veldhoven - nvhgthe added value of long-read amplicon sequencing for clinical applications 12:15...

90
SYMPOSIUM 2019 19 & 20 September VELDHOVEN ANNUAL

Upload: others

Post on 24-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

SYMPOSIUM 2019 19 & 20 September

VELDHOVEN

ANNUAL

Page 2: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

1

Dutch Society for Human Genetics (www.nvhg-nav.nl)

The NVHG encompasses two professional associations: the Dutch Association

of Clinical Geneticists (Vereniging Klinische Genetica Nederland, VKGN), which

unites medical specialists in clinical genetics, the Association of Clinical

Genetic Diagnostic Laboratories (Vereniging Klinisch Genetische

Laboratoriumdiagnostiek, VKGL) and the NACGG (Nederlandse Associatie

voor Community Genetics en Public Health Genomics) which unites

recognised clinical genetics laboratory specialists.

Belgian Society for Human Genetics (www.beshg.be)

The BeSHG represents the community of Belgian geneticists towards the other

national and international Societies of Genetics, and provides a discussion

forum on scientific, professional, social and ethical issues linked to the practice

of human genetics.

Page 3: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

2

Message from Dr. Mieke van Haelst,

president NVHG and Prof. Dr. Gert Matthijs,

president BeSHG

Dear colleagues,

Welcome to the second joint meeting of the Belgian and Dutch Society for Human Genetics.

It is a pleasure to welcome you in Veldhoven!

Both societies are characterized by a genuine interest in research in human genetics in combination

with a strong clinical and societal commitment. Implementation of the scientific results to improve

human health is an everyday goal for the majority of genetic researchers present at this meeting.

Many of us have combine research and clinical activities and obligations, and our PhD students and

post-docs are often implicated in research that stems from clinical observations or deals with

patients’ material.

Human genetics is a broad field, with a myriad of research opportunities and clinical applications.

Our professional societies play a role in promoting scientific research in genetics and genomics, in

its broadest sense. A lot is going on, and clearly, by your attendance, you witness that you wish to

be part of these great times for human genetics.

We are pleased to offer you a star-studded line-up of plenary speakers covering exiting

developments in our field. John van der Oost will teach us about CRISPR-Cas, and guide us

through a plethora of applications that have changed the world of genetic research and are finding

their way to clinical application. Douglas Easton is a global leader in breast cancer genetics

research and will focus on the use of polygenic risk score (PRS) in daily clinical practice. Michel

Georges is a well-known leader in the field of animal genetics, but his work also reaches into the

realms of human genetics, where he made important contributions to better understand GWAS loci

Page 4: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

3

in Inflammatory Bowel Disease. Monogenic diseases – or what we believe are monogenic diseases-

are discussed by Elfride De Baere who studies inherited retinal diseases and will show how

integrated omics lead to better diagnosis and therapeutic applications for specific types of

blindness. Joris Veltman, whom we know from pioneering novel genomic technologies, will focus

on clinical outcomes that provide more insight into the success of assisted reproductive

technologies, as well as the health of future offspring. Malte Spielmann studies genetics from yet

another angle: structural variation in 3D. In his lecture, he will show us how the field is revolutionized

with the study of genomes at the single cell level. Lisanne Vervoort is our youngest invited

speaker. She will bring a wonderful story about the optical mapping of structural variation. Finally,

we are very happy to have Peter de Knijff to entertain us about what he has learned during his 25

year journey in forensic genetic research.

The program also focusses on the challenges we all face. In the interactive panel-discussion on

‘Hurdles for genetic research – what can we do about it?’ we will collectively explore specific issues

that either affect or impede genetic research and clinical services, including diagnostics. We are all

familiar with GDPR (in Dutch: AVG), ethical review procedures, and FAIR data, but really how does

it affect our work? We look forward to interacting with the audience and learn also from your

experiences.

Finally, we are proud to announce that Danielle Posthuma has been awarded the Lodewijk

Sandkuijl price for her ground-breaking work in Complex Genetics and she will enlighten us about

her work in her lecture “From GWAS to Function”.

We sincerely thank all the participants for their contribution. From the abstracts, the Scientific

Committee has been able to select high quality presentations. Unfortunately we have not been able

to offer all abstract submitters a slot in the program, so please also visit the posters.

We also thank the sponsors, not only for their essential and generous financial contribution for the

organization of the meeting, but also because they join us to present their latest innovations. This

interaction between users and providers is very valuable, for all, but mostly for young people and

trainees in the labs.

Importantly, we wish to sincerely thank all the members that actively engage, in the organization

and function of our Societies. We are happy we can count on you to make our societies thrive! We

hope that this year’s meeting will bring you many new ideas and inspiring new collaborations.

Mieke van Haelst, president NVHG

Gert Matthijs, president BeSHG

Page 5: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

4

General information

Venue

Hotel NH Eindhoven Conference Centre Koningshof

Locht 117, 5504 RM Veldhoven

Eindhoven - Nederland

Tel.: +31 40 253 7475

www.nh-hotels.nl/hotel/nh-eindhoven-conference-centre-koningshof

[email protected]

Registration

Holland Foyer: open on Thursday September 19, 2019 09:30 - 10:30hrs

Reception and catering

Holland foyer

Networking event

Brabantzaal

Abstracts

Abstracts guest speakers G 01 to G 08

Abstracts talks: T 01 to T 16

Abstracts posters : P 01 to P 39

Abstracts T 01 - T 16 and P 01 - P 31 are available as download through the NVHG website

For abstracts of the NACGG, VKGN and VKGL sessions we refer to these societies and the NVHG

website

Posters

Poster boards have a size of 200 cm (height) and 100 cm (width)

Please put up your poster immediately after arrival. Do not forget to remove it at the end of the

meeting

Language

The official language of the annual meeting will be English

Accreditation

Accreditation forms are available at the registration desk (GAIA ID number: 370535)

Photos

Photographs will be taken during the meeting which can be used by the NVHG for PR purposes.

Anybody objecting should list their name on the form available at the NVHG registration desk.

Badges

You are requested to hand in your badge at the end of the symposium

Presentations

You are requested to timely hand in an USB stick with your presentation to the chairperson of

your symposium session

Page 6: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

5

Organization

Scientific organization

NVHG board Dr. Mieke van Haelst (President)

Prof. dr. Johan den Dunnen (Secretary)

Dr. ir. Aimée Paulussen (Treasurer)

Dr. Terry Vrijenhoek

Dr. Lidewij Henneman

Dr. Roland Kuiper

Prof. dr. Richard Sinke

Prof. dr. André Uitterlinden

Dr. Lisenka Vissers

Dr. Gijs Santen

BeSHG board

Prof. dr. Gert Matthijs (President)

Prof. Dr. Nisha Limaye (Secretary)

Dr. Lut Van Laer (Treasurer)

Dr. Saskia Bulk

Prof. Dr. Bert Callewaert

Prof. Dr. Kathelijn Keymolen

Dr. Damien Lederer

Dr. Julie Soblet

Administrative organization

Julia Lopez Hernandez ([email protected])

Henny Schurmann ([email protected])

www.nvhg-nav.n

Page 7: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

6

Sponsors

We are very grateful for the

financial support from

Simons Fund Foundation

Page 8: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

7

We thank the following companies for their support:

Golden Sponsors

Agilent Technologies

www.agilent.com

Moon

www.diploid.com/moon

Illumina

www.illumina.com

Takara Bio

www.takarabio.com

Sophia Genetics SA

www.sophiagenetics.com PerkinElmer

www.perkinelmer.com/lifesciences

Page 9: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

8

Silver Sponsors

Sanofi

www.sanofigenzyme.nl

Nimagen B.V.

www.nimagen.com

Pacific Biosciences

www.pacb.com

GE Healthcare

www3.gehealthcare.nl

BIOKÉ B.V.

www.bioke.com

BGI Global Genomic Services

www.bgi.com

Twist Bioscience

www.twistbioscience.com

Sanbio

www.sanbio.nl

Page 10: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

9

Westburg

www.westburg.eu

Hettich

www.hettich.com

Regular Sponsors

Novogene

en.novogene.com

Bionano Genomics

bionanogenomics.com

Alnylan

www.alnylam.com

Thermo Fisher

www.fishersci.nl

Macrogen

dna.macrogen.com

Page 11: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

10

Program

Time Thursday 19th September

09:30 - 10:30 Registration

Room: Holland Foyer

10:30 - 12:40 Opening & Plenary session 1 Room: Auditorium Chair: Mieke van Haelst (NVHG) / Saskia Bulk (BeSHG)

10:30

Opening by Dr. Mieke van Haelst president NVHG / Dr. Saskia Bulk board member BeSHG

10:35 Prof. Dr. John van der Oost (Wageningen, NL)

CRISPR-Cas – from biology to applications

11:15

DNA Day Contest Award

11:20 Prof. Dr. Douglas Easton (Cambridge, UK)

Predicting Breast Cancer Risk using Rare and Common Variants

12:00 Lodewijk Sandkuijl Lecture: Danielle Posthuma (Amsterdam, NL) From GWAS to Function

12:40 - 14:00 Lunch, room: Holland Foyer

13:00 Business/private meetings VKGN

Room: Parkzaal

13:00 Business/private meetings VKGL

Room: Auditorium

14:00 - 16:00 Plenary session 2

Room: Auditorium Chair: Terry Vrijenhoek (NVHG) / Bert Callewaert (BeSHG)

Hurdles for genetic research - what can we do about them? Panel session to explore how genetic research could be further facilitated

14:00 Introduction session

14:15 Panel presentation and hurdle identification

15:00 Power Break

15:15 Plenary discussion - how to overcome the hurdles

15:45 Conclusion in images

Page 12: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

11

16:00 - 17:30 Posters presentation with coffee & tea Room: 65 / Holland Foyer

17:30 - 18:30 Plenary session 3

Room: Auditorium Chair: Johan den Dunnen (NVHG)

17:30 Prof. Dr. Peter de Knijff (Leiden, NL)

A result is only a result if it results in a result

18:30 - 21:00 Social Evening Program

Room: Brabantzaal

18:30 Pre-dinner drinks

19:00 Dinner

21:00 Networking event

Time Friday 20th September

08:30 - 09:30 Registration

Room: Holland Foyer

08:45 - 10:05 Plenary session 4 Room: Auditorium Chair: André Uitterlinden (NVHG) / Saskia Bulk (BeSHG)

8:45 Prof. Dr. Michel Georges (Liege, BE) Identifying causative variants and genes in GWAS-identified IBD risk loci

9:25 Prof. Dr. Elfride De Baere (Ghent, BE)

Integrated omics to accelerate diagnosis and therapy in inherited retinal diseases causing blindness

10:05 - 11:15 Parallel sessions A/B

A. Fertility and Pregnancy Room: Auditorium Chair: Lidewij Henneman (NVHG) / Björn Menten (BeSHG)

10:15 Martine De Rycke

Preimplantation Genetic Testing with HLA matching: from counselling to birth and beyond

10:30 Karuna van der Meij

TRIDENT-2: National Implementation of Genome-Wide Non-Invasive Prenatal Testing as a First-Tier Screening Test in the Netherlands

Page 13: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

12

10:45 Kris Van Den Bogaert

The landscape of pathogenic copy number variations in healthy, reproducing females

11:00 Masoud Zamani Esteki

In vitro fertilization does not increase the incidence of de novo copy number alterations in fetal and placental lineages

B. Personalized Genomics Room: Parkzaal Chair: Gijs Santen (NVHG) / Damien Lederer (BeSHG)

10:15 Inge Lakeman

Longitudinal assessment of the 313-SNP based Polygenic Risk Score for breast cancer risk prediction in a Dutch prospective cohort

10:30 Helen Roessler

Towards the treatment of Cantú syndrome

10:45 Remco Hack

NOTCH3 cysteine altering variants and their phenotypes in 92,456 whole exome sequenced participants of the Geisinger DiscovEHR initiative

11:00 Eline van Hugte

Towards personalized treatment of genetically classified refractory epilepsies using human induced pluripotent stem Cells (hIPSCs) as an ex-vivo tool

11:15 - 11:45 Poster viewing with coffee & tea Room 65 / Holland Foyer

11:45 - 12:45 Parallel sessions C/D

C. Diagnostic Opportunities Room: Auditorium Chair: Richard Sinke (NVHG) / Kris Van Den Bogaert (BeSHG)

11:45 Jeroen van Rooij

Genotyping On ALL patients (GOALL); clinical implementation of high-throughput genotyping arrays.

12:00 Kornelia Neveling

The added value of long-read amplicon sequencing for clinical applications

12:15 Peter Henneman

Reliable application of DNA-methylation signatures in genetic diagnostic testing

12:30 Helga Westers

What if we would use a diagnostic multi-cancer gene panel for opportunistic screening? A study in 2,090 Dutch familial cancer patients

D. Biological Insight into Rare Disease Room: Parkzaal Chair: Lisenka Vissers (NVHG) / Arvid Suls (BeSHG)

11:45 Margot Reijnders

Neurodevelopmental disorders: a next generation

12:00 Lore Pottie

Page 14: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

13

Transcriptome and protein analysis highlight the endosomal pathway in disease pathogenesis of metabolic CL syndrome

12:15 Eva D'haene

Noncoding structural variants disrupt the regulatory architecture of Rett genes

12:30 Daphne Smits

Loss of neutral sphingomyelinase-3 (SMPD4) links neurodevelopmental disorders to cell cycle and nuclear envelope anomalies

12:45 - 14:00

Lunch and poster viewing

13:00

NVHG General Members' Meeting Room: Auditorium

14:00 - 16:30 Plenary session 5

Room: Auditorium Chair: Roland Kuiper (NVHG) / Bert Callewaert (BeSHG)

14:15 Prof. Dr. Joris Veltman (Newcastle, UK)

De novo mutations affecting male reproductive health

14:55 PD. Dr. Malte Spielmann (Berlin, DE)

Human genetics at single cell resolution

15:35 Winner Genetics Retreat 2019: Lisanne Vervoort (Leuven, BE)

Optical Mapping of 22q11.2 Low Copy Repeats reveals structural hypervariability

16:00 Awards 2019

Chair: Mieke van Haelst (NVHG) / Bert Callewaert (BeSHG)

Young Investigator Award (for best thesis) Annual Award (for best oral presentation) Poster Award (for best poster) Puzzle prize

16:15 Closing Remarks

16:30 Meeting ends

Page 15: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

14

Floor plan

Page 16: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

15

Special Guest speaker

Lodewijk Sandkuijl Lecture

From GWAS to Function

Danielle Posthuma, Professor

Complex Trait Genetics, Vrije

Universiteit Amsterdam

Thursday September 19, 2019

Time: 12.00 – 12.40

Room: Auditorium

Genome-wide association studies (GWAS) have successfully identified many novel loci for

neuropsychiatric traits. At the same time the results of GWAS showed that these traits are highly

polygenic, mostly influenced by large numbers of weakly associated variants. Interpreting such

polygenic results is challenging. Recent large-scale initiatives, such as those from the Allen Brain

Institute and the PyschEncode consortium provide fine-scaled atlases of functional genetic

elements at cellular level. This novel information can be used to interpret results from GWAS studies

and facilitate biological understanding of complex traits.

We developed two tools that aid in biological interpretation of GWAS findings and use

biological information at cellular resolution: FUMA, for functional annotation and gene

prioritization, and MAGMA, for gene-based and gene-set analysis. I will discuss recent GWAS

findings for several brain related traits, the biological implications of these findings and possible

routes for functional follow-up, ultimately leading to a mechanistic understanding of complex traits.

Email: [email protected]

Page 17: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

16

Special Guest speaker

A result is only a result if

it results in a result Peter de Knijff, Professor

Population and

Evolutionary Genetics,

Leiden University

Thursday September 19, 2019

Time: 17.30 – 18.30

Room: Auditorium

Peter de Knijff is professor of population and evolutionary genetics at the department of

human genetics of the LUMC. There, he is also, since 1994, head of the forensic laboratory for DNA

research (FLDO).

The research of Peter de Knijff is besides population genetics, it is also targeted at developing

new forensic genetic diagnostic research tools. He was, with two German colleagues, responsible

for the introduction and widespread use of polymorphic Y-chromosome loci in forensics. More

recently, he pioneered in the forensic use of massively parallel sequencing. He started, as research

technician, his scientific career at Leiden University in January 1980. There he obtained his PhD on

June 4, 1992. He was appointed as full professor in 2005. His inaugural lecture was spoken on June

23 2006 and was titled "Crying with the wolves”.

Email: [email protected]

Page 18: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

17

G 01

Abstracts Guest Speakers (G)

CRISPR-Cas – from biology to

applications

John van der Oost

Laboratory of Microbiology, Wageningen University

Ten years ago major discoveries at the level of comparative genomics [1], molecular microbiology

[2] and biochemistry [3] have revealed that CRISPR-Cas is a heritable adaptive immune system of

bacteria and archaea, based on RNA-guided DNA interference. These key findings have initiated a

revolution, including exploration the natural diversity of CRISPR-Cas classes and types [4],

characterizing structure-function relations of CRISPR-associated proteins and RNA guides [5], and

development of a range of applications in biotechnology and medicine [6,7]. Apart from comparing

multi-subunit Cascade-like systems (class-1) and single-protein Cas9/Cas12-like systems (class-2)

systems, both natural and synthetic features will be discussed that are relevant for applications [8,9].

[1] Mojica et al. (2005) J Mol Evol 60, 174-182

[2] Barrangou et al. (2007) Science 315, 1709-1712

[3] Brouns et al. (2008) Science 321, 960-964

[4] Makarova et al. (2015) Nat Rev Microbiol 13, 722-736

[5] Mohanraju et al. (2016) Science 353, aad5147/1-12

[6] Charpentier & Doudna (2013) Nature 495, 50-51

[7] Hsu et al. (2014) Cell 157, 1262-1278

[8] Zetsche et al. (2017) Nat Biotechnol 35, 31-34

[9] Wu et al. (2018) Nat Chem Biol. 14, 642-651

Email: [email protected]

Page 19: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

18

G 02

From GWAS to Function

Prof. dr. Danielle Posthuma

Vu university, dept complex trait genetics & vumc dept clinical genetics

Genome-wide association studies (GWAS) have successfully identified many novel loci for

neuropsychiatric traits. At the same time the results of GWAS showed that these traits are highly

polygenic, mostly influenced by large numbers of weakly associated variants. Interpreting such

polygenic results is challenging. Recent large-scale initiatives, such as those from the Allen Brain

Institute and the PyschEncode consortium provide fine-scaled atlases of functional genetic

elements at cellular level. This novel information can be used to interpret results from GWAS studies

and facilitate biological understanding of complex traits.

We developed two tools that aid in biological interpretation of GWAS findings and use biological

information at cellular resolution: FUMA, for functional annotation and gene prioritization, and

MAGMA, for gene-based and gene-set analysis. I will discuss recent GWAS findings for several brain

related traits, the biological implications of these findings and possible routes for functional follow-

up, ultimately leading to a mechanistic understanding of complex traits.

Email: [email protected]

Page 20: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

19

G 03

Predicting Breast Cancer Risk using

Rare and Common Variants

Prof.dr. Douglas Easton

Centre for Cancer Genetic Epidemiology, Department of Public Health and

Primary Care and Department of Oncology, University of Cambridge,

Cambridge, UK.

The risks of breast cancer are associated with germline genetic variants, lifestyle or hormonal factors

and breast density. Genetic factors include rare coding variants in at least ten susceptibility genes,

and common polymorphisms (mostly SNPs), identified through genome-wide association studies,

that confer more moderate risks1,2. The associations for common variants appear to be largely

independent and can be summarised by a polygenic risk score (PRS). A recently published PRS,

comprising 313 SNPs, confers an estimated a relative risk per 1 standard deviation of

1.61(95%CI:1.57-1.65); women in the top 1% of the risk distribution have an approximately 4-fold

risk of ER-positive disease and 3-fold risk of ER-negative disease3. The risks also appear largely

independent (on a log-scale) of the risks of associated with lifestyle risk factors, breast density and,

approximately, high/moderate risk gene variants. The BOADICEA model, which incorporates all

these components, is being made available for clinical use4.

Despite their now widespread use, the evidence for association with cancer for coding variants in

many genes on genetic testing panels is often weak, and many of the underlying risk estimates are

very imprecise5. However, recent results from the large population studies are providing more

reliable estimates for both truncating and missense variants. The current version of BOADICEA

incorporates five genes (BRCA1, BRCA2, ATM, CHEK2, PALB2) but recent results from the Breast

Cancer Association Consortium BRIDGES project indicate that several other genes, including

BARD1, RAD51C and RAD51D, could now be reliably included.

1. Michailidou, K. et al. Association analysis identifies 65 new breast cancer risk loci. Nature 551, 92-

94, doi:10.1038/nature24284 (2017).

2. Milne, R. L. et al. Identification of ten variants associated with risk of estrogen-receptor-negative

breast cancer. Nat Genet, doi:10.1038/ng.3785 (2017).

3. Mavaddat, N. et al. Polygenic Risk Scores for Prediction of Breast Cancer and Breast Cancer

Subtypes. Am J Hum Genet 104, 21-34, doi:10.1016/j.ajhg.2018.11.002 (2019).

4. Lee, A. et al. BOADICEA: a comprehensive breast cancer risk prediction model incorporating

genetic and nongenetic risk factors. Genet Med, doi:10.1038/s41436-018-0406-9 (2019).

5. Easton, D. F. et al. Gene-panel sequencing and the prediction of breast-cancer risk. N Engl J Med

372, 2243-2257, doi:10.1056/NEJMsr1501341 (2015).

Email: [email protected]

Page 21: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

20

G 03

A result is only a result if it results in

a result

Prof.dr. Peter de Knijff

Department of Human Genetics, Leiden University Medical Center, The

Netherlands.

Performing forensic genetic research essentially boils down to catching criminals and secure a

conviction by an independent Court. During this “hunt” for justice, the forensic DNA expert must

face a complex landscape of adversaries who will have their own (often ill-informed) opinion about

your work and its relevance. Furthermore, despite its extremely high societal relevance, forensic

genetic research is very low - if at all - on the agenda of important scientific organizations such as

the KNAW (not a single forensic-sciences member), NWO (no funding possibilities), de Nationale

Wetenschapsagenda (no interest), and editorial boards of high ranking journals such as Nature,

Science or Cell (unless something scandalous happened). Even, one of the former Deans of my own

organization told me very early in my career “when people see our logo they must think: we are

here to treat patients, not to catch criminals, keep that in mind”. It is in this, sometimes

unhospitable, landscape that I have been travelling the past 25 Years. I will use one recent criminal

case to illustrate how, by being patient, persistent, and creative, such journeys can still be very

rewarding.

Email: [email protected]

Page 22: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

21

G 04

Identifying causative variants and

genes in GWAS-identified IBD risk

loci Prof.dr. Michel Georges

Unit of Animal Genomics, GIGA Institute, University of Liège, Belgium.

More than 250 IBD risk loci have been identified by GWAS accounting for > 50 % of inherited risk.

GWAS identified risk loci typically span 250Kb encompassing thousands of variants and ~5 genes

(range: 0 -100). Identifying the causative variants and genes amongst those is essential to reap the

full benefits of GWAS towards precision medicine and intelligent drug design. For approximately

10 IBD risk loci the causative variants are coding hence allowing effective identification of causative

variants and genes “in one shot”. However, the majority of risk variants underlying common

complex disease are regulatory. Identifying these and the genes they control requires the

application of sophisticated fine-mapping methods and integration of expression QTL information.

The effectiveness of these approaches is limited by LD-structure and tissue heterogeneity,

respectively. We will describe our results in applying these methods as well as our attempts to

overcome their limitations.

Email: [email protected]

Page 23: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

22

G 05

Integrated omics to accelerate

diagnosis and therapy in inherited

retinal diseases causing blindness Prof.dr. Elfride De Baere

Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent,

Belgium

Integrated genomics and transcriptomics reveal an increasing number of non-coding mutations in

Mendelian disorders including inherited retinal diseases (IRD), representing a major cause of early-

onset blindness in 2 million people worldwide. Of these the majority are deep-intronic splicing

mutations, typically leading to pseudo-exon inclusion and amenable to antisense oligonucleotide-

mediated rescue, already implemented in the clinic. Non-coding mutations in cis-regulatory

elements (CREs) are more scarce. Paradigms for regulatory variants are IRD subtypes with

recognizable phenotypes and without or - in case of autosomal recessive disease - monoallelic

coding variants in the presumed disease genes. Examples are Stargardt disease (ABCA4),

choroideremia (CHM), Leber congenital amaurosis 9 (NMNAT1) and North Carolina Macular

Dystrophy (PRDM13 and IRX1). Particularly interesting CREs are ultraconserved non-coding

elements (UCNEs) that are clustered in genomic regulatory blocks (GRBs) and that may act as

distant enhancers. A search for IRD genes in GRBs revealed genes in 12 GRBs, containing 306

UCNEs. In three of these genes (CHM, PRDM13, USH2A), non-coding mutations have already been

reported. In a set of genes under control of the retinal transcription factor CRX, 138 genes were

found in GRBs that harbor 3,424 UCNEs. Only four of these are known IRD genes. These GRBs

strongly coincide with topologically associating domains or TADs, playing key roles in gene

regulation and determined by chromosome conformation capture techniques. Integration of

UCNEs with epigenomic datasets generated in relevant cell types contribute to functional genome

annotations in retina and accelerate diagnosis and therapy in IRD.

Email: [email protected]

Page 24: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

23

G 06

De novo mutations affecting male

reproductive health Prof.dr. Joris A Veltman

Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne,

United Kingdom

Department of Human Genetics, Radboud University Medical Center,

Nijmegen, The Netherlands

During spermatogenesis and oogenesis the 6 billion letters of our diploid genome need to be

accurately copied in order for the gametes to successfully pass on our genetic information. By

studying the genomes of children and their parents, we now know that each human is born with

50-100 de novo mutations. These de novo mutations, by affecting important genes during

development, can result in severe neurodevelopmental disorders such as intellectual disability or

epilepsy. In this presentation, I will discuss our new research focused on studying the role of de

novo mutations in severe forms of male infertility. There is very little known about the role of

genetics of male infertility, unfortunately genomics approaches are not routinely applied in research

and/or diagnostics. I will present our first studies in which we studied the DNA of patients with

azoospermia or severe oligospermia and compared this to that of their unaffected parents. We

identified not only de novo point mutations but also de novo deletions in their genomes,

determined the parent of origin and are currently replicating this work as well as performing

functional studies within the framework of the International Male Infertility Genomics Consortium

(www.IMIGC.org). This work is aimed at furthering our understanding of the genetic causes of male

infertility with the goal to improve diagnostics, provide more insight into the success of assisted

reproductive technologies, as well as the health of future offspring.

Email: [email protected]

Page 25: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

24

G 07

Human genetics at single cell

resolution

PD.dr. Malte Spielmann

Max Planck Institute for Molecular Genetics, Germany

Mammalian embryonic development is an astonishing process. In mice most major internal and

external organs develop within a short window of time, termed organogenesis. The key regulators

of developmental defects can be studied during this critical window, but conventional approaches

lack the throughput and resolution to obtain a global view of the molecular states and trajectories

of a rapidly diversifying and expanding number of cell types.

We have recently developed a three-step combinatorial barcoding method to profile single-cell

transcriptomes ('sci-RNA-seq3') without requiringphysical isolation of each cell. We have used this

new method to profile whole mouse embryos staged between 9.5 and 13.5 days of gestation

withsci-RNA-seq3, and created a transcriptional atlas of mouse organogenesis at single cell

resolution. We identify hundreds of expanding, contracting and transient cell types, many of which

are only detected because of the depth of cellular coverage obtained here, and define the

corresponding sets of cell type-specific marker genes, several of which we validate by whole mount

in situ hybridization. We also delineate and annotate 56 single cell developmental trajectories of

mouse organogenesis. We explore the dynamics of proliferation and gene expression within cell

types over time, including focused analyses of the apical ectodermal ridge, limb mesenchyme and

skeletal muscle.

Our single cell atlas of the development of wild-type mice represents a first step towards

understanding pleiotropic developmental disorders at the organismal scale. We are currently

performing detailed single-cell investigations in mutant mice and patients samples of subtle roles

for genes and regulatory sequences involved in developmental defects.

Email: [email protected]

Page 26: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

25

G 08

Optical Mapping of 22q11.2 Low

Copy Repeats reveals structural

hypervariability Lisanne Vervoort1, Wolfram Demaerel1, Yulia Mostovoy2, Feyza

Yilmaz3,4, Steven Pastor5,6, Matthew S. Hestand7,8, Ann Swillen1, Elfi

Vergaelen1, Elizabeth A. Geiger4, Curtis R. Coughlin4, Stephen K.

Chow2, Donna McDonald-McGinn5,6, Bernice E. Morrow9, Pui-Yan

Kwok2, Ming Xiao10, Beverly S. Emmanuel5,6, Tamim H. Shaikh4, Joris R. Vermeesch1

1Department of Human Genetics, KU Leuven, Leuven, Belgium, 2Cardiovascular Research Institute, UCSF School

of Medicine, San Francisco, CA, United States, 3Department of Integrative Biology, University of Colorado

Denver, Denver, CO, United States, 4Department of Pediatrics, Section of Clinical Genetics and Metabolism,

University of Colorado Denver, Aurora, CO, United States, 5Division of Human Genetics, Children's Hospital of

Philadelphia, Philadelphia, PA, United States, 6Department of Pediatrics, Perelman School of Medicine at the

University of Pennsylvania, Philadelphia, PA, United States, 7Division of Human Genetics, Cincinnati Children's

Hospital Medical Center, Cincinnati, OH, United States, 8Department of Human Genetics, Cincinnati Children's

Hospital Medical Center, Cincinnati, OH, United States, 9Department of Genetics, Albert Einstein College of

Medicine, Bronx, NY, United States, 10School of Biomedical Engineering, Drexel University, Philadelphia, PA,

United States.

Introduction: The 22q11.2 locus is structurally one of the most complex areas of the human genome

due to the presence of low copy repeats (LCR22s). Despite the newest sequencing technologies,

the human reference genome hg38 still comprises three unresolved sequence gaps in LCR22-A.

The recurrent deletion/duplication breakpoints of 22q11.2 deletion syndrome (22q11.2DS) are

embedded within these repeats, but the exact location remains unclarified.

Materials and Methods: We performed an LCR22 de novo assembly using fiber-FISH. Long DNA

molecules were extracted from cells, stretched onto coverslips, and hybridized with LCR22 specific

probes. Following manual signal screening, alleles were de novo assembled by tiling fibers based

on matching colors and distances between the probes.

Results: LCR22 haplotyping in 24 individuals uncovered the presence of 26 different alleles for

LCR22-A. Subunits cluster in larger substructures, which vary in orientation, copy number, and

presence. The rearranged alleles of 22q11.2DS patients were mapped in nine families and

differences in the location of the deletion breakpoints were identified. In addition, LCR22-A alleles

were assembled for the Great Apes to uncover insights in the evolutionary context.

Conclusions: For the first time, the overall architecture of the 22q11.2 locus was successfully

unraveled. The LCR22 hypervariability implicates interindividual gene dosage differences. As a

consequence, copy number variations could influence gene expression profiles. Additionally,

mapped rearrangement breakpoints vary among patients. We hypothesize this variability could

provide a genetic explanation for some of the phenotypic variability characterizing the 22q11.2DS.

Email: [email protected]

Page 27: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

26

Abstracts Talks (T)

No. Name Titel E-mail

A. Fertility and

Pregnancy

T 01 Martine De Rycke Preimplantation Genetic Testing with HLA

matching: from counselling to birth and beyond

[email protected]

T 02 Karuna van der

Meij*

TRIDENT-2: National Implementation of

Genome-Wide Non-Invasive Prenatal Testing as

a First-Tier Screening Test in the Netherlands

[email protected]

T 03 Kris Van Den

Bogaert

The landscape of pathogenic copy number

variations in healthy, reproducing females

[email protected]

T 04 Masoud Zamani

Esteki

In vitro fertilization does not increase the

incidence of de novo copy number alterations

in fetal and placental lineages

[email protected]

B. Personalized Genomics

T 05 Inge Lakeman* Longitudinal assessment of the 313-SNP based

Polygenic Risk Score for breast cancer risk

prediction in a Dutch prospective cohort

[email protected]

T 06 Helen Roessler* Towards the treatment of Cantú syndrome [email protected]

T 07 Remco Hack* NOTCH3 cysteine altering variants and their

phenotypes in 92,456 whole exome sequenced

participants of the Geisinger DiscovEHR

initiative

[email protected]

T 08 Eline van Hugte* Towards personalized treatment of genetically

classified refractory epilepsies using human

induced pluripotent stem Cells (hIPSCs) as an

ex-vivo tool

[email protected]

C. Diagnostic Opportunities

T 09 Jeroen van Rooij* Genotyping On ALL patients (GOALL); clinical

implementation of high-throughput

genotyping arrays.

[email protected]

T 10 Kornelia Neveling The added value of long-read amplicon

sequencing for clinical applications

[email protected]

l

T 11 Peter Henneman Reliable application of DNA-methylation

signatures in genetic diagnostic testing

[email protected]

T 12 Helga Westers What if we would use a diagnostic multi-cancer

gene panel for opportunistic screening? A study

in 2,090 Dutch familial cancer patients

[email protected]

D. Biological Insight into Rare Disease

T 13 Margot Reijnders Neurodevelopmental disorders: a next

generation

[email protected]

T 14 Lore Pottie* Transcriptome and protein analysis highlight

the endosomal pathway in disease

pathogenesis of metabolic CL syndrome

[email protected]

T 15 Eva D'haene* Noncoding structural variants disrupt the

regulatory architecture of Rett genes

[email protected]

T 16 Daphne Smits* Loss of neutral sphingomyelinase-3 (SMPD4)

links neurodevelopmental disorders to cell

cycle and nuclear envelope anomalies

[email protected]

* Eligible for Annual Award

Page 28: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

27

T 01

Preimplantation Genetic Testing with HLA matching: from

counseling to birth and beyond

M. De Rycke1, A. De Vos, F. Belva, V. Berckmoes, M. Bonduelle, A. Buysse, K. Keymolen, I. Liebaers,

J. Nekkebroeck, P. Verdyck, W. Verpoest

Centre for Medical Genetics and Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel, Laarbeeklaan 101 1090

Brussels, Belgium. Vrije Universiteit Brussel (VUB), Reproduction and Genetics, Laarbeeklaan 101, 1090 Brussels, Belgium.

Introduction Preimplantation genetic testing with HLA typing (PGT-HLA) involves HLA typing of a

single or a few cells biopsied from in vitro fertilized preimplantation embryos with the aim of

obtaining a pregnancy where the fetus is HLA compatible with an affected sibling in need of a

hematopoietic stem cell transplantation (HSCT). During PGT-M-HLA the identification of a HLA

compatible embryo is combined with the detection of mutations underlying immunodeficiencies

and hemoglobinopathies. Materials and methods This is a combined retrospective and prospective

cohort analysis of PGT-(M)-HLA procedures from 1998 until 2017 with follow-up of transplantations

until 2019. Data on patient intake and dropout, preclinical workup, cycle outcome, pregnancy, baby

and transplantation follow-up were collected from our patient files, with informed consent from

the couples. Results During the study period, 234 couples from 22 countries were invited for a

multidisciplinary intake. Psychological counselling showed that couples had high hopes on saving

their ill child. Two couples were rejected (no HLA indication) and 70 couples refrained from PGT

before or after preclinical workup (various causes), leaving 162 couples for which 414 clinical cycles

were carried out. The majority of cycles (81%) involved PGT-M-HLA (51% for sickle cell anemia).

Cleavage stage biopsy followed by single cell multiplex PCR for STR-based haplotyping was mostly

applied (98.7%). The diagnostic efficiency was high (94.8%) but genetic selection left only 16.5% of

embryos as genetically suitable for transfer. Fresh embryo transfer cycles mainly included HLA

compatible embryos (91.3%) whereas the majority of cryopreserved embryos were surplus HLA

non-compatible embryos (89.9%). Fresh and frozen-thawed embryo transfer resulted in 67 clinical

pregnancies and 74 liveborn children (52 singletons, 22 twins) of which 60 children were HLA

compatible. This yielded a live birth delivery rate of 30.3% per transfer. Information on neonatal

characteristics showed reassuring outcomes. So far, HSCT was carried out successfully for 24 out of

25 cases. The most striking findings following psychological assessment of a small group of couples

(21%) after unsuccessful PGT treatment were resilience and absence of any regrets. Conclusion Our

data show that PGT-(M)-HLA is a valuable procedure: the high complexity and limited delivery rate

are balanced by the successful HSCT outcome and the positive impact on families.

E-mail: [email protected]

Keywords: preimplantation genetic testing HLA matching hematopoietic stem cell transplantation

sibling donor

Page 29: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

28

T 02

TRIDENT-2: National Implementation of Genome-Wide

Non-Invasive Prenatal Testing as a First-Tier Screening Test

in the Netherlands

Karuna R.M. van der Meij1, Erik A. Sistermans1, Merryn V.E. Macville2, Servi J.C. Stevens2, Caroline

J. Bax3, Mireille N. Bekker4, Caterina M. Bilardo5, Elles M. J. Boon1, Marjan Boter6, Karin E.M.

Diderich6, Christine E.M. de Die-Smulders2, Leonie K. Duin7, Brigitte H. W. Faas8, Ilse Feenstra8,

Monique C. Haak9, Mariëtte J.V. Hoffer10, Nicolette S. den Hollander10, Iris H.I.M. Hollink6,

Fernanda S. Jehee6, Maarten F.C.M. Knapen11, Angelique J.A. Kooper12, Irene M. van Langen13,

Klaske D. Lichtenbelt14, Ingeborg H. Linskens5, Merel C. van Maarle12, Dick Oepkes9, Mijntje J.

Pieters15, G. Heleen Schuring-Blom14, Esther Sikkel16, Birgit Sikkema-Raddatz12, Dominique

F.C.M. Smeets8, Malgorzata I. Srebniak6, Ron F. Suijkerbuijk12, Gita M. Tan-Sindhunata1, A. Jeanine

E.M. van der Ven17, Shama L. van Zelderen-Bhola1, Lidewij Henneman1, Robert-Jan H. Galjaard6,

Diane Van Opstal6 and Marjan M. Weiss1 for the Dutch NIPT Consortium

1 Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands. 2

Department of Clinical Genetics, GROW School for Oncology and Developmental Biology, Maastricht University Medical

Center, Maastricht, The Netherlands. 3 Department of Obstetrics and Gynaecology, Amsterdam UMC, University of

Amsterdam, Amsterdam, The Netherlands. 4 Department of Obstetrics and Gynaecology, Utrecht University Medical Center,

Utrecht, The Netherlands. 5 Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam,

Amsterdam, The Netherlands. 6 Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands. 7

Department of Obstetry and Gynaecology University Medical Centre Groningen, University of Groningen, Groningen, the

Netherlands. 8 Department of Genetics, Radboud Institute for Molecular Life Sciences, Radboud university medical Center,

Nijmegen, The Netherlands. 9 Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands. 10

Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands. 11 Department of Obstetrics

and Fetal medicine, Erasmus Medical Center, Rotterdam, The Netherlands. 12 Department of Clinical Genetics, Amsterdam

UMC, University of Amsterdam, Amsterdam, The Netherlands. 13 Department of Genetics, University Medical Centre

Groningen, University of Groningen, Groningen, the Netherlands. 14 Department of Genetics, Utrecht University Medical

Center, Utrecht, The Netherlands. 15 Department of Obstetrics and Gynecology, Maastricht University Medical Center,

Maastricht, The Netherlands.

The Netherlands is the first country to perform a nationwide implementation study on non-invasive

prenatal testing (NIPT) as a first-tier test in the screening for Down-, Edwards-, and Patau syndrome.

This started April 1st 2017 as the so-called TRIDENT-2 study, licensed by the Dutch Ministry of

Health. All pregnant women are offered prenatal screening and are counseled by certified

counselors, generally midwives. A first-tier NIPT costs women € 175, comparable to the standard

first-trimester combined test (FCT) (~€ 170). Women can choose to have a targeted report of

chromosomes 21, 18, and 13 or a whole-genome report including findings other than trisomy 21,

13, 18 on the remaining autosomes, if over 10-20 Mb. The former applies a targeted filter in the

bioinformatic analysis pipeline while the latter does not. Sex chromosomes are not analyzed. In the

first year, NIPT was performed in 73,239 pregnancies (42% of all pregnancies), 7239 (4%) chose the

FCT, and 54% did not participate. The number of trisomies 21 (239, 0.33%), 18 (49, 0.07%) and 13

(55, 0.08%) reported is in line with earlier studies. The Positive Predictive Values ((PPV), 96% for

trisomy 21, 98% for trisomy 18 and 53% for trisomy 13) were higher than expected in this general

obstetric population. Overall 78% of women chose to have the findings other that trisomy 21, 18

and 13 reported. The number of additional findings was 207 (0.36%), among which other trisomies

(101, 0.18%, PPV 6%), structural chromosomal aberrations (95, 0.16%, PPV 32%) and complex

abnormal profiles indicative of maternal malignancies (11, 0.02%, PPV 64%). The implementation

of genome-wide NIPT is under debate, as the benefits of detecting other fetal chromosomal

Page 30: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

29

aberrations need to be balanced against the risks of false positives, parental anxiety and a potential

increase in (invasive) diagnostic procedures. The first year data provided here including clinical and

laboratory follow-up will fuel this debate. Furthermore, we have shown that NIPT can successfully

be embedded in a national screening program, with a single chain for prenatal care including

counseling, testing and follow-up.

E-mail: [email protected]

Keywords: prenatal screening; non-invasive prenatal testing; cell-free DNA; pregnancy; aneuploidy

Page 31: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

30

T 03

The landscape of pathogenic copy number variations in

healthy, reproducing females

Brison Nathalie, Dehaspe Luc, Melotte Cindy, Dimitriadou Eftychia, Vancoillie Leen, Breckpot Jeroen,

Peeters Hilde, Van Esch Hilde, Van Buggenhout Griet, Vogels Annick, de Ravel Thomy, Legius Eric,

Devriendt Koen, Vermeesch Joris, Van Den Bogaert Kris

Center for Human Genetics, KU Leuven and UZ Leuven, Belgium

Since July 1, 2017, non-invasive prenatal testing (NIPT) is reimbursed by the Belgian Social Security

for all pregnant women resulting in a high uptake. NIPT is an efficient means to detect fetal

aneuploidies. However, the major fraction of cell-free DNA in maternal plasma is derived from

maternal cells. Detection of clinically important variants in the maternal genome provides the

opportunity to diagnose maternal constitutional and acquired disorders and optimize follow-up

and management. We here report our one year experience on reported maternal secondary

variants, detected in a series of 26,123 NIP-tests performed in Leuven. Autosomal rearrangements

with a dominant effect were observed in 21 pregnancies (incidence 1/1243). These included five

microdeletions involving a tumor suppressor gene (NF1, BRIP1, MSH6, BRCA1 and DICER1). Four

different genomics disorders were detected: the 3q29 microdeletion (n=1) , PWS/AS duplication

(n=1), CMT1A (n=3) and HNPP (n=7). Two deletions included a gene causing a developmental

disorder when deleted (ASXL3 (apparently non mosaic); DNMT3A (mosaic)). Two large deletions

were reported: a mosaic 17Mb 9q21 deletion and one non-mosaic 9.5Mb 4qter deletion. With

regard to autosomal rearrangements with a recessive effect, 9 recurrent GJB6 microdeletions were

reported. Not unexpectedly, two thirds of actionable CNV´s were X-linked (65/95). The recurrent

STS deletion was the most common (27 cases), followed by intragenic DMD deletions or

duplications (n=11) and SHOX deletions (n= 4). Known XL-recessive disease genes that were found

to be deleted include Factor 8, COL4A6/COL4A5, and four deletions involved a developmental

disorder gene (IL1RAPL1, FRMPD4, ARSE, RPS6KA3 (n=2)). The recurrent int22h1/int22h2-mediated

Xq28 duplication syndrome was observed 6 times; the reciprocal deletion 4 times. Six large X

chromosomal deletions were detected. Our results show that 1/275 pregnant women carry a

clinically significant secondary finding. Genome-wide NIPT has the potential of informing pregnant

women on significant reproductive risks. However, this potential can only be fully reached on the

condition that the interpretation is performed by an expert team and reporting is based on

predefined guidelines with rapid access to follow-up genetic testing and high quality pre- and

posttest counselling.

E-mail: [email protected]

Keywords: non-invasive prenatal testing, secondary findings, maternal copy number variations

Page 32: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

31

T 04

In vitro fertilization does not increase the incidence of de

novo copy number alterations in fetal and placental lineages

Masoud Zamani Esteki 1,2,3,#,*, Triin Viltrop 4,#, Olga TÅ¡uiko 3,4,#, Airi Tiirats 5, Mariann Koel 6,

Margit Nõukas 7, Olga Žilina 7, Katre Teearu 7, Heidi Marjonen 8, Hanna Kahila 9, Jeroen Meekels

1, Viveca Söderström-Anttila 10, Anne-Maria Suikkari 10, Aila Tiitinen 9, Reedik Mägi 11,12, Sulev

Kõks 13, Nina Kaminen-Ahola 8, Ants Kurg 7, Thierry Voet 3,14,*, Joris Robert Vermeesch 3,* &

Andres Salumets 4,5,9,15,*

1 Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands 2 Department of

Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The

Netherlands 3 Centre for Human Genetics, University Hospital Leuven, Department of Human Genetics, KU Leuven, Leuven,

Belgium 4 Department of Biomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu,

Estonia 5 Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia 6

Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia 7 Department of

Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia 8 Department of Medical and

Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland 9 Department of Obstetrics and Gynecology, University

of Helsinki and Helsinki University Hospital, Helsinki, Finland 10 Väestöliitto Fertility Clinic, Helsinki, Finland 11 Estonian

Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia 12 Department of Bioinformatics, Institute of

Molecular and Cell Biology, University of Tartu, Tartu, Estonia 13 Centre for Comparative Genomics, Murdoch University,

Murdoch, Western Australia and The Perron Institute for Neurological and Translational Science, Nedlands, Western

Australia 14 Sanger-EBI Single Cell Genomics Centre, Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom

15 Competence Centre on Health Technologies, Tartu, Estonia # Co-first author * Co-senior author

We profiled the genomic landscape of fetal and placental tissues postpartum from in vitro fertilized

(IVF)- and naturally-conceived children to investigate the persistence of IVF-related CIN. To this

end, we alnalyzed de novo DNA copy number alterations in DNA samples extracted from cord

blood cells and placental tissue, which are derived from embryonic and extra-embryonic cell

lineages, respectively, of both IVF and naturally conceived neonates using haplarithmisis and

molecular karyotyping. Our innovative haplarithmisis-based approach can be used not only to

estimate fetal and maternal contribution to placental tissue, but also facilitates identification of

mosaic and non-mosaic aberrations in placental and fetal genomes in prenatal diagnostics.

Importantly, the transferred embryos of IVF newborns were not screened nor counter selected for

chromosomal aneuploidy status before intrauterine transfer. Following birth, we scrutinized

genome-wide single nucleotide polymorphism (SNP) profiles in DNA from the father, mother,

placenta and neonate umbilical cord blood of 111 families, of which 49 and 62 quartets were from

IVF and natural pregnancies, respectively. We detected de novoCNVs in placental genomes of

twelve families; of the total fifteen detected aberrations four were also present in newborns and

eleven were exclusively present in the placenta. We did not detect any CNVs present only in

umbilical cord blood DNA, which would indicate genomic alteration in the neonate only. The mosaic

(n= 12) de novoCNVs were 5–70% mosaic and were together with de novo non-mosaic CNVs (n

= 3) randomly located across the genome. Our analysis showed comparable prevalence (P= 0.327

odds ratio test or P= 0.775 Pearson´s chi square test) of de novo CNVs in both cohorts with AGA:

six of the 50 families with naturally conceived neonates (12.0%) and in three of the 48 families with

IVF neonates (6.3%). The high level of chromosomal mosaicism in IVF cleavage-stage embryos is

not preserved at later stages of prenatal development. Although we found a low-level mosaic

trisomy in one IVF placental genome, it was not present in the fetal lineage of the same pregnancy,

suggesting a proliferative defect of trisomic cells. The sporadic de novo genomic aberrations found

in approximately 10.8% of pregnancies were scattered across the genome and represented random

Page 33: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

32

and rare events of embryonic CIN, without any observed functional consequence on placental

biology or fetal health.

E-mail: [email protected]

Keywords: Embryo genetics, prenatal development, chromosome instability, haplarithmisis

Page 34: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

33

T 05

Longitudinal assessment of the 313-SNP based Polygenic

Risk Score for breast cancer risk prediction in a Dutch

prospective cohort

Inge M.M. Lakeman (1), J. van Rooij (2), R. Ruiter (3), C.J. van Asperen (4), André G. Uitterlinden

(2,3), Peter Devilee (1,5)

1. Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands 2. Department of internal

Medicine, Erasmus Medical Center Rotterdam, the Netherlands 3. Department of Epidemiology, Erasmus Medical Center,

Rotterdam, the Netherlands 4. Department of Clinical Genetics, Leiden University Medical Center, the Netherlands 5.

Department of Pathology, Leiden University Medical Center, the Netherlands

Purpose Breast cancer associated SNPs, summarized in a Polygenic Risk Score (PRS) are presently

not used to guide the appropriate starting age for breast cancer screening. At this moment the

most predictive PRS for breast cancer is based on 313 SNPs. The aim of this study is to validate the

performance of the 313-SNP PRS in a Dutch prospective cohort and explore the utility of risk

stratified screening. Materials and Methods We included women from the Rotterdam study, a Dutch

prospective cohort study started in 1989 with >70% response rate. Phenotypic information was

available with 25 years of follow-up. Genotyping was performed with the Illumina 550k and 610k

array for 6670 women aged 45 years or older. After imputation with the HRC1.1 and 1KG reference

panels, the standardized 313-SNP-based PRS (sPRS), per 1 standard deviation (SD) in controls from

the Breast Cancer Association Consortium (BCAC) was calculated. Cox regression analysis, adjusted

for birth year and age at inclusion, was performed to estimate the association between the sPRS

and incident breast cancer risk. Results Of all included women with an average age of 66, 145 had

prevalent breast cancer and 323 developed incident breast cancer during follow-up. These women

had a higher sPRS, mean 0.45 (SD=1.06) for cases with in situ carcinoma (N=37) and 0.51 (SD=1.06)

for cases with invasive carcinoma (N=431) compared to 0 (SD=1.01) in all 6202 controls, and 0.05

(SD=0.98) in 842 of these controls with other tumors than breast cancer. Breast cancer risk increased

by sPRS with an Hazard Ratio (HR) per SD=1.56, 95%CI [1.40-1.73], and compared with the middle

sPRS quintile, the lowest quintile had an HR of 0.57, 95%CI[0.38-0.87] and the highest quintile an

HR of 1.81, 95%CI[1.32-2.50]. We noticed a slightly smaller effect of the sPRS for in situ compared

to invasive breast cancer risk only (HR=1.45, 95%CI[1.02-2.01], and 1.58, 95%CI[1.41-1.76],

respectively). Discussion and conclusion: The 313-SNP PRS can be used for breast cancer risk

stratification in the Dutch population and seems specific for breast tumors and not for other tumor

types. Further analyses are being performed that include other breast cancer risk factors (i.e.,

reproductive and lifestyle factors), which are available for the majority of the included women.

Furthermore, the added value of the 313-SNP PRS on long- and short-term breast cancer risk

prediction and screening stratification will be explored.

E-mail: [email protected]

Keywords: Polygenic Risk Score, Breast Cancer, Genetic Epidemiology, Prospective cohort, Breast

screening

Page 35: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

34

T 06

Towards the treatment of Cantú syndrome

Helen I. Roessler(1), Conor McClenaghan(2), Yan Huang(2), Carmen M. Halabi(2,3), Theresa

Harter(2), Sanne Savelberg(1), Federico Tessadori(1,4), Jeroen Bakkers(4,5), Attila Kovacs(6), Maria

S. Remedi(2,6), Edwin P. Kirk(7,8), Ingrid Scurr(9), Sarah F. Smithson(9), Dorothy K. Grange(2,3), Colin

G. Nichols(2), Mieke M. van Haelst(10,11) and Gijs van Haaften(1)

1 Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The

Netherlands. 2 Center for the Investigation of Membrane Excitability Diseases (CIMED), and Department of Cell Biology and

Physiology, Washington University School of Medicine, St Louis, Missouri, USA. 3 Center for the Investigation of Membrane

Excitability Diseases (CIMED), and Departments of Pediatrics, Washington University School of Medicine, St. Louis, Missouri,

USA. 4 Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, the Netherlands. 5 Department of Medical

Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands. 6 Center for the

Investigation of Membrane Excitability Diseases (CIMED), Medicine, Washington University School of Medicine, St. Louis,

Missouri, USA. 7 Dept of Medical Genetics, Sydney Children's Hospital, Randwick NSW, Australia. 8 School of Women's and

Children's Health, University of New South Wales, Randwick NSW, Australia. 9 Department of Clinical Genetics, St Michael's

Hospital, Bristol, UK. 10 Department of Clinical Genetics, Amsterdam Medical Center, University of Amsterdam, Amsterdam,

The Netherlands. 11 Department of Clinical Genetics, VU Medical Center, VU University Amsterdam, Amsterdam, The

Netherlands.

Cantú Syndrome (CS), first described in 1982, is a rare genetic disorder caused by gain-of-function

(GOF) mutations in genes encoding the pore-forming (Kir6.1, KCNJ8) and regulatory (SUR2, ABCC9)

subunits of ATP-sensitive potassium (KATP) channels. Multiple case reports of individual patients

have described the various clinical features of CS, but systematic studies have been lacking. To

generate a critical mass of CS patients to find further clinical characteristics of the disease as well

as to ensure a rapid progression towards future interventional studies we developed a standardized

REDCap-based registry. We report phenotypic and associated genotypes on 75 CS patients, with

confirmed ABCC9 variants in 72 of the cases. Clinical data is obtained during annual Cantú research

clinics. Polyhydramnios during fetal life, lymphedema, patent ductus arteriosus (PDA),

cardiomegaly, dilated aortic root, vascular tortuosity of thoracic and cerebral arteries and migraine

headaches are common features. CS is debilitating with no targeted therapy available. Clinical

management currently involves symptomatic treatments to address second-order complications

such as heart failure and edema. The off-patent KATP inhibitor glibenclamide which is already

widely applied in clinic to block GOF KATP channels involved in Type 2 diabetes holds promise as

a potential treatment for CS. In order to perform therapeutic drug screening, we have developed

novel CS animal models in which disease-causing mutations were knocked-in to the endogenous

ABCC9 loci in zebrafish and mice using CRISPR/Cas9 genome engineering. Both models

recapitulate key cardiovascular features of CS which significantly reverse after glibenclamide

administration. Hypercontractility and abnormally high-output were reversed in zebrafish larvae

whilst cardiac hypertrophy in mice was similarly attenuated. No sustained hypoglycemia was

observed after chronic exposure to glibenclamide in mice. These results provide key pre-clinical

evidence for the in vivo efficacy of glibenclamide for the treatment of CS. Hence, we intend to

perform a clinical trial to test glibenclamide in CS patients.

E-mail: [email protected]

Keywords: Cantu syndrome, KCNJ8, ABCC9, Polyhydramnios; PDA; Hypertrichosis; Cardiomegaly;

CRISPR/Cas9; zebrafish; cardiovascular system

Page 36: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

35

T 07

NOTCH3 cysteine altering variants and their phenotypes in

92,456 whole exome sequenced participants of the Geisinger

DiscovEHR initiative

R.J. Hack1, J.W. Rutten1*, N. Pearson3*, , J.Li3, A. Khan2, M.A. Iqbal2, J. Hornak2, V. Abedi3, Y.

Zhang4, M.T.M. Lee4, C. Griessenauer2, Regeneron Genetics Center5, R. Zand2†, S.A.J. Lesnik

Oberstein1†* Shared second authors †Shared last authors

1. Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands 2. Neuroscience institute,

Geisinger Health System, Danville, Pennsylvania, USA 3. Biomedical and Translational Informatics Institute, Geisinger,

Danville, Pennsylvania, USA 4. Geisinger Genomic Medicine Institute, Danville, Pennsylvania, USA 5. Regeneron Genetics

Center, Tarrytown, New York, USA

Background: Cysteine altering missense variants in NOTCH3 have been exclusively associated with

CADASIL, a hereditary cerebral small vessel disease (SVD). Recently, we discovered that these

variants, especially those located in the NOTCH3 protein´s epidermal growth factor-like repeat

(EGFr) domains 7-34, have an unexpectedly high frequency in the general population worldwide,

namely 1:300. The goal of this study was to investigate the phenotype of participants with NOTCH3

cysteine altering variants in the Geisinger DiscovEHR initiative. Methods: We utilized whole exome

sequence data from 92,456 Geisinger-Regeneron DiscovEHR participants. We selected individuals

with a cysteine altering variant in one of the NOTCH3 EGFr domains. The control group, matched

for age and sex, had only synonymous variants in NOTCH3. We reviewed and recorded all the

patients´ demographic and clinical information, as well as neuroimaging characteristics. Group

comparisons were done using the ?2 test for categorical variables, and unpaired t-test for normally

distributed continuous variables. SPSS 24.0 was used for all statistical analyses. Results: 135

individuals with a NOTCH3 cysteine altering missense variant were identified (frequency 1:685), of

which 134 had a variant in one of EGFr domains 7-34. Medical records were available for 118 cases,

with a mean age of 58.1 ± 16.9 years. The control group consisted of 184 individuals, mean age

57.9 years ± 16.6. In the case group, 12.7% had a history of stroke, compared to 4.9% of controls

(p=0.014). Age at onset of stroke did not differ between cases and controls: 67,8 years ± 20.4 vs

65,0 years ± 11.72. There was no significant difference in the frequency of migraine headache,

depression, and dementia. Twenty-nine (25%) cases and 45 (24%) controls had an interpretable

MRI. Compared to controls, cases more frequently had large areas of confluent white matter

hyperintensities (Fazekas 3) (p=0.025), and there was a trend to a higher lacune count (p=0.055).

There was no significant difference in the number of microbleeds (p=0.550). Conclusion: NOTCH3

cysteine altering variants in the population, almost exclusively located in EGFr domains 7-34, are

associated with an increased prevalence of stroke and SVD markers on MRI. This suggests that

NOTCH3 EGFr 7-34 variants are a hitherto unknown risk factor for SVD in the general population,

but are rarely associated with a classical CADASIL phenotype.

E-mail: [email protected]

Keywords: CADASIL, cerebral small vessel disease, whole exome sequencing, NOTCH3, biobank

Page 37: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

36

T 08

Towards personalized treatment of genetically classified

refractory epilepsies using human induced pluripotent stem

Cells (hIPSCs) as an ex-vivo tool

E.J.H. van Hugte1,2,3, J. M. Keller1,2, E. Lewerissa1,2, M. Frega2,3, J. Nicolai1, H.J.M. Majoie1, H.J.

Schelhaas1, J.S. Verhoeven1, J.H.L.M van Bokhoven2, N. Nadif Kasri2,3

1 Department of Neurology, Academic Center for Epileptology, Heeze, The Netherlands. 2 Department of Human Genetics,

Radboudumc, Nijmegen, The Netherlands. 3 Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience,

Nijmegen, The Netherlands.

Dravet syndrome (DS) is a severe epileptic encephalopathy (EE) manifesting in the first year of life.

DS is characterized by recurrent and prolonged seizures, behavioral problems and developmental

delay. ln addition, EEs are associated with substantial cognitive and neuropsychological decline as

a result of seizures during brain development. lt is of crucial importance to timely control the seizure

activity. Although numerous antiepileptic drugs are available, it remains a challenge for clinicians

to select drugs with high efficacy but low side effects for each patient individually. Choosing

treatment is based on a rational trial and error approach, but an effective predictive tool to

investigate treatment options is lacking. ln 70-80% of the cases, DS results from de novo mutations

in the SCN1A gene, encoding the voltage gated nav1.1 channel. Conversely, not every patient with

a mutation in SCN1A develops the same symptoms and other genes have been associated with a

DS phenotype, in addition to SCN1A. Both this genetic and phenotypic heterogeneity add to the

difficulties of finding an appropriate treatment for DS. Animal models can neither recapitulate

mutation-specific effects nor genetic background factors, and are not suitable for devising

personalized medicine interventions. Hence, there is a strong need for patient-specific disease

models. The development of human induced pluripotent stem cells that are differentiated into

induced neurons (iNeurons) provides a solution. Here, we show that DS patient iNeurons exhibit an

hyperexitable phenotype on a single cell level and describe how iNeurons, cultured on micro-

electrode arrays (MEA) develop into neuronal networks. Specifically we generate and evaluate a

patient specific neuronal network (PNN) as a disease model for DS by co-culturing both excitatory

and inhibitory iNeurons. By assessing developing and mature PNNs, the MEAs allow us to study the

patient specific network fingerprint, providing a predictive tool for drug efficacy and side effects.

E-mail: [email protected]

Keywords: Epilepsy, human induced Pluripotent Stem Cells, brain-on-a-chip, neuronal networks,

Micro-electrode arrays, anti-epileptic drugs, Dravet Syndrome

Page 38: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

37

T 09

Genotyping On ALL patients (GOALL); clinical

implementation of high-throughput genotyping arrays.

J.G.J. van Rooij1, J.M.H. Verkerk1, B. Sedaghati-khayat1, L. Broer1, J.H. de Vries1, G.M. van Dijk1,

J.B.J. van Meurs1, A.G. Uitterlinden1.

1 Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands

Introduction: Genetic testing is increasingly used in standard clinical practice for most clinical

disciplines. However, genetic tests are expensive and/or must be rerun when additional clinically

relevant variants/genes are identified. Dense genotyping arrays (such as GSA or PMDA) cover a

large portion of genetic variation and provide a cost-effective, high-throughput and highly

standardized alternative. In the GOALL project, we are determining the clinical utility and

applications of genotyping arrays. Methods: We investigate the clinical utility of the GSAMD_v2 and

PMDA arrays by: 1) technical validation of common (vs. WES) and rare variants (vs. clinically

determined genotypes by different methods), 2) prospectively offering polygenic risk scores (PRS)

as primary utility in daily clinical practice, 3) prospectively counseling and testing eligibility of

feeding back secondary findings (pharmacogenomics, ACMG mutations) using array based

genotyping. We are piloting these applications in collaboration with the departments of Oncology

(breast cancer), Ophthalmology (age-related macular degeneration), Clinical Chemistry

(pharmacogenomics), Clinical Genetics (counseling) and Public Health within the Erasmus Medical

Centre. Results: The GSAMD_v2 array contains 12,755 clinical variants from the ClinVar database.

Our preliminary results suggest that >90% of the mutations routinely determined in our hospital

are covered by the array design (e.g. APOE, MTFHR, F5 etcera). We are currently selecting carriers

of clinically relevant mutations to check genotyping concordance. Comparison of array vs. WES of

197 population samples shows an overall non-reference concordance of 98%, and 95% for

singletons among ~30,000 directly genotyped variants and respectively 97% and 93% among

~150,000 imputed variants (QUAL>0.8). Discussion: Our preliminary results suggest that a portion

of current hospital-wide clinical genetic testing can cost-effectively be replaced by array-based

genotyping. In addition to reporting highly concordant results, array-based genotyping allows

additional reporting of several PRS and pharmacogenomics. This data can easily be stored and re-

analyzed when additional genetic loci or PRS are identified, or unblinded when a patient needs

further genetic testing not part of the initial primary clinical question.

E-mail: [email protected]

Keywords: arrays, personalized medicine, pharmacogenomics, polygenic risk scores

Page 39: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

38

T 10

The added value of long-read amplicon sequencing for

clinical applications

Kornelia Neveling, Dorien Lugtenberg, Arjen Mensenkamp, Ronny Derks, Amber den Ouden,

Michael Kwint, Simone van der Heuvel, Menno Tjon-Pon-Fong, Maartje van de Vorst, Linske de

Bruijn, Evelien Hoenselaar, Marjolijn Ligtenberg, Helger Yntema, Marcel Nelen, Lisenka Vissers,

Lonneke Haer-Wigman, Richarda de Voer

Department of Human Genetics, Radboud university medical center, Nijmegen, the Netherlands

Next generation sequencing has revolutionized the field of human genetics by offering new

possibilities to unravel human diseases. Due to limitations of short read sequencing however,

various additional tests including single gene sequencing, MLPA or genescan are still needed for

the analyses of certain difficult genomic regions. Long read high fidelity amplicon sequencing can

provide very accurate, efficient, high throughput sequences from single molecules, with read

lengths largely limited by PCR. The data is much easier to interpret, phased variants and breakpoints

are present within high fidelity individual reads, and additional tests can largely be eliminated. We

here demonstrate how we implemented long read SMRT sequencing in our diagnostic routine. As

examples, we will concentrate on the PMS2 and OPN1 genes. In both cases, homologous regions

make short read sequencing results very difficult to interpret. For PMS2, three amplicons ranging

in size from 11.4 kb to 16.8 kb were designed using primers unique to this gene, covering the entire

36 kb of sequence. SMRT sequencing produced HiFi reads with coverages ranging from 200x to

1500x. We will show examples of various types of aberrations detected by long-range sequencing,

including two deletions >1000kb with precise breakpoint mapping. For the homologous OPN1LW

and OPN1MW, specific 14.0 kb and 15.6 kb amplicons were designed covering the regions of

interest. Long read sequencing results will be shown for several patients in which gene conversions

and hybrid genes made short read sequencing data nearly impossible to analyze. For all cases,

PacBio sequencing confirmed the mutations originally identified by orthogonal technologies.

Moreover, it added further detailed information such as the ability to distinguish between genes

and pseudogenes and included phasing and precise breakpoint mapping. We feel that long-range

sequencing is an attractive alternative strategy to accurately analyze homologous genes, based on

the added value to reduce interpretation complexity and the need for additional confirmatory tests.

E-mail: [email protected]

Keywords: PacBio, long read sequencing, amplicons, PMS2, OPN1

Page 40: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

39

T 11

Reliable application of DNA-methylation signatures in

genetic diagnostic testing

Peter Henneman1,2, Marielle Alders1, Erfan Aref-Eshghi3,4, Jennifer Kerkhof4, Andrea Venema1,

Jet Biek1, Sonna Stolk1, Ruud van den Boogaard1, Bekim Sadikovic3,4 and Marcel Mannens1

1Amsterdam University Medical Centers, department of Clinical genetics, Genome diagnostics; 2Amsterdam University

Medical Centers, department of Clinical genetics, Core Facility Genomics; 3Department of Pathology and Laboratory

Medicine, Western University, London, ON N6A 3K7, Canada;4Molecular Genetics Laboratory, Molecular Di-agnostics

Division, London Health Sciences Centre, London, ON N6A 5W9, Canada.

Current genetic diagnostics of individuals suffering from rare syndromes that include impaired

neurodevelopmental and congenital anomalies, generally involves WES and CNV analysis. A

substantial proportion of such analyses however leaves the phenotype unexplained. Many of these

syndromes have been shown to result from a variant or mutation annotated to chromatin modifying

enzymes or chromatin binding protein complexes. Here, we have validated the clinical diagnostic

utility of genome-wide DNA methylation analysis of peripheral blood in unresolved cases (Kabuki,

SOTOS, CHARGE and others - VUS carriers) using the previously reported methodology and in

collaboration with Aref-Eshghi and Sadikovic et al (AJHG, 2019). We applied a computational model

enabling concurrent detection of 20 syndromes using DNA methylation data with full accuracy. We

were able to fully resolve the diagnosis of individuals with uncertain clinical and genetic diagnosis

for the following disorders: ADNP-related syndromes, BAFopathies, Cornelia de Lange syndrome,

CHARGE syndrome, Kabuki syndrome, SOTOS syndrome, Claes Jensen Syndrome, BWS, SRS and

Angelman syndrome. This study confirms that genomic DNA methylation analysis can facilitate the

molecular diagnosis of unresolved clinical cases and highlights the potential value of genome-wide

DNA-methylation testing in the routine clinical assessment of unresolved cases with rare disorders

involving neurodevelopmental and congenital anomalies. Upcoming research will extend the

number of detectable syndromes up to 40 and the panel will be updated shortly. Moreover, the

number chromatin modifying proteins is estimated to involve approximately 200 genes, which

potentially are candidates for further research and that may be included in future versions of our

panel. Our test (EpiSign) has now been fully implemented in collaboration with, London, Canada,

and is for European requests exclusively available at the AUMC, clinical genetics, genome

diagnostics department (genomediagnostics.amsterdamumc.nl).

E-mail: [email protected]

Keywords: DNA-methylation signature, genetic diagnostics, rare syndrome

Page 41: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

40

T 12

What if we would use a diagnostic multi-cancer gene panel

for opportunistic screening? A study in 2,090 Dutch familial

cancer patients

L.F. Johansson, K.K. van Dijk-Bos, A.H. van der Hout, A.P. Knopperts, B. Leegte, P.C. van den Akker,

K. Kok, I.M. van Langen, M.A. Swertz, R. K. Weersma, R.J. Sinke, B. Sikkema-Raddatz, R.H. Sijmons,

H. Westers

University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands

University of Groningen, University Medical Center Groningen, Department of Gastroenterology, Groningen, the Netherlands

Purpose In familial cancer (FC) diagnostics, analysis of next-generation sequencing data typically

focuses on genes known to be associated with the cancer type that prompted referral. Currently,

however, it is debated whether opportunistic screening should be performed when sequence data

is available for other genes. We aimed to determine how many secondary findings (SFs) would be

detected in cancer-predisposing genes present in our FC gene panel if we offered opportunistic

screening to patients within FC diagnostics. Methods We anonymously reanalyzed sequencing data

of 2,090 FC patients for either 73 genes (original FC panel) or 85 genes (updated panel) for SNVs,

indels and CNVs. To determine the background prevalence of pathogenic variants in FC genes, we

screened 1,326 individuals from the general Dutch population. Results We detected SFs in 3.0% of

patients (excluding heterozygous CHEK2 and MUTYH variants), and a (likely) pathogenic variant

matching their family´s cancer type in 10.1% of patients. In the Dutch population cohort, 3.2% of

individuals had a (likely) pathogenic variant in a cancer-predisposing gene. Conclusion Our results

can assist in the design of future research programs on opportunistic screening. These programs

are needed because there is not yet sufficient evidence to meet international screening program

criteria.

E-mail: [email protected]

Keywords: Genetic screening, diagnostic yield, gene panel, next-generation sequencing, familial

cancer

Page 42: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

41

T 13

Neurodevelopmental disorders: a next generation

Margot R.F. Reijnders1, Tjitske Kleefstra2, Lisenka E.L.M. Vissers2, Han G. Brunner1,2

1 Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands. 2 Department of

Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.

Since 2010 whole exome sequencing (WES) discovered a large numbers of novel genes and

neurodevelopmental disorders (NDDs), denoted here as 'next-generation NDDs´. Yet, ~40% of NDD

patients remain undiagnosed. Therefore, we previously established a cohort of 826 patients with

unexplained ID, a subtype of NDD, for whom trio-based WES was performed. ID severity ranged

from mild (27%) to severe (27%) and 32.2% of patients had other NDD-subtypes. We identified

pathogenic variants in known NDD genes in 28.6% of patients and 386 patients (46.7%) had at least

one de novo mutation in a gene not previously associated with NDDs. Between 2014 and 2018, we

have so far confirmed seven of 586 genes as next-generation NDD-gene

(SON,TLK2,RAC1,RHEB,USP9X,RAB11B,WAC). Matchmaking was successful for all but one (RHEB),

resulting in a maximum of 35 additional patients (TLK2) and underscoring the need for data-sharing

for next-generation NDDs. Additionally, mutations in these genes together only diagnosed ˜1%

of patients of our initial cohort, illustrating the rarity of next-generation NDDs. Of note, for USP9X,

WAC and TLK2, most patients were mildly affected. Further systematic analysis of our cohort

showed that overall a similar percentage of de novo mutations in known NDD genes was present

in patients with mild ID (27.9%) compared to more severe ID (27.2%), indicating that WES should

not be restricted to patients with more severe phenotypes. Finally, we observed a challenge to

translate the discovered next-generation NDDs to clinic. To improve patient care, we propose an

organizational model, in which parents have a key role.

E-mail: [email protected]

Keywords: Rare neurodevelopmental disorders; mild intellectual disability; data sharing; clinical

translation.

Page 43: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

42

T 14

Transcriptome and protein analysis highlight the endosomal

pathway in disease pathogenesis of metabolic CL syndrome

Pottie L, Van Gool W, Van Hooydonck M, De Saffel H, Vermassen P, Coucke P, Sips P and Callewaert

B

Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium

We recently described a novel autosomal recessive cutis laxa (CL) syndrome, caused by missense

mutations in subunit V1E1 of the vacuolar H+-ATPase (ATP6V1E1). The condition is clinically

characterized by loose redundant skin folds, variable mental disability, typical facial characteristics,

lipodystrophy, hypotonia, and cardiopulmonary involvement including pneumothorax,

hypertrophic cardiomyopathy and aortic root dilatation. Similarly to ATP6V0A2-related cutis laxa,

impaired retrograde Golgi transport, glycosylation abnormalities, and a dysfunctional secretory

pathway have been implicated in the pathogenesis, but the pleiotropic manifestations remain

largely unexplained. We report on two knock-out (KO) zebrafish models. The first model has a

proviral insertion in the 5´UTR disrupting atp6v1e1b transcription. The second model has a

premature stop in atp6v1e1b, generated by CRISPR-Cas9 mutagenesis. Both models are

embryonically lethal, show decreased pigmentation, have reduced touch-evoked escape response

and hatching, manifest altered craniofacial cartilage structures, and show abnormal blood vessel

structure and cardiac dysfunction. Transcriptome analysis revealed downregulation of transferrin-a

(tfa), hemoglobin alpha embryonic 1.1 and hemoglobin beta embryonic 1.3. KEGG pathway analysis

indicates a decreased function of the oxidative phosphorylation pathway. These results suggest a

disturbance in iron homeostasis. KO of atp6v1e1b in zebrafish disrupts the maturation from the

early endosome into the late endosome. Protein analysis indicates upregulation of early endosome

antigen 1 and small rab GTPases 5 while small rab GTPases 7 and lysosome associated membrane

glycoprotein show a trend towards downregulation. We hypothesize that reduced acidification of

the endosomes will block the release of iron and the recycling of tfa-tfa receptor complex to the

cell surface implementing pathological manifestations. In conclusion, KO of atp6v1e1b in zebrafish

faithfully recapitulates the human cutis laxa syndrome. The vacuolar H+-ATPase is important for

proper functioning of the endosomal pathway opening avenues for therapeutic strategies.

E-mail: [email protected]

Keywords: cutis laxa syndrome ATP6V1E1 zebrafish transcriptoom endosomal pathway iron

homeostasis

Page 44: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

43

T 15

Noncoding structural variants disrupt the regulatory

architecture of Rett genes

Eva D´haene, Reut Bar-Yaacov, Inbar Bariah, Lies Vantomme, Sien Van Loo, Francisco Avila Cobos,

Karen Verboom, Reut Eshel, Rawan Alatawna, Björn Menten, Ramon Y Birnbaum, Sarah Vergult

Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Belgium Department of Life Sciences,

Faculty of Natural Sciences, Ben-Gurion University of the Negev, Israel Center of Evolutionary Genomics and Medicine, Ben-

Gurion University of the Negev, Israel Cancer Research Institute Ghent (CRIG), Ghent, Belgium Bioinformatics Institute Ghent

from Nucleotides to Networks (BIG N2N), Ghent, Belgium

Rett syndrome is a progressive neurodevelopmental disorder, characterized by a severe

developmental delay, absence of speech, seizures, hypotonia and stereotypic movements. It is

typically caused by mutations in the MECP2 gene, but several other genes, including the

transcription factors MEF2C and FOXG1, have been associated with a Rett-like phenotype as well.

Recently, we and others identified several noncoding structural variants (SVs) in patients with Rett-

like characteristics. All SVs are located proximal to the coding sequence of MEF2C or FOXG1,

suggesting disruption of the regulatory structure governing these genes. Using Circularized

Chromosome Conformation Capture (4C) sequencing in a neuronal cell line, we identified a

complex regulatory interaction network in the MEF2C region. We found that the MEF2C promoter

physically contacts multiple distal enhancer regions upstream of its coding sequence. Based on

epigenetic enhancer marks and sequence conservation, we delineated 16 putative enhancer

elements, of which 14 were active in in vitro luciferase assays and 8 displayed in vivo neuronal

activity during zebrafish development. For FOXG1 as well, 4C-seq experiments have shown that the

promoter interacts with at least three in vivo validated brain enhancers, all situated in a region

affected by deletions or translocations in multiple Rett-like patients. In summary, Rett genes MEF2C

and FOXG1 are part of complex regulatory networks involving multiple distal enhancers. Disruption

of these regulatory structures by noncoding SVs could form the genetic basis of the Rett-like

phenotype observed in some patients.

E-mail: [email protected]

Keywords: Rett syndrome, noncoding structural variation, gene regulation, chromosome

conformation

Page 45: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

44

T 16

Loss of neutral sphingomyelinase-3 (SMPD4) links

neurodevelopmental disorders to cell cycle and nuclear

envelope anomalies

Pamela Magini(1,38), Daphne J. Smits(2,38), Laura Vandervore(2,3), Rachel Schot(2), Marta

Columbaro(4), Esmee Kasteleijn(2), Mees van der Ent(5), Flavia Palombo(6), Maarten H. Lequin(7),

Marjolein Dremmen(8), Marie Claire Y. de Wit(9), Mariasavina Severino(10), Maria Teresa Divizia(11),

Pasquale Striano(12,13), Natalia Ordonez-Herrera(14), Amal Alhashem(15,16), Ahmed Al

Fares(15,16), Malak Al Ghamdi(17), Arndt Rolfs(14), Peter Bauer(14), Jeroen Demmers(18), Frans W.

Verheijen(2), Martina Wilke(2), Marjon van Slegtenhorst(2), Peter J. van der Spek(19), Marco Seri(20),

Anna C. Jansen(3,21), Rolf W. Stottmann(22), Robert B. Hufnagel(23), Robert J. Hopkin(22,24),

Deema Aljeaid(25), Wojciech Wiszniewski(26,27), Pawel Gawlinski(27), Milena Laure-

Kamionowska(28), Fowzan S. Alkuraya(29), Hanah Akleh(30), Valentina Stanley(31), Damir

Musaev(31), Joseph G Gleeson(31), Maha S. Zaki(32), Nicola Brunetti Pierri(33) , Gerarda

Cappuccio(34), Bella Davidov(35), Lina Basel-Vanagaite(35), Lily Bazak(35), Noa Ruhrman

Shahar(35), Aida Bertoli Avella(12), Ghayda M. Mirzaa(36,37), William B. Dobyns(36), Tommaso

Pippucci(1), Maarten Fornerod(5,39), Grazia M. S. Mancini(2,39)

1- Medical Genetics Unit, S.Orsola-Malpighi Hospital, via Massarenti 9, 40138, Bologna, Italy 2- Department of Clinical

Genetics, ErasmusMC University Medical Center Rotterdam, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands. 3- Vrije

Universiteit Brussel, Centrum Medische Genetica, Laarbeeklaan 101, 1090 Brussel 4- Laboratory of Musculoskeletal Cell

Biology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy 5- Department of Cell Biology, ErasmusMC UnMeesiversity

Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands 6- IRCCS Institute of Neurological

Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy 7- Department of Radiology, University medical center Utrecht,

Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands 8- Department of Radiology, Sophia Children´s hospital, ErasmusMC

University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands 9- Department of Child

Neurology, Sophia Children´s hospital, ErasmusMC University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD,

Rotterdam, Rotterdam, NL 10- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy 11- UOC Genetica

Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy 12- Pediatric Neurology and Muscular diseases Unit, IRCCS

Istituto Giannina Gaslini, 16147 Genova, Italy 13- Department of neursciences, Rehabilitation, Ophthalmology, Genetics,

Maternal and Child Health, University of Genova, 16126 Genova, Italy 14- CENTOGENE AG. Am Strande 7. 18055 Rostock,

Germany 15- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia 16- Department of

Pediatrics, College of Medicine, Qassim University, Qassim 52571, Saudi Arabia 17- Department of Pediatrics, College of

Medicine, King Saud University, Riyadh 11451, Saudi Arabia, 18- Proteomics Center, Room Ee-679A, Erasmus University

Medical Center. Wytemaweg 80, 3015 CN Rotterdam 19- Department of Pathology, Clinical Bioinformatics, ErasmusMC

University Medical Center PO Box, 3000CA Rotterdam, The Netherlands. 20- Medical Genetics Unit, Department of Medical

and Surgical Sciences, University of Bologna, via Massarenti 9, 40138, Bologna, Italy 21 - Pediatric Neurology Unit, UZ

Univeristy of Brussel, Laarbeeklaan 101, 1090 Brussel, Belgium 22- Department of Human Genetics, Division of

Developmental Biology Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016. Cincinnati, OH

45229 23- Medical Genetics and Ophthalmic Genomics Unit, National Eye Institute, National Institutes of Health, 10 Center

Drive, Building 10, Room 10N109, Bethesda, MD 20892 24- Department of Pediatrics, University of Cincinnati, College of

Medicine, 3333 Burnet Ave. ML 4006 Cincinnati OH 4522933. 25- Department of Human Genetics, Faculty of Medicine, King

Abdulaziz University, Jeddah 21589, Saudi Arabia 26- Oregon Health & Sciences University 3181 SW Sam Jackson Park

Road L103. Portland, Oregon, U.S.A 27- Institute of Mother and Child, Kasprzaka 17a, 02-211 Warsaw, Poland 28-

Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre Polish Academy of

Sciences, Pawinskiego 5, 02-106 Warsaw, Poland 29- Department of Genetics, King Faisal Specialist Hospital and Research

Center, Riyadh, Saudi Arabia 30- Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research

Center, Riyadh 12713, Saudi Arabia 31- Laboratory for Pediatric Brain Disease, Department of Neurosciences, Howard

Hughes Medical Institute, University of California, San Diego, California, USA 92093. 32 - Human Genetics and Genome

Research Division, Clinical Genetics Department, National Research Centre, Cairo 12622, Egypt. 33 - Brunetti- Department

of Translational Medicine, University of Naples "Federico II", Naples 80131 Italy 34 - Cappuccio - Telethon Institute of

Genetics and Medicine (TIGEM), Pozzuoli 80078, Italy 35 - Department of Medical Genetics, Recanati Genetic Institute Rabin

Page 46: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

45

Medical Center, Beilinson Campus, Petah Tikva, 4941492, Tel Aviv, Israel 36- Department of Pediatrics, University of

Washington, Seattle, WA 98195, Washington, USA 37- Center for Integrative Brain Research, Seattle Children´s Research

Institute, Seattle, WA 98101, Washington, USA. 38- These authors contributed equally 39 - These authors contributed equally

Introduction: Several neurodevelopmental processes including neuronal survival, migration and

differentiation are controlled by sphingolipid metabolism. Sphingomyelinases generate ceramide

from sphingomyelin as a second messenger in intracellular signaling pathways involved in cell

proliferation, differentiation, or apoptosis. While the role of acid sphingomyelinase is well

established, the role of neutral sphingomyelinases in human neurodevelopment has remained

elusive. Methods: Twenty-nine children from 12 families presented with microcephaly with

simplified gyral pattern, cerebellar hypoplasia, severe developmental encephalopathy, congenital

arthrogryposis, diabetes mellitus and early fetal-postnatal demise. All index cases have bi-allelic

loss of function variants in the SMPD4 gene, coding for neutral sphingomyelinase-3.We performed

additional studies to elucidate the mechanisms contributing to disease pathogenesis. Results:

Fibroblasts from affected individuals showed morphologic endoplasmic reticulum (ER) cisternae

abnormalities, consistent with a previously suggested function of SMPD4 in the ER. Overexpression

of human Myc-tagged SMPD4 in HEK392T cells showed localization of the protein to both the

nuclear envelope and the ER. Previous studies localized SMPD4 to the outer nuclear membrane.

Mass spectrometry of SMPD4-associated proteins detected peptides belonging to nuclear pore

complex proteins. We also observed delayed cell cycle progression after downregulation of SMPD4

by siRNA. These data are similar to former studies in HeLa cells showing mitotic abnormalities after

siSMPD4 treatment. Conclusions: Our study describes a novel microcephaly syndrome

characterized by severe neurologic abnormalities. Our results suggest a major function of neutral

sphingomyelinase-3 in regulation of mitosis through modulation of nuclear membrane lipids,

pointing to a novel mechanism and pathway in the pathogenesis of microcephaly.

E-mail: [email protected]

Keywords: microcephaly, arthrogryposis, neutral-sphingomyelinase, SMPD4

Page 47: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

46

Abstracts Posters (P)

No. Name Titel E-mail

P 01 Hanka Venselaar Lost in translation - Experiences from a

Structural Biologist

[email protected]

P 02 Nika Schuermans* NPC2 c.441+1G>A is a likely pathogenic

variant associated with an adult-onset

Alzheimer-like phenotype

[email protected]

P 03 Aude Beyens* A clinical flowchart to classify cutis laxa

patients, supported by electron

microscopy and pathophysiology

[email protected]

P 04 Eline Verberne* Genetic service for birth defects at the

Dutch Caribbean Islands

[email protected]

P 05 Marielle van Gijn Dutch genome diagnostic laboratories

improved variant interpretation quality

and speed by building and

implementing a data sharing platform

[email protected]

P 06 Marian Stevens-

Kroef

Next-generation cytogenetics: high-

resolution optical mapping to replace

FISH, karyotyping and CNA-microarrays

marian.stevens-

[email protected]

P 07 Lennart Raman PREFACE: in silico pipeline for accurate

cell-free fetal DNA fraction prediction

[email protected]

P 08 Jo Vanoevelen DTYMK deficiency is the cause of a

severe congenital neurodegenerative

disease

[email protected]

P 09 Eva Jacobs* A novel neurodevelopmental syndrome

caused by loss-of-function of the Zinc

Finger Homeobox 3 gene (ZFHX3)

[email protected]

P 10 Isabelle Bar Clinical, histopathological and molecular

characterization of a rare case of adult

pilocytic astrocytoma with

differentiation to pilomyxoid

astrocytoma: case report

[email protected]

P 11 Erika Souche Validation of whole genome sequencing

for the diagnosis of rare diseases

[email protected]

P 12 Li-Yen Wong Investigation of the pathogenicity of

LMNA variants in human pluripotent

stem cell-cardiomyocytes using

CRISPR/Cas9 technology

[email protected]

P 13 Mirjam Wamelink Transketolase (TKT) deficiency in 3

patients with short stature,

developmental delay and abnormal

polyols

[email protected]

P 14 Linda Zuurbier Diagnostic yield of Next-Generation

Sequence analysis of a 29 gene panel

related to genetic dyslipidemias

[email protected]

P 15 Machteld Oud Towards a novel diagnostic strategy

using patient-derived cells to diagnose

ciliopathies

[email protected]

P 16 Floor Postema The validation of a clinical screening

instrument for tumor predisposition

syndromes in childhood cancer patients

(TUPS): a prospective, observational,

multi-center study

[email protected]

Page 48: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

47

P 17 Hannie Kremer An in-frame deletion in RIPOR2 is an

important cause of adult-onset hearing

impairment

[email protected]

P 18 Erik-Jan Kamsteeg KIF1A variants are a frequent cause of

autosomal dominant hereditary spastic

paraplegia

[email protected]

P 19 Laura van Zutven 19q13.32 microdeletion syndrome:

further delineation of the clinical

phenotype

[email protected]

P 20 Gido Gravesteijn* NOTCH3 exon skipping as a rational

therapeutic approach for CADASIL:

lessons from a family with naturally

occurring exon 9 skipping

[email protected]

P 21 Zeinab Fadaie* Identification of splice defects due to

non-canonical splice site or deep-

intronic variants in ABCA4

[email protected]

P 22 Eva Sammels A novel missense PLP1 mutation causing

PLP1-related spastic paraplegia: a Case

Report

[email protected]

P 23 Ivo Fokkema* LOVD - supporting DNA variant

interpretation

[email protected]

P 24 Joost Kummeling* SETD1A Loss of function causes a

distinct neurodevelopmental disorder in

human and impaired learning &

memory in Drosophila.

[email protected]

P 25 Mubeen Khan* Towards comprehensive identification

and functional characterization of deep-

intronic ABCA4 variants in 1000

Stargardt disease cases

[email protected]

P 26 Mahdi

Motazacker

Compound heterozygosity for missense

and null variants in CEP55 lead to a

novel viable phenotype

[email protected]

P 27 Jessica Bos Clinical characteristics of patients with

an STXBP1 gene mutation: preliminary

results of a cross-sectional and

retrospective multilevel cohort study

[email protected]

P 28 Vyne van der

Schoot*

The impact of incidental findings in

clinical exome sequencing, a qualitative

interview study

[email protected]

P 29 Shuang Li* CAPICE - a variant prioritization tool for

Consequence-Agnostic Prioritization of

Clinical Exomes variations

[email protected]

P 30 Matthias Van Gils* Targeted compound screening for PXE

using a CRISPR/Cas9 abcc6a mutant

zebrafish model.

[email protected]

P 31 Tamara

Koopmann

Whole-exome sequencing identifies

heterozygous stop-loss mutations in

HBB resulting in an elongated β-globin

chain in two children with severe

anemia.

[email protected]

P 32 Valerie

Jacquemin*

A second family with homozygous

mutation in KIDINS220 presenting as

congenital hydrocephalus and limb

contractures

[email protected]

P 33 Rosanne Ausems* Intrinsic myogenic potential of skeletal

muscle-derived pericytes from patients

with myotonic dystrophy type 1

[email protected]

P 34 Renée

Raaijmakers*

Development and Characterization of

Pericyte-Derived iPSCs and PiPs from

DM1 patients

[email protected]

Page 49: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

48

P 35 Marieke Klein Intellectual Disability-related genes

increase ADHD risk and locomotor

activity in Drosophila melanogaster

[email protected]

P 36 Mala Misra-Isrie An integrative approach to STXBP1-

Encephalopathy: in vitro, in vivo and

patient clinics.

[email protected]

P 37 Diantha Terlouw* The complexities of analysis for APC

mosaicism

[email protected]

P 38 Serwet Demirdas The MAP3K7 gene: further elaboration

on phenotypes, and evidence for

interaction with the RAS

pathway/clinical overlap with Noonan

syndrome

[email protected]

P 39 Lianne Krab Further delineation of phenotypes

related to integral cohesion structural

protein RAD21

[email protected]

* Eligible for Poster Award

Page 50: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

49

P 01

Lost in translation - Experiences from a Structural Biologist

Venselaar

Radboudumc

Structural bioinformatics is of essential importance to understand the molecular effects of genetic

variants. Over the last years, the CMBI at Radboudumc has been involved in many collaborative

projects with (bio-)medical researchers. In these projects, the use of information obtained from 3D-

structures often provided important information to understand genotype-phenotype relations and

often, this information was a used as a lead for therapy or drug-development, or simply as a starting

point for new experiments. We have seen an enormous rise in technological possibilities and the

subsequent growth in data. In contrast, studying a protein in detail remains a laborious task that

requires special skills, time, and experience. Fortunately, many tasks can be automated and we, and

many others, have created automatic tools and software solutions to open up the world of protein

structures to the bio-medical field. However, these tools rely on the completeness and correctness

of the underlying data, which isn't always the case. Besides that, the result as presented by software

isn't always clear for the user without a structural bioinformatics background, and doesn't always

take the inter-human variation into account. In this lecture, we will discuss experiences, challenges

and results from a day with a structural biologist.

E-mail: [email protected]

Keywords: protein structures mutations variant effect prediction

Page 51: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

50

P 02

NPC2 c.441+1G>A is a likely pathogenic variant associated

with an adult-onset Alzheimer-like phenotype

Nika Schuermans1,2, Giulia Ascari1,2, Alexander Gheldof3, Céline Bellenguez4,5,6, Frans W.

Verheijen7, Ariane Van Tongerloo1,2, Florence Pasquier6,8, Gaël Nicolas9, Jean-Charles

Lambert4,5,6, Thorsten Marquardt10, Elfride De Baere1,2, Sandra Janssens1,2, Bart Dermaut1,2

1. Center for Medical Genetics, Ghent University Hospital, Belgium. 2. Department of Biomolecular Medicine, Faculty of

Medicine and Health Sciences, Ghent University, Belgium. 3. Center for Medical Genetics, UZ Brussel, Brussels, Belgium. 4.

Inserm U1167, Molecular Determinants of Aging-Related Diseases, Lille, France. 5. Institut Pasteur de Lille, Lille, France. 6.

Excellence Laboratory LabEx DISTALZ, Lille, University of Lille, France. 7. Department of Clinical Genetics, Erasmus University

Medical Center, Rotterdam, the Netherlands. 8. Department of Neurology, University Hospital of Lille, Lille, France. 9.

Department of Genetics and CNR-MAJ, Normandie Université, UNIROUEN, Inserm U1245 and Rouen University Hospital, F

76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France. 10. Klinik für Kinder- und Jugendmedizin,

Münster, Germany.

Background: Niemann-Pick type C disease (NPC) is an autosomal recessive neurodegenerative lipid

storage disorder caused by mutations in the NPC1 and NPC2 genes. Due to a heterogeneous

phenotype with a variety of clinical symptoms and highly variable ages of onset diagnosis is difficult.

Although the clinical incidence is estimated at ~1/100.000, carrier frequencies of predicted

pathogenic NPC1/2 variants in NGS-based population databases suggest a higher incidence. This

could be due to an important clinical underascertainment of adult-onset neurodegenerative

phenotypes (reduced or age-related penetrance) and/or an overestimation of the pathogenicity of

certain variants (assessed by in silico prediction software). Here we studied the pathogenic role of

NPC2 c.441+1G>A, a canonical splice-site variant with a minor allele frequency of ~1/150 and

conflicting evidence of pathogenicity. Results: We identified a fetal and two adult cases

homozygous for NPC2 c.441+1G>A. The 12-weeks-old fetal case was identified via chorion villus

sampling in an at-risk pregnancy. After pregnancy termination filipin staining of cultured fetal

fibroblasts showed increased levels of unesterified cholesterol confirming the diagnosis of NPC.

Transcript analysis by means of RT-PCR revealed the presence of three aberrant transcripts, whereas

no wild type transcript was present. Western Blotting showed strongly decreased NPC2 protein

levels. One adult case was identified in the French ADES study, consisting of exome data from 1779

Alzheimer´s disease (AD) patients and 1273 controls. This patient was diagnosed with clinical AD at

the age of 54 years, had a cerebrospinal fluid biomarker profile compatible with AD, a negative

family history and was APOE ?4 negative. The other adult patient, homozygous for the NPC2

c.441+1G>A mutation, is a German female with a severe early-onset dystonia who is a also a carrier

of an expanded CAG trinucleotide repeat in the ATXN3 gene, causing Machado-Joseph disease.

Conclusions: Together our results suggest a likely pathogenic nature for NPC2 c.441+1G>A possibly

leading to a late-onset neurodegenerative AD-like phenotype. These findings might have

implications for diagnostics and management of patients with a neurodegenerative clinical

presentation and the genetic counseling of carriers of this mutation.

E-mail: [email protected]

Keywords: Niemann-Pick disease type C, NPC2, rare neurodegenerative diseases, next-generation

sequencing, variant classification, diagnostics

Page 52: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

51

P 03

A clinical flowchart to classify cutis laxa patients, supported

by electron microscopy and pathophysiology

A. Beyens1,2, H. Syryn1, L. Pottie1, R. De Rycke3, B. Fischer-Zirnsak4, T. Van Damme1, I. Hausser5,

M. De Bruyne3, F. Baeke3, M. Morroni6, S. Nampoothiri7, K. Mahesh8, U. Kornak4, Z. Urban9, S.

Hadj-Rabia10, C. Bodemer11, S. De Schepper2, E. C. Davis12, B. Callewaert1

1 Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium 2 Department of Dermatology, Ghent University

Hospital, Ghent, Belgium 3 Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium 4 Institute of

Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany 5 Institute of Pathology,

Universitätsklinikum Heidelberg, Heidelberg, Germany 6 Department of Experimental and Clinical Medicine, Section of

Neuroscience and Cell Biology, School of Medicine, Università Politecnica delle Marche and Electron Microscopy Unit, United

Hospitals, Ancona, Italy 7 Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Center, Kochi,

India 8 Department of Pediatric Cardiology, School of Medicine, Kochi, India 9 Department of Human Genetics, Graduate

School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States 10 Service de Dermatologie, CHU Paris -

Hôpital Necker-Enfants Malades, Paris, France 11 Centre MAGEC (Maladies rares Génétiques a Expression Cutanée), Service

de Dermatologie, CHU Paris - Hôpital Necker-Enfants Malades, Paris, France 12 Department of Anatomy and Cell Biology,

McGill University, Montréal, QC, Canada.

Introduction Cutis laxa (CL) syndromes are a heterogeneous group of rare, multisystem connective

tissue disorders that share a loose, redundant skin as a common feature reflecting elastic fiber

deficiency. The pathogenesis of each of the CL subtypes is different but affects elastogenesis. Light

microscopy of the dermis is non-discriminative and the recently observed vast molecular

heterogeneity mitigates the clinical validity and practicality of the current classification, based on

the mode of inheritance and systemic involvement. We aim to classify the CL subtypes by means

of a simple flowchart and to evaluate correlations between elastic fiber ultrastructural morphology

and clinical presentation. Materials and Methods Transmission electron microscopy was used to

evaluate all CL subtypes. We conducted a systematic search of all existing literature on CL patients

with confirmed molecular defects in one of the known CL genes (ELN, FBLN4, FBLN5, LTBP4,

SLC2A10, ATP6V0A2, ATP6V1E1, ATP6V1A, ALDH18A1, PYCR1, RIN2, GORAB and ATP7A). We

established databases with the clinical data of our in-house cohort and all patients reported in

literature. Results? We developed a clinical flowchart to classify the CL subtypes. As a proof of

principle, we systematically evaluated 67 CL patients from our in-house database and could allocate

95% of patients to the right gene. To confirm final validation of our classification, we evaluated all

495 patients reported in literature and successfully classified more than 90% in a retrospective

manner. Transmission electron microscopy in skin biopsies of all CL subtypes showed discriminative

and specific findings that correlate with the main presenting symptoms (emphysema, arterial

tortuosity, skeletal defects/mental disability with or without glycosylation defects/intrauterine

growth retardation/cataract). Moreover, elastic fiber ultrastuctural morphology reflects the involved

molecular pathogenesis and provides new insights in elastic fiber biogenesis.? Conclusions? Our

novel nosology of the CL syndromes provides a practical approach to the broad differential

diagnosis of CL syndromes. The classification forms a basis to integrate the clinical presentation

with the pathogenesis and ultrastructural elastic fiber defects. We expect this nosology to ease the

diagnostic work-up of CL patients, to be relevant for the interpretation of genotype-phenotype

correlations, and to bode for new management guidelines and therapeutic approaches.

E-mail: [email protected]

Keywords: cutis laxa, flowchart, classification, electron microscopy, elastic fiber

Page 53: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

52

P 04

Genetic service for birth defects at the Dutch Caribbean

Islands

E.A. Verberne, S. Faries, M. Manshande, S. Lo-a-Njoe, P. Philippi, H. Veenhuis, F. Falix, G. Ecury, I.

Angelista, E. Arends, L. Rafael-Croes, W. Ponson, P. Thorsen, A. Oudshoorn, F. Croes, M. de Vroomen,

P. Offringa, M. Tilanus, M.M.A.M. Mannens, M.M. van Haelst.

Amsterdam UMC, University of Amsterdam. St. Elisabeth Hospital, Curacao. Dr. Horacio E. Oduber Hospital, Aruba.

Fundashon Mariadal, Bonaire. St. Maarten Medical Center, St. Maarten.

Introduction: Curacao, Aruba, Bonaire and St. Maarten are islands located in the Caribbean Sea and

part of the Dutch Kingdom. Because of their small size and relative remoteness, they face several

challenges, including limited access to (specialized) health care like medical genetics. We

established a genetic service for pediatric patients on these Dutch Caribbean Islands. The aim of

this study is to evaluate the diagnostic and clinical outcome of 7-year genetic care service for this

pediatric population. Methods: Twice per year the clinical geneticist evaluates patients with birth

defects at the pediatric clinic. If indicated, blood samples are send to the Netherlands for genetic

testing. For this study, we retrospectively included all children (age <18 years) that had a genetic

consultation between November 2011 and November 2018. We excluded children that already

received a definitive diagnosis previously and healthy children that were referred for carrier testing.

Because of the relatively isolated population we hypothesized that specific conditions (caused by

autosomal recessive or founder mutations) could be more frequently identified amongst these

children. Results: In total 479 children were included in this study. In 24% we were able to establish

a definitive clinical and/or molecular diagnosis. Molecular diagnoses included autosomal dominant

conditions (40%), autosomal recessive conditions (10%), microdeletion/-duplication syndromes

(32%), X-linked conditions (7%), aneuploidy (6%) and other (5%). Conclusion: We present our

strategy to provide genetic care in the Dutch Caribbean by establishing a local genetic service and

show the results of the first 7-years. Since genetic results are still pending for part of the cohort, a

final diagnosis could be higher than 24% of the children. Although the population structure of small

islands could suggest a higher level of recessive conditions, we have not (yet) been able to show

this for these Dutch Caribbean islands. A genetic diagnosis helps in ending the diagnostic odyssey

for patients and parents and enables them to make informed reproductive choices. We suggest

that this strategy with a visiting clinical geneticist or via teleconferencing can be used to provide

genetic testing for birth defects in other small island developing states (SIDS).

E-mail: [email protected]

Keywords: Genetic service, diagnostic strategy, birth defects, Dutch Caribbean

Page 54: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

53

P 05

Dutch genome diagnostic laboratories improved variant

interpretation quality and speed by building and

implementing a datasharing platform

Ivo F.A.C. Fokkema, K. Joeri van der Velde, Mariska K. Slofstra, Claudia A.L. Ruivenkamp, Maartje J.

Vogel, Rolph Pfundt, Marinus J. Blok, Ronald H. Lekanne Deprez, Quinten Waisfisz, Kristin M. Abbott,

Richard J. Sinke, Rubayte Rahman, Isaäc J. Nijman, Hanneke W.M. van Deutekom, Bart de Koning,

Gert Thijs, Nienke Wieskamp, Ruben J.G. Moritz, Bart Charbon, Jasper J. Saris, Johan T. den Dunnen,

Jeroen F.J. Laros, Morris A. Swertz, Marielle E. van Gijn

Department of Human Genetics & Department of Clinical Genetics, Leiden University Medical Center, Leiden, the

Netherlands; Genomics Coordination Center & Department of Genetics, University Medical Center Groningen, University of

Groningen, Groningen, The Netherlands; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the

Netherlands; Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Human

Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Clinical Genetics, Maastricht

University Medical Center+, Maastricht, the Netherlands;Department of Clinical Genetics, Academic Medical Center &

Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; DGG

Genomics Software Solutions, Agilent Technologies, Leuven, Belgium; Department of Genetics, University Medical Center

Utrecht, Utrecht, the Netherlands

Each year diagnostic laboratories in the Netherlands test thousands of individuals for heritable

disease using next generation sequencing (NGS). This requires pathogenicity classification of

millions of DNA variants on the standard 5-tier scale. To reduce time spent on data interpretation

and increase data quality and reliability, the nine Dutch labs decided to publicly share their

classifications. Variant classifications of nearly 145,000 unique variants were catalogued and

compared in a centralized MOLGENIS database. Variants classified by more than one center are

labeled as "consensus" when classifications agree, and shared internationally with LOVD and

ClinVar. When classifications oppose (LB/B versus LP/P), they are labeled "conflicting", while other

non-consensus observations are labeled "no consensus". We assessed our classifications using

InterVar to compare to ACMG 2015 guidelines, showing a large overall consistency with only 0.3%

discrepancies. Differences in classifications between Dutch labs or between Dutch labs and ACMG

were mainly present in genes with low penetrance or for late onset disorders and highlight

limitations of the current 5-tier classification system. To address these differences and diminish

confusion a clinical arm could be added to the current classification system, to complement the

ACMG system. Such a system is currently being developed in an ESHG initiative. The data sharing

was implemented successfully in the laboratories in their daily practice and evaluation showed it

boosted the quality of DNA diagnostics in Dutch labs. Publically sharing the data resulted a disease

diagnosis in at least one positive match with a case from outside our consortium. This initiative

proves that data sharing is warranted for good clinical practice in genome diagnostics

E-mail: [email protected]

Keywords: Data sharing, variant pathogenicity classification, VKGL, ACMG classification system

Page 55: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

54

P 06

Next-generation cytogenetics: high-resolution optical

mapping to replace FISH, karyotyping and CNA-microarrays

Marian Stevens-Kroef1*; Tuomo Mantere1*; Kornelia Neveling1*; Marian Stevens-Kroef1*; Daniel

Olde Weghuis1; Marc Pauper1, Guillaume van de Zande1, Susan Vermeulen1, Ellen Kater-Baats1,

Michiel Oorsprong1, Ronald van Beek1, Dominique Smeets1; Alex Hastie2; Alexander Hoischen

1,3,4 * Equal contribution

1 Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands. 2 Bionano Genomics,

San Diego, California, USA 3 Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen,

The Netherlands. 4 Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University

Medical Center, Nijmegen, The Netherlands.

Structural variants (SVs) are an important source of genetic variation in the human genome and

they are involved in a multitude of human diseases. Somatic SVs are important for cancer

development and progression while constitutional SVs are involved in many developmental

disorders. In a diagnostic set-up for leukemia samples, comprehensive analysis of all cytogenetic

aberrations in a given sample still requires a combination of techniques, such as CNA-microarrays,

karyotyping and fluorescence in situ hybridization. We hypothesize that the combination of classical

approaches could be largely replaced by high-resolution optical mapping. Leukemia bone marrow

aspirates and blood samples were analyzed using standard of care workflow and the residual

samples were processed for Bionano Saphyr optical mapping to detect chromosomal aberrations.

We generated up to 400-fold genome coverage by imaging long chromosomal fragments and

detected chromosomal aberrations by comparing optical maps to a reference and control dataset.

In the first leukemia samples we compared the results with those obtained using standard methods.

In each sample, all previously known clinically relevant aberrations were identified. This held true

for deletions, insertions, inversions and translocations, including a 3-way Philadelphia chromosome

(t(9;22;14)(q34;q11;q11)) and even chromothripsis structures were resolved. We were also able to

identify aberrations in samples with cancer cell content less than 40% using Bionano´s latest single-

molecule SV detection tool. Importantly, optical mapping additionally identified multiple novel

events e.g. an inversion of chromosome 11 (chr11:24,875,044-26,299,641) and a translocation

(t(5;14)(q35.2;q32.2)), both validated afterwards. These novel events are now followed-up to identify

possible fusion genes as novel leukemia drivers. We are now conducting a study to systematically

compare the sensitivity and specificity of optical mapping in 100 leukemia samples and 50 samples

with known germline cytogenetic aberrations against the standard of care workflow. Of these, 17

with constitutional aberrations and 19 leukemia samples have been processed successfully so far.

Optical mapping may have the potential to replace most classical cytogenetic tests. In addition this

holds the potential to map new fusion genes as novel leukemia drivers rapidly.

E-mail: [email protected]

Keywords: high-resolution optical mapping, FISH, karyotyping, CNA-microarrays.

Page 56: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

55

P 07

PREFACE: in silico pipeline for accurate cell-free fetal DNA

fraction prediction

Raman Lennart, Baetens Machteld, De Smet Matthias, Dheedene Annelies, Van Dorpe Jo, Menten

Björn

1 Department of Pathology, Ghent University, Ghent University Hospital, Ghent, Belgium. 2 Center for Medical Genetics,

Department for Biomolecular Medicine, Ghent University, Ghent University Hospital, Ghent, Belgium.

OBJECTIVE: During routine non-invasive prenatal testing (NIPT), cell-free fetal DNA fraction is

ideally derived from shallow-depth whole-genome sequencing data, preventing the need for

additional experimental assays. The fraction of aligned reads to chromosome Y enables proper

quantification for male fetuses, unlike for females, where advanced predictive procedures are

required. This study introduces PREFACE (PREdict FetAl ComponEnt), a novel bioinformatics

pipeline to establish fetal fraction in a gender-independent manner. METHODS: PREFACE combines

the strengths of principal component analysis and neural networks to model copy number profiles.

RESULTS: For sets of roughly 1100 male NIPT samples, a cross-validated Pearson correlation of 0.9

between predictions and fetal fractions according to Y chromosomal read counts was noted.

PREFACE enables training with both male and unlabeled female fetuses. Using our complete cohort

(nfemale =2468, nmale =2723), the correlation metric reached 0.94. CONCLUSIONS: Allowing

individual institutions to generate optimized models sidelines between-laboratory bias, as PREFACE

enables user-friendly training with a limited amount of retrospective data. In addition, our software

provides the fetal fraction based on the copy number state of chromosome X. We show that these

measures can predict mixed multiple pregnancies, sex chromosomal aneuploidies and the source

of observed aberrations.

E-mail: [email protected]

Page 57: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

56

P 08

DTYMK deficiency is the cause of a severe congenital

neurodegenerative disease

Vanoevelen JM1,2*, Bierau J1*, Kamsteeg EJ3, Wevers RA4, Bok LA5, Abdel-Salam GMH6, van der

Knaap M7, Bugiani M8, O´Driscoll M9, van den Wijngaard A1, Ferreira CR10, Brunner HG1, Wang

L11, Stumpel CTRM1,2

1.Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands; 2. GROW - School

for Oncology and Developmental Biology, Maastricht, The Netherlands; 3. Department of Human Genetics, Radboud UMC,

Nijmegen, The Netherlands; 4. Translational Metabolic Laboratory, Radboud UMC, Nijmegen, The Netherlands; 5.

Department of Pediatrics, Màxima Medical Center, Veldhoven, The Netherlands; 6. Department of Clinical Genetics, Human

Genetics and Genome Research Division, National Research Centre, Cairo, Egypt; 7. Department of Child Neurology,

Amsterdam UMC, Amsterdam, The Netherlands; 8. Department of Neuropathology, Amsterdam UMC, Amsterdam, The

Netherlands; 9. Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom; 10. National Human

Genome Research Institute, National Institutes of Health, Bethesda, USA; 11. Department of Anatomy, Physiology and

Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden; * shared first authors.

Human nucleotide metabolism is a complex, tightly controlled pathway regulating numerous

cellular processes such as nucleic acid synthesis and repair, cellular proliferation and differentiation.

In this study we describe DTYMK (dTMPK) deficiency as the cause of a severe neurodegenerative

disease in two unrelated families. DTYMK encodes the dTMPK (deoxythymidylate monophosphate

kinase) enzyme which catalyzes the penultimate step in the biosynthesis pathway of dTTP. We

describe two unrelated children showing severe postnatal microcephaly and postnatal growth

retardation. Fascinatingly, in both cases postnatal neurodevelopment was minimal. Imaging of the

brain revealed extensive atrophy of the cerebral cortex with secondary white matter loss and

atrophy of the basal ganglia. Two predicted pathogenic variants in DTYMK were identified;

compound heterozygous variants in one patient and one of the former variants in a homozygous

state in the other patient. No significant dTMPK enzyme activity could be detected in the patients´

fibroblasts, indicating a loss-of-function effect of the variants. Additionally, EdU labelling in

fibroblasts confirmed a marked proliferation defect in these cells. To further substantiate a causal

relationship between DTYMK disruption and the patient´s phenotype, we generated a dtymk loss-

of-function allele in zebrafish. Homozygous dtymk mutant zebrafish are not viable beyond 5dpf

(days post fertilization) and show microcephaly, small eyes, developmental delay, cardiac edema

and prominent edema of the brain. Biochemical analysis of dTMPK activity in mutant zebrafish

larvae confirmed that the allele represents a loss-of-function allele leading to undetectable enzyme

activity. Furthermore, impairment of proliferation was also detected in the brain of mutant zebrafish

larvae, as well as genome instability due to ribonucleotide incorporation and defects in the DNA-

damage response repair mechanism. The striking similarities between the human and zebrafish

phenotype strongly suggest a causal link between dTMPK deficiency and the neurodegenerative

phenotype, observed in both patients. In summary, by combining genetic and biochemical

approaches in a cellular and a zebrafish model we identified loss-of-function in DTYMK as the cause

of a severe neurodegenerative disease. These cases highlight the importance of dTTP synthesis in

the survival of neurons.

E-mail: [email protected]

Keywords: Brain atrophy Nucleotide metabolism zebrafish

Page 58: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

57

P 09

A novel neurodevelopmental syndrome caused by loss-of-

function of the Zinc Finger Homeobox 3 gene (ZFHX3)

Eva Jacobs (1), Sarah Vergult (1), Maria Palomares Braro (2), Sixto Garcia-Minaur (2), Santos Simarro

Fernando (2), Tina Duelund Hjortshøj (3), Marion Gérard (4), Arnaud Molin (4) , Pablo Villavicencio-

Lorini (5), Jorgen Köhlhase (5), Katrin Hoffmann (5), Mareike Bauer (5), Hilde Peeters (6), Peter

Aerssens (7), Nicole Revencu (8), Philippe Lysy (8), Pernille Torring (9), Kristina Sorensen (9), Carlo

Marcelies (10), Lot Snijders Blok (10), Annelies Dheedene (1), Björn Menten (1), Bert Callewaert (1)

(1) Ghent University Hospital, Ghent, Belgium, (2) Hospital Universitario La Paz, Madrid, Spain, (3) Rigshospitalet, Glostrup,

Danemark, (4) Centre Hospitalier Universitaire de Caen, Caen, France, (5) Universitätsklinikum Halle, Halle, Germany, (6)

University Hospital Leuven, Leuven, Belgium, (7) Virga-Jesse Hospital, Hasselt, Belgium, (8) Cliniques Universitaires Saint-

Luc, Brussels, Belgium, (9) Odense University Hospital, Odense, Denmark, (10) Radboud University Medical center, Nijmegen,

the Netherlands

We describe the neurodevelopmental phenotype caused by de novo loss-of-function mutations in

the Zinc Finger Homeobox 3 gene (ZFHX3). In an international collaborative effort, using the

Decipher and Genematcher databases, we identified de novo 16q22.2 microdeletions in 6 patients

with a minimal region of overlap only containing the ZFHX3 gene. Three additional patients

harbored a de novo premature truncating mutation in this gene. All patients show a similar

phenotype, namely intellectual disability and craniofacial malformations. Next to this, ZFHX3 has a

pLI score of 1, which indicates this gene is intolerant for loss-of-function mutations. Therefore, we

hypothesize that ZFHX3 is the causal gene for the observed syndromal intellectual disability. ZFHX3

belongs to the family of zinc-finger homeodomain transcription factors and encodes the ATBF1

protein. This transcription factor plays a role in multiple biological processes including

tumorigenesis and cell differentiation. Expression of ZFHX3 increases during development of the

human cortex and in zebrafish, expression of zfhx3 peaks three days after fertilization. In this period,

the process of brain development takes place in zebrafish. In addition, localization of ZFHX3 to the

nucleus has been shown to induce cell cycle arrest associated with neuronal differentiation. These

data support a role for ZFHX3 in neural differentiation. So far, no human phenotype has been

correlated with molecular defects in ZFHX3. Functional assays to determine how loss-of-function

of ZFHX3 give rise to syndromal intellectual disability are ongoing.

E-mail: [email protected]

Keywords: neurodevelopmental disorder

Page 59: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

58

P 10

Clinical, histopathological and molecular characterization of

a rare case of adult pilocytic astrocytoma with differentiation

to pilomyxoid astrocytoma: case report

I Bar1, A Merhi 2, S Toffoli 3, C DeRop 3 , C Gilliard 4, O Fouard 4, JN Comps 4, M Di Santo 4, T

Gustin 4, P Delree 5

1 Laboratory of Translational Oncology, Institute of Pathology and Genetics/ Grand Hôpital de Charleroi, Gosselies, Belgium.

2 IPG BioBank, Institute of Pathology and Genetics, Gosselies, Belgium. 3 Center for Human Genetics, Institute of Pathology

and Genetics, Gosselies, Belgium. 4 Department of Neurosurgery, CHU UCL Namur, Yvoir, Belgium 5 Department of

Anatomopathology, Institute of Pathology and Genetics, Gosselies, Belgium

Pilocytic astrocytomas (PAs) are the most common pediatric brain tumors. They are grade I tumors

with a good prognosis following surgical resection. Hereby, we describe a rare example of PA in

adult showing a rapid regrowth within 6 months with differentiation to grade III pilomyxoid

astrocytoma (PMA). A 32 years old man was diagnosed with a cystic tumor in the right temporal

region. Histological examination of the tumor was consistent with the diagnosis of pilocytic

astrocytoma (PA). The proliferation index was low, with positive staining for GFAP, Vimentin and

S100. Numerous eosinophilic granular bodies and Rosenthal fibers were observed. The tumor was

resected by surgery. Six months later, the patient was diagnosed with recurrence of the primary PA

into a grade III pilomyxoid astrocytoma (PMA). Rosenthal fibers were absent. The Ki-67 index and

mitotic index were strongly increased. Apoptosis and necrosis areas were present. Comparative

genomic hybridization was performed on both tumor samples. Monosomy 1p and trisomy 19 p

were detected in the primary tumor. The second lesion revealed a more complex genomic profile,

with genomic instability evidenced by an increased number of large copy number aberrations:

chromosomes 2, 4, 5, 10, 11, 13 and 22 monosomy, co-deletion 1p/19q, 19p trisomy and 9p21

homozygous deletion (involving both CDKN2A and CDKN2B). Targeted sequencing of 409

oncogenes and tumor suppressor genes was performed on both tumors and blood leucocytes DNA

(Ion AmpliSeqâ„¢ Comprehensive Cancer Panel). Both primary and secondary tumors had

comparable mutation profiles, with no new somatic mutations in the grade III PMA compared to

grade I PA. Somatic mutations in exon 4 of IDH1 and IDH2 were not detected, but a somatic point

mutation in BRAF ( p.(V600E) – c.1799T>A – NM_004333.5) was present both in primary and

secondary tumor. This was confirmed by BRAF V600E immunostaining. Methylation of MGMT

promoter was also detected. Conclusion: In the present case, tumor progression seems to be

mediated by chromosomal instability rather than acquisition of new somatic mutations. Very few

studies have carefully analyzed the molecular parameters leading to the transformation of a low

grade to a high grade pilocytic astrocytoma in adult. It is important to accumulate data to improve

our understanding of the biology of these tumors, which could help to orientate therapeutics

options.

E-mail: [email protected]

Keywords: Pilocytic astrocytoma, pilomyxoid astrocytoma, adult patient, histopathology,

molecular characterization

Page 60: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

59

P 11

Validation of whole genome sequencing for the diagnosis of

rare diseases

Erika Souche, Luc Dehaspe, Kris Van Den Bogaert, Marijke Bauters, Valérie Race, Anniek Corveleyn,

Anneleen Boogaerts, Jens Van Bouwel, Stephanie Deman, Céline Helsmoortel, Wouter Bossuyt, Gert

Matthijs, Joris Vermeesch

Center for Human Genetics

Whole Genome Sequencing (WGS) is quickly becoming the standard of care to diagnose rare

hereditary diseases. In our project we aimed to implement WGS in a diagnostic setting because it

promises to deliver more information than the current standard of care, namely, a 2-step approach

of arrayCGH and whole exome sequencing (WES). Although these approaches are effective, there

is still room for improvement. Indeed, copy neutral rearrangements cannot be detected by

arrayCGH. Structural variants such as inversions and translocations are difficult to detect using WES

as the breakpoints might not be exonic. Also, WES suffers from capture and PCR biases and some

variants might be missed. WGS allows the detection of CNVs, complex structural variants but also

single nucleotide variants in a single test. Its implementation in a diagnostic setting will homogenize

the wet lab workflow, offer a single test for all conditions, and ultimately decrease cost and time to

diagnosis. However, the capability of WGS to replace arrayCGH and WES has to be proven before

moving forward. Well characterized cell lines have been sequenced on a NovaSeq (PE150) after

shearing and library preparation according to KAPA Hyper PCR free protocol. WGS data has been

compared to both arrayCGH and Platinum and Genome in a Bottle calls. Additionally, clinical exome

data has been compared to WGS data for some real cases.

E-mail: [email protected]

Keywords: Whole genome sequencing diagnostics

Page 61: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

60

P 12

Investigation of the pathogenicity of LMNA variants in

human pluripotent stem cell-cardiomyocytes using

CRISPR/Cas9 technology

Wong, L.Y., van Dijk, W., Veltrop, R., Akbulut, C., van Tienen, F., Antoons, G., Nagy, M., Manders, E.,

Helmes, M., Broers, J., Schurgers, L., Heemskerk, J., Glatz, J., van den Wijngaard, A., Brunner, H.G.

Department of Clinical Genetics, MUMC+, Maastricht University, Maastricht, Netherlands Department of Genetics and Cell

Biology, Maastricht University, Maastricht, Netherlands Department of Biochemistry, CARIM, Maastricht University,

Maastricht, Netherlands Department of Physiology, CARIM, Maastricht University, Maastricht, Netherlands Cytocypher,

VUMC, Amsterdam, Netherlands Department of Human Genetics, RadboudUMC, Nijmegen, Netherlands

Dilated cardiomyopathy (DCM), which is characterized by dilation of left- or both ventricles and

systolic dysfunction, comprises many hospitalized cardiomyopathy cases and has major impact on

public health. About 48% of DCM cases are familial, with over 50 DCM genes identified to date.

Among them, mutations in LMNA gene (encoding nuclear protein Lamin A/C) account for 5-8% of

families with familial DCM. Modern sequencing technologies, e.g. whole-exome-sequencing (WES),

has identified numerous genetic variants in LMNA gene, however the pathogenicity of these

variants and the molecular mechanisms leading to DCM remain unknown. This project aims to

utilize induced pluripotent stem cells (iPSC) as a model system (i) to phenotypically characterize

LMNA mutations causing DCM, and (ii) to design novel genome editing strategies to recapitulate

the disease phenotype by applying the state-of-the-art CRISPR/Cas9 technology. This allows the

interrogation of genetic mutations to decipher the genotype-phenotype relationship in a more

precise and measurable manner. CRISPR/Cas9 genome editing of healthy wild-type IPSCs were

performed to create mutant isogenic IPSC lines by introducing an LMNA variant identified in DCM

patients in order to recapitulate the disease phenotype in comparison with patient-derived IPSCs.

CRISPR-edited and patient-specific IPSCs, together with the relevant control IPSCs were

subsequently differentiated to cardiomyocytes over a period of 2 weeks under feeder-free

conditions. Metabolic selection of IPSC-CMs were performed to yield a high population of purified

IPSC-CMs. Phenotypic and functional analyses such as gene expression, immunocytochemistry,

contractility, and calcium handling will be assessed to distinguish the similarities/differences

between patient-derived IPSCs and CRISPR-edited IPSCs, as compared to wild-type IPSCs. The

powerful combination of IPSC-based disease modeling and CRISPR-Cas9 genome editing will

provide insight into the contribution of these LMNA genetic variants to the pathogenesis of DCM

and to study VUS functional phenotypes in a patient-specific manner. These findings can then be

used for early disease detection, diagnosis and clinical management of patients, leading to an

improvement in patient healthcare. This platform represents a promising tool for assessing DCM-

associated VUS specifically and can significantly contribute to precision medicine for the study of

genetic diseases in the near future.

E-mail: [email protected]

Keywords: Induced pluripotent stem cells, dilated cardiomyopathy, CRISPR-Cas9 genome editing,

cardiomyocyte differentiation

Page 62: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

61

P 13

Transketolase (TKT) deficiency in 3 patients with short

stature, developmental delay and abnormal polyols

Wamelink MMC1, Charles P2,3, Keren B3, Faudet A2, Buratti J3 Roos, B1, Jansen EEW1, Hay E4,

Faravelli, F4

1VUmc, Dept of Clin Chem, Amsterdam, The Netherlands; 2 dept. of Neurology, Hôpital Pitié-Salpêtrière, APHP, Paris, France

3 Dept. Clinical Genetics,, Hôpital Pitié-Salpêtrière, APHP , Paris, France, 4 Clinical Genetics, Great Ormond Street, London,

United Kingdom

BACKGROUND: Transketolase (TKT) deficiency is a rare autosomal recessive defect in the pentose

phosphate pathway (PPP), previously only reported in 5 patients from 3 families. We describe 3

additional patients. CASE REPORT: WES/WGS identified biallelic variants in TKT in 3 females from 2

families, ranging in age from 5 to 32 years. Polyols in urine and plasma and sugar-P in urine were

measured to confirm the pathogenicity. RESULTS: Patient 1, female 5 years old present significant

short stature, microcephaly, severe developmental delay and bilateral frontal lobe polymicrogyria.

Patient 2 and 3, sisters of 32 and 21 years old, present short stature (-2DS and -4DS) with growth

hormone treatment for the oldest, microcephaly, developmental delay, behaviour and character

disorders intellectual disability and morphological features of the face. The youngest presented

tetralogy of fallot with surgical intervention at 8 months and developed bilateral cataract at 21 years

with surgical treatment. The oldest presented dextrocardia and pyramidal signs. Three novel

missense variants in TKT were identified: Patient 1: homozygous for c.362C>T, p.Ala121Val, patient

2 and 3: compound heterozygous for c.1421G>A; p.Arg474His and c.964G>A; p.Gly322Arg. In all 3

elevated urine excretion of erythritol, arabitol, ribitol, erythrose-4-P and pent(ul)ose-5-P and

elevated plasma concentrations of erythritol, arabitol and ribitol, confirmed TKT deficiency.

DISCUSSION: TKT deficiency is now diagnosed in 8 patients (7 female and 1 male). All patients were

small for gestational age, had short stature, and had varying degrees of developmental delay and

morphological features of the face. Congenital heart defects and juvenile cataract are common.

Erythrose-4-P was elevated, indicating that there might be an alternative route. Our findings

provide further support for the biochemical and phenotypic features reported in TKT deficiency.

Given the lack of specific or distinctive diagnostic features in early years, we propose polyol testing

in patients fitting this clinical phenotype.

E-mail: [email protected]

Keywords: Transketolase, pentose phosphate pathway, polyols, TKT, short stature

Page 63: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

62

P 14

Diagnostic yield of Next-Generation Sequence analysis of a

29 gene panel related to genetic dyslipidemias

Linda C. Zuurbier, PhD, Olaf R.F. Mook, PhD, Martin A. Haagmans, Marcel M.A.M. Mannens, PhD,

Joep. C. Defesche, PhD

Introduction: Genetic dyslipidemias, specifically those affecting the metabolism of low-density

lipoprotein cholesterol, high-density lipoprotein cholesterol and triglycerides, are extremely

common in the general population and are strongly associated with premature atherosclerosis,

presenting as myocardial infarctions and sudden death at young age. Because of this abundance,

there is a need for rapid, reliable and comprehensive diagnostic tools. Advances in sequencing

technology, such as Next-Generation Sequencing (NGS), and bioinformatics have made it possible

to analyze large numbers of genes in many patients simultaneously. Method: On more than 4000

consecutive patients that were referred for confirmation of the clinical diagnosis of a genetic

dyslipidemic disorder, NGS analysis of a dyslipidemia-associated 29-gene panel was applied.

Results: Analysis of these data revealed that the clinical diagnosis frequently differed from the

genetic diagnosis. For instance, one third of patients that was clinically diagnosed with

hypercholesterolemia had a pathogenic variant that was causal for hypertriglyceridemia. On the

other hand, compared to the conventional single gene sequencing approach, NGS supplied

additional valuable results, as in more than 10% of cases a second or third pathogenic or

pharmacogenetic variant was identified that was relevant for clinical decision making. Furthermore,

by elaborating in more detail on pharmacogenetic variants, a statin intolerance variant was

observed much more frequently than published in literature. Also other SLCO1B1 variants were

identified that affected therapeutic response. Conclusion: For the detection of genetic

dyslipidemias, NGS is a rapid and very efficient diagnostic tool that increases the yield and quality

of diagnosis, compared to conventional Sanger sequencing. In this particular setting, the unsolicited

findings not only explicate the clinical diagnosis, but also significantly optimize the medical

management of genetic dyslipidemias.

E-mail: [email protected]

Keywords: dyslipidemia, hypercholesterolemia, cardiovascular, pharmacogenetics

Page 64: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

63

P 15

Towards a novel diagnostic strategy using patient-derived

cells to diagnose ciliopathies

Machteld Oud1,2, Ronald van Beek1,2, Dorien Lugtenberg1, Ernie Bongers1, Ronald Roepman1,

Helger Yntema1, Lisenka Vissers1,2

1Department of Human Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands 2Donders Institute for

Brain, Cognition and Behavior, Radboud University Medical Centre, Nijmegen, The Netherlands

Ciliopathies are rare recessive multiorgan disorders that are caused by dysfunction of the cilium.

Cilia are small signaling organelles protruding from the cell membrane of nearly every vertebrate

cell. Diagnostic exome analysis (WES) for ciliopathies leads to an inconclusive molecular diagnosis

in about 30% due to the detection of variants of unknown significance. Functional tests evaluating

the effect of genetic variants on cilium function could elucidate their pathogenicity. Our aim is

therefore to develop a novel diagnostic strategy to improve accurate diagnosis of ciliopathies. We

obtained skin-fibroblasts from 21 unrelated ciliopathy patients and controls, and cultured them in

vitro. Immunocytochemistry techniques were applied to determine cilium phenotype parameters,

including 1) ciliogenesis, based on presence or absence of a base and an axonemal marker, 2) cilium

length, by measurement of the combined signal of an axonemal and a cilium membrane marker,

and 3) intraflagellar transport (IFT), measured by the area of the ciliary tip based on the IFT88 signal.

All experiments were performed in triplicate. We collected cilium phenotypic data from skin-

fibroblasts of 21 (suspected) ciliopathy patients and 8 controls. The data of the eight control

fibroblasts yielded comparable and reproducible results: >85% cells were ciliated, the cilium length

was 2.97±0.381µm, and the IFT88 ciliary tip area was 0.14±0.03µm2. Significant differences in cilium

length and IFT results were identified in nine short-rib thoracic dysplasia (SRTD) ciliopathy patients

when compared to the controls. Interestingly, the data of the different subgroups within the SRTD

patient cohort clustered together and were clearly distinguishable from the other clinical

phenotypes and controls. The three parameters tested did however not result in identifiable clusters

for the remaining seven ciliopathy and five suspected ciliopathy patients. In summary, we show that

a standardized analysis of the cilium phenotype can lead to identifiable clusters for SRTD

ciliopathies and possibly also for other ciliopathies. Based on these results, we believe that

standardized tests based on cilium phenotypic data can provide an additional piece of the puzzle

for conclusive diagnosis of ciliopathies, essential in routine diagnostic care.

E-mail: [email protected]

Keywords: Ciliopathy, diagnostics, functional test

Page 65: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

64

P 16

***WITHROWN*** The validation of a clinical screening

instrument for tumor predisposition syndromes in childhood

cancer patients (TUPS): a prospective, observational, multi-

center study

Floor A.M. Postema1,2, Saskia M.J. Hopman3, Corianne A.J.M. de Borgie4, Cora M. Aalfs3, Jakob K.

Anninga2, Lieke P.V. Berger5, Charlotte J. Dommering6, Natasha K.A. van Eijkelenburg2, Peter

Hammond7, Marry M. van den Heuvel-Eibrink2, Wijnanda A. Kors2, Tom G.W. Letteboer3, Jan L.C.M.

Loeffen2, Lisethe Meijer2, Maran J.W. Olderode-Berends5, Anja Wagner8, Marjolijn C.J. Jongmans3,

Raoul C. Hennekam1, Johannes H.M. Merks2

1. Department of Pediatrics, Emma Children´s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The

Netherlands 2. Princess Máxima Center for Pediatric oncology, Utrecht, The Netherlands 3. Department of Genetics,

University Medical Center Utrecht, Utrecht, The Netherlands 4. Clinical Research Unit, Amsterdam UMC, University of

Amsterdam, Amsterdam, The Netherlands 5. Department of Genetics, University of Groningen, University Medical Center

Groningen, Groningen, The Netherlands 6. Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam,

Amsterdam, The Netherlands 7. Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United

Kingdom 8. Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands

BACKGROUND Up to 10% of children with cancer has a tumor predisposition syndrome (TPS).

Identification of a TPS in a child with cancer is of clinical relevance. Previous studies have shown

that half of TPSs are not recognized prior to treatment. To increase diagnostic accuracy and clinical

efficiency we developed a screening instrument, ensuring evaluation for a TPS in all children with

cancer. METHODS TuPS is a prospective, observational, multi-center study including children newly

diagnosed with cancer in the Netherlands from 2016-2019 in whom no TPS was previously

diagnosed. The screening instrument consists of a clinical assessment, 2D and 3D pictures, and

integrated digital assessment by two independent clinical geneticists. If a TPS was suspected by at

least one clinical geneticist (positive assessment), the patient was referred to previously uninvolved

clinical geneticists for genetic counseling. In addition negative assessed patients were randomized

for genetic consultation as control group. Primary outcome is the clinical value and acceptance of

the instrument. PRELIMINARY RESULTS Results of 315 patients show a positive assessment rate of

57% (180/315) (25% by one assessor, 32% by both assessors). The reason for positive assessment

was based mainly on type of cancer (54%) and family history (38%). In 17% (30/180) the clinical

geneticist would not have assessed the child positive if pictures would not have been available.

Hundred-seventy-one patients were evaluated by genetic consultation thus far. In the positive

assessed group three (3/145) patients were diagnosed with a TPS and in four (4/145) patients a

germline variant of non-conclusive significance was found. In 11 patients further analysis is still

ongoing. Out of the 135 negative assessed patients, 26 patients were referred as a negative control

group in which no TPS was diagnosed. CONCLUSION Preliminary results indicate the screening

instrument seems feasible in routine care. No false negative results were identified in the negative

control group, indicating that negative assessments by the screening instrument were justified.

E-mail: [email protected]

Keywords: Pediatric cancer, morphology, dysmorphology, 3D photography, screening instrument,

tumor predisposition syndrome

Page 66: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

65

P 17

An in-frame deletion in RIPOR2 is an important cause of

adult-onset hearing impairment

Suzanne E. de Bruijn,1,2,6 Jeroen J. Smits, 2,3,6 Chang Liu,4 Cornelis P. Lanting,3 Andy J. Beynon,3

Joëlle Blankevoort,1 Jaap Oostrik,2,3 Wouter Koole,1 Erik de Vrieze,2,3 DOOFNL Consortium, Cor

W.R.J. Cremers,3 Frans P. M. Cremers,1,2 Susanne Roosing,1,2 Helger G. Yntema,1,2 Henricus P.M.

Kunst,3,5 Bo Zhao,4 Ronald J.E. Pennings,2,3,7 Hannie Kremer1,2,3,7,*

1Department of Human Genetics, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands; 2Donders

Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands;

3Hearing & Genes, Department of Otorhinolaryngology, Radboud University Medical Center, 6500 HB, Nijmegen, The

Netherlands; 4Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, IN 46202,

Indianapolis, United States of America; 5Radboud Institute for Health Sciences, Radboud University Medical Center, 6500

HB, Nijmegen, 6,7 These authors contributed equally to this work

An in-frame deletion (c.1696_1707del) in RIPOR2 was identified to underlie dominantly inherited

nonsyndromic hearing impairment (DFNA21) in twelve families of Dutch origin. The age of onset

of the hearing impairment was in adulthood in the majority of the cases but ranged from childhood

to 70 years. Also the configuration of audiograms displayed a high variability. Ex vivo experiments

confirmed a functional effect of the variant as it resulted in aberrant localization of the mutant

RIPOR2 protein throughout the stereocilia of mechanosensory hair cells whereas the wildtype

protein was concentrated at the stereocilia base. Moreover, mutant RIPOR2 could not rescue the

morphological defects observed in RIPOR2-deficient hair cells, in contrast to the wildtype protein.

The variant is indicated to be a founder mutation and is present in one per 1,275 individuals of a

cohort from the South-East of the Netherlands. This suggests that the deletion is the most

important cause of monogenic hearing impairment in the Netherlands, with potentially ~8,600

affected individuals, and also a significant cause of hearing impairment in neighboring Western

European countries. The penetrance of the condition in the studied families is >90%. As this is

biased because of inclusion in the study of hearing impaired index cases, overall penetrance needs

to be further addressed. For adult-onset nonsyndromic hearing impairment the diagnostic yield of

genetic testing is low and decreases with an increasing onset age. RIPOR2 was already described

to be associated with recessive congenital hearing impairment in a single family. Our study

demonstrates that certain apparently 'mild´ variants in genes associated with recessive early-onset

hearing impairment can be an important cause of (late) adult-onset hearing impairment.

E-mail: [email protected]

Keywords: Hearing loss, dominant inheritance, adult-onset

Page 67: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

66

P 18

KIF1A variants are a frequent cause of autosomal dominant

hereditary spastic paraplegia

E. Kamsteeg1, M. Pennings1, M. Schouten1, R. Meyer1, J. van Gaalen1, S. T. de Bot2, M. Kriek2, C.

G. J. Saris1, L. H. van den Berg3, M. A. van Es3, D. M. H. Zuidgeest4, M. W. Elting5, J. M. van de

Kamp5, K. Y. van Spaendonck-Zwarts5, C. de Die-Smulders6, E. H. Brilstra3, C. C. Verschuuren7, B.

B. A. de Vries1, J. Bruijn8, K. Sofou9, B. P. van de Warrenburg1;

1Radboud University Medical Centre, Nijmegen, Netherlands, 2Leiden University Medical Center, Leiden, Netherlands,

3Utrecht University Medical Centre, Utrecht, Netherlands, 4Ikazia Hospital, Rotterdam, Netherlands, 5Amsterdam University

Medical Centre, Amsterdam, Netherlands, 6Maastricht University Medical Centre, Maastricht, Netherlands, 7Groningen

University Medical Centre, Groningen, Netherlands, 8Skaraborg Hospital, Skövde, Sweden, 9The Queen Silvia Children's

Hospital, Gotenborg, Sweden.

Background: Pathogenic variants in the KIF1A gene have been reported in autosomal recessive

spastic paraplegia 30, autosomal recessive hereditary sensory neuropathy, and autosomal (de novo)

dominant mental retardation type 9 . More recently, variants in KIF1A have also been described in

a few cases with autosomal dominant spastic paraplegia. Methods: Clinical exome sequencing data

of 315 unrelated patients with mostly 'pure´ spastic paraplegia were analysed for KIF1A variants.

Clinical characteristics of patients with KIF1A variants were analysed, segregation analyses were

performed, and types of variants were compared. Results: In 21 patients we identified 19 different

KIF1A variants. Patients and affected family members showed a slowly progressive, mostly pure

form of spastic paraplegia, but with a highly variable disease onset (0-57 years). Segregation

analyses showed a de novo occurrence in five cases, and a dominant inheritance pattern in eight

families. The motor domain of KIF1A seems to be the hotspot for pathogenic variants in autosomal

dominant spastic paraplegia, similar to mental retardation type 9 and recessive spastic paraplegia

type 30. Unlike these allelic disorders, however, dominant spastic paraplegia was also caused by

loss-of-function variants outside this domain in six families. Three missense variants were identified

outside the motor domain and need further characterization. Conclusions: KIF1A variants are a

frequent cause of autosomal dominant spastic paraplegia in our cohort (5-7%), with a high de novo

rate. The identification of KIF1A loss-of-function variants suggests haploinsufficiency as a possible

mechanism in autosomal dominant spastic paraplegia.

E-mail: [email protected]

Keywords: spastic paraparesis loss-of-function kinesin

Page 68: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

67

P 19

19q13.32 microdeletion syndrome: further delineation of the

clinical phenotype

L.J.C.M. van Zutven1, C.M.A. van Ravenswaaij-Arts2, T. Dijkhuizen2, M. McGregor-Schuerman3, L.H.

Hoefsloot1, J.J. Saris1, Y. van Bever1, M.I. Srebniak1

1 Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands, 2 Department of Clinical Genetics, UMCG,

Groningen, The Netherlands, 3 Department of Pediatrics, RKZ St. Vincentius, Paramaribo, Suriname

Background Interstitial 19q13.32 microdeletions are rare and have been reported in only five

patients so far. Common features mentioned in the literature include intellectual

disability/developmental delay, facial asymmetry, ptosis, oculomotor paralysis, orofacial clefting,

micrognathia, kyphoscoliosis, cardiac abnormalities and constipation,. Since only a few patients

have been reported, little is known about the phenotypic spectrum of these deletions. Methods To

expand the knowledge on interstitial 19q13.32 microdeletions, we report two new patients with a

de novo microdeletion in this region, and compare them with previously published patients. Results

Evaluation of the phenotypic features in the five published and the two new patients showed that

only developmental delay/intellectual disability was present in all patients. No cardiac

abnormalities, facial asymmetry, ptosis or micrognathia were present in our patients. Patient 1, with

a 1 Mb deletion 19q13.32q13.33, did not have oculomotor dysfunction, scoliosis or constipation,

and patient 2, carrying a 3.2 Mb deletion 19q13.31q13.33, did not have a cleft palate. New features

noted in patients with 19q13.32 microdeletions were ear abnormalities, including hearing loss,

down slanting palpebral fissures and, in male patients, hypospadias. Conclusions Major clinical

features seem to differ between the patients with 19q13.32 microdeletions reported so far. In

addition, some of the previously presented key features for this microdeletion are absent in our

patients. As a consequence, the syndrome may not be as recognizable as previously suggested.

Identification of other patients is necessary to further delineate the clinical phenotype of the

19q13.32 microdeletion and to establish the critical region/genes responsible for intellectual

disability in these patients.

E-mail: [email protected]

Keywords: 19q13.32 microdeletion

Page 69: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

68

P 20

NOTCH3 exon skipping as a rational therapeutic approach

for CADASIL: lessons from a family with naturally occurring

exon 9 skipping

Gido Gravesteijn, MD MSc1; Johannes G. Dauwerse2; Ingrid Hegeman3, Gwendolyn Brouwer, MSc2;

Aat A. Mulder4; Carolina R. Jost, PhD4; Sjoerd van Duinen3; Annemieke Aartsma-Rus, PhD2; Julie

W. Rutten, MD PhD1,2; Saskia A.J. Lesnik Oberstein, MD PhD1.

1 Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands. 2 Department of Human

Genetics, Leiden University Medical Center, Leiden, The Netherlands. 3 Department of Pathology, Leiden University Medical

Center, Leiden, The Netherlands. 4 Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The

Netherlands.

Background CADASIL is a small vessel disease (SVD), caused by cysteine altering missense

mutations in the NOTCH3 gene, which result in NOTCH3 protein aggregation in the

(cerebro)vasculature. We recently described a rational therapeutic approach for CADASIL, aimed at

counteracting NOTCH3 aggregation via antisense oligonucleotide (AON)-mediated skipping of

mutated NOTCH3 exons. We demonstrated the feasibility of this approach in vitro, but in vivo

experiments are so far hampered by inefficient delivery of the AONs. Here, we present clinical and

molecular characteristics of a family with a NOTCH3 cysteine altering mutation located on a splice

site, leading to natural exclusion of the mutation from the RNA via exon skipping. Methods All

family members had brain MRI. Skin punch biopsies were taken and processed for fibroblast culture,

immunohistochemistry and electron microscopy. Exon skipping was analysed using RT-PCR and

Sanger sequencing, on RNA isolated from skin fibroblasts. Results The index (female, 63 years) was

referred because of mild cognitive impairment and white matter hyperintensities on brain MRI

suggestive of SVD. Gene panel testing revealed a novel NOTCH3 mutation in exon 9 (c.1492G>T,

p.Gly498Cys). The mutation is located on the splice donor site of exon 9 and therefore was predicted

to result in exon 9 skipping. Four additional family members with the mutation were identified, all

with a relatively mild phenotype without lacunar strokes. RT-PCR and Sanger sequencing on

fibroblast RNA showed abundant presence of exon 9 skip product, but also low levels of mutant

NOTCH3 from which exon 9 was not skipped. Skin vessels showed only very minimal NOTCH3

aggregation and no granular osmiophilic material. Conclusions These individuals with naturally

occurring NOTCH3 exon skipping have a late-onset SVD, likely attributable to their low levels of

mutant NOTCH3 expression, which is consistent with the strikingly mild NOTCH3 aggregation in

their skin vasculature. These findings also suggest that the abundant skipped NOTCH3 does not

contribute to the SVD phenotype, supporting the potential feasibility of NOTCH3 exon skipping as

a therapeutic strategy in CADASIL.

E-mail: [email protected]

Keywords: CADASIL; NOTCH3; splice site variant; exon skipping; therapeutic strategy

Page 70: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

69

P 21

Identification of splice defects due to non-canonical splice

site or deep-intronic variants in ABCA4

Zeinab Fadaie1, Mubeen Khan1, Marta Del Pozo-Valero1,2, Stéphanie S. Cornelis1, Carmen Ayuso2,

[ABCA4 Studygroup: Rando Allikmets3, Miriam Bauwens4, Mohammad Ghofrani5, Michael B.

Gorin6,7, Mohammad Keramatipur8, Francesca Simonelli9, Naeimeh Tayebi10, Andrea Vincent11,

12, Nicole Weisschuh13] Frans P.M. Cremers1, Susanne Roosing1

1 Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center,

Nijmegen, the Netherlands. 2 Department of Genetics, Instituto de Investigación Sanitaria–Fundación Jiménez Díaz

University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain. 3 Department of Ophthalmology and

Department of Pathology and Cell Biology, Columbia University, New York, NY, USA. 4 Center for Medical Genetics, Ghent

University and Ghent University Hospital, Ghent, Belgium. 5 Cellular and Molecular Research Center, Qom University of

Medical Sciences, Qom, Iran 6 Department of Ophthalmology, David Geffen School of Medicine, Stein Eye Institute, University

of California, Los Angeles, Los Angeles, California, USA. 7 Department of Human Genetics, David Geffen School of Medicine,

University of California, Los Angeles, Los Angeles, California, USA. 8 Department of Medical Genetics, Tehran University of

Medical Sciences, Tehran, Iran. 9 Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences,

University of Campania Luigi Vanvitelli, Naples, Italy. 10 Program in Genetics and Genome Biology, The Hospital for Sick

Children, Toronto, Canada 11 Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and

Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand. 12 Eye Department, Greenlane Clinical

Centre, Auckland District Health Board, Auckland, New Zealand. 13 Molecular Genetics Laboratory, Centre for

Ophthalmology, Institute for Ophthalmic Research, University of Tuebingen, Tuebingen, Germany.

Pathogenic variants in the ATP-binding cassette transporter A4 (ABCA4) gene cause a continuum

of retinal disease phenotypes, including Stargardt disease (STGD1). Non-canonical splice site

(NCSS) and deep-intronic variants constitute a large fraction of disease-causing alleles, defining the

functional consequences of which remains a challenge. We aimed to determine the effect on

splicing of nine previously reported or unpublished NCSS variants, one near exon splice variant and

nine deep-intronic variants in ABCA4, using in vitro splice assays in Human Embryonic Kidney

(HEK293T) cells. Reverse transcription PCR and Sanger sequence analysis revealed splicing defects

for 12 out of 19 variants. Four deep-intronic variants create pseudoexons or elongate the upstream

exon. Furthermore, eight NCSS variants cause a partial deletion or skipping of one or more exons

in mRNAs. Among the 12 variants, nine lead to premature stop codons and predicted truncated

ABCA4 proteins. At least two deep-intronic variants are located in splice enhancers and silencers

and, therefore, these conserved sequences should be carefully evaluated when predicting the

outcome of NCSS and deep-intronic variants.

E-mail: [email protected]

Keywords: ABCA4, Non-canonical splice site variant; deep-intronic variants; splice enhancers;

splice silencers; Stargardt disease

Page 71: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

70

P 22

A novel missense PLP1 mutation causing PLP1-related

spastic paraplegia: a Case Report

Eva Sammels, Nathalie Fieremans, Annelies Fieuw, Elyssa Cannaerts, Ann Van den Bogaert, Elise

Vantroys, Melissa Sys, Kathelijn Keymolen, Boyan Dimitrov

Center for Medical Genetics, UZ Brussels-Vrije Universiteit Brussel

A 12-year old boy presented with lower limb spasticity (first signs by the age of 4), mild facial

dysmorphism (downslanting palpebral fissures) and learning difficulties. There was no particular

familial history, besides a maternal niece with club foot. The patient underwent physiotherapy on a

regular basis during infancy. Array CGH analysis was performed as a first-tier test, but it returned

normal. Subsequently, a neurodevelopmental disorders gene panel (1160 genes) was sequenced in

trio (parents and patient). We detected a novel, hemizygous, likely pathogenic PLP1 missense

mutation (NM_000533.3(PLP1):c.263C>T, p.(Ala88Val)), inherited from the presumably

asymptomatic mother, compatible with PLP1-related spastic paraplegia (X-linked recessive

inheritance). Carrier females occasionally develop mild to moderate signs of the disease, especially

in families with mildly affected males. Therefore, careful clinical follow-up is advised for the

heterozygous mother (and if needed other maternal relatives). This missense mutation seems to

reside within one of the mutational hotspots of the PLP1 gene. Of interest, a number of other

neighbouring amino acid changes and another missense mutation affecting the same codon

(p.(Ala88Asp)) have already been described as pathogenic. The highly conserved Ala88 residue is

located in the transmembrane helix TM2. Mutations affecting this residue are predicted to affect

the organisation of the four transmembrane helices of PLP1, thereby interfering with its biological

function as the primary component of myelin. Patients carrying PLP1 mutations affecting the same

and other nearby residues were reported to have clinical heterogenous phenotypes and different

degrees of severity, making clear genotype-phenotype correlations for PLP1-related disorders still

puzzling. Methodology: Array CGH analysis on DNA isolated from an EDTA blood sample was

performed using a CytoSure Constitutional v3 60k array. Version (v)5 of the UZ Brussel

neurodevelopmental disorders gene panel (NEUDGP) contains the coding regions and 11 basepair

(bp) flanking intronic sequence of 1160 genes. The complete gene list is available on the

BRIGHTcore website: http://www.brightcore.be/mendeliome-neurodev. Clinical follow-up was

done in collaboration with the corresponding clinicians.

E-mail: [email protected]

Keywords: PLP1 - Spastic paraplegia - Pelizaeus-Merzbacher disease - Neurodevelopmental

disorders

Page 72: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

71

P 23

LOVD - supporting DNA variant interpretation

Ivo FAC Fokkema, Julia A Lopez Hernandez, Johan T den Dunnen

Leids Universitair Medisch Centrum

The simplest and cheapest answer to the possible consequences of a variant identified in a patient's

DNA is history: has the variant been seen before, and what were the associated consequences? The

"Global Variome shared LOVD" database builds on a >20 year history of collecting and sharing

information on Individuals (patients), Phenotypes (disease/traits), Screenings and Variants. While it

contains information from published literature, a significant fraction of all data has not yet been

published and was submitted to LOVD only. When performing DNA diagnostics, it is essential to

check LOVD, preventing easily available information to be missed. A growing number of human

genetics journals has made submission of variant/phenotype data to LOVD or ClinVar mandatory

before a manuscript will be accepted for publication. This brings additional advantages, like a free

data quality check and the option to refer to the database as an alternative for supplementary files.

All public LOVD databases together contain information of >6.5 million unique variants linked to

>1,000,000 individuals, and can be queried in many ways including through the LOVD website, the

API, or the GA4GH beacon network. For genes with active curators (incl. BRCA, colon cancer, CFTR,

etc.) specific "Summary records" resume all supporting information whether the variant is or is not

associated with a disease. "Classification records" show the classification regarding variant

pathogenicity as shared by diagnostic labs (incl. data from the Dutch VKGL). Records labelled "In

vitro (cloned)" show the results of assays performed to analyse the functional consequences of

variants. Other features that distinguish LOVD from databases like ClinVar, include data collection

on the geographic origin of the Individual, detailed Phenotype information and showing whether

the variant has been analysed on RNA level and what the consequences were; essential information

for the diagnosis in diseases like Duchenne/Becker muscular dystrophy. LOVD shows the parental

origin of a variant and, when available, full haplotype information. The GV shared LOVD is a

community driven initiative operating under the auspices of Global Variome, a UK charity. Each

gene´s home page links to other resources and the major genome browsers. The data is shared

with public repositories incl. UCSC, EBI and ClinVar, and copies can be downloaded from the gene

home pages. URLs: LOVD.nl/shared, LOVD.nl, LOVD.nl/3.0/search

E-mail: [email protected]

Keywords: LOVD, variant databases, diagnostics

Page 73: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

72

P 24

SETD1A Loss of function causes a distinct

neurodevelopmental disorder in human and impaired

learning & memory in Drosophila.

Joost Kummeling1* Diante E Stremmelaar1* Nicholas Raun2, 20 Margot RF Reijnders3 Marjolein H

Willemsen1 Martina Ruiterkamp-Versteeg1 Marga Schepens1 Calvin CO Man1 Christian Gilissen1

Megan T Cho4 Kirsty McWalter4 Margje Sinnema3 James W Wheless5 Marleen EH Simon6 Casie A

Genetti15,18 Alicia M Casey19 Paulien A Terhal6 Jasper J van der Smagt6 Koen. L. van Gassen6

Pascal Joset7 Angela Bahr7 Katharina Steindl7 Anita Rauch7 Elmar Keller8 Annick Raas-Rothschild9

David A Koolen1 Pankaj B Agrawal10, 15, 18 Trevor L Hoffman11 Nina N Powell-Hamilton12 Isabelle

Thiffault13 Kendra Engleman14 Dihong Zhou14 Olaf Bodamer15 Dirk Schubert21 Rolph Pfundt1

Jamie M Kramer2, 16, 17 Tjitske Kleefstra1

1. Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud university medical center,

P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands. 2. Division of Genetics and Development, Children's Health Research

Institute, London, Ontario, Canada. 3. Department of Clinical Genetics and School for Oncology & Developmental Biology

(GROW), Maastricht University Medical Center, Maastricht 6229 ER, the Netherlands. 4. GeneDx, Gaithersburg, MD 20877,

USA. 5. Division of Pediatric Neurology, University of Tennessee Health Science Center, , Memphis, TN, United States of

America. . Neuroscience Institue & Le Bonheur Comprehensive Epilepsy Program, Le Bonheur Children's Hospital, Memphis,

TN, United States of America. 6. Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The

Netherlands. 7. Institute of Medical Genetics, University of Zurich, Schlieren-Zurich, 8952, Switzerland. 8. Division of

Neuropediatrics, Cantonal Hospital Graubuenden, Chur, Switzerland. 9. Institute of Rare Disease, Danek Gertner Institute of

Human Genetics, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv,

Israel. 10. Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.

11. Regional Department of Genetics, Southern California Kaiser Permanente Medical Group, 1188 N. Euclid Street, Anaheim,

CA 92801, USA 12. Division of Medical Genetics, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA. 13.

Department of Pathology and Laboratory Medicine; Center for Pediatric Genomic Medicine, Children's Mercy Hospital;

University of Missouri-Kansas City School of Medicine. 14. Division of Clinical Genetics, Children's Mercy Hospital;

Department of Pediatrics, Children's Mercy Hospital; University of Missouri-Kansas City School of Medicine. 15. Division of

Genetics and Genomics, Department of Medicine, Boston Children's Hospital/Harvard Medical School, Boston,

Massachusetts, USA. 16. Department of Biology, Faculty of Science, Western University, London, Ontario, Canada. 17.

Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London,

Ontario, Canada. 18. The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical

School, Boston, MA, 02115, USA. 19. Division of Pulmonary and Respiratory Diseases, Boston Children's Hospital and Harvard

Medical School, Boston, MA, 02115, USA. 20. Department of Biology, Faculty of Science, Western University, London, Ontario,

Canada. 21. Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud

University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands

Defects in histone methyltransferases (HMTs) are a major contributing factor to

Neurodevelopmental disorders (NDDs). Loss-of-function (LoF) variants of SETD1A (KMT2F), one of

the genes involved in histone H3 lysine 4 (H3K4) methylation and thus in epigenetic modification,

have been identified in several individuals from large schizophrenia cohort studies. Interestingly,

dominant gene mutations were also identified in children with developmental delay. To provide

further insight in the somatic and behavior profiles, we further characterized the SETD1A associated

Mendelian syndrome by collecting the molecular and clinical features of 15 so far unpublished

individuals with SETD1A variants via a GeneMatcher collaboration. Furthermore, to gain insight into

the potential role of SETD1A in fundamental learning and memory processes, we studied the

behavioral consequences of a Drosophila knock down of the orthologue Set1. The mutations

identified in the patients mainly comprised of de novo variants that predict a loss of function with

Page 74: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

73

c.4582-2_4582delAG being a recurrent mutation resulting in intron retention instead of the

presumed exon skipping. Haploinsufficiency of SETD1A leads to a set of symptoms comprising

global developmental delay or intellectual disability, subtle facial dysmorphisms, behavior and

psychiatric problems. In Drosophila, knockdown of Set1 in postmitotic memory neurons resulted in

significantly reduced short- and long-term courtship memory. Together, this study defines a distinct

neurodevelopmental disorder caused by dominant LoF mutations in the SETD1A and suggests

further support for the important role of H3K4 methyltransferases in memory, developmental

disability and neuron function in general.

E-mail: [email protected]

Keywords: SETD1A; histone methylation; intellectual disability syndrome; behavioral phenotype;

Drosophila Set1 orthologue

Page 75: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

74

P 25

Towards comprehensive identification and functional

characterization of deep-intronic ABCA4 variants in 1000

Stargardt disease cases

Khan, M.,1,2 Cornelis, S.S.,1,2 del Pozo-Valero, M.,1,3 Khan, M.I.,1,2 Stöhr, H.,4 Grassmann, F.,5

Steehouwer, M.,1 Hoischen, A.,1 Ayuso, C.,3 Ramesar, R.,6 Meunier, I.,7 Defoort, S.,8 Weber, B.H.F.,4

Dhaenens, C-M.,9 and Cremers, F.P.M.1

1Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands 2Donders Institute for

Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands 3Department of Genetics,

IIS-Fundación Jiménez Díaz, CIBERER, Madrid, Spain 4Institut für Humangenetik, Universität Regensburg, Regensburg,

Germany 5Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 6Division of Human

Genetics, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa 7Institut

des Neurosciences de Montpellier, INSERM, Université de Montpellier, Montpellier, France. 8Service des explorations de la

fonction visuelle, CHRU de Lille, Lille, France. 9University Lille, Inserm UMR-S 1172, CHU Lille, Biochemistry and Molecular

Biology Department - UF Génopathies, Lille, France

Purpose: Although the gene underlying Stargardt disease (STGD1), ABCA4, has been identified 22

years ago, for many cases the underlying mutations are unknown. This in part is due to the intronic

location of many causal variants. In this study, we aim to develop a cost-effective whole ABCA4

gene sequencing technique to analyze >1000 genetically unsolved STGD1 probands. Methods: We

designed 3866 single molecule molecular inversion probes (smMIPs), each capturing 110 nt of the

128-kb ABCA4 gene proper or ~40 kb of flanking sequences. Sense and antisense strands were

targeted using overlapping smMIPs. Rebalancing of the smMIPs spanning the coding sequences

was performed. We collected STGD1 probands from 21 collaborators worldwide. NextSeq 500

sequence analysis was performed for 20 test cases (16 STGD1, 4 healthy controls) and 200 probands

in each run. Novel deep-intronic variants were tested for splice defects using in vitro splice assays

employing ABCA4 midigenes. Results: smMIPs were designed for 99% of the ABCA4 gene proper

and ~50% of flanking sequences as repeats outside the gene were refractory to smMIPs design.

Sequence analysis of 16 previously genetically solved STGD1 patients revealed all known alleles

with an average smMIP coverage of 933x. Single or double coverage (>100 reads) was achieved for

98.5% of the ABCA4 gene proper and all reported 17 causal deep-intronic variants were covered

with smMIPs. We collected 1000 genetically unsolved STGD1 samples, ~900 of which previously

were scanned for coding variants and ~100 probands lacked genotyping. In the first 200 STGD1

cases analyzed in a single run, the average smMIP coverage was 377x. We identified 7 different

known deep-intronic variants in 23 alleles and 27 different novel deep-intronic variants in 55 alleles.

The latter are being tested using midigene-based splice assays. Conclusions: Due to the paucity of

sizeable repeats, the introns of ABCA4 could readily be captured using smMIPs. Apart from design

and synthesis costs of smMIPs, the sequencing costs for the entire ABCA4 gene are USD25 per

sample, rendering this the most cost-effective ABCA4 sequence analysis for coding and non-coding

sequences. We will generate a comprehensive list of causal deep-intronic variants for ABCA4, which

then can be used to design a minimal diagnostic set of smMIPs that capture 95% of causal ABCA4

alleles in each patient.

E-mail: [email protected]

Keywords: ABCA4, Stargardt disease, smMIPs, deep-intronic variants and midigene

Page 76: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

75

P 26

Compound heterozygosity for missense and null variants in

CEP55 lead to a novel viable phenotype

M. Mahdi Motazacker*, Eline Overwater*, M. Mieke van Haelst

1- Amsterdam UMC, University of Amsterdam, Laboratory of Genome Diagnostics, Meibergdreef 9, Amsterdam, Netherlands

2- Amsterdam UMC, University of Amsterdam, Department of Clinical Genetics, Meibergdreef 9, Amsterdam, The

Netherlands. *These authors contributed equally to this work

The centrosomal protein-55 kDa (CEP55) gene encodes an essential regulator of the final stages of

mitosis during cytokinesis. So far only a few cases with homozygosity for loss-of-function variants

in CEP55 are reported in the literature. These cases present with an autosomal recessive perinatal

or neonatal lethal phenotype similar to Meckel-like syndrome or MARCH syndrome (multinucleated

neurons, anhydramnios, renal dysplasia, cerebellar hypoplasia and hydranencephaly and

dysmorphic features; MIM 236500). Here we report a 40-year-old male born at a gestational age of

38 weeks with mild intellectual disability, developmental delay, microcephaly, bilateral toe

syndactyly, bilateral hypoplastic fifth fingers with clinodactyly and left-sided congenital ptosis. Brain

magnetic resonance imaging (MRI) at the age of 37 years showed no abnormalities except from

nonspecific white matter changes. Whole exome sequencing (WES) revealed compound

heterozygosity for a nonsense c.1274C>A p.(Ser425*) and a missense c.70G>A p.(Glu24Lys) variant

in CEP55 (NM_018131.4). A subsequent enquiry through GeneMatcher network identified multiple

cases with similar combination of variants and a similar non-lethal phenotype. We conclude that

phenotypic spectrum of CEP55 pathogenic variants expands to viable phenotypes with multiple

congenital anomalies. Exome sequencing is expanding the phenotypic spectrum of many disease

causing genes. This should be considered especially when interpreting variants in genes for which

only a few cases with specific class of pathogenic variants are reported.

E-mail: [email protected]

Keywords: MARCH syndrome, CEP55, Genotype-phenotype correlation, WES

Page 77: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

76

P 27

Clinical characteristics of patients with an STXBP1 gene

mutation: preliminary results of a cross-sectional and

retrospective multilevel cohort study

Bos, J.1, Misra-Isrie, M.1, Van Berkel, A. 1,2, Lammertse, H.1,2, Engelen, M.3, Meijers-Heijboer, H.1,

Van Haelst, M.1 and Verhage, M.1,2

1Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Genetics, De Boelelaan 1117, Amsterdam,

Netherlands 2Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam

Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1078, Netherlands 3Amsterdam UMC, University of Amsterdam,

Department of Pediatric Neurology/Emma Children's Hospital, Meibergdreef 9, Amsterdam, Netherlands

STXBP1-encephalopathy is caused by heterozygous mutations in the STXBP1 gene, which encodes

Syntaxin binding protein 1 (STXBP1), also known as Munc18-1. STXBP1 regulates synaptic

transmission by organizing the protein complex that drives synaptic vesicle exocytosis.

Haploinsufficiency of STXBP1 results in a neurodevelopmental disorder, of which the first patients

were reported in 2008, all diagnosed with Ohtahara syndrome. Since then, many case reports and

small cohort studies were reported and the corresponding phenotype has expanded, including

development delay and different forms of epilepsy. A systematic assessment of the phenotypes of

STXBP1-encephalopathy has not been performed thus far and phenotypic features might have

been underreported. The purpose of the current study is to provide a systematical and multilevel

overview of characteristics of STXBP1-encephalopathy patients worldwide. Two clinic days were

organized, during which a semi-structured interview and physical examination by two clinical

geneticists were performed, a neurological examination was performed by a neurologist and a

questionnaire was filled in by parents. Medical records were collected afterwards. The multilevel

approach was complemented by EEG recordings, three-dimensional photographs and skin biopsies

for generation of a cellular in vitro model of STXBP1-encephalopathy. Descriptive statistics were

used to identify common characteristics. Statistical analysis was performed to assess the difference

in prevalence of epilepsy in the current cohort and in literature. Eight of the 13 patients suffered

from epilepsy; a significantly lower proportion than in literature (p 0.002). A high prevalence of

neurological symptoms was observed; tremor and hypotonia were most frequently observed.

Autistic features were observed in 60% of the patients. A characteristic leg cross was recognized by

the parents as (possibly) a typical feature for STXBP1-encephalopathy. Common dysmorphic

features included fetal finger pads and an abnormal shape of the upper lip. Impaired speech-

and/or motor performance was observed in all patients. These results confirm that STXBP1-

encephalopathy is a heterogeneous neurodevelopmental disorder in which epilepsy is not

obligatory, but development delay is invariably observed and the presence of a characteristics leg

cross might be typical. Subtle dysmorphic features and behavioral characteristics such as autistic

features might have been underexposed thus far.

E-mail: [email protected]

Keywords: STXBP1-encephalopathy, STXBP1, Munc18-1, neurodevelopmental disorder,

development delay, intellectual disability, epilepsy, epileptic encephalopathy

Page 78: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

77

P 28

The impact of incidental findings in clinical exome

sequencing, a qualitative interview study

Simone J. Viellevoije2, Femke Tammer3, Ilse Feenstra3, Lisenka E.L.M. Vissers3, Martine van

Koolwijk3 , Han G. Brunner1,3, Yvonne H. J. M. Arens1, Helger G. Yntema3 and Anke J.M.

Oerlemans2

1 Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands; 2 Sscientific Institute

for Quality of Healthcare, Radboud Institute for Health Sciences, Radboud University Nijmegen Medical Centre, Nijmegen;

3 Department of Human Genetics, Radboud University Nijmegen Medical Centre Nijmegen, The Netherlands

Introduction Incidental findings in clinical exome sequencing are (likely) pathogenic variants in

disease genes not related to the initial clinical question, but which could be of medical relevance

to the patients and their families. The management and disclosure of IFs is an internationally

recognised topic of debate. Current recommendations are based upon multiple perspectives

(scientific, ethical, legal, clinical). However, little is known about the impact of IF disclosure on

patients. Materials and methods We conducted ten (20 by October; study ongoing) semi-structured

face-to-face interviews with patients and/or their relatives who underwent genetic testing and to

whom an oncological (n=5) or a cardiological (n=5) IF was disclosed. Results Using thematic analysis

we identified four relevant themes: psychological impact, actionability, comprehension and social

context. Participants described the psychological impact being influenced by the patients´ prior

health condition and the availability of preventive measures. They frequently mentioned the

mitigating nature of actions taken (i.e. cardiological screening, prophylactic surgery) after the

disclosure. Preventive measures and screening options for oncogenetic conditions seemed to

assure a more definite answer to the frequently asked question 'am I sick or am I healthy?´. Difficulty

of comprehending the actual health risk was a recurrent topic. Some participants took future health

risks into account. Patients´ relationships with relatives and the lives of family members did not

seem to be greatly influenced by disclosure of IF. Discussion and conclusion The prior health

condition of the patient appears to influence the impact of incidental findings in clinical exome

sequencing. For patients, the benefit of a diagnosis seems to outweigh the risk of the disclosure of

an IF, noteworthy when preventive measures are available. In this context, we see a difference in

impact between oncological IF and cardiological IF. These results contribute to the ongoing

international debate about managing IFs in clinical DNA sequencing.

E-mail: [email protected]

Keywords: incidental findings, impact, qualitative interview study

Page 79: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

78

P 29

CAPICE - a variant prioritization tool for Consequence-

Agnostic PrIoritization of Clinical Exomes variations

KJ van der Velde, D. de Ridder, A.D.J. van Dijk, D. Soudis, L.R. Zwerwer , M.A. Swertz

1Genomics Coordination Center, Dept of Genetics, University Medical Center Groningen, Groningen, The Netherlands

2Bioinformatics Group, Wageningen University & Research, Wageningen, The Netherlands 3The Center for Information and

Technology, University of Groningen, Groningen, The Netherlands

The past decades have seen rapid advances in sequencing techniques. However, current

interpretation methods determining the pathogenicity or benigness of variants are often too time-

consuming and of limited accuracy, even with the assistance of computation pathogenicity

estimation tools. We here present CAPICE, that has 2x the precision of the next-best variant

classification tool by equal recall of 95%. CAPICE overcomes issues that are common in the great

number of predictors already built for this purpose, several issues exist. Firstly, a lot of predictors

are built specifically for only a subset of SNVs (single nucleotide variants), in particular, non-

synonymous variants. This creates difficulties in applications where variants of different molecular

functions can be the causal variant. Secondly, the general predictors that do give predictions to all

types of variations, such as CADD, often perform much worse compared to specific tools. Moreover,

there is a lack of research examining the predictors´ performance in a general setting where variants

of different molecular functions are considered and rare neutral variants are included. Therefore, to

assist the existing clinical protocols in interpreting patients´ genetic profile the challenge is to

extract all relevant information from different biological databases and integrate those into a robust

classification algorithm to classify all benign and pathogenic variants. CAPICE uses an ensemble

method for prioritizing pathogenic variants for Mendelian disorders in clinical exomes, including

SNVs and short inserts and deletions (InDels). By training and fine-tuning a sophisticated machine

learning model on a balanced large dataset collected from multiple public sources, we showed that

the resulted model outperforms both the specific predictors and general predictors in

distinguishing between pathogenic variants and rare neutral variants. Additionally, we showed that

CAPICE performs consistently good for variants displaying different molecular consequences and

allele frequency. Moreover, we provided suggestions for threshold selection when making

pathogenicity predictions based on CAPICE scores, and showed that the prediction made based on

CAPICE´s scores is of higher precision compared with existing tools for genes involved in different

organ systems. The method for selection of thresholds for making predictions based on the CAPICE

scores can be determined to suit different needs of user cases.

E-mail: [email protected]

Keywords: Variant prioritization, machine learning, molecular consequence, allele frequency,

SNVs, InDels

Page 80: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

79

P 30

Targeted compound screening for PXE using a CRISPR/Cas9

abcc6a mutant zebrafish model.

M. Van Gils; A. Willaert; P. Coucke; O.M. Vanakker

Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium Department of Biomolecular Medicine, Ghent

University, Ghent, Belgium

Introduction: Pseudoxanthoma elasticum (PXE) is an ectopic mineralization disease due to biallelic

ABCC6 mutations. As no curative therapy is available, disease models for rapid compound screening

are necessary. We characterized a zebrafish abcc6a CRISPR/Cas9 knockout model (Cmg52),

identifying spinal hypermineralization as an early, quantifiable phenotypic read-out. In this study,

we evaluated 3 established anti-mineralization compounds - Vitamin K1 (VK1), Etidronate (Etid) and

Sodium Thiosulfate (STS) - for their efficacy in PXE.

Materials & Methods: Following Cmg52+/- incross, Cmg52+/+, Cmg52+/-and Cmg52-/- embryos

were collected. At 3 days post-fertilization (dpf), embryos are randomly distributed per 20.

From 3-10dpf, 60-100 embryos are treated with 8ml compound in E3-medium, refreshed daily.

Based on literature we chose a dosage of 80 µM VK1 and 100µM Etid. Using LC50 analysis we opted

for 30µM STS.

After euthanasia at 10dpf, larvae are fixed, bleached and stained for mineralization. Mineralization

is semi-quantified via ImageJ. After sequencing, mineralization values are linked to genotypes for

statistical analysis (student t-test).

Results: Analysis of Cmg52+/+ and Cmg52+/- data sets revealed no significant changes in spinal

mineralization between treated and control cohorts. However, all STS-treated larvae had no spinal

mineralization and in 33% ventral mineralization foci were observed, regardless of genotype,

indicating putative toxicity. Reducing the dosage to 20µM STS abolished these effects.

In contrast, all compounds significantly (P<0.05) reduced spinal mineralization in Cmg52-/- larvae.

VK1 reduced mineralization by approximately 47% (Mean±SD: Control: 100±66; VK1: 53.5±28.5),

while Etid reduced by 33% (Mean±SD: Control: 100±58.5; Etid: 67±42) and 20µM STS by 41%

(Mean±SD: Control: 100±61; STS:59±35).

Discussion & Conclusion: We demonstrate the validity of our compound screening assay, with

Cmg52-/- spinal hypermineralization as a good read-out. Our data affirm Etid effects in PXE.

Interestingly, contrary to the murine model but similar to PXE patients, it also highlights a role for

VK1 in PXE. The significant effects of STS make it a potentially interesting topical treatment, e.g. for

the skin mineralization in PXE. However, the STS data also underscore the importance of in-depth

dosage analyses. Overall, our zebrafish chemical compound screening assay presents a powerful

new tool for rapid investigation of putative therapeutic drugs for PXE..

E-mail: [email protected]

Keywords: PXE, Zebrafish, CRISPR/Cas9, Compound Screening

Page 81: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

80

P 31

Whole-exome sequencing identifies heterozygous stop-loss

mutations in HBB resulting in an elongated β-globin chain in

two children with severe anemia.

TT Koopmann, GWE Santen, Q Waisfisz, M Elting, FJ Smiers, JL Kerkhoffs, PJ de Pagter, CL Harteveld

1. Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands. 2. Department of Clinical

Genetics, VU University Medical Center, Amsterdam, Netherlands. 3. Department of Pediatric Hematology, University

Medical Center Leiden, Leiden, The Netherlands. 4. Haga Hospital, The Hague, The Netherlands.

Background: ß-Thalassemia is a genetic disease characterized by reduced or absence of ß-globin

gene (HBB) expression. Some mutations in HBB may produce unstable, abnormal hemoglobins

causing (haemolytic) anemia or dominantly inherited ß -thalassemia. Methods: Two unrelated

children presented with macrocephaly and severe congenital anemia. They required stem cell

transplantations at ages 2 and 4 without clear diagnosis. Since the parents of both probands had

no family history of anemia or abnormal Hb, thalassemia was unsuspected. As genetic testing for

variants in genes associated with congenital anemia did not result in an explanation for the

phenotype, whole-exome sequencing was performed. Results: We identified two novel, de novo

missense stop-loss mutations in HBB (c.442T>C and c.442T>A). The mutations identified caused

the loss of a stop codon and an elongation of the translated beta-globin chain of 21 amino acids

(167 a.a. instead of the usual 146) due to a new stop codon in the 3' untranslated region (3'UTR) of

the HBB gene. The predicted ß-globin chains were p.(*148Glnext*21) or p.(*148Lysext*21); the latter

variant was called HbMokum. Discussion: Using whole-exome sequencing, we identified a de novo

mutation in two previously undiagnosed children with severe anemia. Both children carry an almost

identical heterozygous stop-loss mutation in HBB resulting in an elongated ß-globin chain, which

most likely produces an unstable hemoglobin. Frameshift mutations in the HBB gene resulting in

elongated ß-globin chain have been described before, but resulted in shorter ß-chains (max. 157

a.a.) and milder phenotypes than the mutations described here.

E-mail: [email protected]

Keywords: Thalassemia, stop-loss mutation, anemia, HBB gene, whole exome sequencing

Page 82: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

81

P 32

A second family with homozygous mutation in KIDINS220

presenting as congenital hydrocephalus and limb

contractures

Jacquemin V., Antoine M., Duerinckx S., Perazzolo C., Cassart M., Thomas D., Segers V., Drielsma A.,

Desir J., Pirson I., Abramowicz M.

IRIBHM, Université Libre de Bruxelles; Centre Hospitalier Etterbeek-Ixelles, Université Libre de Bruxelles; CHU Brugmann ;

Institut de Pathologie et de Génétique, Gosselies; Department of Genetic Medicine and Development, University of Geneva

Medical School

Background Hydrocephalus is a devastating, potentially lethal neurological condition. A subgroup

with prenatal onset and no apparent cause is called congenital primary hydrocephalus, affecting

1/10,000 newborns. A homozygous mutation in KIDINS220, a large scaffolding protein, was recently

reported in a single family with enlarged cerebral ventricles and limb contractures in four fetuses

(Mero et al 2017, Hum Mol Genet). Heterozygous, de novo mutations of KIDINS220 were also

reported in SINO (spastic paraplegia-intellectual disability-nystagmus-obesity) syndrome. Case

presentation We report a consanguineous family with 3 fetuses presenting with ventriculomegaly

and limb contractures. Exome sequencing revealed as sole potentially disease-causing variant, a

small homozygote in-frame deletion in KIDINS220 present in the three fetuses, resulting in the

deletion of Glutamine 713, Valine 714, and Leucine 715; p.Gln713_Leu715del. This mutation was at

heterozygous state in parents and unaffected siblings, they presented no symptoms of SINO

syndrome. These amino acids are very conserved between species and lie in the most highly

conserved stretch of the protein downstream of the fourth transmembrane domain. This region is

implicated in the interaction with tyrosine receptor kinase TrkA. We co-expressed the mutated

region and the TrkA receptor in a human cell line, and tested their interaction through

immunoprecipitation experiments. The wild-type and mutated fragments produced the same

results. We are now testing the full-length protein. Discussion and Conclusions The co-segregation

in three consanguineous fetuses and the location of the mutation strongly suggest that we

identified the second family with a homozygous, presumably loss-of-function mutation of

KIDINS220, confirming its clinical presentation as a syndrome of congenital hydrocephalus and limb

contractures.

E-mail: [email protected]

Keywords: primary congenitale hydrocephalus, research, exome sequencing, KIDINS220

Page 83: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

82

P 33

Intrinsic myogenic potential of skeletal muscle-derived

pericytes from patients with myotonic dystrophy type 1

Ausems Cornelia Rosanne Maria 1,2,3, Raaijmakers Renée Henrica Lamberta 1,2,3, Van Den Broek

Walterus Johannes Antonius Adriana 3, Willemse Marieke 3, Van Engelen Baziel Gerardus Maria2*,

Wansink Derick Gert3*, Van Bokhoven Hans1*

1. Radboud university medical center, Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics,

6500 HB, Nijmegen, The Netherlands 2. Radboud university medical center, Donders lnstitute for Brain Cognition and

Behavior, Department of Neurology, 6500 HB, Nijmegen, The Netherlands 3. Radboud university medical center, Radboud

Institute for Molecular Life Sciences, Department of Cell Biology, 6500 HB, Nijmegen, The Netherlands

Introduction: We are exploring the possibility of a cell-based therapy to combat muscle problems

in DM1. Accordingly, we have successfully isolated a distinct class of myogenic progenitors, called

pericytes, from skeletal muscle of DM1 patients and transgenic mice. Methods: Muscle biopsies

from DMSXL mice and DM1 patients were cultured under conditions to promote outgrowth of

pericytes, the muscle stem cells wrapped around blood vessels. ALP+/CD31- pericytes were isolated

by flow cytometry and used for characterization of gene expression, cell growth and myogenic

fusion characteristics. Results and Conclusions: Efficient isolation of ALP+/CD31- pericytes was

confirmed by RT-PCR analysis and immunocytochemistry. Expression of expanded DMPK RNA

matched the occurrence of nuclear MBNL foci. Interestingly, patient pericytes maintained myogenic

potential and differentiated efficiently into multinucleated myotubes in vitro. These muscle

progenitors will now be used in CRISPR/Cas9-mediated gene editing to excise the expanded (CTG)n

repeat. Grant Support: Donders Institute, Radboud University Medical Centre.

E-mail: [email protected]

Keywords: cell therapy; mesoangioblast; muscle stem cell; muscular dystrophy; myogenic

progenitor cell; myotonic dystrophy; pericyte; RNA toxicity; triplet repeat expansion;

Page 84: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

83

P 34

Development and Characterization of Pericyte-Derived iPSCs

and PiPs from DM1 patients

Renée H. L. Raaijmakers1,2,3, C. Rosanne M. Ausems1,2,3, Walther J. A. A. van den Broek3, Marieke

Willemse3, Silvia Albert1, Baziel G. M. van Engelen2, Derick G. Wansink3, Hans van Bokhoven1

1 Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center,

Nijmegen, The Netherlands. 2 Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud

university medical center, Nijmegen, The Netherlands. 3 Department of Cell Biology, Radboud Institute for Molecular Life

Sciences, Radboud university medical center, Nijmegen, The Netherlands.

Introduction: We aim to develop a muscle-focused iPSC-based cell therapy for DM1, using

pericytes, the myogenic progenitors wrapped around blood vessels. To ensure an unlimited source

of cells, we will generate pericyte-derived iPSCs. Subsequently, the pathogenic trinucleotide repeat

will be excised from these cells via CRISPR/Cas9-mediated gene editing. Corrected pericyte-derived

iPSCs can then be differentiated to PC-iPSC-derived pericyte-like cells (PiPs), which have shown to

maintain their myogenic potential and have restorative potential. Methods: Skeletal muscle biopsies

were taken from quadriceps muscle from DM1 patients and cultured for pericyte proliferation and

isolation. Cells were sorted for ALP+/CD31- expression by flow cytometry, expanded in vitro, and

transfected with non-integrating episomal vectors containing the Yamanaka factors. Differentiation

of PC-iPSCs to PiPs has been performed following a protocol by Maffioletti et al. (2015, Nature

Protocols) by the use of media components clear from transgene integration. Results: We

successfully isolated ALP+/CD31- pericytes and were able to generated pericyte-derived iPSCs.

Furthermore, preliminary results point to successful differentiation of PC-iPSCs to PiPs. Conclusions:

We are currently applying CRISPR/Cas9-mediated repeat removal on PC-iPSCs and are optimizing

the differentiation protocol for PiPs. Grant Support: Prinses Beatrix Spierfonds [grant W.OR18-18]

E-mail: [email protected]

Keywords: myotonic dystrophy type 1, pericytes, pericyte-derived induced pluripotent stem cells

(PC-iPSCs), PC-iPSC derived pericytes (PiPs)

Page 85: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

84

P 35

Intellectual Disability-related genes increase ADHD risk and

locomotor activity in Drosophila melanogaster

Marieke Klein1uginia Singgih1nne van Rens1, Ditte Demontis2,3,4, Anders D. Børglum2,3,4, Nina

Roth Mota1,5 Anna Castells-Nobau1, Lambertus A. Kiemeney6, Han G. Brunner1, Alejandro Arias-

Vasquez1,5, Annette Schenck1, Monique van der Voet1$, Barbara Franke1,5$*

1Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour, Department of Human Genetics,

Nijmegen, The Netherlands 2Department of Biomedicine, Aarhus University, Aarhus, Denmark 3Centre for Integrative

Sequencing, iSEQ, Aarhus University, Aarhus, Denmark 4The Lundbeck Foundation Initiative for Integrative Psychiatric

Research, iPSYCH, Denmark 5Radboud university medical center, Donders Institute for Brain, Cognition and Behaviour,

Department of Psychiatry, Nijmegen, The Netherlands 6Radboud university medical center, Donders Institute for Brain,

Cognition and Behaviour, Department for Health Evidence, Nijmegen, The Netherlands ual contribution $ shared final

responsibility

Objective: Attention-Deficit/Hyperactivity Disorder (ADHD) is a common, highly heritable

neuropsychiatric disorder. ADHD often co-occurs with Intellectual Disability (ID), and shared

overlapping genetics have been suggested [1]. This study aimed to identify novel ADHD genes by

investigating whether genes carrying rare mutations linked to ID contribute to ADHD risk through

common genetic variants. Validation and characterization of candidates were performed using

Drosophila melanogaster. Method: Common genetic variants in a diagnostic gene panel of 396

autosomal ID genes were tested for association with ADHD risk, through gene-set and gene-wide

analyses [2, 3], using ADHD meta-analytic data of the Psychiatric Genomics Consortium (n=19,210)

for discovery and iPSYCH ADHD data for replication (n=37,076) [4]. The significant genes were

functionally validated and characterized in Drosophila by assessing locomotor activity and sleep

upon knockdown of those genes in brain circuits [5]. Results: The ID gene-set was significantly

associated with ADHD risk in the discovery and replication data-sets. The three genes most

consistently associated were MEF2C, ST3GAL3, and TRAPPC9. Performing functional

characterization of the two evolutionary conserved genes in Drosophila melanogaster, we found

their knockdown in dopaminergic (dMEF2) and circadian neurons (dTRAPPC9) to result in increased

locomotor activity and reduced sleep, concordant with the human phenotype. Conclusions: This

study reveals that a large set of ID-related genes contributes to ADHD risk through effects of

common alleles. Utilizing this continuity, we identified TRAPPC9, MEF2C, and ST3GAL3 as novel

ADHD candidate genes. Characterization in Drosophila suggests that TRAPPC9 and MEF2C

contribute to ADHD-related behavior through distinct neural substrates.

E-mail: [email protected]

Keywords: Intellectual disability, ADHD, genetics, Drosophila melanogaster, MEF2C, TRAPPC9

Page 86: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

85

P 36

An integrative approach to STXBP1-Encephalopathy: in vitro,

in vivo and patient clinics.

Annemiek van Berkel*1,2, Hanna Lammertse*1,2, Jovana Kovacevic*1,2, Mala Misra-Isrie2, Jessica

Bos2, Hanne Meijers-Heijboer2, Ruud Toonen1,2, Mieke van Haelst2 and Matthijs Verhage1,2

1Department of Functional Genomics, Center for Neurogenomics and Cognitive Resarch, Amsterdam Neuroscience, Vrije

Universiteit (VU) Amsterdam, de Boelelaan 1078, 1081 HV Amsterdam, the Netherlands 2 Department of Clinical Genetics,

Amsterdam University Medical Center, location VUmc, 1081 HV Amsterdam, The Netherlands * These authors contributed

equally to this work.

STXBP1 is highly expressed in the brain and important for communication between neurons.

Mutations in the STXBP1 gene cause early infantile encephalopathy (STXBP1-E). Following the first

report of an STXBP1 microdeletion in a patient diagnosed with Ohtahara syndrome in 2008, more

than 100 other cases have been reported of patients with heterozygous missense or nonsense

mutations in the STXBP1 gene. The clinical features presented by patients with an STXBP1 mutation

are highly diverse. All patients display a developmental delay and severe to profound intellectual

disability. The majority of patients present with early-onset epilepsy. The seizure type and

responsivity to anti-epileptic treatments is variable. Many other symptoms are often reported by

families, but not systematically examined in this patient group, such as autism. To date, no therapy

is known beyond seizure control. The pathological mechanisms underlying the symptoms remain

to be elucidated and research has primarily focused on descriptions of individual cases using

diverse methodology and criteria. In the present study, the aim is to generate a standardized,

multilevel assessment of STXBP1-E in affected patients from clinics worldwide. We invite patients

to our special STXBP1 clinics, where patients and their families/caregivers come to a central location

for a multilevel assessment by a multidisciplinary team of researchers. Firstly, the phenotypic

features of patients are described using standardized criteria, through questionnaires and a

structured interview with the patient families/caregivers. Furthermore, patients are examined by a

clinical geneticist (dysmorphologist) and a neurologist. Secondly, an EEG recording is performed.

Thirdly, a skin biopsy is collected from a subgroup of patients. Induced pluripotent stem cell-

technology is being used to generate a cellular in vitro model of STXBP1-E. This allows us to

investigate the effects of STXBP1 mutations at the cellular and molecular level in patient neurons.

Taken together, this multilevel cohort study will provide the first in depth systematic and

quantitative assessment of STXBP1-E patients. Analysis of these data obtained will provide new

insights into the possible disease mechanisms of STXBP1-Encephalopathy. In addition, this

systematic assessment of the phenotypic characteristics can help achieving better diagnoses with

the ultimate aim of providing the best treatment.

E-mail: [email protected]

Keywords: STXBP1-encephalopathy, STXBP1, neurodevelopmental disorder, developmental

delay/intellectual disability, epilepsy, epileptic encephalopathy

Page 87: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

86

P 37

The complexities of analysis for APC mosaicism

D. Terlouw1*, M. Suerink1*, C. Tops1, A. Langers1, F. Hes2, S. ten Broeke3, L. Dams1, T. van Wezel1,

H. Morreau1. Nielsen1

Leiden University Medical Center, Leiden, the Netherlands 2 University Hospital Brussels, Brussels, Belgium 3 University

Medical Center Groningen, Groningen, the Netherlands */ese authors contributed equally to this work

Objectives Mosaic mutations in the APC gene have been identified as a common cause (25%) for

polyposis in patients with >20 adenomas without germline mutation. The frequency remains

unknown in patients with milder phenotypes. Methods The APC gene was sequenced in DNA

isolated from 4 adenomas in our cohort (n=132) using Next Generation Sequencing. Patients were

considered mosaic if an identical mutation was identified in all lesions. An identical variant in only

a subset of the adenomas, was considered a so-called hybrid mosaicism. Detection rates and clinical

characteristics were compared between different subgroups. Results The mosaicism detection rate

was 17% in the entire cohort (22/132), 0% in patients with <10 adenomas (0/20), 7% in those with

10-20 adenomas (3/42) and 12% in patients over age 70 (2/17). Interestingly, 26 hybrid cases were

identified. The mean age of diagnosis and number of adenomas was comparable in hybrid and

non-mosaic cases (both ~61 years and ~23 adenomas) and differed from 'pure´ mosaic cases (45

years, 54 adenomas). Possible explanations for the occurrence of hybrid cases are; chance, field

cancerization, contamination, spread of cells during colonoscopies and partial mosaicism with

incidental adenomas. However, no universal explanation could be found. Conclusions Our results

indicate that mosaic APC mutations also play a role in patients with <20 adenomas and/or older

age at presentation. No universal explanation is identified to explain the occurrence of hybrid cases.

So, case by case evaluation and expansion of the cohort is required.

E-mail: [email protected]

Keywords: unexplained polyposis coli, APC mosaicism

Page 88: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

87

P 38

The MAP3K7 gene: further eloboration on phenotypes, and

evidence for interaction with the RAS pathway/clinical

overlap with Noonan syndrome

REM Senden1*, GM van Woerden2*, C de Konink2, R Avagliano Trezza2, JA Bassetti3, Y van Bever1,

LM Bird4, BW van Bon5, AS Brooks1, I van der Burgt5, EW Klee6, Carlo Marcelis5, JA Morales-

Rosado6, LA Schimmenti7, PA Terhal8, MW Wessels1, H van Wieringen9, M Nicetta10, Y.

Elgersma2d S. Demirdas1

Department of Genetics, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands 2 Department of

Neuroscience and the ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center,

3015 CN Rotterdam, The Netherlands 3 Division of Medical Genetics, Department of Pediatrics, Weill Cornell Medical

College, New York, N.Y, USA 4 Division of Genetics & Genomics, Boston Children's Hospital, Harvard Medical School, Boston,

Massachusetts 5 Department of Human Genetics, Radboud University Medical Center, Radboud University, the Netherlands.

6 Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN,

USA. 7 Department of Otorhinolaryngology, Clinical Genomics and Biochemistry and Molecular Biology, Mayo Clinic,

Rochester, MN, USA. 8 Department of Genetics, University Medical Center Utrecht, Utrecht University, The Netherlands 9

Department of Pediatrics, St. Antonius Hospital, Nieuwegein, The Netherlands 10 Genetics and Rare Diseases Research

Division, Bambino Gesù Children's Hospital, Rome, Italy.

Introduction The MAP3K7 gene is associated with two autosomal dominant inheriting syndromes;

frontomethaphyseal dysplasia type 2 (FMD2, gain of function), and cardiospondylocarpofacial

syndrome (CSCF, loss of function (LOF)). Whole exome sequencing in 5 Dutch patients suspected

to have Noonan Syndrome (NS), showed novel variants in MAP3K7. Clinical assessment confirmed

CSCF. The striking resemblance with NS suggested an upregulation of the RAS pathway caused by

LOF variants in MAP3K7. Furthermore, some patients showed neuronal involvement, suggesting an

effect on neuronal formation. Methods We studied 12 children and 2 adults with a novel variant in

MAP3K7 (12 with CSCF; 2 with FMD2). Clinical assessments were made using a digital database

containing specific clinical features. In order to test the effect of MAP3K7 on the RAS pathway, we

measured levels of activated ERK (pERK) in transfected mice. To test the effect on neuronal

migration (NM) in mice brain we used in vivo transfection via in utero electroporation. Laboratory

technicians were blinded for patient's phenotype. Results The clinical features of CSCF patients

overlapped with NS: short stature (n=7; 58%), congenital cardiac abnormalities (n=7, 58%), low-set

posteriorly rotated ears (n=6; 50%), hypertelorism (n=8; 67%), deeply grooved philtrum (n=6; 50%),

dental malocclusion (n=2, 16%) and epicanthic folds (n=6; 50%). Distinguishing features of CSCF

are the wide nose with bulbous tip (n=6; 50%), peri-orbital fullness (n=4, 32%) and full cheeks (n=8;

67%). Neurological phenotype: ID (n=1), language delay (n=1) and autism (n=1). FMD2 patients

(one with ID) showed phenotypes compatible with previously reported patients. We showed pERK

to be upregulated in CSCF (pERK was stable in FMD2). MAP3K7 LOF variants therefore cause

upregulation of the RAS pathway, explaining the Noonan-like phenotype. The brains of transfected

mice with variants from patients with a neuronal phenotype showed disrupted NM. Conclusion We

investigated the clinical features of patients with novel pathogenic variants in the MAP3K7 gene

leading to either CSCF (n=12) or FMD2 (n=2). We conclude that CSCF should be considered in the

differential diagnosis of NS because there is an overlap in phenotype and an upregulation of the

RAS pathway. In vivo transfection of mice embryos effectively showed which MAP3K7 variants cause

a neurological phenotype.

Page 89: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

88

E-mail: [email protected]

Keywords: MAP3K7, Cardiospondylocarpofacial syndrome, frontometaphyseal dysplasia type 2,

FMD2, RAS pathway, neuronal migration, in utero extrapolation, pERK, Noonan syndrome

Page 90: VELDHOVEN - NVHGThe added value of long-read amplicon sequencing for clinical applications 12:15 Peter Henneman Reliable application of DNA-methylation signatures in genetic diagnostic

89

P 39

Further delineation of phenotypes related to integral cohesin

structural protein RAD21

Lianne C Krab MD PhD, Asuman Zeynep Tümer MD PhD, Raoul C Hennekam MD PhD

Cordaan, Klinkerweg 75, 1033 PK Amsterdam, the Netherlands; [email protected] Department of Clinical Medicine,

Rigshospitalet - Diagnostisk Center, Gamle Landevej 7-9, 2600 Glostrup; Department of Pediatrics, Amsterdam University

Medical Centers, location Academic Medical Center, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands.

Background: Aberrations in RAD21 (RAD21 Cohesin Complex Component) have been associated

with clinical phenotypes that overlap with that of Cornelia de Lange Syndrome (CdLS). Although it

has been stated that RAD21 variants lead to a relatively mild CdLS phenotype, the number of

reported individuals on which this has been based is small, and insight into the cognitive and

behavioral aspects is limited. We aimed to provide an overview of the RAD21 variants reported thus

far, and to gather the associated phenotype data with a special focus on development, cognition

and behavior. Method: Using international inquiry, literature and database search, we identified 54

cases with RAD21 variants (36 index; including 26 sequence variants and 10 CNV´s). With a

dedicated questionnaire, we gathered extensive and updated data. This led to two patient cohorts:

cohort A (29 patients, 22 index) with extensive clinical data, and cohort B (25 cases, 14 index) with

limited clinical data. The phenotype of cohort A were compared to those of CdLS patients with

NIPBL (n=67) and SMC1A (n=51) variants. With the help of in silico analyses of mutation effects,

genotype-phenotype relationships were investigated. Results: All 29 patients in Cohort A had

sufficient CdLS features to warrant molecular genetic testing for CdLS; 13 met clinical criteria for

classical CdLS. Overall, somatic, cognitive and behavioral problems were markedly less prevalent

and less severe in the RAD21 cohort compared to the NIPBL and SMC1A cohorts. Remarkable

findings are a paucity of self-injurious behavior and apparent absence of major limb malformations.

Cohort B included cases with phenotypes not previously associated with CdLS. These will be

elaborated upon at the meeting. Two recurrent mutations were found each in 2 families, with large

interfamilial variation. further genotype-phenotype analyses are in progress and will be presented

at the meeting. At least 10 index cases had a familial variant, with part of the cases were only

identified after diagnosis in a child or sib. The large intrafamilial variation has obvious consequences

for counseling. Conclusion: This case series represents a comprehensive overview of RAD21 variants

reported so far, including 18 new index cases. The data should be of benefit to clinical geneticists

and physicians when counseling families with affected members with a RAD21 variation.

E-mail: [email protected]

Keywords: RAD21 (RAD21 Cohesin Complex Component), Cornelia de Lange Syndrome, deep

phenotyping, genotype phenotype correlations, cognitive functioning, behavior