18.01.2013 Views

14th Annual Meeting of the German Society of Neurogenetics

14th Annual Meeting of the German Society of Neurogenetics

14th Annual Meeting of the German Society of Neurogenetics

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

UNIVERSITÄTSKLINIKUM<br />

Schleswig-Holstein<br />

Campus Lübeck<br />

Dept. <strong>of</strong> Neurology<br />

Institute <strong>of</strong> Human Genetics<br />

Dept. <strong>of</strong> Psychiatry and Psycho<strong>the</strong>rapy<br />

<strong>German</strong><br />

<strong>Society</strong> <strong>of</strong><br />

<strong>Neurogenetics</strong><br />

14 th <strong>Annual</strong> <strong>Meeting</strong><br />

<strong>of</strong> <strong>the</strong> <strong>German</strong> <strong>Society</strong><br />

<strong>of</strong> <strong>Neurogenetics</strong><br />

September 25 – 27, 2008, Lübeck<br />

(c) DGNG e.V.


Wel c o m e<br />

Dear Colleagues,<br />

It is our pleasure to invite you to <strong>the</strong> 14 th <strong>Annual</strong><br />

<strong>Meeting</strong> <strong>of</strong> <strong>the</strong> <strong>German</strong> <strong>Society</strong> <strong>of</strong> <strong>Neurogenetics</strong><br />

held in Lübeck, September 25-27, 2008.<br />

This year’s main topics include<br />

▪ Genetics <strong>of</strong> Movement Disorders<br />

▪ Genetics <strong>of</strong> Abnormal Brain Development<br />

▪ Genetics <strong>of</strong> Psychiatric Diseases<br />

In all three areas, new advances have emerged at<br />

a rapid pace, impacting on our understanding <strong>of</strong><br />

disease mechanisms, genetic testing, and patient<br />

care. While <strong>the</strong>se fascinating developments have<br />

provided important insights, <strong>the</strong>y also sparked novel<br />

questions, revealed fur<strong>the</strong>r complexity <strong>of</strong> a genetic<br />

contribution to neurological and psychiatric disease,<br />

and challenged some <strong>of</strong> <strong>the</strong> previously held concepts.<br />

Bringing toge<strong>the</strong>r neurologists, human geneticists,<br />

psychiatrists, neuropediatricians, and basic science<br />

researchers with world-renowned experts, <strong>the</strong><br />

conference will combine formal lectures, workshops,<br />

platform presentations, and poster sessions.<br />

As Thomas Mann pointed out, „It is impossible for<br />

ideas to compete in <strong>the</strong> marketplace if no forum for<br />

<strong>the</strong>ir presentation is provided or available.“ We are<br />

delighted to provide this forum in Lübeck, home <strong>of</strong><br />

<strong>the</strong> Mann Family, former Queen <strong>of</strong> <strong>the</strong> Hanseatic<br />

League, and last but not least, a modern Baltic city<br />

with a young, vibrant university.<br />

We look forward to sharing ideas at an exciting<br />

conference and to seeing you in Lübeck.<br />

With kind regards<br />

C. Klein C. Zühlke<br />

G. Gillessen-Kaesbach R. Lencer<br />

T. Gasser, President <strong>of</strong> <strong>the</strong> DGNG<br />

3


4<br />

D i r e c t o r y<br />

Page<br />

Program ........................................................................................................................................................6<br />

Workshop: Update on New Techniques .....................................................................................................8<br />

Public Lecture ...........................................................................................................................................16<br />

Key Note Lecture ......................................................................................................................................18<br />

Genetics <strong>of</strong> Movement Disorders .............................................................................................................20<br />

Genetics <strong>of</strong> Abnormal Brain Development ...............................................................................................26<br />

Genetics <strong>of</strong> Psychiatric Diseases .............................................................................................................34<br />

Free Communications ................................................................................................................................42<br />

Poster Abstracts ........................................................................................................................................53<br />

Sponsoring .................................................................................................................................................75<br />

Local Organizers ........................................................................................................................................76<br />

Contact .......................................................................................................................................................77


Fac u l t y<br />

Volker Arolt, Münster, D<br />

Michael Bonin, Tübingen, D<br />

Cornelius Borck, Lübeck, D<br />

Jürgen Deckert, Würzburg, D<br />

Gabriele Gillessen-Kaesbach, Lübeck, D<br />

Paola Giunti, London, UK<br />

Ute Hehr, Regensburg, D<br />

Fritz Hohagen, Lübeck, D<br />

Christine Klein, Lübeck, D<br />

Detlef Kömpf, Lübeck, D<br />

Kerstin Kutsche, Hamburg, D<br />

Rebecca Lencer, Lübeck, D<br />

Susanne Lucae, München, D<br />

Klaus-Armin Nave, Göttingen, D<br />

Markus Nö<strong>the</strong>n, Bonn, D<br />

Michael O’Donovan, Cardiff, UK<br />

Daniela T. Pilz, Cardiff, UK<br />

Stefan-M. Pulst, Salt Lake City, USA<br />

Arndt Rolfs, Rostock, D<br />

Anthony Schapira, London, UK<br />

Constance Schrander-Stumpel, Maastricht,NL<br />

Reiner Siebert, Kiel, D<br />

Andreas Ziegler, Lübeck, D<br />

Christine Zühlke, Lübeck D<br />

5


6<br />

Pr o g r a m<br />

Thursday, Sep 25, 2008<br />

Workshop: Update on New Techniques<br />

Chair: R. Siebert<br />

16.30 Comparative genomic hybridization<br />

R. Siebert<br />

16.30 Array gene expression analysis<br />

M. Bonin<br />

17.00 Next generation sequencing<br />

A. Rolfs<br />

17.30 Association studies<br />

A. Ziegler<br />

Congress Opening<br />

18.30 Welcome Address<br />

Chairs: D. Kömpf / F. Hohagen<br />

18.45 Public lecture<br />

Metaphern und Modelle der Hirnforschung<br />

C. Borck<br />

19.15 Music<br />

19.30 Key note lecture<br />

Neuron-glia interactions and <strong>the</strong> genetics<br />

<strong>of</strong> myelin diseases K.-A. Nave<br />

20.00 Music<br />

20.15 Get-toge<strong>the</strong>r<br />

Friday, Sep 26, 2008<br />

Genetics <strong>of</strong> Movement Disorders<br />

Chairs: C. Zühlke / A. Schapira<br />

9.00 Ion channel dysfunction in <strong>the</strong> SCAs<br />

S. Pulst<br />

9.45 SCA11: Clinical and genetic features<br />

P. Giunti<br />

10.15 C<strong>of</strong>fee break<br />

10.45 Mitochondrial involvement in <strong>the</strong><br />

cause <strong>of</strong> Parkinson’s disease<br />

A. Schapira<br />

11.15 Genetics <strong>of</strong> dystonia C. Klein<br />

Free Communications<br />

11.45 Regulation <strong>of</strong> astrocyte inflammatory<br />

responses by <strong>the</strong> Parkinson’s<br />

disease-associated DJ-1<br />

J. Waak<br />

12.00 Abnormal basal ganglia activity during<br />

response selection in Parkin mutation<br />

carriers J. v.d. Vegt<br />

12.15 Pathogenic HSP gene duplications<br />

and protection <strong>of</strong> female carriers<br />

C. Beetz<br />

12.30 Novel mutations in <strong>the</strong> senataxin gene<br />

in six patients with AOA2<br />

V. Bernard<br />

12.45 Lunch break and poster viewing<br />

13.00 DGNG Board <strong>Meeting</strong>


Pro g r a m<br />

Friday, Sep 26, 2008<br />

Genetics <strong>of</strong> Abnormal Brain Development<br />

Chairs: G. Gillessen-Kaesbach / D. Pilz<br />

14.15 The lissencephalies D. T. Pilz<br />

15.00 Holoprosencephaly: Genetics and<br />

what a face can tell about <strong>the</strong> brain<br />

U. Hehr<br />

15.30 C<strong>of</strong>fee break<br />

16.00 Mutations <strong>of</strong> CASK cause a novel Xlinked<br />

brain malformation phenotype<br />

K. Kutsche<br />

16.30 Congenital hydrocephalus: The<br />

Maastricht experience and <strong>the</strong> concept<br />

<strong>of</strong> ciliopathy<br />

C. Schrander-Stumpel<br />

Free Communications<br />

17.00 Mutations in FOXG1 cause a broad<br />

phenotypic spectrum ...<br />

F. Kortüm<br />

17.15 14-months old girl with holoprosencephaly,<br />

microcephaly, microtia, short<br />

stature and developmental delay. A<br />

new syndrome? J. Stefanova<br />

17.30 Chromosomal inversion combined<br />

with interstitial deletions ...<br />

H. Gabriel<br />

17.45 Refining <strong>the</strong> phenotype <strong>of</strong> α-1a Tubulin<br />

(TUBA1A) mutation ...<br />

D. Morris-Rosendahl<br />

20.00 Conference dinner<br />

Saturday, Sep 27, 2008<br />

Genetics <strong>of</strong> Psychiatric Diseases<br />

Chairs: R. Lencer / V. Arolt<br />

9.00 Schizophrenia genetics:<br />

new insights from new approaches<br />

M. O’ Donovan<br />

9.45 Genetics <strong>of</strong> bipolar affective disorder<br />

M. Nö<strong>the</strong>n<br />

10.15 C<strong>of</strong>fee break<br />

10.45 Genetics <strong>of</strong> <strong>the</strong> response to antidepressant<br />

treatment S. Lucae<br />

11.15 Genetics <strong>of</strong> anxiety disorders<br />

J. Deckert<br />

Free Communications<br />

11.45 MAOA genotype: Impact on amygdala-prefrontal<br />

connectivity in major<br />

depression U. Dannlowski<br />

12.00 Fine-mapping in multiplex families<br />

with Attention-Deficit Hyperactivity<br />

Disorder K. Lin<br />

12.15 Psychiatric symptoms in genetic<br />

forms <strong>of</strong> Parkinson disease<br />

M. Kasten<br />

12.30 DGNG awards and poster prices<br />

12.45 Closing remarks<br />

13.00 Lunch<br />

7


8<br />

Wor k s h o P : uP D a t e on ne W te c h n i q u e s<br />

Reiner Siebert is “Facharzt für Humangenetik”<br />

and since 2006 full Pr<strong>of</strong>essor and Chair <strong>of</strong><br />

Human Genetics at <strong>the</strong> Christian-Albrechts-<br />

University <strong>of</strong> Kiel and Director <strong>of</strong> <strong>the</strong> Institute<br />

<strong>of</strong> Human Genetics at <strong>the</strong> University Hospital<br />

Schleswig-Holstein, Campus Kiel. After studying<br />

Medicine at <strong>the</strong> University <strong>of</strong> Essen he started<br />

his residency at <strong>the</strong> Institute <strong>of</strong> Human Genetic in<br />

Kiel in 1996. He finished his habilitation in 2001<br />

and received a call as Pr<strong>of</strong>essor for Molecular<br />

Oncology in Hamburg in 2002. He is Section<br />

Editor <strong>of</strong> <strong>the</strong> journal “Leukemia” and reviewer for<br />

various international journals and funding bodies.<br />

His research interests among o<strong>the</strong>rs cover<br />

<strong>the</strong> development and application <strong>of</strong> molecular<br />

cytogenetic techniques, genetic changes in<br />

hematologic tumors and <strong>the</strong> role <strong>of</strong> epigenetic<br />

changes in disease development.<br />

Reiner Siebert, MD<br />

Institute <strong>of</strong> Human Genetics<br />

University <strong>of</strong> Kiel, <strong>German</strong>y


Comparative genomic hybridization<br />

Array CGH merges molecular diagnostics with<br />

traditional conventional cytogenetics and opens<br />

a rapidly growing field <strong>of</strong> genetic diagnostics. The<br />

technique is based on <strong>the</strong> competitive hybridization<br />

<strong>of</strong> a test and a reference genome onto a<br />

microarray containing oligonucleotides or BAC<br />

clones. Thus, it provides a high-resolution overview<br />

<strong>of</strong> chromosomal imbalances across <strong>the</strong> genome<br />

and can detect chromosomal microdeletions or<br />

microduplications which are common aberrations<br />

leading to mental retardation and malformation<br />

syndromes.<br />

We apply CGH to oligonucleotide arrays (105k<br />

and 244k arrays) in <strong>the</strong> postnatal diagnostic and<br />

research setting on a routine basis. In <strong>the</strong> postnatal<br />

setting submicroscopic imbalances (


10<br />

Wor k s h o P : uP D a t e on ne W te c h n i q u e s<br />

Dr. Michael Bonin graduated from <strong>the</strong> University<br />

Kassel in 1998 with a first class degree in<br />

Genetics. His PhD studies were conducted at<br />

<strong>the</strong> Genetics Department at <strong>the</strong> same University,<br />

where he worked on <strong>the</strong> biophysics <strong>of</strong> RNA-protein<br />

interactions under <strong>the</strong> mentorship <strong>of</strong> Wolfgang<br />

Nellen. In 2001 Dr. Bonin moved to <strong>the</strong> Medical<br />

Faculty at <strong>the</strong> University <strong>of</strong> Tübingen where he<br />

headed <strong>the</strong> Research Management Group for<br />

one year. After this, he established <strong>the</strong> Microarray<br />

Facility Tübingen, as a Core Facility for microarray<br />

applications. The Facility is an integrated part <strong>of</strong><br />

<strong>the</strong> Institute <strong>of</strong> Human Genetics and he is now<br />

head <strong>of</strong> <strong>the</strong> group for neurotranscriptomic studies.<br />

Michael Bonin, PhD<br />

Institute <strong>of</strong> Human Genetics<br />

University <strong>of</strong> Tübingen, <strong>German</strong>y


Array gene expression analysis<br />

Neurological diseases (ND) are one <strong>of</strong> <strong>the</strong> greatest<br />

challenges facing our population, from medical,<br />

financial, and social perspectives. The application<br />

<strong>of</strong> new research approaches to understand <strong>the</strong><br />

underlying pathogenesis <strong>of</strong> ND is critical. In<br />

this workshop, we review <strong>the</strong> use <strong>of</strong> microarray<br />

analysis in different Neurological diseases.<br />

Microarray technology has become a common tool<br />

for developing expression pr<strong>of</strong>iles. Microarrays<br />

have tremendous power, simultaneously querying<br />

<strong>the</strong> expression <strong>of</strong> tens <strong>of</strong> thousands <strong>of</strong> genes from<br />

a given biological sample. Coupled with impressive<br />

advances in statistical tools for analyzing large,<br />

complex data sets, well-designed microarray<br />

experiments are poised to make a big impact in <strong>the</strong><br />

field <strong>of</strong> ND.<br />

The fact that microarray investigators do not<br />

know which genes or pathways will be discovered<br />

by <strong>the</strong>ir experiments is precisely <strong>the</strong> strength <strong>of</strong><br />

microarrays. The ability to examine <strong>the</strong> expression<br />

pr<strong>of</strong>ile <strong>of</strong> potentially <strong>the</strong> entire genome at once,<br />

without preconceptions, <strong>of</strong>fers <strong>the</strong> possibility <strong>of</strong><br />

completely novel and unexpected insights into<br />

entities as complex as <strong>the</strong> nervous system. But<br />

before <strong>the</strong>se insights can be made, <strong>the</strong> practical<br />

problems <strong>of</strong> experimental design, data analysis,<br />

verification, and interpretation need to be<br />

addressed.<br />

11


12<br />

Wor k s h o P : uP D a t e on ne W te c h n i q u e s<br />

Arndt Rolfs, M.D., studied Medicine at <strong>the</strong><br />

Universities <strong>of</strong> Mainz and Vienna and received<br />

his degree in Medicine from both Universities<br />

in 1985. After working at <strong>the</strong> Free University<br />

<strong>of</strong> Berlin, he got a scholarship from <strong>the</strong> Max-<br />

Planck-<strong>Society</strong> with employment at <strong>the</strong> Max-<br />

Planck-Institute for Molecular Genetics in Berlin.<br />

In 1993, he began working at <strong>the</strong> University <strong>of</strong><br />

Rostock as Senior Physician at <strong>the</strong> Department<br />

<strong>of</strong> Neurology and Pr<strong>of</strong>essor for Neurology and<br />

Psychiatry. In parallel to this he was <strong>the</strong> head<br />

<strong>of</strong> <strong>the</strong> Neurobiological Laboratory. Following<br />

several years in clinical and research working, he<br />

became <strong>the</strong> head <strong>of</strong> <strong>the</strong> Albrecht-Kossel-Institute<br />

for Neuroregeneration in 2008.<br />

Arndt Rolfs<br />

Albrecht-Kossel-Institut für Neuroregeneration<br />

University <strong>of</strong> Rostock, <strong>German</strong>y


Next-generation sequencing<br />

In 2004, <strong>the</strong> National Human Genome Research<br />

Institute (NHGRI) proposed a way to achieve<br />

affordable human genome sequencing by 2014,<br />

in two increments. NHGRI program director Jeff<br />

Schloss explains: “The way <strong>the</strong> [Requests for<br />

Applications] were laid out, at <strong>the</strong> time we launched<br />

<strong>the</strong> program, we were hoping <strong>the</strong> $100,000 genome<br />

might come in five years. The goal for $1,000 was<br />

to be five years after that.” In <strong>the</strong> quest for <strong>the</strong><br />

$1,000 genome, every imaginable approach is<br />

being explored to develop sequencing technologies<br />

with improvements in miniaturization, parallelism<br />

and simplification. Next-generation sequencing is<br />

already several orders <strong>of</strong> magnitude more efficient<br />

than <strong>the</strong> Sanger capillary-array electrophoresis<br />

(CAE) machines. About one year ago, Solexa, <strong>of</strong><br />

Hayward, California, announced <strong>the</strong> completion<br />

<strong>of</strong> an early-access program evaluating its nextgeneration<br />

Genome Analysis system with customers<br />

and reiterated its intention to begin full commercial<br />

sales this quarter. Curagen spin-<strong>of</strong>f 454 Life<br />

Sciences, <strong>of</strong> Branford, Connecticut, and Agencourt<br />

Personal Genomics in Beverly, Massachusetts,<br />

a part <strong>of</strong> Applied Biosystems Group, are already<br />

on <strong>the</strong> market with systems that bring sequencing<br />

costs down several orders <strong>of</strong> magnitude below<br />

<strong>the</strong> millions <strong>of</strong> dollars per genome cost associated<br />

with capillary-array electrophoresis (CAE)<br />

sequencing—<strong>the</strong> technology that made possible<br />

<strong>the</strong> Human Genome Project a mere seven years<br />

ago. Helicos Biosciences, for its part, claims that its<br />

single-molecule sequencing technology, expected<br />

to debut in <strong>the</strong> second half <strong>of</strong> 2008, will enable <strong>the</strong><br />

sought-after ‘$1,000 genome’ price point, although<br />

not immediately. One <strong>of</strong> <strong>the</strong> newest winners <strong>of</strong><br />

NHGRI’s $100,000 genome grant, Intelligent Bio-<br />

Systems, <strong>of</strong> Waltham, Massachusetts, is developing<br />

a four-color sequencing-by-syn<strong>the</strong>sis method<br />

using cleavable fluorescent nucleotide reversible<br />

terminators—an approach similar to that <strong>of</strong> Solexa.<br />

It is placing instruments in selected laboratories for<br />

beta testing, with a technology that features faster<br />

run cycles, less up-front expense and less costly<br />

implementation. On <strong>the</strong> horizon are o<strong>the</strong>r singlemolecule<br />

sequencing technologies from Visigen<br />

Biotechnologies and Pacific Biosciences. These<br />

companies are developing different approaches<br />

based on real-time massively parallel imaging <strong>of</strong><br />

single-molecule base addition events catalyzed by<br />

DNA polymerase. Data are generated at a rate <strong>of</strong><br />

10–50 bases per second per polymerase molecule,<br />

as tens <strong>of</strong> thousands <strong>of</strong> polymerases read DNA<br />

templates. Read lengths <strong>of</strong> 8,000 bases may be<br />

within reach, making <strong>the</strong>se technologies ideal for<br />

<strong>the</strong> most challenging applications, like sequencing<br />

<strong>of</strong> structurally rearranged cancer cell genomes.<br />

Although shotgun-SBH may not be well suited to<br />

tackling genome rearrangements, its potential for<br />

extremely high parallelism through inexpensive<br />

scaling <strong>of</strong> <strong>the</strong> imaging area could give it a cost<br />

advantage in standard genome resequencing<br />

applications. Nearly 20 years have elapsed since<br />

sequencing by hybridization was first envisioned<br />

and <strong>the</strong> sequencing <strong>of</strong> single molecules <strong>of</strong> DNA was<br />

proposed. We are now on <strong>the</strong> cusp <strong>of</strong> a new era in<br />

which widely available high-throughput platforms<br />

will generate DNA sequence information from any<br />

organism at a cost <strong>of</strong> pennies per megabase.<br />

13


14<br />

Wor k s h o P : uP D a t e on ne W te c h n i q u e s<br />

Andreas Ziegler received his Ph.D. in Statistics<br />

(Dortmund) for his work on regression models<br />

for correlated data after receiving his diploma in<br />

Statistics (Munich) in 1992. In 1998, he completed<br />

his habilitation in genetic epidemiology (Marburg)<br />

on linkage analysis <strong>of</strong> quantitative traits for which<br />

he received <strong>the</strong> Johann-Peter Süßmilch Medal<br />

from <strong>the</strong> <strong>German</strong> <strong>Society</strong> for Medical Informatics,<br />

Biometry and Epidemiology. Since 2001, Dr.<br />

Ziegler has served as head <strong>of</strong> <strong>the</strong> Institute <strong>of</strong><br />

Medical Biometry and Statistics and full pr<strong>of</strong>essor<br />

at <strong>the</strong> University <strong>of</strong> Lübeck. He was President <strong>of</strong><br />

<strong>the</strong> <strong>German</strong> Region <strong>of</strong> <strong>the</strong> International Biometric<br />

<strong>Society</strong> and currently is member <strong>of</strong> <strong>the</strong> Board <strong>of</strong><br />

Directors <strong>of</strong> <strong>the</strong> International Biometric <strong>Society</strong> and<br />

<strong>the</strong> International Genetic Epidemiology <strong>Society</strong>.<br />

After his move to Lübeck, his primary interest for<br />

applications has shifted to infectious diseases.<br />

Andreas Ziegler, PhD<br />

Institute <strong>of</strong> Medical Biometry and Statistics,<br />

University <strong>of</strong> Lübeck, <strong>German</strong>y<br />

Thus Dr. Ziegler is currently involved in genetic<br />

studies <strong>of</strong> malaria, tuberculosis and sepsis and<br />

serves as a delegate <strong>of</strong> an expert group <strong>of</strong>f-label<br />

infectious diseases with main area <strong>of</strong> concentration<br />

in HIV/AIDS for <strong>the</strong> BfArM. O<strong>the</strong>r major ongoing<br />

studies relate to cardiovascular diseases. Over <strong>the</strong><br />

past five years, Dr. Ziegler has authored or coauthored<br />

more than 100 peer-reviewed papers and<br />

has authored a textbook on statistical approaches<br />

in genetic epidemiology.


Comparative genomic hybridization<br />

Association studies<br />

To search <strong>the</strong> entire human genome for<br />

association is a promising approach to unravelling<br />

<strong>the</strong> genetic basis <strong>of</strong> complex genetic diseases.<br />

The last year has seen a tremendous number <strong>of</strong><br />

publications on <strong>the</strong>se genome-wide association<br />

studies (GWAs), and many novel loci have<br />

been identified. Because several hundreds <strong>of</strong><br />

thousands <strong>of</strong> single nucleotide polymorphisms<br />

(SNPs) are analyzed at <strong>the</strong> same time, substantial<br />

biostatistical and computational challenges are<br />

posed. In this presentation, we give an overview<br />

<strong>of</strong> approaches to GWAs. We specifically discuss<br />

quality control aspects, adjustments for population<br />

stratification and imputation <strong>of</strong> SNP markers by<br />

using HapMap data.<br />

Finally, we briefly consider multistage approaches<br />

to GWAs and meta-analyses.<br />

15


16<br />

Pu b l i c le c t u r e<br />

Studium der Medizin, Philosophie,<br />

Religionswissenschaften und Medizingeschichte<br />

in Hamburg, Heidelberg und Berlin. 1994 MA<br />

phil. (Berlin), 1995 Promotion zum Dr. med. in der<br />

Aidsforschung (Berlin), 1996 Promotion zum PhD<br />

in Neurosciences (London). 2003 Habilitation für<br />

Medizin- und Wissenschaftsgeschichte (Berlin).<br />

Seit 2007 Pr<strong>of</strong>essor für Geschichte, Theorie<br />

und Ethik der Medizin und Naturwissenschaften<br />

an der Universität zu Lübeck und Direktor des<br />

dortigen Instituts für Medizingeschichte und<br />

Wissenschaftsforschung. Frühere Stationen seines<br />

Werdeganges waren das Max-Planck-Institut für<br />

Wissenschaftsgeschichte in Berlin (1998-2002),<br />

die Bauhaus Universität Weimar (2002-2004) und<br />

die McGill University in Montreal (2004-2007), wo<br />

er einen Canada Research Chair in Philosophy<br />

and Language <strong>of</strong> Medicine innehatte.<br />

Cornelius Borck, MD, PhD<br />

Ethik der Medizin<br />

University <strong>of</strong> Lübeck, <strong>German</strong>y


Modelle und Metaphern der Hirnforschung<br />

Die Hirnforschung pr<strong>of</strong>itiert ohne Zweifel enorm<br />

von technischen Innovationen. Ganze Zweige<br />

der Neurowissenschaften hängen von wichtigen<br />

Entwicklungen wie z.B. neuen Registrier- und<br />

Visualisierungsverfahren ab. Technische<br />

Erfindungen und Apparate haben aber auch noch<br />

andere Funktionen in der Hirnforschung. Immer<br />

wieder dienten sie z.B. als zentrale Modelle für<br />

die Leistungen des Gehirns. Vor gut hundert<br />

Jahren galt so das Gehirn gemeinhin als eine Art<br />

Telefonzentrale, und gerade erst sind wir dabei,<br />

uns von der Vorstellung zu verabschieden, unser<br />

Gehirn sei ein Supercomputer. In ihrem neuen<br />

Paradgima der Neuroplastizität scheint sich die<br />

Hirnforschung immer weniger für programmierte<br />

Schaltpläne und feste Verdrahtungen, aber umso<br />

mehr für Organismus-Umwelt-Interaktionen zu<br />

interessieren. Der Vortrag hinterfragt Genese,<br />

Strukturen und Folgen solcher Leitvorstellungen.<br />

Schon heute liegt dabei die besondere ethische<br />

oder politische Brisanz neuer Erkenntnisse und<br />

Einsichten der Hirnforschung vielleicht weniger<br />

in dem, was die Neurowissenschaft feststellen,<br />

sondern eher darin, wie sie dazu beitragen,<br />

Gehirne zu neuen, bislang unbekannten<br />

Leistungen freizustellen.<br />

17


18<br />

key no t e le c t u r e<br />

Pr<strong>of</strong>. Klaus-Armin Nave graduated in Biology<br />

at <strong>the</strong> University <strong>of</strong> Heidelberg in 1983. As a<br />

DAAD Scholar he obtained his PhD degree in<br />

Neurosciences at <strong>the</strong> University <strong>of</strong> California in<br />

San Diego. Following a postdoctoral fellowship at<br />

<strong>the</strong> Salk Institue for Biological Studies in La Jolla,<br />

he established an independent research group<br />

at <strong>the</strong> Center <strong>of</strong> Molecular Biology Heidelberg<br />

(ZMBH) in 1991 and completed his ‚Habilitation’ in<br />

Molecular Biology and Genetics in 1996. He was<br />

appointed a Full Pr<strong>of</strong>essor <strong>of</strong> Molecular Biology at<br />

<strong>the</strong> University <strong>of</strong> Heidelberg in 1997 and remains<br />

a faculty member on equal terms. Since 1999 he<br />

has been Director <strong>of</strong> <strong>the</strong> Max-Planck Institute for<br />

Experimental Medicine in Göttingen.<br />

Klaus-Armin Nave, MD<br />

Department <strong>of</strong> <strong>Neurogenetics</strong> Max Planck Institute <strong>of</strong> Experimental Medicine<br />

Göttingen, <strong>German</strong>y


Neuron-Glia interactions<br />

and <strong>the</strong> genetics <strong>of</strong> myelin diseases<br />

It is thought that oligodendrocytes myelinate<br />

axons for rapid impulse conduction, but o<strong>the</strong>r<br />

important functions <strong>of</strong> myelinating glial cells have<br />

remained obscure. We have identified genes<br />

expressed exclusively in mature oligodendrocytes<br />

that are not required for myelination, but essential<br />

for maintaining long-term axonal integrity. Also<br />

<strong>the</strong> absence <strong>of</strong> functional peroxisomes selectively<br />

from oligodendrocytes in conditional mouse<br />

mutants causes widespread axonal degeneration<br />

and progressive subcortical demyelination,<br />

without interfering with oligodendrocyte survival.<br />

Surprisingly, glial peroxisomal dysfunction<br />

causes a strong proinflammatory milieu in <strong>the</strong><br />

white matter leading to <strong>the</strong> infiltration <strong>of</strong> B and<br />

activated CD8(+) T cells into brain lesions.<br />

Thus, oligodendrocytes <strong>the</strong>mselves provide<br />

a neuroprotective function against axonal<br />

degeneration and neuroinflammation, a finding<br />

relevant for myelin disease, incl. MS or X-linked<br />

adrenoleukodystrophy. We propose <strong>the</strong> model<br />

that myelination per se causes a risk for axonal<br />

functions unless glial cells are able to support <strong>the</strong><br />

axons that <strong>the</strong>y ensheath.<br />

19


20<br />

ge n e t i c s oF mo v e m e n t Di s o r D e r s<br />

Stefan Pulst is Pr<strong>of</strong>essor and Chair <strong>of</strong> Neurology<br />

and member <strong>of</strong> <strong>the</strong> Brain Institute at <strong>the</strong><br />

University <strong>of</strong> Utah in Salt Lake City. His research<br />

focuses on inherited diseases <strong>of</strong> <strong>the</strong> nervous<br />

system with an emphasis on spinocerebellar<br />

ataxias and Parkinson disease. Ano<strong>the</strong>r interest<br />

relates to tumor suppressor genes controlling<br />

proliferation <strong>of</strong> Schwann cells. Recently, his work<br />

has also branched out into understanding <strong>the</strong><br />

genetic structure <strong>of</strong> human visual attention.<br />

Dr. Pulst was <strong>the</strong> founding chair <strong>of</strong> <strong>the</strong> Section<br />

on <strong>Neurogenetics</strong> and <strong>of</strong> <strong>the</strong> Basic Science<br />

Subcommittee <strong>of</strong> <strong>the</strong> American Academy <strong>of</strong><br />

Neurology and currently serves as chair <strong>of</strong> <strong>the</strong><br />

AAN Science Committee. From 1999 to 2006,<br />

he was Scientific Director <strong>of</strong> <strong>the</strong> National Ataxia<br />

Foundation <strong>of</strong> <strong>the</strong> US.<br />

Stefan Pulst, MD<br />

Department <strong>of</strong> Neurology<br />

University <strong>of</strong> Utah, Salt Lake City, USA


Ion channel dysfunction in <strong>the</strong> SCAs<br />

Spinocerebellar ataxia type 2 (SCA2) is caused<br />

by CAG repeat expansion in <strong>the</strong> ATXN2 gene.<br />

In SCA2 as in o<strong>the</strong>r polyQ diseases, length <strong>of</strong><br />

<strong>the</strong> CAG repeat inversely correlates with age <strong>of</strong><br />

disease onset (AO), but significant variation is<br />

observed within each repeat class. We used <strong>the</strong><br />

Cuban SCA2 founder population to examine age<strong>of</strong>-onset<br />

variance. No evidence <strong>of</strong> gender effects<br />

or imprinting was observed.<br />

We found that polyQ expansion only accounted<br />

for about 60% <strong>of</strong> <strong>the</strong> overall variance, but only<br />

for 30% in patients with repeats from 32 to 40<br />

CAG. Of <strong>the</strong> remainder, 50% was heritable.<br />

Heritability was higher in sib-sib than in parentchild<br />

pairs suggesting presence <strong>of</strong> recessive<br />

modifier alleles. As had been described in<br />

Huntington disease, we observed a higher<br />

heritability in female sibpairs, a phenomenon that<br />

has remained unexplained. We have begun a<br />

candidate gene-based search for modifier alleles<br />

focusing on normal repeat variation in o<strong>the</strong>r<br />

polyQ disease genes. The CACNA1A (SCA6)<br />

repeat modified AO suggesting that ei<strong>the</strong>r polyQ<br />

domain interaction or modified calcium flux may<br />

influence Purkinje cell dysfunction in SCA2. The<br />

SCA1 repeat showed a tendency to influence<br />

SCA2 AO, an observation that was recently<br />

replicated in <strong>the</strong> fly.<br />

Although polyQ ataxias comprise <strong>the</strong> majority<br />

<strong>of</strong> SCAs, recent investigations have identified a<br />

number <strong>of</strong> non-CAG repeat mutations in ataxia<br />

families. We studied a Filipino family with a<br />

late onset pure cerebellar ataxia and identified<br />

a mutation in KCNC3 (Kv3.3), as well as in a<br />

second family that had early-onset ataxia with<br />

mental retardation and seizures. We will report<br />

on a mutation screen <strong>of</strong> more than 400 ataxia<br />

patients that identified a number <strong>of</strong> DNA variants<br />

in KCNC3 with two recurrent mutations. Studies<br />

in Xenopus oocytes confirmed that <strong>the</strong>se variants<br />

change channel function, although genotype-in<br />

vitro phenotype-clinical phenotype correlations<br />

are not straight forward. Some variants change in<br />

vitro function, although genetically <strong>the</strong>y may not<br />

fulfill all genetic criteria to qualify as a mutation.<br />

The identification <strong>of</strong> causative and modifier<br />

alleles in CACNA1A and KCNC3 point to <strong>the</strong><br />

importance <strong>of</strong> ion channels as drug targets both<br />

for symptomatic and disease-modifying <strong>the</strong>rapies.<br />

21


22<br />

ge n e t i c s oF mo v e m e n t Di s o r D e r s<br />

Pr<strong>of</strong>essor Anthony Schapira MD DSc FRCP<br />

FMedSci was appointed in 1990 as Chairman<br />

<strong>of</strong> <strong>the</strong> University Department <strong>of</strong> Clinical<br />

Neurosciences, at <strong>the</strong> Institute <strong>of</strong> Neurology,<br />

Queen Square and Pr<strong>of</strong>essor <strong>of</strong> Neurology at <strong>the</strong><br />

National Hospital and <strong>the</strong> Royal Free Hospital.<br />

He is Director <strong>of</strong> Research and Development,<br />

and Clinical Head <strong>of</strong> Service for Neurosciences at<br />

<strong>the</strong> Royal Free Hospital.<br />

Pr<strong>of</strong>essor Schapira’s research interests<br />

include <strong>the</strong> molecular and clinical aspects<br />

<strong>of</strong> neurodegenerative diseases, with special<br />

emphasis on Parkinson’s disease and o<strong>the</strong>r<br />

movement disorders. He has over 500<br />

publications. Pr<strong>of</strong>essor Schapira is <strong>the</strong> recipient<br />

<strong>of</strong> <strong>the</strong> Harveian Medal, <strong>the</strong> Royal College <strong>of</strong><br />

Physicians Clinical Science Prize, <strong>the</strong> 1998<br />

Anthony Schapira, MD DSc FRCP FMedSci<br />

University Department <strong>of</strong> Clinical Neurosciences<br />

Institute <strong>of</strong> Neurology, Queen Square, UCL, London<br />

European Prize for Clinical Science and <strong>the</strong> 1999<br />

Opprecht Foundation Award. He was elected a<br />

Fellow <strong>of</strong> <strong>the</strong> Academy <strong>of</strong> Medical Sciences in<br />

1999. He was awarded <strong>the</strong> Duchenne Prize in<br />

2005.


Mitochondrial involvement in <strong>the</strong> cause<br />

<strong>of</strong> Parkinson’s disease<br />

Several biochemical abnormalities have been<br />

described in Parkinson’s disease (PD) brain<br />

including oxidative stress and mitochondrial<br />

dysfunction and <strong>the</strong> more recent identification <strong>of</strong><br />

specific gene mutations that cause PD has reenforced<br />

<strong>the</strong> relevance <strong>of</strong> both in <strong>the</strong> causation<br />

<strong>of</strong> familial as well as <strong>the</strong> sporadic forms <strong>of</strong> <strong>the</strong><br />

disease. Importantly, <strong>of</strong> <strong>the</strong> proteins that are<br />

associated with familial PD, two (PINK1 and DJ1)<br />

and possibly a third (parkin) are mitochondrial<br />

proteins, and both alpha-synuclein and LRRK2<br />

appear to be associated with mitochondria.<br />

All <strong>the</strong>se proteins also interface with <strong>the</strong><br />

pathways <strong>of</strong> oxidative stress and free radical<br />

damage. These recent insights into <strong>the</strong> aetiology<br />

and pathogenesis <strong>of</strong> PD provide hope that from<br />

converging ra<strong>the</strong>r than diverging pathways to<br />

cell dysfunction and death, can come drugs or<br />

combinations <strong>of</strong> drugs, that may successfully<br />

intervene in pathogenesis and slow disease<br />

progression.<br />

23


24<br />

ge n e t i c s oF mo v e m e n t Di s o r D e r s<br />

Dr. Christine Klein is a native <strong>of</strong> Hamburg,<br />

<strong>German</strong>y. She attended <strong>the</strong> Medical Schools <strong>of</strong><br />

Hamburg, Heidelberg, and Lübeck, <strong>German</strong>y,<br />

and took her final year studies in England,<br />

including one trimester at The National Hospital<br />

for Neurology and Neurosurgery, Queen Square,<br />

London with Dr. Niall P. Quinn. She moved<br />

to Boston from 1997-1999 for a fellowship in<br />

Molecular <strong>Neurogenetics</strong> under <strong>the</strong> mentorship <strong>of</strong><br />

Dr. X. O. Breakefield.<br />

Dr. Klein completed her neurology training at<br />

Lübeck University with Dr. D. Kömpf in 2004,<br />

followed by a research and clinical fellowship and<br />

two short sabbaticals in movement disorders with<br />

Dr. A. E. Lang in Toronto, Canada in 2004 - 2007.<br />

She was appointed a Lichtenberg Pr<strong>of</strong>essor<br />

<strong>of</strong> Clinical and Molecular <strong>Neurogenetics</strong> at <strong>the</strong><br />

Christine Klein, MD<br />

Department <strong>of</strong> Neurology<br />

University <strong>of</strong> Lübeck, <strong>German</strong>y<br />

Department <strong>of</strong> Neurology <strong>of</strong> Lübeck University<br />

in 2005, where her research has focused on <strong>the</strong><br />

clinical and molecular genetics <strong>of</strong> movement<br />

disorders.<br />

Dr. Klein is a past member <strong>of</strong> <strong>the</strong> Editorial Board<br />

<strong>of</strong> <strong>the</strong> Movement Disorders Journal and a present<br />

member <strong>of</strong> <strong>the</strong> Editorial Board <strong>of</strong> ‘Neurology’. She<br />

has published over 150 scientific papers.


Genetics <strong>of</strong> dystonia<br />

The dystonias are a heterogeneous group <strong>of</strong><br />

movement disorders, characterized clinically by<br />

involuntary twisting and repetitive movements and<br />

abnormal postures.<br />

Familial, early-onset, generalized torsion dystonia<br />

(DYT1) is <strong>the</strong> most common and severe form <strong>of</strong><br />

primary dystonia. It usually begins in childhood,<br />

first starts in a limb and tends to generalize to<br />

o<strong>the</strong>r body parts as <strong>the</strong> disease progresses. The<br />

mode <strong>of</strong> inheritance is autosomal dominant with<br />

reduced penetrance <strong>of</strong> about 30-40%, including<br />

both mild and severe cases. Most cases are<br />

caused by a 3-bp deletion (904_906delGAG) in <strong>the</strong><br />

coding region <strong>of</strong> <strong>the</strong> DYT1 gene, which is widely<br />

expressed in human brain and encodes <strong>the</strong> protein<br />

torsinA.<br />

Dopa-responsive dystonia is characterized by<br />

childhood-onset <strong>of</strong> dystonia, diurnal fluctuation <strong>of</strong><br />

symptoms, and a dramatic response to L-dopa<br />

<strong>the</strong>rapy. Later in <strong>the</strong> course <strong>of</strong> <strong>the</strong> disease,<br />

parkinsonian features may occur. While <strong>the</strong><br />

rare autosomal recessive form <strong>of</strong> DRD (DYT5b)<br />

is associated with mutations in <strong>the</strong> tyrosine<br />

hydroxylase (TH) gene, <strong>the</strong> more frequent<br />

dominantly inherited DRD is <strong>of</strong>ten caused by<br />

mutations in <strong>the</strong> GTP cyclohydrolase I (GCHI)<br />

gene (DYT5a).<br />

In myoclonus-dystonia (DYT 11), a predominantly<br />

myoclonic syndrome is combined with<br />

dystonic features which may rarely be <strong>the</strong> only<br />

manifestation <strong>of</strong> <strong>the</strong> disorder. Symptoms frequently<br />

respond to alcohol. Age <strong>of</strong> onset is usually in <strong>the</strong><br />

first or second decade <strong>of</strong> life. The inheritance<br />

pattern is autosomal dominant with variable<br />

expressivity and incomplete penetrance due to<br />

maternal imprinting <strong>of</strong> <strong>the</strong> epsilon-sarcoglycan<br />

(SGCE) gene.<br />

O<strong>the</strong>r rare forms <strong>of</strong> dystonia-parkinsonism or<br />

dystonia/dyskinesias with known genes include<br />

DYT3 dystonia (X-linked dystonia-parkinsonism;<br />

”lubag”), DYT8 dystonia (paroxysmal<br />

nonkinesigenic dyskinesia), and DYT12 dystonia<br />

(rapid-onset dystonia-parkinsonism), and DYT16<br />

dystonia (recessively inherited juvenile-onset,<br />

generalized dystonia).<br />

25


26<br />

ge n e t i c s oF ab n o r m a l br a i n De v e l o P m e n t<br />

Daniela Pilz is a Consultant in Medical Genetics<br />

at <strong>the</strong> Institute <strong>of</strong> Medical Genetics, University<br />

Hospital <strong>of</strong> Wales, Cardiff. She graduated<br />

from Medical School in Hannover, <strong>German</strong>y,<br />

and trained in paediatrics and clinical genetics<br />

in <strong>the</strong> UK. The topic <strong>of</strong> her MD <strong>the</strong>sis was<br />

“Lissencephaly”. She spent 2 years as a<br />

research associate in a molecular laboratory<br />

at <strong>the</strong> University <strong>of</strong> Chicago in <strong>the</strong> Department<br />

<strong>of</strong> Human Genetics working on cortical<br />

malformations, specifically <strong>the</strong> molecular causes<br />

<strong>of</strong> <strong>the</strong> classical lissencephaly spectrum. Her<br />

clinical expertise lies in dysmorphology and<br />

paediatric neurogenetics, and she continues her<br />

research interest in cortical malformations.<br />

Daniela T. Pilz, MD<br />

Institute <strong>of</strong> Medical Genetics,<br />

University Hospital <strong>of</strong> Wales, Cardiff, UK


The lissencephalies<br />

Cortical malformations are an important cause for<br />

epilepsy and intellectual disability. A significant<br />

subgroup is <strong>the</strong> “lissencephalies”, classified<br />

into Type 1 (classical lissencephaly) and Type<br />

II (cobblestone lissencephaly). The latter is<br />

less common and typically associated with eye<br />

abnormalities and muscular dystrophy (muscleeye-brain<br />

disease spectrum). The molecular<br />

abnormalities identified in <strong>the</strong>se conditions<br />

are associated with abnormal dystroglycan<br />

glycosylation, and <strong>the</strong>y are now <strong>of</strong>ten known as<br />

<strong>the</strong> “dystroglycanopathies”.<br />

In this talk I want to focus on <strong>the</strong> more common<br />

classical (Type 1) lissencephalies (LIS) and<br />

related disorders, lissencephaly with cerebellar<br />

hypoplasia (LCH), and X-linked lissencephaly<br />

with abnormal genitalia (XLAG). Mutations<br />

in LIS1 and DCX are found in about 85% <strong>of</strong><br />

patients with LIS, <strong>the</strong> former associated with<br />

a posterior predominant and <strong>the</strong> latter with an<br />

anterior predominant severity gradient <strong>of</strong> cortical<br />

malformation. Anomalies in <strong>the</strong> recently identified<br />

TUBA1A gene appear to account for some <strong>of</strong> <strong>the</strong><br />

remaining 15%. Their lissencephaly phenotype<br />

seems to show a posterior predominant<br />

severity gradient as seen in association with<br />

LIS1 mutations. However additional structural<br />

anomalies, including callosal and cerebellar<br />

anomalies, appear more prevalent. All 3 genes<br />

ei<strong>the</strong>r interact with or are part <strong>of</strong> <strong>the</strong> cytoskeleton,<br />

which determines cell shape/structure and plays<br />

important roles in mitosis, cell movement and<br />

intracellular transport. Some patients with LCH<br />

have been found to have a RELN mutation,<br />

an extracellular matrix protein required for<br />

normal neuronal migration to <strong>the</strong> appropriate<br />

cortical layers. XLAG is caused by mutations<br />

in <strong>the</strong> transcription factor ARX, thought to be<br />

involved tangential migration <strong>of</strong> neurons during<br />

development.<br />

27


28<br />

ge n e t i c s oF ab n o r m a l br a i n De v e l o P m e n t<br />

Dr. Ute Hehr was born in Dresden, <strong>German</strong>y,<br />

and obtained her formal medical training at <strong>the</strong><br />

University Halle-Wittenberg, where she also<br />

performed mouse studies as part <strong>of</strong> her diploma<br />

<strong>the</strong>sis at <strong>the</strong> Department <strong>of</strong> Biochemistry. Her<br />

laboratory training in molecular cytogenetics was<br />

broadened during a stay at <strong>the</strong> State University <strong>of</strong><br />

Leiden, Ne<strong>the</strong>rlands.<br />

She <strong>the</strong>n received a research grant <strong>of</strong> <strong>the</strong> <strong>German</strong><br />

Academic Exchange Service and in 1993 moved to<br />

Philadelphia to work for 2 years in <strong>the</strong> lab <strong>of</strong> Dr. M.<br />

Münke (Division <strong>of</strong> Human Genetics and Molecular<br />

Biology <strong>of</strong> <strong>the</strong> Children´s Hospital <strong>of</strong> Philadelphia,<br />

University <strong>of</strong> Pennsylvania) on craniosynostosis<br />

and holoprosencephaly. In addition, she pursued<br />

<strong>the</strong>re her training in clinical genetics with Dr. E.H.<br />

Zackai. Dr. Hehr completed her clinical training at<br />

Ute Hehr, MD<br />

Department <strong>of</strong> Human Genetics<br />

University <strong>of</strong> Regensburg, <strong>German</strong>y<br />

<strong>the</strong> University Halle-Wittenberg and in 2000 moved<br />

to Regensburg. Toge<strong>the</strong>r with Dr. B.H.F. Weber she<br />

currently leads <strong>the</strong> Diagnostic Center <strong>of</strong> Human<br />

Genetics at <strong>the</strong> University <strong>of</strong> Regensburg, where<br />

her major research interests include <strong>the</strong> clinical<br />

and molecular genetics <strong>of</strong> crani<strong>of</strong>acial and brain<br />

malformations in collaboration with Dr. G. Uyanik<br />

and Dr. J. Winkler, Department <strong>of</strong> Neurology.<br />

In 2000 she received <strong>the</strong> award <strong>of</strong> <strong>the</strong> Novartis<br />

foundation for <strong>the</strong>rapeutic research. She currently<br />

is president <strong>of</strong> <strong>the</strong> consortium for Reproductive<br />

Genetics <strong>of</strong> <strong>the</strong> <strong>German</strong> <strong>Society</strong> <strong>of</strong> Reproductive<br />

Medicine and in 2004 was appointed as expert <strong>of</strong><br />

<strong>the</strong> National Ethics Council on polar body diagnosis.


Genetics and what a face can tell about <strong>the</strong> brain<br />

Holoprosencephaly (HPE) is a complex brain<br />

malformation, resulting from incomplete cleavage<br />

<strong>of</strong> <strong>the</strong> anterior part <strong>of</strong> <strong>the</strong> forebrain into <strong>the</strong> two<br />

hemispheres and ventricles between days 18 and<br />

28 <strong>of</strong> gestation. A current prevalence <strong>of</strong> about<br />

1.3 : 10 000 live births when compared to 1 : 250<br />

early embryos reflects both, a high intrauterine<br />

lethality as well as a high detection rate during<br />

routine prenatal sonography. The etiology <strong>of</strong> HPE<br />

is very heterogeneous and includes chromosomal<br />

abnormalities, teratogenic effects, monogenic<br />

syndromes as well as familial and sporadic isolated<br />

HPE. Currently, a causal mutation resulting in<br />

monogenic, autosomal dominant inherited HPE<br />

can be identified in about 15-25% <strong>of</strong> HPE patients<br />

with normal chromosomes. For <strong>the</strong>se families<br />

an incomplete penetrance and extremely wide<br />

intrafamilial clinical variability has been observed.<br />

Clinical evaluation <strong>of</strong> <strong>the</strong>se families significantly<br />

expanded <strong>the</strong> spectrum <strong>of</strong> HPE-associated<br />

brain malformations. In particular, <strong>the</strong> additional<br />

distinct heterogeneous entities <strong>of</strong> arrhinencephaly,<br />

syntelencephaly, agenesis <strong>of</strong> <strong>the</strong> corpus callosum,<br />

pituitary anomalies as well as septo-optic dysplasia<br />

are discussed as possible variants <strong>of</strong> <strong>the</strong> HPE<br />

spectrum. In about 70-80 % <strong>of</strong> <strong>the</strong> HPE patients<br />

characteristic crani<strong>of</strong>acial features allow to predict<br />

<strong>the</strong> severity <strong>of</strong> <strong>the</strong> brain malformation („The face<br />

predicts <strong>the</strong> brain“, De Myers et al., 1963). The<br />

spectrum <strong>of</strong> observed facial features includes<br />

cyclopia, proboscis, microphthalmia and median<br />

cleft lip and palate as well as milder manifestations<br />

like hypotelorism or singular median maxillary<br />

incisor. Obligate mutation carriers may present with<br />

such crani<strong>of</strong>acial minor signs and/or microcephaly,<br />

retarded language acquisition or learning problems<br />

as <strong>the</strong> only clinical manifestation. Genetic workup <strong>of</strong><br />

each prenatal and postnatal HPE case should not<br />

only include a chromosome analysis, but also good<br />

cerebral MR imaging as well as a careful search<br />

for associated anomalies in <strong>the</strong> setting <strong>of</strong> a genetic<br />

counseling session. Fur<strong>the</strong>r molecular genetic<br />

testing should be individually assigned based on <strong>the</strong><br />

obtained clinical and anamnestic data. Fur<strong>the</strong>rmore,<br />

genetic workup <strong>of</strong> HPE patients is also providing<br />

important insights into early forebrain development.<br />

Toge<strong>the</strong>r with animal models two current concepts<br />

emerge: most <strong>of</strong> classic HPE appears to result<br />

from direct or indirect disruption <strong>of</strong> <strong>the</strong> ventralizing<br />

effect <strong>of</strong> sonic hedgehog signaling and usually<br />

is accompanied by <strong>the</strong> characteristic HPE facial<br />

gestalt. In contrast, disruption <strong>of</strong> <strong>the</strong> dorsalizing<br />

effect <strong>of</strong> bone morphogenic protein signaling may<br />

account for <strong>the</strong> middle interhemispheric variants.<br />

29


30<br />

ge n e t i c s oF ab n o r m a l br a i n De v e l o P m e n t<br />

Pr<strong>of</strong>essor Dr. Kerstin Kutsche is a native <strong>of</strong><br />

Hannover, <strong>German</strong>y. She attended <strong>the</strong> Universities<br />

<strong>of</strong> Hannover and Bielefeld, <strong>German</strong>y, to study<br />

Biology and performed her PhD in genetics <strong>of</strong><br />

prokaryotes under <strong>the</strong> supervision <strong>of</strong> Pr<strong>of</strong>essor<br />

Dr. Werner Klipp. She moved to Freiburg and<br />

Heidelberg, <strong>German</strong>y, from 1994-1996 for a Post<br />

Doc under <strong>the</strong> mentorship <strong>of</strong> Pr<strong>of</strong>essor Dr. Thomas<br />

Boehm to study <strong>the</strong> winged-helix transcription<br />

factor Foxn1.<br />

In 1996, Dr. Kutsche joined <strong>the</strong> Department <strong>of</strong><br />

Human Genetics at <strong>the</strong> University Medical Center<br />

Hamburg-Eppendorf, Hamburg, <strong>German</strong>y, with<br />

Andreas Gal as Chairman. Since <strong>the</strong>n she has<br />

been working on <strong>the</strong> identification <strong>of</strong> novel disease<br />

genes with <strong>the</strong> help <strong>of</strong> balanced chromosomal<br />

rearrangements. She also performs functional<br />

Kerstin Kutsche, PhD<br />

Institute <strong>of</strong> Human Genetics<br />

University <strong>of</strong> Hamburg, <strong>German</strong>y<br />

studies on selected proteins encoded by disease<br />

genes. She was appointed a C3 Pr<strong>of</strong>essor <strong>of</strong><br />

Molecular Human Genetics at <strong>the</strong> Department <strong>of</strong><br />

Human Genetics <strong>of</strong> Hamburg University in 2004.<br />

Dr. Kutsche did her “Habilitation” in 2005.<br />

She completed her training in Human Genetics in<br />

2006 with <strong>the</strong> degree “Fachhumangenetikerin”.<br />

She has published about 40 scientific papers.


Mutations <strong>of</strong> CASK cause a novel X-linked brain<br />

malformation phenotype with microcephaly and hypoplasia<br />

<strong>of</strong> <strong>the</strong> brainstem and cerebellum<br />

Microcephaly occurs as an isolated malformation<br />

or with associated brain anomalies such as a<br />

simplified gyral pattern or cerebellar hypoplasia.<br />

Recently, homozygous silencing <strong>of</strong> EOMES<br />

(TBR2), encoding a transcription factor <strong>of</strong> <strong>the</strong><br />

brain-specific T-box family, was associated with<br />

microcephaly and additional brain malformations.<br />

TBR2 has a putative function in regulating<br />

cortical neurogenesis and neural identity, and<br />

plays a role in a signalling cascade upstream<br />

<strong>of</strong> TBR1, which is highly related to TBR2. We<br />

describe a novel X-linked brain malformation<br />

phenotype with postnatal microcephaly, hypoplasia<br />

<strong>of</strong> <strong>the</strong> cerebellum and brainstem, and severe<br />

developmental delay that is caused by mutations<br />

<strong>of</strong> CASK in Xp11.4. We mapped one breakpoint<br />

<strong>of</strong> a paracentric Xp inversion within <strong>the</strong> CASK<br />

gene in an affected female. By array comparative<br />

genomic hybridization we identified a heterozygous<br />

deletion <strong>of</strong> ~740 kb encompassing CASK, GPR34<br />

and GPR82 in female patient 2 and two separated<br />

regions <strong>of</strong> copy number loss in female patient 3,<br />

including ~170 kb covering <strong>the</strong> 3’ and ~150 kb<br />

encompassing <strong>the</strong> 5’ region <strong>of</strong> CASK. Mutation<br />

analysis <strong>of</strong> CASK in 13 females and 33 males with<br />

a phenotype similar to patients 1-3 identified a<br />

heterozygous nonsense mutation (c.1915C>T/p.<br />

R639X) in a female and <strong>the</strong> hemizygous c.915G>A<br />

(p.K305) mutation altering splicing in a severely<br />

affected male who died at age 2 weeks. CASK<br />

encodes a multi-domain scaffolding protein that<br />

interacts with <strong>the</strong> transcription factor TBR1 and<br />

regulates expression <strong>of</strong> genes involved in cortical<br />

development, such as RELN. The potential<br />

importance <strong>of</strong> <strong>the</strong> CASK-TBR1-RELN signaling<br />

cascade in brain development is highlighted by our<br />

neuropathological findings in <strong>the</strong> deceased boy,<br />

which show changes nearly identical to <strong>the</strong> Reln<br />

mouse mutant in <strong>the</strong> cerebellum, and overlap with<br />

<strong>the</strong> Tbr1�/� mouse in <strong>the</strong> cerebral cortex.<br />

31


32<br />

ge n e t i c s oF ab n o r m a l br a i n De v e l o P m e n t<br />

Connie Schrander-Stumpel became a<br />

paediatrician in 1984 in Amsterdam. In 1985<br />

she started her carrier in clinical genetics at <strong>the</strong><br />

Maastricht University. Since 1989 she is a clinical<br />

geneticist. From 1985 on she is head <strong>of</strong> <strong>the</strong><br />

outclinic Clinical Genetics and since 1992 head <strong>of</strong><br />

<strong>the</strong> training program for clinical genetics with, in<br />

2008, 7 residents.<br />

X-linked hydrocephalus was <strong>the</strong> topic <strong>of</strong> her<br />

<strong>the</strong>sis in 1994, and research in this area<br />

continues.<br />

In 2002 she became Pr<strong>of</strong>essor <strong>of</strong> Clinical<br />

Genetics, especially for Dysmorphology and<br />

Mental Retardation. The research mainly focuses<br />

on Prader-Willi -, Rett -, and Kabuki syndrome.<br />

Constance T.R.M. Schrander-Stumpel, MD<br />

Department <strong>of</strong> Clinical Genetics<br />

Research Institute Growth and Development (GROW)<br />

Maastricht University, The Ne<strong>the</strong>rlands


Congenital hydrocephalus:<br />

The Maastricht experience and <strong>the</strong> concept <strong>of</strong> ciliopathy<br />

Retrospectively, data <strong>of</strong> 66 individuals with<br />

congenital hydrocephalus were reviewed. Nonsyndromic<br />

hydrocephalus (NSH) was identified<br />

in 25/66 and hydrocephalus “plus” (H+) in<br />

41/66 patients. In 55/66 patients <strong>the</strong> cause <strong>of</strong><br />

hydrocephalus was unknown. The male/female<br />

ratio was 7.8:1, strongly indicating an X-factor.<br />

In <strong>the</strong> group <strong>of</strong> 41 H+ patients, 11 had a known<br />

cause <strong>of</strong> hydrocephalus. Three out <strong>of</strong> 4 familial<br />

cases with an unknown cause were possibly<br />

X-linked. Within <strong>the</strong> group <strong>of</strong> patients with an<br />

unknown cause <strong>of</strong> H+ (30/41), 5/30 patients<br />

had renal abnormalities, 3/30 had congenital<br />

diaphragmatic hernia. 5/30 patients had clinical<br />

features overlapping with those present in<br />

chromosome breakage disorders, e.g. radial<br />

ray defects or VACTERL-H as seen in FANCB<br />

gene defect. Especially in <strong>the</strong> groups with renal<br />

malformations and VACTERL-H with radial<br />

ray defects a ciliopathy may be involved in its<br />

pathogenesis. Congenital hydrocephalus is<br />

reported in about 20% <strong>of</strong> <strong>the</strong> patients with cilia<br />

disorders.<br />

We characterized a spontaneous mouse mutant<br />

with a (new) autosomal recessive form <strong>of</strong><br />

congenital hydrocephalus, cerebellar hypoplasia,<br />

growth retardation, and situs inversus/<br />

heterotaxia in about 20% <strong>of</strong> <strong>the</strong> affected mice.<br />

Electron microscopic studies <strong>of</strong> ependymal cilia<br />

demonstrate an axonemal outer dynein arm<br />

(ODA) defect.<br />

We developed a new fast one-day functional<br />

motile cilia assay to identify immotile ependymal<br />

cilia in <strong>the</strong>se mice. Real time (semi)quantitative<br />

mRNA studies <strong>of</strong> ependymal cilia enriched brain<br />

cDNA excluded 6 <strong>of</strong> <strong>the</strong> known ODA genes:<br />

Dnahc5, Dnahc9, Dnahc11/lrd, Dnaic1, Dnalc1<br />

and Dnalc4 in <strong>the</strong> affected mice.<br />

Mapping procedures combined with<br />

immunocytochemistry/-electronmicroscopy<br />

and fur<strong>the</strong>r candidate gene analysis is ongoing<br />

to identify a possibly new candidate gene for<br />

autosomal recessive congenital hydrocephalus.<br />

33


34<br />

ge n e t i c s oF Ps y c h i a t r i c Di s e a s e s<br />

I began my research career in 1989 as an<br />

MRC Training Fellow, where I developed novel<br />

methods for quantitating mRNA and used <strong>the</strong>se<br />

to show relative expression <strong>of</strong> genes encoding<br />

subunits <strong>of</strong> GABAA receptors was a mechanism<br />

that contributes <strong>the</strong> biology <strong>of</strong> benzodiazepine<br />

tolerance. From 1992-1993, I was an MRC<br />

Travelling Fellow at MIT where I was a member<br />

<strong>of</strong> The Huntington‘s Disease Collaborative<br />

Research Group, a team <strong>of</strong> scientists from leading<br />

genetics institutions in Europe and <strong>the</strong> USA. The<br />

group successfully identified <strong>the</strong> gene causing<br />

Huntington‘s disease in 1993. In 1993, I was<br />

appointed Senior Lecturer in <strong>the</strong> Department<br />

<strong>of</strong> Psychological Medicine, University <strong>of</strong> Wales<br />

College <strong>of</strong> Medicine, and was awarded a chair<br />

in Psychiatric Genetics in 1999. I have a broad<br />

interest in <strong>the</strong> molecular genetics <strong>of</strong> mental<br />

Michael O’Donovan, MD<br />

Department <strong>of</strong> Psychological Medicine<br />

Cardiff University, UK<br />

disorders and hold grants from major funding<br />

bodies (MRC, Wellcome Trust, National Institutes<br />

<strong>of</strong> Health (USA) for molecular genetic studies <strong>of</strong><br />

schizophrenia, bipolar disorder, dyslexia, ADHD,<br />

and Alzheimer’s disease.


Schizophrenia genetics: new insights from new approaches<br />

It has long been known that <strong>the</strong>re is a strong<br />

genetic contribution to schizophrenia, but<br />

identifying <strong>the</strong> specific genes responsible for this<br />

disorder has proven to be a major challenge.<br />

Over <strong>the</strong> past 10 years or so, molecular genetic<br />

approaches to complex diseases in general have<br />

progressed from being small sample studies<br />

based upon at best a handful <strong>of</strong> genetic markers<br />

into large sample studies <strong>of</strong> several hundred<br />

thousand markers with <strong>the</strong> power to extract a high<br />

proportion <strong>of</strong> all common genetic variation in <strong>the</strong><br />

genome. While few studies on this scale have<br />

yet been performed in schizophrenia, <strong>the</strong> early<br />

signs are striking. In <strong>the</strong> first few large studies <strong>of</strong><br />

common variation, <strong>the</strong> new genome-wide approach<br />

at last has provided robust replicable evidence<br />

for <strong>the</strong> involvement <strong>of</strong> specific common genetic<br />

variants in <strong>the</strong> disorder. As a bonus, <strong>the</strong> same<br />

technologies have now for <strong>the</strong> first time provided<br />

<strong>the</strong> capacity to systematically scan <strong>the</strong> genome<br />

for certain types <strong>of</strong> rare variant known as CNVs or<br />

copy number variants representing chromosome<br />

deletions and duplications. Again, <strong>the</strong> results are a<br />

dramatic break with <strong>the</strong> past, with clear replicable<br />

evidence for <strong>the</strong> involvement <strong>of</strong> specific copy<br />

number variants emerging from <strong>the</strong> first two large<br />

studies performed. In this presentation, I will review<br />

what is currently known about <strong>the</strong> genetics <strong>of</strong><br />

schizophrenia with an emphasis on <strong>the</strong> novel but<br />

robust findings.<br />

35


36<br />

ge n e t i c s oF Ps y c h i a t r i c Di s e a s e s<br />

Markus Nö<strong>the</strong>n received his MD degree from<br />

<strong>the</strong> University <strong>of</strong> Würzburg. He was trained as a<br />

human geneticist at <strong>the</strong> University <strong>of</strong> Bonn and<br />

worked <strong>the</strong>re till 2001 as a clinical geneticist<br />

and scientist. From 2001 to 2004 he has been<br />

Pr<strong>of</strong>essor <strong>of</strong> Medical Genetics and Director <strong>of</strong><br />

<strong>the</strong> Center <strong>of</strong> Medical Genetics at <strong>the</strong> University<br />

<strong>of</strong> Antwerp, Belgium. In 2004 Markus Nö<strong>the</strong>n<br />

became <strong>the</strong> Alfried Krupp von Bohlen und<br />

Halbach Pr<strong>of</strong>essor <strong>of</strong> Genetic Medicine at <strong>the</strong><br />

University <strong>of</strong> Bonn. Since 2001 he is founding<br />

head <strong>of</strong> <strong>the</strong> Department <strong>of</strong> Genomics at <strong>the</strong><br />

Life & Brain Research Center and since 2004<br />

Director <strong>of</strong> <strong>the</strong> Institute <strong>of</strong> Human Genetics at <strong>the</strong><br />

University <strong>of</strong> Bonn.<br />

His research interest lies in <strong>the</strong> genetic analysis<br />

<strong>of</strong> complex diseases. He is coordinator <strong>of</strong><br />

Markus M. Nö<strong>the</strong>n, MD<br />

Institute <strong>of</strong> Human Genetics<br />

University <strong>of</strong> Bonn, <strong>German</strong>y<br />

<strong>the</strong> NGFNplus Integrated Genome Research<br />

Network “Systematic Investigation <strong>of</strong> <strong>the</strong><br />

Molecular Causes <strong>of</strong> Major Mood Disorders and<br />

Schizophrenia (MooDS)”.


The Genetics <strong>of</strong> Bipolar Affective Disorder<br />

Bipolar affective disorder (BPD), also known as<br />

manic depressive illness, is a common psychiatric<br />

disorder which imposes a severe burden on <strong>the</strong><br />

individual and society. Family, twin, and adoption<br />

studies provide consistent evidence for a major<br />

genetic contribution to <strong>the</strong> etiology <strong>of</strong> BPD, with<br />

heritability estimates <strong>of</strong> around 85 %.<br />

The first susceptibility genes were proposed<br />

based on candidate gene and linkage-based<br />

positional cloning attempts. At best, <strong>the</strong>se genes<br />

explain only a small fraction <strong>of</strong> <strong>the</strong> genetic risk<br />

<strong>of</strong> <strong>the</strong> disorders, however. Recently, <strong>the</strong> first<br />

genome wide association studies have been<br />

performed for BPD, revealing some points <strong>of</strong><br />

agreement between studies. In contrast to<br />

common diseases, for which genes with larger<br />

affects exist, BPD seems to be truly polygenic,<br />

and <strong>the</strong>se first studies are currently followed by<br />

large collaborative studies to attribute a causal<br />

relationship <strong>of</strong> specific genes with BPD robustly.<br />

The talk will give an update on <strong>the</strong> latest findings<br />

in BPD.<br />

37


38<br />

ge n e t i c s oF Ps y c h i a t r i c Di s e a s e s<br />

Dr. Susanne Lucae was born in Dornbirn, Austria.<br />

She studied biology and medicine at <strong>the</strong> University<br />

<strong>of</strong> Vienna, Austria and completed her PhD <strong>the</strong>sis<br />

at <strong>the</strong> Institute <strong>of</strong> Pathophysiology, University <strong>of</strong><br />

Vienna, where she also spent her postdoctural.<br />

In 2003 she moved to <strong>the</strong> Max-Planck-Institute<br />

<strong>of</strong> Psychiatry in Munich, <strong>German</strong>y, where she<br />

presently works as staff member <strong>of</strong> <strong>the</strong> outpatient<br />

clinic <strong>of</strong> psychiatry. Dr. Lucae has been head <strong>of</strong> <strong>the</strong><br />

research group “Psychiatric Pharmacogenetics” at<br />

<strong>the</strong> MPI in Munich since 2007.<br />

Susanne Lucae, MD<br />

Max-Planck-Institute <strong>of</strong> Psychiatry<br />

Munich, <strong>German</strong>y


Genetics <strong>of</strong> <strong>the</strong> response to antidepressant treatment<br />

Major depression is by far <strong>the</strong> most prevalent<br />

mental disorder frequently characterized by<br />

recurrent episodes or chronicity. Despite a variety<br />

<strong>of</strong> available treatments only 60% <strong>of</strong> severely<br />

depressed patients achieve full remission <strong>of</strong><br />

depressive symptoms, even after several treatment<br />

trials. Both, clinical characteristics and genetic<br />

factors influence <strong>the</strong> likelihood <strong>of</strong> a favourable<br />

treatment outcome.<br />

Genome-wide pharmacogenetic association<br />

studies in 339 depressed inpatients from <strong>the</strong><br />

Munich Antidepressant Response Signature<br />

(MARS) project and in a second independent<br />

sample <strong>of</strong> 361 depressed inpatients were<br />

performed. All patients were <strong>of</strong> Caucasian<br />

descent. 328 SNPs with <strong>the</strong> highest association<br />

with treatment outcome were <strong>the</strong>n genotyped in<br />

a Caucasian depressed outpatients subsample<br />

(n=832) from <strong>the</strong> Sequenced Treatment<br />

Alternatives to Relieve Depression (STAR*D) study<br />

selected based on clinical criteria from <strong>the</strong> initial<br />

samples.<br />

46 SNPs could be replicated at a nominal level <strong>of</strong><br />

significance, which did not withstand correction for<br />

multiple testing. However, a multi-locus analysis<br />

that assumed additivity <strong>of</strong> <strong>the</strong> investigated SNPs<br />

revealed a significant contribution <strong>of</strong> <strong>the</strong> number<br />

<strong>of</strong> responses alleles combined with clinical<br />

characteristics to predict treatment outcome<br />

in both MARS and STAR*D samples. These<br />

findings constitute <strong>the</strong> first empirical evidence<br />

that many genetic variants contribute to treatment<br />

outcome in major depression in a cumulative<br />

way corroborating <strong>the</strong> multifactorial nature <strong>of</strong> this<br />

complex clinical trait.<br />

39


40<br />

ge n e t i c s oF Ps y c h i a t r i c Di s e a s e s<br />

Jürgen Deckert graduated from Würzburg<br />

medical school in 1984. He worked in <strong>the</strong> Unit<br />

on Neurochemistry, Biological Psychiatry Branch,<br />

NIMH from 1985-1988 and in <strong>the</strong> Department<br />

<strong>of</strong> Human Genetics, University <strong>of</strong> Bonn from<br />

1994-1996.<br />

In 1992 he obtained his board license as a<br />

psychiatrist and in 1997 his habilitation in<br />

psychiatry at <strong>the</strong> Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Würzburg. From 1999-2006 he was<br />

vice director and finally pr<strong>of</strong>essor for molecular<br />

psychiatry at <strong>the</strong> Department <strong>of</strong> Psychiatry,<br />

University <strong>of</strong> Münster. In 2006 he moved back<br />

to Würzburg to become department director and<br />

pr<strong>of</strong>essor for psychiatry.<br />

Jürgen Deckert, MD<br />

Department <strong>of</strong> Psychiatry, Psychosomatics and Psycho<strong>the</strong>rapy<br />

University <strong>of</strong> Würzburg, <strong>German</strong>y


Genetics <strong>of</strong> anxiety disorders<br />

Panic disorder with a life-time prevalence <strong>of</strong> 2-3%<br />

is not <strong>the</strong> most common, but <strong>the</strong> most severe<br />

anxiety disorder and according to <strong>the</strong> WHO ranks<br />

among <strong>the</strong> 10 most disabling neuropsychiatric<br />

disorders among middle-aged Europeans and<br />

North Americans.<br />

Its heritability based on twin and family studies<br />

is estimated at about 50%. Segregation studies<br />

though failed to demonstrate Mendelian<br />

inheritance. Accordingly, only a few linkage<br />

studies have been published and proposed<br />

loci on chromosome 1, 4, 9, 13, 18 and 22 are<br />

in need <strong>of</strong> replication. In contrast, numerous<br />

association studies with candidate genes have<br />

been published. Candidates were chosen on<br />

<strong>the</strong> basis <strong>of</strong> <strong>the</strong> known molecular mechanisms<br />

<strong>of</strong> <strong>the</strong>rapeutic drugs or anxiety provoking<br />

agents, on <strong>the</strong> basis <strong>of</strong> our knowledge on <strong>the</strong><br />

pathophysiology <strong>of</strong> anxiety and more recently, on<br />

<strong>the</strong> basis <strong>of</strong> animal models. As to be expected<br />

numerous modest associations were reported,<br />

<strong>of</strong> which, however only few (e.g. COMT, MAO-A,<br />

A2A, CCKRB) could be confirmed in independent<br />

replication studies.<br />

Functional relevance <strong>of</strong> some <strong>of</strong> <strong>the</strong> associated<br />

polymorphisms were confirmed by means <strong>of</strong><br />

cellular, pharmacological or imaging studies<br />

providing in addition hints as to potential geneenvironment<br />

interactions. At present, <strong>the</strong> first<br />

genome-wide association studies are being<br />

performed with preliminary results pointing to a<br />

role for genes hi<strong>the</strong>rto unknown to be involved in<br />

<strong>the</strong> pathophysiology <strong>of</strong> anxiety.<br />

41


42<br />

Fre e co m m u n i c a t i o n<br />

Regulation <strong>of</strong> Astrocyte Inflammatory Responses by <strong>the</strong><br />

Parkinson’s Disease-Associated DJ-1<br />

Jens Waak, 1 Stephanie S. Weber, 1 Daniela Vogt-Weisenhorn, 2 Thu-Trang Pham, 2<br />

Karin Görner, 3 Marianna Alunni-Fabbroni, 3 Wolfgang Wurst, 2 and Philipp J. Kahle1 1 Laboratory <strong>of</strong> Functional <strong>Neurogenetics</strong>, Department <strong>of</strong> Neurodegeneration, Hertie<br />

Institute for Clinical Brain Research, University Clinics, 72076 Tübingen, <strong>German</strong>y<br />

2 Institute <strong>of</strong> Developmental Genetics, Helmholtz Center Munich, <strong>German</strong> Research<br />

Center for Environmental Health, 85764 Neuherberg, <strong>German</strong>y<br />

3 Olympus Life Science Research Europe, BBD, Advalytix Products, 81377 Munich,<br />

<strong>German</strong>y<br />

Mutations <strong>of</strong> DJ-1 cause autosomal-recessive<br />

Parkinson’s disease (PD). In addition to neuronintrinsic<br />

protective functions, its up-regulation<br />

in reactive astrocytes also suggests a glial<br />

contribution <strong>of</strong> DJ-1. Here we show in astrocyterich<br />

primary cultures derived from DJ-1 deficient<br />

mice that DJ-1 regulates pro-inflammatory<br />

responses. When treated with lipopolysaccharide<br />

as well as α-synuclein protein, DJ-1 mutant<br />

astrocytes generated ≈10 times more nitric oxide<br />

(NO) than littermate controls. This effect was<br />

mediated by hyper-stimulation <strong>of</strong> type II NO<br />

synthase (iNOS) via <strong>the</strong> p38MAPK pathway.<br />

Inhibition <strong>of</strong> this pathway with SB203580<br />

suppressed NO production and iNOS induction.<br />

Stimulation <strong>of</strong> p38MAPK in astrocytes was also<br />

detected in DJ-1-/- mice treated systemically<br />

with LPS. DJ-1 most likely regulates <strong>the</strong> p38<br />

pathway by modulating <strong>the</strong> activity <strong>of</strong> Apoptosis<br />

Signal Regulating Kinase (ASK-1) activity. We<br />

identified ASK-1 as <strong>the</strong> main target <strong>of</strong> regulation<br />

<strong>of</strong> <strong>the</strong> ASK/JNK/p38 MAPK pathway by DJ-1.<br />

DJ-1 directly interacts with ASK-1 and suppresses<br />

its ROS-mediated activation. In addition to<br />

iNOS, DJ-1 mutant astrocytes hyper-induced<br />

cyclooxygenase-2 and interleukin-6. These<br />

findings identify DJ-1 is a major regulator <strong>of</strong> proinflammatory<br />

responses in astrocytes and suggest<br />

that loss <strong>of</strong> DJ-1 contributes to PD pathogenesis<br />

by deregulation <strong>of</strong> neuroinflammatory damage.


Abnormal basal ganglia activity during response selection in<br />

Parkin mutation carriers<br />

J.P.M. van der Vegt 1,3 , M. M. Weiss 1 , B.R. Bloem 3 , F. Bink<strong>of</strong>ski 2 , C Klein 2 , H. R.<br />

Siebner 1<br />

1 Dept. <strong>of</strong> Neurology, Christian-Albrechts-University Kiel, <strong>German</strong>y<br />

2 Dept. <strong>of</strong> Neurology, University Lübeck, <strong>German</strong>y<br />

3 Radboud University Medical Centre, Nijmegen, The Ne<strong>the</strong>rlands<br />

Objective: Asymptomatic carriers <strong>of</strong> a single mutant<br />

Parkin allele show a latent nigrostriatal dopaminergic<br />

dysfunction. We examined how <strong>the</strong> presence <strong>of</strong> a<br />

mutant Parkin allele influences task related acvtivity<br />

in <strong>the</strong> basal ganglia when subjects have to select<br />

a movement in <strong>the</strong> context <strong>of</strong> competing response<br />

tendencies.<br />

Methods: Asymptomatic individuals carrying a<br />

single mutant Parkin allele (n=6) and healthy agematched<br />

controls (n=10) performed a Simon task<br />

during fMRI at 3T. Responses are coded by two<br />

symbolic cues in this two-choice reaction time<br />

task. The position <strong>of</strong> <strong>the</strong>se cues is ei<strong>the</strong>r spatially<br />

compatible or incompatible with <strong>the</strong> response<br />

instructed by <strong>the</strong> symbolic cue. We chose <strong>the</strong> Simon<br />

task because patients with Parkinson´s disease are<br />

impaired at resolving <strong>the</strong> conflict induced by spatial<br />

incompatibility (Praamstra & Plat, 2001). Mean<br />

reaction times (RT) and error rates (ER) during fMRI<br />

were analyzed using ANOVA. Task-related BOLD<br />

signal changes were analysed using SPM2 s<strong>of</strong>tware.<br />

Only correct responses were included in <strong>the</strong>se<br />

analyses.<br />

Results: Participants respond faster when relative<br />

spatial positions <strong>of</strong> stimulus and response match.<br />

Incompatible trials were associated with higher<br />

ER in both groups. Parkin mutation carriers made<br />

significantly more errors than healthy non-carriers<br />

in compatible and incompatible trials. No significant<br />

differences in RT were found between groups.<br />

Analysis <strong>of</strong> <strong>the</strong> fMRI data revealed an increased<br />

activation <strong>of</strong> <strong>the</strong> left anterior putamen during spatially<br />

incompatible trials in Parkin mutation carriers relative<br />

to non-mutation carriers. This hyperactivity was only<br />

present in right hand responses. No differences<br />

between groups were found during spatially<br />

compatible trials.<br />

Conclusions: The fMRI results indicate that a<br />

mutant Parkin allele is associated with overactivity<br />

<strong>of</strong> <strong>the</strong> basal ganglia when a correct action has to<br />

be selected in <strong>the</strong> context <strong>of</strong> conflicting response<br />

tendencies, due to a latent dopaminergic dysfunction.<br />

Given <strong>the</strong> overall increase in error rate, this may<br />

indicate basal ganglia dysfunction associated with<br />

latent nigrostriatal dopaminergic degeneration.<br />

43


44<br />

Fre e co m m u n i c a t i o n s<br />

Pathogenic HSP gene duplications and protection <strong>of</strong> female<br />

carriers<br />

Christian Beetz 1 , Anders O.H. Nygren 2 , Rebecca Schüle 3 , Ludger Schöls 3 ,<br />

Thomas Deufel 1<br />

1 Institut für Klinische Chemie und Laboratoriumsdiagnostik, Universitätsklinikum,<br />

Jena, <strong>German</strong>y<br />

2 MRC-Holland, Amsterdam, The Ne<strong>the</strong>rlands<br />

3 Hertie Institut für Klinische Hirnforschung, Tübingen, <strong>German</strong>y<br />

Background: Hereditary spastic paraplegias<br />

(HSP) are neurodegenerative disorders primarily<br />

affecting lower limb movement. The two major<br />

disease genes SPG3A and SPG4 are associated<br />

with autosomal dominant (AD) inheritance.<br />

Mutational class in <strong>the</strong>se genes does not predict<br />

age at onset (aao).<br />

Objective: To screen AD-HSP patients for copy<br />

number aberrations in SPG3A and SPG4.<br />

Methods: A multiplex ligation-dependent probe<br />

amplification assay simultaneously targeting both<br />

genes was applied to index patients for which<br />

“small” mutations had been excluded.<br />

Results: Large deletions affecting SPG4 were<br />

frequently identified; aao in <strong>the</strong>se patients<br />

was as in o<strong>the</strong>r cases <strong>of</strong> SPG4 HSP. A single<br />

SPG3A whole gene deletion was apparently non-<br />

pathogenic. The only instance <strong>of</strong> a (partial) SPG4<br />

duplication was found in a large Brazilian pedigree<br />

where male carriers (n=14) showed aao typical for<br />

SPG4 HSP (median 30 years) whereas females<br />

carriers (n=13) were significantly protected from<br />

developing <strong>the</strong> disease (median aao >67 years)<br />

(p=0.017). An SPG3A whole gene triplication<br />

was found in an early onset male index patient,<br />

in his similarly affected male cousin, and in <strong>the</strong>ir<br />

unaffected mo<strong>the</strong>rs (both > 50 years <strong>of</strong> age). Of<br />

three related patients, which have not yet been<br />

genotyped, two males show early onset whereas<br />

one female started to develop mild symptoms<br />

in her fifties. In addition, <strong>the</strong>re is ano<strong>the</strong>r seven<br />

females representing potential carriers but not<br />

being affected.<br />

Conclusions: It appears that female carriers <strong>of</strong><br />

SPG3A and SPG4 amplifications are less likely<br />

to develop <strong>the</strong> typical phenotype associated with<br />

mutations in <strong>the</strong>se HSP genes. The underlying<br />

mechanisms are unclear at present and can only<br />

be speculated about.


Novel Mutations in <strong>the</strong> Senataxin Gene in Six Patients<br />

Affected by Ataxia-oculomotor Apraxia 2<br />

Veronica Bernard 1 , Ulf Edener 1 , Friedmar Kreuz 2 , Martina Minnerop 3 , Gabriele<br />

Gillessen-Kaesbach 1 , Christine Zühlke 1<br />

1 Institut für Humangenetik, Universität Lübeck<br />

2 Praxis für Humangenetik, Berlin<br />

3 Klinik für Neurologie, Universität Bonn<br />

Background: The recessively inherited<br />

ataxia-oculomotor apraxia 2 (AOA2) is a<br />

neurodegenerative disorder characterised by<br />

adolescent age at onset, gait ataxia, cerebellar<br />

atrophy, axonal sensorimotor neuropathy,<br />

oculomotor apraxia, and elevated serum AFP level.<br />

AOA2 is caused by mutations within <strong>the</strong> senataxin<br />

gene (SETX) that is localised on chromosome<br />

9q34. The SETX gene encodes a large 2677<br />

amino acid protein, named senataxin. The protein<br />

senataxin is <strong>the</strong> ortholog <strong>of</strong> <strong>the</strong> yeast RNA helicase<br />

Sen1p and contains a DNA/RNA helicase domain<br />

at its C terminus and a putative protein interaction<br />

domain at its N terminus.<br />

Objective and Methods: We screened six patients<br />

showing a clinical phenotype consistent with<br />

AOA2 and for mutations in <strong>the</strong> SETX gene. We<br />

sequenced all 24 coding exons flanking intronic<br />

sequences <strong>of</strong> <strong>the</strong> SETX gene, to confirm <strong>the</strong><br />

molecular genetic diagnosis in <strong>the</strong>se patients.<br />

Results: Sequence analysis revealed 12 novel<br />

DNA variations: patient 1 was homo-zygous for<br />

<strong>the</strong> missense mutation R2414Q, patient 2 showed<br />

homozygosity for <strong>the</strong> amino acid exchange K992R<br />

and <strong>the</strong> missense mutation R2444H. The o<strong>the</strong>r<br />

four patients were compound heterozygous for<br />

different mutations in <strong>the</strong> SETX gene. Patients 3<br />

and 4 carried <strong>the</strong> missense mutations L2155W and<br />

K2382E, and D2207V and T2373P, respectively.<br />

The missense mutation R2444C and a 2 bpdeletion<br />

(c.4095_4096delTT) were found in<br />

patient 5, as well as <strong>the</strong> unknown polymorphism<br />

R20H. Patient 6 was heterozygous for <strong>the</strong><br />

missense mutation P311L and a four-nucleotide<br />

deletion affecting <strong>the</strong> 5’ splice site <strong>of</strong> exon 22<br />

(c.6847_6850delACAG).<br />

Conclusion: Remarkably, most <strong>of</strong> <strong>the</strong>se mutations<br />

are located in <strong>the</strong> DNA/RNA helicase domain,<br />

which is reported to span amino acid residue 1931<br />

to 2456. Although <strong>the</strong> function <strong>of</strong> <strong>the</strong> human protein<br />

is still unknown, <strong>the</strong> correct function <strong>of</strong> <strong>the</strong> helicase<br />

domain seems to be important, to prevent <strong>the</strong><br />

AOA2 phenotype.<br />

45


46<br />

Fre e co m m u n i c a t i o n s<br />

Mutations in FOXG1 cause a broad phenotypic spectrum incl.<br />

microcephaly, frontal pachygyria + agenesis <strong>of</strong> <strong>the</strong> corpus callosum<br />

Fanny Kortüm 1 , William B. Dobyns 2 , Ute Hehr 3 , Gökhan Uyanik 1,4 , and Kerstin<br />

Kutsche 1<br />

1 Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Hamburg,<br />

<strong>German</strong>y<br />

2 Departments <strong>of</strong> Human Genetics, Neurology and Pediatrics, University <strong>of</strong> Chicago,<br />

Chicago, Illinois, USA<br />

3 Zentrum für Humangenetik und Institut für Humangenetik, Universität Regensburg,<br />

Regensburg, <strong>German</strong>y<br />

4 Klinik und Poliklinik für Neurologie, Universität Regensburg, Regensburg, <strong>German</strong>y<br />

Background: Heterozygous loss-<strong>of</strong>-function<br />

mutations in FOXG1, located in 14q12, have been<br />

reported in two patients showing neurological<br />

symptoms consistent with <strong>the</strong> diagnosis <strong>of</strong> a<br />

congenital variant <strong>of</strong> Rett syndrome. They also had<br />

microcephaly and <strong>the</strong>ir MRI scans revealed corpus<br />

callosum hypoplasia. FOXG1 encodes a winged<br />

helix transcriptional factor <strong>of</strong> <strong>the</strong> forkhead protein<br />

family which is mainly expressed in brain and<br />

regulates early steps in cortical development.<br />

Objective: We wanted to explore <strong>the</strong> phenotypic<br />

spectrum associated with a FOXG1 mutation.<br />

Therefore, we investigated a female patient<br />

with a balanced de novo 2;14 translocation<br />

[46,XX,t(2;14)(q11.2;q12)]. She had postnatal<br />

microcephaly, seizures, mental retardation, and<br />

a gastroesophageal reflux. Selected MRI images<br />

showed subtle to mild pachygyria over <strong>the</strong> anterior<br />

frontal lobe, a mildly thickened cortex in <strong>the</strong> affected<br />

area, and a thin and poorly formed corpus callosum.<br />

We also performed FOXG1 mutation analysis in a<br />

patient cohort with mental retardation, microcephaly<br />

and pachygyria.<br />

Methods: We mapped <strong>the</strong> breakpoints <strong>of</strong> <strong>the</strong> 2;14<br />

translocation by fluorescence in situ hybridization<br />

and sequenced <strong>the</strong> FOXG1 gene in 36 patients.<br />

Results: The translocation breakpoint in 14q12 was<br />

located ~265 kb downstream <strong>of</strong> FOXG1, suggesting<br />

that a position effect causes transcriptional<br />

misregulation <strong>of</strong> FOXG1. We detected <strong>the</strong><br />

heterozygous de novo missense mutation<br />

c.757A>G (p.N253D) in FOXG1 in a female patient.<br />

Asparagine at position 253 is an invariant residue<br />

located in <strong>the</strong> DNA-binding domain <strong>of</strong> FOXG1. MRI<br />

images <strong>of</strong> <strong>the</strong> affected girl showed mild pachygyria<br />

<strong>of</strong> <strong>the</strong> anterior-middle frontal lobe, moderately<br />

thickened cortex in abnormal areas frontally and<br />

a normal corpus callosum. She is mildly mentally<br />

retarded and did not develop seizures although her<br />

EEG was abnormal.<br />

Conclusions: We conclude that de novo mutations<br />

in FOXG1 are a cause <strong>of</strong> autosomal dominant<br />

mental retardation in patients with microcephaly<br />

and various brain malformations that could include<br />

frontal pachygyria and abnormal corpus callosum.


14-months old girl with microtia, short stature and<br />

developmental delay. A new syndrome?<br />

Irina Stefanova 1 , Almuth Caliebe 2 , Reiner Siebert 2 , Jürgen Sperner 3 , Holger<br />

Tönnies 2 , Gabriele Gillessen-Kaesbach 1<br />

1 Institut für Humangenetik, Universität zu Lübeck, Lübeck, <strong>German</strong>y<br />

2 Institut für Humangenetik, Christian-Albrechts-Universität zu Kiel, Kiel, <strong>German</strong>y<br />

3 Klinik für Kinder- und Jugendmedizin, Universität zu Lübeck, Lübeck, <strong>German</strong>y<br />

Background: Holoprosencephaly is a complex<br />

brain malformation resulting from incomplete<br />

cleavage <strong>of</strong> <strong>the</strong> prosencephalon, occuring between<br />

<strong>the</strong> 18th and 28th day <strong>of</strong> gestation. In most cases<br />

facial abnormalities are observed in association<br />

with holoprosencephaly, like hypotelorism,<br />

cyclopia, cleft lip or palate. Holoprosencephaly is<br />

a genetically heterogenous anomaly and is known<br />

to be part <strong>of</strong> different syndromes and chromosomal<br />

abnormalities. Several genes, such as SHH, ZIC2,<br />

SIX3, TGIF, PTCH, GLI2 and TDGF1 have been<br />

positively implicated in holoprosencephaly.<br />

Clinical report: Here we report a 14-months<br />

old girl with a semilobar holoprosenencephaly,<br />

microcephaly, bilateral microtia with absent auditory<br />

canals, short stature and developmental delay.<br />

Semilobar holoprosencephaly and an arachnoidal<br />

cyst were noted at 33 weeks <strong>of</strong> gestation by fetal<br />

MRI. After spontaneous delivery at 40 weeks <strong>of</strong><br />

gestation body measurements at birth were: length<br />

48 cm (-1,7 SD), weight 2490 g (-2,3 SD), head<br />

circumference 31,5 (-2,6 SD). Microtia with absent<br />

auditory canals was diagnosed after birth. Later on<br />

<strong>the</strong> child developed feeding difficulties and diabetes<br />

insipidus. At <strong>the</strong> age <strong>of</strong> 14 months she showed<br />

pronounced developmental delay. Both parents are<br />

healthy and fur<strong>the</strong>r family history is unremarkable.<br />

Methods: Cytogenetic analysis on blood<br />

lymphocytes, CGH to oligonucleotide arrays (105 K,<br />

Agilent) <strong>of</strong> peripheral blood DNA <strong>of</strong> <strong>the</strong> patient and<br />

both parents.<br />

Results: Cytogenetic analysis revealed a normal<br />

karyotype 46,XX in <strong>the</strong> patient. Array-CGH analysis<br />

detected an approximately 0,3Mb-sized duplication<br />

at 2q37.2 <strong>of</strong> paternal origin.<br />

Discussion and conclusions: We report a patient<br />

with semilobar holoprosencephaly, microcephaly,<br />

bilateral microtia with missing auditory canals.<br />

The combination <strong>of</strong> <strong>the</strong>se features has not been<br />

described in <strong>the</strong> literature until now. We suggest that<br />

our patient might have a new previously unknown<br />

syndrome.<br />

47


48<br />

Fre e co m m u n i c a t i o n s<br />

Chromosomal inversion combined with interstitial deletions<br />

on chromosomes 4q and 14q revealed by oligo-arrayCGH in<br />

a patient with autism<br />

Lott, A. 1 ; Kukuckova, M. 2 ; Čmelova, E. 3 ; Gencik, M. 1,2<br />

1 Zentrum für Medizinische Genetik, Osnabrück, <strong>German</strong>y<br />

2 Medgene, Bratislava, Slovakia<br />

3 Oddelenie klinickej genetiky, Bratislava, Slovakia<br />

Background: Autism is a behaviorally defined<br />

syndrome with a variety <strong>of</strong> causes, mostly on a<br />

complex genetic basis with many genes involved.<br />

Especially genes that are expressed during brain<br />

development are candidate genes for autism.<br />

Objective:In this case study we present an 8 year<br />

old male patient with psychomotoric retardation,<br />

autistic features and cytogenetically visible<br />

paracentric inversion <strong>of</strong> chromosome 14. To reveal<br />

cryptic chromosomal imbalances, high resolution<br />

arrayCGH was performed.<br />

Methods: Array CGH was performed using a<br />

44k 60mer oligonucleotide array (Agilent) with a<br />

<strong>the</strong>oretical resolution <strong>of</strong> about 100kb.<br />

Results: A deletion <strong>of</strong> 3,2 Mb was found at<br />

one inversion breakpoint at 14q12q13.1. The<br />

deleted region contains 5 known genes, one <strong>of</strong><br />

<strong>the</strong>m NPAS3. This gene codes for a neuronal<br />

transcription factor that is especially expressed in<br />

<strong>the</strong> developing human fetal brain and defects <strong>of</strong><br />

this gene are implicated in psychiatric disorders.<br />

Additionally, a deletion <strong>of</strong> 5,0 Mb was found at<br />

4q31.3q32.1. The deleted region comprises<br />

22 known genes, including AMPA2 and TDO2.<br />

The AMPA2 gene product is a neurotransmitter<br />

receptor and plays a key role in several<br />

aspects <strong>of</strong> developmental and adult synaptic<br />

plasticity. The TDO2 gene product is involved in<br />

serotonin production. Serotonin is pivotal during<br />

development and disturbed levels may contribute<br />

to structural brain abnormalities and autistic<br />

features.<br />

Conclusions: Cases <strong>of</strong> autism in which<br />

chromosomal inbalances occur are very important<br />

to highlight chromosome regions that are <strong>the</strong> sites<br />

<strong>of</strong> autism candidate genes. Oligo-arrayCGH is a<br />

powerful tool to search for such imbalances with<br />

high resolution. This case reveals <strong>the</strong> possible<br />

involvement <strong>of</strong> some genes in autism.


Refining <strong>the</strong> phenotype <strong>of</strong> α-1a Tubulin (TUBA1A) mutation in<br />

patients with classical lissencephaly<br />

Morris-Rosendahl DJ 1 *, Najm J2, Lachmeijer AMA 3 , Sztriha L 4 , Kuechler A 5 , Haug<br />

V 6 , Zeschnigk C 1 , Martin P 7 , Santos M8, Omran H 6 , Kraus U 7 , Van der Knaap MS 9 ,<br />

Schuierer G 10 , Kutsche K 2 , Uyanik G 11<br />

1 Inst. for Human Genetics and Anthropology, Albert-Ludwigs University <strong>of</strong> Freiburg, D<br />

2 Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, D<br />

3 Dept. <strong>of</strong> Clinical Genetics, VU University Medical Center, Amsterdam, NL<br />

4 Dept. <strong>of</strong> Paediatrics, University <strong>of</strong> Szeged, Szeged, Hungary<br />

5 Institute for Human Genetics, University <strong>of</strong> Essen, Essen, <strong>German</strong>y<br />

6 Centre for Paediatrics and Adolescent Medicine, University <strong>of</strong> Freiburg, Freiburg, D<br />

7 Epilepsy Center Kork, Kehl-Kork, <strong>German</strong>y<br />

8 Neuropediatrics Service, Hospital Maria Pia, Porto, Portugal<br />

9 Child Neurology, VU University Medical Center, Amsterdam, The Ne<strong>the</strong>rlands<br />

10 Institute <strong>of</strong> Neuroradiology, Bezirksklinikum Regensburg, Regensburg, <strong>German</strong>y<br />

11 Department <strong>of</strong> Neurology, University <strong>of</strong> Regensburg, Regensburg, <strong>German</strong>y<br />

Background: Classical or Type 1 lissencephaly<br />

is an autosomal dominantly inherited disorder<br />

<strong>of</strong> neuronal migration which results in agyria<br />

or pachygria, a thickened cortex and may be<br />

accompanied by various o<strong>the</strong>r structural brain malformations.<br />

Patients with classical lissencephaly<br />

have severe psychomotor retardation and epilepsy.<br />

Mutations in <strong>the</strong> α-1a Tubulin (TUBA1A) gene have<br />

recently been found to cause cortical malformations<br />

resemblant <strong>of</strong> classical lissencephaly, but with a<br />

specific combination <strong>of</strong> features. To date, TUBA1A<br />

mutations have been described in five patients and<br />

three foetuses.<br />

Objective: Our aim was to establish how common<br />

TUBA1A mutations are in patients with lissencephaly<br />

and to contribute to defining <strong>the</strong> phenotype<br />

associated with TUBA1A mutation.<br />

Methods: We performed mutation analysis in<br />

<strong>the</strong> TUBA1A gene in 46 patients with classical<br />

lissencephaly. In 44 <strong>of</strong> <strong>the</strong> patients, mutations in<br />

<strong>the</strong> LIS1 and/or DCX genes had previously been<br />

excluded; in two patients mutation analysis was only<br />

performed in TUBA1A, based on MRI findings.<br />

Results: We identified three new mutations and<br />

one recurrent mutation in five patients with variable<br />

patterns <strong>of</strong> lissencephaly on brain MRI. Four <strong>of</strong> <strong>the</strong><br />

five patients had congenital microcephaly, all had<br />

dysgenesis <strong>of</strong> <strong>the</strong> corpus callosum and cerebellar<br />

hypoplasia, and variable cortical malformations,<br />

including subtle subcortical band heterotopia, and<br />

absence or hypoplasia <strong>of</strong> <strong>the</strong> anterior limb <strong>of</strong> <strong>the</strong><br />

internal capsule.<br />

Conclusions: The new findings that mutations<br />

in TUBA1A also cause a disorder <strong>of</strong> neuronal<br />

migration, adds a fifth gene to <strong>the</strong> molecular genetic<br />

diagnosis for patients with classical lissencephaly.<br />

We estimate <strong>the</strong> frequency <strong>of</strong> mutation in<br />

TUBA1A in patients with classical lissencephaly<br />

to be approximately 4%, and although not as<br />

common as mutations in <strong>the</strong> LIS1 or DCX genes,<br />

mutation analysis in TUBA1A should be included<br />

in <strong>the</strong> molecular genetic diagnosis <strong>of</strong> classical<br />

lissencephaly, particularly in patients with <strong>the</strong><br />

combination <strong>of</strong> features highlighted in this paper.<br />

49


50<br />

Fre e co m m u n i c a t i o n s<br />

MAOA genotype: Impact on amygdala-prefrontal connectivity<br />

in major depression<br />

Udo Dannlowski 1,2,5 , Patricia Ohrmann 1 , Carsten Konrad 1,2 , Katharina Domschke 1 ,<br />

Jochen Bauer 1 , Harald Kugel 3 , Christa Hoh<strong>of</strong>f 1 , Sonja Schöning 1,2 , Anette<br />

Kersting 1 , Bernhard T. Baune 1,4 , Lena S. Mortensen 1 , Volker Arolt 1 , Pienie<br />

Zwitserlood 5 , Jürgen Deckert 1,6 , Walter Heindel 3 , Thomas Suslow 1 ,*<br />

1 Department <strong>of</strong> Psychiatry, University <strong>of</strong> Münster, <strong>German</strong>y<br />

2 IZKF-Research Group 4, IZKF Münster, University <strong>of</strong> Münster, <strong>German</strong>y<br />

3 Department <strong>of</strong> Clinical Radiology, University <strong>of</strong> Münster, <strong>German</strong>y<br />

4 Department <strong>of</strong> Psychiatry, James Cook University, Townsville, Australia<br />

5 Department <strong>of</strong> Psychology, University <strong>of</strong> Münster, <strong>German</strong>y<br />

6 Department <strong>of</strong> Psychiatry, University <strong>of</strong> Würzburg, <strong>German</strong>y<br />

Background: The amygdala plays a pivotal role in<br />

a prefrontal-limbic circuitry implicated in emotion<br />

processing and regulation. Major depression has<br />

been associated with a common functional variable<br />

number tandem repeat (VNTR) polymorphism in <strong>the</strong><br />

promoter region <strong>of</strong> <strong>the</strong> monoamine oxidase A gene<br />

(MAOA u-VNTR) as well as reduced amygdalaprefrontal<br />

functional connectivity, which could be a<br />

useful intermediate phenotype for depression.<br />

Objective: In <strong>the</strong> present study, we sought to<br />

investigate <strong>the</strong> impact <strong>of</strong> MAO-A genotype on<br />

<strong>the</strong> functional connectivity <strong>of</strong> amygdala-prefrontal<br />

emotion regulation circuitries in depressed<br />

patients.<br />

Methods: Functional connectivity <strong>of</strong> <strong>the</strong> amygdala<br />

with prefrontal areas involved in emotion<br />

regulation was investigated during a facial<br />

expression processing task in a sample <strong>of</strong> 34<br />

depressed inpatients and 31 healthy controls. All<br />

patients were genotyped for <strong>the</strong> MAOA u-VNTR<br />

polymorphism.<br />

Results: We observed strong coupling <strong>of</strong> <strong>the</strong><br />

amygdala and dorsal prefrontal areas comprising<br />

<strong>the</strong> dorsolateral prefrontal cortex (DLPFC), dorsal<br />

parts <strong>of</strong> <strong>the</strong> anterior cingulate cortex (dACC), and<br />

lateral orbit<strong>of</strong>rontal cortex in healthy subjects.<br />

Amygdala-prefrontal connectivity was significantly<br />

reduced in depressed patients compared with<br />

controls. Among both samples, carriers <strong>of</strong> <strong>the</strong><br />

higher active MAOA risk alleles (MAOA-H)<br />

showed weaker amygdala-prefrontal coupling.<br />

Fur<strong>the</strong>rmore, reduced coupling <strong>of</strong> this circuitry<br />

predicted more than 40% variance <strong>of</strong> clinical<br />

variables characterizing a longer and severer<br />

course <strong>of</strong> disease.<br />

Conclusions: We conclude that genetic variation<br />

in <strong>the</strong> MAOA gene may affect <strong>the</strong> course <strong>of</strong> major<br />

depression by disrupting cortico-limbic connectivity,<br />

a potential neural substrate for emotion regulation.


Fine-mapping in multiplex families with Attention-Deficit<br />

Hyperactivity Disorder<br />

Michelle K. Lin 1 , Haukur Palmason 1 , Christine Freitag 4 , Christiane Seitz 4 , Tobias<br />

J. Renner 2 , Marcel Romanos 2 , Susanne Walitza 2 , Christian Jacob 3 , Klaus P.<br />

Lesch 3 , Jobst Meyer 1<br />

1 Department <strong>of</strong> Neurobehavioral Genetics, University <strong>of</strong> Trier, <strong>German</strong>y<br />

2 Department <strong>of</strong> Child and Adolescent Psychiatry and Psycho<strong>the</strong>rapy, University <strong>of</strong><br />

Wuerzburg, <strong>German</strong>y<br />

3 Department <strong>of</strong> Psychiatry and Psycho<strong>the</strong>rapy, University <strong>of</strong> Wuerzburg, <strong>German</strong>y<br />

4 Institution:Department <strong>of</strong> Child and Adolescent Psychiatry, Saarland University<br />

Hospital, Homburg, <strong>German</strong>y<br />

Attention-deficit hyperactivity disorder (ADHD)<br />

is a neuropsychiatric disorder characterized<br />

by symptoms <strong>of</strong> inattention, hyperactivity and<br />

increased impulsiveness. Despite many studies<br />

over <strong>the</strong> last decades, <strong>the</strong> etiology <strong>of</strong> ADHD<br />

remains unknown.<br />

Eight large ADHD-affected families were recruited<br />

for this study. Using genetic data derived from<br />

related individuals within each <strong>of</strong> <strong>the</strong> large families,<br />

an attempt is made to identify predisposing<br />

chromosomal regions in <strong>the</strong>se families. Finemapping<br />

<strong>of</strong> <strong>the</strong> significant loci reported in a recent<br />

paper (Romanos et al, 2008) <strong>of</strong> a genome-wide<br />

linkage study <strong>of</strong> <strong>the</strong> eight large familes was carried<br />

out. These families were recruited in a multicenter<br />

collaboration study conducted by three clinical<br />

units <strong>of</strong> child and adult psychiatry (University <strong>of</strong><br />

Trier, Homburg/Saar, and Wuerzburg). These<br />

families comprised <strong>of</strong> 191 individuals, <strong>of</strong> which 95<br />

were affected with ADHD.<br />

From <strong>the</strong> preliminary results, in one <strong>of</strong> <strong>the</strong> large<br />

families, a 20 cM interval on chromosome 18<br />

was narrowed down to 18q11-18q2. All affected<br />

individuals in this family share <strong>the</strong> same haplotype.<br />

ADHD is believed to be a complex, polygenic<br />

disease in which many genes <strong>of</strong> small effect<br />

contribute to disease susceptibility. However,<br />

<strong>the</strong>re may be monogeneous Mendelian<br />

inheritance in large families. Currently, interesting<br />

candidate genes in <strong>the</strong> narrowed down region<br />

<strong>of</strong> chromosome 18 are being selected for fur<strong>the</strong>r<br />

functional studies.<br />

51


52<br />

Fre e co m m u n i c a t i o n s<br />

Psychiatric symptoms in genetic forms <strong>of</strong> Parkinson disease<br />

M. Kasten 1 , L. Kertelge 1,2 , C. Klein 2<br />

1 Dept. <strong>of</strong> Psychiatry and Psycho<strong>the</strong>rapy, University <strong>of</strong> Lübeck, <strong>German</strong>y<br />

2 Dept. <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, <strong>German</strong>y<br />

Background: Psychiatric symptoms are recognized<br />

as important features in monogenic forms <strong>of</strong><br />

parkinsonism.<br />

Objective: To review current knowledge on non<br />

motor symptoms, particularly psychiatric features in<br />

monogenic forms <strong>of</strong> parkinsonism in <strong>the</strong> literature.<br />

Methods: A Medline search was performed using<br />

<strong>the</strong> search terms “parkinson” and <strong>the</strong> name <strong>of</strong> known<br />

PD genes including all publications from January<br />

1966 to January 2008. This resulted in 1,855<br />

citations, 305 <strong>of</strong> those included genetic information<br />

on PD patients. While avoiding double counts 119<br />

articles containing any information on any type <strong>of</strong><br />

non motor symptom (990 cases) could be included.<br />

We focused on presence <strong>of</strong> depression, anxiety,<br />

hallucinations, and dementia for each <strong>of</strong> <strong>the</strong> genes<br />

with available information.<br />

Results: The number <strong>of</strong> cases with available<br />

information was variable, ranging from 12 cases for<br />

DYT1 to 314 cases for LRRK2. Likewise methods<br />

differed widely across studies. Fur<strong>the</strong>rmore,<br />

cases differed regarding ages at onset, disease<br />

durations, ethnicities, treatments, and comorbidities.<br />

Overall, hallucinations and dementia were most<br />

frequently commented on, followed by depression<br />

and anxiety. Detailed information on psychiatric<br />

symptom frequencies linked to SNCA, Parkin,<br />

PINK1, and LRRK2 mutations include hallucinations:<br />

SNCA: 10/43(23%); Parkin: 7/206(3%); PINK1:<br />

9/60(15%); LRRK2: 37/216(17%); dementia:<br />

SNCA: 11/43(26%); Parkin: 3/62(5%); PINK1:<br />

6/54(11%); LRRK2: 35/314(11%); depression:<br />

SNCA: 16/43(37%); Parkin: 22/207(8%); PINK1:<br />

25/64(30%); LRRK2: 58/204(28%); anxiety: SNCA:<br />

3/24(13%); Parkin: 14/192(7%); PINK1: 15/41(37%);<br />

LRRK2: 28/140(20%).<br />

Conclusions: There is currently only very limited<br />

data on psychiatric features in genetic forms <strong>of</strong><br />

parkinsonism. Overall, <strong>the</strong> frequency <strong>of</strong> psychiatric<br />

features in patients with genetic parkinsonism does<br />

not appear to be higher and may even be lower<br />

than in idiopathic PD. Possible exceptions include<br />

<strong>the</strong> occurrence <strong>of</strong> specific psychiatric disorders<br />

in PINK1-linked disease or <strong>of</strong> dementia in SNCAassociated<br />

parkinsonism. Psychiatric symptoms<br />

have a high impact on <strong>the</strong> patients’ quality <strong>of</strong> life<br />

and caregiver burden and should be considered as<br />

important and <strong>of</strong>ten treatable concomitant features<br />

<strong>of</strong> genetic parkinsonism.


Pos t e r ab s t r a c t P1<br />

Enhanced stress reactivity in NOS2 mutant mice: findings in<br />

support <strong>of</strong> astrocytic nitrosative modulation <strong>of</strong> behavior.<br />

Abu-Ghanem Yasmin 1,2 , Cohen Hagit 3 , Buskila Yossi 1,2 , Grauer Ettie 4 , Amitai<br />

Yael 1,2<br />

1 Dept <strong>of</strong> Physiology<br />

2 Zlotowski Center for Neuroscience<br />

3 Mental Health Center, Ben-Gurion University Beer-Sheva<br />

4 Israel Institute for Biological Research, Ness-Ziona<br />

Alterations <strong>of</strong> nitric oxide (NO) metabolism in <strong>the</strong><br />

brain have been associated with modifications <strong>of</strong><br />

stress-related behavior in animal models. It has<br />

been generally assumed that <strong>the</strong>se behavioral<br />

changes are due to <strong>the</strong> neuronal nitrosative<br />

activity. On <strong>the</strong> o<strong>the</strong>r hand, glial NO production<br />

has been demonstrated mainly as a slow reaction<br />

to brain insults through <strong>the</strong> activity <strong>of</strong> an inducible<br />

NOS is<strong>of</strong>orm (NOS2). Recently we uncovered<br />

increased NOS activity in astrocytes <strong>of</strong> mice with a<br />

NOS2 mutation. Interestingly, <strong>the</strong>se mice revealed<br />

a behavioral phenotype suggestive <strong>of</strong> increased<br />

susceptibility to stress.<br />

In <strong>the</strong> present study we investigated <strong>the</strong> responses<br />

<strong>of</strong> <strong>the</strong>se mutants to stress by exposing <strong>the</strong>m<br />

to predator scent. Seven days later, mutant<br />

mice exhibited significantly higher anxiety like<br />

behavior in <strong>the</strong> elevated-plus maze, increased<br />

acoustic startle responses, and higher plasma<br />

corticosterone levels compared to <strong>the</strong>ir controls.<br />

Systemic administration <strong>of</strong> a NOS inhibitor prior to<br />

<strong>the</strong> stress exposure reversed <strong>the</strong>se stress-related<br />

effects without affecting controls’ behavior. These<br />

findings are in agreement with previous studies<br />

showing an association between increased NO<br />

levels and enhanced anxiety-like responses. In<br />

addition, mutant mice performed better in <strong>the</strong><br />

Morris water maze prior to stress exposure, but <strong>the</strong><br />

two animal groups performed alike in an objectrecognition<br />

test.<br />

Taken toge<strong>the</strong>r, our results suggest <strong>the</strong><br />

involvement <strong>of</strong> astrocytic-derived NO in modulating<br />

behavior.<br />

53


54<br />

Pos t e r ab s t r a c t P2<br />

Mapping <strong>of</strong> a novel syndrome involving spastic paraplegia,<br />

ataxia, and mental retardation<br />

Christian Beetz 1 , Nihal O. Dündar 2 , Rebecca Schüle 3 , Senay Haspolat 2 , Peter<br />

Nürnberg 4 , Ozgur Duman 2 , Ludger Schöls 3 , Thomas Deufel 1 , Christian Hübner 1<br />

1 Institut für Klinische Chemie und Laboratoriumsdiagnostik, Universitätsklinikum, Jena,<br />

<strong>German</strong>y<br />

2 Department <strong>of</strong> Child Neurology, Faculty <strong>of</strong> Medicine, Akdeniz University Antalya, Turkey<br />

3 Hertie Institut für Klinische Hirnforschung, Tübingen, <strong>German</strong>y<br />

4 Cologne Center <strong>of</strong> Genomics, Universität Köln, <strong>German</strong>y<br />

Background: Hereditary spastic paraplegias<br />

(HSP) and cerebellar ataxias (CA) are<br />

neurodegenerative conditions with highly<br />

heterogeneous genetic backgrounds. Spastic<br />

ataxias (SA) are increasingly recognized as a<br />

separate group <strong>of</strong> disorders showing phenotypic<br />

features <strong>of</strong> both HSP and CA.<br />

Objective: To characterise a large Turkish<br />

pedigree for which <strong>the</strong> index patient had been<br />

diagnosed with spastic paraplegia.<br />

Methods: Available family members were clinically<br />

examined. The disease status <strong>of</strong> unavailable<br />

or deceased family members was inferred from<br />

reports provided by relatives. Genomic DNA <strong>of</strong> six<br />

individuals was analysed on 10K SNP arrays.<br />

Results: The index case, whose parents are<br />

first cousins, shows pure spastic paraplegia that<br />

started at age 5. His paternal aunt and uncle<br />

have also suffered from spastic paraplegia since<br />

<strong>the</strong>ir first decade <strong>of</strong> life. Later on, <strong>the</strong>y developed<br />

ataxia, dysarthria, and mental retardation. Their<br />

parents, i.e. <strong>the</strong> index patient’s grandparents,<br />

are first cousins too. Four additional individuals<br />

representing <strong>of</strong>fspring from yet ano<strong>the</strong>r but more<br />

distant consanguineous marriage are reported to<br />

have had similar symptoms; <strong>the</strong>y died at relatively<br />

young age. SNP genotyping using DNA from <strong>the</strong><br />

three patients available and from three unaffected<br />

relatives revealed linkage to a single region on<br />

chromosome 1p. This region, being homozygous<br />

in <strong>the</strong> patients, spans 54MB and contains<br />

approximately 500 genes. It does not overlap with<br />

any known locus for recessive HSP, CA, and SA.<br />

Conclusions: Our data suggest <strong>the</strong> identification<br />

<strong>of</strong> a novel form <strong>of</strong> recessive SA that is complicated<br />

by mental retardation and, possibly, short lifespan.


Pos t e r ab s t r a c t P3<br />

Heterozygous ATP13A2 mutations in Parkinson´s disease<br />

Butz, E. 1 , Djarmati, A. 1 , Hagenah, J. 1 , Winkler, S. 1 , Lohmann, K. 1 , Berg, D. 3 , Kostić,<br />

V.S. 2 , Volkmann, J. 4 , Gasser, T. 3 , Klein, C. 1<br />

1 Depart. <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, Lübeck, <strong>German</strong>y;<br />

2 Institute <strong>of</strong> Neurology CCS, Belgrade, Serbia;<br />

3 Department <strong>of</strong> Neurodegenerative Diseases, Hertie-Institut for Clinical Brain<br />

Research, University <strong>of</strong> Tübingen, <strong>German</strong>y;<br />

4 Department <strong>of</strong> Neurology, Christian-Albrechts University, Kiel, <strong>German</strong>y<br />

Background: Juvenile parkinsonism and<br />

young-onset Parkinson´s disease (YOPD) are<br />

neurodegenerative disorders with an early disease<br />

onset (< 21 years and 21-40 years, respectively). To<br />

date, four genes linked to <strong>the</strong> recessively inherited<br />

form <strong>of</strong> parkinsonism have been identified. Since<br />

<strong>the</strong> year 2006, when <strong>the</strong> ATP13A2 gene was first<br />

associated with parkinsonism, seven different<br />

homozygous, compound and single heterozygous<br />

mutations have been reported in patients with<br />

an autosomal recessive form <strong>of</strong> early-onset<br />

parkinsonism with dementia (Kufor-Rakeb syndrome)<br />

and parkinsonism <strong>of</strong> juvenile or young onset.<br />

Objective: To fur<strong>the</strong>r investigate <strong>the</strong> role <strong>of</strong> <strong>the</strong><br />

ATP13A2 gene in <strong>the</strong> etiology <strong>of</strong> parkinsonism in a<br />

large cohort <strong>of</strong> patients with juvenile parkinsonism<br />

and YOPD.<br />

Methods: We performed a mutational screen <strong>of</strong><br />

all 29 exons <strong>of</strong> ATP13A2 in 112 patients and 55<br />

neurologically healthy individuals. Our patients were<br />

mainly <strong>of</strong> European ethnic origin and were affected<br />

with juvenile parkinsonism (n=16; 66.7% men; mean<br />

age <strong>of</strong> onset±SD: 15.1±4.5 years; range: 8-20 years)<br />

and YOPD (n=96; 55.4% men; mean age <strong>of</strong> onset ±<br />

SD: 31.8±3.9 years; range: 21-40 years). A subset<br />

<strong>of</strong> patients (n=22; 19.6%) had mutations in o<strong>the</strong>r PD<br />

genes.<br />

Results: We identified four different novel single<br />

heterozygous mutations in 3 patients with YOPD<br />

and one with juvenile parkinsonism (3.6%). None <strong>of</strong><br />

<strong>the</strong>se were found in healthy, neurologically examined<br />

control individuals. All <strong>of</strong> <strong>the</strong>se mutations affected<br />

amino acids highly conserved among mammalian<br />

species. Interestingly, <strong>the</strong> carrier <strong>of</strong> one <strong>of</strong> <strong>the</strong>se<br />

changes also harbored a heterozygous deletion and<br />

duplication in <strong>the</strong> Parkin gene, thus representing<br />

a possible case <strong>of</strong> digenic parkinsonism. A fur<strong>the</strong>r<br />

mutation was present in one YOPD patient and in<br />

one control individual.<br />

Conclusion: Although this work represents <strong>the</strong><br />

most comprehensive mutational analysis for<br />

mutations in ATP13A2 published to date, no<br />

definitive conclusions can be drawn on <strong>the</strong> potential<br />

significance <strong>of</strong> this gene in <strong>the</strong> etiology <strong>of</strong> earlyonset<br />

parkinsonism. The presence <strong>of</strong> single,<br />

possibly pathogenic missense mutations might<br />

suggest that even single heterozygous mutations<br />

in ATP13A2 may have a role as a susceptibility<br />

factor for parkinsonism.<br />

55


56<br />

Pos t e r ab s t r a c t P4<br />

Identification <strong>of</strong> SOD1 Ala4Val mutation in a Mexican family<br />

with lateral amyotrophic sclerosis<br />

Castañeda-Cisneros G 1,2 , García-Cruz D 3 , Moran-Moguel MC 1 , Rosales-Gómez<br />

R1,5, Gutiérrez-Rubio SA 1 , Dávalos-Rodríguez IP 4 , Nuñez-Revélez N 1,5 , Sánchez-<br />

Corona J 1 .<br />

1 División de Medicina Molecular, Centro de Investigación Biomédica de Occidente<br />

(CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara, México.<br />

2 Departamento de Neurocirugía, Hospital de Especialidades, IMSS, Guadalajara,<br />

México.<br />

3 Instituto de Genética Humana, Universidad de Guadalajara, Guadalajara, México.<br />

4 División de Genética, CIBO, IMSS, Guadalajara, México.<br />

5 Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud<br />

(CUCS), Universidad de Guadalajara, Guadalajara, México.<br />

Background: The amyotrophic lateral sclerosis<br />

(ALS) is included within <strong>the</strong> group <strong>of</strong> neurogenetic<br />

and neurodegenerative diseases. It is a condition<br />

that involves upper and lower motor neurons,<br />

typically presents asymmetrical focal weakness<br />

<strong>of</strong> <strong>the</strong> limbs, and/or findings at bulbar level like<br />

dysarthria/dysphagia, and lacking <strong>of</strong> sensory<br />

disturbances. ALS familial cases correspond to <strong>the</strong><br />

10 %, and 20 % <strong>of</strong> <strong>the</strong>m are related to mutations<br />

in <strong>the</strong> SOD1 gene. The mutation Ala4Val covers<br />

50 % <strong>of</strong> affected individuals in familial ALS and is<br />

characterized by rapid progression and severe<br />

clinical signs.<br />

Objective: To describe a Mexican family<br />

that corresponds to <strong>the</strong> clinical diagnosis <strong>of</strong><br />

amyotrophic lateral sclerosis according to <strong>the</strong><br />

established clinical criteria and <strong>the</strong>ir correlation<br />

with SOD1 Ala4Val mutation.<br />

Methods: Clinical, electromyography and<br />

neuroimaging studies for diagnosis <strong>of</strong> ALS; and<br />

polymerase chain reaction (PCR) with HaeIII<br />

digestion to identification <strong>of</strong> SOD1 Ala4Val mutation.<br />

Results: The pattern <strong>of</strong> inheritance is autosomal<br />

dominant, with variable expressivity and a probable<br />

phenomenon <strong>of</strong> anticipation. The number <strong>of</strong><br />

affected individuals in <strong>the</strong> present family is eight<br />

in three generations and average age <strong>of</strong> onset<br />

35.7 years, with a male/female ratio <strong>of</strong> 0.6/1. The<br />

molecular studies showed <strong>the</strong> presence <strong>of</strong> <strong>the</strong><br />

mutation SOD1 Ala4Val in two affected individuals<br />

and six unaffected individuals (generation IV: 43-50<br />

years; generation V: 6 -20 years), corresponding to<br />

100 % and 43.75 % respectively.<br />

Conclusions: In Mexico, <strong>the</strong>re are not familial<br />

reports with ALS related to SOD1 Ala4Val mutation,<br />

<strong>the</strong>refore is unknown if this is <strong>the</strong> most frequent<br />

one and presents severe symptoms in individuals<br />

affected by this disease.


Pos t e r ab s t r a c t P5<br />

Spinocerebellar Ataxia Type 11: Novel variations in <strong>the</strong><br />

TTBK2 gene<br />

Ulf Edener 1 , Veronica Bernard 1 , Katrin Bürk 2 , Gabriele Gillessen-Kaesbach 1 ,<br />

Christine Zühlke 1<br />

1 Institut für Humangenetik, Universität Lübeck<br />

2 Klinik für Neurologie, Universität Marburg<br />

Background: The autosomal dominant<br />

spinocerebellar ataxias (SCAs) are a heterogeneous<br />

group <strong>of</strong> neurodegenerative disorders characterised<br />

by poor coordination, impairment <strong>of</strong> speech<br />

and swallowing, abnormal eye movements and<br />

pyramidal signs. To date 30 SCA loci have been<br />

discovered and for at least 16 <strong>the</strong> respective gene<br />

or mutation has been determined. In 2007 Houlden<br />

and colleagues described mutations in <strong>the</strong> TTBK2<br />

gene to be associated with SCA11. This gene<br />

localised on chromosome 15q15.2 encodes tau<br />

tubulin kinase 2 which is highly expressed in <strong>the</strong><br />

cerebellum Purkinje cells and seems to play an<br />

important role in <strong>the</strong> tau cascade. They identified a<br />

one-base insertion (c.1328_1329insA) at codon 444<br />

in a British family and a deletion <strong>of</strong> a dinucleotide<br />

(c.1284_1285delGA) at codons 428 and 429 in<br />

a family <strong>of</strong> Pakistani ancestry. Both frameshift<br />

mutations occured in exon 13 <strong>of</strong> <strong>the</strong> 15-exontranscript<br />

and created premature stop sites at<br />

codons 450 and 449 respectively.<br />

Objective and Methods: To determine <strong>the</strong><br />

frequency and to search for fur<strong>the</strong>r mutations<br />

concerning TBK2 we sequenced <strong>the</strong> 18 translated<br />

exons and flanking intronic sequences <strong>of</strong> <strong>the</strong><br />

currently longest 19-exon-transcript for 48 unrelated<br />

patients with dominant ataxia.<br />

Results: We found six variations at DNA level.<br />

A 157-bp-frameshift-deletion in exon 11 was<br />

detected in 8,5 % <strong>of</strong> ataxia patients and in 12,5 %<br />

<strong>of</strong> 95 unrelated control individuals. Remarkably,<br />

<strong>the</strong> 19-exon-transcript being <strong>the</strong> only transcript<br />

harbouring exon 11 could not be detected in<br />

lymphoblasts <strong>of</strong> patients and control individuals by<br />

RT-PCR. Fur<strong>the</strong>rmore three novel DNA variations<br />

were found in <strong>the</strong> TTBK2 coding region: a missense<br />

exchange E1247G in one ataxia patient <strong>of</strong> Arabian<br />

ancestry and <strong>the</strong> two silent mutations S758S and<br />

E1269E in one and four patients respectively. Beside<br />

<strong>the</strong>se novel variations two known 1-bp-substitutions<br />

in <strong>the</strong> 5’-UTR at mRNA positions -37 and -67 were<br />

detected in more than 30 % <strong>of</strong> <strong>the</strong> ataxia patients.<br />

Conclusion: In conclusion SCA11 is not a frequent<br />

cause for dominantly inherited ataxias. The presence<br />

<strong>of</strong> <strong>the</strong> 157-bp-deletion in samples from patients and<br />

unaffected individuals argues against a disease<br />

causing potential. The E1247G missense exchange<br />

should be fur<strong>the</strong>r investigated to determine whe<strong>the</strong>r<br />

it is a mutation or a polymorphism.<br />

57


58<br />

Pos t e r ab s t r a c t P6<br />

Differential effect <strong>of</strong> PINK1 nonsense and missense<br />

mutations on mitochondrial function and morphology<br />

A. Grünewald, MS 1,2 , M. Gegg, PhD 3 , J-W. Taanman, PhD 3 , C. Klein, MD 1,2 , A.H.V.<br />

Schapira, MD 3<br />

1 2 Departments <strong>of</strong> Neurology and Human Genetics, Lübeck University, Lübeck,<br />

<strong>German</strong>y<br />

3 University Department <strong>of</strong> Clinical Neurosciences, Royal Free and University College<br />

Medical School, University College London, London, UK<br />

Background: Recessive mutations in PTEN-induced<br />

putative kinase 1 (PINK1) are responsible for a<br />

familial form <strong>of</strong> early-onset parkinsonism. PINK1<br />

localizes to <strong>the</strong> inner mitochondrial membrane.<br />

Objective: The aim <strong>of</strong> <strong>the</strong> present study was to<br />

investigate fibroblasts <strong>of</strong> patients with parkinsonism<br />

for functional and morphological effects <strong>of</strong> nonsense<br />

and missense mutations in PINK1.<br />

Methods: We examined 4 members <strong>of</strong> Family<br />

W with <strong>the</strong> homozygous p.Q456X nonsense<br />

mutation and one patient with <strong>the</strong> homozygous<br />

p.V170G missense mutation. Additionally, 3<br />

healthy members <strong>of</strong> Family W and 5 independent<br />

age-matched individuals were included.<br />

Fibroblasts were cultured under standard<br />

conditions and treated with paraquat (PQ).<br />

The activities <strong>of</strong> complexes I-IV (CI-IV) were<br />

measured spectrophotometrically in mitochondrial<br />

preparations. For determination <strong>of</strong> <strong>the</strong> cellular<br />

ATP levels [ATP] a luminometric kit was used.<br />

The mitochondrial membrane potential (mMP)<br />

was detected by means <strong>of</strong> 5,5’,6,6’-tetrachloro-<br />

1,1’,3,3’-tetraethylbenzimidazolocarbocyanine<br />

iodide. The cellular gluthatione concentration<br />

[GSH] was quantified by reverse-phase HPLC. The<br />

mitochondrial DNA level [mtDNA] was assessed by<br />

real-time PCR. The mitochondrial morphology was<br />

studied by electron microscopy.<br />

Results: In <strong>the</strong> nonsense samples no significant<br />

differences were detected when <strong>the</strong> activities <strong>of</strong><br />

CI-IV were compared with controls. However,<br />

relative to <strong>the</strong> mean control values, <strong>the</strong> missense<br />

mutant revealed a 112% increased CII+III activity<br />

and a 43% decreased CIV activity. The average<br />

[ATP] in <strong>the</strong> nonsense cases amounted to 75%<br />

<strong>of</strong> <strong>the</strong> control group (p


Pos t e r ab s t r a c t P7<br />

Blood expression pr<strong>of</strong>iles <strong>of</strong> symptomatic DYT1 dystonia<br />

patients differ from asymptomatic mutation carriers – a<br />

signature for penetrance in DYT1 dystonia<br />

K. Grundmann 1 , M. Walter 1 , E. Valente 2 , M. Gambarin 3 , M. Tinazzi 3 , C. Kamm 4 , M.<br />

Bonin 1 , C. Klein5, O. Riess 1<br />

1 Dept <strong>of</strong> Medical Genetics, University <strong>of</strong> Tübingen, <strong>German</strong>y<br />

2 <strong>Neurogenetics</strong> Unit, IRCCS CSS-Mendel, Rome, Italy,<br />

3 Unità operativa di Neurologia, Ospedale Civile Maggiore Borgo Trento Verona, Italy<br />

4 Department <strong>of</strong> Neurodegenerative Diseases and Hertie-Institute for Clinical Brain<br />

Research University <strong>of</strong> Tübingen, <strong>German</strong>y<br />

5 Clinical and Molecular <strong>Neurogenetics</strong>, Department <strong>of</strong> Neurology, University <strong>of</strong><br />

Lübeck, <strong>German</strong>y,<br />

DYT1 dystonia is a neurodevelopmental<br />

movement disorder which is inherited in an<br />

autosomal dominant pattern. Most cases <strong>of</strong> this<br />

early-onset form <strong>of</strong> primary dystonia are due to<br />

a GAG deletion in <strong>the</strong> torsinA gene. Although <strong>the</strong><br />

underlying genetic defect has been identified, <strong>the</strong><br />

determination <strong>of</strong> <strong>the</strong> GAG deletion in a subject is<br />

<strong>of</strong> limited use with regard to factual manifestation<br />

<strong>of</strong> <strong>the</strong> disease, as <strong>the</strong> penetrance <strong>of</strong> <strong>the</strong> mutation<br />

is reduced to ~30-40%. Thus it is <strong>of</strong> extreme<br />

importance to develop new diagnostic tools.<br />

In <strong>the</strong> present study, we tried to implement a<br />

novel genetic paradigm: Affymetrix oligonucleotide<br />

microarrays were used to analyze global gene<br />

expression in blood samples <strong>of</strong> 24 DYT1 patients,<br />

22 nonmanifesting mutation carriers and controls.<br />

We identified 269 mRNAs that showed a<br />

significantly altered expression in DYT1 patients<br />

compared to nonmanifesting mutation carriers<br />

(P


60<br />

Pos t e r ab s t r a c t P8<br />

Functional characterization <strong>of</strong> <strong>the</strong> murine Mlc1 promoter with<br />

respect to stress-related behavior and psychoses<br />

Darja Henseler, Thorsten Kranz, Jobst Meyer<br />

Department <strong>of</strong> Neurobehavioural Genetics, University <strong>of</strong> Trier<br />

Background: Mutations in <strong>the</strong> MLC1 cause a<br />

severe neurological disorder, megalencephalic<br />

leukoencephalopathy with subcortical cysts, that is<br />

clinically characterized by macrocephaly and <strong>the</strong><br />

deterioration in motor functions, ataxia, spasticity<br />

and mental decline.<br />

Fur<strong>the</strong>rmore, a L309M mutation was found to cosegregate<br />

with catatonic schizophrenia in a large<br />

family. Aside from genes, o<strong>the</strong>r environmental<br />

factors, like stress, have been suggested to<br />

play a role in <strong>the</strong> susceptibility to psychoses like<br />

schizophrenia.Like <strong>the</strong> human MLC1, <strong>the</strong> murine<br />

Mlc1 is mainly expressed in astrocytes. The<br />

genomic organization <strong>of</strong> <strong>the</strong> murine Mlc1 was<br />

described by Steinke and coworkers (2003) which<br />

revealed 12 exons encoding a membrane protein<br />

consisting <strong>of</strong> 382 amino acids.<br />

Objective: The aim <strong>of</strong> <strong>the</strong> current work is to study<br />

<strong>the</strong> effect <strong>of</strong> stress on Mlc1 expression in mice.<br />

Fur<strong>the</strong>rmore, <strong>the</strong> function <strong>of</strong> <strong>the</strong> MLC1 protein,<br />

which still remains unclear, will be investigated.<br />

Methods: We assayed <strong>the</strong> murine Mlc1 promoter<br />

region using reporter gene constructs. We focus<br />

on changes in <strong>the</strong> regulation <strong>of</strong> gene expression,<br />

especially with respect to glucocorticoids.<br />

Results: We found a predicted promoter 3.9<br />

kb upstream <strong>of</strong> <strong>the</strong> Mlc1 gene, followed by a<br />

transcribed region including a pseudogene <strong>of</strong><br />

Pomp. NFкB binding sites in <strong>the</strong> regulatory region<br />

suggest a potential effect <strong>of</strong> stress on Mlc1<br />

expression.<br />

Conclusions: According to <strong>the</strong> literature and our<br />

findings, <strong>the</strong> murine Mlc1 seems to be differentially<br />

regulated compared to <strong>the</strong> human orthologue. This<br />

implies <strong>the</strong> need for a knock-out mouse model for<br />

both schizophrenia and MLC1. Currently, we are<br />

creating Mlc1 knock-out mouse to elucidate <strong>the</strong><br />

role <strong>of</strong> Mlc1 in brain development and behaviour.


Pos t e r ab s t r a c t P9<br />

Late-onset PEO and proximal myopathy in a patient carrying<br />

mutations in <strong>the</strong> POLG2 gene<br />

Rita Horvath. 1,2,4 , Maggie Walter 1 , Birgit Czermin 2 , Joanna Stewart 4 , Gavin<br />

Hudson 4 , Stefanie Bulst1, Peter Schneiderat1, Angela Abicht 1,2 , Hanns<br />

Lochmüller 3 , Patrick F. Chinnery 4 , Thomas Klopstock 1<br />

1 Friedrich-Baur-Institute and 2 Medical Genetic Centre, Munich, <strong>German</strong>y<br />

3 Institute <strong>of</strong> Human Genetics and 4 Mitochondrial Research Group, University <strong>of</strong><br />

Newcastle upon Tyne, UK<br />

Aims: Polymerase gamma 1 (POLG1) mutations<br />

are a frequent cause <strong>of</strong> both autosomal dominant<br />

and recessive complex neurological phenotypes.<br />

In contrast, a pathogenic mutation in POLG2 was<br />

detected in one patient only world-wide.<br />

Here we describe a 62 year-old woman carrying<br />

two novel heterozygous sequence variants in <strong>the</strong><br />

POLG2 gene.<br />

Patient and Methods: The first symptoms started<br />

at age 30 years with bilateral ptosis, followed by<br />

exercise intolerance and proximal weakness in <strong>the</strong><br />

late forties. CK was occassionally elevated up to<br />

500 U/L. Her 22 year old daughter and one <strong>of</strong> her<br />

three bro<strong>the</strong>rs also show ptosis.<br />

Results: Histological examination <strong>of</strong> skeletal<br />

muscle and biochemical measurement <strong>of</strong> <strong>the</strong><br />

respiratory enzymes showed normal results. Long<br />

range PCR analysis detected multiple mtDNA<br />

deletions in muscle DNA <strong>of</strong> <strong>the</strong> patient. The<br />

molecular genetic analysis <strong>of</strong> POLG1, Twinkle and<br />

ANT1 did not reveal any abnormalities. Two novel<br />

heterozygous mutations were detected in POLG2,<br />

an intronic variant c.1192-16_1192-15hetinsT and<br />

a complex mutation predicted to insert 8 additional<br />

amino acids into <strong>the</strong> protein. The analysis <strong>of</strong> fur<strong>the</strong>r<br />

family members and cDNA studies are underway.<br />

Conclusions: POLG2 mutations are a rare<br />

cause <strong>of</strong> autosomal dominant PEO. The clinical<br />

presentation <strong>of</strong> our patient is in keeping with <strong>the</strong><br />

previously described clinical phenotype.<br />

61


62<br />

Pos t e r ab s t r a c t P10<br />

Analysis <strong>of</strong> <strong>the</strong> SCN1A gene for idiopathic epilepsy<br />

Kuhn, M. 1 , Gabriel, H. 1 , Jagiello, P. 1 and Gencik, M. 1,2<br />

1 Zentrum für Medizinische Genetik, Osnabrück, <strong>German</strong>y<br />

2 Medgene, Bratislava, Slovakia<br />

Background: Idiopathic epilepsies represent<br />

~40% <strong>of</strong> <strong>the</strong> epilepsy spectrum. So far, about<br />

~12 genes for ‘single gene’ epilepsies have been<br />

identified, mostly encoding ion channels joining <strong>the</strong><br />

epilepsy phenotype to <strong>the</strong> channelopathies. Ion<br />

channels are critical for neuronal excitability and,<br />

<strong>the</strong>refore for <strong>the</strong> delicate balances that maintain<br />

electric stability in <strong>the</strong> CNS.<br />

Objective: Here, we present <strong>the</strong> diagnostic <strong>of</strong> <strong>the</strong><br />

SCN1A gene in 35 children suffering from GEFS+<br />

type 2 (n=15) and SMEI (n=20). Both phenotypes<br />

belong to <strong>the</strong> spectrum <strong>of</strong> GEFS+. GEFS+2 is a<br />

relatively mild form <strong>of</strong> <strong>the</strong> GEFS+ spectrum and<br />

<strong>the</strong>rapeutic strategies work well, SMEI (Dravet<br />

syndrome) is <strong>the</strong> most severe phenotype with<br />

a poor outcome. In 33-100% (different studies)<br />

mutations in <strong>the</strong> SCNA1 gene in SMEI are de novo.<br />

Methods: We perform this test by <strong>the</strong> DGGE<br />

method in addition to direct sequencing <strong>of</strong> selected<br />

exons <strong>of</strong> <strong>the</strong> SCN1A gene including <strong>the</strong> splicing<br />

sites. Additionally, we have screened for deletions<br />

or duplications <strong>of</strong> single exons by MLPA technique.<br />

Results: We detected missense-mutations in<br />

two <strong>of</strong> our GEFS+2 patients. In 6/20 SMEI cases<br />

(all de novo) including small deletions/insertions,<br />

truncating and missense mutations, mostly located<br />

in <strong>the</strong> pore and voltage sensitive region <strong>of</strong> <strong>the</strong><br />

SCN1A protein. In one patient a novel homozygous<br />

sequence variation was found. Both parents were<br />

heterozygous carriers <strong>of</strong> this variation. It is unlikely<br />

that this variant is a causative mutation. Although<br />

unknown for SCN1A mutations a recessive trait<br />

can not be excluded.<br />

Conclusion: The molecular genetic diagnostic <strong>of</strong><br />

epilepsies particularly in children is challenging,<br />

but will facilitate <strong>the</strong> early diagnosis, prognosis and<br />

also <strong>the</strong>rapeutic strategy.


Pos t e r ab s t r a c t P11<br />

Huntingtin-associated protein-1 is a modifier <strong>of</strong> <strong>the</strong><br />

age-at-onset <strong>of</strong> Huntington’s disease<br />

Silke Metzger 1 , Juan Rong 2 , Huu-Phuc Nguyen 1 , Jürgen Tomiuk 1 , Anne Söhn 1 ,<br />

Peter Propping 3 , Yun Freudenberg-Hua 3 , Jan Freudenberg 4 , Liang Tong 5 , Shi-Hua<br />

Li 2 , Xiao-Jiang Li 2 , Olaf Riess 1<br />

1 Medical Genetics, Tübingen, <strong>German</strong>y<br />

2 Human Genetics, Atlanta, US<br />

3 Human Genetics, Bonn, <strong>German</strong>y<br />

4 Neurology, UCSF, US<br />

5 Biological Sciences, Columbia University, US<br />

Background: Huntington’s disease (HD) is caused<br />

by an unstable CAG repeat expansion in <strong>the</strong> HD<br />

gene that is inversely correlated with <strong>the</strong> age-atonset<br />

<strong>of</strong> <strong>the</strong> disease. Since <strong>the</strong> size <strong>of</strong> <strong>the</strong> CAG<br />

repeat only explains about 42-73% <strong>of</strong> <strong>the</strong> variance<br />

<strong>of</strong> <strong>the</strong> age-at-onset, o<strong>the</strong>r factors might modify <strong>the</strong><br />

course <strong>of</strong> <strong>the</strong> disease.<br />

Objective: We aimed to identify and characterize<br />

a genetic modifier in Huntingtin-associated<br />

protein-1 (HAP1), which participates in intracellular<br />

trafficking, colocalizes and interacts with huntingtin,<br />

thus representing a good candidate for a genetic<br />

modifier.<br />

Methods: We analyzed HAP1 polymorphisms<br />

by association studies in 980 European HD<br />

patients. As one <strong>of</strong> <strong>the</strong>se turned out to modify <strong>the</strong><br />

disease, <strong>the</strong> functional analyzes <strong>of</strong> HAP1 and this<br />

polymorphism concentrated on <strong>the</strong> examination<br />

<strong>of</strong> protein-protein interactions by yeast two-hybrid<br />

assays and coimmunoprecipitation, aggregate<br />

formation, cellular localization and apoptotic<br />

properties in cell culture.<br />

Results: We identified one HAP1 polymorphism<br />

as a genetic modifier for HD age-at-onset. Patients<br />

homozygous for this polymorphism develop <strong>the</strong><br />

first symptoms about 8 years later than patients<br />

with o<strong>the</strong>r genotypes. Functional analyses<br />

showed that polymorphic HAP1 binds more<br />

mutant huntingtin and reduces its degradation.<br />

Additionally, it increases <strong>the</strong> number <strong>of</strong> large<br />

aggregates, prevents <strong>the</strong> localization <strong>of</strong> huntingtin<br />

in <strong>the</strong> nucleus and protects against huntingtinmediated<br />

toxicity in transfected cells.<br />

Conclusion: We identified <strong>the</strong> first genetic modifier<br />

for HD that acts protective and has a functional<br />

importance to huntingtin-mediated toxicity and thus<br />

to HD pathogenesis.<br />

63


64<br />

Pos t e r ab s t r a c t P12<br />

PolyQ-expanded TBP leads to aggregation and neuronal<br />

death in primary neurons and transgenic mice<br />

Nguyen HP 1 , Then F 2 , Osmand A 3 , Wolburg H 4 , Golub Y 1 , Ott T 1 , Bauer C 1 ,<br />

Teismann P 5 , Hennek T 1 , Krainc D 2 , Riess O 1 , Bauer P 1<br />

1 Department <strong>of</strong> Medical Genetics, Tübingen, <strong>German</strong>y<br />

2 Department <strong>of</strong> Neurology, Charlestown, US<br />

3 Department <strong>of</strong> Medicine, Knoxville, US<br />

4 Department <strong>of</strong> Pathology, Tübingen, <strong>German</strong>y<br />

5 Institute <strong>of</strong> Medical Sciences, Aberdeen, UK<br />

Background and objective: SCA17 is a<br />

progressive neurodegenerative disease leading<br />

to cerebellar ataxia and dementia. Genetically, a<br />

CAG/CAA expansion in <strong>the</strong> TATA binding protein<br />

(TBP) is found in SCA17 patients. To fur<strong>the</strong>r study<br />

<strong>the</strong> pathogenesis <strong>of</strong> this disorder, we aimed to<br />

develop a transgenic mouse model for SCA17.<br />

Methods and results: Overexpression <strong>of</strong><br />

polyQ-expanded TBP in primary neurons lead to<br />

aggregate formation and neuronal death. This<br />

prompted us to generate transgenic mice which<br />

express 64 CAG/CAA repeats containing human<br />

TBP gene under <strong>the</strong> control <strong>of</strong> <strong>the</strong> truncated<br />

human prion protein promoter. Transgenic<br />

protein expression throughout different brain<br />

regions was clearly demonstrable. Onset <strong>of</strong><br />

motor dysfunction started by 7 months and<br />

progressed with age. By electron microscopy and<br />

immunohistochemical methods we were able<br />

to detect neurodegeneration and aggregation<br />

selectively in <strong>the</strong> cerebellum, but not in striatum<br />

and cortex. This could be confirmed using filter trap<br />

assay with mouse brain lysates. SDS-insoluble<br />

aggregates in <strong>the</strong> membrane were seen in lysates<br />

from transgenic mice, whereas wild-type brain<br />

lysates did not reveal significant retention <strong>of</strong><br />

aggregated material in cellulose acetate.<br />

Conclusion: We present detailed morphological<br />

and phenotypical data for this rodent model <strong>of</strong><br />

SCA17, which will be a valuable tool to conduct<br />

preclinical <strong>the</strong>rapeutic studies and to fur<strong>the</strong>r study<br />

<strong>the</strong> pathogenesis <strong>of</strong> this so far incurable disease.


Pos t e r ab s t r a c t P13<br />

Late onset hereditary sensory neuropathie type 1 (HSN1)<br />

caused by a novel p.C133R missense mutation in SPTLC1<br />

Bernd Rautenstrauss 1,2 , Birgit Neitzel 1 , Christoph Muench 3 , Judith Haas 3 , Elke<br />

Holinski-Feder 1,4 , Angela Abicht 1,2<br />

1 Medical Genetics Center Munich, <strong>German</strong>y<br />

2 Friedrich-Baur-Institute, Ludwig-Maximilian-University Munich, <strong>German</strong>y<br />

3 Jewish Hospital Berlin, <strong>German</strong>y<br />

4 Ludwig-Maximilian-University Munich, <strong>German</strong>y<br />

The hereditary sensory and autonomic<br />

neuropathies (HSAN), which are also referred to<br />

as hereditary sensory neuropathies (HSN) in <strong>the</strong><br />

absence <strong>of</strong> significant autonomic features, are a<br />

genetically and clinically heterogeneous group <strong>of</strong><br />

disorders associated with sensory dysfunction.<br />

HSN1 is a dominantly inherited sensorimotor<br />

axonal neuropathy usually with onset in <strong>the</strong> first<br />

or second decades <strong>of</strong> life. We investigated a<br />

female patient with age-<strong>of</strong>-onset at 50 years.<br />

Her mo<strong>the</strong>r was also affected but died. Her<br />

bro<strong>the</strong>r was diagnosed as chronic inflammatory<br />

demyelinating neuropathy (CIDP) but - typical<br />

for an inherited peripheral neuropathy - steroid<br />

treatment failed. Neurological examination <strong>of</strong> <strong>the</strong><br />

index patient revealed a normal nerve conduction<br />

velocity (NCV) <strong>of</strong> N. fibularis at 46 m/s. No motor<br />

impairment was found in EMG and NCV studies,<br />

also no atrophies were present. Major symptom<br />

is a distal, symmetric hypes<strong>the</strong>sia. Sequence<br />

analysis <strong>of</strong> all coding exons <strong>of</strong> <strong>the</strong> gene for <strong>the</strong><br />

Serine Palmitoyltransferase, Long-Chain subunit<br />

1 (SPTLC1) revealed a heterozygous variation<br />

c.397T>C resulting in a missense mutation<br />

p.C133R. Serine palmitoyltransferase (SPT;<br />

EC 2.3.1.50) is <strong>the</strong> key enzyme in sphingolipid<br />

biosyn<strong>the</strong>sis. It catalyzes <strong>the</strong> pyridoxal-5-primephosphate-dependent<br />

condensation <strong>of</strong> L-serine<br />

and palmitoyl-CoA to 3-oxosphinganine. The<br />

p.C133Y and p.C133W mutations were earlier<br />

described as cause <strong>of</strong> severe HSN1 accompanied<br />

by deafness and ulcerations. Bejaoui et al. (2002)<br />

found that <strong>the</strong> p.C133Y and p.C133W mutations<br />

do not alter <strong>the</strong> steady state levels <strong>of</strong> <strong>the</strong> SPTLC1<br />

or <strong>the</strong> SPTLC2 subunit but result in reduced<br />

SPT activity and sphingolipid syn<strong>the</strong>sis. These<br />

functional results indicated that both <strong>of</strong> <strong>the</strong>se<br />

mutations have a dominant-negative effect on <strong>the</strong><br />

SPT enzyme. The here reported p.C133R mutation<br />

may follow <strong>the</strong> same loss-<strong>of</strong>-function mechanism,<br />

but <strong>the</strong> resulting phenotype is relatively mild<br />

indicating a reduced but not completely missing<br />

SPT activity.<br />

65


66<br />

Pos t e r ab s t r a c t P14<br />

Clinical and genetic investigation <strong>of</strong> a multigenerational<br />

Parkinson disease family<br />

K. Rondorf 1 , W. Külper 1 , N. Brüggemann 1 , P. Vieregge 2 , J. Hagenah 1 , C. Klein 1 , K.<br />

Lohmann 1<br />

1 Department <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, Lübeck, <strong>German</strong>y<br />

2 Department <strong>of</strong> Neurology, Klinikum Lippe-Lemgo, Lemgo, <strong>German</strong>y<br />

Background: Parkinson disease (PD) is a<br />

common neurodegenerative disorder. In <strong>the</strong> past<br />

10 years at least six genes have been identified<br />

to be associated with PD. We performed a clinical<br />

and genetic investigation <strong>of</strong> a multigenerational PD<br />

family for whom known genetic factors had been<br />

excluded through linkage or mutational analysis.<br />

Objective: To clinically and genetically<br />

characterize a family with PD.<br />

Patients/Methods: Twenty-six family members<br />

underwent a detailed neurological examination<br />

by a movement disorder specialist. A consensus<br />

diagnosis was obtained based on <strong>the</strong> findings <strong>of</strong><br />

<strong>the</strong> onsite examiner and <strong>the</strong> results <strong>of</strong> a videotape<br />

review by two additional experts in movement<br />

disorders. A genome-wide linkage analysis was<br />

performed on 13 family members with a set <strong>of</strong><br />

450 microsatellite markers. LOD scores were<br />

calculated using <strong>the</strong> ALLEGRO s<strong>of</strong>tware. Regions<br />

with a multipoint LOD score <strong>of</strong> >1.0 or a single<br />

point LOD score <strong>of</strong> >1.5 were analyzed in all family<br />

members by fine-mapping. Selected genes from<br />

regions with potentially shared haplotypes were<br />

sequenced.<br />

Results: Four family members were definitely<br />

affected with PD, two were considered to be<br />

probably, and two possibly affected. The age <strong>of</strong><br />

onset was between 45 and 70 years. The pattern<br />

<strong>of</strong> disease transmission was compatible with<br />

autosomal dominant inheritance.Twelve regions<br />

were investigated by fine-mapping and linkage<br />

to all but one <strong>of</strong> <strong>the</strong>se regions was excluded.<br />

Thus, we identified a likely new PD gene locus<br />

with a maximum lod score <strong>of</strong> 2.45. A haplotype<br />

in a 10.7Mb region was shared by all 8 affected<br />

patients. Sequencing <strong>of</strong> <strong>the</strong> first five candidate<br />

genes in this region did not yet result in <strong>the</strong><br />

identification <strong>of</strong> a disease-associated mutation.<br />

Conclusions: Linkage studies localized a new<br />

gene locus for clinically typical, late-onset,<br />

dominantly inherited PD in this family. Fur<strong>the</strong>r<br />

mutational analyses are underway to identify <strong>the</strong><br />

causative gene.


Pos t e r ab s t r a c t P15<br />

Parkin protects <strong>the</strong> mitochondrial genome against<br />

oxidative stress<br />

Rothfuss, O. 1 , Hasegawa, T. 1 , Fischer, H. 1 , Sharma, M. 1 , Leitner, P. 1 ,<br />

Bornemann, A. 2 , Berg, D. 1 , Gasser, T. 1 , and Patenge, N. 1<br />

1 Hertie Institute for Clincical Brain Research (HIH), Center for Neurology, University<br />

Hospital Tübingen<br />

2 Institute for Brain Research, University Hospital Tübingen<br />

Background: Parkinson’s disease (PD) is a<br />

neurodegenerative disorder, characterized by<br />

<strong>the</strong> progressive loss <strong>of</strong> dopaminergic neurons<br />

in <strong>the</strong> substantia nigra pars compacta. Both<br />

environmental and genetic factors have been<br />

implicated in <strong>the</strong> development <strong>of</strong> PD. Mutations<br />

in <strong>the</strong> parkin gene (prkn) are <strong>the</strong> most common<br />

cause <strong>of</strong> recessive familial PD. Recently,<br />

evidence accumulated that parkin is involved in<br />

<strong>the</strong> maintenance <strong>of</strong> mitochondrial function and<br />

biogenesis.<br />

Objectives: The objective <strong>of</strong> this study was to<br />

investigate <strong>the</strong> influence <strong>of</strong> parkin on mitochondrial<br />

function.<br />

Methods: We have employed Chromatin<br />

Immunoprecipitation to identify parkin specific<br />

binding sites in SH-SY5Y cells as well as in brain<br />

tissue. In SH-SY5Y cell lines stably expressing<br />

wildtype parkin and six pathogenic point<br />

mutants, we evaluated <strong>the</strong> influence <strong>of</strong> parkin on<br />

mitochondrial DNA (mtDNA) using LightCycler<br />

approaches.<br />

Results: Parkin is associated physically with<br />

mtDNA in proliferating as well as in differentiated<br />

SH-SY5Y cells. This phenomenon was not limited<br />

to <strong>the</strong> regulatory sequences <strong>of</strong> <strong>the</strong> D-loop region,<br />

but was detected all over <strong>the</strong> mitochondrial<br />

genome. In vivo, <strong>the</strong> association <strong>of</strong> parkin with<br />

mtDNA could be confirmed in brain tissue <strong>of</strong> mouse<br />

and human origin. Replication and Transcription<br />

<strong>of</strong> mtDNA were enhanced in SH-SY5Y cells overexpressing<br />

<strong>the</strong> parkin gene. The ability <strong>of</strong> parkin<br />

to support mtDNA-metabolism was impaired by<br />

pathogenic parkin point mutations. Moreover, we<br />

show that parkin protects mtDNA from oxidative<br />

damage and stimulates mtDNA repair.<br />

Conclusions: Our data suggest a novel<br />

neuroprotective pathway <strong>of</strong> parkin by supporting<br />

mitochondrial function and protecting mitochondrial<br />

genetic integrity from oxidative stress.<br />

67


68<br />

Pos t e r ab s t r a c t P16<br />

Analysis <strong>of</strong> CYP7B1 in non-consanguineous cases <strong>of</strong><br />

hereditary spastic paraplegia<br />

Rebecca Schüle 1 , Elisabeth Brandt 2 , Kathrin N. Karle 1 , Maria Tsaousidou 3 ,<br />

Stephan Klebe 4 , Sven Klimpe 5 , Michaela Auer-Grumbach 6 , Andrew H. Crosby 3 ,<br />

Christian A. Hübner 2 , Ludger Schöls 1 , Thomas Deufel 2 , Christian Beetz 2<br />

1 Research Division Clinical <strong>Neurogenetics</strong>, Hertie-Institute for Clinical Brain Research<br />

and Department <strong>of</strong> Neurodegenerative Diseases, Center <strong>of</strong> Neurology, Tübingen,<br />

<strong>German</strong>y<br />

2 Institute for Clinical Chemistry and Laboratory Diagnostics, University <strong>of</strong> Jena,<br />

<strong>German</strong>y<br />

3 Department <strong>of</strong> Medical Genetics, St George’s University, London, UK<br />

4 Department <strong>of</strong> Neurology, Christian-Albrechts-University, Kiel, <strong>German</strong>y<br />

5 Department <strong>of</strong> Neurology, Johannes-Gutenberg-University Mainz, <strong>German</strong>y<br />

6 Center for Medical Research, Medical University, Graz, Austria<br />

Background: Hereditary spastic paraplegia (HSP)<br />

is a neurodegenerative condition characterised by<br />

lower limb spasticity and weakness. Homozygous<br />

mutations in CYP7B1 have been identified in<br />

several consanguineous families that represented<br />

HSP type 5 (SPG5), one <strong>of</strong> <strong>the</strong> many genetic forms<br />

<strong>of</strong> <strong>the</strong> disease.<br />

Objective: To determine frequency and phenotypic<br />

spectrum <strong>of</strong> SPG5.<br />

Methods: We used direct sequencing and<br />

multiplex ligation-dependent probe amplification to<br />

screen apparently sporadic HSP patients (n=12) as<br />

well as index patients from non-consanguineous<br />

recessive (n=8) and from dominant families (n=8)<br />

for alterations in CYP7B1.<br />

Results: One sporadic patient showing HSP<br />

as well as optic atrophy carried a homozygous<br />

nonsense mutation. Compound heterozygosity<br />

was observed in a recessive family with a clinically<br />

pure phenotype. A heterozygous missense<br />

change segregated in a small dominant family. We<br />

also found a significant association <strong>of</strong> a known<br />

coding polymorphism with cerebellar signs as<br />

complicating a primary HSP phenotype.<br />

Conclusions: Our findings suggest CYP7B1<br />

alterations to represent a ra<strong>the</strong>r frequent cause<br />

<strong>of</strong> HSP that should be considered in patients with<br />

various clinical presentations.


Pos t e r ab s t r a c t P17<br />

Genome-Wide Association Study in Parkinson’s Disease:<br />

First stage analysis<br />

C. Schulte 1 *, M. Sharma 1 *, M. Bonin 2 , P. Lichtner 3 , D. Berg 1 , C. Gieger 4 , R. Krüger 1 ,<br />

C. Klein5, G. Deuschl 6 , O. Riess 2 , T. Gasser 1<br />

1 Hertie Institute <strong>of</strong> Clinical Brain Research, Tübingen, <strong>German</strong>y;<br />

2 Department <strong>of</strong> Medical Genetics, University <strong>of</strong> Tübingen, <strong>German</strong>y;<br />

3 Institute <strong>of</strong> Human Genetics, GSF National Research Institute, Neuherberg, <strong>German</strong>y<br />

4 Institute <strong>of</strong> Epidemiology, Helmholtz Zentrum München, <strong>German</strong> Research Center for<br />

Environmental Health, Neuherberg, <strong>German</strong>y;<br />

5 Department <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, <strong>German</strong>y.<br />

6 Dept. <strong>of</strong> Neurology, Christian-Albrechts-University Kiel, Kiel, <strong>German</strong>y<br />

Background: Parkinson’s disease (PD) is<br />

<strong>the</strong> second most common neurodegenerative<br />

disorder. The etiology <strong>of</strong> PD is largely unknown.<br />

Epidemiological findings along with genetic<br />

association studies highlight a significant genetic<br />

contribution to disease risk. However, <strong>the</strong> genetics<br />

<strong>of</strong> PD have only been partially explained by genes<br />

which were identified in large multigenerational<br />

families. Genome wide association scans <strong>of</strong>fer<br />

a powerful approach to identify common genetic<br />

factors that influence <strong>the</strong> common form <strong>of</strong> PD.<br />

Objective: To identify variants influencing <strong>the</strong><br />

susceptibility to sporadic PD.<br />

Methods: We performed a genome-wide<br />

association study in PD in 757 cases and 976<br />

neurologically healthy controls. Each individual was<br />

genotyped on Illumina genotyping chips (Illumina<br />

humanhap550) yielding 561 466 genotypes/<br />

individual. SNPs with minor allele frequencies<br />

(MAF) > 5 %, Hardy-Weinberg Equilibrium p ><br />

0.01 and genotype call rates > 95 % were included<br />

in <strong>the</strong> analysis (498 560 SNPs). We performed<br />

association tests using <strong>the</strong> trend test in PLINK<br />

s<strong>of</strong>tware.<br />

Results: We identified a number <strong>of</strong> different<br />

chromosomal regions across <strong>the</strong> genome which<br />

may potentially harbour susceptibility genes for PD.<br />

Conclusions: Our data suggest that <strong>the</strong>re are<br />

genetic variants contributing to <strong>the</strong> pathogenesis<br />

<strong>of</strong> sporadic PD. The associated SNPs are currently<br />

in <strong>the</strong> genotyping phase in an independent cohort<br />

consisting <strong>of</strong> 1100 cases and 2200 controls.<br />

69


70<br />

Pos t e r ab s t r a c t P18<br />

Frequency <strong>of</strong> SPG15 in complicated hereditary spastic<br />

paraplegia (cHSP)<br />

A. Seibel 1 , N. Schlipf 2 , D. Möcke 1 l, U. Hehr 3 , S. Klebe 4 , S. Klimpe 5 , M. Syn<strong>of</strong>zik 6 ,<br />

J. Winkler 7,8 , B. Winner 7 , L. Schöls 1,6 , P. Bauer 2 , R. Schüle 1,6<br />

1 Research Division for Clinical <strong>Neurogenetics</strong>, Hertie-Institute for Clinical Brain<br />

Research, Tübingen<br />

2 Department <strong>of</strong> Medical Genetics, University <strong>of</strong> Tübingen<br />

3 Department <strong>of</strong> Human Genetics, University <strong>of</strong> Regensburg<br />

4 Department <strong>of</strong> Neurology, University <strong>of</strong> Schleswig-Holstein, Kiel<br />

5 Department <strong>of</strong> Neurology, University <strong>of</strong> Mainz<br />

6 Department <strong>of</strong> Neurodegenerative Diseases, Center for Neurology, Tübingen<br />

7 Department <strong>of</strong> Neurology, University <strong>of</strong> Regensburg<br />

8 Department <strong>of</strong> Neurology, Clinical Center Landshut<br />

MutatioBackground: Hereditary spastic<br />

paraplegias (HSP) are genetically exceedingly<br />

heterogenous. Since 2007, two genes for<br />

complicated HSP were identified: spatacsin<br />

(SPG11) and spastizin (SPG15). Both forms <strong>of</strong><br />

HSP share a similar clinical phenotype including<br />

thin corpus callosum, cognitive impairment,<br />

peripheral neuropathy mild cerebellar signs as<br />

main complicating features.<br />

Objective: To determine <strong>the</strong> frequency <strong>of</strong> SPG15<br />

in complicated early-onset HSP.<br />

Methods: All 42 exons and exon-intron boundaries<br />

<strong>of</strong> <strong>the</strong> SPG15 gene spastizin were sequenced in a<br />

cohort <strong>of</strong> 35 index patients. Patients were selected<br />

based on <strong>the</strong> following criteria: early-onset HSP<br />

(C) were identified in a boy with<br />

sporadic complicated spastic paraplegia. The first<br />

presenting symptom was slurring <strong>of</strong> speech at <strong>the</strong><br />

age <strong>of</strong> three, whereas spastic gait disturbances<br />

started at age 16. Predominantly posterior thinning<br />

<strong>of</strong> <strong>the</strong> corpus callosum and mild periventricular<br />

white matter changes were noted on MRI. A<br />

heterozygous sequence change <strong>of</strong> unknown<br />

significance (c.1925C>T, p.A642V) was found in a<br />

child with early onset spastic paraplegia that was<br />

absent in ~400 control chromosomes. No second<br />

mutation however was identified in this child.<br />

Additionally, two unpublished SNPs were identified<br />

in <strong>the</strong> sample as well as in controll samples:<br />

c.282G>A, p.M942I and c.1844C>T, p.S615F.<br />

Conclusions: SPG15 is rare (1/35) in complicated<br />

HSP. As its phenotype is virtually indistinguishable<br />

from SPG11 it is certainly advisable to screen <strong>the</strong><br />

SPG11 gene prior to SPG15 testing.


Pos t e r ab s t r a c t P19<br />

Cellular model for monogenic parkinsonism<br />

Philip Seibler, Norman Kock, Slobodanka Orolicki, Christine Klein<br />

Department <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, Lübeck, <strong>German</strong>y<br />

Background: Despite intensive, international<br />

research efforts, <strong>the</strong> cause <strong>of</strong> degeneration <strong>of</strong><br />

dopaminergic neurons in <strong>the</strong> substantia nigra<br />

observed in Parkinson’s disease (PD) is still<br />

unknown. To date, several genes have been<br />

identified, mutations in which are clearly linked<br />

to PD-specific cell loss. The availability <strong>of</strong> human<br />

neuronal cells is limited, thus requiring a model<br />

system that reflects in vivo conditions. One <strong>of</strong> <strong>the</strong><br />

disadvantages <strong>of</strong> current animal and cell culture<br />

models for monogenic PD is overexpression <strong>of</strong><br />

mutated sequences.<br />

Objective: To generate a model <strong>of</strong> “dopaminergic”<br />

fibroblasts to study <strong>the</strong> influence <strong>of</strong> Parkin<br />

mutations on dopamine-producing human skin<br />

fibroblasts.<br />

Methods: Human skin fibroblast cultures were<br />

obtained from 12 patients with Parkin mutations<br />

(two homozygous, one compound heterozygous<br />

and nine heterozygous) and 3 control individuals<br />

and grown. In order to establish dopaminergic<br />

metabolism in <strong>the</strong>se cells, <strong>the</strong> following genes were<br />

cloned and transfected via Nucle<strong>of</strong>ection (Amaxa<br />

Biosystems, Köln, <strong>German</strong>y) into fibroblasts:<br />

Tyrosine hydroxylase 2 (TH2), GTP cyclohydrolase<br />

1 (GCH1) and L-DOPA decarboxylase (DDC).<br />

Results: First, expression <strong>of</strong> <strong>the</strong> Parkin protein in<br />

fibroblasts was confirmed in <strong>the</strong> control individuals<br />

and <strong>the</strong> heterozygous mutation carriers. Thirtysix<br />

hours after transfection with TH2, GCH1 and<br />

DDC, <strong>the</strong> cells were analyzed. Overexpression<br />

<strong>of</strong> <strong>the</strong> three genes was shown by Western blot<br />

and production <strong>of</strong> dopamine was revealed highperformance<br />

liquid chromatography.<br />

Conclusions: Here we report, for <strong>the</strong> first time, a<br />

model <strong>of</strong> human skin fibroblasts, originating from<br />

PD patients, with a fully expressed dopamine<br />

metabolism. Future investigations <strong>of</strong> this cellular<br />

model regarding levels <strong>of</strong> dopamine production,<br />

cytotoxicity levels or activities <strong>of</strong> Parkin<br />

substrates, will yield new insights into disease<br />

mechanisms <strong>of</strong> PD.<br />

71


72<br />

Pos t e r ab s t r a c t P20<br />

Dopamine Receptor D3 Gene and Essential tremor in Large<br />

Series <strong>of</strong> <strong>German</strong>, Danish and French Patients<br />

Thier, S 1 , Klebe S 1 , Lorenz D 1 , Stevanin G 2,3,4 , Nebel A 5 , Feingold J 2,6 , Frederiksen<br />

H 7 , Denis E 2 , Christensen K 7 , Schreiber S 2 , Brice A 2,3,4,8 , Deuschl G 1 , Dürr A 2,3,4<br />

1 Department <strong>of</strong> Neurology, UK S-H, Campus Kiel, Kiel, <strong>German</strong>y<br />

2 AP-HP, Pitié-Salpêtrière Hospital, Department <strong>of</strong> Genetics and Cytogenetics, Paris,<br />

France<br />

3 INSERM, UMR_S679, Paris, France<br />

4 Université Pierre et Marie Curie-Paris, Pitié-Salpêtrière Hospital, Paris, France<br />

5 Institute <strong>of</strong> Clinical Molecular Biology, UK S-H, Campus Kiel, Kiel, <strong>German</strong>y<br />

6 Université Pierre et Marie Curie-Paris, Modelling in Clinical Research, Paris, France<br />

7 Danish Twin Registry, Epidemiology, Institute <strong>of</strong> Public Health, University <strong>of</strong> Sou<strong>the</strong>rn<br />

Denmark, Denmark<br />

8 AP-HP, Pitié-Salpêtrière Hospital, Federation <strong>of</strong> Neurology, Paris, France.<br />

Aim <strong>of</strong> this study: To examine <strong>the</strong> DRD3 variant<br />

in three different populations from <strong>German</strong>y,<br />

Denmark and France Background: The genetic<br />

causes <strong>of</strong> ET seem to be heterogeneous. Recently,<br />

ET has been found associated with a functional<br />

variant (Ser9Gly) <strong>of</strong> <strong>the</strong> dopamine D3-receptor,<br />

located close to <strong>the</strong> 1997-first-time described<br />

ETM1 locus on chromosome 3q13.3.Methods: We<br />

undertook an association study <strong>of</strong> <strong>the</strong> Ser9Gly<br />

variant in 299 ET patients and 528 healthy<br />

controls from three different European populations<br />

(<strong>German</strong>y, France, Denmark). ET patients contain<br />

97 sporadic cases and 202 familial cases. In<br />

addition, linkage analysis was carried out in 22<br />

informative ET families.<br />

Results: Comparing <strong>the</strong> whole study sample<br />

(n=299) with healthy controls (n=528) <strong>of</strong> <strong>the</strong><br />

same origins no difference <strong>of</strong> genotypes or allele<br />

frequency was shown between ET patients and<br />

controls. Regarding ET patients with a positive<br />

(n=202) or no family history (n=97) no difference<br />

between ET patients and healthy controls could<br />

be found. Age <strong>of</strong> onset <strong>of</strong> tremor, tremor duration<br />

and tremor severity did not show an association<br />

with <strong>the</strong> DRD3 variant. In addition, <strong>the</strong> G allele <strong>of</strong><br />

<strong>the</strong> variant was not found linked to <strong>the</strong> disease in<br />

a subset <strong>of</strong> informative ET families.Conclusion:<br />

We did not find a significant association nor<br />

linkage <strong>of</strong> <strong>the</strong> DRD3 variant to ET for <strong>the</strong> so far<br />

largest sample <strong>of</strong> European ET patients from three<br />

different national cohorts, suggesting that it is<br />

unlikely a common risk factor for ET in Caucasians.


Pos t e r ab s t r a c t P21<br />

Generation and expression characterization <strong>of</strong> BAC-HD<br />

transgenic rats with full-length mutant huntingtin<br />

Libo Yu 1 , Silke Metzger 1 , Alexandra Schwienbache r1 , Thomas Ott1, Xia<strong>of</strong>eng Gu 2 ,<br />

Michelle Gray 2 , William Yang 2 , Olaf Riess 1 and Huu Phuc Nguyen 1<br />

1 Department <strong>of</strong> Medical Genetics, Tübingen, <strong>German</strong>y<br />

2 Department <strong>of</strong> Psychiatry & Biobehavioral Sciences, Los Angeles, USA<br />

Background and objective: Our group has<br />

previously generated and characterized transgenic<br />

rats, which express a fragment <strong>of</strong> mutant<br />

huntingtin. This rat model mirrors many aspects<br />

<strong>of</strong> Huntington’s disease, but it lacks <strong>the</strong> full-length<br />

mutant protein and <strong>the</strong>refore some aspects <strong>of</strong> <strong>the</strong><br />

human condition might be imperfectly replicated.<br />

To overcome this potential disadvantage, we<br />

aimed to generate transgenic rats, which express<br />

full-length mutant human huntingtin in <strong>the</strong> same<br />

developmental and tissue- and cell-specific<br />

manner seen in patients.<br />

Methods: Bacterial artificial chromosomes (BACs)<br />

containing human genomic DNA spanning <strong>the</strong> fulllength<br />

gene with 97 CAGs including all regulatory<br />

elements, were microinjected into oocytes <strong>of</strong><br />

Sprague–Dawley rats. Founders and F1 transgenic<br />

rats were screened for copy number <strong>of</strong> BAC<br />

insertion, number <strong>of</strong> integration sites, integrity <strong>of</strong><br />

<strong>the</strong> BAC insertion and level <strong>of</strong> mutant huntingtin<br />

RNA by QPCR. Protein expression in brain was<br />

measured by Western Blot.<br />

Results: Initially we established one line, in<br />

which <strong>the</strong> full-length huntingtin expression was<br />

detected in all <strong>the</strong> brain regions at low level. 24<br />

new founders were <strong>the</strong>refore generated, which all<br />

showed germline transmission <strong>of</strong> <strong>the</strong> transgene.<br />

Many potential founders had multiple copies <strong>of</strong><br />

<strong>the</strong> transgene integrated into <strong>the</strong>ir genome, in<br />

some cases more than 5-fold in comparison to <strong>the</strong><br />

low-expressing line. Also 22 potential founders<br />

expressed <strong>the</strong> full-length mutant huntingtin gene.<br />

Expression <strong>of</strong> <strong>the</strong> full-length protein was confirmed<br />

in <strong>the</strong> brains <strong>of</strong> F1 transgenic rats, in several lines<br />

at high levels.<br />

Conclusion: We have established several lines <strong>of</strong><br />

transgenic rats, which express full-length mutant<br />

huntingtin at high levels.<br />

73


74<br />

Pos t e r ab s t r a c t P22<br />

Abnormal interhemispheric interactions and sensorimotor<br />

integration in ATP13A2 mutation carriers: a TMS study<br />

S Zittel 1 , J Kröger 1 , T Bäumer 1 , C Gerl<strong>of</strong>f 1 , HR Siebner 2 , N Brüggemann 3 ,<br />

A Schmidt 3 , J Hagenah 3 , ME Behrens 4 , A Ramirez 5 , C Klein 3 , A Münchau 1<br />

1 Department <strong>of</strong> Neurology, University Medical Center Eppendorf, Hamburg<br />

2 Department <strong>of</strong> Neurology, University <strong>of</strong> Kiel, Kiel, <strong>German</strong>y<br />

3 Department <strong>of</strong> Neurology, University <strong>of</strong> Lübeck, Lübeck, <strong>German</strong>y<br />

4 University <strong>of</strong> Santiago de Chile, Chile<br />

5 Institute <strong>of</strong> Human Genetics, University <strong>of</strong> Cologne, Cologne, <strong>German</strong>y<br />

Background: Kufor Rakeb syndrome (PARK 9) is an<br />

autosomal recessive nigro-striatal-pallidal-pyramidal<br />

neurodegeneration characterized by juvenile-onset<br />

parkinsonism, pyramidal signs, dementia and<br />

supranuclear gaze palsy. Sensorimotor integration<br />

and interhemispheric interactions investigated by<br />

transcranial magnetic stimulation (TMS) are altered<br />

in idiopathic Parkinson’s disease (IPD) with a<br />

reduced inhibitory tone. As yet it is unclear whe<strong>the</strong>r<br />

<strong>the</strong>se alterations are due to neurodegeneration<br />

per se or represent compensatory adaptive<br />

mechanisms. Here we investigated a Chilean PARK<br />

9 family using a number <strong>of</strong> TMS paradigms.<br />

Methods: Five heterozygous asymptomatic carriers<br />

<strong>of</strong> an ATP13A2 mutation, one affected homozygous<br />

carrier, two unaffected family members, and nine<br />

healthy subjects were investigated. All mutation<br />

carriers were members <strong>of</strong> one Chilean family. We<br />

studied short latency afferent inhibition (SAI) after<br />

digital nerve stimulation, interhemispheric inhibition<br />

(IHI) with a paired pulses paradigm and <strong>the</strong> ipsilateral<br />

silent period (iSP) and postinhibitory excitability to<br />

probe sensorimotor integration and interhemispheric<br />

interaction in mutation carriers. ISP and corticospinal<br />

excitability following iSP for an interval <strong>of</strong> 100 msec<br />

were measured using TMS applied over <strong>the</strong><br />

left and right primary motor cortex (M1) with an<br />

intensity <strong>of</strong> 175% resting motor threshold (RMT)<br />

during ipsilateral pinch grips <strong>of</strong> maximal voluntary<br />

contraction. IHI was investigated by a subthreshold<br />

conditioning stimulus (intensity 120% RMT) applied<br />

over left and consecutively right M1 followed by a test<br />

stimulus (intensity 120% RMT) over <strong>the</strong> contralateral<br />

motor cortex at interstimulus intervals (ISI) <strong>of</strong> 6 – 10<br />

msec. SAI was tested by electrical stimulation <strong>of</strong> <strong>the</strong><br />

right index finger followed by magnetic stimuli over<br />

<strong>the</strong> left M1 at ISIs <strong>of</strong> 25, 30 and 40 msec.<br />

Results: Postinbitory facilitation following iSP was<br />

enhanced in <strong>the</strong> group <strong>of</strong> mutation carriers at an<br />

ISI <strong>of</strong> 40 msec. SAI was stronger at an ISI <strong>of</strong> 40<br />

msec in mutation carriers as compared to healthy<br />

controls but this was not significant. IHI was normal<br />

in mutation carriers.<br />

Conclusion: Interhemispheric interactions are<br />

altered in carriers <strong>of</strong> <strong>the</strong> ATP13A2 mutation. In<br />

contrast to reduced SAI in IDP and heterozygous<br />

Parkin mutation carriers SAI was increased in<br />

ATP13A2 mutation carriers. These alterations might<br />

be a direct consequence <strong>of</strong> <strong>the</strong> PARK 9 mutation or<br />

represent compensatory adaptive changes.


sPo n s o r i n g<br />

Wir danken der Deutschen Forschungsgemeinschaft<br />

und folgenden Firmen für Ihre<br />

freundliche Unterstützung:<br />

75


76<br />

loc a l or g a n i z e r s<br />

Department <strong>of</strong> Neurology<br />

Pr<strong>of</strong>. Dr. Detlef Kömpf<br />

Pr<strong>of</strong>. Dr. Christine Klein<br />

Dr. Katja Lohmann<br />

Institute <strong>of</strong> Human Genetics<br />

Pr<strong>of</strong>. Dr. Gabriele Gillessen-Kaesbach<br />

Pr<strong>of</strong>. Dr. Christine Zühlke<br />

Department <strong>of</strong> Psychiatry<br />

and Psycho<strong>the</strong>rapy<br />

Pr<strong>of</strong>. Dr. Fritz Hohagen<br />

PD Dr. Rebekka Lencer<br />

Dr. Meike Kasten<br />

The congress has been CME-certified.


con t a c t<br />

Congress Secretariat<br />

UNIVERSITÄTSKLINIKUM Schleswig-Holstein<br />

Institut für Humangenetik<br />

Marianne Schirr<br />

Ratzeburger Allee 160<br />

23538 Lübeck, <strong>German</strong>y<br />

Phone: +49 451 500-26 21, Fax -41 87<br />

E-Mail: Marianne.Schirr@uk-sh.de<br />

www.neuro.uni-luebeck.de/dgng2008<br />

Congress Venue<br />

Audimax, AM3, University <strong>of</strong> Lübeck<br />

77


©<br />

UNIVERSITÄTSKLINIKUM<br />

Schleswig-Holstein<br />

Stabsstelle UEM, G. Weinberger; Stand September 2008

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!