurrent and non-recurrent CNVs.

Recurrent CNVs occur at specific sites of the genome due to the recombination of DNA between highly homologous chromosomal regions (non-allelic homologous recombination). During meiosis, these highly homologous regions can misalign and an unequal crossover can occur, resulting in deletions

or duplications of the DNA. These deletions and duplications can include single or multiple genes.47

Recurrent CNVs are associated with different syndromes. For example, a 15q11.2-q13 microdeletion is associated with Angelman and Prader-Willi syndrome (OMIM 105830 and 176270, respectively) and a 22q13.33 microdeletion with Phelan-McDermid Syndrome (OMIM 606232). Epilepsy might occur in the context of these microdeletion or microduplication syndromes.

Non-recurrent CNVs are unique for every individual. These CNVs occur due to unequal crossover of DNA during meiotic (or less frequently mitotic) DNA repair processes with inconsistent breakpoints

throughout the genome in regions with limited homology.47 Identifying a critical region of overlap

(CRO) between such non-recurrent CNVs of patients with similar phenotypes can elucidate new genes for epilepsy. For example, GRIN2A was identified as a candidate gene for epilepsy based on

the identification of a CRO betewen 16p13.2 deletions in three patients with similar phenotypes.48

INTRODUCTION

(17)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 16PDF page: 16PDF page: 16PDF page: 16 Subsequently, disease-causing variants in GRIN2A were identified in patients with epilepsy within

the epilepsy-aphasia-spectrum.49–51

Several studies have shown that causal CNVs are present in ~5% of patients from different cohorts

with generalized epilepsy, focal epilepsy or epileptic encephalopathy.52–55 In addition, a few recurrent

CNVs have been found significantly more often in patients with epilepsy compared to controls, and these are now referred to as susceptibility CNVs because they can increase the risk of developing epilepsy. Deletions on chromosomes 1q21.1, 15q11.2, 15q13.3, 16p11.2 and 16p13.11 and duplications

on chromosome 16p11.2 are well-known examples of susceptibility CNVs.52,55–57

Next generation sequencing.

With the introduction of Next Generation Sequencing (NGS) in 2008-2009, we entered a new, very exciting era in epilepsy genetics research and diagnostics. NGS enables sequencing of multiple genes, of the whole exome or of the whole genome in a single test run. This has enabled two major advances. First, variants can be identified in genes that would otherwise not have been selected for genetic testing. NGS may therefore identify many novel candidate genes for epilepsy and, indeed,

the list of epilepsy candidate genes has grown tremendously since its introduction.58–60 Second,

gene-related phenotypic spectra can be expanded when gene variants are found in patients with phenotypes that were not yet associated with this gene. For example, SYNGAP1 was identified as a

gene for an epileptic encephalopathy after it was first identified as a gene for intellectual disability.61,62

GENETIC DATA INTERPRETATION

The genome of each individual, affected or non-affected, differs at 4-5 million sites from the

reference human genome and harbors 2,100 to 2,500 structural genetic variants.63 Not surprisingly,

the genome-wide genetic techniques discussed above also identify numerous variants that are not associated with disease. Therefore, our challenge is to interpret these variants and pinpoint the disease-causing variant in our patients. International guidelines have been established to classify variants based on their pathogenicity as benign, likely benign, of unknown significance,

likely pathogenic or definitely pathogenic.64 Variants are first filtered based on their prevalence in

national and international population databases such as the genome of the Netherlands (GoNL)65,

genome Aggregation Database (gnomAD)66, 1000 Genomes63, dbSNP67 and the database for

genomic variants (DGV)68. A variants is considered likely benign if it is present in more than 1% of

the population.64 For the variants that are not considered likely benign, the variant’s inheritance,

location, in silico predicted or functionally tested effects, presence in other patients with similar

phenotypes are evaluated to determine its pathogenicity.64 With the introduction of whole exome

sequencing, and possibly whole genome sequencing in the near future, the interpretation of genetic testing is becoming more difficult and time-consuming as ever more variants are being identified.

(18)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 17PDF page: 17PDF page: 17PDF page: 17

17

T

able 1:

Genetic t

echniques and their indications

, advantages , disadvantages Human genome le v e l for detec ting var ian ts Te st Va ri a ti o n s dete ct e d Ty pical indicat ion A dvan tages Disadvan tages Ka ry o ty p in g La rg e chr o m os o mal re ar ra nge me n ts Su spicion of a chr o m os o mal di so rd er - Ab le t o det ec t r ing chr o m o so m es an d b alanc ed re ar ra nge me n ts - L o w r es o lution c o ver age of genom e - Po ss ibilit y of inciden tal fi n d ing s for di se as es o th er than epileps y Micr o ar ra y C N Vs , r egion s of homo zy go si ty , unip ar en tal is o d is o m y Ep ileps y “ p lu s” (d evelopm en tal dela y, b eh aviora l pr oble m s o r d ysm o rp h isms ) - High r es o lution co ve rage of ge nome - Mi ss es b alanc ed chr o m o so mal re ar ra nge me n ts - Po ss ibilit y of inciden tal fi n d ing s for di se as es o th er than epileps y Sa nger se q u en ci ng Se qu en ce v ar ian ts in s ingle or up t o a few gen es Ep ile p sy sy n d ro m e kn ow n t o b e c au se d by a s ingle gen e va ria n t - N o p o ss ibilit y of in ci de n ta l fi nd in g s due t o c ar ef u l se le ct ion of gen es - Pr e-se le ct ion of gen e b as ed on clinic al pr es en ta tion i s n ec ess ar y - Ti m e- an d c o st -e xp en si ve t o s equenc e g en es in di vi duall y - N o t p o ss ib le t o det ec t in tr ag enic C N Vs Ep ile p sy gen e p an el Se qu en ce v ar ian ts within a s u bs et of ge nes G en etic epileps y - Lo w p o ss ibilit y of in ci de n ta l fi nd in g s due t o c ar ef u l se le ct ion of gen es - G en e p an el s n ee d t o b e up da te d fr eq uen tl y - So m e lab o ra to ri es : no t p o ss ib le t o det ec t i n trage n ic C N Vs Whol e ex o m e se q u en ci ng with epileps y fi lt er Se qu en ce v ar ian ts in th e e xom es, but on ly a n al yz es of subs et o f g en es G en etic epileps y - Lo w p o ss ibilit y of in ci de n ta l fi nd in g s due t o c ar ef u l se le ct ion of gen es, which c an b e ex te nde d e as ily - So m e lab o ra to ri es : no t p o ss ib le t o det ec t i n trage n ic C N Vs Whol e ex o m e se q u en ci ng Se qu en ce v ar ian ts within in th e ex o m es G en etic epileps y without a gen e va rian t iden tifi e d u sing a gen e p an el - N o n ee d for s ele ct ion of gen es if no clinic al clu e Whol e ge nome se q u en ci ng Se qu en ce v ar ian ts an d C N Vs in th e whole ge nome (int ro ns + e xo n s) C u rr en tl y onl y app lie d in re se ar ch se tt in g s - N o n ee d for s ele ct ion of gen es if no clinic al clu e - Da ta a vailab le t o det ec t i n tr on ic var ian ts an d e xo n ic co py numb er v ar ian ts - N o t yet p oss ib le t o re liab ly in te rp ret th e re lev anc e of in tr onic v ar ian ts - Po ss ibilit y of inciden tal fi n d ing s for di se as es o th er than epileps y INTRODUCTION

1

(19)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 18PDF page: 18PDF page: 18PDF page: 18

CONCEPTUALIZING EPILEPSY GENETICS

Over the last few decades our understanding of the genetic basis of epilepsies has increased tremendously and this knowledge can be summarized in three important concepts.

Epilepsy gene products are part of different pathways

When the first ion-channel-coding genes for epilepsies were discovered, the idea was raised that

epilepsy could be channelopathy.60 This concept has however been overturned by the discovery

in 2002 of LGI1, a non-ion-channel-coding gene for autosomal dominant lateral temporal lobe

epilepsy.69 Following the identification of many non-ion-coding-channel genes for epilepsy, we

realized that epilepsy gene products can be part of different pathways and thereby affect neuronal function differently. These pathways include DNA repair, transcriptional regulation, axon myelination, metabolite and ion transport, peroxisomal function, cell-cell adhesion, neurite formation,

interneuron migration, apoptosis and blood-brain barrier transport.70 Notably, gene products

within the same pathway can often result in a similar epilepsy phenotype. The identification of DEPDC5 as the causal gene for focal epilepsy and cortical brain malformations in the mammalian target of rapamycin (mTOR) pathway, known to be involved in tuberous sclerosis, nicely illustrates

this.71,72 Even more striking, sequencing of other genes in the mTOR pathway identified variants in

NPRL2 and NPRL3, both associated with a similar phenotype.73

Epilepsy encompasses different modes of inheritance

We have learned that epilepsy can be inherited following different modes of inheritance. Although some epilepsies are caused by a single gene variant (monogenic inheritance), the majority of epilepsies follow a complex inheritance where many genes (polygenic inheritance) or a combination of genes

and environmental factors (multifactorial inheritance) increase the risk of developing epilepsy.74 In

monogenic epilepsies, the disease-causing gene variant does not necessarily lead to disease in all relatives carrying this variant, a phenomenon called incomplete penetrance. Alternatively, a relative might be affected yet not carry the disease-causing variant, a condition referred to as a phenocopy.

Phenotypic and genetic heterogeneity play a key role in epilepsy

Phenotypic heterogeneity (also called pleiotropy) refers to a single genetic disorder leading to different epilepsy syndromes: e.g. mutations in KCNQ2 can cause benign neonatal and infantile epilepsy

as well as severe neonatal epileptic encephalopathy.75,76 Genetic heterogeneity means that a single

epilepsy syndrome can be caused by pathogenic variants in different genes. For example, benign

infantile epilepsy can by caused by variants in KCNQ2, KCNQ3, SCN2A or PRRT2.75 For some genes,

such as SCN1A, clear genotype-phenotype correlations have been identified that explain the SCN1A phenotypic heterogeneity by focusing on the mutation type and location of SCN1A variants in

relation to the phenotype.77 For other genes, such as SYNGAP1 and GRIN2A, we still lack a clear

(20)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 19PDF page: 19PDF page: 19PDF page: 19

19

ADVANCES AND CHALLENGES IN EPILEPSY GENETICS IN

CURRENT CLINICAL PRACTICE

The yield of genetic testing

Not long ago it was unimaginable that we would now know of 700 genes related to epilepsy.58

Even so, we still cannot identify the genetic cause in a significant number of epilepsy cases that are suspected to be genetic. Only one large and two very small studies addressed the yield of microarray in clinical hospital cohorts of patients with all types of epilepsies: the yield varied between 9% and 40%.55,80,81 In one of these studies, new loci of interest for epilepsy were identified.55 In chapter 2, we describe the diagnostic yield of microarray in a large clinical hospital cohort of children with epilepsy in the Netherlands and the possible novel CNVs for epilepsy.

With our increasing ability to identify the underlying genetic causes of epilepsy, genetic testing is offered more and more in clinical practice. Using these techniques, we hope not only to improve epilepsy care but also to provide answers to longstanding questions from patients and parents about the epilepsy cause and prognosis. However, the impact on patients and their parents of genetic

counseling and receiving a genetic diagnosis has not yet been studied.82,83 In chapter 3, we present

the results of a study evaluating empowerment and anxiety during a genetic counseling trajectory in patients with epilepsy or their parents in whom genetic causes were and were not identified.

Genotype-phenotype studies

With the introduction of NGS, many new genes for epilepsy have been identified in different cohorts of patients selected based on their phenotype. Due to this ascertainment, the clinical features associated with these genes is often biased in the original papers. To unravel the full gene-related phenotypic spectrum and the genotype-phenotype correlations, patients should be selected based on their genotype and subsequently be phenotyped (a process called reverse phenotyping). In chapter 4 and chapter 5, we report the phenotypic spectra and genotype-phenotype correlations for SYNGAP1 and GRIN2A, respectively. In chapter 6, we show that psychotic disorders are a new, later-onset manifestation of PCDH19 Girls Clustering Epilepsy. Chapter 7 describes a case report of a boy with a deletion of the STX1B gene and a new phenotype: epilepsy with myoclonic-atonic seizures (previously called myoclonic astatic epilepsy).

In Chapter 8, we evaluate the presence of PRRT2-related phenotypes in patients with 16p11.2 microdeletions including PRRT2 and the 16p11.2 microdeletion syndrome.

All efforts to identify genes for epilepsy are driven by the hope of improving epilepsy care. For now, however, the identification of new epilepsy genes has been translated into better management of seizures in only a few cases. Currently, ‘personalized’ medicine for epilepsy has been established in only two scenarios: a ketogenic diet for patients with SLC2A1 variants and treatment with pyridoxine

for patients with ALDH7A1 variants.84 In chapter 9, we report the efficacy of carbamazepine as a

INTRODUCTION

(21)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 20PDF page: 20PDF page: 20PDF page: 20 sodium-channel blocker in the case of a girl with infantile epilepsy due to a SCN2A sequence variant

and. This case report is an example of precision medicine.

In chapter 10 we discuss our results in a general perspective and we propose a diagnostic algorithm for genetic testing for epilepsy in clinical practice.

(22)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 21PDF page: 21PDF page: 21PDF page: 21

21

REFERENCES

1. Jones WHS [translator]. Hippocrates, vol II. Cambridge: Harvard; 1923.

2. Fisher RS, van emde Boas W, Blume W, et al. Epileptic seizures and epilepsy: Definitions proposed by the International League Against Epilepsy (ILAE) and the International Bureau for Epilepsy (IBE). Epilepsia 2005; 46: 470-472.

3. Fisher RS, Acevedo C, Arzimanoglou A, et al. ILAE Official Report: A practical clinical definition of epilepsy. Epilepsia. 2014; 55: 475–482.

4. Camfield P, Camfield C. Incidence, prevalence and aetiology of seizures and epilepsy in children. Epileptic Disord. 2015; 17: 117–123.

5. Classification PI, Seizures E. A Proposed International Classification of Epileptic Seizures. Epilepsia. 1964; 5: 297–306.

6. Gastaut H. Clinical and Electro-encephalographical Classification of Epileptic Seizures. Epilepsia 1970; 11: 102–112.

7. Merlis JK. Proposal for an international classification of the epilepsies. Epilepsia 1970; 11: 114–119.

8. Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for Revised Classification of Epilepsies and Epileptic Syndromes: Commission on Classification and Terminology of the International League Against Epilepsy. Epilepsia 1981; 22: 489–501.

9. Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for the classification of epilepsies and epileptic syndromes. Epilepsia 1985; 26: 268–278.

10. Commission on Classification and Terminology of the International League Against Epilepsy. Proposal for Revised Classification of Epilepsies and Epileptic Syndromes. Epilepsia 1989; 30: 389–399.

11. Berg AT, Berkovic SF, Brodie MJ, et al. Revised terminology and concepts for organization of seizures and epilepsies: Report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia 2010;51:676–685. 12. Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification

of the epilepsies: Position paper of the ILAE Commission for Classification and Terminology. Epilepsia 2017; **: 1–10. doi: 10.111/epi.13709

13. Fisher RS, Cross JH, D’Souza C, et al. Instruction manual for the ILAE 2017 operational classification of seizure types. Epilepsia 2017; **: 1–9. doi: 10.1111/epi. 13670. 14. Laufs H. Functional imaging of seizures and epilepsy:

evolution from zones to networks. Curr Opin Neurol 2012; 25: 194–200.

15. Abela E, Rummel C, Hauf M, Weisstanner C, Schindler K, Wiest R. Neuroimaging of epilepsy: Lesions, networks, oscillations. Clin Neuroradiol 2014; 24: 5–15. 16. Berg AT, Scheffer IE. New concepts in classification of the

epilepsies: Entering the 21st century. Epilepsia 2011; 52: 1058-1062.

17. Blumenfeld H, Meador KJ. Consciousness as a useful concept in epilepsy classification. Epilepsia 2014; 55: 1145–1150.

18. McTague A, Howell KB, Cross JH, Kurian MA, Scheffer IE. The genetic landscape of the epileptic encephalopathies of infancy and childhood. Lancet Neurol 2016; 15: 304-316.

19. Lennox WG. The heredity of epilepsy as told by relatives and twins. JAMA 1951; 146: 529–536.

20. Annegers J, Hauser W, Anderson V, Kurland L. The risks of seizure disorders among relatives of patients with childhood onset epilepsy. Neurology 1982; 32: 174–179. 21. Ottman R, Annegers JF, Hauser WA, Kurland LT. Seizure

Risk in Offspring of Parents with Generalized Versus Partial Epilepsy. Epilepsia 1989; 30: 157–161.

22. Aziz H, Ali SM, Frances P, Khan MI, Hasan KZ. Epilepsy in Pakistan: a population-based epidemiologic study. Epilepsia 1994; 35: 950–958.

23. Monetti VC, Granieri E, Casetta I, et al. Risk-Factors for Idiopathic Generalized Seizures - a Population-Based Case-Control Study in Copparo, Italy. Epilepsia 1995; 36: 224–229.

24. Jain S, Padma MV, Puri A, Jyoti, Maheshwari MC. Occurrence of epilepsies in family members of Indian probands with different epileptic syndromes. Epilepsia 1997; 38: 237–244.

25. Callenbach PM, Geerts AT, Arts WFM, et al. Familial occurrence of epilepsy in children with newly diagnosed multiple seizures: Dutch Study of Epilepsy in Childhood. Epilepsia 1998; 39: 331–336.

26. Berkovic SF, Howell RA, Hay D, Hopper J. Epilepsies in twins: genetics of the major epilepsy syndromes. Ann Neurol 1998; 43: 435–445.

27. Corey LA, Berg K, Pellock JM, Solaas MH, Nance WE, DeLorenzo RJ. The occurrence of epilepsy and febrile seizures in Virginian and Norwegian twins. Neurology 1991; 41: 1433–1436.

28. Sillanpaa M, Koskenvuo M, Romanov K, Kaprio J. Genetic factors in epileptic seizures: evidence from a large twin population. Acta Neurol Scand 1991; 84: 523–526. 29. Zhang YH, Burgess R, Malone JP, et al. Genetic epilepsy

with febrile seizures plus. Neurology 2017; 89: 1210-1219. 30. Abou-Khalil B, Afawi Z, Allen AS, et al. Phenotypic analysis of 303 multiplex families with common epilepsies. Brain 2017; 140: 2144–2156.

31. Greenberg D, Delgado-Escueta A, Widelitz H, et al. Juvenile myoclonic epilepsy (JME) may be linked to the BF and HLA loci on human chromosome 6. Am J Med Genet 1988; 31: 185–192.

32. Chen T, Giri M, Xia Z, Subedi YN, Li Y. Genetic and epigenetic mechanisms of epilepsy: A review. Neuropsychiatr Dis Treat 2017; 13: 1841–1859.

33. Koeleman BPC. What do genetic studies tell us about the heritable basis of common epilepsy? Polygenic or complex epilepsy? Neurosci Lett 2017; doi: 10.1016/j. neulet. 2017.03.042.

INTRODUCTION

(23)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 22PDF page: 22PDF page: 22PDF page: 22 34. Chung W-H, Hung S-I, Hong H-Sh, et al. A marker for

Stevens-Johnson syndrome. Nature 2004; 428: 486. 35. Tan NCK, Mulley JC, Berkovic SF. Genetic association

studies in epilepsy: “The truth is out there.” Epilepsia 2004; 45: 1429–1442.

36. Risch NJ. Searching for genetic determinants in the new millennium. Nature. 2000; 405: 847-856.

37. Steinlein OK, Mulley JC, Propping P, et al. A missense mutation in the neuronal nicotinic acetylcholine receptor alpha 4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy. Nat Genet 1995; 11: 201–203.

38. Sanger F, Nicklen S. DNA sequencing with chain-terminating. 1977; 74: 5463–5467.

39. Wallace RH, Wang DW, Singh R, et al. Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B. Nat Genet 1998; 19: 366–370.

40. Singh NA, Charlier C, Stauffer D, et al. A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns. Nat Genet 1998; 18: 25–29.

41. Charlier C, Singh NA, Ryan SG, et al. A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family. Nat Genet 1998; 18: 53–55.

42. Seidner G, Alvarez MG, Yeh JI, et al. GLUT-1 deficiency syndrome caused by haplo-insufficiency of the blood- brain barrier hexose carrier. Nat Genet 1998; 18: 188–191. 43. Escayg A, MacDonald BT, Meisler MH, et al. Mutations

of SCN1A, encoding a neuronal sodium channel, in two families with GEFS+2. Nat Genet 2000; 24: 343–345. 44. Cossette P, Liu L, Brisebois K, et al. Mutation of GABRA1

in an autosomal dominant form of juvenile myoclonic epilepsy. Nat Genet 2002; 31: 184–189.

45. Baulac S, Huberfeld G, Gourfinkel-An I, et al. First genetic evidence of GABA(A) receptor dysfunction in epilepsy: a mutation in the gamma2-subunit gene. Nat Genet 2001; 28: 46–48.

46. Mefford HC. CNVs in Epilepsy. Curr Genet Med Rep 2014; 2: 162–167.

47. Hastings PJ, James R Lupski SMR and GI. Mechanisms of change in gene copy number. Nat Rev Genet 2010; 10: 551–564.

48. Reutlinger C, Helbig I, Gawelczyk B, et al. Deletions in 16p13 including GRIN2A in patients with intellectual disability, various dysmorphic features, and seizure disorders of the rolandic region. Epilepsia 2010; 51: 1870– 1873.

49. Endele S, Rosenberger G, Geider K, et al. Mutations in GRIN2A and GRIN2B encoding regulatory subunits of NMDA receptors cause variable neurodevelopmental phenotypes. Nat Genet 2010; 42: 1021–1026.

50. Lemke JR, Lal D, Reinthaler EM, et al. Mutations in GRIN2A cause idiopathic focal epilepsy with rolandic spikes. Nat Genet 2013; 45: 1067-1072.

51. Lesca G, Rudolf G, Bruneau N, et al. GRIN2A mutations in acquired epileptic aphasia and related childhood focal epilepsies and encephalopathies with speech and language dysfunction. Nat Genet 2013; 45: 1061–1066.

52. Mefford HC, Muhle H, Ostertag P, et al. Genome-wide copy number variation in epilepsy: novel susceptibility loci in idiopathic generalized and focal epilepsies. PLoS Genet 2010; 6: 1–9.

53. Heinzen EL, Radtke RA, Urban TJ, et al. Rare Deletions at 16p13.11 Predispose to a Diverse Spectrum of Sporadic Epilepsy Syndromes. Am J Hum Genet 2010; 86: 707–718. 54. Mefford HC, Yendle SC, Hsu C, et al. Rare copy

number variants are an important cause of epileptic encephalopathies. Ann Neurol 2012; 70: 974–985. 55. Olson H, Shen Y, Ph D, et al. Copy number variation plays

an important role in clinical epilepsy. Ann Neurol 2014; 75: 943–958.

56. De Kovel CGF, Trucks H, Helbig I, et al. Recurrent microdeletions at 15q11.2 and 16p13.11 predispose to idiopathic generalized epilepsies. Brain 2010; 133: 23–32. 57. Mullen SA, Carvill GL, Bellows S, et al. Copy number

variants are frequent in genetic generalized epilepsy with intellectual disability. Neurology 2013; 81: 1507– 1514.

58. Wang J, Lin Z-J, Liu L, et al. Epilepsy-associated genes. Seizure 2017; 44: 11–20.

59. Møller RS, Dahl HA, Helbig I. The contribution of next generation sequencing to epilepsy genetics. Expert Rev Mol Diagn 2015; 15: 1531-1538.

60. Helbig I, Abou Tayoun AN. Understanding genotypes and phenotypes in epileptic encephalopathies. Mol. Syndromol 2016; 7: 172-181.

61. Hamdan FF, Gauthier J, Spiegelman D, et al. Mutations in SYNGAP1 in Autosomal Nonsyndromic Mental Retardation. N Engl J Med 2009; 360: 599–605.

62. Carvill GL, Heavin SB, Yendle SC, et al. Targeted resequencing in epileptic encephalopathies identifies de novo mutations in CHD2 and SYNGAP1. Nat Genet 2013; 45: 825-830.

63. Auton A, Abecasis GR, Altshuler DM, et al. A global reference for human genetic variation. Nature 2015; 526: 68–74.

64. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J. 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: 405–424.

65. Francioli LC, Menelaou A, Pulit SL, et al. Whole-genome sequence variation, population structure and demographic history of the Dutch population. Nat Genet 2014; 46: 818–825.

66. Lek M, Karczewski KJ, Minikel E V, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016; 536: 285–291.

67. Sherry S, Ward M, Kholodov M, et al. dbSNP: the NCBI database of genetic variation. Nucleic Acids Res 2001; 29: 308–311.

68. MacDonald JR, Ziman R, Yuen RKC, Feuk L, Scherer SW. The Database of Genomic Variants: A curated collection of structural variation in the human genome. Nucleic Acids Res 2014; 42: 986–992.

(24)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 23PDF page: 23PDF page: 23PDF page: 23

23

69. Kalachikov S, Evgrafov O, Ross B, et al. Mutations in LGI1 cause autosomal-dominant partial epilepsy with auditory features. Am J Hum Genet 2008; 30: 335–341. 70. McTague A, Howell KB, Helen Cross J, Kurian MA,

Scheff er IE. The genetic landscape of the epileptic encephalopathies of infancy and childhood. Lancet Neurol 2016; 15:304–316.

71. Dibbens LM, De Vries B, Donatello S, et al. Mutations in DEPDC5 cause familial focal epilepsy with variable foci. Nat Genet 2013; 45: 546–551.

72. Scheffer IE, Heron SE, Regan BM, et al. Mutations in mammalian target of rapamycin regulator DEPDC5 cause focal epilepsy with brain malformations. Ann Neurol 2014; 75: 782–787.

73. Ricos MG, Hodgson BL, Pippucci T, et al. Mutations in the mammalian target of rapamycin pathway regulators NPRL2 and NPRL3 cause focal epilepsy. Ann Neurol 2016; 79: 120–131.

74. Berkovic SF, Mulley JC, Scheffer IE, Petrou S. Human epilepsies: interaction of genetic and acquired factors. Trends Neurosci 2006; 29:391–397.

75. Zara F, Specchio N, Striano P, et al. Genetic testing in benign familial epilepsies of the first year of life: Clinical and diagnostic significance. Epilepsia 2013; 54: 425–436. 76. Weckhuysen S, Mandelstam S, Suls A, et al. KCNQ2 encephalopathy: Emerging phenotype of a neonatal epileptic encephalopathy. Ann Neurol 2012; 71: 15–25.

77. Zuberi SM, Brunklaus A, Birch R, Reavey E, Duncan J, Forbes GH. Genotype-phenotype associations in SCN1A-related epilepsies. Neurology 2011; 76: 594–600. 78. Mignot C, Stülpnagel C Von, Nava C, et al. Genetic and

neurodevelopmental spectrum of SYNGAP1 -associated intellectual disability and epilepsy. J Med Gen 2016; 53: 511-522.

79. Myers KA, Scheffer IE. GRIN2A-Related Speech Disorders and Epilepsy. GeneReviews®. Washington: Seattle. 2016. 80. Faheem M, Nasser M, Chaudhary A, et al.

Array-comparative genomic hybridization analysis of a cohort of Saudi patients with epilepsy. CNS Neurol Disord Drug Targets 2015; 14: 468–475.

81. Naseer MI, Faheem M, Chaudhary AG, et al. Genome wide analysis of novel copy number variations duplications / deletions of different epileptic patients in Saudi Arabia. BMC Genomics 2015; 16: S10.

82. Shostak S, Ottman R. Ethical, legal, and social dimensions of epilepsy genetics. Epilepsia 2006; 47: 1595–1602. 83. Shostak S, Zarhin D, Ottman R. What’s at stake? Genetic

information from the perspective of people with epilepsy and their family members. Soc Sci Med 2011; 73: 645–654.

84. Balestrini S, Sisodiya SM. Pharmacogenomics in epilepsy. Neurosci Lett 2017. doi: 10.1016/ j.neulet.2017.01.014.

INTRODUCTION

(25)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

(26)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 25PDF page: 25PDF page: 25PDF page: 25

25 25

PART I

(27)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

(28)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 27PDF page: 27PDF page: 27PDF page: 27

27

27

Chapter 2

Copy number variation in

a hospital-based cohort of

children with epilepsy

Published as: DRM Vlaskamp, PMC Callenbach, P Rump, et al. Copy number variation in a hospital-based cohort of children with epilepsy. Epilepsia Open, 2017; 2: 244–254.

(29)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 28PDF page: 28PDF page: 28PDF page: 28

Danique RM Vlaskamp1,2, Petra MC Callenbach1*, Patrick Rump2*, Lucia AA Giannini2, Trijnie

Dijkhuizen2, Oebele F Brouwer1, Conny MA van Ravenswaaij-Arts2

1 University of Groningen, University Medical Center Groningen, Department of Neurology,

Groningen the Netherlands. 2 University of Groningen, University Medical Center Groningen,

Department of Genetics, Groningen, the Netherlands. * These authors contributed equally to this work

Acknowledgements. P.M.C. Callenbach received an unrestricted research grant from UCB Pharma BV, the Netherlands. UCB Pharma BV had no role in the study design, data collection, and analysis, decision to publish, or preparation of the manuscript. We thank K. McIntyre for editing the manuscript and J. Anderson for helping identifying CNVs for epilepsy reported in the literature.

Disclosures. None of the authors has any conflicts of interest to disclose. We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.

(30)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 29PDF page: 29PDF page: 29PDF page: 29

29

ABSTRACT

Objective. To evaluate the diagnostic yield of microarray analysis in a hospital-based cohort of children with seizures and to identify novel candidate genes and susceptibility loci for epilepsy. Methods. Of all children who presented with their first seizure in the University Medical Center Groningen (January 2000 through May 2013) (n = 1,368), we included 226 (17%) children who underwent microarray analysis before June 2014. All 226 children had a definite diagnosis of epilepsy. All their CNVs on chromosomes 1-22 and X that contain protein-coding genes and have a prevalence of <1% in healthy controls were evaluated for their pathogenicity.

Results. Children selected for microarray analysis more often had developmental problems (82% vs 25%, p < 0.001), facial dysmorphisms (49% vs 8%, p < 0.001) or behavioral problems (41% vs 13%, p < 0.001) than children who were not selected. We found known clinically relevant CNVs for epilepsy in 24 of the 226 children (11%). Seventeen of these 24 children had been diagnosed with symptomatic focal epilepsy not otherwise specified (71%) and five with West syndrome (21%). Of these 24 children, many had developmental problems (100%), behavioral problems (54%) or facial dysmorphisms (46%). We further identified five novel CNVs comprising four potential candidate genes for epilepsy: MYT1L, UNC5D, SCN4B and NRXN3.

Significance. The 11% yield in our hospital-based cohort underscores the importance of microarray analysis in diagnostic evaluation of children with epilepsy.

COPY NUMBER VARIATION IN A HOSPITALBASED COHORT OF CHILDREN WITH EPILEPSY

(31)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 30PDF page: 30PDF page: 30PDF page: 30

INTRODUCTION

Genetic factors play an important role in the etiology of epilepsy,1 as demonstrated by the large

number of genes and regions that cause or predispose to epilepsy newly identified by various

genome-wide technologies.2 Chromosomal microarray analysis, in particular, enables the

identification of chromosomal deletions (losses) or duplications (gains), called Copy Number Variants

(CNVs).3 CNVs may contribute to epilepsy in two ways. First, CNVs that comprise epilepsy-related

genes could lead to epilepsy following a Mendelian inheritance. For example, both KCNQ2 sequence

variants and whole gene deletions can cause benign familial neonatal seizures.4,5 Second, CNVs

that occur more frequently in patients compared to healthy controls may increase an individual’s susceptibility to developing epilepsy, with the responsible haploinsufficient gene(s) often being unknown. Large cohort studies have identified such susceptibility CNVs in several chromosomal

regions, including well-known CNVs located at 15q11.2 (BP1-BP2), 15q13.3 and 16p13.11.6-8

Studies using microarray analysis have most often been performed in research cohorts of children who were selected based on their epilepsy diagnosis, e.g. idiopathic generalized epilepsies, focal

epilepsies and/or fever-associated epilepsies.6-8 Only a few studies have addressed the yield of

microarray analysis in clinical cohorts of all children presenting with any type of seizures in a clinical

setting.9-11 We therefore aimed to evaluate the diagnostic yield of microarray analysis in a

hospital-based cohort of children with epilepsy for whom detailed phenotypic information was available, with the further goal of identifying novel candidate genes or susceptibility loci for epilepsy.

MATERIAL AND METHODS

Study cohort

The study cohort was derived from the childhood seizure database of the University Medical Center Groningen (UMCG), a regional referral center for children with epilepsy. In this database, we retrospectively included all children who presented with their first febrile or afebrile seizure before the age of 18 years between January 2000 and June 2013, and who were seen and/or treated by a child neurologist of the UMCG (n = 1,368). Epilepsy was diagnosed in 91% of these children using the current International League Against Epilepsy (ILAE) practical clinical definition

of epilepsy.12 Of the remaining children (9%), 7% had febrile seizures only and 2% had only one

afebrile seizure. The UMCG database contains phenotype information and was independently completed by two researchers (DRMV and PMCC). Phenotypic inconsistencies and epilepsy classification were discussed until agreement was reached using the information in the database as well as in the original medical records (PMCC and OFB). Epilepsy syndromes and seizure types

were classified according to the 2006 ILAE classification.13 Children were included in this study if

(32)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 31PDF page: 31PDF page: 31PDF page: 31

31

Formal independent review board evaluation was waived by the Institutional Medical Ethical Committee of the UMCG because of the retrospective and observational character of this study.

Chromosomal microarray analysis and data interpretation

Microarray analyses were performed using an oligonucleotide array (Agilent 105K or 180K custom HD-DGH microarray; Agilent Technologies, Santa Clara, USA) or a single nucleotide polymorphism (SNP) array (Illumina Omni Express 12-V1.0; Illumina, San Diego, USA). Cartagenia Bench Lab CNV software was used for storage, analysis and reporting of the structural genomic data (Cartagenia, Leuven, Belgium; part of Agilent Technologies, Santa Clara, USA). The chromosomal coordinates of CNVs were reported relative to the Genome Reference Consortium Human Reference genome version 37 (GRCh37/hg19).

CNVs on chromosome 1-22 or X identified by at least three (SNP microarray) or four (oligonucleotide microarray) consecutive probes were evaluated for their pathogenicity (Figure 1). CNVs were excluded from further analysis when they did not contain (protein-coding) genes or had ≥90% overlap with CNVs seen in ≥1% of healthy controls. The prevalence of CNVs in healthy controls was calculated using the International Database of Genomic Variance (n =

14,316, last updated February 2013),14 the Low Lands Consortium database of oligonucleotides (n

= 2,402, last updated December 2012) and SNP microarray results of healthy parents of children who underwent microarray analysis in five Dutch genetic centers (n = 749, last updated October 2014). Remaining CNVs were categorized into two groups: (1) CNVs with <90% overlap with CNVs observed in healthy controls and (2) CNVs with ≥90% overlap with CNVs observed in <1% of the healthy controls (Figure 1). CNVs in both groups were marked as potentially clinically relevant if they had overlap with genetic regions previously associated with epilepsy. These regions were identified by performing a literature search using PubMed, complemented with information from the Decipher database and Cartagenia Bench Lab CNV software. The remaining CNVs in both groups were evaluated for novel candidate genes or susceptibility loci for epilepsy. CNVs with <90% overlap with CNVs of healthy controls were of interest if they contained a gene with an expression or function in the brain or a gene associated with an autosomal dominant or X-linked neuropsychiatric disease, and if they occurred in at least one (for deletions) or two (for duplications) unrelated children in our cohort. In the group of CNVs with ≥90% overlap with CNVs observed in <1% of the healthy controls, overlapping regions between CNVs in at least two unrelated children were of interest if these regions contained protein-coding genes and were 10 times more prevalent in our cohort compared to healthy controls.

Statistical analyses

SPSS Statistics Version 22.0 (IBM Corporation, NY, USA) was used to perform descriptive and comparative statistics. Differences in categorical and ordinal phenotypic data between children were analyzed using Fisher’s exact and Mann-Whitney U tests, respectively.

COPY NUMBER VARIATION IN A HOSPITALBASED COHORT OF CHILDREN WITH EPILEPSY

(33)

525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp 525699-L-sub01-bw-Vlaskamp Processed on: 29-10-2018 Processed on: 29-10-2018 Processed on: 29-10-2018

Processed on: 29-10-2018 PDF page: 32PDF page: 32PDF page: 32PDF page: 32

RESULTS

Characteristics of the study cohort

In 226 (17%) children, microarray analysis was performed in the context of their diagnostic work-up. Their phenotypic characteristics are summarized in Table 1. All children had a definite diagnosis of epilepsy, except for one who had a single febrile status epilepticus.

Children with epilepsy who underwent microarray analysis had significantly more developmental problems (82% vs 25%, p < 0.001), facial dysmorphisms (49% vs 8%, p < 0.001) or behavioral problems (41% vs 13%, p < 0.001) when compared to children in our database who did not undergo microarray analysis. To reduce bias, our comparisons were limited to children with epilepsy onset after December 31, 2005, when microarray analysis was introduced in our center (n = 158 for children with array; n = 271 for children without array or another identified genetic cause). The presence of a positive family history for epilepsy, known in 141/158 children who did and in 206/271 children who did not undergo microarray, did not differ significantly (33% vs 30%, p = 0.56) between the two groups.

ϭ͕ϵϴϮEsƐ ŝŶϮϮϲĐŚŝůĚƌĞŶ ϭϴϰEsƐǁŝƚŚŶŽŐĞŶĞƐŝŶϭϯϬĐŚŝůĚƌĞŶ ϰϯϳEsƐǁŝƚŚŶŽƉƌŽƚĞŝŶĐŽĚŝŶŐŐĞŶĞƐŝŶϭϵϯĐŚŝůĚƌĞŶ ϭ͕ϯϲϭŐĞŶĞͲĐŽŵƉƌŝƐŝŶŐ EsƐ ŝŶϮϮϰĐŚŝůĚƌĞŶ ϰϬϴEsƐ ŝŶϭϴϭĐŚŝůĚƌĞŶ ϵϱϯEsƐǁŝƚŚшϵϬйŽǀĞƌůĂƉŝŶшϭйŽĨŚĞĂůƚŚLJ ĐŽŶƚƌŽůƐ EsƐǁŝƚŚшϵϬйŽǀĞƌůĂƉŝŶфϭйŚĞĂůƚŚLJĐŽŶƚƌŽůƐ ϯϮϴEsƐŝŶϭϳϬĐŚŝůĚƌĞŶ EsƐǁŝƚŚфϵϬйŽǀĞƌůĂƉŝŶŚĞĂůƚŚLJĐŽŶƚƌŽůƐ ϴϬEsƐŝŶϱϯĐŚŝůĚƌĞŶ <ŶŽǁŶĐůŝŶŝĐĂůůLJƌĞůĞǀĂŶƚEsƐ ϭϱEsƐŽĨϭϭƌĞŐŝŽŶƐŝŶϭϰĐŚŝůĚƌĞŶ <ŶŽǁŶĐůŝŶŝĐĂůůLJƌĞůĞǀĂŶƚEsƐ ϭϬEsƐŽĨϲƌĞŐŝŽŶƐŝŶϭϬĐŚŝůĚƌĞŶ EŽǀĞůEsƐ ϱEsƐŽĨϰƌĞŐŝŽŶƐŝŶϱĐŚŝůĚƌĞŶ EŽǀĞůEsƐ;ϭϬdžŵŽƌĞĨƌĞƋƵĞŶƚͿ ϭϲEsƐŽĨϴƌĞŐŝŽŶƐŝŶϭϲĐŚŝůĚƌĞŶ ϯϬϮEsƐŽĨƵŶŬŶŽǁŶƐŝŐŶŝĨŝĐĂŶĐĞŝŶϭϲϭĐŚŝůĚƌĞŶ ϲϬEsƐŽĨƵŶŬŶŽǁŶ ƐŝŐŶŝĨŝĐĂŶĐĞŝŶϯϵĐŚŝůĚƌĞŶ

Figure 1: Flowchart for evaluating Copy Number Variants (CNVs) in our hospital-based cohort of

Referenties

GERELATEERDE DOCUMENTEN

We found significant differences for other phenotypes only between individuals with mis TMD+Linker and those with mis ATD+LBD or null variants, but not between those with mis ATD+LBD

The pedigrees show striking inter- and intrafamilial phenotypic heterogeneity of features including a spectrum of severity for epilepsy, intellectual disability, behavioral

Recently, sequence variants in the STX1B gene (MIM 601485) have been shown to cause a broad spectrum of fever-associated epilepsy syndromes, ranging from simple febrile seizures

To increase the validity of any observed differences, we compared the phenotypes of patients with a 16p11.2 deletion in our cohort (n=33) with those of patients with a

Our case illustrates the possible positive effect of sodium channel blockers in neonatal-infantile seizures associated with a SCN2A mutation. A precise family history and

First, we showed that genetic services for epilepsies in childhood not only have important diagnostic value, but that patients and parents also show an increased empowerment

In hoofdstuk 7 presenteren wij de casus van een 18-jarige jongen met epilepsie met een specifiek aanvalstype (myoclone-atone aanvallen) op basis van een deletie op chromosoom

Jullie hebben mij doen beseffen hoe leuk en waardevol een goede samenwerking en communicatie tussen de verschillende vakgebieden is, niet alleen voor wetenschappelijk onderzoek