24.02.2013 Views

Research Report - Nikolaus-Fiebiger-Zentrum für Molekulare Medizin

Research Report - Nikolaus-Fiebiger-Zentrum für Molekulare Medizin

Research Report - Nikolaus-Fiebiger-Zentrum für Molekulare Medizin

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Nikolaus</strong>-<strong>Fiebiger</strong>-<strong>Zentrum</strong><br />

<strong>für</strong><br />

<strong>Molekulare</strong> <strong>Medizin</strong><br />

<strong>Research</strong> <strong>Report</strong><br />

2011


Publisher<br />

<strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center for Molecular Medicine<br />

Glückstrasse 6<br />

D-91054 ERLANGEN<br />

Tel.: + 49 (9131) 85 29109<br />

Fax: + 49 (9131) 85 29111<br />

Acting Director: Prof. Dr. Jürgen Behrens<br />

Email: jbehrens@molmed.uni-erlangen.de<br />

Editing and Layout<br />

Dr. Jürgen Behrens and Angela Döbler<br />

Cover Layout<br />

Angela Döbler


PREFACE<br />

Our present report covers the period from 2009 to 2012 and documents the scientific<br />

achievements at the <strong>Nikolaus</strong>-<strong>Fiebiger</strong> Center which precipitated in a number of high<br />

impact publications in both basic and applied research. The report also highlights the<br />

personal turnover at our institution. In 2011, we were pleased to welcome Prof. Dr.<br />

Dominik Müller, expert in hypertension research as the successor of Prof. von der<br />

Mark at the chair of Experimental Medicine I. Also two new IZKF junior researchers,<br />

Dr. Beate Winner and Dr. Jens Titze who have their research focus in clinical neurobiology<br />

and kidney physiology, respectively, started in 2011. They replace Dr. Reinhard<br />

Voll, now Director of the Division of Rheumatology and Clinical Immunology, Freiburg,<br />

and Dr. Michael Wiesener, now Professor at the Medical Clinic 4, Erlangen. Several of<br />

the clinical research group leaders left to take up new positions, including Dr. Falk<br />

Nimmerjahn, now Professor of Genetics, Erlangen, Dr. Diana Dudziak, now professor<br />

of Dendritic Cell Biology, Dematology, Erlangen. We are grateful to all previous and<br />

current members for their cooperation, their assistance in the organisation of the<br />

house and most importantly for contributing to the high scientific standard of our centre<br />

by their outstanding publications.<br />

The goal of our centre is to promote scientific excellence and to foster interactions between<br />

clinical and basic researcher. A number of publications in internationally distinguished<br />

and high-impact journals can be found in the individual reference lists. Moreover,<br />

the center is a meeting place for students taking courses in Molecular Medicine,<br />

Medicine and Biology and remains attractive for a large number of PhD students and<br />

PostDocs, frequently from foreign countries. Core facilities in the house, in particular<br />

the state-of-the art cell sorting facility are used by many scientists from other institutions<br />

of the University Erlangen. Altogether these various interactions contribute to the<br />

lively atmosphere of the NFZ.<br />

On behalf of the steering committee of the NFZ I would like to take the opportunity to<br />

thank the University and University Clinics for their support, the Advisory Board of the<br />

NFZ for help in evaluation our research and the many research units from the Medical<br />

Faculty for their continued interest in our house.<br />

Erlangen, August 2012 Prof. Jürgen Behrens<br />

Acting chairman of the NFZ


TABLE OF CONTENTS<br />

SCIENTIFIC ADVISORY BOARD 1<br />

GENERAL OVERVIEW 2<br />

RESEARCH UNITS 5<br />

DEPARTMENT OF EXPERIMENTAL MEDICINE I 6<br />

DEPARTMENT OF EXPERIMENTAL MEDICINE II 26<br />

DIVISION OF MOLECULAR IMMUNOLOGY 37<br />

HEMATOPOIESIS UNIT 49<br />

JUNIOR RESEARCH GROUPS 62<br />

CLINICAL PROJECT GROUPS 80<br />

TEACHING ACTIVITIES 166<br />

SEMINARS 167


SCIENTIFIC ADVISORY BOARD<br />

The scientific standard, productivity and interactions within the <strong>Research</strong> Center for<br />

Molecular Medicine are evaluated by an International Scientific Advisory Board. The<br />

members of the Board are nominated by the Medical Faculty. The board monitors and<br />

judges the quality and stringency of current and future research projects within the<br />

Center. Furthermore the Advisory Board evaluates the proposals by young clinicians<br />

for research terms and makes recommendations to the Extended Steering Committee.<br />

Prof. Dr. C. Huber<br />

Medical Clinic III<br />

Div. of Hematology<br />

University of Mainz<br />

55101 Mainz, Germany<br />

Prof. Dr. K. Knight<br />

Professor and Chair<br />

Department of Microbiology<br />

& Immunology<br />

Loyola University of Chicago<br />

Maywood, IL 60153, U.S.A.<br />

Prof. Dr. D. Heinegard<br />

Dept. Of Cell and Molecular Biology<br />

University of Lund<br />

Box 94<br />

22100 Lund, Sweden<br />

Prof. Dr. F. Melchers<br />

Max-Planck-Institute<br />

Of Infection Biology<br />

Charitéplatz 1<br />

10117 Berlin<br />

1


GENERAL OVERVIEW<br />

Overall Structure<br />

The <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center of Molecular Medicine is a research institution of the<br />

Medical Faculty at the Friedrich-Alexander University (FAU) of Erlangen-Nuremberg.<br />

The center harbours two chairs of Experimental Medicine I and II (Connective Tissue<br />

<strong>Research</strong> and Molecular Tumor <strong>Research</strong>, respectively), a division of Molecular<br />

Immunology as part of the Department of Internal Medicine III, a division of Genetics<br />

which belongs to the Department of Genetics of the Science Faculty, as well as two<br />

junior research groups of the Interdisciplinary Clinical <strong>Research</strong> Center (IZKF) of the<br />

Medical Faculty. The intention of the research center is to strengthen biomedical<br />

research in the Medical School by stimulating cooperations between basic and clinical<br />

researchers and by giving young clinicians the opportunity to carry out competitive<br />

biomedical research projects under the infrastructure of a modern research center.<br />

The <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center of Molecular Medicine is well equipped with modern<br />

research facilities required for cell and molecular biological research and offers a<br />

variety of biochemical, immunological and cell biological seminars, guest lectures and<br />

common graduate student seminars. Central equipment such as DNA sequencing,<br />

fluorescence activated cell sorting, confocal laser microcopy, surface plasmon<br />

resonance are accessible to all scientists and technical personell. All scientists of the<br />

center are encouraged to seek interactions and cooperations with clinicians inside and<br />

outside the center, and to participate in the maintenance of the central facilities and<br />

equipment.<br />

Steering Committee<br />

The two full professors of Experimental Medicine, the associate professor and the<br />

Speaker of the Interdisciplinary Center for Clinical <strong>Research</strong> (IZKF) are members of<br />

the Steering Committee that is responsible for the scientific and administrative<br />

organization of the <strong>Research</strong> Center. The Steering Comittee elects a speaker every 2<br />

years. Young clinicians and scientists who are members of the Medical School or the<br />

University Hospital and whose research is supported by extramural grants are<br />

encouraged to apply for a maximum of up to 3 years for research space in the center.<br />

Decisions on their applications are made by the Extended Steering Committee (see<br />

Scheme) based on recommendations of the Scientific Advisory Board. These research<br />

groups have access to all central large equipment, library, computers, isotope<br />

laboratory and other central facilities; in turn, it is expected that they will participate in<br />

research seminars, journal clubs and teaching.<br />

2


Financial Concept (Running Costs)<br />

Running costs of the <strong>Research</strong> Center (electricity, water, heating, gas, cleaning,<br />

building maintenance, salaries for central administrative and technical personell etc.)<br />

are shared by the University and the University Hospital. Running cost for central large<br />

equipment, equipment maintenance and renewal, liquid nitrogen, carbon dioxide,<br />

central computer software, library, isotope waste, dish washing, animal costs will be<br />

shared among the users.<br />

♦ Library<br />

♦ Seminar rooms<br />

Facilities and Equipment<br />

Central laboratories and facilities<br />

♦ S2 Biological Safety Laboratory<br />

♦ Isotope laboratory (S2) for experiments with 125 Iodine, 51 Cr, 45 Ca etc<br />

♦ Temporary animal rooms<br />

♦ Confocal microscope<br />

Central major equipment<br />

♦ High-speed cell sorter, analytical fluorescence-activated cell sorters<br />

♦ DNA Sequencer<br />

♦ BIAcore Surface Plasmon Resonance<br />

♦ Phosphoimager<br />

♦ Luminoimager<br />

♦ Preparative liquid chromatography (BioPerseptive)<br />

♦ Microinjection unit<br />

♦ Liquid nitrogen cell storage system<br />

4


RESEARCH UNITS<br />

� Department of Experimental Medicine I<br />

Head: Prof. K. von der Mark until 3/2011<br />

Prof. D. Müller from 4/2011<br />

� Department of Experimental Medicine II<br />

(Molecular Tumor <strong>Research</strong>)<br />

Head: Prof. J. Behrens<br />

� Division of Molecular Immunology<br />

Department of Internal Medicine III<br />

Head: Prof. H.-M. Jäck<br />

� Hematopoiesis Unit, Chair of Genetics,<br />

Department of Biology<br />

Faculty of Natural Sciences II<br />

Head: Prof. T. Winkler<br />

� Junior <strong>Research</strong> Groups of the IZKF<br />

Heads: PD Dr. R. Voll*<br />

PD Dr. M. Wiesener*<br />

PD Dr. B. Winner #<br />

PD Dr. J. Titze #<br />

� Clinical <strong>Research</strong> Groups<br />

PD Dr. J. Distler* Prof. A. Bozec #<br />

Prof. D. Dudziak* PD Dr. K. Gelse #<br />

Prof. F. Nimmerjahn* PD Dr. U. Kneser #<br />

PD Dr. J. Titze* Dr. G. Krönke #<br />

Prof. R. Voll* Dr. M. Stock #<br />

Prof. H. Taubert #<br />

Prof. E. Ullrich #<br />

PD Dr. M. Wiesener #<br />

5<br />

* previous<br />

# current


DEPARTMENT OF EXPERIMENTAL MEDICINE I<br />

Matrix Biology/ Molecular Pathology<br />

Head Klaus von der Mark, Dr. rer.nat<br />

(till 3/2011) Prof. of Experimental Medicine<br />

Address: Department of Experimental Medicine I<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Phone: + 49 (9131) 85 29124<br />

Fax: + 49 (9131) 85 26341<br />

E-mail: kvdmark@molmed. uni-erlangen.de<br />

Homepage: www.em1.molmed.uni-erlangen.de<br />

Supporting Staff<br />

Irene Bielmeier, Secretary<br />

Postdoctoral Fellows<br />

.<br />

Jung Park, Dr,med. dent.<br />

Helga von der Mark, Dr.rer. nat<br />

Takako Sasaki, Ph.D.<br />

Michael Stock, Dr. rer. nat.<br />

Cordula Surman-Schmitt, Dr. rer. nat.<br />

Nicole Eitzinger<br />

( � 10(2008)<br />

(��9/2008<br />

(11/2008 �)<br />

(��10/2010<br />

(��9/2011<br />

Doctoral Students (Biology)<br />

6<br />

PhD 8/2011


.Svitlana Golovchenko<br />

Doctoral Students (Molecular Medicine)<br />

( 10/2011)<br />

Diploma Students<br />

Catharina Müller(2008)<br />

Svitlana Ryabova-Golovchenko (2010)<br />

MatthiasGebhard (2011)<br />

Eva Bauer<br />

Friederike Pausch<br />

Britta Schlund<br />

Technicians<br />

General<br />

The legal time as head of the department of Experimental Medicine I ended for Prof.<br />

K. von der Mark with his official retirement date at Oct. 1, 2008. Until the installment<br />

of the new head of the Department, Prof. Dominik Müller, in April 2011, K. von der<br />

Mark continued to conduct research and teaching in the Department, but with<br />

strongly reduced staff and student numbers. With retirement of Dr. Helga von der<br />

Mark, the integrin research project was terminated, but collaborative studies with Dr.<br />

Jung Park, now Childrens Hospital, FAU (Prof. Holm Schneider) and Prof. Schmuki,<br />

Dept. of Material Sciences, FAU, on translational aspects of Titandioxide nanotubes<br />

are still going on. The research work of K. von der Mark focuses on two topics: 1) the<br />

role of hypertrophic chondrocytes in controlling subchondral trabecular bone<br />

formation by deleting �-catenin with a BACCol10Cre –deleter mouse, and 2) cell<br />

fate tracing and transdifferentiation of hypertrophic chondrocytes to osteoblasts using<br />

a BAC Col10Cre;ROSA-YFP reporter mice. The research projects on the new<br />

cartilage-specific proteins UCMA and the Wnt –inhibitor Wif 1 were continued under<br />

the supervision of Dr. M. Stock, now as research staff member in Internal Medicine<br />

III (Prof. G.Schett).<br />

A new research project on the role of fibulin-4 was started by Dr. Takako Sasaki in<br />

Nov. 2008 who received an independent research grant from the DFG in 2010.<br />

7


<strong>Research</strong> projects:<br />

I. Analyzing the role of hypertrophic chondrocytes in endochondral bone<br />

formation using BACCol10Cre deleter mice (K.von der Mark)<br />

II. Cellular response to nanoscale patterns of Titandioxide surfaces (J.Park)<br />

III. Control of collagen crosslinking and connective tissue stability by fibulin-4<br />

(Takako Sasaki)<br />

IV. Characterization of new cartilage matrix proteins (Michael Stock)<br />

<strong>Research</strong><br />

I. Analyzing the role of hypertrophic chondrocytes in endochondral bone<br />

formation using BACCol10Cre deleter mice (P.I. :Klaus von der Mark)<br />

A) Deletion of �- catenin in hypertrophic growth plate chondrocytes impairs<br />

trabecular bone formation<br />

Svitlana Golovchenko, Sonja Gebhard, Britta Schlund, Matthias Gebhardt, Andreas<br />

Hess, Klaus von der Mark<br />

In numerous studies it has been shown that canonical Wnt signalling plays a central<br />

but complex role in the regulation of chondrogenic and osteogenic differentiation<br />

during skeletal development Continuous Wnt/ �-catenin signalling in nascent<br />

cartilaginous skeletal elements blocks chondrocyte hypertrophy and endochondral<br />

ossification, whereas �-catenin overexpression at later stages stimulates hypertrophy<br />

and Col10a1 expression, mediated by binding to the Runx2 promoter and activating<br />

thereby expression of Col10a1. Furthermore, activation of Wnt/ �-catenin signalling in<br />

mature chondrocytes stimulated the expression of markers of late hypertrophy<br />

including MMP13 and VEGF and the expression other MMP genes.<br />

The role of Wnt/ �-catenin signalling in bone development is even more complicated<br />

in light of report showing that Wnt signalling is also involved in the control of bone<br />

remodelling by regulating the expression RANKL and osteoprotegerin in osteoblasts,<br />

thereby controlling osteoclast differentiation. Since RANKL and OPG are also<br />

expressed in hypertrophic chondrocytes, we tested the hypothesis that inhibition of �catenin<br />

signalling in hypertrophic chondrocytes might interfere with the RANKL/OPG<br />

balance and thus affect trabecular bone formation in growing bones.<br />

For this purpose, we specifically deleted the �-catenin gene in hypertrophic<br />

chondrocytes by crossing a ctnnb1 fl/fl mouse with a BAC-Col10a1-Cre deleter<br />

mouse. Surprisingly, the Cre-positive, ctnnb1 -/- mice (BCat-ko mice) revealed<br />

morphologically normal growth plate cartilage, but a substantial deficiency in<br />

trabecular bone, with increasing loss during postnatal development. Collagen I<br />

immunostaining and Alcian Blue staining for proteoglycans showed that not only the<br />

osteoid seam of bone trabeculae was missing, but also the calcified cartilage core,<br />

indicating excess of osteoclast activity. This notion was confirmed by TRAP staining<br />

8


and in situ hybridisation analysis of MMP9 which revealed a dramatically enhanced<br />

concentration of osteoclasts at the cartilage-bone marrow border in BCat-ko mice.<br />

Quantitative analysis of mRNA isolated from hypertrophic chondrocytes or whole<br />

bones of BCat–ko mice and wt littermates indicated significantly enhanced RANKL<br />

mRNA levels in BCat ko samples. In light of recent reports indicating that not<br />

osteoblasts, but osteocytes and hypertrophic chondrocytes are the main sources for<br />

RANKL synthesis, our findings support the provocative concept that it is the �-catenin<br />

in hypertrophic chondrocytes which predominantly controls the extent of trabecular<br />

bone formation in postnatal and juvenile development by regulating RANKL<br />

expression.<br />

Fig.1a,b: In situ hybridization analysis of newborn wild type and BACCol10Cre; ctnnb1 -/-<br />

(= BCat-ko) embryonic littermates (2d) shows that ��catenin mRNA expression is<br />

substantially reduced in the BCat-ko hypertrophic zones (b) as compared to wt cartilage<br />

(a), but equal in proliferating chondrocytes and in the bone marrow zone. c,d: Alcian blue<br />

staining marks growth plate cartilage and the calcified cartilage in subchondral trabeculae of<br />

wt mice (c), and demonstrates substantial loss of trabeculae in BCat-ko (d). (P7 tibia). e,f:<br />

Staining for TRAP reveals a high concentration of osteoclasts (Blue arrows) at the erosion<br />

zone of hypertrophic cartilage in BCat-ko mice (f), while in the wt spongiosa osteoclasts are<br />

evenly distributed between cartilage erosion zone and bone trabeculae (e). (P17,<br />

humerus).g) Real Time PCR analysis mRNA levels of hypertrophic chondrocytes isolated<br />

from growth plates of wt and BCAT-ko mice (P5) confirm reduction of �-catenin mRNA and<br />

indicate enhanced RANKL expression in Cat-ko hypertrophic chondrocytes.<br />

B) Do trabecular osteoblasts originate from hypertrophic chondrocytes?<br />

Matthias Gebhardt, Cordula Surmann-Schmitt, Jung Park, Britta Schlund, Klaus von der<br />

Mark<br />

Although the above findings indicate that the lack of trabecular bone caused by<br />

inactivation of the �-catenin gene in hypertrophic chondrocytes is due to enhanced<br />

9


osteoclast activity, we cannot rule out the possibility that the deletion of �-catenin in<br />

hypertrophic chondrocytes also impairs the differentiation of osteoblasts. This would<br />

be in line with a hypothesis by H. Roach et al (1996) who proposed<br />

transdifferentiation of terminally differentiated hypertrophic chondrocytes at the<br />

cartilage-bone marrow interface into osteoblasts, although the general<br />

understanding is that terminally differentiated chondrocytes die by apoptosis. To test<br />

this hypothesis, our BAC-Col10Cre deleter mice were mated to ROSA26LacZ<br />

reporter mice, and LacZ expression was used to trace the cell fate of hypertrophic<br />

chondrocyte. In fact, while X-gal staining of BAC Col10cre; ROSA26LacZ embryos<br />

labeled exclusively hypertrophic chondrocytes as expected, in postnatal stages<br />

LacZ activity was also seen in bone marrow and even in osteoblasts-like cells lining<br />

endosteal bone trabeculae. In order to exclude unspecific expression of Cre in cells<br />

other than hypertrophic chondrocytes, e.g. in bone marrow cells or osteoblast<br />

precursors, resulting in unspecific recombination of the floxed ROSA;LacZ locus,<br />

Cre expression in BACCol10Cre;ROSALacZ mice was analyzed by in situ<br />

hybridization. Cre signals were only seen hypertrophic chondrocytes, but not in<br />

other cells or tissues of the mouse embryo.<br />

To confirm the osteogenic phenotype of the LacZ-positive cells in the bone marrow,<br />

BACCol10Cre;ROSAYFP mice were generated and analyzed for expression of YFP<br />

expression osteogenic markers such as collagen I, osteocalcin and osterix.<br />

Immunofluorescence analysis revealed YFP and collagen I positive cells lining<br />

subchondral trabeculae, confirming their osteogenic phenotype. Also the<br />

immunofluorescence analysis of endosteal cells obtained after flushing long bones<br />

of 2-3 wk old BACCol10Cre;ROSA-YFP mice with collagenase showed YFP<br />

positive cells staining for collagen I and osteocalcin. These results strongly support<br />

the notion that not all terminal differentiated hypertrophic chondrocytes die by<br />

apoptosis, but may – at least to some extent – transdifferentiate to trabecular<br />

osteoblasts.<br />

a b<br />

c<br />

d e<br />

Figure 2: X-gal staining detects LacZ reporter gene activity in BACCol10Cre; ROSA26-<br />

LacZ transgenic mice in hypertrophic chondrocytes in E14.5-E18.5 embryos (a), in<br />

postnatal stages (P6) also in bone marrow and trabecular osteoblasts (c,d, arrows). A<br />

similar reactivity is seen in vertebrae (D) and long bones (e) of BACCol10Cre; ROSA-YFP<br />

mice.<br />

10


The mechanism of this transdifferentiation process remains to be elucidated. We<br />

have preliminary evidence that after opening of the chondrocyte lacunae by<br />

osteoclasts hypertrophic chondrocytes de-differentiate to bone marrow stem cells<br />

which may then differentiate into osteoblasts precursor cells and other<br />

mesenchymal cells. Studies to confirm this hypothesis are under investigation.<br />

Fig.3 Fig. 4<br />

Figure 3. YFP positive endosteal and bone marrow cells express osteoblast marker<br />

gene mRNA. Adherent bone marrow and endosteal cells were isolated from long bones of<br />

P21 BAC Col10 Cre;Rosa26-YFP reporter mice by bone flushing with collagenase P and<br />

cultured for two weeks. (A) YFP positive cells were sorted by FACS. Gating for viable cells<br />

(89.5% of total cells) is illustrated in the upper panel (pink circle). Lower panel: analysis of<br />

viable cells for YFP fluorescence reveals a clearly defined population of positive cells<br />

(12.4%). (B) RT-PCR analysis of RNA isolated from YFP + and YFP - cells after sorting and<br />

reverse transcription. Expression of osteoblast markers was analyzed using specific primers<br />

for the marker genes osterix, Runx2, osteocalcin, and Col1a1). Ppia (cyclophilin a) was used<br />

as a housekeeping gene control. Gel electrophoresis of PCR products shows, that YFP<br />

positive cells express high message levels of all four osteoblast marker genes (green box).<br />

Cal.: calvarial cDNA.<br />

Figure 4: Hypertrophic chondrocytes are an additional source of osteoblasts<br />

<strong>Report</strong>er gene expression is initiated in hypertrophic chondrocytes (HCs). The progeny of<br />

HCs continues reporter gene expression and can be detected as YFP positive trabecular<br />

osteoblasts (OBs). Some HCs are eliminated from the growth plate by apoptosis.<br />

Additionally, osteoblast precursor cells (OBP) invade the cartilage template and give rise to<br />

trabecular osteoblasts. Grey: cartilage matrix. Brown: bone trabeculae. Sand: bone marrow.<br />

PCs: proliferating chondrocytes. RCs: resting chondrocytes<br />

II: Synergistic control of mesenchymal stem cell differentiation by<br />

nanoscale surface geometry and immobilized growth factors on TiO2<br />

nanotubes ( P.I.: Dr. Jung Park)<br />

Park J, Bauer S, Pittrof A, Killian MS, Schmuki P, von der Mark K.<br />

11


Previously we have shown that cellular responses of bone marrow mesenchymal<br />

stem cells including adhesion, proliferation, migration, differentiation and apoptosis<br />

on titandioxide nanotube surfaces are strictly dependent on the nanoscale diameter<br />

of the tubular surfaces, with an optimum response on 15 nm diameter TiO2<br />

nanotubes (Park et al, 2007). In subsequent studies we asked the question whether<br />

combined environmental signals provided by nanoscale topography and by growth<br />

factors control cell behavior of mesenchymal stem cells (MSCs) in a synergistic or<br />

simply additive manner. Differentiation of MSCs to osteoblasts, chondrocytes and<br />

endothelial cells was studied on vertically aligned TiO2 nanotubes of size 15 and 100<br />

nm with and without immobilized bone morphogenetic protein-2 (BMP-2) and other<br />

growth factors such as EGF. Covalent immobilization of these growth factors onto the<br />

oxide surfaces was achieved by N,N-carbonyldiimidazole (CDI) coupling via binding<br />

to amine groups of the proteins either directly or via a spacer, namely 11-hydroxyundecylphosphonic<br />

acid (PhoA). Although BMP-2 coating stimulates both<br />

chondrogenic and osteogenic differentiation of MSCs, the response strongly<br />

depended on the surface nanoscale geometry of the BMP-2-coated nanotubes.<br />

Chondrogenic differentiation was strongly supported on 100 nm BMP-2-coated<br />

nanotubes, but not on 15 nm nanotubes, which induce spreading and dedifferentiation<br />

of chondrocytes. A similar response was observed with primary<br />

chondrocytes, which maintained their chondrogenic phenotype on BMP-2-coated 100<br />

nm nanotubes, but de-differentiated on 15 nm nanotubes. In contrast, osteogenic<br />

differentiation was greatly enhanced on 15 nm but not on 100 nm BMP-2-coated<br />

nanotubes as shown previously. Furthermore, covalent immobilization of BMP-2<br />

rescued MSCs from apoptosis occurring on uncoated 100 nm TiO2 nanotube<br />

surfaces. Thus, combined signals provided by BMP-2 immobilized to a defined lateral<br />

nanoscale spacing geometry seem to contain environmental cues that are able to<br />

modulate a lineage-specific decision of MSC differentiation and cell survival in a<br />

synergistic manner.<br />

III. The role of fibulin-4 in development and homeostasis of elastic<br />

components of the skeletal connective tissue (P.I. Dr.Takako Sasaki)<br />

Takako Sasaki, Eva Bauer, K.von der Mark<br />

The goal of this project is to elucidate the role of fibulin-4, a component of fibrillin<br />

microfibrils, in skeletal development and homeostasis. A recent gene targeting study<br />

has revealed fibulin-4 to be an essential molecule for the elastic fiber assembly<br />

during embryonic development. Mice deficient in fibulin-4 die perinatally due to<br />

cardiovascular and lung abnormalities, and similar phenotypes were reported for<br />

fibrillin-1 and lysyl oxidase (LOX) deficient mice. It has been proposed that fibrillin-1<br />

and fibulin containing microfibrils have an important role in sequestering cytokines,<br />

since perturbation of this function contributes to the pathogenesis of the disease.<br />

Thus, a hypomorphic in-frame deletion in the fibrillin-1 mutant mice, an accepted<br />

model of Marfan syndrome, leads to dysregulation of TGF-� activation and signaling.<br />

12


Increased TGF-� signaling in the cardiovascular system has also been found in mice<br />

hypomorphic for fibulin-4 and in the patient positive FBLN4 mutation. Recently, we<br />

found fibulin-4 expression also in fetal growth plate cartilage. Preliminary data on<br />

Fbln4 null mice showing reduced bone mass and increased bone fragility, and<br />

mutations found in human patients indicate that fibulin-4 may be also involved in<br />

skeletal development. The goal of this study is to obtain further insight into molecular<br />

changes causing the observed of skeletal alterations in fibulin-4 deficient mice such<br />

as limb contracture, detached distal phalanges and reduced bone mass.<br />

Results:<br />

Biochemical analyses on bone revealed that collagen I from fibulin-4 null mice was<br />

less cross-linked compared with those from wild type and heterozygous mice.<br />

Reduction of proteolytic activation of lysyl oxidase (LOX) which is essential for<br />

collagen crosslinking was detected not only in osteoblast culture but also in<br />

chondrocyte and fibroblast culture from fibulin-4 null mice. The mechanism how<br />

fibulin-4 is involved in LOX activation will be elucidated in vitro. It is still necessary to<br />

quantify the crosslink markers hyroxylyslpyridinoline (HP)/lysylpyridinoline (LP) of<br />

some more samples.<br />

Fig.5 SDS Page of collagens extracted with<br />

acid form bones of fibulin-4 k.o. mice<br />

shows significantly enhanced amounts of<br />

soluble collagen chains (a1(I) and a2(I)) ,<br />

indicating impaired crosslink formation,<br />

while collagens from wild type or<br />

heterozygote bones remain mostly insoluble<br />

2) There is ample evidence in the literature for a role of fibulin-4 in extracellular<br />

assembly of collagen and elastin. Therefore, particular attention was paid to fibulin-4<br />

binding proteins such as LTBPs and lysyloxidases in order to elucidate the<br />

mechanisms by which the absence of fibulin-4 leads to abnormalities in mice and<br />

human. To this aim, four mutations in fibulin-4 found in human patients were<br />

introduced in recombinant fibulin-4. The C276Y mutant fibulin-4 was not secreted<br />

from transfected 293 cells indicating that this cysteine residue is important for the<br />

protein folding. Other mutants can be secreted and purified except R279C mutant.<br />

These data suggest that the homozygous missense mutation of C276Y and the<br />

compound heterozygosity resulted in null mutation of fibulin-4 therefore those<br />

patients died very early, similar to fibulin-4 null mice. It will be tested whether cells<br />

other than 293 cells can secrete mutant fibulin-4. Some of fibulin-4 mutants exhibited<br />

reduced binding activities with different ligands. The E57K and E126K mutants were<br />

more susceptible to the proteases tested, suggesting that these mutation resulted in<br />

loss of calcium, causing instability in the conformation of EGF-like domain .<br />

13


The proposed studies will provide novel insights into the role of fibulin-4 in skeletal<br />

system as well as in the development and homeostasis of cardiovascular tissue.<br />

IV. Characterization of new cartilage matrix proteins<br />

(P.I.: Dr .Michael Stock)<br />

1. mWif-1 is expressed at cartilage-mesenchyme interfaces and impedes<br />

Wnt3a-mediated inhibition of chondrogenesis<br />

Cordula Surmann-Schmitt, Nathalie Widmann, Uwe Dietz, Bernhard Saeger, Nicole<br />

Eitzinger, Yukio Nakamura, Marianne Rattel, Richard Latham, Christine Hartmann,<br />

Helga von der Mark, Georg Schett, Klaus von der Mark and Michael Stock<br />

Skeletal development is controlled by a complex network of growth factors. Important<br />

signalling cascades regulating chondrocyte differentiation and maturation include<br />

signals mediated by the TGFβ/BMP puderfamily of growth factors, fibroblast growth<br />

factors (FGF), the Ihh/PTHrp system and Wnt factors. Wnt proteins are involved in<br />

the regulation of all steps of cartilage development. Their activity to a large extent<br />

regulated at the extracellular level by factors like the Dkk family, sFRPs, Cerberus<br />

and Wnt inhibitory factor 1 (Wif-1).<br />

In this study we provide evidence that Wif-1 is highly expressed at cartilagemesenchyme<br />

interfaces of the early developing skeleton. In fetal and postnatal<br />

skeletal development, Wif-1 expression is mainly confined to a zone of only a few cell<br />

layers in the upper hyaline layer of epiphyseal and articular cartilage. Significant Wif-<br />

1 expression was also detected in trabecular bone. In order to identify cartilagerelated<br />

Wnt factors that Wif-1 might interact with in vivo, we performed<br />

coimmunoprecipitation and pull-down assays using recombinant Wif-1 and Wnt<br />

factors. Thereby, we could identify specific binding of Wif-1 to Wnt3a, Wnt4, Wnt5a,<br />

Wnt7a, Wnt9a and Wnt11. We could demonstrate that Wif-1 was able to block Wnt3a<br />

mediated activation of the canonical Wnt signalling pathway, probably mediated by<br />

14


Figure 6: Wif-1 blocks Wnt3a activity<br />

The chondrogenic cell line 4C6 was stimulated as indicated with 25 µl/ml or 100 µl/ml Wnt3aconditioned<br />

medium in the presence or absence (Ctrl.) of 10 µg/ml recombinant Wif-1.<br />

Wnt3a-dependent accumulation and nuclear translocation of β-catenin was detected by<br />

immunofluorescence 5 hours after stimulation.<br />

B: Mouse limb-bud cells were grown in micromass cultures and treated with Wnt3a and Wif-1<br />

as indicated. After RNA extraction, Col2a1 mRNA levels were determined by real-time PCR<br />

using cyclophilin A levels for normalisation. Values are means ± s.d.<br />

physical interaction of Wif-1 and Wnt3a. Consequently, Wif-1 impaired growth of<br />

mesenchymal precursor cells and neutralised Wnt3a-mediated inhibition of<br />

chondrogenesis in micromass cultures of embryonic chick limb bud cells.<br />

These results identify Wif-1 as a novel extracellular Wnt modulator in cartilage<br />

biology.<br />

2. The Wnt Antagonist Wif-1 interacts with CTGF and Inhibits CTGF<br />

Activity<br />

Cordula Surmann-Schmitt, Takako Sasaki, Takako Hattori, Nicole Eitzinger, Georg<br />

Schett, Klaus von der Mark, and Michael Stock<br />

A recent study showed that shifted, the Drosophila orthologue of Wif-1, is involved in<br />

the control of hedgehog signalling. Therefore, we hypothesized that in mammals Wif-<br />

1 may also be associated with signalling pathways other than the Wnt cascade.<br />

Since Wif-1 exerts its regulatory role in Wnt signalling by direct interaction with Wnt<br />

ligands, we searched for novel protein interaction partners of Wif-1, which are<br />

involved in signalling pathways other than Wnt signalling. Therefore, we performed a<br />

yeast-two-hybrid approach to identify such novel protein interactions. Thereby, we<br />

identified the matricellular signalling molecule connective tissue growth factor<br />

(CTGF/CCN2) as a potential Wif-1-interacting protein. We confirmed that Wif-1<br />

physically binds to connective tissue growth factor (CTGF/CCN2) in vitro,<br />

predominantly by interaction with the C-terminal cysteine knot domain of CTGF. In<br />

vivo such an interaction appears also likely since the expression patterns of these<br />

two secreted proteins overlap in peripheral zones of epiphyseal cartilage. In<br />

chondrocytes CTGF has been shown to induce the expression of cartilage matrix<br />

genes such as aggrecan (Acan) and collagen2a1 (Col2a1). Here we could<br />

demonstrate that Wif-1 is capable to interfere with CTGF-dependent induction of<br />

Acan and Col2a1 gene expression in primary murine chondrocytes. Conversely,<br />

CTGF does not interfere with Wif-1-dependent inhibition of Wnt signalling.<br />

These results indicate that Wif-1 may be a multifunctional modulator of signalling<br />

pathways in the cartilage compartment.<br />

3. Ucma is not Necessary for Normal Development of the Mouse Skeleton<br />

Nicole Eitzinger, Cordula Surmann-Schmitt, Michael Bösl, Georg Schett, Klaus<br />

Engelke, Andreas Hess, Klaus von der Mark, and Michael Stock<br />

15


We recently introduced Ucma (Upper zone of growth plate and Cartilage Matrix<br />

Associated protein), a highly conserved tyrosine,sulphated, secreted protein of Mw<br />

17 kDa, which is expressed by juvenile chondrocytes. Initial in vitro experiments<br />

indicated that Ucma may be involved in skeletal development by affecting<br />

differentiation of pre-osteoblasts. Here we present the generation and analysis of a<br />

Ucma-deficient mouse strain, which should help to identify the physiological role of<br />

Ucma. Ucma gene-deficient mice were generated by introducing a lacZ/neoRcassette<br />

into the first exon of the Ucma gene. This mutation results in the complete<br />

loss of Ucma mRNA and protein expression. As expected due to the cartilage-<br />

specific expression of Ucma, these mice are viable and fertile. Surprisingly, however,<br />

Ucma-deficient mice appear to develop normally. Neither ossification and calcification<br />

of the skeleton, nor cartilage development were affected by Ucma-deficiency. This is<br />

particularly surprising since in a recent study, the knockdown of Ucma in zebrafish<br />

resulted in a severely disturbed formation of craniofacial cartilage.<br />

Our earlier studies have shown that Ucma is most prominently expressed perinatally,<br />

a finding that supported our hypothesis that Ucma may play a role in skeletal<br />

development. In the study presented here we could show that Ucma is additionally<br />

expressed in the cartilage of ribs and growth plate in adult mice. This may indicate<br />

that Ucma might be involved in skeletal homeostasis and in the mechanical<br />

properties of the skeleton during challenging conditions such as ageing or disease.<br />

Publications<br />

2012<br />

Eitzinger N, Surmann-Schmitt C, Bösl M, Schett G, Engelke K, Hess A, von der<br />

Mark K, Stock M. Ucma is not necessary for normal development of the mouse<br />

skeleton. Bone. 2012 Mar;50(3):670-80. Epub 2011 Dec 2. PubMed PMID: 22155508.<br />

Park J, Bauer S, Pittrof A, Killian MS, Schmuki P, von der Mark K. Synergistic<br />

control of mesenchymal stem cell differentiation by nanoscale surface geometry<br />

and immobilized growth factors on TiO2 nanotubes. Small. 2012 Jan 9;8(1):98-107.<br />

doi: 10.1002/smll.201100790. Epub 2011 Nov 18. PubMed PMID: 22095845.<br />

Surmann-Schmitt C, Sasaki T, Hattori T, Eitzinger N, Schett G, von der Mark K,<br />

Stock M. The Wnt antagonist Wif-1 interacts with CTGF and inhibits CTGF activity.<br />

J Cell Physiol. 2012 May;227(5):2207-16. doi: 10.1002/jcp.22957. PubMed PMID:<br />

21928342.<br />

2011<br />

Bauer S, Park J, Pittrof A, Song YY, von der Mark K, Schmuki P. Covalent<br />

functionalization of TiO2 nanotube arrays with EGF and BMP-2 for modified<br />

behavior towards mesenchymal stem cells. Integr Biol (Camb). 2011<br />

Sep;3(9):927-36. Epub 2011 Aug 10. PubMed PMID: 21829821.<br />

Gelse K, Klinger P, Koch M, Surmann-Schmitt C, von der Mark K, Swoboda B,<br />

Hennig FF, Gusinde J. Thrombospondin-1 prevents excessive ossification in<br />

16


cartilage repair tissue induced by osteogenic protein-1. Tissue Eng Part A. 2011<br />

Aug;17(15-16):2101-12. Epub 2011 Jun 1. PubMed PMID: 21513464.<br />

Klinger P, Surmann-Schmitt C, Brem M, Swoboda B, Distler JH, Carl HD, von der<br />

Mark K, Hennig FF, Gelse K. Chondromodulin 1 stabilizes the chondrocyte phenotype and<br />

inhibits endochondral ossification of porcine cartilage repair tissue.<br />

Arthritis Rheum. 2011 Sep;63(9):2721-31. doi: 10.1002/art.30335. PubMed PMID:<br />

21391200.<br />

2010<br />

Hattori T, Müller C, Gebhard S, Bauer E, Pausch F, Schlund B, Bösl MR, Hess A,<br />

Surmann-Schmitt C, von der Mark H, de Crombrugghe B, von der Mark K. SOX9 is a<br />

major negative regulator of cartilage vascularization, bone marrow formation and<br />

endochondral ossification. Development. 2010 Mar;137(6):901-11. PubMed PMID:<br />

20179096.<br />

Dragu A, Schnürer S, Surmann-Schmitt C, von der Mark K, Stürzl M, Unglaub F,<br />

Wolf MB, Leffler M, Beier JP, Kneser U, Horch RE. Gene expression analysis of<br />

ischaemia and reperfusion in human microsurgical free muscle tissue transfer. J<br />

Cell Mol Med. 2011 Apr;15(4):983-93. doi: 10.1111/j.1582-4934.2010.01061.x.<br />

PubMed PMID: 20345846.<br />

Belluoccio D, Etich J, Rosenbaum S, Frie C, Grskovic I, Stermann J, Ehlen H,<br />

Vogel S, Zaucke F, von der Mark K, Bateman JF, Brachvogel B. Sorting of growth<br />

plate chondrocytes allows the isolation and characterization of cells of a<br />

defined differentiation status. J Bone Miner Res. 2010 Jun;25(6):1267-81. PubMed<br />

PMID: 20200945.<br />

Krönke G, Uderhardt S, Kim KA, Stock M, Scholtysek C, Zaiss MM, et al. R-spondin 1<br />

protects against inflammatory bone damage during murine arthritis by modulating the Wnt<br />

pathway. Arthritis Rheum. 2010 Aug; 62(8):2303-2312.<br />

2009<br />

Bauer S, Park J, Faltenbacher J, Berger S, von der Mark K, Schmuki P. Size<br />

selective behavior of mesenchymal stem cells on ZrO(2) and TiO(2) nanotube<br />

arrays. Integr Biol (Camb). 2009 Sep;1(8-9):525-32. Epub 2009 Jun 19. PubMed<br />

PMID: 20023767.<br />

Bruckner-Tuderman L, von der Mark K, Pihlajaniemi T, Unsicker K. Cell<br />

interactions with the extracellular matrix. Cell Tissue Res. 2010 Jan;339(1):1-5.<br />

PubMed PMID: 19902257.<br />

von der Mark K, Park J, Bauer S, Schmuki P. Nanoscale engineering of<br />

biomimetic surfaces: cues from the extracellular matrix. Cell Tissue Res. 2010<br />

Jan;339(1):131-53. Epub 2009 Nov 7. Review. PubMed PMID: 19898872.<br />

Surmann-Schmitt C, Widmann N, Mallein-Gerin F, von der Mark K, Stock M.<br />

Stable subclones of the chondrogenic murine cell line MC615 mimic distinct stages<br />

of chondrocyte differentiation. J Cell Biochem. 2009 Oct 15;108(3):589-99. PubMed<br />

PMID: 19670270.<br />

Park J, Bauer S, Schmuki P, von der Mark K. Narrow window in nanoscale<br />

dependent activation of endothelial cell growth and differentiation on TiO2<br />

nanotube surfaces. Nano Lett. 2009 Sep;9(9):3157-64. PubMed PMID: 19653637.<br />

17


Park J, Bauer S, Schlegel KA, Neukam FW, von der Mark K, Schmuki P. TiO2<br />

nanotube surfaces: 15 nm--an optimal length scale of surface topography for cell<br />

adhesion and differentiation. Small. 2009 Mar;5(6):666-71. PubMed PMID: 19235196.<br />

von der Mark K, Bauer S, Park J, Schmuki P. Another look at "Stem cell fate<br />

dictated solely by altered nanotube dimension". Proc Natl Acad Sci U S A. 2009<br />

Jun 16;106(24):E60; author reply E61. Epub 2009 Jun 8. PubMed PMID: 19506261;<br />

Surmann-Schmitt C, Widmann N, Dietz U, Saeger B, Eitzinger N, Nakamura Y,<br />

Rattel M, Latham R, Hartmann C, von der Mark H, Schett G, von der Mark K, Stock<br />

M. Wif-1 is expressed at cartilage-mesenchyme interfaces and impedes<br />

Wnt3a-mediated inhibition of chondrogenesis. J Cell Sci. 2009 Oct 15;122(Pt<br />

20):3627-37. Epub 2009 Sep.<br />

18


DEPARTMENT OF EXPERIMENTAL MEDICINE I<br />

(Hypertension-Induced Target Organ Damage)<br />

Head: Dominik N. Müller, Dr. rer. nat.<br />

Professor of Experimental Medicine<br />

Address: Department of Experimental Medicine I<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 29100<br />

Fax: + 49 (9131) 85 26341<br />

E-mail: dmueller@molmed.uni-erlangen.de<br />

Homepage: http://www.molmed.uni-erlangen.de/<br />

Head<br />

Dominik N. Müller, Prof. Dr. rer. nat.<br />

Professor of Experimental Medicine<br />

Dr. rer. nat. Katrina Binger<br />

Dr. rer. nat. Kristina Tanneberger<br />

Dr. rer. nat. Agnes Schröder<br />

Eva Bauer<br />

Supporting Staff<br />

Irena Bielmeier, Secretary<br />

Postdoctoral Fellows<br />

Technicians<br />

19<br />

Dr. med. Anke Dahlmann<br />

Dr. med. Wolfgang Freisinger<br />

Britta Schlund


<strong>Research</strong><br />

Hypertension is the primary risk factor for cardiovascular disease, the major<br />

cause of death worldwide. Our group’s major research interests are the reninangiotensin<br />

system (RAS), the immune system and how both systems cause<br />

hypertension-induced target organ damage. In a translational approach, the Müller<br />

lab focuses on the vessels, hearts, kidneys with our recent work extending these<br />

concepts to analyze how epigenetic factors, such as high salt, influence immune cells<br />

and target organ damage.<br />

Projects<br />

The immune system, salt and hypertension-induced target organ damage<br />

Our research is aimed at better understanding the role of innate and adaptive<br />

immunity in hypertension-induced organ damage. T cells, macrophages, and<br />

dendritic cells all harbor the AT1 receptor. Ang II stimulates T cell proliferation and<br />

dendritic cell migration. We showed that mice lacking the transcription factor Id2, a<br />

pivotal factor for Langerhans dendritic cells, are resistant to Ang II-induced<br />

hypertension and sequelae. We also observed that regulatory T cells modulate Ang<br />

II-induced cardiac damage to a point of therapeutic utility. Since our data suggested<br />

an interaction of the renin-angiotensin system, immune system and target organ<br />

damage (Figure), we are investigating the role of angiotensin II in autoimmunity.<br />

In collaboration with Ralf Linker (Univ. Erlangen), we investigated whether blockade<br />

of the renin-angiotensin system improves non-cardiovascular autoimmunity. Our<br />

results suggest that by<br />

blocking the aspartyl<br />

protease renin, ACE<br />

and the AT1 receptor,<br />

autoimmune<br />

encephalitis in rodents<br />

can be effectively<br />

inhibited.<br />

We are now<br />

extending our<br />

immunological studies<br />

to the investigation of<br />

inflammatory<br />

activation in the<br />

interstitium by<br />

hyperosmolarity through sodium. This line of investigation was initiated with Jens<br />

Titze (Univ. Erlangen/ Vanderbilt Univ.), with whom we have an intensive<br />

collaboration. With dietary sodium excess, sodium accumulates in the skin and<br />

activates the osmotic stress gene tonicity-responsive enhancer binding protein<br />

(TonEBP/NFAT5) in macrophages. TonEBP activity in macrophages results in<br />

secretion of VEGF-C promoting the clearance of hypertonic fluid from the interstitium.<br />

This circuit is critically dependent on macrophages and their ability to ward off<br />

hypertension in case of excess sodium supply. We have initiated a program to<br />

determine how hypertonicity induced by deranged sodium chloride storage affects<br />

the differentiation of T cells and macrophages.<br />

20


Cardiovascular and non-cardiovascular functions of the (pro)renin receptor<br />

The (pro)renin receptor (PRR) is a relatively newly discovered member of the<br />

renin-angiotensin system (RAS). Initially, PRR was believed to directly contribute to<br />

cardiovascular disease by activating the RAS and several signaling cascades.<br />

However, recent publications in Science have shown that PRR plays an essential<br />

and non-RAS related role in the activation of Wnt signal transduction and cellular<br />

development. In light of this, our understanding of the role of the prorenin receptor<br />

(PRR) in physiology and pathology has changed dramatically and we are now<br />

focused on determining these non-RAS functions of PRR.<br />

In collaboration with Michael Bader (MDC), we have determined that complete<br />

knockout of PRR in mouse embryonic stem cells, fails to generate chimeras when<br />

injected into blastocysts, indicating an important role for this protein in cellular<br />

development. We have subsequently initiated several PRR tissue-specific conditional<br />

knockout models. Generation of podocyte-specific PRR knockout mice (cKO)<br />

resulted in the death of the animals ~2-3 weeks after birth. Within 14 days, these<br />

cKO animals developed nephrotic syndrome and albuminuria, due to podocyte foot<br />

process fusion (Figure) and cytoskeletal changes.<br />

Figure legend EM shows normal epithelial cells and foot processes in control animals (left). cKO mice<br />

(right) developed foot process fusion (c=capillary, p=podocyte).<br />

Our in vivo and in vitro findings indicated a functional block in autophagosomelysosome<br />

fusion and overload of the proteasome protein degradation machinery.<br />

These results suggest that the PRR is essential for podocyte function and survival by<br />

maintaining autophagy and protein turnover machinery. We are now investigating the<br />

effect of PRR deletion in other cell types, namely T cells, pancreatic b-cells and<br />

renin-producing cells.<br />

We are also focused on understanding the molecular mechanism by which<br />

PRR is important for cellular homeostasis and development. The PRR was initially<br />

proposed to have no homology to other proteins, however it is now apparent that the<br />

nucleotide sequence of PRR is identical to that of ATP6AP2; a gene identified as an<br />

21


accessory protein of the H+ vacuolar-ATPase (V-ATPase). The V-ATPase is<br />

responsible for establishing and maintaining intracellular pH gradients along the<br />

secretory and endocytic pathways in all cell types. Acidification of vacuoles along<br />

these pathways is essential for many cellular functions including the processing and<br />

secretion of proteins, receptor endocytosis and recycling, and membrane fusion<br />

events, which are essential for processes such as autophagy. We are undertaking<br />

experiments to determine the protein-protein interactions important for PRR to<br />

maintain this function, and its role in Wnt signal activation.<br />

Interestingly, the PRR also exists as a soluble receptor (sPRR), and it has<br />

been hypothesized that this sPRR may have an important biological role. We have<br />

developed a sensitive ELISA to quantitate sPRR in biological fluids, and we are<br />

currently investigating if sPRR levels may be an important biomarker for<br />

cardiovascular disease and cancer.<br />

CV<br />

Dominik N. Müller studied pharmacy at the Free University of Berlin (1991-1992).<br />

He completed his Ph.D. thesis in 1996 and achieved faculty rank (Habilitation) for<br />

Experimental Medicine in 2004. After his postdoctoral work at Franz-Volhard Clinic<br />

and Max-Delbrück-Center Berlin (1996-2004), he was appointed a Delbrück Fellow at<br />

the MDC (2004-2010). In 2011, Dominik N. Müller was recruited to a W3 (full)<br />

professorship for Experimental Medicine at the University of Erlangen.<br />

Publications (2010-2012)<br />

Original Articles<br />

Five most important publications since 2007<br />

Riediger, F., Quack, I., Qadri, F., Hartleben, B., Park, J.K., Potthoff, S.A., Sohn, D.,<br />

Sihn. G., Rousselle, A., Fokuhl, V., Maschke, U., Pur<strong>für</strong>st, B., Schneider, W., Rump,<br />

L.C., Luft, F.C., Dechend, R., Bader, M., Huber, T.B., Nguyen, G. and Müller, D.N.<br />

(2011) Prorenin receptor is essential for podocyte autophagy and survival. J Am Soc<br />

Nephrol. 22, 2193-2202.<br />

Stegbauer, J., Lee, D.H., Seubert, S., Ellrichmann, G., Manzel, A., Kvakan, H.,<br />

Müller, D.N., Gaupp, S., Rump, L.C., Gold, R.,Linker, R.A. (2009). Role of the reninangiotensin<br />

system in autoimmune inflammation of the central nervous system. Proc<br />

Natl Acad Sci U S A 106, 14942-14947.<br />

Machnik, A., Neuhofer, W., Jantsch, J., Dahlmann, A., Tammela, T., Machura, K.,<br />

Park, J.-K., Beck, F.-X., Müller, D.N., Derer, W., Goss, J., Ziomber, A., Dietsch, P.,<br />

Wagner, H., van Rooijen, N., Kurtz, A., Hilgers, K.F., Alitalo, K., Eckardt, K.U., Luft,<br />

22


F.C., Kerjaschki, D., Titze, J. (2009) Macrophages regulate salt-dependent volume<br />

and blood pressure by a VEGF-C dependent buffering mechanism. Nat Med 15, 545-<br />

52.<br />

Kvakan, H., Kleinewietfeld, M., Qadri, F., Park, J.-K., Fischer, R., Schwarz, I., Rahn,<br />

H.-P., Plehm, R., Wellner, M., Elitok, S., Gratze, P.., Dechend, R, Luft, F.C., and<br />

Müller, D.N. (2009) Regulatory T cells ameliorate angiotensin II-induced cardiac<br />

damage. Circulation. 119, 2904-2912.<br />

Henke, N., Schmidt-Ullrich, R., Dechend, R., Park, J.-K., Qadri, F., Wellner, M., Obst,<br />

M., Gross, V., Dietz, R., Luft, F.C., Scheidereit, C., Müller, D.N. Vascular endothelial<br />

cell-specific NF-kB suppression attenuates hypertension-induced renal damage. Circ<br />

Res. 2007; 101:268-76.<br />

Publications since 2010<br />

2012<br />

Markó L, Kvakan H, Park J-K, Qadri F, Spallek B, Binger KJ, Bowman EP,<br />

Kleinewietfeld M, Fokuhl V, Dechend R, Muller DN. Interferon gamma (IFN-γ)<br />

signaling inhibition ameliorates Ang II-induced cardiac damage Hypertension 2012<br />

(in press)<br />

Kopp, C., Linz, P., Wachsmuth, L., Dahlmann, A., Horbach, T., Schofl, C., Renz, W.,<br />

Santoro, D., Niendorf, T., Muller, D.N., Neininger, M., Cavallaro, A., Eckardt, K.-U.,<br />

Schmieder, R.E., Luft, F.C., Uder, M., Titze, J. (2012). 23Na Magnetic Resonance<br />

Imaging of Tissue Sodium. Hypertension 59 167-172<br />

Finckenberg, P., Eriksson, O., Baumann, M., Merasto, S., Lalowski, M.M., Levijoki,<br />

J., Haasio, K., Kyto, V., Muller, D.N., Luft, F.C., Oresic., M, Mervaala, E. (2012).<br />

Caloric Restriction Ameliorates Angiotensin II-Induced Mitochondrial Remodeling and<br />

Cardiac Hypertrophy. Hypertension 59:76-84.<br />

2011<br />

Riediger, F., Quack, I., Qadri, F., Hartleben, B., Park, J.K., Potthoff, S.A., Sohn, D.,<br />

Sihn, G., Rousselle, A., Fokuhl, V., Maschke, U., Pur<strong>für</strong>st, B., Schneider, W., Rump,<br />

L.C., Luft, F.C., Dechend, R., Bader, M., Huber, T.B., Nguyen, G., Müller D.N.<br />

(2011). Prorenin receptor is essential for podocyte autophagy and survival. J Am<br />

Soc Nephrol 22, 2193-2202.<br />

Park, J.B., Kim, B.K., Kwon, Y.W., Muller, D.N., Lee, H.C., Youn, S.W., Choi, Y.E.,<br />

Lee, S.W., Yang, H.M., Cho, H.J., Park, K.W., Kim, H.S. (2011). Peroxisome<br />

proliferator-activated receptor-gamma agonists suppress tissue factor overexpression<br />

in rat balloon injury model with Paclitaxel infusion. PLoS One 6, e28327.<br />

Batenburg, W.W., Lu, X., Leijten, F., Maschke, U., Muller, D.N., and Danser, A.H.<br />

(2011). Renin- and prorenin-induced effects in rat vascular smooth muscle cells<br />

overexpressing the human (pro)renin receptor: does (pro)renin-(pro)renin receptor<br />

interaction actually occur? Hypertension 58, 1111-1119.<br />

23


Searle, J., Mockel, M., Gwosc, S., Datwyler, S.A., Qadri, F., Albert, G.I., Holert, F.,<br />

Isbruch, A., Klug, L., Muller, D.N., Dechend, R., Muller, R., Vollert, J.O., Slagman, A.,<br />

Mueller, C., Herse, F. (2011). Heparin strongly induces soluble fms-like tyrosine<br />

kinase 1 release in vivo and in vitro--brief report. Arterioscler Thromb Vasc Biol 31,<br />

2972-2974.<br />

Falck, J.R., Wallukat, G., Puli, N., Goli, M., Arnold, C., Konkel, A., Rothe, M., Fischer,<br />

R., Muller, D.N., and Schunck, W.H. (2011). 17(R),18(S)-epoxyeicosatetraenoic acid,<br />

a potent eicosapentaenoic acid (EPA) derived regulator of cardiomyocyte<br />

contraction: structure-activity relationships and stable analogues. J Med Chem 54,<br />

4109-4118.<br />

Wenzel, K., Rajakumar, A., Haase, H., Geusens, N., Hubner, N., Schulz, H., Brewer,<br />

J., Roberts, L., Hubel, C.A., Herse, F., Hering, L., Qadri, F., Lindschau, C., Wallukat,<br />

G., Pijnenborg, R., Heidecke, H., Riemekasten, G., Luft, F.C., Müller, D.N., Lamarca,<br />

B., Dechend, R. (2011). Angiotensin II type 1 receptor antibodies and increased<br />

angiotensin II sensitivity in pregnant rats. Hypertension 58, 77-84.<br />

Herse, F., Fain, J.N., Janke, J., Engeli, S., Kuhn, C., Frey, N., Weich, H.A.,<br />

Bergmann, A., Kappert, K., Karumanchi, S.A., Luft, F.C., Müller, D.N., Staff, A.C.,<br />

Dechend, R. (2011). Adipose tissue-derived soluble fms-like tyrosine kinase 1 is an<br />

obesity-relevant endogenous paracrine adipokine. Hypertension 58, 37-42.<br />

Riemekasten, G., Philippe, A., Nather, M., Slowinski, T., Muller, D.N., Heidecke, H.,<br />

Matucci-Cerinic, M., Czirjak, L., Lukitsch, I., Becker, M., Kill, A., van Laar, J.M., Catar,<br />

R., Luft, F.C., Burmester, G.R., Hegner, B., Dragun, D. (2011). Involvement of<br />

functional autoantibodies against vascular receptors in systemic sclerosis. Ann<br />

Rheum Dis 70, 530-536.<br />

Hoff, U., Lukitsch, I., Chaykovska, L., Ladwig, M., Arnold, C., Manthati, V.L., Fuller,<br />

T.F., Schneider, W., Gollasch, M., Müller, D.N., Flemming, B., Seeliger, E., Luft,<br />

F.C., Falck, J.R., Dragun, D., Schunck, W.H. (2011). Inhibition of 20-HETE synthesis<br />

and action protects the kidney from ischemia/reperfusion injury. Kidney Int 79, 57-<br />

65.<br />

2010<br />

Schmerbach, K., Pfab, T., Zhao, Y., Culman, J., Mueller, S., Villringer, A., Müller,<br />

D.N., Hocher, B., Unger, T., and Thoene-Reineke, C. (2010). Effects of aliskiren on<br />

stroke in rats expressing human renin and angiotensinogen genes. PLoS One 5,<br />

e15052.<br />

Biala, A., Martonen, E., Kaheinen, P., Levijoki, J., Finckenberg, P., Merasto, S.,<br />

Louhelainen, M., Müller, D.N., Luft, F.C., and Mervaala, E. (2010a). Levosimendan<br />

improves cardiac function and survival in rats with angiotensin II-induced<br />

hypertensive heart failure. Hypertens Res 33, 1004-1011.<br />

Arnold, C., Markovic, M., Blossey, K., Wallukat, G., Fischer, R., Dechend, R., Konkel,<br />

A., von Schacky, C., Luft, F.C., Müller, D.N., Rothe, M., Schunck, W.H. (2010).<br />

24


Arachidonic acid-metabolizing cytochrome P450 enzymes are targets of {omega}-3<br />

fatty acids. J Biol Chem 285, 32720-32733.<br />

Verlohren, S., Geusens, N., Morton, J., Verhaegen, I., Hering, L., Herse, F.,<br />

Dudenhausen, J.W., Müller, D.N., Luft, F.C., Cartwright, J.E., Davidge, S.T.,<br />

Pijnenborg, R., Dechend, R. (2010). Inhibition of trophoblast-induced spiral artery<br />

remodeling reduces placental perfusion in rat pregnancy. Hypertension 56, 304-310.<br />

Hering, L., Herse, F., Geusens, N., Verlohren, S., Wenzel, K., Staff, A.C., Brosnihan,<br />

K.B., Huppertz, B., Luft, F.C., Müller, D.N., Pijnenborg, R., Cartwright, J., Dechend,<br />

R. (2010). Effects of circulating and local uteroplacental angiotensin II in rat<br />

pregnancy. Hypertension 56, 311-318.<br />

Wenzel, K., Wallukat, G., Qadri, F., Hubner, N., Schulz, H., Hummel, O., Herse, F.,<br />

Heuser, A., Fischer, R., Heidecke, H., Luft F.C., Müller, D.N., Dietz, R., Dechend, R.<br />

(2010). Alpha1A-adrenergic receptor-directed autoimmunity induces left ventricular<br />

damage and diastolic dysfunction in rats. PLoS One 5, e9409.<br />

Machnik, A., Dahlmann, A., Kopp, C., Goss, J., Wagner, H., van Rooijen, N., Eckardt,<br />

K.U., Müller, D.N., Park, J.K., Luft, F.C., Kerjaschki, D., Titze, J. (2010).<br />

Mononuclear phagocyte system depletion blocks interstitial tonicity-responsive<br />

enhancer binding protein/vascular endothelial growth factor C expression and<br />

induces salt-sensitive hypertension in rats. Hypertension 55, 755-761.<br />

Mervaala, E., Biala, A., Merasto, S., Lempiainen, J., Mattila, I., Martonen, E.,<br />

Eriksson, O., Louhelainen, M., Finckenberg, P., Kaheinen, P., Müller, D.N., Luft,<br />

F.C., Lapatto, R., Oresic, M. (2010). Metabolomics in angiotensin II-induced cardiac<br />

hypertrophy. Hypertension 55, 508-515.<br />

25


DEPARTMENT OF EXPERIMENTAL MEDICINE II<br />

(Molecular Tumour <strong>Research</strong>)<br />

Head: Jürgen Behrens, Dr. rer. nat.<br />

Professor of Experimental Medicine<br />

Address: Department of Experimental Medicine II<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 29109<br />

Fax: + 49 (9131) 85 29111<br />

E-mail: jbehrens@molmed.uni-erlangen.de<br />

Homepage: www.em2.molmed.uni-erlangen.de<br />

Martin Sachs, Dr. rer. nat.<br />

Jean Schneikert, Dr. rer. nat.<br />

Head<br />

Jürgen Behrens, Prof. Dr. rer. nat.<br />

Professor of Experimental Medicine<br />

Supporting Staff<br />

Angela Döbler, Secretary<br />

Postdoctoral Fellows<br />

26<br />

Michel Hadjihannas, Dr. rer. nat.<br />

Ingrid Wacker, Dr. rer. nat.


Katharina Brauburger<br />

° PhD received<br />

° PhD received<br />

Doctoral Students (Biology)<br />

Sandra Mattauch°<br />

Astrid Pfister°<br />

Doctoral Students (Molecular Medicine)<br />

Martina Brückner<br />

Gabriele Daum<br />

* part of the time reported<br />

Dominic Bernkopf<br />

Kristina Tanneberger°<br />

Technicians<br />

27<br />

Shree Harsha Vijaya°<br />

Birgit Saffer<br />

Sabine Lukat*


<strong>Research</strong><br />

Our group analyses molecular mechanisms that control tumour development and<br />

progression, focussing on oncogenic Wnt and Activin signalling.<br />

Functional analysis of Wnt signaling components<br />

Wnts are a family of secreted glycoproteins that bind to frizzled seventransmembrane<br />

span receptors and LRP coreceptors. Intracellularly, the Wnt<br />

signaling cascade blocks degradation of β-catenin in proteasomes and thereby leads<br />

to accumulation of β-catenin in the cytoplasm. β-Catenin then enters the nucleus and<br />

activates the expression of Wnt-specific target genes by interacting with LEF-1/TCF<br />

transcription factors. Conductin (also named Axin2), and its relative axin act as<br />

scaffolding components that simultaneously bind to β-catenin, the serine/threonine<br />

kinases GSK3β and CK1ε, and APC. In this complex GSK3β and CK1ε<br />

phosphorylate β-catenin at serine and threonine residues, which leads to<br />

ubiquitination and subsequent proteasomal degradation of β-catenin. It is suggested<br />

that axin proteins block Wnt signaling by assembling the essential components of the<br />

β-catenin degradation pathway. In colorectal tumours mutations of the tumour<br />

suppressor APC (adenomatous polyposis coli) lead to stabilization of β-catenin and<br />

constitutive signaling to the nucleus independent of Wnts. APC is a co-factor<br />

indispensable for axin-mediated degradation of β-catenin. APC binds to β-catenin<br />

and axin/conductin via multiple interaction domains thereby supporting β-catenin<br />

degradation. Mutations of β-catenin have been observed that also lead to<br />

stabilization of β-catenin. Thus, aberrant activation of the Wnt signaling pathway is<br />

considered a major oncogenic mechanism for many tumour types.<br />

In the past years we have identified and functionally characterized several key<br />

components of the pathway using our highly efficient yeast two hybrid screening<br />

system. Among these are Lef-1 and conductin/axin2 as interaction partners of βcatenin,<br />

and more recently the Amer family of APC interactors, as well as EB1 as an<br />

interaction partner of Amer2.<br />

Role of conductin/axin2 in the cell cycle<br />

Michel Hadjihannas, Martina Brückner<br />

We found that conductin levels are regulated during the cell cycle with lowest levels<br />

present during the G1/S phase and highest during G2/M. Following exit from mitosis<br />

conductin expression levels decline in parallel with those of mitotic regulators, such<br />

as cyclin B1. In line, Wnt/β-catenin target genes are low at G2/M and high at G1/S,<br />

28


and β-catenin phosphorylation oscillates during the cell cycle in a conductindependent<br />

manner. Conductin is degraded by the anaphase-promoting<br />

complex/cyclosome cofactor CDC20. Knockdown of CDC20 blocks Wnt signalling<br />

through conductin. CDC20-resistant conductin inhibits Wnt signalling and attenuates<br />

colony formation of colorectal cancer cells. We propose that CDC20-mediated<br />

degradation of conductin regulates Wnt/β-catenin signalling for maximal activity<br />

during G1/S (Hadjihannas et al., 2012).<br />

We also found that conductin localizes at the centrosomes by binding to the centrioleassociated<br />

component C-Nap1. Knockout or knockdown of conductin leads to<br />

premature centrosome separation which is abolished by knockdown of β-catenin.<br />

Conductin promotes phosphorylation of the amino-terminal serine (Ser 33/37) and<br />

threonine (Thr 41) residues of centrosome-associated β-catenin. β-Catenin mutated<br />

at these residues causes centrosomal separation, whereas a phospho-mimicking<br />

mutant of β-catenin does not. Treatment with Wnts and inhibition of glycogen<br />

synthase kinase 3 block β-catenin phosphorylation and induce centrosomal splitting.<br />

These data indicate that Wnt signalling and conductin regulate centrosomal cohesion<br />

by altering the phosphorylation status of β-catenin at the centrosomes (Hadjihannas<br />

et al., 2010).<br />

Functional dissection of APC mutants truncated in colorectal cancer<br />

Jean Schneickert, Eva Kohler, Shree Harsha Vijaya Chandra<br />

The tumour suppressor Adenomatous Polyposis Coli (APC) is truncated in most<br />

colon cancers, but is not completely lost. It is not clear why colon cancer cells retain<br />

the truncated APC fragment. The mutations affecting both APC alleles are<br />

interdependent, the position of the first APC mutation determining where the second<br />

hit will occur. This results in a complex pattern of mutation distribution in the APC<br />

sequence that translates into the stabilization of β−catenin that in turn feeds the<br />

affected cells with a permanent mitogenic signal. We found a new APC domain, the<br />

β−catenin inhibitory domain (CID) of APC located between the second and third 20<br />

amino acid repeats and therefore present in many truncated APC products found in<br />

human tumours. In truncated APC, the CID is absolutely necessary to down-regulate<br />

the transcriptional activity and the level of β−catenin, even when an axin/conductin<br />

binding site is present (Kohler et al., 2009).<br />

The four 15 amino acid repeats (15R) and the seven 20 amino acid repeats (20R) of<br />

APC are beta-catenin-binding sites, but their role in beta-catenin degradation has<br />

remained unclear. We showed that binding of β-catenin to the 15R of APC is<br />

necessary and sufficient to target β-catenin for degradation. The first 15R displays<br />

the highest affinity for beta-catenin in the 15R-20R module. The analysis of the<br />

distribution of truncating mutations along the APC sequence in colorectal tumours<br />

from FAP patients revealed that the first 15R is one target of the positive selection of<br />

mutations that lead to tumour development (Kohler et al., 2010).<br />

29


We found that the transcriptional repressor C-terminal binding protein (CtBP)<br />

promotes the oligomerization of truncated APC through binding to the 15 amino acid<br />

repeats of truncated APC. CtBP can bind to either first, third or fourth 15 amino acid<br />

repeats, but not to the second. CtBP-mediated oligomerization requires both<br />

dimerization domains of truncated APC as well as CtBP dimerization. This suggests<br />

that the sensitivity of truncated APC to oligomerization by CtBP constitutes an<br />

essential facet of tumour development (Schneikert et al., 2011).<br />

RNA interference was used to down-regulate truncated APC in several colorectal<br />

cancer cell lines expressing truncated APCs of different lengths, thereby performing<br />

an analysis covering most of the mutation cluster region. The consequences on<br />

proliferation in vitro, tumour formation in vivo and the level and transcriptional activity<br />

of β-catenin were investigated. Down-regulation of truncated APC results in an<br />

inhibition of tumour cell population expansion in vitro in 6 cell lines out of 6 and<br />

inhibition of tumour outgrowth in vivo as analysed in one of these cell lines, HT29.<br />

Down-regulation of truncated APC is accompanied by an up-regulation of the<br />

transcriptional activity of β-catenin and in most cases β-catenin levels, indicating that<br />

truncated APC can still modulate wnt signalling through controlling the level of βcatenin.<br />

Thus, truncated APC is an essential component of colorectal cancer cells,<br />

required for cell proliferation, possibly by adjusting β-catenin signalling to the "just<br />

right" level (Vijaya Chandra et al., 2012).<br />

The AMER family of Wnt pathway regulators<br />

Kristina Tanneberger, Astrid Alzner, Katharina Brauburger, Martin Sachs<br />

Amer 1, Amer2 and Amer3 constitute a new family of Wnt pathway regulators that<br />

are characterized by the presence of conserved APC binding domains which interact<br />

with the armadillo repeats of APC. We have initially identified Amer1 in a yeast two<br />

hybrid screen for interaction partners of APC and have cloned the other family<br />

members as open reading frames from database information.<br />

Amer1<br />

We previously identified Amer1 (APC membrane recruitment 1) as a novel<br />

membrane-associated protein that interacts with APC and recruits it away from<br />

microtubule ends to the plasma membrane. The N-terminus of Amer1 contains two<br />

distinct phosphatidylinositol(4,5)-bisphosphate [PtdIns(4,5)P(2)]-binding domains<br />

composed of clusters of lysines, which mediate its localization to the plasma<br />

membrane.<br />

Amer1 is identical to the tumour suppressor WTX, which is mutated in a proportion of<br />

Wilms tumours. Amer1/WTX acts as an inhibitor of Wnt signaling by inducing βcatenin<br />

degradation. Amer1 directly interacts with the armadillo repeats of β-catenin<br />

30


via a domain consisting of repeated arginine-glutamic acid-alanine (REA) motifs, and<br />

assembles the β-catenin destruction complex at the plasma membrane by recruiting<br />

β-catenin, APC, and Axin/Conductin. Deletion or specific mutations of the membrane<br />

binding domain of Amer1 abolish its membrane localization and abrogate negative<br />

control of Wnt signaling. Knockdown of Amer1 leads to the activation of Wnt target<br />

genes, preferentially in dense compared with sparse cell cultures, suggesting that<br />

Amer1 function is regulated by cell contacts. Amer1 stabilizes Axin and counteracts<br />

Wnt-induced degradation of Axin, which requires membrane localization of Amer1.<br />

The data suggest that Amer1 exerts its negative regulatory role in Wnt signaling by<br />

acting as a scaffold protein for the β-catenin destruction complex and promoting<br />

stabilization of Axin at the plasma membrane (Tanneberger et al., 2011a).<br />

Phosphorylation of the Wnt receptor low-density lipoprotein receptor-related protein 6<br />

(LRP6) by glycogen synthase kinase 3β (GSK3β) and casein kinase 1γ (CK1γ) is a<br />

key step in Wnt/β-catenin signalling, which requires Wnt-induced formation of<br />

phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)). We found that Amer1 is<br />

essential for the activation of Wnt signalling at the LRP6 receptor level. Knockdown<br />

of Amer1 reduced Wnt-induced LRP6 phosphorylation, Axin translocation to the<br />

plasma membrane and formation of LRP6 signalosomes. Overexpression of Amer1<br />

promotes LRP6 phosphorylation, which requires interaction of Amer1 with<br />

PtdIns(4,5)P(2). Amer1 translocates to the plasma membrane in a PtdIns(4,5)P(2)dependent<br />

manner after Wnt treatment and is required for LRP6 phosphorylation<br />

stimulated by application of PtdIns(4,5)P(2). Amer1 binds CK1γ, recruits Axin and<br />

GSK3β to the plasma membrane and promotes complex formation between Axin and<br />

LRP6. We propose a mechanism for Wnt receptor activation by which generation of<br />

PtdIns(4,5)P(2) leads to recruitment of Amer1 to the plasma membrane, which acts<br />

as a scaffold protein to stimulate phosphorylation of LRP6.<br />

Altogether our data show that Amer1 has a dual functional role in Wnt signalling<br />

acting as an activator at the level of the Frizzled/LRP receptor complex and as an<br />

inhibitor by assembling the β-catenin destruction complex (Fig. 1). Of note, Amer1<br />

makes use of similar interaction partners, namely axin and GSK3 for both activities<br />

(Tanneberger et al., 2011b).<br />

31


Fig. 1 Dual functional role of Amer1 in Wnt signalling. Amer1 recruits the b-catenin<br />

destruction complex to the plasma membrane by binding to PIP2 which promotes b-catenin<br />

degradation and blocks Wnt signalling. When Wnts bind to Frizzled/LRP6 receptors more<br />

PIP2 is generated probably in vicinity to the receptors, Amer1 becomes recruited and<br />

stimulates phosphorylation of LRP6 receptors through interaction with GSK3 and CK1γ.<br />

Thus, Amer1 can act both as a repressor and an activator of the pathway.<br />

Amer2<br />

Amer2 (APC membrane recruitment 2; FAM123A) is a direct interaction partner of<br />

APC, related to Amer1/WTX. We showed that Amer2 recruits APC to the plasma<br />

membrane by binding to phosphatidylinositol 4,5-bisphosphate lipids via lysine-rich<br />

motifs and that APC links β-catenin and the destruction complex components axin<br />

and conductin to Amer2. Knockdown of Amer2 increased Wnt target gene expression<br />

and reporter activity in cell lines, and overexpression reduced reporter activity, which<br />

required membrane association of Amer2. In Xenopus embryos, Amer2 is expressed<br />

mainly in the dorsal neuroectoderm and neural tissues. Down-regulation of Amer2 by<br />

specific morpholino oligonucleotides altered neuroectodermal patterning, which could<br />

be rescued by expression of a dominant-negative mutant of Lef1 that interferes with<br />

β-catenin-dependent transcription. Our data characterize Amer2 for the first time as a<br />

negative regulator of Wnt signaling both in cell lines and in vivo and define Amer<br />

proteins as a novel family of Wnt pathway regulators.<br />

In yeast two hybrid screen using a C-terminal fragment of Amer2 as a bait we found<br />

that Amer2 is a direct interaction partner of EB1, which is a key factor in the<br />

32


organisation of the microtubule cytoskeleton. Of note, EB1 interacts with APC to<br />

stabilise microtubules. Amer2 binds to EB1 via specific S/TxIP motifs and recruits it to<br />

the plasma membrane. Coexpression of Amer2 and EB1 generates stabilised<br />

microtubules whereas knockdown of Amer2 leads to destabilisation of microtubules.<br />

Knockdown of Amer2, APC, or EB1 reduces cell migration, and morpholino-mediated<br />

downregulation of Xenopus Amer2 blocks convergent extension cell movements<br />

suggesting that the Amer2/EB1/APC complex regulates cell migration by altering<br />

microtubule stability (Pfister et al., JBC in revision). The Amer2 knock-out in mice has<br />

recently been generated in our lab but shows no obvious phenotypes, possibly due to<br />

compensation by Amer2. We are currently generating double-knockout animals of<br />

Amer1 and Amer2 for further characterization.<br />

Fig. 2 Scheme of Amer2 interactions with APC and EB1 in microtubule stabilization.<br />

Amer3<br />

Amer3 does not interact with the plasma membrane and is mainly located in the<br />

cytoplasm and occasionally in the cell nucleus. Moreover, in contrast to Amer1 and<br />

Amer2 this protein acts as an activator rather than inhibitor of Wnt signalling.<br />

Ongoing work aims at identifying the precise function of Amer3 and its interplay with<br />

the other Amers in the Wnt pathway.<br />

33


HIF target gene expression in renal cell carcinomas<br />

A key role for Activin B in cellular transformation after loss of the von Hippel-<br />

Lindau (VHL) tumour suppressor<br />

Ingrid Wacker, Martin Sachs,<br />

Clear cell renal cell carcinoma (clear cell RCC) is the most frequent tumour disease<br />

in the kidney. Most of these tumours contain mutations in the VHL tumour suppressor<br />

gene, which leads to stabilization of HIF1α and Hif2α, and causes increased<br />

transcription of HIF target genes. We found that VHL suppresses key features of cell<br />

transformation solely through downregulation of the HIF-dependent expression of<br />

Activin B, a member of the TGFβ superfamily. Activin B expression is repressed by<br />

restoration of VHL in VHL-deficient RCC cells and upregulated by hypoxia. RCC<br />

tumour samples show increased expression of Activin B as compared to normal<br />

kidney. Importantly, knockdown of Activin B reduces tumour growth of RCC cell lines<br />

in nude mice. Our data indicate that Activin B is a key mediator of VHL/HIF induced<br />

transformation in RCC (Wacker et al., 2009). We are currently searching for ways to<br />

block Activin B signalling using biological reagents in order to prevent RCC<br />

tumourigenesis.<br />

Liprinα4 is a novel HIF1α target gene<br />

Sandra Mattauch<br />

Liprin-α1 to liprin-α4 constitute a family of cytoplasmic proteins, which have been<br />

found in various multiprotein complexes. For liprin-α1 roles in synapse formation and<br />

cell spreading were described but other liprin family members are not well<br />

characterized. We found that liprin-α4 is upregulated in human clear cell renal cell<br />

carcinomas as compared to normal kidney tissue and that it is regulated the VHL/HIF<br />

system. The liprin-α4 gene promoter is directly activated by binding of the hypoxiainducible<br />

factor 1α (HIF-1α) to HRE consensus binding sites as shown by reporter<br />

assays and chromatin immunoprecipitations. RNAi mediated knockdown of liprin-α4<br />

leads to reduced E-cadherin and β-catenin levels at cell junctions and to dissociation<br />

of epithelial cell contacts. Our data describe for the first time liprin-α4 as a hypoxiainduced<br />

gene potentially involved in cell-cell adhesion (Mattauch et al., 2010).<br />

34


2012<br />

Publications (2009-2012)<br />

Original Articles<br />

Vijaya Chandra, S.H., Wacker, I., Appelt, U.K., Behrens, J., and Schneikert, J.<br />

(2012). A common role for various human truncated adenomatous polyposis<br />

coli isoforms in the control of Beta-catenin activity and cell proliferation. PLoS<br />

One 7, e34479.<br />

Pfister, A.S., Tanneberger, K., Schambony, A., and Behrens, J. (2012). Amer2<br />

protein is a novel negative regulator of Wnt/beta-catenin signaling involved in<br />

neuroectodermal patterning. J Biol Chem 287, 1734-1741.<br />

Hadjihannas, M.V., Bernkopf, D.B., Bruckner, M., and Behrens, J. (2012). Cell cycle<br />

control of Wnt/beta-catenin signalling by conductin/axin2 through CDC20.<br />

EMBO Rep 13, 347-354.<br />

2011<br />

Tanneberger, K., Pfister, A.S., Kriz, V., Bryja, V., Schambony, A., and Behrens, J.<br />

(2011a). Structural and Functional Characterization of the Wnt Inhibitor APC<br />

Membrane Recruitment 1 (Amer1). J Biol Chem 286, 19204-19214.<br />

Tanneberger, K., Pfister, A.S., Brauburger, K., Schneikert, J., Hadjihannas, M.V.,<br />

Kriz, V., Schulte, G., Bryja, V., and Behrens, J. (2011b). Amer1/WTX couples<br />

Wnt-induced formation of PtdIns(4,5)P2 to LRP6 phosphorylation. EMBO J 30,<br />

1433-1443.<br />

Schneikert, J., Brauburger, K., and Behrens, J. (2011). APC mutations in colorectal<br />

tumours from FAP patients are selected for CtBP-mediated oligomerization of<br />

truncated APC. Hum Mol Genet 20, 3554-3564.<br />

2010<br />

Schietke, R., Warnecke, C., Wacker, I., Schodel, J., Mole, D.R., Campean, V.,<br />

Amann, K., Goppelt-Struebe, M., Behrens, J., Eckardt, K.U., et al. (2010). The<br />

lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress Ecadherin<br />

in hypoxia: insights into cellular transformation processes mediated<br />

by HIF-1. J Biol Chem 285, 6658-6669.<br />

Mattauch, S., Sachs, M., and Behrens, J. (2010). Liprin-alpha4 is a new hypoxiainducible<br />

target gene required for maintenance of cell-cell contacts. Exp Cell<br />

Res 316, 2883-2892.<br />

Kohler, E.M., Brauburger, K., Behrens, J., and Schneikert, J. (2010). Contribution of<br />

the 15 amino acid repeats of truncated APC to beta-catenin degradation and<br />

selection of APC mutations in colorectal tumours from FAP patients.<br />

Oncogene 29, 1663-1671.<br />

Hadjihannas, M.V., Bruckner, M., and Behrens, J. (2010). Conductin/axin2 and Wnt<br />

signalling regulates centrosome cohesion. EMBO Rep 11, 317-324.<br />

35


2009<br />

Wacker, I., Sachs, M., Knaup, K., Wiesener, M., Weiske, J., Huber, O., Akcetin, Z.,<br />

and Behrens, J. (2009). Key role for activin B in cellular transformation after<br />

loss of the von Hippel-Lindau tumour suppressor. Mol Cell Biol 29, 1707-1718.<br />

Rotte, A., Bhandaru, M., Foller, M., Biswas, R., Mack, A.F., Friedrich, B., Rexhepaj,<br />

R., Nasir, O., Ackermann, T.F., Boini, K.M., et al. (2009). APC Sensitive<br />

Gastric Acid Secretion. Cell Physiol Biochem 23, 133-142.<br />

Nasir, O., Wang, K., Foller, M., Gu, S., Bhandaru, M., Ackermann, T.F., Boini, K.M.,<br />

Mack, A., Klingel, K., Amato, R., et al. (2009). Relative resistance of SGK1<br />

knockout mice against chemical carcinogenesis. IUBMB Life 61, 768-776.<br />

Kohler, E.M., Chandra, S.H., Behrens, J., and Schneikert, J. (2009). Beta-catenin<br />

degradation mediated by the CID domain of APC provides a model for the<br />

selection of APC mutations in colorectal, desmoid and duodenal tumours.<br />

Hum Mol Genet 18, 213-226.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Astrid Pfister<br />

Klonierung und funktionelle Charakterisierung von AMER2<br />

Katharina Brauburger<br />

<strong>Molekulare</strong> Funktionsanalyse von APC<br />

Sandra Mattauch<br />

Identifizierung und Charakterisierung von HIF-Zielgenen im Nierenzellkarzinom<br />

Shree Harsha Vijaya Chandra<br />

Blockade des Wachstums von Dickdarmkrebs durch RNA Interferenz<br />

Doctoral theses (Molecular Medicine)<br />

Dominic Bernkopf<br />

<strong>Molekulare</strong>r und funktioneller Vergleich von Axin und Conductin/Axin2<br />

Kristina Tanneberger<br />

<strong>Molekulare</strong> Charakterisierung des Tumoursuppressors AMER1/WTX<br />

36


DIVISION OF MOLECULAR IMMUNOLOGY<br />

(Department of Internal Medicine III)<br />

Head: Hans Martin Jäck, Dr. rer. nat.<br />

Professor of Immunology<br />

Address: Division of Molecular Immunology<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 35912<br />

Fax: + 49 (9131) 85 39343<br />

E-mail: hjaeck@molmed.uni-erlangen.de<br />

Homepage: www.molim.uni-erlangen.de<br />

Dirk Mielenz, Dr. rer. nat.<br />

Jürgen Wittmann, Dr. rer. nat.<br />

Wolfgang Schuh, Dr. rer. nat.<br />

Head<br />

Hans-Martin Jäck, Prof. Dr. rer. nat.<br />

Professor of Immunology<br />

Supporting Staff<br />

Elisabeth Lang, Secretary<br />

Postdoctoral Fellows<br />

37<br />

Barbara Fürnrohr, Dr. rer. nat.<br />

Rebecca Winkelmann, Dr. rer. nat.<br />

Martina Porstner, Dr. rer. nat.


Andreas Brandl*<br />

Sebastian Dütting*<br />

Sandra Schreiber*<br />

* PhD received<br />

Doctoral Students (Biology)<br />

Pavitra Purohit<br />

Julia Schmid<br />

Sebastian Brachs<br />

Patrick Daum<br />

Carmen Kroczek*<br />

Stefanie Thiele*<br />

Anke Lang*<br />

Doctoral Students (Molecular Medicine & Medicine)<br />

Martina Porstner*<br />

Rebecca Winkelmann*<br />

* PhD received<br />

Edith Roth<br />

Christiane Lang<br />

Heike Danzer<br />

Sandra Hagen<br />

Technicians<br />

Manuela Hauke<br />

Lena Sandrock<br />

Katrin Weiß<br />

Professor Emeritus<br />

Prof. Dr. med. Dr.<br />

mult. h.c. Joachim R.<br />

Kalden<br />

38<br />

Alexandra Klej<br />

Uwe Appelt


<strong>Research</strong><br />

Our research concentrates on molecular mechanisms that guide the development of<br />

antibody-producing B cells and control the adaptive humoral immunity in healthy<br />

organisms. In addition, we are interested in strategies to target pathologic plasma<br />

cells in autoimmune disease and in patients with multiple myeloma.<br />

Nonsense-mediated mRNA decay in lymphocytes<br />

Jürgen Wittmann, Carmen Baumgarten, Manuela Hauke, Heike Danzer and Hans-<br />

Martin Jäck<br />

Identification of novel factors involved in nonsense-mediated mRNA decay in<br />

higher eukaryotes<br />

Nonsense-mediated mRNA decay (NMD) is an essential eukaryotic pathway that<br />

recognizes and degrades erroneous mRNAs that contain a premature termination<br />

codon (nonsense mRNAs). By doing so, a negative influence on physiologic<br />

functions within a cell by faulty mRNAs is prevented, and error-free gene expression<br />

is guaranteed. Three evolutionary conserved proteins, UPF1, UPF2 and UPF3, play<br />

important roles in NMD. In particular, UPF2 fulfils a central role as adaptor protein,<br />

because it interacts with UPF1 and UPF3. However, not much is known about the<br />

biochemical role of UPF2 and its precise function in the different effector arms of<br />

NMD. We are therefore planning to identify interaction partners of key NMD factors<br />

by immunoprecipitation followed by mass spectrometric analysis to shed some light<br />

on mechanistic details of UPF function during NMD.<br />

Collaboration with Henning Urlaub (Göttingen)<br />

Function of the nonsense-mediated mRNA decay pathway during murine<br />

lymphocyte development<br />

As a result of the programmed rearrangements that generate the diverse<br />

immunoglobulin repertoire, nonsense codons are frequently generated in B- and Tcell<br />

receptor genes. Therefore, during B- and T-cell-development, a large number of<br />

lymphoid precursors with non-functional mRNAs are produced, which have to be<br />

detected and degraded early by the NMD pathway to avoid the production of<br />

deleterious products. To start addressing the role of NMD during lymphocyte<br />

development, we have generated two different conditional knock-out mice for one of<br />

the key factors of NMD. Although NMD is predicted to be important during<br />

embryogenesis, our conditional knock-out strategy allows us to study the role of NMD<br />

not only in lymphocyte development, but also in disease prevention in pre-selected<br />

tissues in the adult. Therefore, we will address presently unanswered fundamental<br />

questions about the role of NMD in B cell development and the basic NMD<br />

mechanism.<br />

Collaboration with Bo Porse (Copenhagen)<br />

39


microRNAs in lymphocytes in health and disease<br />

Jürgen Wittmann, Markus Zwick, Ina Wirries, Sandra Schreiber, Patrick Daum,<br />

Andreas Brandl, Martina Porstner, Manuela Hauke, Heike Danzer and Hans-Martin<br />

Jäck<br />

Role of microRNAs in B cell development in health and disease<br />

MicroRNAs (miRNAs) are a class of small endogenous non-coding RNAs that<br />

regulate gene expression at the post-transcriptional level by targeting partially<br />

sequence-complementary mRNAs. To investigate the role of miRNAs in B cell<br />

development, we used deep sequencing analysis of sorted primary B cells of several<br />

developmental stages to profile their miRNome. These studies allowed us to<br />

determine a set of marker miRNAs characteristic of the respective developmental<br />

stage. In addition, stage-specific novel miRNAs and other small RNA classes were<br />

discovered, opening up new research avenues for elucidating the fine-tuning of gene<br />

expression in murine B cell development.<br />

To expand the known regulatory function of miRNAs in early hematopoiesis and to<br />

test the hypothesis that miRNAs play a role in the pre-B cell receptor (pre-BCR)mediated<br />

control of early B cell development, we used a tetracycline-inducible µ<br />

heavy chain mouse model to identify differently expressed miRNAs in pro- and pre-B<br />

cells by miRNA microarray- and deep sequencing analysis. Several miRNAs were<br />

found to be up- or downregulated after µ heavy chain induction. In silico analysis<br />

identified several potential mRNA targets of these miRNAs that are involved in early<br />

B cell development, BCR signal transduction, opening of the Ig light chain locus as<br />

well as proliferation and cancer development. Functional assays with overexpressed<br />

miRNAs and reporter gene assays revealed that they indeed regulate key processes<br />

in early B cell development.<br />

Role of microRNAs in rheumatic diseases<br />

Rheumatoid arthritis is a chronic, systemic, inflammatory autoimmune disease that<br />

has its primary target in synovial tissues. It principally attacks the joints, which<br />

produce an inflammatory reaction that often progresses to destruction of the articular<br />

cartilage and bones and may eventually result in ankylosis of the joints. The major<br />

goal of this project is to gain insights into the molecular circuits that control the<br />

development of rheumatoid- and autoimmune diseases. Our short-term aim is to<br />

identify prognostic, diagnostic and potentially therapeutic miRNA markers and to<br />

better understand the role miRNAs play in the development of rheumatoid diseases.<br />

Plasma cell homing and B cell homeostasis controlled by Krüppellike<br />

factor 2 (KLF2)<br />

Rebecca Winkelmann, Lena Sandrock, Wolfgang Schuh and Hans-Martin Jäck<br />

Plasma cells are powerful micro factories producing thousands of antibody molecules<br />

per second and thereby protecting the organism from harmful pathogens and<br />

infections. Once a B cell encounters its specific antigen, a fascinating differentiation<br />

program is initiated that leads to morphological cellular restructuring and converting<br />

40


the B cell into an antibody-producing cell, the so-called plasma cell. Plasma cells can<br />

be subdivided into short- and long-lived plasma cells. Short-lived plasma cells<br />

predominantly produce IgM and have a life-span of approximately one week; in<br />

contrast long-lived plasma cells continuously produce high affinity IgG or IgA<br />

molecules and survive for years. Long-lived plasma cells, which are generated in<br />

secondary lymphoid tissues, have to reach specialized survival niches formed by<br />

stromal and other cells in the bone marrow. Still, it is puzzling how migration, homing<br />

and survival of these cells are controlled.<br />

Krüppel-like factor 2 (KLF2/LKLF) belongs to the family of Krüppel-like transcription<br />

factors, which consists of at least 17 members in mammals. Krüppel-like<br />

transcription factors bind to GC rich DNA domains via three C-terminal zinc fingers<br />

and are involved in controlling proliferation and terminal differentiation of various cell<br />

types. KLF2 was originally discovered in lung tissue and was shown to be important<br />

for cardiovascular development. Apart from its role in lung and cardiovascular<br />

development, KLF2 plays an important role in activation and migration of T<br />

lymphocytes.<br />

Using mice with a B cell specific deletion of KLF2, we demonstrated that KLF2 is<br />

crucial for proper homing of antigen-specific plasma cells to the bone marrow and for<br />

B cell homeostasis. In KLF2 deficient animals all splenic B cell subsets including B1<br />

cells were present, but their numbers were increased with a clear bias for marginal<br />

zone (MZ) B cells. In contrast, less peyers patches harboring fewer B cells were<br />

found and B1 cells in the peritoneal cavity were almost absent. B cell specific deletion<br />

of KLF2 results in a clear reduction of plasma cells in the bone marrow, whereas<br />

plasma cell numbers in the spleen and the blood were fairly normal, indicating that<br />

KLF2 plays an important role in homing of plasma cells to the bone marrow (Fig. 1).<br />

Since we found diminished levels of ß7 Integrin and L-Selectin on KLF2-deficient B<br />

cells, we propose that KLF2 regulates plasma cell homing to the bone marrow via<br />

regulation of these two cell adhesion molecules. Future experiments using a plasma<br />

cell-specific GFP reporter mouse will clarify the role of KLF2 in humoral immunity and<br />

provide new ways to interfere with the generation of pathologic plasma cells in<br />

patients with multiple myeloma and autoimmune disease.<br />

Fig. 1 Elispot assay of TNP-specific IgG secreting cells in spleen and bone marrow 14 days after<br />

boost immunization with TNP-KLH. A triplicate of one representative experiment is shown to the left,<br />

and results of all analyzed littermates are summarized with one dot representing the mean value of<br />

triplicates of one mouse is shown to the right.<br />

41


Function of EFhd1 and EFhd2 in B cells and brain in vivo<br />

Pavitra Purohit, Sebastian Brachs, Sebastian Dütting, Carmen Kroczek, Christiane<br />

Lang and Dirk Mielenz<br />

Antigen-induced B cell activation depends on a subset of antigen-B cell receptor<br />

(BCR) complexes that partition into plasma membrane microdomains, thereby,<br />

controlling the BCR-elicited calcium (Ca 2+ ) efflux from the endoplasmatic reticulum.<br />

This, in turn, regulates the total cytosolic Ca 2+ concentration [Ca 2+ ]i after BCR<br />

stimulation. Amplitude and duration of [Ca 2+ ]i in response to BCR activation governs<br />

activity of pro- or anti-apoptotic transcription factors, NF-AT and NF-�B. To identify<br />

mechanisms mediating membrane microdomain association of the BCR, a couple of<br />

years ago we defined the membrane microdomain proteome of B cells by separating<br />

purified membrane microdomains on 2D gels, followed my mass spectrometry.<br />

Thereby, we identified the new Ca 2+ binding protein Swiprosin-1/EFhd2 (EFhd2).<br />

EFhd2 regulates proximal BCR signaling and [Ca 2+ ]i via a mechanism that involves<br />

low-affine (110 �M) and cooperative Ca 2+ binding to EFhd2 in a 2:1 molar ratio, as<br />

well as membrane microdomain association of EFhd2 and proximal BCR signaling<br />

proteins. In addition to enhancing proximal BCR signals, EFhd2 interferes with antiapoptotic<br />

NF-�B activation, thereby, acting pro-apoptotically in B cells. Thus, loss of<br />

EFhd2 was expected to affect B cell selection and activation. To elucidate the<br />

function of EFhd2 in vivo, we generated EFhd2 deficient mice (EFhd2 K.O.).<br />

Surprisingly, B cell development and basal immunoglobulin (Ig) titers of all<br />

subclasses were normal in EFhd2 K.O. mice. However, EFhd2 K.O. mice responded<br />

slightly better to T-independent and T-dependent immunizations. More clearly, EFhd2<br />

K.O. mice developed higher affine antibodies towards a T-dependent antigen after<br />

recall immunization. We tested whether that could be due to increased competition<br />

amongst B cells, which would favour selection of high-affine B cell clones by T cells.<br />

In fact, EFhd2 K.O. mice developed larger germinal centers after immunization with<br />

sheep red blood cells. Along these lines, EFhd2 K.O. mice develop anti double<br />

stranded DNA antibodies after 1 year of age, pointing to either a defect in negative B<br />

cell selection or enhanced polyclonal activation. We obtained evidence for the latter<br />

point through polyclonal stimulation of purified B cells in vitro with lipopolysaccharide,<br />

demonstrating that EFhd2 K.O. B cells are 1) pre-activated and 2) switch faster from<br />

this pre-activated state into the plasma cell differentiation programme. In summary,<br />

EFhd2 is a negative regulator of B cell activation through an as yet to be defined<br />

mechanism.<br />

In contrast to EFhd2 that is expressed in all B cell stages, the related EFhd1<br />

becomes down-regulated during early B cell development by the pre-BCR. Activity of<br />

the transcription factor Foxo1, that controls stress responses, senescence and<br />

expression of recombinase activating genes (rag), is controlled by the pre-BCR as<br />

well as Interleukin-7 (IL-7). Interestingly we found a physical interaction of<br />

endogenous EFhd1 with endogenous Foxo1 in the cytoplasm of the pro B cell line<br />

38B9, as well as in 293 cells transfected with EFhd1. EFhd2 does not interact with<br />

Foxo1. To circumvent down-regulation of EFhd1 by the pre-BCR we generated<br />

transgenic mice which ectopically express EFhd1 (EFhd1tg) both in B as well as in T<br />

cells. In contrast to data obtained with an in vitro differentiation system, ectopic<br />

expression of EFhd1 did not affect steady state early B cell development in vivo.<br />

However, transgenic B and T cells expressed more CD62L, a Foxo1 target gene, and<br />

revealed increased Foxo1 expression on the protein level. We hypothesize that<br />

EFhd1 regulates Foxo1 function positively. This interaction should contribute to<br />

42


Foxo1 activity in early B cells in the context of IL-7 and pre-BCR signaling. Loss of<br />

EFhd1 should induce loss of function of Foxo1, thereby, reducing rag expression and<br />

slowing down early B cell development. Both gain and loss of function of EFhd1 may<br />

only be revealed under competitive conditions. Thus, we will create mixed bone<br />

marrow chimeras of wt and EFhd1tg bone marrow as well as wt and EFhd1 K.O.<br />

bone marrow and analyze the putative contribution of EFhd1 to early B cell<br />

development<br />

Since there was evidence that EFhd2 might be involved in neurodegeneration<br />

mediated by the microtubule-stabilizing protein tau we tested whether EFhd2<br />

modulates tau function. Surprisingly, we observed that co-expression of EFhd2 with<br />

mutant tau proteins (tau P301S and tau �K280), that cause frontotemporal dementia<br />

associated with parkinsonism (FTDP), but not with wildtype tau, caused degradation<br />

of EFhd2, but not of the co-expressed GFP. We are currently elucidating the<br />

proteolytic pathway underlying this degradation process. In line with a putative<br />

interaction of tau and EFhd2, we detected EFhd2 with newly generated monoclonal<br />

antibodies in ordered presynaptic complexes in murine as well as human brain<br />

sections. Interestingly, the presynaptic, ordered expression of EFhd2 was completely<br />

abolished in barins of Alzheimer’s patients. Likewise, EFhd2 co-localized with tau and<br />

with tau tangles in brains of Alzheimer’s patients. Finally we observed that the<br />

presynaptic marker Bassoon was largely absent from synaptic structures in brains of<br />

EFhd2 K.O. mice whereas tau expression was stronger and more confined to<br />

presynaptic complexes than in wildtype mice. Taken together we hypothesize that<br />

EFhd2 modulates tau function to control the composition of pre-synaptic complexes.<br />

This might be important during onset and progression of tau-mediated<br />

neurodegenerative diseases and for learning or rewarding processes.<br />

Genetic and functional analysis of Lupus-associated risk alleles<br />

Katrin Weiß and Barbara Fürnrohr<br />

Systemic Lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease<br />

involving multiple organs including skin, joints, kidneys, brains, the cardiovascular<br />

system and serosal membranes. Immunologically, SLE is characterised by the<br />

presence of antinuclear autoantibodies, especially against double stranded (ds) DNA<br />

and nucleosomes, activation of the complement system during flares and type I<br />

interferon secretion. The etiology of SLE remains elusive, however, an interplay of<br />

genetic and environmental factors is considered to ultimately cause immune<br />

dysregulation, resulting in clinical symptoms. Twin studies have reported a<br />

concordance of 24-57% and 2-5% in monozygotic and dizygotic twins, respectively,<br />

therefore SLE has an estimated heritability of 66%. Recent genome-wide association<br />

studies together with candidate gene studies have uncovered four major candidate<br />

gene loci, namely the MHC class II locus IKZF1, STAT4 and ITGAM which are most<br />

probably of functional relevance in SLE.<br />

Genetic variation at the ITGAM gene has been identified as a key genetic<br />

susceptibility effect in human systemic lupus erythematosus (SLE). It is one of the<br />

strongest known genetic risk factors in SLE, and is common (minor allele frequency<br />

approximately 10%) in all except East Asian populations. Interest has specifically<br />

focussed on the rs1143679 variant, which encodes an arginine to histidine amino<br />

acid change at position 77 in the �-propeller of the extracellular domain in CD11b<br />

which is encoded by ITGAM. As part of the complement receptor CR3, CD11b is<br />

43


mainly expressed on phagocytes, functioning as an adhesion molecule and a<br />

phagocytic receptor.<br />

At the Department of Medical and Molecular Genetics at King’s College in London in<br />

the group of Professor Timothy Vyse we have studied together with Dr. Benjamin<br />

Rhodes ex vivo monocytes/monocyte-derived macrophages from healthy volunteers<br />

homozygous for the 77H variant compared with homozygous wild-type (WT) controls.<br />

In cells isolated from donors homozygous for 77H we observed a reduction in the<br />

phagocytosis of iC3b opsonised sheep erythrocytes (sRBCiC3b) in comparison to WT<br />

cells.<br />

1160<br />

Ch04 Ch02/C h05 Ch05<br />

Fig. 1 Phagocytic Assay with red-fluorescent labelled iC3b-opsonised sheep red blood cells<br />

internalised by green-fluorescent labelled human macrophages.<br />

In addition, the adhesion of 77H monocytes was reduced to a range of CR3 ligands,<br />

such as iC3b, fibrinogen, fibronectin and DC-SIGN. To confirm the functionality of<br />

R77H specific ITGAM variants were transfected into COS7 cells and defective<br />

phagocytosis was observed in COS7 cells expression the 77H variant. We could also<br />

demonstrate differences in the release of Toll-like receptor-induced pro-inflammatory<br />

cytokines from monocytes of donors with different ITGAM genotypes following preengagement<br />

of CR3 using sRBCiC3b. The R77H variant therefore impairs a broad<br />

range of CR3 effector functions in human monocytes.<br />

CR3 ligation is likely to be important in immune complex and apoptotic cell clearance,<br />

both of which may contribute to the pathogenesis of SLE. Our future experiments will<br />

include phagocytic assays with monocytes from donors with different ITGAM<br />

genotype using fluorescent labelled apoptotic cells. Protein purification and structural<br />

analysis of the individual CD11b variants will hopefully unravel the functional<br />

differences observed in adhesion and phagocytosis.<br />

Collaboration with Prof. Timothy Vyse and Dr. Ben Rhodes (King’s College London)<br />

44


2009<br />

Publications (2009-2012)<br />

Original Articles (members are underlined)<br />

Trageser, D., Iacobucci, I., Nahar, R., Duy, C., Von Levetzow, G., Klemm, L., Park,<br />

E., Schuh, W., Gruber, T., Herzog, S., Kim, Y.M., Hofmann, W.K., Li, A.,<br />

Storlazzi, C.T., Jäck, H.M., Groffen, J., Martinelli, G., Heisterkamp, N., Jumaa,<br />

H., and Müschen, M. (2009). Pre-B cell receptor-mediated cell cycle arrest in<br />

Philadelphia chromosome-positive acute lymphoblastic leukemia requires<br />

IKAROS function. J Exp Med, 206 (8): 1739-1753.<br />

Urbonaviciute, V., Meister, S., Fürnrohr, B.G., Frey, B., Gückel, W., Schett, G.,<br />

Herrmann, M., and Voll, R.E. (2009). Oxidation of the alarmin high-mobility<br />

group box 1 protein (HMGB1) during apoptosis. Autoimmunity. 42 (4):305-7.<br />

2010<br />

Danke, C., Grünz, X., Wittmann, J., Schmidt, A., Agha-Mohammadi, S., Kutsch, O.,<br />

Jäck, H. M., Hillen, W., and Berens, C. (2010). Adjusting transgene expression<br />

levels in lymphocytes with a set of inducible promoters. J Gene Med, 12:501-<br />

515.<br />

Duy, C., Yu, J.J., Nahar, R., Swaminathan, S. , Kweon, S.M., Polo, J.M., Valls, E.,<br />

Klemm, L., Shojaee, S., Cerchietti, L., Schuh, W., Jäck, H.M., Hurtz, C.,<br />

Ramezani-Rad, P., Herzog, S., Jumaa, H., Koeffler, H.P., De Alborán, I.M.,<br />

Melnick, A.M., Ye, B.H., and Müschen, M. (2010). BCL6 is critical for the<br />

development of a diverse primary B cell repertoire. J Exp Med, 207 (6), pp.<br />

1209-1221.<br />

Fürnrohr, B.G., Wach, S., Haslbeck, M., Weber, C.K., Stach, C.M., Hueber, A., Graef,<br />

D., Spriewald, B., Manger, K., Herrmann, M., Kaufmann, K.M., Summer, G.F.,<br />

James, J.A., Goodmon, E., Kelly, J.A., Schett, G., Winkler, T., Harley, J.B.,<br />

and Voll, R.E. (2010). Hsp70 polymorphisms are genetically associated with<br />

systemic lupus erythematosous. Ann Rheum Dis, 69 (11):1983-9.<br />

Hakkim, A., Fürnrohr, B.G., Amann, K., Laube, B., Abu Abed, U., Brinkmann, V.,<br />

Herrmann, M., Voll, R.E., and Zychlinsky, A. (2010). Impairment of neutrophil<br />

extracellular trap degradation is associated with lupus nephritis. PNAS, 107<br />

(21):9813-8.<br />

Kroczek, C., Lang, C., Brachs, S., Grohmann, M., Dutting, S., Schweizer, A.,<br />

Nitschke, L., Feller, H. M., Jäck, H.M. and Mielenz, D. (2010). Swiprosin-<br />

1/EFhd2 controls B cell receptor signaling through the assembly of the B cell<br />

receptor, Syk, and phospholipase C gamma2 in membrane rafts. J Immunol,<br />

184:3665-3676.<br />

45


Lang, V. R., Mielenz, D., Neubert, K., Bohm, C., Schett, G., Jäck, H.M., Voll, R.E.,<br />

and Meister, S. (2010). The early marginal zone B cell-initiated T-independent<br />

type 2 response resists the proteasome inhibitor bortezomib. J Immunol,<br />

185:5637-5647.<br />

Metzner, M., Schuh, W., Roth, E., Jäck, H.M., and Wabl, M. (2010). Two forms of<br />

activation-induced cytidine deaminase differing in their ability to bind agarose.<br />

PloS one, 5 (1).<br />

Weber, C. K., Haslbeck, M., Englbrecht, M., Sehnert, B., Mielenz, D., Graef, D.,<br />

Distler, J. H., Mueller, R. B., Burkhardt, H., Schett, G., Voll, R. E., and<br />

Fürnrohr, B. G. (2010). Antibodies to the endoplasmic reticulum-resident<br />

chaperones calnexin, BiP and Grp94 in patients with rheumatoid arthritis and<br />

systemic lupus erythematosus. Rheumatology (Oxford), (12):2255-63.<br />

2011<br />

Avouac, J., Fürnrohr, B.G., Tomcik, M., Palumbo, K., Zerr, P., Horn, A., Dees, C.,<br />

Akhmetshina, A., Beyer, C., Distler, O., Schett, G., Allanore, Y., Distler, J.H.<br />

(2011). Inactivation of the transcription factor STAT4 prevents inflammationdriven<br />

fibrosis in systemic sclerosis animal models. Arthritis Rheum, 63<br />

(3):800-9.<br />

Brandl, A., Wittmann, J., and Jäck, H. M. (2011). Increased retroviral titers of miRNAencoding<br />

retroviruses by inhibition of the RNaseIII enzyme Drosha. Eur J<br />

Immunol, 41:549-551.<br />

Dedman, A. M., Majeed, Y., Tumova, S., Zeng, F., Kumar, B., Munsch, C., Bateson,<br />

A. N., Wittmann, J., Jäck, H. M., Porter, K. E., and Beech, D. J.. (2011).<br />

TRPC1 transcript variants, inefficient nonsense-mediated decay and low upframeshift-1<br />

in vascular smooth muscle cells. BMC Mol Biol, 12:30.<br />

Marschall, J.S., Wilhelm, T., Schuh, W., and Huber, M. (2011). MEK/Erk-based<br />

negative feedback mechanism involved in control of Steel Factor-triggered<br />

production of Krüppel-like factor 2 in mast cells. Cell Signal, (4):879-88.<br />

Rosenbaum, S., Kreft, S., Etich, J., Frie, C., Stermann, J., Grskovic, I., Frey, B.,<br />

Mielenz, D., Pöschl, E., Gaipl, U., Paulsson, M., and Brachvogel, B. (2011).<br />

Identification of novel binding partners (annexins) for the cell death signal<br />

phosphatidylserine and definition of their recognition motif. J Biol Chem,<br />

286:5708-5716.<br />

Weber, C.K., Mielenz, D., Voll, R.E., and Fürnrohr, B.G. (2011). Comment on:<br />

Antibodies to the endoplasmic reticulum-resident chaperones calnexin, BiP<br />

and Grp94 in patients with rheumatoid arthritis and systemic lupus<br />

erythematosus: Reply. Rheumatology (Oxford), 50:629-631.<br />

46


Winkelmann, R., Sandrock, L., Porstner, M., Roth, E., Mathews, M., Hobeika, E.,<br />

Reth, M., Kahn, M.L., Schuh, W., and Jäck, H.M. (2011). B cell homeostasis<br />

and plasma cell homing controlled by Krüppel-like factor 2. PNAS, 108 (2), pp.<br />

710-715.<br />

2012<br />

Buettner, M., Lang, A., Tudor, C.S., Meyer, B., Cruchley, A., Barros, M.H., Farrell, P.,<br />

Jäck, H.M., Schuh, W., and Niedobitek, G. (2012). Lytic EBV infection in<br />

epithelial cells but not in B lymphocytes is dependent on Blimp1. J Gen Virol,<br />

[Epub ahead of print].<br />

Hagen, S., Brachs, S., Kroczek, C., Fürnrohr, B.G., Lang, C., and Mielenz, D. (2012).<br />

The B cell receptor-induced calcium flux involves a calcium mediated positive<br />

feedback loop. Cell Calcium, [ePub 06Feb].<br />

Preußner, M., Schreiner, S., Hung, L.H., Porstner, M., Jäck, H.M., Benes, V., Rätsch,<br />

G., and Bindereif, A. (2012). HnRNP L and L-like cooperate in multiple-exon<br />

regulation of CD45 alternative splicing. Nucleic Acids Res. 2012 Mar 8. [Epub<br />

ahead of print]<br />

Books and Reviews<br />

Dütting, S., Brachs, S., and Mielenz, D. (2011). Fraternal twins: Swiprosin-1/EFhd2<br />

and Swiprosin-2/EFhd1, two homologous EF-hand containing calcium binding<br />

adaptor proteins with distinct functions. Cell Commun Signal, 9:2.<br />

Wittmann, J., and Jäck, H.M. (2011). microRNAs in rheumatoid arthritis: midget<br />

RNAs with a giant impact. Ann Rheum Dis, 70:Suppl 1:i92-i96.<br />

Wittmann, J., Jäck H.M., and Mashreghi, M.F. (2011). microRNAs in B- and T cells<br />

as regulators of inflammation. Z Rheumatol, 70:507-510.<br />

Rhodes B., Fürnrohr B.G., and Vyse, T.J. (2011). C-reactive protein in rheumatology:<br />

biology and genetics. Nat Rev Rheumatol. 7 (5):282-9.<br />

Sestak, A.L., Fürnrohr, B.G., Harley J.B., Merrill, J.T., and Namjou, B. (2011). The<br />

genetics of systemic lupus erythematosus and implications for targeted<br />

therapy. Ann Rheum Dis, 70 Suppl 1:i37-43.<br />

Wittmann, J., and Jäck, H.M. (2010). Serum microRNAs as powerful biomarkers in<br />

disease. Biochim Biophys Acta – Reviews on Cancer, 1806:200-207.<br />

Wittmann, J., and Jäck, H.M. (2010). New surprises from the deep - The family of<br />

small regulatory RNAs increases. ScientificWorldJournal, 10:1239-1243.<br />

47


TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Andreas Brandl<br />

MicroRNA-vermittelte Kontrolle der Antigen-induzierten B-Zell-Differenzierung und<br />

deren Rolle in der B-Zell-Entwicklung<br />

Sebastian Dütting<br />

Charakterisierung von Swiprosin-2/EFHD1 und dessen Funktion während der frühen<br />

B-Zellentwicklung<br />

Carmen Kroczek<br />

Funktionelle Analyse von Swiprosin-1/EFhd2 im proximalen B-Zellrezeptor Signalweg<br />

Anke Lang<br />

Mechanismen der lytischen Reaktivierung des Epstein-Barr Virus<br />

Sandra Schreiber<br />

Die Rolle von microRNAs in der frühen B-Zellentwicklung<br />

Stefanie Thiele<br />

Die Rolle der microRNAs und Dicer in Entzündungszellen von<br />

Atemwegserkrankungen<br />

Doctoral theses (Molecular Medicine)<br />

Rebecca Winkelmann<br />

The function of Krüppel-like factor 2 in B cell development<br />

Martina Porstner<br />

Untersuchungen zur Rolle von microRNAs in der Plasmazelldifferenzierung und dem<br />

Multiplen Myelom<br />

48


Hematopoiesis Unit<br />

Chair of Genetics – Department Biology<br />

Head: Thomas Winkler, Dr. rer. nat.<br />

Professor for Genetics<br />

Address: Hematopoiesis Unit<br />

Glückstr. 6<br />

D-91054 ERLANGEN<br />

Telefone: + 49 (9131) 85 29136<br />

Fax: + 49 (9131) 85 29106<br />

E-mail: thomas.winkler@molmed.rz.uni-erlangen.de<br />

Homepage: http://www.genetik.nat.uni-erlangen.de<br />

Ute Wellmann, Dr. rer. nat. *<br />

* part of the time reported<br />

Sven Brenner °<br />

Agnes Giniewski<br />

Kristin Schröder<br />

° PhD received<br />

Tanja Fisch*<br />

Markus Mroz*<br />

* part of the time reported<br />

Head<br />

Thomas Winkler, Prof. Dr. rer. nat.<br />

Professor for Genetics<br />

Postdoctoral Fellows<br />

Doctoral Students (Biology)<br />

Sonja Pötzsch °<br />

Martina Seefried<br />

Technicians<br />

49<br />

Sabrina Sell<br />

Florian Weisel °<br />

Andrea Schneider<br />

Hannes Tittlbach*


<strong>Research</strong><br />

Our research focus lies on the selection processes at different stages of the<br />

development of B-lymphocytes. We investigate the genetic mechanisms for the early<br />

development of B-lymphocytes, the mechanisms for the loss of self-tolerance in the<br />

autoimmune disease systemic lupus erythematosus and antibody-mediated<br />

protection against cytomegalovirus. In our research we predominantly work with<br />

mouse models that we also create in our laboratory by the techniques of gene<br />

targeting (knock-out mice, knock-in mice, transgenic mice).<br />

B cell development and regulation of VDJ-recombination<br />

During the differentiation of B-lymphocytes the genes for the antibody molecules are<br />

assembled in a unique process called VDJ-recombination. VDJ-recombination is<br />

responsible for the enormous diversity of antibody molecules. This diversity is<br />

necessary to defend all possible invaders of our body. Whereas the molecular<br />

mechanism of the VDJ-recombination is understood in very much detail today, the<br />

regulation of this process is less well understood. All 7 different antigen-receptor<br />

gene loci (3 for immunoglobulins and 4 for T-cell-receptors) use the same VDJrecombination<br />

machinery, but still the expression of the different genes is tighly<br />

regulated. Antibody receptors are expressed only on B-lymphocytes and T-cell<br />

receptors only on T-lymphocytes. In addition, during development of B-lymphocytes<br />

the genes for the heavy chain of the immunoglobulin is recombined before the genes<br />

for the light chain and additional regulation exists that only one antigen-receptor is<br />

expressed on each B-lymphocyte.<br />

Our research focus is to investigate the accessibility of the immunoglobulin heavy<br />

chain genes for the VDJ-recombinase. The epigenetic mechanisms for the regulation<br />

of the chromatin state at the immunoglobulin loci is analyzed by modern methods<br />

(ChIP-on-ChiP, ChIP-Seq). In addition we try to identify the essential regulatory<br />

sequences at the immunoglobulin locus by gene-targeting.<br />

Regulation of VDJ-recombination in precursor B-lymphocytes<br />

Agnes Giniewski and Thomas Winkler<br />

VDJ recombination is strictly confined to the early precursors of T- and B-cells, where<br />

only T-cells recombine the T-cell-receptor loci and only B-cells recombine the B-cellreceptor<br />

loci. The differential regulation of VDJ recombination remains unknown as<br />

the Rag1 and Rag2 enzymes that are critical for this reaction are expressed in early<br />

B-cells as well as in early T-cells. It has been proposed, that the differentially<br />

regulated accessibility of the T- and B-cell receptor loci is the basis of this regulation.<br />

As chromatin accessibility is regulated by histone modifications, we generated a high<br />

50


esolution map of histone modifications at the complete IgH-locus (app. 2.8 Mb) from<br />

early B-cell progenitors. Chromatin immunoprecipitation (ChIP) experiments using<br />

primary pro-B cells from Rag -/- mice and specific antibodies for acetylated histone H3<br />

and several different histone H3 methylations were performed. For the quantification<br />

of the results we use custom-made high-density arrays for a high-resolution analysis<br />

of histone modifications of the complete IgH locus (ChIP-on Chip analysis). The IgH<br />

Eµ intron enhancer region from the switch region to the first D segment (DQ52) is<br />

found to be enriched for the two activating histone modifications H3Ac and<br />

H3K4Me2. We also have indications that within the VH gene cluster histone<br />

acetylation and histone H3 K4 methylation is limited to the VH gene segments.<br />

In the ChIP-on Chip analysis we discovered three new regions in the intergenic part<br />

of the distal VH cluster (we call them IVARs), which are associated with high levels of<br />

active chromatin marks (H3Ac and H3K4Me2/3) only in pro B cells but not in pro T<br />

cells.<br />

To elucidate the role of the IVARs in VH -> DJH recombination we were generating a<br />

knock out mouse, which is lacking the IVAR #3 element. By analyzing the VDJrecombination<br />

repertoire using high-throughput next generation sequencing we found<br />

that the usage of distal VH gene segments is altered in mutant mice, supporting our<br />

hypothesis that the IVAR elements are involved in the VDJ-recombination of distal<br />

VH-elements.<br />

A hypomorph mutation of the B cell receptor<br />

Sven Brenner and Thomas Winkler<br />

As a result from a gene targeting experiment in which we wanted to introduce a GFPreporter<br />

gene into the immunoglobulin locus, we incidentally created a hypomorphic<br />

IgH-gene allele. Due to a poly-adenylation site introduced into the enhancer intron<br />

the levels of Igµ heavy chain mRNA and protein are reduced approximately 5-fold. In<br />

heterozygous mice B cells expressing the targeted IgH allele are undetectable,<br />

although VDJ recombination at the targeted allele is normal.<br />

We could show for these novel gene-targeted mice that hypomorphic expression of<br />

µHC leads to augmented selection processes at all stages of B-cell development,<br />

noticeably at the expansion of pre-B cells, the positive selection of immature B<br />

lymphocytes in the bone marrow and the selection into the follicular, marginal zone<br />

and B1 B lymphocyte compartments in peripheral lymphoid organs. Immature as well<br />

as mature follicular and marginal zone B lymphocytes in the peripheral lymphoid<br />

organs express lower levels of the receptor for B-cell activating factor (BAFF). In<br />

addition, hypomorphic expression of the BCR favors receptor editing. Together, our<br />

results highlight the critical importance of pre-BCR and BCR receptor levels for the<br />

normal development of B-lymphocyte-subpopulations in the context of intact VDJ<br />

recombination and a diverse antibody repertoire.<br />

51


During the last 3 years we had successful collaborations with Dr. Lars Nitschke (Dep.<br />

Biology, Genetics, Erlangen).<br />

The role of B lymphocytes in the chronic autoimmune disease<br />

systemic Lupus erythematosus<br />

SLE is a multisystem autoimmune disease characterized by autoantibodies against<br />

nuclear antigens, particularly antibodies against native (double-stranded) DNA (antidsDNA).<br />

Glomerulonephritis is one of the most serious manifestations of the disease.<br />

The incidence of glomerulonephritis in SLE patients is about 40-60%. The nephritis<br />

that accompanies SLE is mediated by immune complexes that deposit in the kidney<br />

and initiate an inflammatory response leading to organ damage and failure. The<br />

presence of anti-dsDNA antibodies correlates with nephritis in both human patients<br />

and mice with a spontaneous lupus like disease. Our research focus is on both the<br />

mechanisms of the generation of anti-dsDNA antibodies and on the role of antidsDNA<br />

antibodies in the pathogenesis of lupus nephritis.<br />

Molecular mechanisms of glomerular deposition of autoantibodies and<br />

pathogenesis of lupus nephritis<br />

Ute Wellmann, and Thomas Winkler, Technician: Markus Mroz<br />

A monoclonal anti-dsDNA antibody from a SLE patient binds in vivo to the<br />

mesangium upon injection in mice. Recently we have identified a crossreactivity of<br />

this antibody with nucleolin, a multifunctional protein that had been shown to be<br />

displayed on the cell surface of endothelial cells. We can show that nucleolin-specific<br />

rabbit antisera also bind specifically in the glomerulus of mice upon injection<br />

supporting the notion that nucleolin is accessible for autoantibodies in the<br />

glomerulus. Furthermore, we found a strong correlation of anti-nucleolin<br />

autoantibodies with more severe glomerulonephritis in several mouse models for<br />

SLE. In addition sera from SLE patients with severe glomerulonephritis contain antinucleolin<br />

autoantibodies in elevated titres.<br />

To further elucidate the potential pathogenetic role of anti-nucleolin (crossreactive)<br />

autoantibodies we eluted immune complexes from kidneys from proteinuric SLE mice<br />

(NZB/W mice). The eluted antibodies were highly enriched for anti-nucleolin reactivity<br />

further supporting the pathogenic role of these autoantibodies.<br />

(Collaborations with Dr. Ole-Petter Rekvig, University Tromsö, Norway)<br />

The evolution of anti-dsDNA autoantibodies in germinal centers<br />

Kristin Kruse, Ute Wellmann and Thomas Winkler, Technician: Andrea Schneider<br />

and Markus Mroz<br />

52


It has been proposed that the anti-double-stranded DNA (dsDNA) response in<br />

patients with systemic lupus erythematosus (SLE) is antigen driven and that DNA or<br />

nucleosomes select anti-DNA reactive, somatically mutated B cells. Recently we<br />

showed that high-affinity binding to dsDNA and nucleosomes is acquired by somatic<br />

replacement mutations in a stepwise manner, presumably in germinal center<br />

reactions. Importantly, autoreactivity seems to by acquired de novo as antibodies that<br />

have been backmutated to the germline sequence lack any autoreactivity towards<br />

DNA (Wellmann et al. 2005, PNAS 102: 9258-63). Nucleosomes accumulating due to<br />

defects in clearing of apoptotic debris, might positively select high affinity anti-DNA B<br />

cells.<br />

To directly test this hypothesis we were generating knock-in mice expressing a<br />

backmutated, non-autoreactive, lupus autoantibody. Only 3 somatic mutations are<br />

necessary and sufficient to create high affinity anti-dsDNA reactivity in this antibody.<br />

We targeted the BCR heavy- and light chain loci to obtain 33.C9gl heavy chain and<br />

33.C9gl light chain double knockin mice. As expected, B cells expressing the<br />

revertant B-cell receptor are developing normally and show no evidence for<br />

tolerization. Upon immunization with a surrogate phage antigen the transgenic B cells<br />

form germinal centers and undergo somatic hypermutation. The three critical somatic<br />

hypermutations, which are necessary and sufficient for the acquisition of high affinity<br />

anti-dsDNA binding, were not observed in a large collection of sequences analyzed.<br />

In accordance with this, anti-DNA autoantibodies do not develop, even after repetitive<br />

immunizations and when FDCs were MFG-E8 deficient. These results strongly<br />

suggest a self-tolerance checkpoint by deletion of autoreactive clones during or after<br />

the germinal center reaction.<br />

(Collaboration with Dr. Martin Herrmann, Med. Dept. III, Erlangen)<br />

We had further successful collaborations with Dr. Reinhard Voll at the NFZ on the<br />

role of plasma cells in SLE (Starke et al., 2011).<br />

Antibody-based immune protection against Cytomegalovirus<br />

Infections with Cytomegalovirus are usually asymptomatic and 50 – 90 % of the<br />

human population is persistently infected with this herpes virus. Severe disease<br />

becomes apparent when the immune system of the infected individual is suppressed,<br />

for example after transplantation. In addition, human cytomegalovirus is an important<br />

congenital infection.<br />

We aim at the better understanding of humoral immune-responses against the virus<br />

to develop new therapeutic concepts and potential therapeutic antibodies for<br />

treatment. We carry out all research in this field in close collaboration with the group<br />

of Prof. Michael Mach at the Virology Institute.<br />

For our research we use the human cytomegalovirus (HCMV) as well as its close<br />

relative in the mouse, the murine cytomegalovirus (MCMV).<br />

53


Adoptive transfer of memory B cells as a new cell based therapy for infection<br />

with Cytomegalovirus after transplantation<br />

Florian Weisel, Martina Seefried, Anna Bootz*, Monika Dietz*, Astrid Karbach*, Julia<br />

Winkler # , Andreas Mackensen # , Michael Mach*, Thomas Winkler, Technicians:<br />

Andrea Schneider, Hannes Tittlbach<br />

* Institute for Clinical and Molecular Virology<br />

# Department of Internal Medicine 5, Hematology and Oncology<br />

Severe disease associated with cytomegalovirus (CMV) infection is still a major<br />

problem in transplant patients. Support of the patient’s immune defense against the<br />

virus is therefore a major goal in transplantation medicine. We use the murine model<br />

of CMV (MCMV) to investigate the potential of a cell-based strategy to support the<br />

humoral antiviral immune response. We have previously shown that a transfer of<br />

memory B cells was effective in protecting from an ongoing viral infection indicating a<br />

therapeutic potential of virus specific memory B cells. In the recent period for this<br />

report we analyzed the effector mechanisms for the protective effect of anti-CMV<br />

antibodies. We have clear data that Fc-receptor (FcR) mediated mechanisms,<br />

presumably antibody dependent cellular cytotoxicity (ADCC) play a major role in the<br />

protective effect of antibodies in this viral disease. Determination of viral load in<br />

organs (Fig. 1A, B) and analysis of survival (Fig. 1C) after serotherapy confirmed that<br />

FcγR-deficient mice are not protected by administration of serum from MCMVimmune<br />

donors. Further experiments with individual FcγRs revealed a complex and<br />

partly redundant function of the different FcγRs. Whereas genetic deletion or antibody<br />

blockade of either FcγRIII or FcγRIV alone have any measurable effect on antibody<br />

protection, a combined defect of FcγRIII and FcγRIV had strong effects on protective<br />

function of antibodies. With regard to effector cells we have the surprising finding that<br />

NK cells, which are believed to be efficient effectors for ADCC, have surprisingly no<br />

major role in antibody-mediated protection in CMV-infection as NK cell-depleted<br />

animals were fully protected by serum therapy. Preliminary results indicate that<br />

certain monocyte subpopulations that can be specifically depleted by clodronate<br />

liposomes are important for antibody protection. We are currently studying the<br />

function of these monocyte subpopulation and the role of FcγRI in antibody-mediated<br />

protection in CMV-infection.<br />

54


Fig. 1 Fc_Rs are essential for antibody-mediated protection from MCMV infection.<br />

Mice were infected with 10 5 plaque forming units MCMV157luc and treated with 100µl serum from<br />

naïve or MCMV-immune donors. (A) Bioluminescence imaging of mice 10 days after serum treatment.<br />

The pseudocolor scale indicates viral load. (B) Relative viral load in organs in mice 14days after<br />

serotherapy. Luciferase activity was measured in organ homogenates and relative light units (RLU)/15<br />

µg protein are shown. (C) Survival curve of 5 mice/group after serotherapy.<br />

Our data provide evidence that a cell based strategy using memory B-lymphocytes to<br />

support the humoral immune response can be effective to combat infectious<br />

pathogens in severely immunodeficient hosts, like patients after stem cell<br />

transplantation. To bring this strategy into the clinics, the preclinical technology<br />

development including registration and implementation according to AMG –<br />

guidelines were mandatory prerequisites for the initiation of a phase I/II clinical trial,<br />

particularly as no experience was available for the GMP-qualified production of Blymphocytes.<br />

We therefore developed protocols for the production of a GMP-qualified<br />

B-lymphocyte product from leukapheresis material during the last 2 years by using<br />

the CliniMacs-technology in cooperation with Miltenyi-Biotec. A two-step separation<br />

strategy was developed and validated under GMP-conditions in a validated GMPlaboratory<br />

(Department of Transfusion Medicine). The manufacturing license for the<br />

B-cell product will soon be obtained.<br />

As a second prerequisite for the initiation of a first-in-human phase I/IIa clinical trial<br />

we developed a clinical protocol for a phase I/II study in accordance with GCP<br />

guidelines. An adoptive transfer of B-lymphocytes after allogeneic stem cell<br />

55


transplantation represents a novel, world-wide first-in-man cellular therapy.<br />

Therefore, this therapy has to be evaluated in a Phase I/IIa clinical trial primarily for<br />

safety and feasibility. The B-lymphocyte product will be the investigational medicinal<br />

product (IMP) in this trial. After extensive discussion with stem cell transplantation<br />

specialists we considered that a phase I/II trial with patients at risk for HCMVreactivation<br />

shortly after stem cell transplantation would potentially provide<br />

inconclusive data, especially regarding safety, which has to be the primary endpoint<br />

of the study. We therefore intend a transfer of the B-lymphocyte product<br />

approximately 4-5 months after transplantation when B-lymphocyte numbers are still<br />

significantly reduced in patients receiving matched-unrelated grafts and revaccination<br />

of the patient is routinely performed in the clinic. For the patient, a<br />

significant benefit for the reconstitution of the humoral immune function would be<br />

expected by the transfer of the B-lymphocyte product. At the same time, routinely<br />

performed re-vaccinations would offer the unique possibility to assess the function of<br />

the transferred B-lymphocytes from the donor. For the immune monitoring of patients<br />

in the clinical trial we already developed methods for the analysis of the frequencies<br />

of antigen-specific memory B-lymphocytes and plasma-blasts for various viral<br />

antigens and vaccine antigens.<br />

Currently we complete the documents for the trial protocol, the investigator brochure<br />

and the IMP dossier for the application to Paul-Ehrlich-Institure with the help of the<br />

Center of Clinical Studies (CCS) of the University hospital. These preclinical and<br />

clinical aspects of our research are performed in close collaboration with Dr. Julia<br />

Winkler and Dr. Andreas Mackensen, Department of Internal Medicine 5 and Miltenyi<br />

Biotec and are supported by the BayImmuNet.<br />

Furthermore, we refined our murine model further including viral reactivation from<br />

latency as well as induction of graft-versus-host disease (GVHD) after bone marrow<br />

transplantation to adopt it as close as possible to the clinical situation. Using this<br />

model, we were able to demonstrate a vital importance of antibodies in prevention of<br />

virus reactivation following bone marrow transplantation. When B-cell deficient mice<br />

unable to produce antibodies in the serum were infected with MCMV, a rapid<br />

clearance of the virus was observed in the B-cell deficient mice, as expected,<br />

because these mice contain a normal T-cell compartment being able to control<br />

infection to latency. When these recipients were treated by irradiation and<br />

transplanted with autologous or allogeneic bone marrow a massive reactivation of<br />

MCMV was observed in B-cell deficient bone marrow recipient mice whereas mice<br />

with serum antibodies only showed a low level of reactivation of the virus Fig. 2). By<br />

day 30 after transplantation the virus in surviving B-cell deficient animals was under<br />

control, due to the newly formed immune system derived from the bone marrow<br />

transplant.<br />

When a graft-versus-host reaction was induced in this transplant setting we observed<br />

a massive and uncontrolled reactivation of MCMV, even in the presence of antibodies<br />

(data not shown). These preliminary data demonstrate a major clinical problem in<br />

56


stem cell transplantations for the first time in the mouse model and will enable us to<br />

refine our therapy concepts under these clinically relevant conditions.<br />

Fig. 2 Antibodies are important for prevention of MCMV reactivation after bone marrow<br />

transplantation<br />

C57Bl/6 mice and B-cell deficient JH -/- were infected with 10 5 plaque forming units of MCMV157luc<br />

and transplanted with bone marrow after lethal irradiation. Bioluminescence imaging of mice after<br />

primary infection, latency (d24) and after bone marrow transplantation.<br />

These experiments were in part (GVHD-model) performed in close collaboration with<br />

the group of Dr. Evelyn Ulrich, Department of Internal Medicine 5.<br />

Neutralizing human antibodies against HCMV<br />

Sonja Pötzsch and Thomas Winkler, Technicians: Tanja Fisch, Hannes Tittlbach and<br />

Uwe Appelt<br />

Human cytomegalovirus (HCMV) infections can lead to clinically manifested, often<br />

life-threatening disease among immunocompromised hosts, in particular solid organ<br />

or bone marrow transplant recipients. Owing to lack of immune response in these<br />

patients, virus replication and dissemination occurs uncontrollably. Therefore,<br />

57


passive immunization for prevention or amelioration of CMV disease in high-risk<br />

individuals is a major goal not only in transplant medicine. The administration of CMV<br />

neutralizing monoclonal antibodies could be a feasible approach and has shown to<br />

be very effective in the mouse model.<br />

In addition, infection during pregnancy has been recognized as a major problem for<br />

the newborn. Congenital (present at birth) CMV infection causes more long-term<br />

problems and childhood deaths than Down syndrome, fetal alcohol syndrome, and<br />

neural tube defects. Antibodies delivered to pregnant mothers at risk would have the<br />

potential to prevent severe sequelae, such as hearing loss and other CNS defects in<br />

about 30,000 children which are born with congenital CMV infection each year<br />

(numbers from CDC for the USA, http://www.cdc.gov/cmv/trends-stats.html).<br />

Target antigens of CMV neutralizing antibodies usually are glycoproteins in the viral<br />

membrane, the most immunogenic being glycoprotein B (gB). Glycoprotein B (gB) is<br />

a well conserved protein among herpesviruses. In the case of HCMV, gB is a major<br />

target for the virus neutralizing humoral immune response. Two antigenic domains<br />

(AD) on gB which are targets of neutralizing antibodies have been identified and<br />

characterized in some detail. AD-1 is located between aa 552-635 and AD-2 between<br />

aa 68-77 of gB strain AD169. A third antigenic domain (AD-3) which is located<br />

intravirally and which is not target of neutralizing antibodies has also been identified.<br />

We have used data from the recently<br />

established 3D structure of HSV-1 gB for<br />

molecular modelling of HCMV gB and<br />

based on the model we have identified<br />

two domains on HCMV gB which<br />

resemble structural compact domains on<br />

the protein surface that could be targets of<br />

antibodies. Domain 1 comprises aa 132-<br />

343 and domain 2, a discontinuous<br />

sequence, comprises aa 116-132 + 344-<br />

440 of gB strain AD169 (Fig. 3)<br />

Fig. 3 Antigenic domains (ADs) on HCMV<br />

glycoprotein B<br />

(3D model generated by H. Sticht)<br />

In a comprehensive screening of over 800<br />

human monoclonal antibodies binding to<br />

gB derived from normal blood donors we identified several strongly neutralizing<br />

antibodies reacting against newly defined epitopes on domain 1 and domain 2 (AD-5<br />

and AD-4, respectively. These new antibodies certainly represent lead candidates for<br />

further preclinical and clinical development for therapeutic applications in prenatal<br />

HCMV infections and HCMV reactivation in transplant patients.<br />

(Collaboration with Dr. Michael Mach, Institute for Virology, Erlangen, Dr. Heinrich<br />

Sticht, Bioinformatics, Erlangen and 4-Antibody AG, Basel, Switzerland).<br />

58


2011/12<br />

Publications (2009-2012)<br />

Original Articles<br />

Schietke, R.E., T. Hackenbeck, M. Tran, R. Gunther, B. Klanke, C.L. Warnecke, K.X.<br />

Knaup, D. Shukla, C. Rosenberger, R. Koesters, S. Bachmann, P. Betz, G. Schley, J.<br />

Schodel, C. Willam, T. Winkler, K. Amann, K.U. Eckardt, P. Maxwell, and M.S.<br />

Wiesener. 2012. Renal Tubular HIF-2alpha Expression Requires VHL Inactivation<br />

and Causes Fibrosis and Cysts. PLoS ONE 7:e31034.<br />

Sitte, S., J. Glasner, J. Jellusova, F. Weisel, M. Panattoni, R. Pardi, and A. Gessner.<br />

2012. JAB1 is essential for B cell development and germinal center formation and<br />

inversely regulates Fas ligand and Bcl6 expression. J Immunol 188:2677-2686<br />

Pötzsch, S., N. Spindler, A.K. Wiegers, T. Fisch, P. Rucker, H. Sticht, N. Grieb, T.<br />

Baroti, F. Weisel, T. Stamminger, L. Martin-Parras, M. Mach, and T.H. Winkler. 2011.<br />

B Cell Repertoire Analysis Identifies New Antigenic Domains on Glycoprotein B of<br />

Human Cytomegalovirus which Are Target of Neutralizing Antibodies. PLoS Pathog<br />

7:e1002172.<br />

Brenner, S., D. Drewel, T. Steinbart, F. Weisel, E. Hartel, S. Pötzsch, H. Welzel, A.<br />

Brandl, P. Yu, G.C. Mudde, A. Schweizer, L. Nitschke, and T.H. Winkler. 2011. A<br />

hypomorphic IgH-chain allele affects development of B-cell subsets and favours<br />

receptor editing. Embo J 30:2705-2718.<br />

Ackermann, J.A., D. Radtke, A. Maurberger, T.H. Winkler, and L. Nitschke. 2011.<br />

Grb2 regulates B-cell maturation, B-cell memory responses and inhibits B-cell Ca(2+)<br />

signalling. Embo J 30:1621-1633.<br />

Starke, C., S. Frey, U. Wellmann, V. Urbonaviciute, M. Herrmann, K. Amann, G.<br />

Schett, T. Winkler, and R.E. Voll. 2011. High frequency of autoantibody-secreting<br />

cells and long-lived plasma cells within inflamed kidneys of NZB/W F1 lupus mice.<br />

Eur J Immunol 41:2107-2112<br />

2010<br />

Furnrohr, B.G., S. Wach, J.A. Kelly, M. Haslbeck, C.K. Weber, C.M. Stach, A.J.<br />

Hueber, D. Graef, B.M. Spriewald, K. Manger, M. Herrmann, K.M. Kaufman, S.G.<br />

Frank, E. Goodmon, J.A. James, G. Schett, T.H. Winkler, J.B. Harley, and R.E. Voll.<br />

2010. Polymorphisms in the Hsp70 gene locus are genetically associated with<br />

systemic lupus erythematosus. Ann Rheum Dis 69:1983-1989.<br />

Jellusova, J., U. Wellmann, K. Amann, T.H. Winkler, and L. Nitschke. 2010. CD22 x<br />

Siglec-G double-deficient mice have massively increased B1 cell numbers and<br />

develop systemic autoimmunity. J Immunol 184:3618-3627.<br />

59


Weisel, F.J., U.K. Appelt, A.M. Schneider, J.U. Horlitz, N. van Rooijen, H. Korner, M.<br />

Mach, and T.H. Winkler. 2010. Unique Requirements for Reactivation of Virus-<br />

Specific Memory B Lymphocytes. J Immunol 185:4011-4021.<br />

2009<br />

Burkhardt, C., S. Himmelein, W. Britt, T. Winkler, and M. Mach. 2009. Glycoprotein N<br />

subtypes of human cytomegalovirus induce a strain-specific antibody response<br />

during natural infection. J Gen Virol 90:1951-1961.<br />

Books and Reviews<br />

Almqvist, N., T.H. Winkler, and I.L. Martensson. 2011. Autoantibodies: Focus on anti-<br />

DNA antibodies. Self Nonself 2:11-18.<br />

Kruse, K., C. Janko, V. Urbonaviciute, C.T. Mierke, T.H. Winkler, R.E. Voll, G. Schett,<br />

L.E. Munoz, and M. Herrmann. 2010. Inefficient clearance of dying cells in patients<br />

with SLE: anti-dsDNA autoantibodies, MFG-E8, HMGB-1 and other players.<br />

Apoptosis 15:1098-1113.<br />

60


TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Florian Weisel<br />

Reaktivierung Virus-spezifischer Gedächnis B Zellen<br />

Sven Brenner<br />

Selection processes guided by B-cell receptor signal strength<br />

Sonja Pötzsch<br />

Neutralizing human monoclonal antibodies against Cytomegalovirus<br />

Kristin Kruse<br />

Die Evolution von anti-DNA Autoantikörpern in Keimzentren<br />

Agnes Giniewski<br />

Charakterisierung eines neuen regulatorischen Elementes im IgH-Genlokus<br />

Martina Seefried<br />

Immunological mechanisms of reactivation of murine CMV after bone marrow<br />

transplantion<br />

Sabrina Sell<br />

The protective role of γ/δ T-lymphocytes in murine CMV infection<br />

61


IZKF Junior Group III<br />

BMBF <strong>Research</strong> Group Neurosciences<br />

Head: Beate Winner, PD Dr. med.<br />

Address: IZKF Junior Group III<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 39301<br />

Fax: + 49 (9131) 85 39311<br />

E-mail: beate.winner@med.uni-erlangen.de<br />

Homepage:<br />

http://www.izkf.uk-erlangen.de/e37/e38/e87/e1593/index_ger.html<br />

Head<br />

Beate Winner, PD Dr. med.<br />

<strong>Research</strong> associates Doctoral students (Biology)<br />

Francesc Perez-Branguli, PhD*<br />

Iryna Prots, Dr. rer. nat.<br />

Haixin Zhang, MD/ PhD*<br />

Master Students (M)/<br />

Student helpers (S)<br />

Stefanie Brey (M) *<br />

Naime Denguir (S)<br />

Theresa Halder (S)<br />

* part of the time reported<br />

62<br />

Steven Havlicek<br />

Himanshu Mishra<br />

Technicians<br />

Daniela Gräf*<br />

Domenika Saul*<br />

Holger Wend


<strong>Research</strong><br />

The overall goal of research in our laboratory is to model neurodegenerative<br />

diseases in stem cell based models. Specifically we investigate neurodegeneration<br />

and regeneration in neurites. The two diseases studied are Parkinson’s disease (PD)<br />

and hereditary spastic paraplegia (HSP), a specific genetic form of motor neuron<br />

disease.<br />

Hereditary spastic paraplegia (HSP) and other motor neuron diseases<br />

We are specifically interested in understanding the involvement of selected hereditary<br />

spastic paraplegia (HSP) proteins in axonal degeneration. HSP is a genetic condition<br />

leading to corticospinal tract degeneration. HSPs selectively involve a lengthdependent<br />

axonopathy from long projecting corticospinal motorneurons (CSMN),<br />

while sparing the neuronal cell bodies. We study CSMN to understand the molecular<br />

mechanisms crucial for axonal maintenance and degeneration. The models we use<br />

are human induced pluripotent stem cell (hiPSC)-derived neurons generated from<br />

HSP patient’s fibroblasts (SPG4 and SPG11). The overall aim is to understand the<br />

normal function of selected HSP proteins, to determine how abnormality of these<br />

functions lead to the disease and to use this knowledge to rationally design new<br />

therapeutic strategies for the disorder.<br />

63


Figure 1: Induced pluripotent stem cells (iPSC) to model neurodegenerative<br />

diseases. Human iPSC from patients (e.g. HSP) and controls are generated after<br />

reprogramming of somatic tissue into iPSCs. Neural progenitor cells (NPC) are<br />

generated from these iPSCs and are further differentiated into neurons and/or glial<br />

cells. The neuronal phenotype is analyzed. Once a distinct disease-related<br />

phenotype is identified, drug-screening platforms can be developed to test<br />

compounds. Therapeutic compounds could emerge from the screenings, potentially<br />

benefiting neurologic patients.<br />

Understanding aggregation in Parkinson’s disease (PD)<br />

Protein aggregation of mis-folded proteins is associated with several<br />

synucleinopathies. Lewy bodies, an aggregated fibrillar form of multiple proteins<br />

(among them α-synuclein), are the major pathological hallmark in PD. During the<br />

recent year we have shown in vivo that small oligomeric species of the protein, rather<br />

than its aggregated fibrillar counterpart, are the toxic culprit in a rat model of<br />

synucleinopathies. We are investigating the impact of oligomerisation of α-synuclein<br />

in human in vitro models. Specifically, we are interested in studying the mechanism<br />

and functional consequences of oligomerization for neurite degeneration, axonal<br />

transport, and cellular membranes.<br />

Important results<br />

In vivo demonstration that oligomers are toxic<br />

We tested, whether Glu -> Lys mutations in the core area of α-synuclein interfere<br />

with the formation of α-synuclein fibrils and form oligomers instead. Several single<br />

point mutations in the core region (30-110) of α-synuclein have been investigated for<br />

their tendency to form fibrils and oligomers when compared with wild-type (WT) α-<br />

synuclein. In collaboration with Roland Riek, ETH Zürich, these results were obtained<br />

by incubating the different (artificial) variants of α-synuclein in a cell-free system. α-<br />

synuclein variants were analyzed by electron microscopy (EM), which indicated that<br />

these artificial mutants in the core region (30-110) were able to form oligomers but<br />

did not form fibrils. Interestingly, some preparations of these artificial mutants did not<br />

show amyloid fibrils even after long-term incubation, but formed very pronounced,<br />

ring/pore-like structures resembling large oligomers. We found two α-synuclein single<br />

point mutations, E35K and E57K, that showed a strongly decreased tendency of fibril<br />

formation when compared with WT α-synuclein, as evidenced by time-resolved<br />

amyloid formation. Neither variant E35K nor E57K showed amyloid fibrils under<br />

64


certain conditions, but they formed very pronounced ring/pore-like structures. In<br />

contrast, human WT and familial α-synuclein variants formed amyloid fibrils as<br />

expected. We injected lentiviral vectors of the artificial mutants into the rat substantia<br />

nigra and examined survival of dopaminergic neurons after 3 weeks. Our data<br />

indicate, that an enormous loss of dopaminergic cells in the substantia nigra of rats<br />

upon 3 weeks of lentiviral infection is observed for the artificial α-synuclein variants<br />

that form large quantities of oligomers, whereas the variant that forms fibrils very<br />

quickly is less toxic.<br />

The correlation between the extent of oligomer formation in vitro and toxicity in vivo is<br />

apparent. Furthermore, there is a lack of correlation between the tendency of amyloid<br />

fibril formation in vitro and toxicity in vivo and between the amount of monomer in<br />

vitro and toxicity in vivo. Combining the in vitro and in vivo experiments, we suggest<br />

that the in vivo-derived SDS-stable oligomers may be the building blocks for a larger<br />

alpha-synuclein oligomer that causes toxicity. In conclusion, this structure-toxicity<br />

relationship study of human α-synuclein strongly supports the notion that α-synuclein<br />

oligomers are the toxic species in PD and that the amyloid fibrils are not directly toxic,<br />

despite being present in the Lewy bodies, which are the pathological hallmark of PD<br />

(published in PNAS 2011).<br />

We are currently investigating the role of α-synuclein oligomers for axonal transport,<br />

membrane stability and the effect of α-synuclein on dendrites in stem cell based<br />

rodent and human models.<br />

Figure 2: In vitro aggregation of wild-type α-synuclein (B) compared to artificial<br />

oligomer-forming mutants (e.g. A) in a cell-free system. EM shows an abundance of<br />

fibrils in the wild-type α-synuclein (B), whereas only pore-like oligomers are found in<br />

the artificial oligomerizing mutant (A). Lentivirus injection into the rat substantia nigra<br />

of adult rats (C: paradigm): decrease in TH-positive cells following injection with the<br />

65


oligomerizing mutant (α-syn mut, F) compared to wild-type α-synuclein (α-syn, E)<br />

and GFP control (D, PNAS 2011).<br />

Figure 3: Neuronal differentiation of dopaminergic neurons from human embryonic<br />

stem cell (hESC)-derived neural precursor cells (NPCs). (A) Neurons were<br />

differentiated from NPCs over 6 weeks using neurotrophic factors and characterized<br />

by immunofluorescence for neuronal markers (B) and by electrophysiology (C,<br />

collaboration with Dr. Tobias Huth, Institute of Physiology and Pathophysiology,<br />

Head: Prof. Dr. Christian Alzheimer).<br />

Publications (2010-current)<br />

Bracko O, Singer T, Aigner S, Knobloch M, Winner B, Ray J, Clemenson GD Jr, Suh<br />

H, Couillard-Despres S, Aigner L, Gage FH, Jessberger S (2012). Gene expression<br />

profiling of neural stem cells and their neuronal progeny reveals IGF2 as a regulator<br />

of adult hippocampal neurogenesis. J Neurosci. 32(10):3376-87.<br />

Kohl Z, Winner B, Ubhi K, Rockenstein E, Mante M, Münch M, Barlow C, Carter T,<br />

Masliah E, Winkler J (2012) Fluoxetine rescues impaired hippocampal neurogenesis<br />

in a transgenic A53T synuclein mouse model. Eur J Neurosci. 35(1):10-9.<br />

Winner B, Kohl Z, Gage FH. (2011) Neurodegenerative disease and adult<br />

neurogenesis. Eur J Neurosci. 33(6):1139-51.<br />

Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner S, Hetzer C, Loher T,<br />

Vilar M, Campioni S, Tzitzilonis C, Soragni A, Jessberger S, Mira H, Consiglio A,<br />

Pham E, Masliah E, Gage FH, Riek R. (2011) In vivo demonstration that alphasynuclein<br />

oligomers are toxic. Proc Natl Acad Sci U S A. 108(10):4194-9.<br />

McCrone P, Payan CA, Knapp M, Ludolph A, Agid Y, Leigh PN, Bensimon G;<br />

NNIPPS Study Group (2011) The economic costs of progressive supranuclear palsy<br />

and multiple system atrophy in France, Germany and the United Kingdom. PLoS<br />

One. 6(9):e24369.<br />

Payan CA, Viallet F, Landwehrmeyer BG, Bonnet AM, Borg M, Durif F, Lacomblez L,<br />

Bloch F, Verny M, Fermanian J, Agid Y, Ludolph AC, Leigh PN, Bensimon G;<br />

NNIPPS Study Group (2011), Disease severity and progression in progressive<br />

supranuclear palsy and multiple system atrophy: validation of the NNIPPS—<br />

Parkinson Plus Scale. PLoS One. 6(8):e22293.<br />

Rolland Y, Vérin M, Payan CA, Duchesne S, Kraft E, Hauser TK, Jarosz J, Deasy N,<br />

Defevbre L, Delmaire C, Dormont D, Ludolph AC, Bensimon G, Leigh PN; NNIPPS<br />

66


Study Group (2011). A new MRI rating scale for progressive supranuclear palsy and<br />

multiple system atrophy: validity and reliability. J Neurol Neurosurg Psychiatry.<br />

82(9):1025-32.<br />

Winner B, Melrose HL, Zhao C, Hinkle KM, Yue M, Kent C, Braithwaite AT,<br />

Ogholikhan S, Aigner R, Winkler J, Farrer MJ, Gage FH (2011). Adult neurogenesis<br />

and neurite outgrowth are impaired in LRRK2 G2019S mice. Neurobiol Dis.,<br />

41(3):706-16.<br />

Marchetto MC, Winner B, Gage FH (2010). Pluripotent stem cells in<br />

neurodegenerative and neurodevelopmental diseases. Hum Mol Genet. R71–R76.<br />

Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH (2010). Mechanism<br />

underlying inflammation in neurodegeneration. Cell. 19;140(6):918-34.<br />

Kohl Z, Regensburger M, Aigner R, Kandasamy M, Winner B, Aigner L, Winkler J.<br />

(2010) Impaired adult olfactory bulb neurogenesis in the R6/2 mouse model of<br />

Huntington’s disease. BMC Neurosci. 11:114.<br />

Kandasamy M, Couillard-Despres S, Raber KA, Stephan M, Lehner B, Winner B,<br />

Kohl Z, Rivera FJ, Nguyen HP, Riess O, Bogdahn U, Winkler J, von Hörsten S,<br />

Aigner L (2010). Stem cell quiescence in the hippocampal neurogenic niche is<br />

associated with elevated transforming growth factor-beta signaling in an animal<br />

model of Huntington disease. J Neuropathol Exp Neurol. 69(7):717-28.<br />

Doctoral (Biology)<br />

TRAINING OF GRADUATE AND MASTER STUDENTS<br />

Steven Havlicek<br />

Modeling familial motor neuron disease (SPG4) by the use of induced pluripotent<br />

stem cells<br />

Himanshu Mishra<br />

Modeling familial motor neuron disease (SPG11) by the use of induced pluripotent<br />

stem cells<br />

Master (Molecular Medicine)<br />

Stefanie Brey<br />

Impact of oligomerisation of α-synuclein on membranes in human neural in vitro<br />

models<br />

67


INTERDISCILINARY CENTER OF CLINICAL<br />

RESEARCH<br />

Junior <strong>Research</strong> Group 2 (IZKF – N2)<br />

Head: Jens Titze, Prof. Dr. med.<br />

Address: IZKF – N2<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 39300<br />

Fax: + 49 (9131) 85 36386<br />

E-mail: jens.titze@uk-erlangen.de<br />

jens.m.titze@vanderbilt.edu<br />

Homepage: http://www.izkf.ukerlangen.de/e1615/inhalt1880/IZKF_JB11_05.2012.pdf<br />

Agnes Schröder, Dr. rer. nat.<br />

Peter Linz, Dr. rer. nat.<br />

Natalia Rakova, Dr.<br />

* part of the time reported<br />

Head<br />

Jens Titze, Prof. Dr. med.<br />

Supporting Staff<br />

Doris Bittner, Secretary<br />

Postdoctoral Fellows<br />

68<br />

Anke Dahlmann, Dr. med.<br />

Christoph Kopp, Dr. med.<br />

Sven Brenner, Dr. rer. nat.


Kathrin Jüttner<br />

° PhD received<br />

Florian Eicher<br />

° PhD received<br />

Ulrike Goller<br />

Jenny Goß<br />

* part of the time reported<br />

Doctoral Students (Biology)<br />

Diana Friedrich<br />

Doctoral Students (Medicine)<br />

Kathrin Dörfelt<br />

Technicians<br />

Sabrina Cabric<br />

69


<strong>Research</strong><br />

Primary essential hypertension is the major cardiovascular disease risk factor.<br />

Dietary salt intake is a putative driving force of blood pressure elevation; however,<br />

the mechanisms of this effect remain unclear. The central nervous system, heart and<br />

blood vessels, and kidney are primary participants and the kidney is the putative<br />

grand regulator of salt disposition and blood pressure. The overall goal of this work is<br />

to address whether or not local regulation of skin electrolyte metabolism is important<br />

for blood pressure control. Clinical evidence from pilot studies in humans and<br />

preliminary experimental data accumulated in our laboratory, support this hypothesis.<br />

Our data point to macrophage-derived vascular endothelial growth factor C (VEGF-C)<br />

as a crucial factor controlling skin electrolyte homeostasis. VEGF-C promotes<br />

interstitial electrolyte clearance through the cutaneous lymph capillary network.<br />

Macrophages induce hyperplasia of subcutaneous lymph capillaries after sensing<br />

local Na + or Cl - overload in the interstitium. The sensing function is accomplished by<br />

binding of the transcription factor tonicity-enhancer binding protein (TonEBP) to the<br />

promoter region of the VEGF-C gene. The cells exert their regulatory function by<br />

increasing VEGF-C expression and secretion. Blockade of this VEGF-C response<br />

from macrophages leads to skin electrolyte accumulation and arterial hypertension.<br />

Significance and Background<br />

The importance of a constant milieu intérieur, maintained through extracellular<br />

electrolyte and water content, has long been appreciated for blood pressure control.<br />

The underlying concept is that extracellular fluids readily equilibrate, resulting in a<br />

homogenous and uniform composition of the extracellular fluid space. The<br />

predominant role of the kidney for internal environment composition and blood<br />

pressure control relies on the idea that renal regulation of blood composition is<br />

sufficient for concomitant maintenance of interstitial fluid composition. Any inability of<br />

the body to excrete Na + is thus regarded as a major pathophysiological component<br />

for development of hypertension – and traditionally considered due to perturbations<br />

of renal function (Fig. 1A).<br />

We have previously shown that Na + is stored independent of water in the skin. This<br />

finding resulted in a hypothesis-driven research program in which we searched for<br />

extrarenal mechanisms for regulation of local interstitial electrolyte and water<br />

homeostasis. Approximately 70-80% of the extracellular fluids are interstitial and<br />

thereby not directly controlled by renal salt and water excretion. Therefore,<br />

identification of novel local extrarenal regulatory mechanisms for interstitial fluid<br />

70


homeostasis at the skin tissue level would provide a novel research avenue towards<br />

maintenance of electrolyte and water homeostasis and introduce new potential<br />

therapeutic targets for salt, water, and blood pressure disorders.<br />

We first identified a potential mechanism for storage and release of Na + from the<br />

skin. We have shown that a high-salt diet results in specific changes in the negative<br />

charge density of the extracellular matrix in the skin interstitium. Dietary salt led to<br />

increased glycosaminoglycan (GAG) polymerization and sulfatation in the skin<br />

interstitium. The resulting increase in the negative interstitial charge density was<br />

paralleled with Na + storage in the skin. In reverse, a low-salt diet led to reduced GAG<br />

polymerization and sulfatation, and the resulting decrease in the negative GAG<br />

charge density was paralleled by release of Na + from the skin. We interpreted these<br />

findings in a way that the skin interstitium actively modulated its extracellular GAG<br />

charge density in order to store or release Na + from an interstitial reservoir. Because<br />

Na + would electrostatically interact with GAGs, this storage process could result in<br />

water-free, osmotically inactive Na + storage in the skin. We concluded that the skin<br />

interstitium holds the capacity to locally control its electrolyte composition via<br />

extrarenal regulatory mechanisms.<br />

We then found that Na + storage was additionally paralleled by macrophage infiltration<br />

into the Na + -loaded interstitium (Fig. 1B), where the cells exert an important<br />

extrarenal regulatory cascade for interstitial electrolyte homeostasis and systemic<br />

blood pressure control. In the Na + -loaded skin, macrophages express the<br />

osmoprotective transcription factor TonEBP, suggesting that Na + storage results in<br />

A B<br />

Fig. 1 (A) Traditional research approach for body electrolyte balance and blood pressure<br />

homeostasis, based on the oversimplified concept of passive body fluid equilibrium in closed<br />

systems where known forces are balanced and electrolyte concentrations are not remarkably<br />

different between blood volume and interstitial volume (equilibrium). Suggesting that blood<br />

purification is sufficient for interstitial clearance of electrolytes and water, this research relies on<br />

investigation of renal electrolyte and water clearance mechanisms. (B) Novel research approach for<br />

body electrolyte balance and blood pressure homeostasis, based on the finding that interstitial<br />

electrolyte concentrations are higher than in blood (“skin Na + storage”). Interstitial electrolyte<br />

balance is not achieved by renal blood purification alone, but relies on additional extrarenal<br />

regulatory mechanisms within the skin interstitium. Macrophages act as local osmosensors that<br />

regulate local interstitial electrolyte composition via a TonEBP/VEGF-C dependent mechanism,<br />

enhancing electrolyte clearance via VEGF-C/VEGFR3 mediated modulation of the lymph capillary<br />

network in the skin. We are aware that peripheral vascular resistance in hypertension is increased<br />

throughout the entire body, including obviously the kidneys. Our skin storage mechanism implies an<br />

additional compartment that clearly impacts on general vascular resistance, and does not contradict<br />

the infinite-gain theory that links internal environment composition with blood pressure control.<br />

71


hypertonic interstitial fluid accumulation. TonEBP binds to the promoter of the VEGF-<br />

C gene in macrophages, resulting in gene expression and secretion of VEGF-C into<br />

the interstitial Na + reservoir. VEGF-C binds to vascular endothelial growth factor<br />

receptor 2 (VEGFR2) and increases eNOS expression in blood vessels. VEGF-C<br />

also binds to VEGFR3 on lymph endothelial cells, resulting in hyperplasia of<br />

interstitial lymph capillaries. When we blocked the physiologic macrophage-driven<br />

VEGF-C response experimentally, eNOS expression in blood vessels did not<br />

increase, the lymph capillary network showed no hyperplasia, and blood pressure<br />

was increased. These findings suggest that interstitial skin macrophages regulate<br />

blood pressure via a TonEBP/VEGF-C-dependent mechanism, either by increasing<br />

NO production in small blood vessels, or by enhancing local interstitial electrolyte<br />

clearance in the skin through the lymph capillary network (Fig. 1B).<br />

Our findings have added a new and unexpected research field for hypertension<br />

research. They suggest that skin electrolyte and water homeostasis is important for<br />

blood pressure regulation. We furthermore suggest that macrophages can modulate<br />

skin electrolyte and water metabolism via a TonEBP/VEGF-C regulatory axis, and we<br />

have identified the lymph capillary system in the skin as a new target for modulation<br />

of interstitial electrolyte composition and blood pressure.<br />

Macrophages regulate interstitial electrolyte composition via<br />

TonEBP/VEGF-C driven modulation of lymph capillaries<br />

We have shown that rats, in which we blocked de-novo infiltration of MPS cells into<br />

the skin interstitium with clodronate liposome treatment, were unable to increase<br />

VEGF-C in skin in response to a high-salt diet, and developed salt-sensitive<br />

hypertension. Clodronate liposomes are phagocytized by blood circulatory<br />

macrophages / MPS cells and induce apoptosis. We concluded that increased<br />

TonEBP / VEGF-C expression in skin in response to high salt was induced by<br />

circulatory monocytes/macrophages which entered the Na + overloaded<br />

subcutaneous interstitium. This finding was corroborated by cell culture experiments,<br />

where we showed that macrophages express VEGF-C in response to osmotic stress<br />

by TonEBP binding to the VEGF-C promoter. In additional in-vitro experiments in<br />

cells with TonEBP overexpression or silencing, we furthermore demonstrated that<br />

TonEBP promoter binding was essential for VEGF-C secretion in macrophages in<br />

response to osmotic stress. However, we have so far not yet demonstrated in vivo<br />

that TonEBP is essential for VEGF-C driven modulation of interstitial electrolyte<br />

composition and blood pressure.<br />

To test the hypothesis that TonEBP deficiency results in reduced skin VEGF-C<br />

levels, reduced lymph capillary density, electrolyte accumulation, and blood pressure<br />

increase in vivo, we have designed experiments with TonEBP (gene name: Nfat5)<br />

deficient mice. TonEBP-null mouse embryos die prenatally or late in gestation and<br />

have pronounced renal medullary atrophy and impaired cardiac development. In<br />

72


contrast, studies utilizing haploinsufficient TonEBP mice report normal development.<br />

TonEBP +/- lymphocytes proliferate normal at 330-350 mosmol/kg osmotic stress, and<br />

only when osmolality in cell culture media is >350 mosmol/kg is their proliferation<br />

impaired. No data on renal function in TonEBP +/- mice are available to the best of<br />

our knowledge. Our preliminary data on skin electrolyte composition suggest that<br />

reduced TonEBP expression in skin in TonEBP +/- mice fed a high-salt diet (Fig. 2A)<br />

is paralleled by reduced VEGF-C gene expression and reduced lymph capillary<br />

density in skin (Fig. 2B), which should lead to impaired skin electrolyte clearance.<br />

Indeed, TonEBP mice with high-salt diet were characterized by an almost two-fold<br />

increase in skin Na + and Cl - content<br />

(Fig. 2C), while the ratio of skin<br />

(Na + +K + )-to-water ratio was<br />

unchanged in the mice (Fig. 2D).<br />

We interpret these preliminary<br />

findings that the skin interstitium in<br />

mice is characterized by hypertonic<br />

interstitial fluid accumulation<br />

([Na + +K + ] = 180 mmol/L; Fig. 2D),<br />

and that immune cells are exposed<br />

to osmotic stress when leaving blood<br />

vessels and entering the interstitial<br />

Fig. 2. VEGF-C expression and skin electrolytes in<br />

TonEBP +/- mice. (A) TonEBP mRNA expression in<br />

skin in wild type (wt; n=6) and TonEBP -/+ (n=5)<br />

mice. (B) VEGF-C mRNA expression and average<br />

lymph capillary diameter in wt (n=3) and TonEBP -/+<br />

(n=3) mice fed a high-salt diet. (C & D) Skin Na +<br />

and Cl - content, and skin (Na + +K + )-to-water ratio in<br />

the same mice. * P


its soluble receptor sVEGFR3). This systemic depletion of VEGF-C resulted in<br />

increased skin electrolyte accumulation and blood pressure. We concluded that a<br />

loss of lymph capillary clearance function for skin electrolytes might lead to saltsensitive<br />

hypertension, and that VEGF-C initiates electrolyte clearance from skin<br />

tissue. We tested in an animal model with selective depletion of VEGF-C driven<br />

lymphangiogenesis in the skin whether primary disorders in skin electrolyte and water<br />

metabolism result in experimental arterial hypertension.<br />

We reasoned that mice with over-expression of soluble VEGFR3 (sVEGFR-3)<br />

receptor (gene name: flt-4) under the control of the keratinocyte promoter K14 (K14-<br />

FLT4 mice) may serve as a model to study the effect of targeted overexpression of<br />

sVEGFR3 in the skin on skin electrolyte metabolism and systemic blood pressure.<br />

High-dose overexpression of sVEGFR3 in keratinocytes in mice homozygous for the<br />

transgene leads to<br />

embryonic death. In<br />

contrast, K14 FLT4<br />

mice with<br />

heterozygous<br />

overexpression of<br />

the sVEGFR3 breed<br />

well and show<br />

selective hypoplasia<br />

of subdermal lymph<br />

capillaries with only<br />

discrete skin edema.<br />

In our preliminary<br />

experiments, we<br />

found that a high-salt<br />

diet induced lymph<br />

capillary hyperplasia<br />

in wt mice, while<br />

subdermal lymph<br />

Fig. 3. Selective blockade of the MPS/VEGF-C regulatory axis in the<br />

skin by over-expression of sVEGFR3 by skin keratinocytes (VEGF-C<br />

trap) in K14-FLT4 mice results in exaggerated salt-sensitive<br />

hypertension. (A-D) Subcutaneous lymph capillaries, visualized with a<br />

LYVE-1 specific antibody (green) in wt (n=14) and K14-FLT4 mice<br />

(n=12) fed a low-salt (LSD) or a high-salt (HSD) diet for two weeks. (E)<br />

CD68 and VEGF-C expression in skin in the same mice. β-actin<br />

expression was used as a loading control. (F) eNOS and p eNOS<br />

expression in mouse skin. (G) Lymph capillary density in ear and mean<br />

arterial blood pressure in the mice. * P < 0.05 versus wt/LSD; † P <<br />

0.05 versus K14 Flt4/LSD; ‡ P < 0.05 versus wt/HSD.<br />

capillaries remained hypoplastic in heterozygous K14-FLT4 mice (Fig. 3 A-D),<br />

because increased VEGF-C protein expression by macrophages (Fig. 3E) was<br />

blocked by sVEGFR3 from skin keratinocytes which acted as a skin-specific VEGF-C<br />

trap. In support of this skin specificity, renal macrophage count, VEGF-C mRNA and<br />

protein expression, and lymph capillary density was unchanged in the same animals.<br />

Targeted depletion of subdermal lymphcapillaries resulted in Na + , Cl - , and water<br />

accumulation in skin in K14-FLT4 mice, even with low salt diet. Salt-sensitive<br />

hypertension was augmented in K14-FLT4 mice, and blood pressure tended to be<br />

higher in K14-FLT4 than in wt mice with low-salt diet (Fig. 3G).<br />

We conclude that targeted depletion of skin VEGF-C with hypoplasia of intradermal<br />

lymph capillaries result in skin electrolyte and water retention and blood pressure.<br />

This hypertension comes about an external regulatory mechanism within the skin,<br />

presumably because of reduced electrolyte and water clearance by hypoplastic skin<br />

74


vessels. This animal model underscores the importance of skin lymph capillaries for<br />

skin electrolyte homeostasis and systemic blood pressure control. Perhaps more<br />

importantly, this animal model provides a valuable model that is complementary to<br />

our animal model with pharmacological blockade of VEGF-C/VEGFR3 driven lymph<br />

capillary hyperplasia.<br />

Macrophages control skin electrolyte composition via VEGF-C<br />

driven electrolyte clearance by lymph capillaries<br />

As predicted by our model (Fig. 1B), we have shown previously that blockade of the<br />

macrophage driven VEGF-C response in rats prevents hyperplasia of the<br />

lymphcapillary network with electrolyte accumulation in the skin, inhibits an increase<br />

in eNOS expression in blood vessels, and leads to salt-sensitive hypertension.<br />

However, it has been unclear whether macrophages exert this blood-pressure<br />

buffering effect in response to high-salt diet by VEGF-C/VEGFR2-driven modulation<br />

of vascular eNOS expression (vasodilation), or by VEGF-C/VEGFR3-driven<br />

hyperplasia of interstitial skin lymph capillaries (enhanced electrolyte clearance).<br />

Other investigators have shown previously that selective VEGFR-2 blockade reduces<br />

vascular eNOS expression and increases blood pressure independently of salt<br />

intake.<br />

We have data suggesting that VEGF-C/VEGFR3-driven modulation of the lymph<br />

capillary bed is<br />

decisive for the blood<br />

pressure buffering<br />

mechanism of<br />

macrophages (Fig. 4).<br />

We fed mice<br />

(background: FVB) a<br />

low or a high-salt diet<br />

for 2 consecutive<br />

weeks and also treated<br />

Fig. 4. Selective blockade of the VEGF-C/VEGFR-3 regulatory axis in<br />

the skin by systemic treatment with anti-VEGFR-3 antibody (mF4-<br />

31c1) blocks lymph capillary hyperplasia and results in exaggerated<br />

salt-sensitive arterial hypertension. (A-D) Subcutaneous lymph<br />

capillaries, visualized with a LYVE-1 specific antibody (green) in<br />

untreated (n=20) and mF4-31c1-treated mice (n=24) fed a low-salt<br />

(LSD) or a high-salt (HSD) diet for two weeks. (E) CD68 (macrophage<br />

surface marker) and VEGF-C expression in skin in the same mice. βactin<br />

expression was used as a loading control. (F) eNOS and p<br />

eNOS expression in mouse skin. (G) Lymph capillary density in ear<br />

and mean arterial blood pressure in the mice. * P < 0.05 versus<br />

wt/LSD; † P < 0.05 versus K14 Flt4/LSD; ‡ P < 0.05 versus wt/HSD.<br />

the mice with an anti-<br />

VEGFR3 antibody<br />

(mF4-31c1, 45 μg/g<br />

body weight i.p. every<br />

48 hours). The<br />

resulting blockade of<br />

VEGF-C/VEGFR3driven<br />

lymph capillary<br />

hyperplasia allowed us<br />

to specifically<br />

investigate the role of lymph capillaries for macrophage/VEGF-C-driven blood<br />

75


Fig. 5. Selective blockade of the VEGF-<br />

C/VEGFR-3 regulatory axis in the skin by<br />

systemic treatment with anti-VEGFR-3 antibody<br />

(mF4-31c1) blocks lymph capillary hyperplasia<br />

and results in tissue Cl - retention, which parallels<br />

salt-sensitive hypertension. (A): Relationship<br />

between lymphcapillary density and skin Na +<br />

content relative to tissue dry weight (rSKNa + ).<br />

(B): Relationship between lymphcapillary density<br />

and skin water content relative to tissue dry<br />

weight (rSKW). (C): Relationship between<br />

lymphcapillary density and skin Cl - content<br />

relative to tissue dry weight (rSKCl - ). (D):<br />

Relationship between lymph capillary density in<br />

skin and mean arterial blood pressure (MAP,<br />

intra-arterial measurement and the end of the<br />

experiment). LSD: low-salt diet, HSD: high-salt<br />

diet.<br />

pressure control. We found that mF4-<br />

31c1 did not change lymph capillary<br />

density in low-salt diet (LSD) mice;<br />

however, skin lymph capillary<br />

hyperplasia with high-salt diet (HSD)<br />

was completely prevented with anti-<br />

VEGFR-3 treatment (Fig. 4A-D).<br />

VEGFR-3 blockade was characterized<br />

by increased macrophage recruitment<br />

and increased VEGF-C production in<br />

the skin (Fig. 4E), presumably by a<br />

compensatory effect of the<br />

marcophages to overcome VEGFR3<br />

blockade. As expected, high VEGF-C<br />

levels in both untreated and mF4-<br />

31c1-treated mice with HSD diet<br />

resulted in increased p-eNOS<br />

expression (Fig. 4F). We conclude<br />

that mF4-31c1 treatment selectively<br />

eliminates the macrophage/VEGF-Cdriven<br />

hyperplastic response of<br />

cutaneous lymph capillaries, while<br />

VEGF-C/VEGFR2-driven increase in<br />

eNOS expression in blood vessels<br />

remains intact. This selective<br />

blockade of the hyperplastic response<br />

of lymph capillaries to HSD diet increased mean arterial blood pressure by 19 mmHg<br />

within two weeks (Fig. 4G), despite normal p-eNOS expression (Fig. 4F). The<br />

findings support the idea that macrophages exert their blood-pressure buffering effect<br />

by VEGF-C driven lymphatic regulation of blood pressure.<br />

Lymph capillaries serve as a transit system for rapid movement of interstitial immune<br />

cells into secondary lymphatic tissue, such as lymph nodes. They also serve as a<br />

drainage system of interstitial fluid into the blood stream. Having shown that saltsensitive<br />

hypertension “goes” with subcutaneous lymph capillaries in the skin, we<br />

next tested whether or not blockade VEGF-C/VEGFR3-driven hyperplasia of the<br />

lymph capillary system resulted in increased Na + , K + , Cl - , and water content in the<br />

skin. This finding would support the hypothesis that macrophages induce interstitial<br />

electrolyte clearance via an unknown clearance process within the subcutaneous<br />

lymph capillary system (Fig. 1B). Therefore, we chemically analyzed electrolyte<br />

composition in the skin in the same mice. We found that VEGFR-3 blockade in HSD<br />

mice neither increased Na + (Fig. 5A), nor water content in skin tissue (Fig. 5B).<br />

Instead, VEGFR-3 blockade resulted in a selective increase in skin Cl - content (Fig.<br />

5C). This finding suggests that macrophages selectively influence Cl - metabolism in<br />

the skin via VEGF-C/VEGFR3-driven modulation of lymph capillary function. In<br />

76


Tab. 1. Skin electrolyte and water composition in comparison to<br />

serum electrolyte composition in the same animals. Note that<br />

selective blockade of the VEGF-C/VEGFR-3 regulatory axis in<br />

the skin by systemic treatment with anti-VEGFR-3 antibody<br />

(mF4-31c1) tissue led to Cl - retention and increased Cl -<br />

concentration at the skin tissue level, but not in serum. Skin Na +<br />

content was more stable compared with Cl - . Depending on<br />

intervention, skin Na + content was 2-4 fold higher than Cl -<br />

content, while serum Na + content was exactly 1.2-fold higher<br />

than serum Cl - .<br />

parallel with increased<br />

tissue Cl - content, blood<br />

pressure was elevated<br />

(Fig. 5D), suggesting that<br />

Cl - metabolism is of major<br />

importance for saltsensitive<br />

hypertension.<br />

This finding is in line with<br />

data from humans, where<br />

salt-sensitive<br />

hypertension is mainly<br />

linked with NaCl<br />

ingestion, while similar<br />

NaHCO3 loading does not<br />

increase blood pressure.<br />

Our results bring saltsensitive<br />

hypertension<br />

into a novel biological<br />

context, as we suggest<br />

that hyperplastic lymph capillaries specifically control Cl - clearance from<br />

subcutaneous skin tissue, while hyperplasia of an otherwise intact lymph capillary<br />

system is not coupled with Na + or water clearance. We interpret our data in a way<br />

that skin lymph capillaries apparently do not only collect and pump interstitial fluid,<br />

but also actively modulate its electrolyte concentration. The unknown underlying<br />

concentration mechanism is controlled by VEGF-C from skin macrophages: we found<br />

that modulation of lymph capillary density by anti-VEGFR3 treatment resulted in<br />

increased Cl - concentration in skin tissue, but not in blood (Tab. 1).<br />

2012<br />

Publications (2008-2012)<br />

Original Articles<br />

Kopp C, Linz P, Wachsmuth L, Dahlmann A, Horbach T, Schofl C, Renz W, Santoro D,<br />

Niendorf T, Muller DN, Neininger M, Cavallaro A, Eckardt KU, Schmieder RE, Luft FC, Uder<br />

M, Titze J. (23)Na magnetic resonance imaging of tissue sodium. Hypertension.<br />

2012;59:167-172<br />

77


2011<br />

Slagman MC, Kwakernaak AJ, Yazdani S, Laverman GD, van den Born J, Titze J, Navis G.<br />

Vascular endothelial growth factor c levels are modulated by dietary salt intake in proteinuric<br />

chronic kidney disease patients and in healthy subjects. Nephrol Dial Transplant.<br />

2011;27:978-982.<br />

2010<br />

Machnik A, Dahlmann A, Kopp C, Goss J, Wagner H, van Rooijen N, Eckardt KU, Muller DN,<br />

Park JK, Luft FC, Kerjaschki D, Titze J. Mononuclear phagocyte system depletion blocks<br />

interstitial tonicity-responsive enhance binding protein/vascular endothelial growth factor c<br />

expression and induces salt-sensitive hypertension in rats. Hypertension. 2010;55:755-761.<br />

2009<br />

Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, Park JK, Beck FX,<br />

Muller DN, Derer W, Goss J, Ziomber A, Dietsch P, Wagner H, van Rooijen N, Kurtz A,<br />

Hilgers KF, Alitalo K, Eckardt KU, Luft FC, Kerjaschki D, Titze J. Macrophages regulate saltdependent<br />

volume and blood pressure by a vascular endothelial growth factor-c-dependent<br />

buffering mechanism. Nat Med. 2009;15:545-552<br />

Heer M, Frings-Meuthen P, Titze J, Boschmann M, Frisch S, Baecker N, Beck L. Increasing<br />

sodium intake from a previous low or high intake affects water. Br J Nutr. 2009;101:1286-<br />

1294.<br />

2008<br />

Gratze P, Dechend R, Stocker C, Park JK, Feldt S, Shagdarsuren E, Wellner M, Gueler F,<br />

Rong S, Gross V, Obst M, Plehm R, Alenina N, Zenclussen A, Titze J, Small K, Yokota Y,<br />

Zenke M, Luft FC, Muller DN. Novel role for inhibitor of differentiation 2 in the genesis of<br />

angiotensin ii-induced hypertension. Circulation. 2008;117:2645-2656<br />

Ziomber A, Machnik A, Dahlmann A, Dietsch P, Beck FX, Wagner H, Hilgers KF, Luft FC,<br />

Eckardt KU, Titze J. Sodium-, potassium-, chloride-, and bicarbonate-related effects on blood<br />

pressure and electrolyte homeostasis in deoxycorticosterone acetate-treated rats. Am J<br />

Physiol Renal Physiol. 2008;295:F1752-1763<br />

78


2012<br />

Reviews/Comments<br />

Harrison DG, Marvar PJ, Titze JM. Vascular inflammatory cells in hypertension. Front<br />

Physiol. 2012;3:128.<br />

2010<br />

Titze J, Machnik A. Sodium sensing in the interstitium and relationship to hypertension. Curr<br />

Opin Nephrol Hypertens. 2010 Jul;19(4):385-92.<br />

Titze, J., Muller, D. N., Luft, F. C. Response to Blood Pressure Control: A Facelift for<br />

Macrophages? Hypertension. 2010;56(2): E24-E24<br />

2009<br />

Titze J. “Water-free sodium accumulation”. Semin Dial. 2009 May-Jun;22(3):253-5.<br />

Titze J, Ritz E. Salt and its effect on blood pressure and target organ damage: New pieces in<br />

an old puzzle. J Nephrol. 2009;22:177-189<br />

Titze J. Salz- und Wasserhaushalt: Neue Salzspeicher. Der Nephrologe. 2009;4:488–496<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Agnes Schröder (2009)<br />

Macrophages as regulators of volume and blood pressure homeostasis.<br />

Doctoral theses (Medical Students)<br />

Markus Schafflhuber (2009)<br />

Gycosaminoglycan polymerization and Na + storage in the skin<br />

Christoph Ilies (2010)<br />

Osmotically inactive Na + storage in the skin<br />

Katharina Bauer (2010)<br />

Internal Na + balance in desoxycorticosterone acetate-treated rats<br />

79


Matrix Biology Group<br />

(Dpt. Of Internal Medicine III)<br />

Head: PD Dr. med. Jörg Distler<br />

Address: Department of Internal Medicine III<br />

Krankenhausstr. 12<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 39226<br />

Fax: + 49 (9131) 85 35467<br />

E-mail: joerg.distler@uk-erlangen.de<br />

Christian Beyer, Dr. med. *<br />

* part of the time reported<br />

Head<br />

PD Dr. med. Jörg Distler<br />

Supporting Staff<br />

--<br />

Postdoctoral Fellows<br />

80<br />

Alfiya Akhmetshina, Dr. rer. nat. *


Nicole Reich°<br />

Pawel Zerr<br />

Katrin Palumbo<br />

° PhD received<br />

Maria Halter<br />

Anna Maria Herrmann*<br />

Julia Gebhardt*<br />

* part of the time reported<br />

Jerome Avouac, Dr.<br />

med., Paris<br />

Doctoral Students (Biology)<br />

Clara Dees<br />

Alfiya Akhmetshina°<br />

Ekatherina Skirtladze<br />

Technicians<br />

Guest Scientists<br />

Michal Tomcik, Dr.<br />

med., Prag<br />

81


<strong>Research</strong><br />

Our group is mainly interested in mechanisms of fibroblast activation and in the<br />

development of novel anti-fibrotic approaches. Tissue fibrosis can affect virtually<br />

every organ system and represents a major medicinal problem, because effective<br />

therapies are currently not available. During our time in the <strong>Nikolaus</strong>-<strong>Fiebiger</strong> Center<br />

from 2009 to 2010, we used systemic sclerosis (SSc), a prototypic systemic fibrotic<br />

disease, to investigate the role of the following pathways for fibroblast activation and<br />

fibrosis. We very particularly interested (i) in AP-1 signaling as a downstream<br />

mediator of the pro-fibrotic effects of TGFβ, (ii) in the role of morphogenic pathways<br />

such as hedgehog and notch-signaling in fibrosis and (iii) in cannabinoid receptors as<br />

potential targets for therapeutic approaches in fibrotic diseases.<br />

AP-1 signaling<br />

Transforming growth factor-β (TGF-β) is a key-player in fibrotic diseases. However,<br />

the molecular mechanisms by which TGF-β activates fibroblasts are incompletely<br />

understood. We have previously shown that the expression of the AP-1 transcription<br />

factor Fra-2 is upregulated in SSc and that Fra-2 stimulates the release of<br />

extracellular matrix. We hypothesized that other members of the AP-1 family may<br />

also contribute to the aberrant release of collagen in SSc. Indeed, we observed that<br />

the mRNA as well as the protein levels of three other members of the AP-1 family, c-<br />

Jun, JunD and cFos were strongly increased in fibrotic skin of SSc patients compared<br />

to healthy individuals. The overexpression persisted in cultured fibroblasts from SSc<br />

patients. c-Jun, JunD and cFos were induced by TGF-β in a time- and dosedependent<br />

manner. Moreover, the expression of those AP-1 family members colocalized<br />

with nuclear pSmad 3 in cultured fibroblasts and in fibrotic skin. Inhibition of<br />

canonical Smad signaling by siRNA mediated knockdown of either Smad3 or Smad4<br />

prevented the stimulatory effects of TGF-β and normalized the expression of c-Jun,<br />

JunD and cFos in SSc fibroblasts. To further evaluate the role of AP-1 signaling as a<br />

downstream mediator of the pro-fibrotic effects of TGF-β in vitro and in vivo, we<br />

employed knockout and knockdown strategies as well as selective chemical<br />

82


inhibitors. The small molecule inhibitor T-5224 abrogated the stimulatory effects of<br />

TGF-β on collagen synthesis and differentiation of resting fibroblasts into<br />

metabolically active myofibroblasts. Similar results were obtained upon siRNA<br />

mediated knockdown of cJun, JunD and cFos and in JunD deficient murine<br />

fibroblasts. Moreover, JunD -/- mice were protected from bleomycin induced fibrosis<br />

with reduced dermal thickening, decreased myofibroblast counts and lower collagen<br />

content of lesional skin. AP-1 signaling may also be a potential target for therapeutic<br />

intervention. T-5224, which is currently evaluated in clinical trials for rheumatoid<br />

arthritis, exerted potent anti-fibrotic effects in murine models of SSc in well tolerated,<br />

pharmacologically relevant doses. T-5224 prevented fibrosis in the mouse models of<br />

bleomycin-induced dermal fibrosis and fibrosis induced by overexpression of a<br />

constitutively active TGF-β receptor 1. Treatment with T-5224 dose-dependently<br />

reduced dermal thickening, myofibroblast counts and the collagen content in those<br />

models without toxic effect. Given the central role of AP-1 signaling as a downstream<br />

mediator of TGF- β signaling and the availability of well tolerated, selective inhibitors<br />

such as T-5224, AP-1 signaling might be a promising molecular target for the<br />

treatment of SSc and other fibrotic diseases.<br />

Hedgehog and Notch signaling<br />

Hedgehog and Notch signaling are so-called morphogenic pathways that do not only<br />

play major roles during embryonic development, but that are also essential to<br />

maintain tissue homeostasis in the adult. Aberrant activation of hedgehog- or notch<br />

signaling has both been implicated in the pathogenesis of various diseases in adults,<br />

including a variety of different malignancies. Given their important role in cellular<br />

differentiation, we hypothesized that hedgehog- and notch signaling may contribute<br />

to the activation of fibroblasts and may play a role in tissue fibrosis.<br />

For hedgehog signaling, we demonstrated an overexpression of the ligand sonic<br />

hedgehog (Shh) with accumulation of the downstream transcription factors Gli-1 and<br />

Gli-2 in SSc. The expression of hedgehog target genes such as ptch-1, ptch-2 and<br />

cyclin D were also upregulated, confirming the activation of hedgehog signaling in<br />

SSc. Stimulation of cultured fibroblasts with recombinant Shh induced an activated<br />

phenotype with differentiation of resting fibroblasts into myofibroblasts and increased<br />

release of collagen. Overexpression of Shh in the skin of mice induced fibrosis with<br />

83


accumulation of collagen, dermal thickening and myofibroblast differentiation,<br />

demonstrating that activation of hedgehog signaling alone is sufficient to induce<br />

fibrosis in vivo. Consistent with these findings, mice lacking one allele of the inhibitory<br />

receptor ptch-1 were more sensitive to experimental fibrosis. Conversely, inhibition of<br />

the activating receptor smoothened (Smo) either pharmacologically by the selective<br />

inhibitor LDE223 or by siRNA prevented bleomycin induced dermal fibrosis. Inhibition<br />

of the hedgehog pathway also exerted potent anti-fibrotic effects in the tight skin 1<br />

mouse model.<br />

We also demonstrated a role of aberrant notch signaling in the pathogenesis of SSc.<br />

Notch signaling was activated in SSc in vivo as well as in cultured SSc fibroblasts in<br />

vitro as shown by overexpression of the notch ligand Jagged-1 (Jag-1) and by<br />

elevated transcription of notch target genes such as hes-1. Stimulation of healthy<br />

dermal fibroblasts with the Jag-1-Fc induced an SSc-like phenotype with expression<br />

of contractile proteins and increased release of collagen. Inhibition of Notch signaling<br />

by the γ-secretase inhibitor DAPT or by siRNA mediated knockdown of Notch-1<br />

efficiently reduced the release of collagen from SSc fibroblasts. Overexpression of a<br />

Notch antisense construct or treatment with DAPT also prevented bleomycin-induced<br />

fibrosis and fibrosis in tight-skin 1 mice. Moreover, targeting Notch signaling resulted<br />

in almost complete regression of pre-established experimental fibrosis. Thus, we<br />

demonstrated that Notch signaling plays an important role in fibroblast activation and<br />

tissue fibrosis.<br />

Collectively, our results describe a central role of hedgehog and notch signaling for<br />

fibroblast activation in fibrosis. These findings might have direct translational<br />

implications because inhibitors of Smo as well as γ-secretase inhibitors are available<br />

and were well tolerated in clinical trials of cancer patients.<br />

Cannabinoid receptors<br />

Cannabinoids are derivates of the marijuana component Δ 9 -tetrahydrocannabinol that<br />

exert their effects via CB1 and CB2 receptors. Cannabinoids can be subdivided into<br />

three different groups according to their origin. The family of cannabinoids includes<br />

plant derived cannabinoids, synthetic cannabinoids and endogenous cannabinoids<br />

(endocannabinoids) that are synthesized within the human body. Besides their<br />

effects in the CNS, endocannabinoids regulate physiological and pathophysiological<br />

processes in non-neuronal tissues. Recent data demonstrated that cannabinoids<br />

84


egulate the activation of hepatic stellate cells, which contribute to the pathogenesis<br />

of liver fibrosis. We therefore aimed to evaluate the role of cannabinoids and their<br />

receptors in fibroblast activation and evaluate targeting of cannabinoid receptors as a<br />

therapeutic approach in fibrotic diseases.<br />

We demonstrated that mice deficient for CB1 (CB1 -/- ) were protected from<br />

bleomycin-induced dermal fibrosis with reduced dermal thickening, hydroxyproline<br />

content and myofibroblast counts. Inactivation of CB1 decreased the number of<br />

infiltrating T cells and macrophages in lesional skin. In contrast, activation of CB1<br />

with the selective small molecule inhibitor ACEA increased leukocyte infiltration and<br />

enhanced the fibrotic response to bleomycin. The phenotype of CB1 -/- mice was<br />

mimicked by transplantation of CB1 -/- bone marrow into CB1 +/+ mice, demonstrating<br />

that leukocytes mediate the pro-fibrotic effects of CB1. Consistently, knockdown of<br />

CB1 did not prevent fibrosis in the inflammation-independent Tsk-1 model.<br />

In contrast to CB1 -/- mice, CB2 -/- mice were more sensitive to bleomycin induced<br />

dermal fibrosis. Leukocytes counts were significantly higher in lesional skin of CB2 -/-+<br />

mice than CB2 +/+ mice. Treatment with the CB2 antagonist AM-630 also increased<br />

dermal fibrosis, whereas the CB2 agonist JWH-133 reduced leukocyte infiltration and<br />

dermal fibrosis. As for CB1, the altered sensitivity of CB2 -/- mice to fibrosis was<br />

mediated by bone marrow derived cells rather than by resident fibroblasts, because<br />

the phenotype of CB2 -/- mice was mimicked by transplantation of CB2 -/- bone<br />

marrow into CB2 +/+ mice, but not by CB2 -/- mice transplanted with bone marrow from<br />

CB2 +/+ mice. Consistent with this hypothesis, neither knockdown or inhibition, nor<br />

activation of CB1 or CB2 in cultured fibroblasts altered fibroblast activation or the<br />

release of collagen.<br />

We demonstrated that CB1 and CB2 have opposing effects in fibrosis. CB1<br />

stimulates leukocyte infiltration and enhances tissue fibrosis, whereas CB2 limits<br />

leukocyte influx and ameliorates experimental dermal fibrosis. Since selective CB1<br />

antagonists as well as selective CB2 agonists are available, the endocannabiod<br />

system and both of its receptors might be interesting molecular targets for the<br />

treatment of early inflammatory stages of fibrotic diseases, whereas they are unlikely<br />

to be effective in later, less inflammatory stages with predominant endogenous<br />

activation of fibroblasts.<br />

85


Original Articles<br />

Publications (2009-2010)<br />

1. Balistreri E, Garcia-Gonzalez E, Selvi E, Akhmetshina A, Palumbo K,<br />

Lorenzini S, Maggio R, Lucatelli M, Galeazzi M, Distler J. The cannabinoid<br />

WIN55, 212-2 abrogates dermal fibrosis in scleroderma bleomycin model. Ann<br />

Rheum Dis. 2010 Dec 21.<br />

2. Dieudé P, Guedj M, Truchetet ME, Wipff J, Revillod L, Riemekasten G,<br />

Matucci-Cerinic M, Melchers I, Hachulla E, Airo P, Diot E, Hunzelmann N,<br />

Mouthon L, Cabane J, Cracowski JL, Riccieri V, Distler J, Amoura Z, Valentini<br />

G, Camaraschi P, Tarner I, Frances C, Carpentier P, Brembilla NC, Meyer O,<br />

Kahan A, Chizzolini C, Boileau C, Allanore Y. Association of the CD226<br />

307Ser variant with systemic sclerosis: Evidence for a contribution of costimulation<br />

pathways in SSc pathogenesis. Arthritis Rheum. 2010 Dec 15.<br />

3. Dieudé P, Guedj M, Wipff J, Ruiz B, Riemekasten G, Airo P, Melchers I,<br />

Hachulla E, Cerinic MM, Diot E, Hunzelmann N, Caramaschi P, Sibilia J, Tiev<br />

K, Mouthon L, Riccieri V, Cracowski JL, Carpentier PH, Distler J, Amoura Z,<br />

Tarner I, Avouac J, Meyer O, Kahan A, Boileau C, Allanore Y. NLRP1<br />

influences the systemic sclerosis phenotype: a new clue for the contribution of<br />

innate immunity in systemic sclerosis-related fibrosing alveolitis pathogenesis.<br />

Ann Rheum Dis. 2010 Dec 13.<br />

4. Polzer K, Neubert K, Meister S, Frey B, Baum W, Distler JH, Gückel E, Schett<br />

G, Voll RE, Zwerina J. Proteasome inhibition aggravates TNF-mediated bone<br />

resorption. Arthritis Rheum. 2010 Dec 6.<br />

5. Avouac J, Fürnrohr BG, Tomcik M, Palumbo K, Zerr P, Horn A, Dees C,<br />

Akhmetshina A, Beyer C, Distler O, Schett G, Allanore Y, Distler JH.<br />

Inactivation of the transcription factor STAT4 prevents inflammation-driven<br />

fibrosis in systemic sclerosis animal models. Arthritis Rheum. 2010 Nov 30.<br />

6. Heiland GR, Zwerina K, Baum W, Kireva T, Distler JH, Grisanti M, Asuncion F,<br />

Li X, Ominsky M, Richards W, Schett G, Zwerina J. Neutralisation of Dkk-1<br />

protects from systemic bone loss during inflammation and reduces sclerostin<br />

expression. Ann Rheum Dis. 2010 Sep 21.<br />

7. Weber CK, Haslbeck M, Englbrecht M, Sehnert B, Mielenz D, Graef D, Distler<br />

JH, Mueller RB, Burkhardt H, Schett G, Voll RE, Fürnrohr BG. Antibodies to<br />

the endoplasmic reticulum-resident chaperones calnexin, BiP and Grp94 in<br />

patients with rheumatoid arthritis and systemic lupus erythematosus.<br />

Rheumatology (Oxford). 2010 Aug 17.<br />

8. Dallos T, Heiland GR, Strehl J, Karonitsch T, Gross W, Moosig F, Holl-Ulrich<br />

C, Distler JH, Manger B, Schett G, Zwerina J. Chemokine CCL17/TARC in<br />

Churg-Strauss syndrome. Arthritis Rheum. 2010 Jul 28.<br />

86


9. Marquart S, Zerr P, Akhmetshina A, Palumbo K, Reich N, Tomcik M, Horn A,<br />

Dees C, Engel M, Zwerina J, Distler O, Schett G, Distler JH. Inactivation of the<br />

cannabinoid receptor CB1 prevents leukocyte infiltration and experimental<br />

fibrosis. Arthritis Rheum. 2010 Jul 8.<br />

10. Wipff J, Dieudé P, Guedj M, Ruiz B, Riemekasten G, Cracowski J, Matucci-<br />

Cerinic M, Melchers I, Humbert M, Hachulla E, Airo P, Diot E, Hunzelmann N,<br />

Caramaschi P, Sibilia J, Valentini G, Tiev K, Girerd B, Mouthon L, Riccieri V,<br />

Carpentier P, Distler JH, Amoura Z, Tarner I, Degano B, Avouac J, Meyer O,<br />

Kahan A, Boileau C, Allanore Y. Association of KCNA5 gene polymorphism<br />

with systemic sclerosis-associated pulmonary arterial hypertension in the<br />

European Caucasian population. Arthritis Rheum. 2010 Jun 15.<br />

11. Avouac J, Walker U, Tyndall A, Kahan A, Matucci-Cerinic M, Allanore Y;<br />

EUSTAR, Miniati I, Muller A, Iannone F, Distler O, Becvar R, Sierakowsky S,<br />

Kowal-Bielecka O, Coelho P, Cabane J, Cutolo M, Shoenfeld Y, Valentini G,<br />

Rovensky J, Riemekasten G, Vlachoyiannopoulos P, Caporali R, Jiri S, Inanc<br />

M, Zimmermann Gorska I, Carreira P, Novak S, Czirjak L, Oliveira Ramos F,<br />

Jendro M, Chizzolini C, Kucharz EJ, Richter J, Cozzi F, Rozman B, Mallia CM,<br />

Gabrielli A, Farge D, Kiener HP, Schöffel D, Airo P, Wollheim F, Martinovic D,<br />

Trotta F, Jablonska S, Reich K, Bombardieri S, Siakka P, Pellerito R, Bambara<br />

LM, Morovic-Vergles J, Denton C, Hinrichs R, Van den Hoogen F, Damjanov<br />

N, Kötter I, Ortiz V, Heitmann S, Krasowska D, Seidel M, Hasler P, Van Laar<br />

JM, Kaltwasser JP, Foeldvari I, Juan Mas A, Bajocchi G, Wislowska M,<br />

Pereira Da Silva JA, Jacobsen S, Worm M, Graniger W, Kuhn A, Stankovic A,<br />

Cossutta R, Majdan M, Damjanovska Rajcevska L, Tikly M, Nasonov EL,<br />

Steinbrink K, Herrick A, Müller-Ladner U, Dinc A, Scorza R, Sondergaard K,<br />

Indiveri F, Nielsen H, Szekanecz Z, Silver RM, Antivalle M, Espinosa IB,<br />

García de la Pena Lefebvre P, Midtvedt O, Launay D, Valesini F, Tuvik P,<br />

Ionescu RM, Del Papa N, Pinto S, Wigley F, Mihai C, Sinziana Capranu M,<br />

Sunderkötter C, Jun JB, Alhasani S, Distler JH, Ton E, Soukup T, Seibold J,<br />

Zeni S, Nash P, Mouthon L, De Keyser F, Duruöz MT, Cantatore FP, Strauss<br />

G, von Mülhen CA, Pozzi MR, Eyerich K, Szechinski J, Keiserman M,<br />

Houssiau FA, Román-Ivorra JA, Krummel-Lorenz B, Aringer M, Westhovens<br />

R, Bellisai F, Mayer M, Stoeckl F, Uprus M, Volpe A, Buslau M, Yavuz S,<br />

Granel B, Valderílio Feijó A, Del Galdo F, Popa S, Zenone T, Ricardo<br />

Machado X, Pileckyte M, Stebbings S, Mathieu A, Tulli A, Tourinho T, Souza<br />

R, Acayaba de Toledo R, Stamp L, Solanki K, Veale D, Francisco Marques<br />

Neto J, Bagnato GF, Loyo E, Toloza S, Li M, Ahmed Abdel Atty Mohamed W,<br />

Cobankara V, Olas J, Salsano F, Oksel F, Tanaseanu CM, Foti R, Ancuta C,<br />

Vonk M, Caramashi P, Beretta L, Balbir A, Chiàla A, Pasalic Simic K, Ghio M,<br />

Stamenkovic B, Rednic S, Host N, Pellerito R, Hachulla E, Furst DE.<br />

Characteristics of joint involvement and relationships with systemic<br />

inflammation in systemic sclerosis: results from the EULAR Scleroderma Trial<br />

and <strong>Research</strong> Group (EUSTAR) database. J Rheumatol. 2010 Jul;37(7):1488-<br />

501. Epub 2010 Jun 15.<br />

12. Dieudé P, Guedj M, Wipff J, Ruiz B, Riemekasten G, Matucci-Cerinic M,<br />

Melchers I, Hachulla E, Airo P, Diot E, Hunzelmann N, Cabane J, Mouthon L,<br />

Cracowski JL, Riccieri V, Distler J, Meyer O, Kahan A, Boileau C, Allanore Y.<br />

Association of the TNFAIP3 rs5029939 variant with systemic sclerosis in the<br />

European Caucasian population. Ann Rheum Dis. 2010 May 28.<br />

87


13. Krönke G, Uderhardt S, Kim KA, Stock M, Scholtysek C, Zaiss MM, Surmann-<br />

Schmitt C, Luther J, Katzenbeisser J, David JP, Abdollahi-Roodsaz S, Tran K,<br />

Bright JM, Binnerts ME, Akhmetshina A, Böhm C, Distler JH, Joosten LA,<br />

Schett G, Abo A. R-spondin1 protects against inflammatory bone damage<br />

during murine arthritis by modulating the wnt pathway. Arthritis Rheum. 2010<br />

Apr 6.<br />

14. Reich N, Beyer C, Gelse K, Akhmetshina A, Dees C, Zwerina J, Schett G,<br />

Distler O, Distler JH. Microparticles stimulate angiogenesis by inducing ELR(+)<br />

CXC-chemokines in synovial fibroblasts. J Cell Mol Med. 2010 Mar 8.<br />

15. Maurer B, Stanczyk J, Jüngel A, Akhmetshina A, Trenkmann M, Brock M,<br />

Kowal-Bielecka O, Gay RE, Michel BA, Distler JH, Gay S, Distler O. miR-29 is<br />

a key regulator of collagen expression in systemic sclerosis. Arthritis Rheum.<br />

2010 Mar 3.<br />

16. Akhmetshina A, Beer J, Polzer K, Englbrecht M, Palumbo K, Dees C, Reich N,<br />

Zwerina J, Szucs G, Gusinde J, Nevskaya T, Distler O, Kerjaschki D, Schett<br />

G, Distler JH. Decreased lymphatic vessel counts in systemic sclerosis -<br />

Association with fingertip ulcers. Arthritis Rheum. 2010 Feb 12.<br />

17. Reich N, Maurer B, Akhmetshina A, Venalis P, Dees C, Zwerina J, Nevskaja<br />

T, Schett G, Distler JH. The transcription factor fos-related antigen-2 (Fra-2)<br />

regulates the production of extracellular matrix in systemic sclerosis. Arthritis<br />

Rheum 2010 Jan;62(1):280-90.<br />

18. Functional autoantibodies against serpin E2 in rheumatoid arthritis.<br />

Maciejewska-Rodrigues H, Al-Shamisi M, Hemmatazad H, Ospelt C, Bouton<br />

MC, Jäger D, Cope AP, Charles P, Plant D, Distler JH, Gay RE, Michel BA,<br />

Knuth A, Neidhart M, Gay S, Jüngel A.. Arthritis Rheum. 2010 Jan;62(1):93-<br />

104.<br />

19. Maurer B, Busch N, Jüngel A, Pileckyte M, Gay RE, Michel BA, Schett G, Gay<br />

S, Distler JH, Distler O. Fra-2 induces progressive peripheral vasculopathy in<br />

mice closely resembling human systemic sclerosis. Circulation. 2009 Nov 23.<br />

20. Kurowska-Stolarska M, Distler JH, Jüngel A, Rudnicka W, Neumann E, Pap T,<br />

Wenger RH, Michel BA, Müller-Ladner U, Gay RE, Maslinski W, Gay S and<br />

Distler O. Id-2 induced by hypoxia promotes synovial fibroblast-dependent<br />

osteoclastogenesis. Arthritis Rheum. 2009 Nov 30;60(12):3663-3675.<br />

21. Ruiz Heiland G, Aigner E, Dallos T, Sahinbegovic E, Krenn V, Thaler C, Weiss<br />

G, Distler JH, Datz C, Schett G, Zwerina J. Synovial immunopathology in<br />

hemochromatosis arthropathy. Ann Rheum Dis. 2009 Nov 23.<br />

22. Dieudé P, Wipff J, Guedj M, Ruiz B, Melchers I, Hachulla E, Riemekasten G,<br />

Diot E, Hunzelmann N, Sibilia J, Tiev K, Mouthon L, Cracowski JL, Carpentier<br />

PH, Distler J, Amoura Z, Tarner I, Avouac J, Meyer O, Kahan A, Boileau C,<br />

Allanore Y. BANK1 is a genetic risk factor for diffuse cutaneous systemic<br />

sclerosis and has additive effects with IRF5 and STAT4. Arthritis Rheum. 2009<br />

Nov;60(11):3447-54.<br />

88


23. Mieliauskaite D, Venalis P, Dumalakiene I, Venalis A, Distler J. Relationship<br />

between serum levels of TGF-beta1 and clinical parameters in patients with<br />

rheumatoid arthritis and Sjögren's syndrome secondary to rheumatoid arthritis.<br />

Autoimmunity. 2009 May;42(4):356-8.<br />

24. Guiducci S, Distler JH, Milia AF, Miniati I, Rogai V, Manetti M, Falcini F, Ibba-<br />

Manneschi L, Gay S, Distler O, Matucci-Cerinic M. Stiff skin syndrome:<br />

evidence for an inflammation-independent fibrosis? Rheumatology (Oxford).<br />

2009 May 25.<br />

25. Hemmatazad H, Rodrigues HM, Maurer B, Brentano F, Pileckyte M, Distler<br />

JH, Gay RE, Michel BA, Gay S, Huber LC, Distler O, Jüngel A. Histone<br />

deacetylase 7, a potential target for the antifibrotic treatment of systemic<br />

sclerosis. Arthritis Rheum. 2009 May;60(5):1519-29.<br />

26. Akhmetshina A, Dees C, Busch N, Beer J, Zimmer A, Distler O, Schett G,<br />

Distler JH. The cannabinoid receptor CB2 exerts anti-fibrotic effects in<br />

experimental dermal fibrosis. Arthritis Rheum, 2009 Apr;60(4):1129-36.<br />

27. Polzer K, Joosten L, Gasser J, Distler JH, Ruiz G, Baum W, Redlich K,<br />

Bobacz K, Smolen JS, van den Berg W, Schett G, Zwerina J. IL-1 is essential<br />

for systemic inflammatory bone loss. Ann Rheum Dis. 2009 Feb 5.<br />

28. Akhmetshina A, Venalis P, Dees C, Busch N, Zwerina J, Jüngel A, Schett G,<br />

Distler O, Distler JH. Treatment with imatinib does not only prevent fibrosis in<br />

different preclinical models of SSc, but also induces regression of established<br />

fibrosis. Arthritis Rheum 2009 Jan;60(1):219-24.<br />

29. Hanitsch LG, Burmester GR, Witt C, Hunzelmann N, Genth E, Krieg T,<br />

Lehmacher W, Melchers I, Meurer M, Müller-Ladner U, Schulze-Lohoff E,<br />

Becker M, Sunderkoetter C; Worm M, Klaus P, Rubbert A, Steinbrink K,<br />

Grundt B, Hein R, Scharffetter-Kochanek K, Hinrichs R, Walker K, Szeimies<br />

RM, Karrer S, Müller A, Seitz C, Schmidt E, Lehmann P, Foeldvári I,<br />

Reichenberger F, Gross L, Kuhn A, Haust M, Reich K, Böhm M, Saar P,<br />

Fierlbeck G, Kötter I, Lorenz HM, Blank N, Gräfenstein K, Juche A, Aberer E,<br />

Bali G, Fiehn C, Stadler R, Bartels V, Buslau M, Distler J, Sticherling M,<br />

Riemekasten G. Skin sclerosis is only of limited value to identify SSc patients<br />

with severe manifestations--an analysis of a distinct patient subgroup of the<br />

German Systemic Sclerosis Network (DNSS) Register. Rheumatology<br />

(Oxford). 2009 Jan;48(1):70-3.<br />

89


Reviews<br />

1. Iwamoto N, Distler JH, Distler O. Tyrosine Kinase Inhibitors in the Treatment<br />

of Systemic Sclerosis: From Animal Models to Clinical Trials. Curr Rheumatol<br />

Rep. 2010 Nov 2.<br />

2. Beyer C, Schett G, Distler O, Distler JH. Animal Models of Systemic Sclerosis:<br />

Prospects and Limitations. Arthritis Rheum. 2010 Jul 8.<br />

3. Beyer C, Distler JH, Distler O. Are tyrosine kinase inhibitors promising for the<br />

treatment of systemic sclerosis and other fibrotic diseases? Swiss Med Wkly.<br />

2010 Apr 26.<br />

4. Distler JH and Distler O. Tyrosine kinase inhibitors for the treatment of fibrotic<br />

diseases such as systemic sclerosis: towards molecular targeted therapies.<br />

Ann Rheum Dis. 2010 Jan;69 Suppl 1:i48-51.<br />

5. Beyer C and Distler JH. The scientific basis for novel treatments of systemic<br />

sclerosis. f1000 Medicine <strong>Report</strong>s 2009, 1:95.<br />

6. Distler JH, Beyer C, Schett G, Lüscher TF, Gay S, Distler O. Endothelial<br />

Progenitor Cells - Novel players in the pathogenesis of rheumatic diseases.<br />

Arthritis Rheum. 2009 Nov;60(11):3168-79.<br />

7. Abraham DJ, Krieg T, Distler J, Distler O. Overview of pathogenesis of<br />

systemic sclerosis. Rheumatology (Oxford). 2009 Jun;48 Suppl 3:iii3-7.<br />

8. Beyer C, Schett G, Gay S, Distler O, Distler JH. Hypoxia in the pathogenesis<br />

of systemic sclerosis. Arthritis Res Ther. 2009;11(2):220. Epub 2009 Apr 21.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Alfiya Akhmetshina, Nicole Busch, Michal Tomcik (PhD part time in Erlangen),<br />

Jerome Avouac (PhD part time in Erlangen)<br />

Doctoral theses (Molecular Medicine)<br />

Sieglinde Marquart, Constantin Huhn, Tobias Strapatsas, Christina Bergmann, Michael<br />

Balzer<br />

90


Laboratory of Dendritic Cell Biology<br />

(Department of Dermatology)<br />

Head: Diana Dudziak, Dr. rer. nat.<br />

Professor of Dendritic Cell Biology<br />

Address: Laboratory of Dendritic Cell Biology<br />

Department of Dermatology<br />

Hartmannstr. 14<br />

D-91052 Erlangen<br />

Telephone: + 49 (9131) 85 39346<br />

Fax: + 49 (9131) 85 39347<br />

E-mail: diana.dudziak@uk-erlangen.de<br />

Homepage: http://www.hautklinik.ukerlangen.de/e1585/e1893/e2171/index_ger.html<br />

Head<br />

Diana Dudziak, Prof. Dr. rer. nat.<br />

Professor of Dendritic Cell Biology<br />

Postdoctoral Fellows<br />

Kirsten Neubert, Dr. rer. nat.<br />

Nathalie Eissing, Dr. med. vet.<br />

Doctoral Students (Biology)<br />

Gordon Heidkamp<br />

Christian Lehmann<br />

Anna Baranska<br />

Lukas Heger<br />

91


* PhD received<br />

Doctoral Students (Medicine)<br />

Kirsten Ehrenspeck<br />

Nathalie Eissing*<br />

Yang Jiao<br />

Technicians<br />

Simone Beck<br />

Christina Weiss<br />

92


<strong>Research</strong><br />

Our research is focussed on the function of Dendritic cell subpopulations in the<br />

process of induction of immunity and maintenance of peripheral tolerance. Dendritic<br />

cells are very important antigen presenting cells. We are especially interested in the<br />

analysis of antigen uptake, antigen processing and antigen presentation by Dendritic<br />

cells, if and how Dendritic cell subpopulations differ in antigen processing and<br />

presentation, and how Dendritic cell subpopulations migrate in the lymphoid tissues<br />

when they have encountered their antigens under immunizing and tolerizing<br />

conditions. Moreover, to understand antigen processing and presentation<br />

mechanisms we focus on the responding T cells. Depending on the Dendritic cell<br />

subpopulation we found that different Dendritic cell subpopulations can induce<br />

different T cell responses in vivo (Dudziak et al., 2007). These findings will have<br />

important implications in the development of new therapeutic vaccines.<br />

Dendritic cells and antigen targeting<br />

Dendritic cells maintain peripheral tolerance and induce immunity<br />

The immune system controls invading pathogenic organisms by innate and adaptive<br />

immune mechanisms. To enable recognition of virtually any antigen, developing B<br />

and T cells randomly rearrange their receptors to produce unique clones with a wide<br />

variety of specificities. It is well accepted that this also leads to the generation of<br />

receptors that can recognize ‘self’ antigens. By central tolerance mechanisms at<br />

several checkpoints self reactive B cells are deleted in the bone marrow whereas self<br />

reactive T cells undergo negative selection mechanisms in the thymus where they<br />

are either deleted or undergo anergic mechanisms. As some of the T cells can<br />

escape negative selection in the thymus, checkpoints in the periphery have to exist to<br />

prevent autoimmune reactions by those potentially self-reactive T cells. Only recently<br />

it became clear that DCs which are essential for the induction of adaptive immune<br />

responses are also key players not only for the maintenance of central tolerance but<br />

also for the maintenance of peripheral tolerance.<br />

Dendritic cells (DCs) are the most important antigen presenting cells in the immune<br />

system. In non-lymphoid organs DCs act as sentinels and take up and process<br />

antigens from the periphery (skin, airways, intestine, interstitial spaces of various<br />

organs). Antigen loaded DCs migrate to the lymphoid tissues and interact with<br />

antigen-specific T cells. Besides the uptake of soluble antigens, DCs are also able to<br />

ingest apoptotic cells, tumor cells, or infected cells to present their antigen. The<br />

induction of peripheral tolerance or immunity depends on the DC differentiation state.<br />

In the steady-state immature DCs express low levels of CD80, CD86 or MHC class II<br />

molecules on their cell surface. Recognition of the MHC peptide complex by a T cell<br />

in the steady-state causes deletion or anergy of self reactive T cells after an initial<br />

phase of proliferation. This process is resulting in peripheral tolerance (Dudziak et al.,<br />

2007, Dudziak, 2009).<br />

In contrast, during a microbial or viral infection antigen uptake is paralleled by DC<br />

activation and maturation. In this process pattern-recognition receptors (PRRs)<br />

expressed on DCs, such as Toll-like receptors (TLRs) and proteins of the C-type<br />

lectin receptor family, recognize conserved parts of pathogens such as lipids, sugar<br />

93


esidues, or nucleic acids. This stimulation causes upregulation of cell surface<br />

activation markers (e.g. CD80, CD83, and CD86) and expression of cytokines and<br />

chemokines such as TNFα, IL6, IL12 or Rantes 9 (Dudziak et al., 2005, Dudziak,<br />

2009). Under these circumstances T cells that recognize bacterial or viral antigens<br />

become activated, clonally expand and develop into memory T cells (Dudziak et al.,<br />

2007; Dudziak, 2009).<br />

Production of antigen carrying recombinant antibodies against different<br />

Dendritic cell subpopulations<br />

Diana Dudziak, Gordon Heidkamp, Nathalie Eissing, Kirsten Ehrenspeck<br />

At least 6 different subtypes of murine DCs can be distinguished based on their<br />

localization and expression of cell surface molecules (Fig. 1, Dudziak, 2009). We are<br />

only just beginning to understand why such a great variety of DC subtypes has<br />

evolved and what their precise role is in the maintenance of peripheral tolerance or in<br />

the induction of immunity. The two main subpopulations of conventional DCs in the<br />

spleen can be distinguished by the expression of CD11c and CD8 as CD11c + CD8 -<br />

(myeloid) and CD11c + CD8 + (lymphoid) DCs (Dudziak, 2009). We found that these<br />

DC subpopulations express different endocytosis receptors and C-type lectin<br />

receptors on their cell surface, e.g. CD11c + CD8 - DCs express DCIR2 whereas<br />

CD11c + CD8 + DCs express DEC205 (CD205) (Fig. 1, Dudziak et al., 2007; Soares et<br />

al., 2007; Yamazaki et al., 2008; Do et al., 2010, O’Kamphorst et al., 2010; Loschko<br />

et al., 2011(1); Loschko et al., 2011(2)). We could show that DCIR2 and DEC205<br />

were rapidly internalized after interaction with antibodies against these endocytosis<br />

receptors (Dudziak et al., 2007). In addition to the internalization of the receptors we<br />

found that also the bound antibodies were internalized into the DCs. These findings<br />

let us to the idea of producing recombinant endocytosis receptor antibodies that are<br />

genetically fused with an antigen of choice.<br />

94<br />

Figure 1. Murine splenic<br />

DCs. A) Overview of<br />

different murine DC subpopulations<br />

and their localization.<br />

B) Immunofluorescence<br />

of murine spleen<br />

showing B cells (B220),<br />

CD11c + CD8 - (DCIR2/<br />

33D1 + ) in the bridging area<br />

and red pulp and<br />

CD11c + CD8 + (DEC205 + )<br />

DCs in the T cell area. C)<br />

FACS analysis of CD11c +<br />

DCs in murine spleen and<br />

the expression of CD8,<br />

DEC205 and DCIR2/33D1.<br />

(Dudziak et al., 2009)


We cloned the variable regions of the DEC205 and the 33D1 rat antibodies (the latter<br />

we identified by microarray analysis and expression cloning to be DCIR2) and fused<br />

them by overlap PCR to the constant regions of the heavy and light chain of murine<br />

IgG1. The Fc region of the murine IgG1 was mutated and therefore binding to Fc<br />

receptors is inhibited. In the heavy chain we inserted the Ovalbumin antigen (and for<br />

some experiments Hen egg lysozyme) as model antigen of choice. The light and<br />

Ovalbumin carrying heavy chains of DEC205 and 33D1 antibodies were transiently<br />

transfected into 293T cells and recombinant antibody containing supernatants were<br />

enriched with protein G. We found that the recombinant antibodies DEC205-Ova and<br />

33D1/DCIR2-Ova were binding to the DC subsets CD11c + CD8 + or CD11c + CD8 - ,<br />

respectively, in vivo and in vitro, were internalized and thus were therefore ready for<br />

antigen targeting experiments (Dudziak et al., 2007). Currently we are producing new<br />

antibodies for future targeting of other DC subpopulations in vivo.<br />

Differential Antigen processing and presentation by Dendritic cell<br />

subpopulations<br />

Diana Dudziak, Anna Baranska, Gordon Heidkamp, Kirsten Ehrenspeck<br />

To analyze if DC subpopulations can be targeted with our recombinant Ovalbumin<br />

carrying antibodies we injected C57BL/6 mice with 33D1-Ova and DEC205-Ova<br />

antibodies to analyze how the different DC subpopulations uptake, process and<br />

present the antigens to T cells. In our experiments we are using transgenic mice that<br />

contain T cells that express either a T cell receptor that recognizes Ovalbumin<br />

peptide when it is presented as peptide MHCI complex or transgenic mice whose T<br />

cell receptor recognizes Ovalbumin antigen when it is presented as peptide MHCII<br />

complex on the surface of antigen presenting DCs. Therefore, both cytotoxic and T<br />

helper cell responses can be analyzed in vivo. First, we performed in vivo antigen<br />

targeting and in vitro T cell stimulation experiments with transgenic Ovalbumin<br />

specific T cells. After antigen targeting of only 10μg DEC205-Ova, or 33D1/DCIR2-<br />

Ova (normally, uncoupled Ovalbumin-protein needs to be injected as 1000-5000<br />

μg/mouse to receive T cell responses) we found, that delivering Ovalbumin antigens<br />

with DEC205-Ova to CD11c + CD8 + DCs induces a strong CD8 T cell response and<br />

antigen targeting with 33D1/DCIR2-Ova to CD11c + CD8 - DCs induced a more<br />

prominent CD4 T cell response in vitro.<br />

Further, we analyzed how T cells respond to antigen loaded DC subpopulations in<br />

vivo. Within 3 days we found that CD4 or CD8 transgenic T cells showed a<br />

proliferative response after interaction with antigen loaded DCs in vivo. We found that<br />

with less than 30 ng of the recombinant antibodies we were able to induce a CD8 T<br />

cell response when DEC205-Ova antibodies were targeted to CD11c + CD8 + DCs,<br />

whereas when we targeted 33D1-Ova to CD11c + CD8 - DCs in vivo we were able to<br />

induce a strong CD4 T cell response in vivo. By microarray analysis and the use of<br />

transgenic mice that express human DEC205 on all DC subpopulations in vivo we<br />

were able to demonstrate that the DC subpopulations were different in antigen<br />

processing and presentation and that the differences were not due to the receptors<br />

we have had targeted in vivo. Thus, we can conclude that CD11c + CD8 + DCs excel in<br />

antigen presentation as peptide MHCI complexes whereas CD11c + CD8 - DCs excel in<br />

antigen presentation on MHCII in the steady state.<br />

95


Targeting antigens under immunizing conditions induced a strong prolonged<br />

proliferative response in either CD4 transgenic T cells (targeting CD11c + CD8 - with<br />

33D1-Ova) or CD8 transgenic T cells (targeting of CD11c + CD8 + DCs with DEC205-<br />

Ova). On the other hand when we targeted antigens under tolerizing conditions<br />

transgenic T cells started to proliferate till day 3, but were deleted afterwards. Left T<br />

cells clearly were of regulatory T cell phenotype (Yamazaki et al., 2008). We further<br />

could show by 2-Photon microscopy that T cells that were interacting with DCs that<br />

presented the antigen under tolerizing conditions or presented the antigen under<br />

immunizing conditions were behaving very similar in dependency on interaction time,<br />

velocity and activation markers expressed on the T cells (Lindquist et al., 2004;<br />

Shakhar et al., 2005). Regarding the induced immune responses we induced under<br />

immunizing conditions with anti CD40 antibody, as gold standard activation stimulus,<br />

we found that targeting antigens to CD11c + CD8 - DCs predominantly induced a TH2 T<br />

cell response when we analyzed Balb/c mice but on the other hand a TH1 T cell<br />

response when CD11c + CD8 + DCs were targeted (Soares et al., 2007).<br />

Altogether, our data indicate that targeting of CD11c + CD8 + DCs by anti-<br />

DEC205 antibody seems to be an efficient way to induce cross presentation in the<br />

classical MHCI pathway. In contrast, CD11c + CD8 - DCs preferentially processed and<br />

presented antigens in the traditional MHCII pathway. Currentlc we are investigating<br />

the antigen processing machineries when DCs were activated with TLR ligands.<br />

Analysis of early T cell tolerance and T cell immunity<br />

Diana Dudziak, Kirsten Neubert<br />

We and others could show that antigen targeted DCs elicited a peripheral tolerance<br />

in antigen-specific T cells under non-inflammatory conditions. In contrast, delivery of<br />

recombinant antigen-carrying antibodies together with a stimulatory anti CD40<br />

antibody established an antigen-specific immune response. Because of severe side<br />

effects such as systemic inflammation and the non-specific activation of all immune<br />

cells that express CD40 on their cell surface the usage of systemic anti CD40<br />

antibody cannot be a feasible costimulatory strategy for human therapy.<br />

As TLR stimulation is an approach already used in clinical trials we believe that TLR<br />

stimulation in combination with our antibody targeting strategy might be an alternative<br />

approach to induce protective immune responses in vivo. TLRs are very well<br />

characterized class of pattern-recognition-receptors (PRRs) in mammalian species.<br />

The TLRs are differentially expressed on various cells of the immune system<br />

including DCs. TLRs 1, 2, 4, 5, and 6 are expressed on the cell surface and seem to<br />

have evolved to recognize bacterial products. In contrast TLRs 3, 7, 8, and 9 are<br />

expressed intracellularly in endosomes and lysosomes and recognize nucleic acids.<br />

Several pathogen-associated molecules such as double stranded RNA (poly(I:C) =<br />

pIC), bacterial or viral double stranded DNA (CpG), bacterial lipopolysaccharides<br />

(LPS), lipoproteins (Pam3Cys = Pam3), lipoteichoide acids, zymosan, or flagellin<br />

have been identified to bind to TLR receptors resulting in a strong inflammatory and<br />

protective immune response.<br />

96


To determine if TLR agonists are able to support the initiation of an immune response<br />

in combination of in vivo antigen targeting of C-type lectin receptors we are analyzing<br />

the early T cell activation and proliferation after interaction of the T cells with antigen<br />

loaded DC subsets in vivo to identify 1) if T cells can respond to TLR ligands similar<br />

to anti CD40 stimulating antibody, 2) to analyze different pathways of TLR stimulation<br />

(MyD88 dependent versus MyD88 independent), 3) to identify which kind of T cell<br />

subset emerged after antigen targeting and TLR stimulation, and 4) to analyze what<br />

type of antibody response can be induced.<br />

To investigate if T cells can respond to antigen loaded and TLR ligand activated DCs<br />

in vivo, we perform T cell transfer experiments in antibody targeted mice. To target<br />

DC subsets in vivo we use the recombinant chimeric antibody DEC205, which<br />

recognizes predominantly CD11c + CD8 + splenic DCs and the 33D1 antibody which<br />

binds uniquely to the molecule DCIR2 on CD11c + CD8 - splenic DCs. For the<br />

phenotypical and functional analysis of the emerged T cells it is planned to reisolate<br />

CD4 + or CD8 + transgenic T cells after activation with TLR-ligands and targeting<br />

antibody in vivo. By CBA-cytokine bead array, FACS-analysis, Western blots and<br />

microarray analyses we will be able to identify important transcription factors,<br />

cytokines, and genes involved in establishing immunity or tolerance in T cells. With<br />

these data we will be able to identify which stimulus needs to be given to induce a<br />

strong and long lasting immune response in vivo.<br />

Analysis of cell migration in steady state und immunity in lymphoid<br />

organs<br />

Anna Baranska; Christina Weiss<br />

In this project we focus on the role of DC migration for the induction of efficient and<br />

life long memory T cell responses. Therefore, we inject mice with different TLR<br />

ligands or anti CD40 antibody to analyze the expression of activation markers by<br />

FACS-analysis, DC migration by confocal microscopy, and the produced cytokines by<br />

CBA-assay at different time points after activation. We found that the C-type lectin<br />

receptor DCIR2 was not influenced by the DC activation, whereas DEC205 was up<br />

regulated by diverse TLR ligands. In addition, we found that the upregulation of<br />

MHCII and the activation markers CD80 and CD86 were different dependent on the<br />

TLR stimulus as well as the DC subpopulation (Baranska et al., in preparation). We<br />

further established a 6 color confocal microscopy analysis to identify where the<br />

different DC subsets localize after activation. Our next steps will be the simultaneous<br />

targeting of antigens to either CD11c + CD8 - or CD11c + CD8 + DCs under stimulating or<br />

tolerizing conditions to analyze when and were DCs interact with T cells in vivo.<br />

These data will help to understand the important role of DCs in initiation of an<br />

immune response.<br />

New molecules for Antigen targeting in vivo<br />

Targeting inhibitory and activating Fc-receptors in vivo<br />

Christian Lehmann<br />

Fc-receptors are widely accepted as key mediators of specific, antibody-dependent<br />

reactions of the immune system against pathogens (Biburger et al., 2011,<br />

97


Baerenwald et al., 2011; Nimmerjahn et al., 2010). In the last couple of years it<br />

became clear, that antibodies can also down-regulate immune-responses by acting<br />

on inhibitory Fc-receptors like Fc-gamma-RIIb (which contains an inhibitory signaling<br />

motif, ITIM). The Fc-gamma-RIIB is an Fc-receptor expressed on B cells, and<br />

therefore gives a negative feed-back to the antibody producing B cells. On the other<br />

hand all other Fc-receptors e.g. Fc-gamma RIV (which interacts with the common<br />

gamma chain and therefore induces an activating signal, ITAM), activate the cells<br />

after binding antibodies. As we are interested to understand how the immune system<br />

maintains peripheral tolerance and induces immunity, we would like to study the role<br />

of intrinsic signaling properties of the targeted receptors. As Fc receptors have a<br />

strong endocytosis capacity, and as they are expressed on a variety of antigen<br />

presenting cells, Fc receptors seem to be ideal molecules for targeting antigens in<br />

vivo. Targeting these molecules can model reactions to antigens bound by<br />

antibodies. In our studies we concentrate on the activating Fc-gamma-RIV and the<br />

inhibitory Fc-gamma-RIIb. We already know from literature that monocytes, but not B<br />

cells express Fc-gamma-RIV, whereas Fc-gamma-RIIb is expressed on both cell<br />

types.<br />

For addressing these questions we have cloned the variable regions of the antibodies<br />

directed against the two mentioned Fc-gamma receptors as mouse IgG1 isotype.<br />

Since the constant region is mutated these antibodies are only able to bind the Fc<br />

receptors via their variable regions and not via their Fc part. In addition, we<br />

genetically fused the model antigen Ovalbumin to the heavy chains, which has been<br />

widely used for investigating specific CD4 and CD8 T cell responses in vivo (Dudziak<br />

et al., 2007, Soares et al., 2007; Do et al., 2010, O’Kamphorst et al., 2010). We are<br />

currently producing these antibodies in HEK293T cells by transient transfection.<br />

Afterwards they will be used to compare T cell responses after targeting different<br />

cells (e.g. CD11c + CD8 + DCs, CD11c + CD8 - DCs, B cells, monocytes) and receptors for<br />

the induction of CD4 and CD8 T cells responses. In future, we are planning to use<br />

our antibodies against the inhibitory Fc-gamma-RIIb to protect mice prone to diabetes<br />

from the disease.<br />

Functional characterization of human Dendritic cell<br />

subpopulations<br />

Characterization of Dendritic cell subpopulations in human tissues<br />

Gordon Heidkamp; Nathalie Eissing, Lukas Heger; Simone Beck, Yang Jiao<br />

As everything we know about immune responses is restricted to the murine system,<br />

we are investigating the function of DC subpopulations in human lymphoid organs. In<br />

order to obtain single cell suspensions, a method to efficiently process human splenic<br />

tissue needed to be established (Heidkamp et al., manuscript in preparation).<br />

Extensive multi-colour FACS analysis was used to identify Dendritic cell<br />

subpopulations. Here, Dendritic cells in the human spleen, tonsils, thymus, bone<br />

marrow, blood and cord blood was analyzed according to the already described<br />

expression profiles of blood DCs. So far, two major human blood DC lineages are<br />

defined: myeloid DCs and plasmacytoid DCs. The latter is characterized by the coexpression<br />

of BDCA-2, BDCA-4, CD123 and HLA-DR, whereas myeloid DCs can be<br />

further subdivided into mDC type 1 (BDCA-1 + , CD14 + , CD11c hi ) and mDC type 2<br />

98


(BDCA-3 + , CD14 + , CD11c lo ). These subpopulations have now been characterized for<br />

the expression profil of their cell surface receptors, by their RNA-expression profile in<br />

genearray analyses and by the localization of the DC subpopulations in the tissue on<br />

the tissues mentioned above with the focus on antigen targeting receptors and<br />

antigen processing machinery in the human DC subpopulations.<br />

Antigen targeting of human DCs with anti C-type lectin antibodies<br />

Gordon Heidkamp, Nathalie Eissing, Kirsten Ehrenspeck,<br />

As 33D1 antibody targeting induced a strong CD4 T cell response in mice and<br />

antigen targeting by DEC205 antibody induced a strong CD8 T cell response in vivo<br />

we wanted to investigate the role of the human counterpart of DCIR2 (33D1-antigen)<br />

and DEC205 in peripheral blood and human organs. As the human DEC205 antibody<br />

already existed, we concentrated on the production of an antibody against human<br />

DCIR. ClecSF6/DCIR/LLIR is the counterpart of the murine DCIR2. The antibodies<br />

were first tested by ELISA, FACS, immunofluorescence, Western blot, and<br />

immunoprecipitations if they bind specifically to the C-type lectin hDCIR (Heidkamp et<br />

al., 2010). Later we have chosen the clone 15E12 which showed the best binding<br />

specificities. We cloned the variable regions of the antibody and produced<br />

recombinant hDCIR antibodies in the tissue culture. In addition, our recombinant<br />

antibodies contained the Hemagglutinin antigen of Influenza virus as model antigen.<br />

With the recombinant HA-conjugated hDCIR-HA and hDEC205-HA antibodies we<br />

were able to target human monocyte derived DCs in the tissue culture and to induce<br />

a restimulation of Influenza-specific CD4 and CD8 T cells (Heidkamp et al.,<br />

manuscript submitted). New antibodies are currently produced against newly<br />

identified endocytic receptors specifically expressed in human tissue DCs for future<br />

antigen targeting as therapeutic option.<br />

2011/2012<br />

Publications (2006-2008)<br />

Original Articles<br />

Heidkamp GF, Eissing N, Heger L, Schmitt V, Neubert K, Baranska A, Bozzacco F,<br />

Trumpfheller C, Zebroski H, Schuler G, Nimmerjahn F, Nussenzweig MC, , Dudziak<br />

D. Differential and directed antigen presentation after targeting of DEC205 and DCIR<br />

on human dendritic cells. manuscript submitted<br />

Biburger M, Albert H, Woigk M, Dudziak D, Mack M, Ravetch JV, Nimmerjahn F.<br />

Monocyte subsets involved in immunoglobulin G mediated effector functions in vivo.<br />

Immunity. 2011 Dec 23;35(6):932-44<br />

Loschko J, Heink S, Hackl D, Dudziak D, Reindl W, Korn T, Krug AB. Antigen<br />

Targeting to Plasmacytoid Dendritic Cells via Siglec-H Inhibits Th Cell-Dependent<br />

Autoimmunity. J Immunol. 2011 Dec 15;187(12):6346-56.<br />

Baerenwaldt A, Lux A, Danzer H, Spriewald BM, Ullrich E, Heidkamp G, Dudziak D,<br />

Nimmerjahn F. Fcγ receptor IIB (FcγRIIB) maintains humoral tolerance in the human<br />

immune system in vivo. Proc Natl Acad Sci U S A. 2011 Nov 15;108(46):18772-<br />

18777.<br />

99


Loschko J, Hackl D, Dudziak D, Reindl W, Krug A. Tuning T cell responses by<br />

antigen targeting to plasmacytoid dendritic cells in vivo. J Immunol. 2011 Jun<br />

15;186(12):6718-25<br />

2010<br />

Nimmerjahn F, Lux A, Albert H, Woigk M, Lehmann C, Dudziak D, Smith P, Ravetch<br />

JV. FcγRIV deletion reveals its central role in mediating IgG2a and IgG2b activity in<br />

vivo. Proc Natl Acad Sci U S A. 2010 Nov 9;107(45):19396-401.<br />

Kamphorst, AO, Guermonprez P, Dudziak D, Nussenzweig MC. Route of antigen<br />

uptake differentially impacts presentation by dendritic cells and activated monocytes.<br />

J Immunol. 2010 Sep 15;185(6):3426-35. Epub 2010 Aug 20.<br />

Do Y, Koh H, Park CG, Dudziak D, Seo P, Mehandru S, Choi JH, Cheong C, Park S,<br />

Perlin DS, Powell BS, Steinman RM. Targeting of LcrV virulence protein from<br />

Yersinia pestis to dendritic cells protects mice against pneumonic plague. Eur J<br />

Immunol. 2010 Oct;40(10):2791-6.<br />

Heidkamp GF, Neubert K, Haertel E, Nimmerjahn F, Nussenzweig MC, Dudziak D*.<br />

Efficient generation of a monoclonal antibody against the C-type Lectin receptor<br />

DCIR by targeting murine dendritic cells. Immunol Lett. 2010 Aug 16;132(1-2):69-78.<br />

Epub 2010 Jun 8.<br />

2009-2011/2012<br />

Review Articles<br />

Ullrich E, Bosch J, Aigner M, Voelkl S, Kroeger I, Hoffmann P, Kreutz M, Dudziak D,<br />

Gerbitz A. Advances in cellular therapy: 6th international symposium on the clinical<br />

use of cellular products, March 24 and 25, 2011, Erlangen, Germany. Cancer<br />

Immunol Immunother. 2011 Dec 27.<br />

Ullrich E, Bosch J, Aigner M, Völkl S, Dudziak D, Spriewald B, Schuler G, Andreesen<br />

R, Mackensen R. Advances in Cellular Therapy: 5th International Symposium on the<br />

Clinical Use of Cellular Products, March 19 and 20, 2009, Nürnberg, Germany.<br />

Cancer Immunology Immunotherapy. 2010.<br />

Dudziak D, Dendritic cells as Master regulators of T cell responses. BioForum<br />

Europe. 2009.<br />

100


TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology, Dr. rer. nat.)<br />

Gordon Heidkamp (Dipl. Biol.)<br />

Characterization of human Dendritic cell subpopulations<br />

Anna Baranska (Dipl. Biol.)<br />

Antigen targeting and antigen processing in maturing Dendritic cell subpopulations<br />

Christian Lehmann (Dipl. Chem.)<br />

Antigen targeting to inhibitory and activating Fc-receptors in vivo<br />

Doctoral theses (Molecular Medicine, Dr. rer. nat.)<br />

Lukas Heger<br />

Chemotactic characterization of human DC subpopulations in lymphoid and nonlymphoid<br />

tissues<br />

Doctoral theses (Medicine, Dr. med.)<br />

Yang Jiao<br />

Characterization of pDC subpopulations in human tissues<br />

Doctoral theses (Veterinarian Medicine, Dr. med. vet.)<br />

Nathalie Eissing (maiden name: Nathalie Bleny)<br />

Characterization of Dendritic cell subpopulations and their expression of C-type lectin<br />

receptors in human tissues by immunofluorescence microscopy<br />

Kirsten Ehrenspeck<br />

Characterization of stem cells in human tissues and their differentiation into Dendritic<br />

cells<br />

101


Experimental Immunology and Immunotherapy<br />

(Medical Department III) until March 2010<br />

Head: Falk Nimmerjahn, Dr. rer. nat.<br />

Associate Professor of Experimental Immunology and<br />

Immunotherapy (until March 2010)<br />

Since April 2010:<br />

Professor and Chairman of the Institute of Genetics at<br />

the Department of Biology<br />

Address: Chair of Genetics<br />

Erwin-Rommelstr. 5<br />

D-91058 Erlangen<br />

Telefone: + 49 (9131) 85 25050<br />

Fax: + 49 (9131) 85 28526<br />

E-mail: fnimmerj@biologie.uni-erlangen.de<br />

Markus Biburger, Dr. rer. nat.<br />

Anne Bärenwaldt (PhD<br />

received in 2011)<br />

Susanne Aschermann<br />

(PhD received in 2012)<br />

Head<br />

Falk Nimmerjahn, Prof. Dr. rer. nat.<br />

Professor of Genetics<br />

Postdoctoral Fellows<br />

Doctoral Students<br />

Technicians<br />

102<br />

Anja Lux, Dr. rer. nat.<br />

Inessa Schwab<br />

Birgit Lehmann<br />

Sybille Böhm


<strong>Research</strong><br />

Heike Albert Melissa Woigk<br />

Heike Danzer<br />

The research of our laboratory focuses on understanding the mechanisms that underlie the<br />

different activities of immunoglobulin G (IgG) antibodies in vivo. IgG antibodies are the<br />

primary mediators of protective humoral immunity against pathogens and have been used<br />

therapeutically for over a century. They were first used as antitoxins for the treatment of<br />

infectious diseases in the pre-antibiotic era. Today, hyperimmune sera from human donors<br />

recovering from infection with specific viruses, such as hepatitis B, cytomegalovirus, and<br />

varicella zoster, are used to provide protective immunity to susceptible populations.<br />

Moreover, tumor specific antibodies have been successfully used in human cancer therapy<br />

and have been included in standard therapeutic regimens in lymphoma and breast cancer.<br />

Besides these protective activities, IgG autoantibodies are the principal mediators of<br />

autoimmune diseases such as immune thrombocytopenia (ITP), autoimmune haemolytic<br />

anemia (AHA), and systemic lupus erythematosus (SLE). In addition to this pro-inflammatory<br />

activity antibodies also are known to have an anti-inflammatory activity. If infused at high<br />

doses, IgG can effectively suppress autoimmune mediated inflammation (IVIG therapy).<br />

Recent evidence suggests that both the pro- and anti-inflammatory activity of IgG is<br />

regulated by the sugar side chain that is attached to the CH2-domain of all IgG subclasses.<br />

Subtle variations in the composition of this sugar moiety will either enhance or decrease the<br />

pro-inflammatory activity. Using a variety of in vivo model systems of autoimmune diseases<br />

and syngeneic tumor models we are trying to delineate which effector mechanisms the<br />

different IgG subclasses depend on and which effector cell populations are involved in this<br />

process.<br />

Focus I: Effector cells responsible for IgG activity (Biburger,<br />

Aschermann, Lehmann, Schwab)<br />

Immunoglobulins (Ig) are important mediators of the immunological defence against<br />

pathogens, however, autoantigen-specific antibodies may elicit autoimmunediseases.<br />

Due to these double-edged effects, immunoglobulin-mediated responses<br />

are strictly regulated. This is achieved by the existence of both activating and<br />

inhibitory receptors recognizing these antibodies (Fc receptors). Such Fc receptors<br />

are present in different combinations on the cell surface of various cell types and are<br />

characterized by different affinities for the respective IgG subclasses. In spite of the<br />

physiological importance of the interactions between immunoglobulins and their<br />

103


activating or inhibitory Fc receptors, there are still many unresolved questions. In this<br />

project we aim to understand how antibody-dependent cell-mediated cytotoxicity<br />

(ADCC) reactions work in vivo using a model of autoantibody mediated<br />

thrombocytopenia (ITP). During the last funding period we were able to show that a<br />

certain activating Fc-receptor, FcγRIV, which is crucial for autoantibody mediated<br />

platelet depletion is selectively expressed on tissue macrophages, neutrophils and on<br />

resident monocytes. By using a variety of cell depletion strategies we could<br />

demonstrate that neither neutrophils nor macrophages but resident monocytes were<br />

responsible for IgG dependent platelet removal (Figure 1).<br />

Figure 1: (A) Quantification of cell numbers of Ly6C hi and Ly6C lo monocytes in mice that were either<br />

left untreated, injected with PBS or with 10μl of clodronate liposomes to deplete the Ly6C lo monocyte<br />

subset. (B) Shown are liver sections stained with an F4/80 antibody specific for liver resident<br />

macrophages two hours after injection of 10µl clodronate liposomes or PBS. (C) Blood neutrophil<br />

counts in mice left either untreated or injected with PBS or 10µl of clodronate liposomes two hours<br />

after injection. (D) Platelet depletion activity of the 6A6-IgG2a antibody in mice either left untreated or<br />

pretreated with PBS or clodronate liposomes 2 hours before antibody injection. (E) Platelet depleting<br />

activity of the 6A6-IgG2a antibody in the indicated mouse strains twenty four hours after pretreatment<br />

with diphtheria toxin (+DT) or PBS (-DT). (F) Residual platelet count in C57BL/6 and Fcgr1 -/- Fcgr4 -/-<br />

mice four hours after injection of the 6A6-IgG2a anti-platelet antibody. * indicates p< 0.05.<br />

104


In addition to these passive models of autoimmunity we are also investigating more<br />

complex spontaneous mouse models of chronic autoimmune diseases. Specifically<br />

we are trying to elucidate which parts of the immune system contribute to the severe<br />

systemic autoimmune disease in Scurfy mice, which have a mutation in the<br />

transcription factor Foxp3, the master regulator of CD4+ CD25+ regulatory T cell<br />

development and function. As a member of the forkhead/winged-helix family of<br />

transcriptional regulators Foxp3 is essential for normal immune homeostasis. Mice<br />

hemizygous for the X-linked (Xsf/Y) mutation (scurfy mice) suffer from a fatal<br />

lymphoproliferative disorder caused by an impaired development of regulatory T cells<br />

(CD4+ CD25+ Foxp3+). Mice develop multi-organ lymphocytic infiltrates, resulting in<br />

lethal autoimmunity 3-6 weeks after birth. In our studies during the last years, we<br />

could identify autoantibody specificities associated with arthritis as well as murine<br />

lupus, which is the prototype of immune complex (IC)-mediated autoimmune disease.<br />

In addition, renal function was severely impaired. Interestingly, depletion of B cells<br />

either with a monoclonal antibody or by crossing Scurfy mice to B cell deficient<br />

mouse strains resulted in a prolonged survival of mice, suggesting that B cells and<br />

the respective autoantibodies play a critical role in the systemic inflammatory<br />

response.<br />

Focus II: Humanized mouse models to understand human IgG<br />

activity and pathways of humoral tolerance (Lux, Bärenwaldt,<br />

Böhm)<br />

Mice and humans differ in several aspects of IgG and FcγR biology. Therefore, one<br />

aim of our group is to establish in vivo models that allow studying the mechanism of<br />

human IgG activity and checkpoints of humoral tolerance in the setting of a human<br />

immune system in vivo. Humanized mice are generated by reconstitution of<br />

immunodeficient, irradiated mice (e.g. NOD/SCID/gamma-chain -/- ) with human CD34 +<br />

hematopoetic stem cells (HSC) purified from umbilical cord blood. Following<br />

reconstitution, the development of the human immune system is confirmed by flow<br />

cytometry and immunofluorescence staining. Human immune cells such as B, T, NK<br />

and dendritic cells are present in peripheral blood, spleen and bone marrow and<br />

human haematopoetic stem cells are able to persist in the bone marrow for months<br />

after reconstitution ensuring an ongoing human hematopoesis. As a model for human<br />

IgG activity we study B cell depletion in the peripheral blood, spleen, lymph nodes<br />

and bone marrow by IgG subclass variants of the CD20 specific antibody rituximab.<br />

In the second major project we investigated how impaired FcγRIIB alleles associated<br />

with the development of SLE in humans are involved in the maintenance of humoral<br />

tolerance. For this, humanized mouse colonies were generated that carried either the<br />

functionally normal FcγRIIB-232I allele or the functionally impaired FcγRIIB-232T<br />

allele. By following the development of the human immune system in these mouse<br />

cohorts we were able to show that mice carrying a human immune system with<br />

impaired FcγRIIB function showed a more rapid production of serum immunoglobulin<br />

105


M and G levels. This was mirrored by a higher number of late B cell developmental<br />

stages including memory B cells and plasma cells, consistent with previous data<br />

obtained in classical mouse models. More importantly, however, we could also show<br />

that human B cells in mice with impaired human FcγRIIB function started to produce<br />

autoantibodies (Figure 2). These findings establish an important function of human<br />

FcγRIIB as a late checkpoint in the human humoral immune system, preventing the<br />

expansion of autoantibody producing B cells.<br />

Figure 2: Development of autoantibodies in humanized mice. (A-B) The levels of serum IgM and<br />

serum IgG were analyzed in humanized mice carrying the different FcγRIIB allelic variants at 4 and 6<br />

months of age. (C-F) Detection of IgM autoantibody responses to double stranded DNA (anti-DNA),<br />

glucose 6-phosphate-isomerase (anti-GPI), rheumatoid factor (RF) and cyclic citrullinated peptides<br />

(anti-CCP) in humanized mice at 16 and 24 weeks of age. The level of autoantibody production is<br />

shown as arbitrary units (AU) for anti-DNA, -GPI, and –RF responses and as optical density (OD) for<br />

anti-CCP responses. Statistical significance was calculated with the Mann-Whitney U test. *P < 0.05.<br />

Data points of mice that received human hematopoetic stem cells from the same donor are depicted in<br />

the same color.<br />

106


Publications (2009-2012)<br />

Original Articles<br />

2012<br />

Biburger, M., and Nimmerjahn, F. (2012). Low level of FcgammaRIII expression on<br />

murine natural killer cells. Immunol Lett 143, 53-59.<br />

Bohm, S., Schwab, I., Lux, A., and Nimmerjahn, F. (2012). The role of sialic acid as a<br />

modulator of the anti-inflammatory activity of IgG. Semin Immunopathol.<br />

Harre, U., Georgess, D., Bang, H., Bozec, A., Axmann, R., Ossipova, E., Jakobsson,<br />

P.J., Baum, W., Nimmerjahn, F., Szarka, E., et al. (2012). Induction of<br />

osteoclastogenesis and bone loss by human autoantibodies against citrullinated<br />

vimentin. J Clin Invest 122, 1791-1802.<br />

Kasperkiewicz, M., Nimmerjahn, F., Wende, S., Hirose, M., Iwata, H., Jonkman, M.F.,<br />

Samavedam, U., Gupta, Y., Moller, S., Rentz, E., et al. (2012). Genetic identification<br />

and functional validation of FcgammaRIV as key molecule in autoantibody-induced<br />

tissue injury. J Pathol.<br />

Lehmann, B., Schwab, I., Bohm, S., Lux, A., Biburger, M., and Nimmerjahn, F.<br />

(2012). FcgammaRIIB: a modulator of cell activation and humoral tolerance. Expert<br />

Rev Clin Immunol 8, 243-254.<br />

Moldt, B., Shibata-Koyama, M., Rakasz, E.G., Schultz, N., Kanda, Y., Dunlop, D.C.,<br />

Finstad, S.L., Jin, C., Landucci, G., Alpert, M.D., et al. (2012). A Nonfucosylated<br />

Variant of the anti-HIV-1 Monoclonal Antibody b12 Has Enhanced FcgammaRIIIa-<br />

Mediated Antiviral Activity In Vitro but Does Not Improve Protection against Mucosal<br />

SHIV Challenge in Macaques. J Virol 86, 6189-6196.<br />

Ruiz-Heiland, G., Horn, A., Zerr, P., Hofstetter, W., Baum, W., Stock, M., Distler, J.H.,<br />

Nimmerjahn, F., Schett, G., and Zwerina, J. (2012). Blockade of the hedgehog<br />

pathway inhibits osteophyte formation in arthritis. Ann Rheum Dis 71, 400-407.<br />

Schwab, I., Biburger, M., Kronke, G., Schett, G., and Nimmerjahn, F. (2012). IVIgmediated<br />

amelioration of ITP in mice is dependent on sialic acid and SIGNR1. Eur J<br />

Immunol 42, 826-830.<br />

Uderhardt, S., Herrmann, M., Oskolkova, O.V., Aschermann, S., Bicker, W., Ipseiz,<br />

N., Sarter, K., Frey, B., Rothe, T., Voll, R., et al. (2012). 12/15-lipoxygenase<br />

107


orchestrates the clearance of apoptotic cells and maintains immunologic tolerance.<br />

Immunity 36, 834-846.<br />

2011<br />

Baerenwaldt, A., Lux, A., Danzer, H., Spriewald, B.M., Ullrich, E., Heidkamp, G.,<br />

Dudziak, D., and Nimmerjahn, F. (2011). Fcgamma receptor IIB (FcgammaRIIB)<br />

maintains humoral tolerance in the human immune system in vivo. Proc Natl Acad<br />

Sci U S A 108, 18772-18777.<br />

Biburger, M., Aschermann, S., Schwab, I., Lux, A., Albert, H., Danzer, H., Woigk, M.,<br />

Dudziak, D., and Nimmerjahn, F. (2011). Monocyte subsets responsible for<br />

immunoglobulin G-dependent effector functions in vivo. Immunity 35, 932-944.<br />

Lux, A., and Nimmerjahn, F. (2011). Impact of differential glycosylation on IgG<br />

activity. Adv Exp Med Biol 780, 113-124.<br />

Nimmerjahn, F., and Lunemann, J.D. (2011). Expression and function of the<br />

inhibitory Fcgamma-receptor in CIDP. J Peripher Nerv Syst 16 Suppl 1, 41-44.<br />

Nimmerjahn, F., and Ravetch, J.V. (2011). FcgammaRs in health and disease. Curr<br />

Top Microbiol Immunol 350, 105-125.<br />

Santiago-Raber, M.L., Baudino, L., Alvarez, M., van Rooijen, N., Nimmerjahn, F., and<br />

Izui, S. (2011). TLR7/9-mediated monocytosis and maturation of Gr-1(hi)<br />

inflammatory monocytes towards Gr-1(lo) resting monocytes implicated in murine<br />

lupus. J Autoimmun 37, 171-179.<br />

2010<br />

Anthony, R.M., and Nimmerjahn, F. (2010). The role of differential IgG glycosylation<br />

in the interaction of antibodies with FcgammaRs in vivo. Curr Opin Organ Transplant.<br />

Aschermann, S., Lux, A., Baerenwaldt, A., Biburger, M., and Nimmerjahn, F. (2010).<br />

The other side of immunoglobulin G: suppressor of inflammation. Clin Exp Immunol<br />

160, 161-167.<br />

Baerenwaldt, A., Biburger, M., and Nimmerjahn, F. (2010). Mechanisms of action of<br />

intravenous immunoglobulins. Expert Rev Clin Immunol 6, 425-434.<br />

Comabella, M., Montalban, X., Kakalacheva, K., Osman, D., Nimmerjahn, F., Tintore,<br />

M., and Lunemann, J.D. (2010). B cell expression of the inhibitory Fc gamma<br />

receptor is unchanged in early MS. J Neuroimmunol 223, 135-137.<br />

Heidkamp, G.F., Neubert, K., Haertel, E., Nimmerjahn, F., Nussenzweig, M.C., and<br />

Dudziak, D. (2010). Efficient generation of a monoclonal antibody against the human<br />

108


C-type lectin receptor DCIR by targeting murine dendritic cells. Immunol Lett 132, 69-<br />

78.<br />

Lux, A., Aschermann, S., Biburger, M., and Nimmerjahn, F. (2010). The pro and antiinflammatory<br />

activities of immunoglobulin G. Ann Rheum Dis 69 Suppl 1, i92-96.<br />

Najjar, I., Deglesne, P.A., Schischmanoff, P.O., Fabre, E.E., Boisson-Dupuis, S.,<br />

Nimmerjahn, F., Bornkamm, G.W., Dusanter-Fourt, I., and Fagard, R. (2010).<br />

STAT1-dependent IgG cell-surface expression in a human B cell line derived from a<br />

STAT1-deficient patient. J Leukoc Biol 87, 1145-1152.<br />

Nimmerjahn, F., Lux, A., Albert, H., Woigk, M., Lehmann, C., Dudziak, D., Smith, P.,<br />

and Ravetch, J.V. (2010). FcgammaRIV deletion reveals its central role for IgG2a<br />

and IgG2b activity in vivo. Proc Natl Acad Sci U S A 107, 19396-19401.<br />

Nimmerjahn, F., and Ravetch, J.V. (2010). Antibody-mediated modulation of immune<br />

responses. Immunol Rev 236, 265-275.<br />

Ritter, M., Gross, O., Kays, S., Ruland, J., Nimmerjahn, F., Saijo, S., Tschopp, J.,<br />

Layland, L.E., and Prazeres da Costa, C. (2010). Schistosoma mansoni triggers<br />

Dectin-2, which activates the Nlrp3 inflammasome and alters adaptive immune<br />

responses. Proc Natl Acad Sci U S A 107, 20459-20464.<br />

Schoenen, H., Bodendorfer, B., Hitchens, K., Manzanero, S., Werninghaus, K.,<br />

Nimmerjahn, F., Agger, E.M., Stenger, S., Andersen, P., Ruland, J., et al. (2010).<br />

Cutting edge: Mincle is essential for recognition and adjuvanticity of the<br />

mycobacterial cord factor and its synthetic analog trehalose-dibehenate. J Immunol<br />

184, 2756-2760.<br />

Tackenberg, B., Nimmerjahn, F., and Lunemann, J.D. (2010). Mechanisms of IVIG<br />

efficacy in chronic inflammatory demyelinating polyneuropathy. J Clin Immunol 30<br />

Suppl 1, S65-69.<br />

Zaiss, M.M., Frey, B., Hess, A., Zwerina, J., Luther, J., Nimmerjahn, F., Engelke, K.,<br />

Kollias, G., Hunig, T., Schett, G., et al. (2010). Regulatory T cells protect from local<br />

and systemic bone destruction in arthritis. J Immunol 184, 7238-7246.<br />

Zaiss, M.M., Sarter, K., Hess, A., Engelke, K., Bohm, C., Nimmerjahn, F., Voll, R.,<br />

Schett, G., and David, J.P. (2010). Increased bone density and resistance to<br />

ovariectomy-induced bone loss in FoxP3-transgenic mice based on impaired<br />

osteoclast differentiation. Arthritis Rheum 62, 2328-2338.<br />

2009<br />

109


Grevers, L.C., van Lent, P.L., Koenders, M.I., Walgreen, B., Sloetjes, A.W.,<br />

Nimmerjahn, F., Sjef Verbeek, J., and van den Berg, W.B. (2009). Different<br />

amplifying mechanisms of interleukin-17 and interferon-gamma in Fcgamma<br />

receptor-mediated cartilage destruction in murine immune complex-mediated<br />

arthritis. Arthritis Rheum 60, 396-407.<br />

Guo, S., Muhlfeld, A.S., Wietecha, T.A., Peutz-Kootstra, C.J., Kowalewska, J., Yi, K.,<br />

Spencer, M., Pichaiwong, W., Nimmerjahn, F., Hudkins, K.L., et al. (2009). Deletion<br />

of activating Fcgamma receptors does not confer protection in murine<br />

cryoglobulinemia-associated membranoproliferative glomerulonephritis. Am J Pathol<br />

175, 107-118.<br />

Santiago-Raber, M.L., Amano, H., Amano, E., Baudino, L., Otani, M., Lin, Q.,<br />

Nimmerjahn, F., Verbeek, J.S., Ravetch, J.V., Takasaki, Y., et al. (2009). Fcgamma<br />

receptor-dependent expansion of a hyperactive monocyte subset in lupus-prone<br />

mice. Arthritis Rheum 60, 2408-2417.<br />

Syed, S.N., Konrad, S., Wiege, K., Nieswandt, B., Nimmerjahn, F., Schmidt, R.E.,<br />

and Gessner, J.E. (2009). Both FcgammaRIV and FcgammaRIII are essential<br />

receptors mediating type II and type III autoimmune responses via FcRgamma-LATdependent<br />

generation of C5a. Eur J Immunol 39, 3343-3356.<br />

Tackenberg, B., Jelcic, I., Baerenwaldt, A., Oertel, W.H., Sommer, N., Nimmerjahn,<br />

F., and Lunemann, J.D. (2009). Impaired inhibitory Fcgamma receptor IIB expression<br />

on B cells in chronic inflammatory demyelinating polyneuropathy. Proc Natl Acad Sci<br />

U S A 106, 4788-4792.<br />

Werninghaus, K., Babiak, A., Gross, O., Holscher, C., Dietrich, H., Agger, E.M.,<br />

Mages, J., Mocsai, A., Schoenen, H., Finger, K., et al. (2009). Adjuvanticity of a<br />

synthetic cord factor analogue for subunit Mycobacterium tuberculosis vaccination<br />

requires FcRgamma-Syk-Card9-dependent innate immune activation. J Exp Med<br />

206, 89-97.<br />

Current <strong>Research</strong> Funding<br />

2007-2012 Bayerisches Genomforschungsnetzwerk<br />

2009-2012 DFG Forschergruppe 832: Regulatoren der humoralen Immunität<br />

2008-2012 SFB 643: Strategien zur zellulären Immunintervention<br />

2010-2013 SPP 1468: Immunobone<br />

110


2011-2015 GK1660: Schlüsselsignale der adaptiven Immunität<br />

2011-2013 Bill and Melinda Gates Foundation<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses<br />

Anja Lux (PhD in 2011)<br />

Anne Bärenwaldt (PhD in 2011)<br />

Susanne Aschermann (PhD in 2012)<br />

Inessa Schwab<br />

Birgit Lehmann<br />

Sybille Böhm<br />

111


Clinical <strong>Research</strong> Group<br />

(Reinhard Voll, Med.3)<br />

Head: Reinhard Voll, Dr. med.<br />

Professor of Rheumatology and Clinical Immunology<br />

(Since Dec. 2010 director of the Division Rheumatology<br />

and Clinical Immunology, University Medical Center<br />

Freiburg)<br />

Address: Clinical <strong>Research</strong> Group<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 39303<br />

Fax: + 49 (9131) 85 39311<br />

E-mail: reinhard.voll@uniklinik-freiburg.de<br />

Head<br />

Reinhard Voll, Prof. Dr. med.<br />

Professor of Rheumatolgy and Clinical Immunology<br />

Vilma Urbonaviciute, Dr. rer. nat.*<br />

Silke Meister, Dr. rer. nat. *<br />

* part of the time reported<br />

Supporting Staff<br />

Postdoctoral Fellows<br />

112<br />

Bettina Sehnert, Dr. rer. nat. *


Charlotte Starke<br />

° PhD received<br />

Sandy Pohle<br />

° PhD received<br />

Doctoral Students (Biology)<br />

Eva Gückel°<br />

Doctoral Students (Molecular Medicine)<br />

Daniela Graef *<br />

* part of the time reported<br />

<strong>Research</strong><br />

Technicians<br />

Guest Scientists<br />

The clinical research group is mainly interested in the etiology and pathogenesis of<br />

autoimmune diseases, especially systemic lupus erythematosus and rheumatoid<br />

arthritis. Based on the insights into the pathogenesis, new treatment strategies are<br />

going to be developed, initially in mouse models and, eventually, transferred to<br />

clinics. Targeting plasma cells for the treatment of autoantibody-mediated diseases<br />

and selective inhibition of the transcription factor NF-κB within activated endothelial<br />

cells represent such novel treatment strategies.<br />

113


From the immunopathogenesis of systemic lupus erythematosus to<br />

new treatment strategies<br />

Autoantibodies against nuclear antigens, especially double stranded (ds) DNA and<br />

nucleosomes represent a hallmark of SLE. However, the mechanisms involved in<br />

breaking the immunological tolerance against these normally poorly immunogenic<br />

nuclear components are not understood yet. We have shown previously that impaired<br />

phagocytosis of apoptotic cells with consecutive release of nuclear antigens may<br />

critically contribute to the immune pathogenesis of SLE. The architectural<br />

chromosomal protein and alarmin high mobility group box protein 1 (HMGB1) is<br />

tightly attached to the chromatin of apoptotic cells. Our results demonstrate that<br />

HMGB1 remains bound to nucleosomes released from late apoptotic cells in vitro. In<br />

addition, HMGB1-nucleosome complexes were also detected in the blood of SLE<br />

patients. Biochemically purified HMGB1-containing nucleosomes from apoptotic cells<br />

induced secretion of proinflammatory cytokines from monocytes/macrophages and<br />

maturation as well as activation of dendrtic cells. Immunizaton of non-autoimmune<br />

mice with nucleosomes from apoptotic cells, but not with those from living cells,<br />

induced autoantibodies to dsDNA. Further investigations are focused on the<br />

characterization of receptors for HMGB1-nucleosome complexes such as TLR2 and<br />

their roles in the pathogenesis of SLE. In addition, we are investigating if TLR2 and<br />

HMGB1 are suitable therapeutic targets in SLE.<br />

Refractory disease courses of SLE might be caused by long-lived plasma cells<br />

producing pathogenic autoantibodies. Hence, the elimination of long-lived plasma<br />

cells appears to represent an important treatment goal. However, long-lived plasma<br />

cells are resistant toward conventional therapies. We found that proteasome<br />

inhibitors can efficiently eliminate plasma cells including long-lived ones in murine<br />

SLE models.<br />

Recently, we described that long-lived plasma cells can be also found within the<br />

inflamed kidneys of diseased NZB/W F1 lupus mice. Interestingly, the frequency of<br />

autoreactive antibody-producing cells in the kidneys was markedly higher than in<br />

bone marrow and spleen (Starke et al., 2011). Currently we try to characterize these<br />

pathogenic plasma cells within kidneys and explore ways of their efficient elimination.<br />

Selective inhibition of NF-kB in activated endothelium as<br />

treatment strategy in immune-mediated diseases<br />

One research focus of our group is the investigation of the role of the transcription<br />

factor NF-κB in the pathogenesis of inflammatory diseases and the exploration of<br />

new strategies for therapeutic NF-κB inhibition. NF-κB represents a master switch of<br />

the inflammatory and immune responses. Most pro-inflammatory cytokines, tissue<br />

degrading enzymes, and several adhesion molecules are expressed in an NF-κB-<br />

114


dependent manner. Due to its central position in the inflammatory process<br />

interference with NF-κB activation represents a promising target for the treatment of<br />

inflammatory and autoimmune diseases. However, NF-κB plays also an important<br />

role in function and survival of several cell types, tissues, and organs. Therefore, we<br />

explore also the physiological function of NF-κB in lymphocyte development and the<br />

consequences of NF-κB inhibition for developing and mature lymphocytes.<br />

In collaboration with Stefan Dübel (Braunschweig), Harald Burkhardt (Frankfurt),<br />

Alexander Steinkasserer (Erlangen), Falk Nimmerjahn (Erlangen), Georg Schett<br />

(Erlangen) we developed a strategy for cell-type specific interference with NF-κB<br />

activation. Prototypic “sneaking ligand constructs” (SLC) were designed to target<br />

selectively activated endothelial cells via E selectin-binding peptides. The second<br />

domain of the fusion protein contains an endosomal release sequence, which<br />

translocates the IKK2 inhibiting NEMO-binding peptide into the cytoplasm. We could<br />

demonstrate that SLC1 efficiently ameliorates immune-mediated diseases such as<br />

murine arthritis models and experimental autoimmune encephalomyelitis (Sehnert et<br />

al. submitted).<br />

2011/12<br />

Publications (2009-2012)<br />

Original Articles<br />

Hainz N, Thomas S, Neubert K, Meister S, Benz K, Rauh M, Daniel C, Wiesener M, Voll RE,<br />

Amann K. The Proteasome Inhibitor Bortezomib Prevents Lupus Nephritis in the NZB/W F1<br />

Mouse Model by Preservation of Glomerular and Tubulointerstitial Architecture. Nephron<br />

Exp Nephrol. 2012;120(2):e47-58. Epub 2012 Jan 26.<br />

Pullerits R, Urbonaviciute V, Voll RE, Forsblad-D'Elia H, Carlsten H. Serum Levels of<br />

HMGB1 in Postmenopausal Patients with Rheumatoid Arthritis: Associations with<br />

Proinflammatory Cytokines, Acute-phase Reactants, and Clinical Disease Characteristics. J<br />

Rheumatol. 2011 Jul;38(7):1523-5.<br />

Starke C, Frey S, Wellmann U, Urbonaviciute V,Herrmann M, Amann K, Schett G, Winkler ,<br />

Voll RE High frequency of autoantibody-secreting cells and long-lived plasma cells within<br />

inflamed kidneys of NZB/W F1 lupus mice. Eur J 2011 Jul;41(7):2107-12<br />

Gückel E, Frey S, Zaiss MM, Schett G, Ghosh S, Voll RE. Cell-Intrinsic NF-kappaB Activation<br />

Is Critical for the Development of Natural Regulatory T Cells in Mice. PLoS One<br />

2011;6(5):e20003. Epub 2011 May 18<br />

Polzer K, Neubert K, Meister S, Frey B, Baum W, Distler JH, Gückel E, Schett G, Voll RE,<br />

Zwerina J.Proteasome inhibition aggravates tumor necrosis factor-mediated bone resorption<br />

in a mouse model of inflammatory arthritis.Arthritis Rheum. 2011 Mar;63(3):670-80.<br />

Gomez AM, Vrolix K, Martínez-Martínez P, Molenaar PC, Phernambucq M, van der Esch E,<br />

Duimel H, Verheyen F, Voll RE, Manz RA, De Baets MH, Losen M. Proteasome inhibition<br />

115


with bortezomib depletes plasma cells and autoantibodies in experimental autoimmune<br />

myasthenia gravis. J Immunol. 2011 Feb 15;186(4):2503-13.<br />

Mosca M, Govoni M, Tomietto P, Aringer M, Boumpas D, Cervera R, Conti F, D'Cruz D,<br />

Doria A, De La Fuente D, Galeazzi M, Houssiau F, Huizinga TW, Khamashta MA, Ines L,<br />

Duarte C, Couto M, Meroni P, Montecucco C, Norkuviene E, Riemekasten G, Rios V,<br />

Schneider M, Shoenfeld Y, Steup-Beekman GM, Szmyrka-Kaczmarek M, Tani C, Tincani A,<br />

Tzioufas AG, Voll R, Bencivelli W, Salaffi F, Bombardieri S. The development of a simple<br />

questionnaire to screen patients with SLE for the presence of neuropsychiatric symptoms in<br />

routine clinical practice. Lupus. 2011;20(5):485-92.<br />

2010<br />

Lang VR, Mielenz D, Neubert K, Böhm C, Schett G, Jäck HM, Voll RE, Meister S. The early<br />

marginal zone B cell-initiated T-independent type 2 response resists the proteasome inhibitor<br />

bortezomib. J Immunol. 2010 Nov 1;185(9):5637-47.<br />

Jellusova J, Düber S, Gückel E, Binder CJ, Weiss S, Voll R, Nitschke L.: Siglec-g regulates<br />

B1 cell survival and selection. J Immunol. 2010 Sep, 15;185(6):3277-84.<br />

Weber CK, Haslbeck M, Englbrecht M, Sehnert B, Mielenz D, Graef D, Distler JH, Mueller<br />

RB, Burkhardt H, Schett G, Voll RE, Fürnrohr BG. Antibodies to the endoplasmic reticulumresident<br />

chaperones calnexin, BiP and Grp94 in patients with rheumatoid arthritis and<br />

systemic lupus erythematosus. Rheumatology (Oxford). 2010 Aug 27. [Epub ahead of print]<br />

van Bavel CC, Dieker JW, Kroeze Y, Tamboer WP, Voll R, Muller S, Berden JH, van der<br />

Vlag J. Apoptosis-induced histone H3 methylation is targeted by autoantibodies in systemic<br />

lupus erythematosus. Ann Rheum Dis. 2010 Aug 10. [Epubahead of print]<br />

Meister S, Frey B, Lang VR, Gaipl US, Schett G, Schlötzer-Schrehardt U, Voll RE. Calcium<br />

channel blocker verapamil enhances endoplasmic reticulum stress and cell death induced by<br />

proteasome inhibition in myeloma cells. Neoplasia. 2010, Jul;12(7):550-61.<br />

Zaiss MM, Sarter K, Hess A, Engelke K, Böhm C, Nimmerjahn F, Voll R, Schett G, David JP.<br />

Increased bone density and resistance to ovariectomy-induced bone loss in FoxP3transgenic<br />

mice based on impaired osteoclast differentiation. Arthritis Rheum. 2010<br />

Aug;62(8):2328-38.<br />

Fürnrohr BG, Wach S, Kelly JA, Haslbeck M, Weber CK, Stach CM, Hueber AJ, Graef D,<br />

Spriewald BM, Manger K, Herrmann M, Kaufman KM, Frank SG, Goodmon E, James JA,<br />

Schett G, Winkler TH, Harley JB, Voll RE. Polymorphisms in the Hsp70 gene locus are<br />

genetically associated with systemic lupus erythematosus. Ann Rheum Dis. 2010 May 24.<br />

[Epub ahead of print]<br />

Vogelbacher R, Meister S, Gückel E, Starke C, Wittmann S, Stief A, Voll R, Daniel C, Hugo<br />

C. Bortezomib and sirolimus inhibit the chronic active antibody-mediated rejection in<br />

experimental renal transplantation in the rat. Nephrol Dial Transplant. 2010 Apr 28. [Epub<br />

ahead of print]<br />

Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE,<br />

Zychlinsky A. Impairment of neutrophil extracellular trap degradation is associated with lupus<br />

nephritis. Proc Natl Acad Sci U S A. 2010 May, 25;107(21):9813-8. Epub 2010 May 3.<br />

Kruse K, Janko C, Urbonaviciute V, Mierke CT, Winkler TH, Voll RE, Schett G, Muñoz LE,<br />

Herrmann M. Inefficient clearance of dying cells in patients with SLE: anti-dsDNA<br />

autoantibodies, MFG-E8, HMGB-1 and other players. Apoptosis. 2010, Sep;15(9):1098-<br />

113.<br />

116


Rödel F, Frey B, Capalbo G, Gaipl U, Keilholz L, Voll R, Hildebrandt G, Rödel, C.<br />

Discontinuous induction of X-linked inhibitor of apoptosis in EA.hy.926 endothelial cells is<br />

linked to NF-kappaB activation and mediates the anti-inflammatory properties of low-dose<br />

ionising-radiation. Radiother Oncol. 2010 Feb 17. [Epub ahead of print]<br />

2009<br />

Rödel F, Keilholz L, Herrmann M, Weiss C, Frey B, Voll R, Gaipl U, Rödel C. Activator<br />

protein 1 shows a biphasic induction and transcriptional activity after low dose X-irradiation in<br />

EA.hy.926 endothelial cells. Autoimmunity. 2009, May;42(4):343-5.<br />

Krönke G, Katzenbeisser J, Uderhardt S, Zaiss MM, Scholtysek C, Schabbauer G, Zarbock<br />

A, Koenders MI, Axmann R, Zwerina J, Baenckler HW, van den Berg W, Voll RE, Kühn H,<br />

Joosten LA, Schett G. 12/15-lipoxygenase counteracts inflammation and tissue damage in<br />

arthritis. J Immunol. 2009 Sep 1;183(5):3383-9.<br />

Rödel F, Keilholz L, Herrmann M, Weiss C, Frey B, Voll R, Gaipl U, Rödel C. Activator<br />

protein 1 shows a biphasic induction and transcriptional activity after low dose X-irradiation in<br />

EA.hy.926 endothelial cells. Autoimmunity. 2009 May;42(4):343-5.<br />

Urbonaviciute V, Meister S, Fürnrohr BG, Frey B, Gückel E, Schett G, Herrmann M, Voll RE.<br />

Oxidation of the alarmin high-mobility group box 1 protein (HMGB1) during apoptosis.<br />

Autoimmunity. 2009 May;42(4):305-7.<br />

Muñoz LE, Janko C, Grossmayer GE, Frey B, Voll RE, Kern P, Kalden JR, Schett G, Fietkau<br />

R, Herrmann M, Gaipl US. Remnants of secondarily necrotic cells fuel inflammation in<br />

systemic lupus erythematosus. Arthritis Rheum. 2009 Jun;60(6):1733-42.<br />

Raaz D, Herrmann M, Ekici AB, Klinghammer L, Lausen B, Voll RE, Leusen JH, van de<br />

Winkel JG, Daniel WG, Reis A, Garlichs CD. FcgammaRIIa genotype is associated with<br />

acute coronary syndromes as first manifestation of coronary artery disease.<br />

Atherosclerosis. 2009 Jan 21 [Epub ahead of print].<br />

Books and Reviews<br />

Urbonaviciute V, Voll RE: HMGB1 represents a potential marker of disease activity and novel<br />

therapeutic target in SLE. J Intern Med. 2011 Jul 27. doi: 10.1111/j.1365-<br />

2796.2011.02432.x. [Epub ahead of print]<br />

Feuchtenberger M, Voll RE, Kneitz C.: Impfungen in der Rheumatologie. Z Rheumatol. 2010<br />

Nov;69(9):803-812.<br />

Nikolova KA, Mihaylova NM, Voynova EN, Tchorbanov AI, Voll RE, Vassilev TL. Selective<br />

silencing of autoreactive B lymphocytes-Following the Nature's way. Autoimmun Rev. 2010<br />

Sep;9(11):775-9. Epub 2010 Jul 1.<br />

Weiss EM, Frey B, Rödel F, Herrmann M, Schlücker E, Voll RE, Fietkau R, Gaipl US.: Ex<br />

vivo- and in vivo-induced dead tumor cells as modulators of antitumor responses. Ann N Y<br />

Acad Sci. 2010 Oct;1209:109-17.<br />

Muñoz LE, Lauber K, Schiller M, Manfredi AA, Schett G, Voll RE, Herrmann M.[The role of<br />

incomplete clearance of apoptotic cells in the etiology and pathogenesis of SLE]. Z<br />

Rheumatol. 2010 Mar;69(2):152, 154-6.<br />

117


Voll R, Hiepe F. [Depletion of plasma cells - a novel strategy in the therapy of systemic lupus<br />

erythematosus in mice and man.]. Z Rheumatol. 2009, Mar;68(2):150-3.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Eva Gückel<br />

Die Rolle des Transkriptionsfaktors NF-kB <strong>für</strong> die Entwicklung und Funktion natürlich<br />

vorkommender regulatorischer T-Zellen<br />

Charlotte Starkte<br />

Charakterisierung von Antikörper-sezernierenden Zellen in der entzündeten Niere im<br />

NZB/W-F1 Lupusmausmodell.<br />

Doctoral theses (Molecular Medicine)<br />

Sandy Pohle<br />

Influence of dietary salt intake on the immune response and inflammatory diseases<br />

Doctoral theses (Medicine)<br />

Veronika Lang<br />

Resistenz der von Marginalzonen B- Zellen initiierten frühen Phase der T-Zellunabhängigen<br />

Immunantwort Typ 2 gegenüber dem Proteasominhibitor Bortezomib<br />

Ramona Peukert<br />

Proteasominhibition zur Therapie des SLE<br />

Christian Weber<br />

ER-Chaperone als Autoantigene bei der chronischen Polyarthritis<br />

118


DEPARTMENT OF ORTHOPAEDIC TRAUMA<br />

SURGERY<br />

(Molecular Cartilage <strong>Research</strong>)<br />

Head: Prof. Dr. med. F.F. Hennig<br />

(Head of Department of Orthopaedic Trauma Surgery)<br />

Priv.-Doz. Dr. med. K. Gelse<br />

(Leader of <strong>Research</strong> Group)<br />

Address: Department of Orthopaedic Trauma Surgery, Molecular<br />

Cartilage <strong>Research</strong> Group)<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 42121<br />

Fax: + 49 (9131) 85 33300<br />

E-mail: kolja.gelse@uk-erlangen.de<br />

Patricia Klinger, Dr. rer. nat.<br />

Head<br />

Friedrich F. Hennig, Prof. Dr. med.<br />

Professor of Orthopaedic Trauma Surgery<br />

Leader of <strong>Research</strong> Group<br />

Kolja Gelse, Priv.-Doz. Dr. med.<br />

Postdoctoral Fellows<br />

119


Matthias Koch<br />

Franziska Cipa<br />

Melanie Pflügner<br />

<strong>Research</strong><br />

Doctoral Students (Medicine)<br />

Simon Obier<br />

Technicians<br />

Our group analyses molecular mechanisms that control cartilage and bone biology<br />

focussing on the mechanisms of chondrogenesis and terminal chondrocyte<br />

differentiation. The investigations further include the role of hypoxia and epigenetic<br />

mechanisms on the differentiation of chondroprogenitor cells and mesenchymal stem<br />

cells.<br />

Role of HIF-1α for the chondrogenic phenotype<br />

The aim of this project was to investigate the chondrogenic potential of growth factorstimulated<br />

periosteal cells with respect to the activity of Hypoxia-inducible Factor 1a<br />

(HIF-1a). In this project, scaffold-bound autologous periosteal cells, which had been<br />

activated by Insulin-like Growth Factor 1 (IGF-1) or Bone Morphogenetic Protein 2<br />

(BMP-2) gene transfer using both adeno-associated virus (AAV) and adenoviral (Ad)<br />

vectors, were applied to chondral lesions in the knee joints of miniature pigs. Six<br />

weeks after transplantation, the repair tissues were investigated for collagen type I<br />

and type II content as well as for HIF-1a expression. The functional role of<br />

phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) and<br />

mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK)<br />

signaling on BMP-2/IGF-1-induced HIF-1a expression was assessed in vitro by<br />

employing specific inhibitors.<br />

Unstimulated periosteal cells formed a fibrous extracellular matrix in the superficial<br />

zone and a fibrocartilaginous matrix in deep zones of the repair tissue. This zonal<br />

difference was reflected by the absence of HIF-1a staining in superficial areas, but<br />

moderate HIF-1a expression in deep zones. In contrast, Ad/AAVBMP-2-stimulated<br />

periosteal cells, and to a lesser degree Ad/AAVIGF-1-infected cells, adopted a<br />

120


chondrocyte-like phenotype with strong intracellular HIF-1a staining throughout all<br />

zones of the repair tissue and formed a hyaline-like matrix. In vitro, BMP-2 and IGF-1<br />

supplementation increased HIF-1a protein levels in periosteal cells, which was based<br />

on posttranscriptional mechanisms rather than de novo mRNA synthesis, involving<br />

predominantly the MEK/ERK pathway. The data of this project indicated that<br />

chondrogenesis by chondrogenic precursor cells is facilitated in deeper hypoxic<br />

zones of cartilage repair tissue and is stimulated by growth factors which enhance<br />

HIF-1a activity.<br />

Fig. 1.<br />

O2<br />

Toluidine<br />

blue<br />

Hypoxia<br />

HIF-1α<br />

Hypoxia facilitates<br />

chondrogenesis.<br />

Mesenchymal stem<br />

cells overexpressing<br />

BMP-2 were<br />

transplanted into<br />

cartilage defects in<br />

the knee joints of a<br />

minipig model.<br />

Chondrogenesis was<br />

facilitated in deep<br />

hypoxic tissue layers.<br />

The role of anti-angiogenic factors for stabilizing the<br />

chondrocyte phenotype<br />

This study investigated the effect of the anti-angiogenic factors Thrombospondin-1<br />

(TSP-1) and Chondromodulin-I (Chm-I) on the formation of cartilage repair tissue in<br />

combination with stimulation by osteogenic protein-1 (OP-1). In miniature pigs,<br />

articular cartilage lesions in the femoral trochlea were treated by the microfracture<br />

technique and either received no further treatment (MFX), or were treated by<br />

additional application of recombinant osteogenic protein-1 (MFX+OP1), recombinant<br />

TSP-1 (MFX+TSP1), or a combination of both proteins (MFX+TSP1+OP1). Since<br />

Chm-I is not available as a recombinant protein, we applied self-complementary<br />

Adeno-Associated-Virus (AAV) vectors carrying the Chm-I cDNA to achieve<br />

overexpression of Chm-I within the defects. Six and 26 weeks after surgery, the<br />

repair tissue and the degree of endochondral ossification were assessed by<br />

histochemical and immunohistochemical methods detecting collagen types I, II, X,<br />

TSP-1 and CD31. Microfracture treatment merely induced the formation of inferior<br />

fibrocartilaginous repair tissue. OP-1 stimulated chondrogenesis, but also induced<br />

chondrocyte hypertrophy, characterized by synthesis of collagen type X, and<br />

excessive bone formation. Application of TSP-1 inhibited inadvertant endochondral<br />

ossification, but failed to induce chondrogenesis. In contrast, the simultaneous<br />

121


application of both TSP-1 and OP-1 induced and maintained a permanent, nonhypertrophic<br />

chondrocyte-like phenotype within cartilage repair tissue. Similar<br />

favourable results were observed following Chm-I gene transfer. Overexpression of<br />

Chm-I stabilized the chondrocyte phenotype within the repair tissue and prevented<br />

excessive endochondral ossification. Gene expression analyses in vitro revealed the<br />

cell cycle inhibitor p21 cip1/waf1 as one target upregulated by AAVChm-I. The data of<br />

this study demonstrate that treatment by a combination of OP-1 and TSP-1, as well<br />

as overexpression of Chm-I stabilizes the chondrocyte phenotype by supporting<br />

chondrogenesis but inhibiting chondrocyte hypertrophy and endochondral<br />

ossification.<br />

Molecular differentiation between transient osteophytic and<br />

permanent articular cartilage<br />

The aim of this project to identify the molecular differences between the transient and<br />

permanent chondrocyte phenotype in osteophytic and articular cartilage. Total RNA<br />

was isolated from the cartilaginous layer of osteophytes and from intact articular<br />

cartilage from knee joints of 15 adult human donors and subjected to cDNA<br />

microarray analysis. The differential expression of relevant genes between these two<br />

cartilaginous tissues was additionally validated by quantitative RT-PCR and by<br />

immunohistochemistry. Among 47,000 screened transcripts, 600 transcripts were<br />

differentially expressed between osteophytic and articular chondrocytes. Osteophytic<br />

chondrocytes were characterized by increased expression of genes involved in the<br />

endochondral ossification process (BGLAP, BMP8B, COL1A2, SOST, GADD45ß,<br />

Runx2), and genes encoding tissue remodelling enzymes (MMP-9, -13, HAS1).<br />

Articular chondrocytes expressed increased transcript levels of antagonists and<br />

inhibitors of the BMP- and Wnt-signalling pathways (GREM1, FRZP, WISP3), as well<br />

as factors that inhibit terminal chondrocyte differentiation and endochondral bone<br />

formation (PTHLH, SOX9, STC2, S100A1, S100B). Immunohistochemistry of tissue<br />

sections for GREM1 and BGLAP, the two most prominent differentially expressed<br />

genes, confirmed selective detection of GREM1 in articular chondrocytes and that of<br />

BGLAP in osteophytic chondrocytes and bone.<br />

As a conclusion, osteophytic and articular chondrocytes significantly differ in their<br />

gene expression pattern. In articular cartilage, a prominent expression of antagonists<br />

inhibiting the BMP- and Wnt-pathway may serve to lock and stabilize the permanent<br />

chondrocyte phenotype and thus prevent their terminal differentiation. In contrast,<br />

osteophytic chondrocytes express genes with roles in the endochondral ossification<br />

process, which may account for their transient phenotype.<br />

Epigenetic mechanisms for stabilizing the chondrocyte<br />

phenotype<br />

This study investigates the differences in epigenetic mechanisms between transient<br />

and permanent cartilage. We performed a Methylation Array which demonstrated<br />

distinct differences in the epigenetic pattern of articular chondrocytes and<br />

chondrocytic differentiated mesenchymal stem cells. The promotor region of FoxS1<br />

was one of the most striking regions with a high degree of methylation in articular<br />

chondrocytes. The hypothesis of this study is that an epigenetic suppression of<br />

122


FoxS1 increases the transcriptional activity of Foxo3a which stabilizes the postmitotic<br />

chondrocyte phenotype. Currently, we further analyse the promotor region of Foxs1<br />

by Pyrosequencing and the role of DNA-methyltransferases. The aim of this project is<br />

to evaluate the methylation status as tool for screening the potency of chondrogenic<br />

differentiation of mesenchymal stem cells.<br />

Publications (2009-2011)<br />

Original Articles<br />

Gelse K, Ekici, AB, Cipa F, Swoboda B, Carl HD, Olk A, Hennig FF, Klinger P.<br />

Molecular differentiation between osteophytic and articular cartilage – clues for a<br />

transient and permanent chondrocyte phenotype. Osteoarthritis Cartilage.<br />

2012;20(2) :162-71.<br />

Gelse K, Klinger P, Koch M, Surmann-Schmitt C, von der Mark K, Swoboda B,<br />

Hennig FF, Gusinde J. Thrombospondin-1 prevents excessive ossification in cartilage<br />

repair tissue induced by osteogenic protein-1. Tissue Eng Part A. 2011 Aug;17(15-<br />

16):2101-12.<br />

Klinger P, Schietke RE, Warnecke C, Swoboda B, Wiesener M, Hennig FF, Gelse K.<br />

Deletion of the oxygen-dependent degradation domain results in impaired<br />

transcriptional activity of hypoxia-inducible factors. Transcription. 2011 Nov<br />

1;2(6):269-75.<br />

Klinger P, Surmann-Schmitt C, Brem M, Swoboda B, Distler JH, Carl HD, von der<br />

Mark K, Hennig FF, Gelse K. Chondromodulin 1 stabilizes the chondrocyte<br />

phenotype and inhibits endochondral ossification of porcine cartilage repair tissue.<br />

Arthritis Rheum. 2011 Sep;63(9):2721-31.<br />

Gelse K, Olk A, Eichhorn S, Swoboda B, Schoene M, Raum K. Quantitative<br />

ultrasound biomicroscopy for the analysis of healthy and repair cartilage tissue. Eur<br />

Cell Mater. 2010;19:58-71.<br />

Blanke M, Carl HD, Klinger P, Swoboda B, Hennig F, Gelse K. Transplanted<br />

chondrocytes inhibit endochondral ossification within cartilage repair tissue. Calcif<br />

Tissue Int. 2009;85(5):421-33.<br />

Gelse K, Brem M, Klinger P, Hess A, Swoboda B, Hennig F, Olk A. Paracrine effect<br />

of transplanted rib chondrocyte spheroids supports formation of secondary cartilage<br />

repair tissue. J Orthop Res. 2009;27(9):1216-25.<br />

123


Books and Reviews<br />

Gelse K, Beyer C. The prostaglandin E(2) system: a toolbox for skeletal repair?<br />

Arthritis Rheum. 2011 Apr;63(4):871-3.<br />

Gelse K, Schneider H. In vivo evaluation of gene transfer into mesenchymal cells (in<br />

view of cartilage repair). Methods Mol Biol. 2011;737:391-405.<br />

Linke R, Gelse K, Schuch F. The indication for radiosynoviorthesis. From the<br />

perspective of the nuclear medicine expert, rheumatic orthopedist and internist. Z<br />

Rheumatol. 2011 Jan;70(1):34-44.<br />

Gelse K, Swoboda B. Zell- und Gentherapie – eine kurative Therapieoption bei<br />

Arthrose? Der Internist 2009;50:73.<br />

Gelse K, Swoboda B. Therapie der Arthrose. Der Allgemeinarzt 2009;5:36-40.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Medicine)<br />

Simon Obier<br />

Die Rolle des Transkriptionsfaktors HIF-1a <strong>für</strong> die Integrität des murinen<br />

Gelenkknorpels<br />

Matthias Koch<br />

Die Rolle von Thrombospondin-1 <strong>für</strong> die Stabilität des Knorpelzellphänotyps<br />

Franziska Cipa<br />

<strong>Molekulare</strong>r Vergleich von „transienten“ Osteophytenknorpel und „permanenten“<br />

Gelenkknorpel<br />

124


DEPARTMENT OF PLASTIC AND HAND SURGERY<br />

(Tissue Engineering)<br />

Dr. Subha N. Rath<br />

Leonie Strobel<br />

Daniel Hiller<br />

Head: Raymund E. Horch, Prof. Dr. med.<br />

Vice chairman: Ulrich Kneser, PD Dr. med.<br />

Amelie Balzer<br />

Jennifer Covi<br />

Address: Department of Hand and Plastic Surgery<br />

Krankenhausstraße 12<br />

D-91054 Erlangen<br />

E-mail: ulrich.kneser@uk-erlangen.de<br />

Postdoctoral Fellows<br />

Dr. Andreas Brandl<br />

Doctoral Students (Medicine)<br />

Quan Yuan<br />

Johannes Hilgert<br />

Gregor Bührer<br />

Doctoral Students (Veterinary Medicine)<br />

125


Stefan Fleischer<br />

Technicians<br />

Marina Milde<br />

Guest Scientists<br />

126<br />

Ilse Arnold


<strong>Research</strong><br />

Since the late 1980s, Tissue Engineering has been one of the key technologies in<br />

regenerative medicine. Regarding bone tissue, an approach based on scaffolds as<br />

carriers for patient’s own cells and suitable growth factors has been proposed. Our<br />

group focuses on engineering bioartificial bone tissues and vascularisation models.<br />

The combination of smart biomaterials, osteoinductive factors and osteogenic cells<br />

lead to new perspectives in bone regeneration therapy. Endothelial cell (EC) culture,<br />

osteoblast (OB) culture and marrow stromal cell (MSC) culture are utilised in vitro.<br />

Furthermore, we use in vivo subcutaneous models and vascularisation models (e.g.<br />

arteriovenous loop model) in rats (in cooperation with Franz-Penzoldt-<strong>Zentrum</strong>).<br />

Bone Tissue Engineering<br />

Bone defects and impaired bone healing represent substantial challenges in<br />

reconstructive surgery. By bone tissue engineering it may be possible to heal<br />

complex bone defects in the future. Our group works on the application of novel<br />

biomaterials, innovative cell culture models and co-cultures.<br />

Induction of bone formation in biphasic calcium phosphate ceramics<br />

Subha N. Rath, Leonie Strobel, Ulrich Kneser<br />

In cooperation with the Institute of Materials Science (Prof. P. Greil), Erlangen<br />

Novel biphasic calcium phosphate (BCP) matrices were generated by 3D printing and<br />

high porosity was achieved by starch consolidation (Department of Materials Science<br />

(Glass and Ceramics), FAU Erlangen). This project aimed to characterise the porous<br />

BCP scaffold’s properties and the interaction of osteogenic cells and growth factors<br />

under in vitro and in vivo conditions.<br />

Bioreactors are advised for improved cell survival, as they are able to provide a<br />

controlled flow through the scaffold. Osteogenic cells (rat primary osteoblasts (OB)<br />

and bone marrow stromal cells (MSCs)) were isolated, cultured and seeded onto 3D<br />

scaffolds. Samples in static culture without osteoinduction, static culture with<br />

osteoinduction and dynamic culture with osteoinduction were compared over 6<br />

weeks. Analyses showed that a continuous flow bioreactor not only preserves the<br />

number of osteogenic cells, but also keeps their differentiation ability in balance<br />

providing a suitable cell-seeded scaffold product for applications in regenerative<br />

medicine (Rath, Strobel et al. 2012).<br />

Furthermore, these very BCP scaffolds were evaluated in vivo. Five differently<br />

treated constructs were implanted subcutaneously in syngeneic male Lewis rats:<br />

127


plain BCP constructs, constructs pre-treated with BMP-2, seeded with osteoblasts<br />

(OB), seeded with OB and BMP-2 and constructs with OB that were pre-cultivated in<br />

a flow bioreactor for 6 weeks. Specimens were explanted and subjected to histology<br />

and molecular biological analysis after 2, 4 and 6 weeks.<br />

All explanted constructs were invaded by fibrovascular tissue. Samples with both OB<br />

and BMP-2 demonstrated higher osteogenic gene expression and significantly<br />

increased bone formation compared to all other groups. Pre-cultivation in the flow<br />

bioreactor induced bone formation comparable with the freshly seeded group. In<br />

summary, the combination of OB and BMP-2 enhanced bone formation in the novel<br />

ceramic scaffolds (Strobel, Rath et al. 2012).<br />

This work was supported by the German <strong>Research</strong> Foundation (DFG).<br />

Rath SN, Strobel LA, Meier AK. 2012, Osteoinduction of osteoblasts and bone marrow stromal cells in<br />

3D BCP scaffolds: the effect of bioreactor on cell survival and differentiation, Journal of<br />

Cellular and Molecular Medicine; 2012 Feb 3. doi: 10.1111/j.1582-4934.2012.01545.x.<br />

Strobel LA, Rath SN, Maier AK. 2012, Induction of Bone Formation in Biphasic Calcium Phosphate<br />

Scaffolds by Bone Morphogenetic Protein-2 and Primary Osteoblasts Journal of Tissue<br />

Engineering and Regenerative Medicine; (accepted)<br />

Haematoxylin-eosin (HE) staining of an explanted biphasic calcium phosphate (BCP) scaffold at 6<br />

weeks after subcutaneous implantation. The scaffold was seeded with primary osteoblasts previous to<br />

implantation. Newly formed bone areas (arrows) are visible in the pores of the BCP construct.<br />

Human marrow stromal cells (hMSCs) on innovative bioglass scaffolds*<br />

Subha N. Rath, Daniel Hiller, Ulrich Kneser<br />

In cooperation with the Institute of Biomaterials (Prof. A. Boccaccini), Erlangen<br />

The aim of this study is to analyse hMSCs response to new bioactive copper-doped<br />

glass scaffolds. 45S5 bioglass scaffolds containing different concentrations of copper<br />

are provided by the Institute of Biomaterials. First, these scaffolds were evaluated<br />

regarding copper and bioactive ion release in vitro. For this purpose, we observed 2dimensional<br />

cell culture in vicinity to bioactive glass. The constructs proved non-toxic<br />

and provided continuous release of copper (depending on the initial copperconcentration).<br />

Furthermore, scaffolds are seeded with hMSCs and evaluated under<br />

128


different culture conditions. This will provide the basis for further in vivo studies with<br />

bioactive glass materials.<br />

Bioglass scaffolds in the arteriovenous (AV) loop model*<br />

Amelie Balzer, Gregor Bührer, Andreas Arkudas<br />

Generation of axially vascularised bone tissue engineering constructs in vivo is<br />

performed using an arteriovenous (AV) loop model. After bioactive glass scaffolds<br />

proved biocompatible in vitro, scaffolds are evaluated in the AV loop model in rats.<br />

Different scaffold materials, osteogenic cells and bioactive molecules such as bone<br />

morphogenetic protein-2 (BMP-2) can be incorporated in the matrix that is inserted in<br />

the AV-loop (in cooperation with Franz-Penzoldt-<strong>Zentrum</strong>, Erlangen).<br />

Co-cultures of human marrow stromal cells (hMSCs) and endothelial cells<br />

(hECs) in vitro*<br />

Subha N. Rath, Andreas Brandl, Oliver Bleiziffer, Ulrich Kneser<br />

Previous reports have shown that hMSCs can be osteo-differentiated into boneforming<br />

cells and thus can be used for bone tissue engineering. However, when<br />

applied in vivo in larger dimensions, the lack of development of neovascularization<br />

into the defect substantially limits their survival. It was also reported that hECs when<br />

cultured in vitro forms endothelial tube like structures, which vascularizes upon in<br />

vivo implantation to host blood vessels to supply blood relatively quicker than without<br />

hECs.<br />

Therefore, the combined co-culture of hMSCs and hECs can be attempted for a bone<br />

tissue engineered product which can later be vascularized upon in vivo implantation.<br />

A perfect scaffold material to study the effect is in bioactive glass, which not only<br />

have osteoconductive property, but also have vascularization inducing ability. To<br />

study this effect, our group developes co-culture models to investigate the<br />

interactions and influences of these cells upon each other and the effect of bio-active<br />

glass in inducing VEGF from hMSCs.<br />

*These projects are supported by the Emerging Fields Initiative, FAU Erlangen.<br />

Neovascularization of bioartificial tissues<br />

Neovascularization of tissues and organs is based upon two distinct processes: de<br />

novo formation of blood vessels via the assembly of progenitor cells (vasculogenesis)<br />

and expansion of pre-existing vascular networks by endothelial cell sprouting<br />

(angiogenesis). The latter one is the main mechanism in postnatal life. Evidence exist<br />

that bone marrow-derived progenitor cells can contribute to the formation of new<br />

vessels by incorporation into sites of active angiogenesis (Rüger et al.; 2008). The<br />

discovery of putative endothelial progenitor cells (EPC) gave rise to a new era of<br />

vascular biology to use their highly promising multipotent character for therapeutic<br />

angiogenesis equally with regard to tissue engineering where they could be used as<br />

129


a source to establish a reliable perfusion of bioartificial tissues and organs. Bone<br />

marrow-derived progenitors are known to be heterogenous. Therefore it is absolutely<br />

necessary to clearly characterize isolated populations from bone marrow that are<br />

designated to be EPCs, with regard to their morphology and performance in<br />

endothelial-specific functional assays in vitro prior to using them in in vivo studies.<br />

Effect of rat primary EPCs on neovascularization using the (AV) loop model*<br />

Andreas Brandl, Quan Yuan, Oliver Bleiziffer<br />

Previous studies used a mouse endothelial progenitor cell (EPC) line (T17b) to study<br />

vascularization in a highly standardarized (AV) loop model in rat. One aim of this<br />

project is to isolate and characterize a bone marrow-derived EPC-population from rat<br />

to obtain an allogenic cell population to study angiogenesis in this well-established in<br />

vivo system.<br />

First results show that rat bone marrow-derived mononuclear cells can be<br />

differentiated into E(P)Cs in vitro. MNCs (mononuclear cells) from rat bone marrow<br />

were isolated using density gradient centrifugation and a differential adhesion<br />

method as described in Kähler et al., 2007. Cells were followed up concerning their<br />

morphology for several weeks and they exhibit the typical “cobblestone”-shape after<br />

about two to three weeks in culture (Fig.1).<br />

a) b)<br />

Fig.1: Mononuclear cells from rat bone marrow ( a) day of isolation) can be differentiated<br />

into endothelial cells ( b) 3 weeks post isolation) in vitro using special differentiation<br />

medium.<br />

The cells were also characterized with regard to their surface marker expression<br />

using flow cytometric analyses and their ability to form tube-like structures on<br />

Matrigel. The flow cytometry analyses showed that the cultivated cells gain<br />

endothelial-specific surface marker expression like CD31, VEGFR-2 and CD146 in<br />

culture. Four weeks old EPC are able to form tube-like structures and sprouts on the<br />

130


specialized extracellular matrix (ECM) Matrigel as potent as pure endothelial cell<br />

lines (HDMECs and EC52) do (Fig. 2).<br />

a) b)<br />

Fig.2: Isolated primary endothelial cells ( a) 4 weeks after isolation) perform just as pure<br />

commercially available cell lines ( b) HDMECs) in sprouting experiments on the<br />

specialized extracellular matrix (ECM) Matrigel.<br />

Future experiments will investigate the ability of these primary endothelial cells to<br />

efficiently support or promote neovascularization in bioartificial tissues using the wellestablished<br />

AV-loop model.<br />

Distribution and Impact of Hypoxia on blood vessel formation in a developing<br />

three-dimensional vascular network in the AV-Loop HIF investigated by<br />

detection of hypoxia-inducible factor 1 alpha (HIF-1 α)<br />

Hypoxia inducible factor (HIF) is a transcriptional factor that is the master regulator of<br />

oxygen homeostasis and plays an essential role in angiogenesis. In normoxic<br />

conditions, HIF could be degraded by prolyl hydroxylases (PHDs). Suppression of<br />

PHD activity increases endogenous HIF levels, which is probably able to promote the<br />

angiogenesis. Dimethyloxallyl Glycine (DMOG) is a cell permeable, competitive<br />

inhibitor of prolyl hydroxylase domain-containing proteins (PHDs and HIF-PHs).<br />

Future experiments will apply DMOG in Lewis rats with AV-loop systemically; detect<br />

the angiogenesis-related target genes such as HIF-1a, HIF-2a, VEGFA, eNOS, Flt-1,<br />

VE-Cadherin, KDR and vWF at transcription level in the loop at certain time points,<br />

and evaluate the outgrowth of the capillary network with morphology methods.<br />

* These projects are supported by the Else-Kröner-Fresenius-Stiftung.<br />

131


Original Articles<br />

Rath SN, Strobel LA, Meier AK. 2012, Osteoinduction of osteoblasts and bone marrow stromal cells in<br />

3D BCP scaffolds: the effect of bioreactor on cell survival and differentiation, Journal of<br />

Cellular and Molecular Medicine; 2012 Feb 3. doi: 10.1111/j.1582-4934.2012.01545.x.<br />

Strobel LA, Rath SN, Maier AK. 2012, Induction of Bone Formation in Biphasic Calcium Phosphate<br />

Scaffolds by Bone Morphogenetic Protein-2 and Primary Osteoblasts Journal of Tissue<br />

Engineering and Regenerative Medicine; (accepted)<br />

Bleiziffer O, Horch RE, Hammon M, Arkudas A, Naschberger E, Rath S, Pryymachuk G, Beier JP,<br />

Hatzopoulos AK, Stürzl M, Kneser U, T17b murine embryonal endothelial progenitor cells can<br />

be induced towards both proliferation and differentiation in a fibrin matrix. J Cell Mol Med.<br />

2009; 13: 926-35.<br />

Bleiziffer O, Hammon M, Naschberger E, et al. Endothelial Progenitor Cells are integrated in newly<br />

formed capillaries and alter adjacent fibrovascular tissue after subcutaneous implantation in a<br />

fibrin matrix. J Cell Mol Med 2010; doi: 10.1111/j.1582-4934.2010.01247.x.<br />

Arkudas A, Tjiawi J, Saumweber A, Beier JP, Polykandriotis E, Bleiziffer O, Horch RE, Kneser U.<br />

Evaluation of blood vessel ingrowth in fibrin gel subject to type and concentration of growth<br />

factors. J Cell Mol Med. 2009; 13: 2864-74<br />

Arkudas A, Pryymachuk G, Hoereth T, Beier JP, Polykandriotis E, Bleiziffer O, Horch RE, Kneser U.<br />

Dose-finding study of fibrin gel-immobilized vascular endothelial growth factor 165 and basic<br />

fibroblast growth factor in the arteriovenous loop rat model. Tissue Eng Part A. 2009; 15:<br />

2501-11.<br />

Fiegel HC, Pryymachuk G, Rath S, Bleiziffer O, Beier JP, Bruns H, Kluth D, Metzger R, Horch RE, Till<br />

H, Kneser U. Foetal hepatocyte transplantation in a vascularized AV-Loop transplantation<br />

model in the rat. J Cell Mol Med. 2010; 14: 267-74.<br />

Arkudas A, Beier JP, Pryymachuk G, Hoereth T, Bleiziffer O, Polykandriotis E, Hess A, Gulle H, Horch<br />

RE, Kneser U. Automatic quantitative micro-computed tomography evaluation of angiogenesis<br />

in an axially vascularized tissue-engineered bone construct. Tissue Eng Part C Methods.<br />

2010; 16: 1503-14.<br />

132


AG Krönke<br />

(Department of Internal Medicine 3)<br />

Principal Investigator: Gerhard Krönke, Dr. med.<br />

Address: AG Krönke<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 43012<br />

E-mail: gerhard.kroenke@uk-erlangen.de<br />

Homepage: www..medizin3.ukerlangen.de/e1846/e741/e614/index_ger.html<br />

Carina Scholtysek<br />

Tobias Rothe<br />

StefanUderhardt°<br />

Arnd Kleyer<br />

Susanne Weig<br />

°MD received<br />

Cornelia Stoll<br />

Martin Steffen<br />

Doctoral Students (Biology)<br />

Natacha Ipseiz<br />

Réne Pfeifle<br />

Students (Medicine)<br />

Tobias Fillep<br />

Technicians<br />

Isabell Schmidt<br />

133<br />

Edith Bottesch


<strong>Research</strong><br />

Our group is interested in common mechanisms regulating the initiation and<br />

resolution of the inflammatory and adaptive immune response. Our aim is to<br />

elucidate the pathogenesis of inflammatory and autoimmune diseases and the<br />

consequences of chronic inflammation on tissue homeostasis. To answer these<br />

questions, we focus on the role and function of cells of the monocytic lineage such as<br />

macrophages, dendritic cells and osteoclasts as well as on the contribution of lipid<br />

mediators and lipid-activated nuclear receptors to inflammation, immunity and<br />

regulation of tissue homeostasis.<br />

Regulation of the immune response by lipid mediators<br />

Lipids such as fatty acids, phospholipids and triglycerides are not only an integral<br />

component of cellular membranes and essential for energy homeostasis, but are also<br />

involved in the regulation of inflammation and the adaptive immune response.<br />

Multiple enzymes including different cyclooxygenases and lipoxygenases metabolize<br />

lipids such as free and esterified polyunsaturated fatty acids and thereby generate<br />

bioactive lipid mediators including prostaglandins, leukotriens and lipoxins as well as<br />

less well characterized species of distinct phospholipid oxidation products. Such lipid<br />

mediators act via multiple mechanisms, which include both activation and inhibition of<br />

different cell-surface receptors and of nuclear receptors that are involved in the<br />

modulation of the innate and adaptive immune response.<br />

Enzymatic lipid oxidation orchestrates the resolution of inflammation<br />

Gerhard Krönke<br />

Inflammation is the physiologic response to infection and tissue damage,<br />

respectively. The inflammatory response, however, has to be tightly regulated and<br />

controlled, since overwhelming and prolonged inflammation would exacerbate tissue<br />

damage and prevent the healing process. Typically, different mechanisms cooperate<br />

to initiate the resolution of inflammation. Interestingly, different lipid oxidation<br />

products such as lipoxin A4 have been recognized as major factors driving this<br />

resolving process.<br />

To study the role of lipoxin A4 during the pathogenesis of chronic inflammatory<br />

diseases, we are analyzing mice with a targeted deletion in the gene for 12/15lipoxygenase<br />

(12/15-LO) as the major enzyme involved in the generation of this proresolving<br />

mediator. These mice display a defect in the resolution of inflammation as<br />

well as a dramatic exacerbation of the inflammatory response as observed in two<br />

models of inflammatory arthritis (Fig. 1; Krönke et al., 2009). Likewise, 12/15-LO<br />

deficiency results in increased inflammation-associated tissue damage, in an altered<br />

healing response and overwhelming post-inflammatory fibrosis (Krönke et al., 2012).<br />

Interestingly additional data indicate that 12/15-LO is also involved in the<br />

pathogenesis of acute lung injury (Zarbock et al., 2009) as well as in the process of<br />

osteoclast differentiation (Krönke et al., 2009).<br />

134


Fig. 1 Experimental Arthritis in TNF-transgenic (TNFtg) mice exacerbates in the absence of 12/15-<br />

LO. (A) survival, (B) weight gain, (C) histological analysis of paws, (D) mRNA expression in joints and<br />

(E) clinical score of arthritis in TNFtg mice in the presence and absence of 12/15-LO.<br />

12/15-lipoxygenase orchestrates the clearance of apoptotic cells<br />

Stefan Uderhardt<br />

During the inflammatory response and the resolution of inflammation, the immune<br />

system is confronted with both pathogens and self-antigens in the form of necrotic<br />

and apoptotic cells (AC). Uptake and processing of AC by inflammatory monocytes<br />

and dendritic cells would eventually result in an immune response to self antigens.<br />

Therefore, the non-inflammatory removal and clearance of AC is essential to<br />

maintain immunologic tolerance. To study the clearance of AC in an inflammatory<br />

setting, we performed phagocytosis assays with monocytes that were isolated from<br />

murine peritonitis. Here we observed that, during inflammation, uptake of ACs is<br />

confined to a population of 12/15-LO-expressing, alternatively activated resident<br />

macrophages (resMΦ), which actively block uptake of ACs into freshly recruited<br />

inflammatory Ly6C high monocytes in a 12/15-LO-dependent manner (Fig. 2). On a<br />

mechanistic basis, we could identify novel 12/15-LO-derived oxidation-products of<br />

phosphatidylethanolamine, which are exposed on the plasma membranes of resMΦ.<br />

Thereby these cells generate a sink for distinct soluble receptors for ACs such as<br />

milk fat globule-EGF factor 8, which are essential for the uptake of ACs into<br />

inflammatory monocytes. Loss of 12/15-LO-activity, in turn, results in an aberrant<br />

phagocytosis of ACs by inflammatory monocytes, subsequent antigen-presentation of<br />

AC-derived antigens, and a lupus-like autoimmune disease (Fig. 2; Uderhardt et al.,<br />

2012). These data indicate that enzymatic phospholipid oxidation products are<br />

135


essential regulatory mediators during the clearance of AC and in the maintenance of<br />

self-tolerance.<br />

Fig. 2 12/15-LO orchestrates the clearance of apoptotic cells. (A and B) Macrophages isolated from<br />

a thioglycollate-induced peritonitis of WT and 12/15-LO-deficient mice were incubated with apoptotic<br />

cells (AC; green) and analysed by immunofluorescence. Alternatively activated resident macrophages,<br />

which ingest most of the AC in WT mice, were identified by their expression of 12/15-LO (blue in A) or<br />

Tim4 (red in B). (C) FACS-based analysis of the uptake of CFSE-labelled AC by Ly6Chigh<br />

inflammatory monocytes during a thioglycollate-induced peritonitis in vivo. (D and E) 12/15-LOdeficient<br />

mice develop a systemic autoimmune disease with signs of glomerulonephritis and different<br />

classes of autoantibodies.<br />

Oxidized phospholipids control the maturation of dendritic cells<br />

Tobias Rothe<br />

The analysis of murine bone marrow-derived dendritic cells (DC) revealed a high<br />

expression of 12/15-LO in these cells. Likewise we could identify different 12/15-LOderived<br />

phospholipid oxidation products in these cells. Our data demonstrates that<br />

this enzyme and its oxidation products critically modulate the maturation status and<br />

gene expression profile of DC. Absence of 12/15-LO results in an enhanced DC<br />

maturation, whereas addition of 12/!5-LO-specific phospholipid oxidation products<br />

interfered with the maturation process of DC. As a consequence, 12/15-LO-dficient<br />

mice show an altered activation of T-cells and an exacerbation of T-cell-driven<br />

autoimmune diseases such as experimental autoimmune encephalomyelitis (Rothe<br />

et al.; in preperation).<br />

136


Nuclear receptors as modulators of tissue homeostasis<br />

Nuclear receptors comprise a superfamily of ligand-activated transcription factors and<br />

regulate multiple homeostatic processes. Different nuclear receptors such as PPARγ<br />

have been identified as key regulators of fat and glucose metabolism as well as<br />

crucial factors influencing inflammation and the adaptive immune response.<br />

The nuclear receptor NR4A1 mediates anti-inflammatory effects of apoptotic<br />

cells<br />

Natacha Ipseiz<br />

The non-inflammatory clearance of apoptotic cells (AC) is essential to dispose selfantigens<br />

and to maintain immunologic tolerance. Mechanisms and factors<br />

contributing to this process are poorly understood, though. We observed a rapid<br />

induction of the expression of the nuclear receptor NR4A1 in macrophages ingesting<br />

AC. This induction of NR4A1 was critically involved in the anti-inflammatory effects<br />

AC exerted on macrophages. Indeed, AC efficiently blocked the inflammatory<br />

response in WT, but not NR4A1-deficient macrophages. Moreover, we observed a<br />

critical role of NR4A1 during the modulation of the activity of the pro-inflammatory<br />

transcription factor NFκB and the expression of different sets of pro-inflammatory<br />

genes such as IL12 and TNF. In line with these results, NR4A-/- mice display a break<br />

in self-tolerance in a model of murine lupus erythematodes (Ipseiz et al. in<br />

preperation).<br />

The nuclear receptor PPARβ acts as a key regulator of bone homeostasis<br />

Carina Scholtysek, Edith Bottesch and Arnd Kleyer,<br />

The nuclear receptor PPARγ acts as a master regulator of adipocyte differentiation.<br />

In contrast, PPARγ negatively affects Wnt-signalling in osteoblasts and thereby<br />

interferes with osteoblast differentiation and bone formation. The role of its family<br />

member PPARβ during bone homeostasis has not been studied, though. To address<br />

the role of PPARβ during bone homeostasis, we analyzed its role in osteoblasts and<br />

observed that this nuclear receptor acted in a permissive manner on Wnt-signaling in<br />

these cells. Activation of PPARβ by specific ligands induced expression of the Wntco-receptor<br />

LRP5, promoted nuclear accumulation of β-catenin and consequently<br />

enhanced TCF-driven transcriptional activity. Thereby, PPARβ augmented<br />

expression of different Wnt-dependent genes, such as osterix and osteoprotegerin<br />

(opg), in osteoblasts. Consequently, activation of PPARβ in osteoblasts blocked the<br />

differentiation of bone- resorbing osteoclasts. Mice deficient in PPARβ, displayed<br />

reduced Wnt-signaling activity and low serum levels of OPG resulting in an increased<br />

differentiation of osteoclasts and osteopenia. Conversely, pharmacological treatment<br />

with a PPARβ-specific agonist blocked the formation of osteoclasts in vivo and<br />

protected mice from ovariectomy-induced bone loss in a therapeutic manner. These<br />

data reveal a so far unrecognized role for PPARβ in the crosstalk between energy<br />

metabolism and bone homeostasis and highlights its potential to serve as a target for<br />

the treatment of osteoporosis (Fig. 3; Scholtysek et al., in revision).<br />

137


Fig. 3 The nuclear receptor PPARβ acts as key regulator of bone homeostasis. (A) Gene expression<br />

analysis in primary osteoblasts after stimulation with specific agonists for PPARα, PPARβ or PPARγ in<br />

the absence or presence of the Wnt agonist Wnt3a. (B) Analysis of the mineralization of primary<br />

osteoblasts after treatment with the indicated concentrations of the PPARβ agonist GW501516. (C)<br />

Measurement of the transcriptional activity of a TCF-luciferase reporter after overexpression of PPARβ<br />

and stimulation with Wnt3a or a vehicle, respectively. (D) In vitro quantification of osteoclast<br />

differentiation in osteoblast/osteoclast co-cultures after stimulation with the indicated concentration of<br />

the PPAR β agonist GW501516 and Wnt3a. (E) Micro-CT analysis of the bone structure and (F)<br />

determination of the mineral apposition in sham-trated mice and mice after ovariectomy (OVX), which<br />

have been treated with a vehicle or the PPARβ agonist GW501516 over a period of 6 weeks.<br />

Publications (2009-2012)<br />

2011/12<br />

Uderhardt S, Herrmann M, Oskolkova O, Aschermann S, Bicker W, Ipseiz N, Sarter<br />

K, Frey B, Rothe T, Voll R, Nimmerjahn F, Bochkov VN, Schett G, Krönke G. 12/15-<br />

Lipoxygenase Orchestrates the Clearance of Apoptotic Cells and Maintains<br />

Immunologic Tolerance, Immunity (2012), doi:10.1016/j.immuni.2012.03.010. [Epub<br />

ahead of print]<br />

Schwab I, Biburger M, Krönke G, Schett G, Nimmerjahn F. IVIg-mediated<br />

amelioration of ITP in mice is dependent on sialic acid and SIGNR1. Eur J Immunol.<br />

2012 Jan 26. doi: 10.1002/eji.201142260. [Epub ahead of print]<br />

Krönke G, Reich N, Scholtysek C, Akhmetshina A, Uderhardt S, Zerr P, Palumbo K,<br />

Lang V, Dees C, Distler O, Schett G, Distler JH. The 12/15-lipoxygenase pathway<br />

138


counteracts fibroblast activation and experimental fibrosis. Ann Rheum Dis. 2012<br />

Jan 20.<br />

Dees C, Akhmetshina A, Zerr P, Reich N, Palumbo K, Horn A, Jüngel A, Beyer C,<br />

Krönke G, Zwerina J, Reiter R, Alenina N, Maroteaux L, Gay S, Schett G, Distler O,<br />

Distler JH. Platelet-derived serotonin links vascular disease and tissue fibrosis.<br />

J Exp Med. 2011 May 9;208(5):961-72.<br />

2010<br />

Krönke G, Uderhardt S, Kim KY, Zaiss MM, Katzenbeisser J, Schett G, Abo A. R-<br />

Spondin-1 protects against inflammatory bone damage during murine arthritis by<br />

modulating the Wnt pathway. Arthritis Rheum. 2010 Aug;62(8):2303-12.<br />

Uderhardt S, Diarra D, Katzenbeisser J, David JP, Zwerina J, Richards WG, Krönke<br />

G, Schett G. Blockade of Dickkopf-1 induces fusion of sacroiliac joints.<br />

Ann Rheum Dis. 2010 Mar;69(3):592-7<br />

Stach CM, Bäuerle M, Englbrecht M, Krönke G, Engelke K, Manger B, Schett G.<br />

Periarticular bone structure in rheumatoid arthritis patients and healthy individuals<br />

assessed by high-resolution computed tomography.<br />

Arthritis Rheum. 2010 Feb;62(2):330-9.<br />

2009<br />

Böhm C, Hayer S, Kilian A, Zaiss MM, Finger S, Hess A, Engelke K, Kollias G,<br />

Krönke G, Zwerina J, Schett G, David JP. The {alpha}-Isoform of p38 MAPK<br />

Specifically Regulates Arthritic Bone Loss.<br />

J Immunol. 2009 Oct 14. [Epub ahead of print]<br />

Zarbock A, Distasi MR, Smith E, Sanders JM, Krönke G, Harry BL, von Vietinghoff S,<br />

Buscher K, Nadler JL, Ley K. Improved Survival and Reduced Vascular Permeability<br />

by Eliminating or Blocking 12/15-Lipoxygenase in Mouse Models of Acute Lung Injury<br />

(ALI).<br />

J Immunol. 2009 Oct 1;183(7):4715-22. Epub 2009 Sep 14.<br />

Schett G, Stolina M, Dwyer D, Zack D, Uderhardt S, Krönke G, Kostenuik P, Feige U.<br />

Tumor necrosis factor alpha and RANKL blockade cannot halt bony spur formation in<br />

experimental inflammatory arthritis.<br />

Arthritis Rheum. 2009 Sep;60(9):2644-54.<br />

Axmann R, Böhm C, Krönke G, Zwerina J, Smolen J, Schett G. . Inhibition of<br />

interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo.<br />

Arthritis Rheum. 2009 Sep;60(9):2747-56.<br />

Krönke G, Katzenbeisser J, Uderhardt S, Zaiss M, Scholtysek C, Schabbauer G,<br />

Zarbock A, Koenders MI, Axmann R, Zwerina J, Baenckler HW, van den Berg W, Voll<br />

RE, Kühn H, Joosten LAB, Georg Schett G. 12/15-lipoxygenase counteracts<br />

inflammation and tissue damage in arthritis.<br />

J Immunol. 2009 Sep 1;183(5):3383-9. Epub 2009 Aug 12.<br />

Krönke G, Uderhardt S, Katzenbeisser J, Schett G. The 12/15-Lipoxygenase-<br />

Pathway Promotes Osteoclast Development and Differentiation.<br />

Autoimmunity. 2009 May;42(4):383-5.<br />

139


Books and Reviews<br />

Scholtysek C, Krönke G, Schett G. Inflammation-associated changes in bone<br />

homeostasis.<br />

Inflamm Allergy Drug Targets. 2012 Jan 20.<br />

140


DEPARTMENT OF UROLOGY<br />

Div. Molecular Urology<br />

Head: Helge Taubert, Dr. rer. nat.<br />

Professor of Molecular Urology<br />

Address: Division of Molecular Urology<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 23373<br />

Fax: + 49 (9131) 85 23374<br />

E-mail: helge.taubert@uk-erlangen.de<br />

Homepage: www.urologie.uk-erlangen.de<br />

Sven Wach, Dr. rer. nat.<br />

Head<br />

Helge Taubert, Prof. Dr. rer. nat.<br />

Professor of Molecular Urology<br />

Supporting Staff<br />

Postdoctoral Fellows<br />

141


Elke Nolte geb. Löprich,<br />

Dipl. Biologin<br />

Omar Al-Janabi , Dipl.<br />

Biologe<br />

Christine Ellmann<br />

Kathrin Holzer<br />

Katrin Weigelt, MTA<br />

Doctoral Students (Biology)<br />

Doctoral Students (Medicine)<br />

Technicians<br />

Guest Scientists<br />

142<br />

Anne Theil, BTA


<strong>Research</strong><br />

Our group analyses molecular mechanisms that control tumor development and<br />

progression, focusing on microRNA expression and their target genes and gene<br />

expression that is hypoxia- or stem cell-associated in solid tumors (urologic tumors,<br />

soft tissue sarcomas).<br />

Tumor genetic research with focus on identification of biomarkers<br />

Dr. rer. nat. S. Wach<br />

The identification and characterization of specific biological properties of the prostate<br />

carcinoma as well as other malignant tumors like kidney carcinoma is the main focus<br />

of the biological research projects. By assessing changes in microRNA (miRNA)<br />

expression profiles it is already possible to distinguish between samples of tumor and<br />

non-malignant tissue. Furthermore the prognostic value of miRNA expression profiles<br />

is currently examined. MiRNAs directly regulate the expression of numerous other<br />

proteins in cells. Therefore, experimental methods for analyzing protein expression<br />

(Fig.1) are a vital component of our research.<br />

The complete spectrum of molecular cytogenetic techniques including fluorescence<br />

in situ DNA and RNA hybridization is established in the laboratory. The quantification<br />

of micro-RNA and gene expression as well as the determination of gene copy<br />

numbers using real time PCR approaches is a central part of the experimental<br />

methods.<br />

The role of hypoxia and hypoxia-associated signal transduction<br />

pathways in solid tumors<br />

Prof. Dr. rer. nat. Helge Taubert<br />

The lack of oxygen (hypoxia) is a situation seen in many solid tumors. Especially<br />

locally advanced malignancies rapidly outgrow the blood vessels that supported their<br />

growth. By this tumor cells are confronted with a lack of oxygen and nutrition. As a<br />

consequence, more than 70 genes are activated by the HIF-1 transcription factor.<br />

Signal molecules are produced that stimulate the growth of new blood vessels,<br />

enzymes are produced that support a survival of cells under hypoxic conditions and<br />

stem cell-associated genes are expressed. We are mainly interested in the regulation<br />

of miRNA genes by HIF-1. Because miRNAs themselves regulate numerous target<br />

genes, it is obvious that hypoxia has a vital influence on any cell. Using cell culture<br />

models we examine the functional consequences of hypoxia on tumor cells.<br />

Projects<br />

Is the detection of members of the urokinase plasminogen activator<br />

family and of splice variants of survivin suitable as marker for<br />

143


prognosis and metastases of prostate cancer in the Clinic of<br />

Urology?<br />

Rudolf und Irmgard Kleinknecht-Stiftung (Grant No. 01.10.11-30.9.2013)<br />

Prof. Dr. H. Taubert<br />

Although prostate cancer belongs is the most frequent tumor in men there are except<br />

clinical factors no markers that are valid for an evaluation of the individual disease<br />

course ort he individual prognosis. We will investigate members of the urokinase<br />

plasminogen activator family and splice variants of the apoptosis inhibitor survivin on<br />

the mRNA level in prostate cancer retrospectively and prospectively. The molecular<br />

results will be correlated with the clinical parameters and tested for their relevance for<br />

the tumor biological behavior (development of relapses and/or metastases) and the<br />

individual prognosis. We expect to better characterize factors that can improve<br />

individual prognosis evaluation and help to support future therapy decisions.<br />

Alterations of the microRNA expression profile of primary prostate<br />

cancer<br />

Wilhelm Sander-Stiftung (Grant No. 2007.025.01)<br />

Prof. Dr. B. Wullich, Prof. Dr. F. Grässer<br />

MicroRNAs (miRNAs) belong to the family of non-coding RNAs. Their biological role<br />

is the regulation of gene expression on a post-transcriptional level. In every human<br />

tumor entity examined so far, there exists a characteristic pattern of deregulated<br />

miRNAs. Hereby, miRNAs contribute to tumorigenesis by promoting the expression<br />

of oncogenes or suppressing the tumor suppressor genes. In prostate cancer we<br />

were able to establish comprehensive and reliable miRNA expression signatures.<br />

Using these miRNA expression signatures, we were able to distinguish between<br />

tumor- and normal tissue with high accuracy. Therefore, miRNAs have the potential<br />

of being valuable biomarkers for the diagnosis of prostate cancer in the future.<br />

Besides their properties as biomarkers, miRNAs lead the way to the discovery of<br />

novel tumor-relevant genes because miRNA deregulation is not merely a symptom<br />

but a driving force in the process of tumorigenesis. We are currently investigating<br />

several specific target genes of deregulated miRNAs to unravel their intrerplay and<br />

functional relevance in prostate carcinogenesis.<br />

MiRNAs in papillary renal cell carcinoma<br />

ELAN-Funds (Grant No. 09.11.11.1)<br />

Dr. Sven Wach<br />

Papillary renal cell carcinomas (pRCCs) represent the second most common entity of<br />

renal cell carcinomas. According to histopathological criteria, pRCCs can even be<br />

further subdivided into two distinct subtypes. Recent insights point towards the fact,<br />

that pRCCs of type 1 represent a low-malignant phenotype whereas type 2 pRCCs<br />

are high-risk carcinomas associated with a worse clinical outcome. So far, there is<br />

little insight into the molecular basis of the differentiation pattern or the different<br />

clinical behavior of these two subtypes of pRCC. Using our established method of<br />

miRNA expression profiling, we are currently identifying miRNAs that discriminate<br />

144


etween the two pRCC subtypes. When target genes of these miRNAs will be<br />

defined, this may lead to a better understanding of the molecular pathways affected<br />

in both subtypes of pRCC.<br />

2012<br />

Publications (2009-2012)<br />

Original Articles<br />

Wach S, Nolte E, Szczyrba J, Stöhr R, Hartmann A, Orntoft T, Dyrskjøt L, Eltze E, Wieland<br />

W, Keck B, Ekici AB, Grässer F, Wullich B. MicroRNA profiles of prostate carcinoma<br />

detected by multiplat- form microRNA screening. Int J Cancer 2012;130(3):611-21<br />

Greither T, Würl P, Grochola L, Bond G, Bache M, Kappler M, Lautenschläger C,<br />

Holzhausen HJ, Wach S, Eckert AW, Taubert H. Expression of microRNA 210 associates<br />

with poor survival and age of tumor onset of soft-tissue sarcoma patients. Int J Cancer 2012<br />

130:1230-5.<br />

2011<br />

Szczyrba J, Nolte E, Wach S, Kremmer E, Stöhr R, Hartmann A, Wieland W, Wullich B,<br />

Grässer FA. Down-regulation of Sec23A protein by miRNA-375 in prostate carcinoma. Mol<br />

Cancer Res. 2011;9(6):791-800.<br />

Keck B, Stoehr R, Wach S, Rogler A, Hofstaedter F, Lehmann J, Montironi R, Sibonye M,<br />

Fritsche HM, Lopez-Beltran A, Epstein JI, Wullich B, Hartmann A. The plasmacytoid<br />

carcinoma of the bladder- rare variant of aggressive urothelial carcinoma. Int J Cancer.<br />

2011;129(2):346-54.<br />

Eckert AW, Lautner MH, Schütze A, Taubert H, Schubert J, Bilkenroth U. (2011)<br />

Coexpression of hypoxia-inducible factor-1α and glucose transporter-1 is associated with<br />

poor prognosis in oral squamous cell carcinoma patients. Histopathology 2011;58:1136-47.<br />

Kappler M, Taubert H, Eckert AW. Oxygen sensing, homeostasis, and disease. N Engl J<br />

Med. 2011;365(19):1845-6 (letter/author reply).<br />

Rot S1, Taubert H1, Bache M, Greither T, Würl P, Eckert AW, Schubert J, Vordermark D,<br />

Kappler M. A novel splice variant of the stem cell marker LGR5/GPR49 is correlated with the<br />

risk of tumor-related death in soft-tissue sarcoma patients. BMC Cancer. 2011 Oct 6;11:429.<br />

(1equally contributed)<br />

Bache M, Zschornak MP, Passin S, Keßler J, Wichmann H, Kappler M, Paschke R,<br />

Kaluđerović GN, Kommera H, Taubert H, Vordermark D. Increased betulinic acid induced<br />

cytotoxicity and radiosensitivity in glioma cells under hypoxic conditions. Radiat Oncol. 2011<br />

Sep 9;6(1):111.<br />

Kotzsch M, Magdolen V, Greither T, Kappler M, Bache M, Lautenschlager C, Fussel S,<br />

Eckert AW, Luther T, Baretton G, Wurl P, Taubert H. Combined mRNA expression levels of<br />

members of the urokinase plasminogen activator (uPA) system correlate with diseaseassociated<br />

survival of soft-tissue sarcoma patients. BMC Cancer. 2011 Jun 25;11(1):273.<br />

[Epub ahead of print]<br />

145


Grochola LF, Taubert H, Greither T, Bhanot U, Udelnow A, Würl P. Elevated transcript levels<br />

from the MDM2 P1 promoter and low p53 transcript levels are associated with poor<br />

prognosis in human pancreatic ductal adenocarcinoma. Pancreas. 2011 Mar;40(2):265-70.<br />

2010<br />

Szczyrba J, Loprich E, Wach S, Jung V, Unteregger, G, Barth S, Grobholz R, Wieland W,<br />

Stohr R, Hartmann A, Wullich B, Grässer F. The MicroRNA Profile of Prostate Carcinoma<br />

Obtained by Deep Sequencing. Mol Cancer Res. 2010;8(4):529-38.<br />

Goebell PJ, Keck B, Wach S and Wullich B. [Value of biomarkers in urology]. Urologe A<br />

2010, 49, 547-559.<br />

Fürnrohr BG, Wach S, Kelly JA, Haslbeck M, Weber CK., Stach CM, Hueber AJ, Graef, D,<br />

Spriewald BM, Manger K Herrmann M, Kaufman KM, Frank SG, Goodmon E, James JA,<br />

Schett G, Winkler TH, Harley JB, Voll RE. Polymorphisms in the Hsp70 gene locus are<br />

genetically associated with systemic lupus erythematosus. Ann Rheum Dis.<br />

2010;69(11):1983-9.<br />

Taubert H, Heidenreich C, Holzhausen HJ, Schulz A, Bache M, Kappler M, Eckert AW, Wurl<br />

P, Melcher I, Hauptmann K, Hauptmann S, Schaser KD. Expression of survivin detected by<br />

immunohistochemistry in the cytoplasm and in the nucleus is associated with prognosis of<br />

leiomyosarcoma and synovial sarcoma patients. BMC Cancer. 2010,10(1):65.<br />

Taubert H, Würl P, Greither T, Kappler M, Bache M, Lautenschläger C, Füssel S, Meye A,<br />

Eckert AW, Magdolen V, Kotzsch M. Co-detection of members of the urokinase plasminogen<br />

activator system in tumor tissue and in serum correlates with a poor prognosis for soft-tissue<br />

sarcoma patients. Brit J Cancer 2010 Feb 16;102(4):731-7.<br />

Greither T, Grochola L, Udelnow A, Lautenschläger C, Würl P, Taubert H. Elevated<br />

expression of microRNAs 155, 203, 210 and 222 in pancreatic tumours associates with<br />

poorer survival. Int J Cancer. 2010 Jan 1;126(1):73-80.<br />

Vazquez A, Grochola LF, Bond EE, Levine AJ, Taubert H, Müller TH, Würl P, Bond GL.<br />

Chemosensitivity Profiles Identify Polymorphisms in the p53 Network Genes 14-3-3{tau} and<br />

CD44 That Affect Sarcoma Incidence and Survival. Cancer Res. 2010; 70(1):172-80.<br />

Bache M, Kappler M, Wichmann H, Rot S, Hahnel A, Greither T, Said HM, Kotzsch M, Würl<br />

P, Taubert H, Vordermark D. (2010) Elevated tumor and serum levels of the hypoxiaassociated<br />

protein osteopontin are associated with prognosis for soft tissue sarcoma<br />

patients. BMC Cancer. 10:132.<br />

Eckert AW, Schubert J, Taubert H. Optimising the therapeutic ratio in head and neck cancer.<br />

Lancet Oncol. 2010 Jun;11(6):511-2. (letter/author reply)<br />

Eckert AW, Schutze A, Lautner MH, Taubert H, Schubert J, Bilkenroth U. HIF-1a is a<br />

prognostic marker in oral squamous cell carcinomas. Int J Biol Markers. 2010 Apr-<br />

Jun;25(2):87-92.<br />

Eckert AW, Lautner MHW, Schuetze A, Bolte K, Bache M, Kappler M, Schubert J, Taubert H,<br />

Bilkenroth U. (2010) Co-expression of Hif1a and CAIX is associated with poor prognosis in<br />

oral squamous cell carcinoma patients. J Oral Pathol Med. 39(4):313-7.<br />

Hahnel A, Wichmann H, Kappler M, Kotzsch M, Vordermark D, Taubert H, Bache M. Effects<br />

of osteopontin inhibition on radiosensitivity of MDA-MB-231 breast cancer cells. Radiat<br />

Oncol. 2010;5:82.<br />

Grochola LF, Müller TH, Bond GL, Taubert H, Udelnow A, Würl P. MDM2 SNP309<br />

Associates With Accelerated Pancreatic Adenocarcinoma Formation. Pancreas. 2010<br />

Jan;39(1):76-80.<br />

146


2009<br />

Hoffman AC, Danenberg KD, Taubert H, Danenberg PV, Wuerl P. A three-gene signature for<br />

outcome in soft tissue sarcoma. Clin Cancer Res. 2009 15(16):5191-8.<br />

Bluemke K, Bilkenroth U, Meye A, Fuessel S, Lautenschlaeger C, Goebel S, Melchior A,<br />

Heynemann H, Fornara P, Taubert H. Detection of circulating tumor cells in peripheral blood<br />

of patients with renal cell carcinoma correlates with prognosis. Cancer Epidemiol Biomarkers<br />

Prev 2009; 2009 Aug;18:2190-4.<br />

Grochola LF, Vazquez A, Bond EE, Würl P, Taubert H, Müller TH, Levine AJ, Bond GL.<br />

Recent Natural Selection Identifies a Genetic Variant in a Regulatory Subunit of Protein<br />

Phosphatase 2A that Associates with Altered Cancer Risk and Survival. Clin Cancer Res.<br />

2009;15(19):6301-8.<br />

Schmidt H, Taubert H, Lange H, Kriese K, Schmitt WD, Hoffmann S, Bartel F, Hauptmann<br />

S.Small polydispersed circular DNA contains strains of mobile genetic elements and occurs<br />

more frequently in permanent cell lines of malignant tumors than in normal lymphocytes.<br />

Oncol Rep. 2009 Aug;22:393-400.<br />

Seifert A, Taubert H, Hombach-Klonisch S, Fischer B, Navarrete Santos A.: TCDD mediates<br />

inhibition of p53 and activation of ERalpha signaling in MCF-7 cells at moderate hypoxic<br />

conditions. Int J Oncol. 2009;35:417-24.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

147


DEPARTMENT OF INTERNAL MEDICINE 5<br />

(Haematology / Oncology)<br />

Head: Andreas Mackensen, Prof. Dr. med.<br />

<strong>Research</strong> Cellular Immunoregulation<br />

Group: Evelyn Ullrich, Prof. Dr. med.<br />

Address: Department of Internal Medicine 5<br />

Ulmenweg 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 43091<br />

Fax: + 49 (9131) 85 35958<br />

E-mail: evelyn.ullrich@uk-erlangen.de<br />

Homepage: http://www.medizin5.uk-erlangen.de<br />

Postdoctoral Fellows<br />

Ruth Bauer, Dr. rer. nat.<br />

Doctoral Students (Biology)<br />

Kathrin Meinhardt<br />

Johanna Rothamer<br />

Doctoral Students (Molecular Medicine)<br />

Stephanie Krieg<br />

148


<strong>Research</strong><br />

Technicians<br />

Julia Schneider<br />

Franziska Ganß<br />

Our group analyses human and murine natural killer cells in different lymphoid and<br />

non-lymphoid organs. NK cells are a heterogeneous population of immune cells with<br />

cytotoxic capacity and multiple immunoregulatory properties. Until recently, studies<br />

on human NK cells have been mainly focused on NK cells from peripheral blood. The<br />

project based in the <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center aims to characterize the genomic,<br />

phenotypical and functional profile of human NK cell subsets from different organs<br />

with a special interest in early forms of thymic NK cells in comparison to mature<br />

peripheral blood NK cells from adults and children.<br />

Phenotypical, functional and genomic analyses of human NK cell<br />

subpopulations<br />

Human NK cells can be classified according to the expression of CD56 and CD16<br />

into immunoregulatory, cytokine producing CD56 high CD16 dim and mature cytotoxic<br />

CD56 dim CD16 high NK cells. In contrast to murine NK cell subpopulations that have<br />

been extensively studied in different compartments, studies on human NK cells have<br />

been mainly focused on NK cells from peripheral blood. Little is known about the<br />

functional role of human NK cell subpopulations in different lymphoid organs.<br />

Therefore, a translation of murine studies, mainly performed on splenic NK cells, into<br />

the human system remains quite difficult. In our first analyses of human NK cell<br />

subsets from peripheral blood, spleen, bone marrow, and thymus, we observed<br />

relevant differences concerning the distribution of CD56 high CD16 dim and<br />

CD56 dim CD16 high NK cells. In addition, the expression of functionally relevant surface<br />

molecules varies on the NK cell subsets.<br />

One important observation from our phenotypic analyses was that especially the<br />

composition of NK cell subpopulations in the thymus differs in particular from those in<br />

spleen and peripheral blood. For that reason that thymus is already well-known for<br />

being an organ for immune cell development, we further focused on the<br />

characterization of thymic NK cells. NK cells from thymus have not only a lower<br />

CD56-expression but also a different ratio of CD16 high / CD16 dim NK cells compared<br />

to peripheral blood NK cells (Fig.1). While peripheral blood NK cells from adults have<br />

a ration of 10 / 1, children have a ratio of only 4 / 1 and those of child thymus have an<br />

even lower ratio of < 1 (Fig.2).<br />

149


Fig. 1 FACS analysis of NK cell subpopulations in child PBMC and<br />

thymic MNC. Two populations can be distinguished according to<br />

the expression of CD56 and CD16. CD16 dim thymic NK cells have a<br />

lower CD56-expression compared to peripheral blood NK cells.<br />

Importantly, the NK cell subset ratio shows an age-dependency that differs in<br />

peripheral blood and thymus. Especially the number of CD16 dim immunoregulatory<br />

NK cells increases in thymus, but decreases in peripheral blood with age so that the<br />

ratio of CD16 high / CD16 dim NK cells increases in total (Fig.2). This process might be<br />

explained by acquisition of surface molecules that regulate migration, homing and<br />

differentiation of early forms of NK precursors, immature, immunoregulatory or<br />

mature NK cells.<br />

Fig. 2 Ratio of CD16 high / CD16 dim NK cells in peripheral blood and<br />

thymus of children. This NK subset ratio is lower in thymus and<br />

showed a negative correlation with increasing donor’s age.<br />

To get further insights, we performed microarray analyses that will provide us with<br />

detailed information on the gene expression profile of NK cell populations from<br />

different organs. In addition to the genomic and phenotypical analysis, we<br />

investigated the cytokine production and cytotoxicity of these different NK cell<br />

150


subpopulations. Our data clearly demonstrates that NK cells from thymus as well as<br />

from peripheral blood of young children produce only low amounts of IFN-γ and need<br />

additional IL-2 stimulation to develop an antitumor capacity that is still under<br />

averaged compared to adult NK cells (data not shown).<br />

In summary, this comparative study of NK cell subsets provides important information<br />

on the development and function of NK cells that will be of great value for the<br />

optimization of cellular therapy.<br />

Publications (2006-2011)<br />

Original Articles<br />

Ullrich E, Bosch J, Aigner M, Völkl S, Dudziak D, Spriewald B, Schuler G,<br />

Andreesen R, Mackensen A. Advances in Cellular Therapy: 5th International<br />

Symposium on the Clinical Use of Cellular Products, March 19 and 20, 2009,<br />

Nürnberg, Germany. Cancer Immunol. Immunother. 2009; Oct 28. [Epub ahead of<br />

print]<br />

Terme M, Ullrich E, Delahaye N, Chaput N, Zitvogel L. NK cell-directed<br />

therapies: from unexpected results to successful strategies. Nat.Immunol.<br />

2008;9(5):486-94.<br />

Ullrich E, Bonmort M, Mignot G, Jacobs B, Bosisio D, Sozzani S, et al. Transpresentation<br />

of Interleukin-15 dictates IKDC effector functions. J.Immunol.<br />

2008;180(12):7887-97.<br />

Mignot G, Ullrich E, Bonmort M, Menard C, Apetoh L, Taieb J, et al. The<br />

critical role of IL-15 in the antitumor effects mediated by the combination therapy<br />

imatinib and IL-2. J.Immunol. 2008; 180(10):6477-83.<br />

Ullrich E, Chaput N, Zitvogel L. Killer dendritic cells and their potential role in<br />

immunotherapy. Hormone and Metabolic <strong>Research</strong> 2008;40(2):75-81.<br />

Ullrich E, Bonmort M, Mignot G, Kroemer G, Zitvogel L. Tumor stress, cell<br />

death and the ensuing immune response. Cell Death.Differ. 2008;15(1):21-8.<br />

Ullrich E, Menard C, Flament C, Terme M, Mignot G, Bonmort M, et al.<br />

Dendritic cells and innate defense against tumor cells. Cytokine Growth Factor Rev.<br />

2008; 19(1):79-92.<br />

151


Ullrich E, Bonmort M, Mignot G, Chaput N, Taieb J, Menard C, et al. Therapy-<br />

Induced Tumor Immunosurveillance Involves IFN-Producing Killer Dendritic Cells.<br />

Cancer Res. 2007;67(3):851-3.<br />

Taieb J, Chaput N, Menard C, Apetoh L, Ullrich E, Bonmort M, et al. A novel<br />

dendritic cell subset involved in tumor immunosurveillance. Nat.Med. 2006;12(2):214-<br />

9.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Kathrin Meinhardt<br />

Isolation and characterization of murine NK cell subpopulations in vitro und in vivo<br />

Johanna Rothamer<br />

Investigation of the role of TH17 cells in a murine model of GVHD<br />

Doctoral theses (Molecular Medicine)<br />

Stephanie Krieg<br />

Functional, phenotypical und genomic analyses of human NK cells subpopulations<br />

152


AG Wiesener<br />

Department of Nephrology and Hypertension<br />

Group leader: Michael Wiesener, Prof. Dr. med.<br />

Address: Hypoxia <strong>Research</strong> Group<br />

Glückstr. 6<br />

D-91054 Erlangen<br />

Telefone: + 49 (9131) 85 36353<br />

Fax: + 49 (9131) 85 39209<br />

E-mail: michael.wiesener@uk-erlangen.de<br />

Karl Knaup, Dr. rer. nat.<br />

Ruth Schietke, Dr. rer. nat.*<br />

* period of time reported<br />

Head<br />

Michael Wiesener, Prof. Dr. med.<br />

Postdoctoral Fellows<br />

153<br />

Thomas Hackenbeck, Dr. rer. nat.


Tilman Jobst-Schwan<br />

Sina Grupp<br />

* period of time reported<br />

Juliana Monti*<br />

* period of time reported<br />

Doctoral Students (Medicine)<br />

Stefan Thieme<br />

Regina Günther<br />

Technicians<br />

Guest Scientists<br />

154<br />

-<br />

Johanna Stöckert


<strong>Research</strong><br />

The constant availability of molecular oxygen is crucial for function and survival of<br />

any cell and tissue of organisms of higher order. All mammalian cells investigated to<br />

date are capable of utilising an important adaptive mechanism, which is strongly<br />

influenced by oxygen tensions. Hypoxia-Inducible transcription-Factors (HIF) are<br />

activated by a decline of oxygen supply and stimulate numerous target genes that<br />

convey adaptation to hypoxia. This system is probably of great importance for the<br />

growth of solid tumours. Inactivation of the “von Hippel Lindau” tumour suppressor<br />

gene, which is part of the destruction apparatus of HIF, leads to strong upregulation<br />

of HIF in clear cell renal carcinoma. Importantly, early HIF activation may play an<br />

important role in tumorigenesis of VHL associated tumours. These tumours could<br />

therefore function as a model system to study causes and consequences of<br />

pronounced HIF expression.<br />

Our group mainly investigates the role and effect of overexpression of both HIFα<br />

subunits, HIF-1α and HIF-2α, in tumour tissues, cell culture models, as well as rodent<br />

models. The role of selected HIF target genes in tumour growth is being analysed.<br />

Genetically modified mice for HIF overexpression in the kidney have been generated,<br />

to investi-gate whether HIF plays a role in tumorigenesis.<br />

The role of HIF in renal epithelial cells and tumorigenesis<br />

Renal tubular HIF-2α expression requires VHL inactivation and causes fibrosis<br />

and cysts<br />

A number of publications have indicated tumorigenic potential of HIF, which may<br />

primarily concern HIF-2α and particularly for clear cell RCC. We have studied the<br />

expression patterns of HIFα isoforms in the renal tubules in detail. In three different<br />

species (mouse, rat and human) microenvironmental stimulation by hypoxia always<br />

leads to tubular HIF-1α activation, but never to HIF-2α stabilization. Since HIF-2α is<br />

widely expressed in human RCC and may be functionally more important in terms of<br />

tumorigenesis, de novo expression of HIF-2α may indicate an oncogenic switch of<br />

renal tubular cells. In line with this, we have studied two different kidney specific<br />

knockout models for the tumour suppressor VHL, which shows de novo expression of<br />

HIF-2α, next to HIF-1α. Thus, the deletion of VHL may enable expression of HIF-2α,<br />

which may have an oncogenic potential.<br />

We have therefore established transgenic mice for overexpression of HIF-2α in distal<br />

tubuli of the kidney, driven by the Ksp-Cadherin promotor (Schietke and Hackenbeck<br />

et al., PLoSOne 2012). We have not observed any malignant tumors in this model,<br />

but the transgenic HIF-2α animals develop an interesting phenotype at 12 months of<br />

age. The kidneys showed obvious morphological differences on the surface in<br />

155


comparison to the control animals. Control animals displayed a uniform and smooth<br />

kidney surface, whereas the kidneys of HIF-2 expressing mice had an irregular and<br />

rough appearance and a significantly reduced kidney weight as judged by kidney to<br />

body weight ratio.<br />

Histological analysis of the kidneys from the transgenic mice showed a strong<br />

increase of fibrotic tissue in comparison to healthy kidneys and an increase of<br />

fibrosis-associated genes like TGFβ1. We additionally determined plasma<br />

parameters for renal function. Furthermore, transgenic mice had significantly<br />

increased creatinine levels confirming impaired renal function (Fig. 1)<br />

Figure 1: Renal tubular HIF-2α expression induces fibrosis and results in reduced kidney<br />

function. A. Immunohistochemical analysis of HIF-2α and collagen I in a kidney from a transgenic<br />

HIF-2 expressing mouse. B. Blinded analysis of renal fibrosis after SiriusRed (total collagen) staining<br />

C. mRNA expression (RT-PCR) of TGFβ1 from total kidney lysates. D. Kidney function as a<br />

measurement of serum creatinine of transgenic HIF-2α mice (* p < 0.05). All figures: tmHIF-2α.HA(-):<br />

transgen negative; tmHIF-2α-HA(+): transgen positive.<br />

In addition to the fibrotic phenotype, the histological analysis of the tmHIF-2α.HA(+)<br />

transgenic mice revealed frequent formation of cysts in the renal cortex, which was<br />

not observed in the control strain (Fig. 2).<br />

156


Figure 2: Renal cyst development in aged HIF-2α transgenic mice. 12 month and older HIF-2α<br />

expressing mice develop multiple renal cysts, mainly in the kidney cortex. These partly form around<br />

glomeruli, where the glomerular tuft can be seen (arrows in A). Other cysts arise from distal tubular<br />

segments.<br />

In summary we could demonstrate, that continuous transgenic expression of HIF-2α<br />

by the Ksp-Cadherin promotor leads to renal fibrosis and insufficiency, next to<br />

multiple renal cysts. Our data cleary show, that unphysiological activation of HIF<br />

contributes to renal epithelial dedifferentiation.<br />

Differential role of HIFalpha subunits<br />

HIF-1 or HIF-2 induction is sufficient to achieve cell cycle arrest in NIH3T3<br />

mouse fibroblasts independent from hypoxia<br />

Hypoxia is a severe stress which induces physiological and molecular adaptations,<br />

where the latter is dominated by the Hypoxia-inducible transcription Factor (HIF). A<br />

well described response on cellular level upon exposure to hypoxia is a reversible<br />

cell cycle arrest, which probably renders the cells more resistant to the difficult<br />

environment. The individual roles of hypoxia itself and of the isoforms HIF-1α and<br />

HIF-2α in cell cycle regulation are poorly understood and discussed controversially.<br />

In order to characterize the isolated effect of both HIFα isoforms on the cell cycle we<br />

generated tetracycline inducible, HIF-1α and -2α expressing NIH3T3 cells<br />

(Hackenbeck et al., Cell Cycle 2009). The cDNAs for HIFα were mutated to generate<br />

stable and active HIF under normoxia. Upon activation of both HIFα subunits, the<br />

total number of living cells was reduced and long-term stimulation of HIF led to<br />

complete loss of transgene expression, implicating a strong negative selection<br />

pressure. Equally, colony forming activity was reduced by activation of both HIFα<br />

subunits. Cell cycle analyses showed that HIF activation resulted in a prominent cell<br />

cycle arrest in G1-phase, similarly to the hypoxic effect. Both, HIF-1α and HIF-2α<br />

157


were able to induce the expression of the cyclin-dependent kinase inhibitor p27 on<br />

reporter gene and protein level.<br />

Our study shows that HIF-1 and HIF-2 can individually arrest the cell cycle<br />

independent from hypoxia. These findings have implications for the resistance of<br />

tumor cells to the environment and treatment, but also for physiological cells.<br />

Importantly, recent approaches to stabilize HIFα in normoxia could have deleterious<br />

effects on proliferating tissues.<br />

Regulation of HIF by the mammalian target of rapamycin (mTOR)<br />

Consequences of genetic mTOR activation on HIF expression<br />

Stabilisation of the Hypoxia Inducible Transcriptionfactor (HIF) is a global<br />

phenomenon in most solid tumors. The predominant regulation of HIF is controlled<br />

via a prolyl hydroxylase (PHD) mediated oxygen-dependent, posttranslational<br />

degradation. The PI3K/TSC/mTOR pathway is an alternative pathway, which is<br />

capable of enhancing HIF levels. mTOR itself is negatively regulated via the<br />

Tuberous Sclerosis Complex (TSC), which consists of and is only functional in<br />

combination of both subunits (TSC1 and TSC2). In the disease of “Tuberous<br />

Sclerosis” (TS) patients harbour a heterozygous germline mutation of either the<br />

TSC1 or TSC2 gene. Due to a somatic “second hit” these individuals develop a<br />

variety of tumors, including angiomyolipomas of the kidney. Our study aims to<br />

analyze the impact and the consequences of this genetic mTOR activation on HIF<br />

accumulation. We are approaching this by pursuing three experimental avenues:<br />

1. in vitro analysis of the molecular mechanisms with an doxycyclin inducible siTSC2<br />

HeLa cell line (in cooperation with Dr. Kathrin Thedick, Albert-Ludwigs-University of<br />

Freiburg) shows that HIF is stabilized quicker, if there is previous mTOR activation to<br />

hypoxic exposure. (Fig. 3).<br />

158<br />

Fig. 3: Immunoblots of doxycyclin inducible<br />

siTSC2 HeLa cells. TSC2 reduction leads to an<br />

(A) activation of mTOR by increased rpS6<br />

phosphorylation at Ser 235/236 and (B) an<br />

earlier accumulation of HIF-1α under mild<br />

hypoxia (3% O2). ß-actin is implied as a loading<br />

control.


Apart from our previously described interplay of the mTOR and the HIF system<br />

(Knaup et al., Mol Canc Res 2009) this supports the thesis of an oxygen independent<br />

regulation pathway of HIF by mTOR.<br />

2. Immunohistochemical analysis of the mTOR and the HIF pathway in kidney samples<br />

of patients with TS reveals an unphysiological HIF accumulation in the tubules.<br />

This is probably due to the genetic inactivation of TSC and the resulting subsequent<br />

activation of mTOR (Fig. 4). Whether this unphysiological HIF expression in the<br />

healthy kidney directly influences the development and/or the growth of the tumors<br />

(angiomyolipomas) is subject of ongoing analysis.<br />

Figure 4: Immunohistohemical analysis of mTOR activation (P-rpS6) and HIF-1α stabilization in both<br />

angiomyolipoma and adjacent kidney samples from human tuberous sclerosis patients.<br />

HIF and the HIF target gene HIG2 (data not shown) are strongly expressed in angiomyolipomas.<br />

This could be due to either regional hypoxia as well as mTOR<br />

activation, possibly a combination of both scenarios.<br />

3. We are currently analysing the role of HIF activation in the tumor development and<br />

growth in a TSC2 knockout mouse model which was supplied to us by the group of<br />

Prof. T. Noda, Tokyo, Japan (Kobayashi, T. et al, Cancer Res. 1999 Mar<br />

15;59(6):1206-11).<br />

159


Figure 5: A. Kidney of a 7 months old TSC2 knockout mouse (supplied to us from the group of Prof.<br />

T. Noda, Tokyo, Japan), which developed a macroscopically visible tumor on the kidney surface. B.<br />

Morpholgical evaluation classifies this as being a papillary tumor. C and D. mTOR activation in the<br />

tumor can be detected by immunohistochemical staining of P-rpS6 (Ser 235/236).<br />

The knockout mice develop macroscopically visible tumors on the surface of the kidneys<br />

(Fig. 5, A). These tumors have papillary structures (B) and express high levels<br />

of phospho-rpS6 (Ser235/236) (C and D), which is a profound marker of mTOR<br />

activation. Our current data suggests that this genetic mTOR activation can enhance<br />

accumulation of HIF and therefore support tumor development and growth. We are<br />

currently analyzing the kidneys and the developing tumors of these mice in this<br />

context.<br />

In this ongoing study we hope to gain more insight into the molecular mechanisms of<br />

the mTOR-HIF pathway. Further, we are aiming to investigate the impact of<br />

pharmacological mTOR inhibition on tumor development and growth and its<br />

consequences on HIF expression and functionality in vitro and in vivo. Ongoing<br />

clinical studies with TS-patients and mTOR inhibitors are showing promising results,<br />

encouraging us in our effort to dissect the details and mechanisms of the mTOR-HIF<br />

interplay.<br />

Identification and characterisation of new HIF target genes<br />

In order to understand the function and mechanism of action of the HIF transcription<br />

factors it is important to characterise the spectrum of its target genes. We have<br />

previously performed affymetrix arrays to identify novel HIF-1α and HIF-2α targets<br />

(Warnecke et al., Exp. Cell Res. 2008) and selected a number of those for further<br />

analysis with potential functionality in tumor biology.<br />

160


The GTPase Rab20 is a HIF target with mitochondrial localization mediating<br />

apoptosis in hypoxia<br />

We were able to demonstrate that Rab20, which is a member of the Rab family of<br />

small GTP-binding proteins, regulating intracellular trafficking and vesicle formation,<br />

is a new HIF-1 target gene. Rab20 is directly regulated by HIF-1, resulting in rapid<br />

upregulation of Rab20 mRNA as well as protein under hypoxia. Furthermore,<br />

exogenous as well as endogenous Rab20 protein colocalizes with mitochondria.<br />

Knockdown studies revealed that Rab20 is involved in hypoxia induced apoptosis.<br />

Since mitochondria play a key role in the control of cell death, we suggest that<br />

regulating mitochondrial homeostasis in hypoxia is a key function of Rab20.<br />

Furthermore, our study implicates that cellular transport pathways play a role in<br />

oxygen homeostasis. Hypoxia-induced Rab20 may influence tissue homeostasis and<br />

repair during and after hypoxic stress. (Hackenbeck et al., Biochim Biophys Acta<br />

2011).<br />

The lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress Ecadherin<br />

in hypoxia: insights into cellular transformation processes mediated<br />

by HIF-1.<br />

In our array experiments, the lysyl oxidases Lox and LoxL2 showed a very high level<br />

of regulation, where LOX has previously been shown to be a HIF-1 target gene (Erler<br />

et al., Nature 2006). We were able to show that LOXL2 is a direct target as well. Both<br />

genes are very highly expressed in clear cell RCC and are involved in repression of<br />

the epithelial marker E-cadherin – a hallmark of the epithelial to mesenchymal<br />

transformation (EMT). EMT is of great importance for tumour cells to progress into an<br />

increasingly aggressive phenotype, as well as for diseased tissues into fibrosis. Thus,<br />

we believe that the lysyl oxidases represent a relevant mechanism in tumour biology,<br />

possible an effective area for pharmacological intervention (Schietke et al., J Biol<br />

Chem. 2010).<br />

Miscellaneous kidney related research projects<br />

The inhibitor of apoptosis protein surviving accumulates at the apical<br />

membrane of renal proximal tubules, which colocolises with megalin<br />

In the past report period we also analysed the inhibitor of apoptosis protein survivin<br />

and its role in kidney function. Survivin is a bifunctional molecule, which regulates<br />

cellular division and survival. A paradigm of a restricted oncofetal role for survivin has<br />

been formulated, where most of the published work implicates a function merely in<br />

embryonic and malignant tissues. In contrast, we have previously shown that survivin<br />

161


protein is highly expressed in the adult kidney, particularly in the proximal tubules and<br />

to a weaker extent in podocytes (Lechler et al., Am J Pathol 2007).<br />

We were able to confirm and expand our previous work, which identified survivin as<br />

being strongly present in healthy adult kidneys. Survivin predominantly accumulates<br />

at the apical membranes of proximal tubules, demonstrated clearly in vivo by<br />

immunostaining and electron microscopy. The apical localisation at the membrane of<br />

the proximal tubules reveals a pattern, which resembles that of many absorbed<br />

proteins. Survivin is a small protein of approximately 14 kDa which could, if<br />

circulating, be freely filtered by the glomerular apparatus. Many proteins in primary<br />

urine are internalised by megalin, an endocytosis receptor, of which the expression<br />

pattern is in principle comparable to the observed survivin pattern. Therefore, we<br />

generated a hypothesis of an active renal uptake mechanism for survivin, possibly by<br />

a megalin dependent process. We could clearly demonstrate in vitro, that polarised<br />

primary renal tubular cells store survivin at the apical membrane, which is true for<br />

both, endogenous and exogenously added recombinant survivin protein. Finally,<br />

knockout animals for renal megalin showed accumulation of survivin in their urine.<br />

We are currently in the process of preparing a manuscript for submission for these<br />

data (Jobst-Schwan et al.).<br />

Analyses of molecular causes of different genetic renal diseases<br />

We have recently been engaged in attempting to clarify the genetic cause of a<br />

number of hereditary diseases, which lead to end-stage renal disease. A moderate<br />

number of families are treated in our department, where we have undertaken linkage<br />

and locus analyses by next generation sequencing. We have identified families with<br />

medullary cystic kidney disease type 1 (MCKD1), as well as Alport´s disease and<br />

membranoproliferative glomerulonephritis, as well as so far uncharacterised<br />

hereditary diseases. These studies are ongoing. We hope to be able to take some of<br />

these genetic studies on to functional and molecular biology investigations after<br />

identification of the causative genes.<br />

2011/12<br />

Publications (2009-2011/12)<br />

Original Articles<br />

162


Buettner M, Xu H, Böhme R, Seliger B, Jacobi J, Wiesener M, Benz K, Amann K.<br />

Predominance of Th2 cells and plasma cells in polyoma virus nephropathy: a role for<br />

humoral immunity? Hum Pathol. 2012 Mar 8.<br />

Schietke RE, Hackenbeck T, Tran M, Günther R, Klanke B, Warnecke CL, Knaup<br />

KX, Shukla D, Rosenberger C, Koesters R, Bachmann S, Betz P, Schley G, Schödel<br />

J, Willam C, Winkler T, Amann K, Eckardt KU, Maxwell P, Wiesener MS. Renal<br />

tubular HIF-2α expression requires VHL inactivation and causes fibrosis and cysts.<br />

PLoS One. 2012;7(1):e31034.<br />

Hainz N, Thomas S, Neubert K, Meister S, Benz K, Rauh M, Daniel C, Wiesener M,<br />

Voll RE, Amann K. The Proteasome Inhibitor Bortezomib Prevents Lupus Nephritis in<br />

the NZB/W F1 Mouse Model by Preservation of Glomerular and Tubulointerstitial<br />

Architecture. Nephron Exp Nephrol. 2012;120(2):e47-58.<br />

Klinger P, Schietke RE, Warnecke C, Swoboda B, Wiesener M, Hennig FF, Gelse K.<br />

Deletion of the oxygen-dependent degradation domain results in impaired<br />

transcriptional activity of hypoxia-inducible factors. Transcription. 2011 Nov-<br />

Dec;2(6):269-75.<br />

Schley G, Klanke B, Schödel J, Forstreuter F, Shukla D, Kurtz A, Amann K, Wiesener<br />

MS, Rosen S, Eckardt KU, Maxwell PH, Willam C. Hypoxia-inducible transcription<br />

factors stabilization in the thick ascending limb protects against ischemic acute<br />

kidney injury. J Am Soc Nephrol. 2011 Nov;22(11):2004-15.<br />

Hackenbeck T, Huber R, Schietke R, Knaup KX, Monti J, Wu X, Klanke B, Frey B,<br />

Gaipl U, Wullich B, Ferbus D, Goubin G, Warnecke C, Eckardt KU, Wiesener MS.<br />

The GTPase RAB20 is a HIF target with mitochondrial localization mediating<br />

apoptosis in hypoxia. Biochim Biophys Acta. 2011 Jan;1813(1):1-13.<br />

2010<br />

Bernhardt WM, Wiesener MS, Scigalla P, Chou J, Schmieder RE, Günzler V, Eckardt<br />

KU. Inhibition of prolyl hydroxylases increases erythropoietin production in ESRD. J<br />

Am Soc Nephrol. 2010 Dec;21(12):2151-6.<br />

Gimm T, Wiese M, Teschemacher B, Deggerich A, Schödel J, Knaup KX,<br />

Hackenbeck T, Hellerbrand C, Amann K, Wiesener MS, Höning S, Eckardt KU,<br />

Warnecke C. Hypoxia-inducible protein 2 is a novel lipid droplet protein and a specific<br />

target gene of hypoxia-inducible factor-1. FASEB J. 2010 Nov;24(11):4443-58.<br />

Schietke R, Warnecke C, Wacker I, Schödel J, Mole DR, Campean V, Amann K,<br />

Goppelt-Struebe M, Behrens J, Eckardt KU, Wiesener MS. The lysyl oxidases LOX<br />

163


and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: insights<br />

into cellular transformation processes mediated by HIF-1. J Biol Chem. 2010 Feb<br />

26;285(9):6658-69.<br />

2009<br />

Morris MR, Hughes DJ, Tian YM, Ricketts CJ, Lau KW, Gentle D, Shuib S, Serrano-<br />

Fernandez P, Lubinski J, Wiesener MS, Pugh CW, Latif F, Ratcliffe PJ, Maher ER.<br />

Mutation analysis of hypoxia-inducible factors HIF1A and HIF2A in renal cell<br />

carcinoma. Anticancer Res. 2009 Nov;29(11):4337-43.<br />

Bernhardt WM, Gottmann U, Doyon F, Buchholz B, Campean V, Schödel J,<br />

Reisenbuechler A, Klaus S, Arend M, Flippin L, Willam C, Wiesener MS, Yard B,<br />

Warnecke C, Eckardt KU. Donor treatment with a PHD-inhibitor activating HIFs<br />

prevents graft injury and prolongs survival in an allogenic kidney transplant model.<br />

Proc Natl Acad Sci U S A. 2009 Dec 15;106(50):21276-81.<br />

Weidemann A, Kerdiles YM, Knaup KX, Rafie CA, Boutin AT, Stockmann C, Takeda<br />

N, Scadeng M, Shih AY, Haase VH, Simon MC, Kleinfeld D, Johnson RS. The glial<br />

cell response is an essential component of hypoxia-induced erythropoiesis in mice. J<br />

Clin Invest. 2009 Nov;119(11):3373-83.<br />

Tanaka T, Wiesener M, Bernhardt W, Eckardt KU, Warnecke C. The human HIF<br />

(hypoxia-inducible factor)-3alpha gene is a HIF-1 target gene and may modulate<br />

hypoxic gene induction. Biochem J. 2009 Oct 23;424(1):143-51.<br />

Kroening S, Neubauer E, Wessel J, Wiesener M, Goppelt-Struebe M. Hypoxia<br />

interferes with connective tissue growth factor (CTGF) gene expression in human<br />

proximal tubular cell lines. Nephrol Dial Transplant. 2009 Nov;24(11):3319-25.<br />

Schödel J, Klanke B, Weidemann A, Buchholz B, Bernhardt W, Bertog M, Amann K,<br />

Korbmacher C, Wiesener M, Warnecke C, Kurtz A, Eckardt KU, Willam C. HIF-prolyl<br />

hydroxylases in the rat kidney: physiologic expression patterns and regulation in<br />

acute kidney injury. Am J Pathol. 2009 May;174(5):1663-74.<br />

Hackenbeck T, Knaup KX, Schietke R, Schödel J, Willam C, Wu X, Warnecke C,<br />

Eckardt KU, Wiesener MS. HIF-1 or HIF-2 induction is sufficient to achieve cell cycle<br />

arrest in NIH3T3 mouse fibroblasts independent from hypoxia. Cell Cycle. 2009 May<br />

1;8(9):1386-95.<br />

164


Wacker I, Sachs M, Knaup K, Wiesener M, Weiske J, Huber O, Akçetin Z, Behrens J.<br />

Key role for activin B in cellular transformation after loss of the von Hippel-Lindau<br />

tumor suppressor. Mol Cell Biol. 2009 Apr;29(7):1707-18.<br />

Knaup KX, Jozefowski K, Schmidt R, Bernhardt WM, Weidemann A, Juergensen JS,<br />

Warnecke C, Eckardt KU, Wiesener MS. Mutual regulation of hypoxia-inducible factor<br />

and mammalian target of rapamycin as a function of oxygen availability. Mol Cancer<br />

Res. 2009 Jan;7(1):88-98.<br />

Books and Reviews<br />

Wiesener MS, Maxwell PH, Eckardt KU. Novel insights into the role of the tumor<br />

suppressor von Hippel Lindau in cellular differentiation, ciliary biology, and cyst<br />

repression. J Mol Med (Berl). 2009 Sep;87(9):871-7.<br />

TRAINING OF GRADUATE AND MEDICAL STUDENTS<br />

Doctoral theses (Biology)<br />

Ruth Schietke (2009)<br />

Thomas Hackenbeck (2009)<br />

Doctoral theses (Medicine)<br />

Stefan Thieme<br />

Tilman Jobst-Schwan<br />

Sina Grupp<br />

Regina Günther<br />

165


TEACHING ACTIVITIES<br />

at the <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center<br />

LECTURES, SEMINARS AND PRACTICAL COURSES<br />

Members of the <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center participate in courses and lectures that are<br />

currently offered to students enrolled in the Graduate Programs in Biology at the<br />

School of Basic Sciences II (Naturwissenschaftliche Fakultät II) and in Molecular<br />

Medicine at the Medical School as well as to medical students. The focus is on basic<br />

and advanced molecular cell biology (Behrens, von der Mark, Müller) and basic and<br />

clinical immunology (Jäck).<br />

166


SEMINARS AND CONFERENCES<br />

at the <strong>Nikolaus</strong>-<strong>Fiebiger</strong>-Center<br />

Selection of Speakers since January 2009:<br />

NIKOLAUS-FIEBIGER-SEMINAR<br />

K. Förstemann, München<br />

Gene Regulation by Small RNAs- General Principles and Examples from Drosophila<br />

28.01.2009<br />

C. Stocking, Hamburg<br />

Activated FLT3 Tyrosine Receptor Kinase in Acute Lymphoblastic Leukemiea<br />

20.05.2009<br />

H. Urlaub, Göttingen<br />

Quantitative Mass Sepctrometry to Elucidate B Cell Receptor Signaling<br />

29.07.2009<br />

K. Toellner, Birmingham, UK<br />

Imprinting B Cells: Germinal Centres and Plasma Cells – Two sides of the Coin<br />

21.10.2009<br />

E. Izaurralde, Tübigen<br />

Mechansim of miRNA-mediated gene Silencing<br />

31.03.2010<br />

T. Nguyen, Paris<br />

The (pro)renin receptor: important for blood pressure and organ damage or just in<br />

development?<br />

23.02.2011<br />

G. Weidinger, Dresden<br />

Wnt-signaling in Development and Regeneration<br />

11.05.2011<br />

R. Renkawitz-Pohl, Marburg & S. Lindner, Hamburg, D. Mielenz, Erlangen<br />

Interdisciplinary Minisymposium<br />

18.05.2011<br />

R. Obst, München<br />

Timed antigen presentation and T cell responses<br />

25.05.2011<br />

M. Flajnik, Baltimore, USA<br />

Evolution of the Immune System<br />

15.06.2011<br />

167


G. Riemekasten, Berlin<br />

Systemic sclerosis as prototypic disease for functional autoantibodies against vascular<br />

receptors<br />

06.07.2011<br />

H. Bastians, Göttigen<br />

Mechanisms underlying Chromosomal Instability in Human Cancer<br />

12.07.2011<br />

N. Abdalah<br />

The interplay of behavior and adult neurogenesis for cognitive and affective disorders<br />

27.07.2011<br />

B. Blomberg, Miami, USA<br />

Molecular mediators of autonomous B cell deficiencies in aging mice and humans<br />

19.09.2011<br />

R. Blum, Würzburg<br />

The sodium channel Nav1.9 regulates activity-dependent axon growth and might serve as a<br />

target for axon regeneration and maintenance<br />

05.10.2011<br />

S. Herzog, Freiburg<br />

BIOSS – Centre for Biological Signaling Studies<br />

23.05.2012<br />

A. Blesch, Heidelberg<br />

Challenges and Opportunities in Spinal Cord Regeneration<br />

20.06.2012<br />

168

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!