EINFÜHRUNG Das CCR blickt nun auf sieben Jahre

Transcription

EINFÜHRUNG Das CCR blickt nun auf sieben Jahre
Einführung
EINFÜHRUNG
D
as CCR blickt nun auf sieben Jahre seines
Bestehens zurück. Gegründet wurde es im
Jahre 2003 als ein interdisziplinäres kardiovaskuläres Forschungszentrum an der Charité, einer
renommierten, traditionsreichen Stätte medizinischer
Forschung, die dieses Jahr auf ihr 300-jähriges Bestehen zurückblickt.
Zum Gründungszeitpunkt des CCR hatte die „alte“
Charité, also das Klinikum in Berlin-Mitte, das als
Medizinische Fakultät der Berliner Humboldt Universität bis zur deutschen Wiedervereinigung im Jahr 1990
der medizinische Leuchtturm der Deutschen Demo­
kratischen Repu­blik (DDR)
war, bereits eine Fusion mit
dem Westberliner VirchowKlinikum hinter sich; eine
weitere Fusion wurde in der
folgenden Dekade mit dem
Universitätsklinikum „Benjamin Franklin“ der Frei­en
Universität (West-) Berlins
vollzogen. Die Charité wurde
damit als eigenständige Körperschaft mit beinahe
15.000 Mitarbeitern zum grössten Universitäts­klinikum
Europas. Ähnliche Entwicklungen hat es in anderen
europäischen Hauptstädten gegeben, z.B. in London
und Paris. Die Hintergründe dieser
Fusionen waren im Wesentlichen
ökonomischer, nicht unbedingt wissenschaftlicher oder Patienten-orientierter Natur.
rité auf durchaus hohem
Niveau betrieben, allerdings nicht in wünschenswertem Maße koordiniert.
Verschiedene Hintergründe und Traditionen der einzelnen Fakultäten und Klinika sowie die mangelnde
inhaltliche und strukturelle Vernetzung innerhalb der
Berliner kardiovaskulären Forschungsinstitutionen,
zwischen Kardiologen, Nephrologen sowie kardiovaskulär orientierten Physiologen und Pharmakologen, zwischen Grundlagenforschern und Klinikern
an verschiedenen Standorten der Charité kann als
eine Ursache dafür angesehen werden, dass es in
Berlin - mit Ausnahme eines „Transregio“-Verbundes
der Deutschen Forschungsgemeinschaft (DFG) zum
Thema Myokarditis - bisher nicht zu grösseren institutionellen Forschungsverbünden kam, wie etwa zu
einem kardiovaskulären Sonderforschungsbereich
der DFG.
Mit der Gründung des CCR war die Hoffnung verbunden, diese Lücke zu schliessen, die teilweise
divergierenden kardiovaskulären Forschungsaktivitäten einzufangen und zu gemeinsamen Projektanstrengungen zu bündeln. Dies ist bisher zum Teil
gelungen.
Kardiovaskuläre Forschung im
Grundlagen- und klinischen Berei­ch
wurde an allen Standorten der Cha5
Einführung
Eine Initiative für einen Sonderforschungsbereich zum
Thema Vaskulärer Stress ist auf den Weg gebracht. Sie
wird wesentlich von Mitgliedern des CCR bestritten.
Vielfache räumliche Umstrukturierungen sowie zeitraubende Vorbereitungen für einen neuen medizinischen
Studiengang an der Charité, beide insbesondere den
Bereich der Vorklinik betreffend, haben dieses Projekt
verzögert. Parallel dazu beteiligt sich eine Reihe von
Wissenschaftlern des CCR an einem Projektantrag
der Charité für ein vom Bundesministerium für Bildung
und Forschung (BMBF) in Zusammenarbeit mit der
Helmholtz-Gesellschaft initiiertes deutschlandweites
Vernetzungsprojekt namens „Deutsches Zentrum für
Herz-Kreislauf-Forschung (DZHK)“. Die Initia­tivanträge
zu diesem Projekt sind eingereicht; eine erste Auswahl unter den Antragstellern wird Ende des Jahres
stattfinden.
Eine weitere Bereicherung des CCR stellt die Aufnahme von Dr. Michael Schupp im August 2010 dar,
eines ehemaligen Doktoranden aus dem Institut für
Pharmakologie im CCR, der nach einem fünfjährigen
Forschungsaufenthalt in den USA als Stipendiat der
Emmy-Noether-Stiftung der DFG ans CCR zurückgekehrt ist (siehe spezifischer Text).
Diesen erfreulichen Entwicklungen steht ein grosser
Verlust entgegen: die Arbeitsgruppe von Prof. Patricia
Ruiz Noppinger hat Ende 2009 das CCR verlassen,
da sich Frau Prof. Ruiz anderen Aufgaben im wissenschaftlich-administrativen Bereich zugewandt hat.
Die organisatorische Struktur des CCR hat sich im
Berichtszeitraum nicht wesentlich verändert; sie
ist durch die im Jahre 2008 vom Fakultätsrat der
Charité verabschiedete neue Satzung des CCR
vorgegeben. Der CCR-Vorstand setzt sich wie folgt
zusammen:
Im Mai 2010 hat das CCR eine wichtige Bereicherung erfahren durch AufCCR-ORGANISATION :
nahme des von Prof. Pontus
Vorstand
Persson geleiteten Institutes für
Vegetative Physiologie an der Cha• Prof. Thomas Unger (Direktor)
• Prof. Axel R. Pries (stellvertretender Direktor)
rité. Die Umsiedlung dieses Institutes
• Prof. Ulrich Kintscher
war erforderlich geworden, da die
• Prof. Harm Peters (kooptiert im Januar 2010 nach
Ausscheiden von Prof. Ruiz Noppinger)
Charité sich von einer Vielzahl von
• Dekan/Dekanin der Charité
Immobilien ausserhalb ihrer eigentSatzungsgemäß gibt es weiterhin ein wichtiges Gremium, den Nutzerrat, der sich
lichen Standorte getrennt hat. Die
aus Projektleitern und technischen Mitarbeitern zusammensetzt.
Einsicht in den gemeinsamen wisIn jüngster Zeit hat sich ein weiteres Gremium etabliert, die Arbeitsgruppen- und
senschaftlichen Vorteil der VereiniProjektleiterkonferenz, die zwar in der CCR-Satzung nicht vorgesehen ist, sich
aber zu einer wichtigen Informations- und Austauschbörse entwickelt hat, die einmal
gung sowie die Bereitschaft beider
im Monat tagt.
Seiten zu räumlichen EinschränkunSeit 2010 hat das CCR eine wissenschaftliche Koordinatorin in der Person von
gen haben die Eingliederung der
Frau Dr. rer. nat. Karin Effertz. Sie kümmert sich um die Belange der Öffentlichkeits­
Physiologie in ein bereits voll belegarbeit und unterstützt Verbundanträge und Verbundprojekte am CCR.
tes Forschungszentrum erleichtert.
6
Einführung
Die einzelnen wissenschaftlichen Schwerpunkte des CCR haben
sich in den vergangenen
Jahren unterschiedlich
entwickelt:
Die kardiovaskuläre Geschlechterforschung ist
ihren Weg konsequent weiter gegangen. Die von
Prof. Regitz-Zagrosek geleitete DFG-Forschergruppe
zum Thema geschlechtsspezifischer Mechanismen
der Myokardhypertrophie, die wesentlich von CCRWissenschaftlern getragen wird, sieht einer zweiten
Förderperiode entgegen (siehe gesonderter Textbeitrag); das DFG-Graduiertenkolleg 754-Myokardiale
Genexpression- und Funktion
befindet sich in der Auslaufphase.
Dieses Kolleg hat mit seinen zahlreichen Stipendiaten/innen die
wissenschaftliche Arbeit am CCR
seit seiner Gründung bereichert.
Der relativ neue Forschungsschwerpunkt Drug Deve­
lopment hat sich besonders rapide entwickelt: Die
der Arbeitsgruppe von PD Heiko Funke-Kaiser vom
BMBF im Rahmen des Go Bio-Programms sowie von
der Investitionsbank Berlin (IBB) im Rahmen des Pro­
fit-Programms bereitgestellten Mittel in Millionenhöhe
für das Projekt zur Entwicklung eines Hemmstoffs des
vor einigen Jahren entdeckten (Pro)-Renin-Rezeptors
wurden erfolgreich zur Findung von Molekülen mit den
gewünschten Eigenschaften eingesetzt. Die Forderung des BMBF, mit Hilfe des Go Bio-Projektes eine
Start-Up Firma aus der Charité heraus zu gründen,
wurde erfüllt: Mit den Gründern PD Heiko FunkeKaiser, Dr. Jan Schefe, Prof. Thomas Unger und Dr.
Frank Zollmann entsteht die Firma CCR-Pharma
GmbH (siehe spezifischer Text zu diesem Projekt).
Grosse Fortschritte hat ein weiteres Projekt aus diesem
Bereich gemacht: die Entwicklung eines Agonisten:
des Angiotensin AT2-Rezeptors zum Medikament. Die
von Dr. Ulrike Steckelings geführte Arbeitsgruppe hat
den ersten Vertreter dieser neuen Substanzgruppe,
Compound 21, erfolgreich in einer Vielzahl von experimentellen Ansätzen in vitro und in vivo getestet.
Zahlreiche Kooperationen im In- und Ausland sind im
Rahmen dieses Projektes entstanden. Compound 21
wird nach Abschluss gegenwärtiger toxikologischer
Untersuchungen voraussichtlich Anfang 2011 in die
Phase I der klinischen Erprobung gehen.
Das in Zusammenarbeit mit der Firma Vicore in
Göteborg/Schweden sowie der Universität Uppsala/
Schweden durchgeführte Projekt wird seit neuestem
aus
Europäischen
Fördermitteln
im
Rahmen des Euro­
stars-Programms
gefördert (siehe spezifischer Text zu diesem Projekt).
Die Arbeitsgruppe um Prof. Ulrich Kintscher beschäftigt sich im Rahmen ihrer Untersuchungen zellulärer
Transkriptionsfaktoren weiterhin mit der Entwicklung
von Modulatoren des Transkriptionsfaktors PPARγ zu
Medikamenten mit metabolischer Indikation auf der
Basis bereits bestehender Strukturen von Arzneimitteln.
Die Entwicklung von Arzneimitteln, in der Vergangenheit fast ausschliesslich eine Domaine der Pharmazeutischen Industrie, hat sich, wie am Beispiel CCR
7
Einführung
zu sehen, zum Teil in den akademischen Bereich verlagert, insbesondere, was die frühen Phasen der Entwicklung angeht. Schlagworte in dieser Hinsicht sind
die sog. Innovationslücke bei der Industrie, aber auch
die sog. Translationslücke bei der Umsetzung von
neuen, der akademischen Forschung entstammenden, Substanzen in die industrielle Weiterentwicklung
zu Arzneimitteln. Angesichts der grossen Bedeutung
dieses aktuellen Umbruchs für die Weiterentwicklung
des CCR findet sich in diesem Report ein Artikel von
Prof. Garret A. FitzGerald, Direktor des Institute for
Translational Medicine and Therapeutics an der University of Pennsylvania, zum Thema „Drug, Industry
and Academia “.
den verschiedenen Charité-Standorten, als auch aus
dem Max-Delbrück Centrum für Molekulare Medizin
(MDC) in Berlin-Buch.
Im Bereich der vaskulären Forschung am CCR hat
sich weiterhin das von PD Ivo Buschmann initiierte und
geleitete internationale Arteriogenese Netzwerk (Art.
Net) mittlerweile fest etabliert. Diese Arbeitsgruppe,
die ein gemeinsames Labor mit derjenigen von Ferdinand LeNoble am MDC betreibt, hat erfolgreich
Fördermittel von Seiten öffentlicher und industrieller
Drittmittelgeber eingeworben und bietet ein gutes Beispiel translationeller Forschung von den theoretischen
Grundlagen bis zur erfolgreichen klinischen Anwendung (siehe spezifischer Text).
Der dritte Forschungsschwerpunkt Vascular Plasti­
city hat in den vergangenen Jahren eine Modifikation
erfahren, im Wesentlichen bedingt durch die schon
erwähnte SFB-Initiative Vascular Stress. In dieser Initiative, die zu mehr als einem Drittel von Mitgliedern
des CCR getragen und von Prof. Axel Pries und Prof.
Thomas Unger geleitet wird, bündeln sich jetzt die
ursprünglichen vaskulären Projekte des CCR zum
Thema Angiogenese, Arteriogenese, Aneurysma und
Transplantationsvaskulopathie, bereichert durch neu
dazu gekommene Forschungsprojekte etwa aus dem
Bereich Genetik und genetisch modifizierter Tiermodelle sowie der mikrovaskulären Funktion
bis zu neuen systembiologischen Ansätzen. Hervorzuheben
ist, dass in dieser Initiative viele kardiovaskulär tätige Arbeitsgruppen in Berlin vereint sind, sowohl aus
Der Forschungsschwerpunkt Metabolism ist weiter
verstärkt worden. Mit der Einbindung der Abteilung
für Endokrinologie, Diabetes und Ernährungsmedizin
(Prof. J. Spranger) ist es gelungen, eine DFG-Nachwuchsgruppe im Emmy-Noether Programm an das
CCR zu rekrutieren. Dr. Michael Schupp hat nach seinem Postdoc-Aufenthalt an der University of Pennsylvania in Philadelphia, USA, im August 2010 seine
Arbeit am CCR aufgenommen.
8
Der Bereich der angewandten Kachexieforschung um
Prof. Stefan Anker konnte mit der SICA-HF Studie eine
von der EU unterstützte Multizenterstudie zur Prävalenz, Persistenz, und Phänotypisierung von Adipositas, Kachexie und Typ 2 Diabetes Mellitus bei Patienten mit chronischer Herzinsuffizienz einwerben und
übernimmt die Studienleitung. Hierdurch konnte ein
wesentlicher Baustein der metabolischen Forschung
des CCRs gestärkt werden.
Die Integration des Forschungsscherpunktes Metabolism in thematisch verwandte Forschungsverbünde
der Charité ist ebenfalls erfolgreich fortgeführt worden.
Einführung
So bestehen nun enge aktive Kooperationen mit den
DFG-geförderten klinischen Forschergruppen 218/1
Hormonal Regulation of Body Weight Maintenance,
und 192/2 Skeletal Muscle Growth and Regulation,
und ermöglichen eine transdisziplinäre angewandte
Metabolismusforschung am CCR.
Forscher aus dem
CCR, insbesondere der jüngst
dazu gestossenen Vegetativen
Physiologie unter Prof. Pontus Persson sind weiterhin massgeblich an einer neuen nierenphysiologisch/
klinisch-nephrologischen Forschergruppe der DFG
beteiligt (FG 1368, Mechanisms of acute kidney injury),
die vor kurzem im CCR begutachtet wurde mit grossen Chancen auf Förderung. Ob sich daraus ein CCRspezifischer Forschungsschwerpunkt ent­wickeln
wird, bleibt abzuwarten; der Vorstand des CCR und
die an der FG 1368 Beteiligten würden eine solche
Entwicklung begrüssen.
Netzwerke und Forschungsverbünde innerhalb
des CCR und mit weiteren Berliner Institutionen
innerhalb und ausserhalb der Charité sind mittlerweile
selbstverständlich geworden. Darüber hinaus sind
die am CCR tätigen Wissenschaftler natürlich auch
bemüht, sich mit Forschern anderer Einrichtungen
national und international zusammen zu schliessen.
Beispiele sind u.a. Anträge bei der transatlantischen Fondation Leducq (Thomas Unger, Ulrike
Steckelings) zusammen mit der US-amerikanischen
Arbeitsgruppe um Irvin Zucker (University of Nebraska) und einer Reihe weiterer Wissenschaftler aus
dem In- und Ausland; das Europäische Forschungsnetzwerk Ingenious HyperCare (Heiko Funke-Kai-
ser), das Berliner Excellenzcluster Neurocure (Ivo
Buschmann) oder die wissenschaftliche Koordination
der NIH-WARCEF-Studie und der oben erwähnten
SICA-HF-Studie durch Stefan Anker.
Weitere Einzelheiten sind der Zusammenstellung weiter unten zu entnehmen und in den Berichten der einzelnen Arbeitsgruppen angeführt.
Geplant ist, unter der Leitung von Ulrich Kintscher und
PD Kai Kappert, wieder ein EU-finanziertes Graduier­
tenprogramm ans CCR zu holen als Fortsetzung der
bisher so erfolgreichen Graduiertenprogramme der
DFG und der Europäischen Union (EU) (Graduiertenkolleg 754; EU Graduiertenprogramm Cardiovasc /
Marie Curie Early Stage Training Program) sowie das
gemeinsame Graduierten- und Trainingsprogramm in
Vascular Medicine der Universitäten Padua, Gdansk
und Charité/CCR weiter zu führen. Die oben erwähnte
SFB Initiative Vascular Stress beinhaltet ebenfalls ein
Graduiertenprogramm.
Am 15./16. Februar dieses Jahres fand die zweite wissenschaftliche Vor-Ort Begutachtung des CCR
durch den Wissenschaftlichen Beirat des CCR statt,
ein Gremium international renommierter deutscher und
ausländischer Wissenschaftler (siehe Kasten).
CCR – EXTERNE GUTACHTER 2010
•
•
•
•
•
•
•
•
•
•
Prof. Matthias Blüher, Leipzig, Germany
Prof. Thomas Eschenhagen, Hamburg, Germany
Prof. Jürgen Floege, Aachen, Germany
Prof. Xavier Jeunemaitre, Paris, France
Prof. Michael Mulvany, Aarhus, Denmark
Prof. Jeremy Pearson, London, UK
Prof. Thomas Philipp, Essen, Germany
Prof. Eberhard Ritz, Heidelberg, Germany
Prof. Heikki Ruskoaho, Oulu, Finland
Prof. Bernward Schölkens, Frankfurt, Germany
9
Einführung
An dieser Begutachtung nahm auch der Prodekan
für Forschung der Charité, Prof. Rudolf Tauber, teil.
Vorbereitet wurde die Begutachtung seitens des CCR
durch eine Klausurtagung aller Projektleiter im Januar
in Bad Saarow / Brandenburg. Ein Auszug aus dem
Bericht der Gutachter an die Medizinische Fakultät der
Charité ist im Folgenden abgedruckt:
"General
Founded in 2003, the Center for Cardiovascular Research (CCR) is an interdisciplinary center with 12 research
groups and about 130 projects. These impressive numbers are echoed by the scientific output, multiple
graduate and training programs, participation in comprehensive and collaborative scientific projects, the
external funding record, the structural organization representing all research groups and the rich stream of
research proposals. Overall, there was a unanimous feeling of an excellent centre of cardiovascular research
that should be encouraged to continue based on its remarkable performances and possibly reinforce its
activities.
Strengths
The committee was favourably impressed by the high quality and the diversity of research performed at the
CCR, from basic science and experiments to clinical trials and drug development. There was a consensus
that this centre conducts research which is really original, innovative and productive.….The activities related
to education and training of young scientists and students are also one of the strengths of the centre. Nine
students got their Ph.D. through the Marie Curie program in the last 4 years. This program as well as the
Graduate course coordinated by Prof Vera Regitz-Zagrosek are important instruments not only for the students, but also to stimulate discussions and collaborations between the groups of the CCR.
Limitations
Cardiovascular research in Berlin probably suffers from not being sufficiently visible and structured. As a first
step, Prof Unger informed us that he would submit an application to the SFB programme, joining 16 teams of
cardiovascular research, many of them being in the CCR, others being outside, especially at the MDC (Prof
Bader, Prof Hübner). The committee unanimously supported this initiative, hoping that it would get funded
– the project being already well ranked in 2009. Success would provide a platform for a complementary
application in 2011 for a Graduate School in Cardiovascular Medicine.
Another limitation is the very limited amount of financial and structural support given by the Faculty to the
CCR…. The committee believes that it is essential for the viability of the CCR that it has at least some central
funds to allow coordination. Even though the committee understands that the Charité has to face difficult
times with a 25% reduction in its overall budget, it was highlighted that the CCR on its own provided approximately 8% of the external funding of the Faculty. This should be taken into account, and closer support and
connections are strongly requested…
10
Einführung
The third limitation that appeared…was the extent of new joint research projects between groups of the
CCR. Links and participation to common projects exist, but there is still room for more formal established
and previously discussed common projects. In that regard, the number of scientific publications involving
several groups at the CCR appeared to be still too limited.
Finally, the advisors also agreed that some very few projects did not really fit into the overall concept. It would
be an option for the CCR to further focus the activities and to redistribute resources and space to support
the more active and collaborating groups. In that regard, the expectation that a young researcher, Dr Michael
Schupp at the moment in the USA, who has obtained funds for the DFG on a project on metabolism, will
return and join the CCR, perfectly fits with the renewal of the structure and research foci. Clear procedures
for reallocating space in the light of scientific development should however be developed…
Concluding remarks
During the past three years, the CCR has successfully continued to be a unique model of an interdisciplinary
centre for cardiovascular research with very good productivity and performance. The output of the last 3
years is the most appropriate justification for continued support, development and growth. The scientific
direction towards 4 research foci and a greater impact on drug development has been favourably appreciated by the committee.
Since its start in 2003, the CCR has developed into an innovative and productive entity at the university.
The advisors recommend that the university Charité takes actions to further expand the CCR to strengthen
interdisciplinary and translational approaches in cardiovascular research in Berlin. Core functions and infrastructures, in particular, deserve active support from the university. In addition, some improvements in the
CCR governance (especially increased networking between groups, increased common budget, better
possibilities for space reallocation with the Faculty) should improve the visibility and the performance of this
excellent research centre.
CCR should under all circumstances to be continued based on the excellent track record, the
past performance and future plans”.
Soweit zum Bericht des externen Wissenschaftlichen
Gutachtergremiums über das CCR im Februar 2010.
Der Vorstand des CCR möchte den Gutachtern an
dieser Stelle nochmals für ihre Arbeit und die Zeit, die
sie für unser Zentrum aufgebracht haben, ganz herzlich danken.
11
Einführung
Um dem Gutachterurteil noch Substanz hinzuzufügen, seien im Folgenden einige Zahlen und Fakten
angeführt:
Schliesslich gilt unser
Dank wiederum auch
der Firma Boehringer
Ingelheim, die nun schon im
siebten Jahr kontinuierlich die
CCR Seminarreihe BI-Lectures
grosszügig unterstützt und damit den internationalen
wissenschaftlichen Austausch im CCR fördert (siehe
spezifischer Text).
Sieben Jahre CCR: besser als in der Anfangsphase
dieses Zentrums kann man heute abschätzen, was im
Hinblick auf die ursprünglichen Ziele bereits gelungen
und was noch verbesserungsbedürftig ist. Ablesen
lässt sich das auch im Urteil der externen Gutachter:
Sie bestätigen den Wert der Interdisziplinarität, mahnen aber eine höhere Intensität der Zusammenarbeit
innerhalb der Arbeitsgruppen des CCR an. Sie anerkennen das unter erheblichen finanziellen Restriktionen bisher Erreichte, wünschen sich aber eine stärkere
Förderung durch die Charité sowie mehr Visibilität des
Zentrums nach aussen. Sie begrüssen die individuellen wissenschaftlichen Leistungen, sprechen sich
aber auch für eine Verstärkung der Vernetzung in grösseren Forschungsverbünden sowie für Expansion des
Zentrums aus, um der kardiovaskulären Forschung an
der Charité mehr Gewicht zu verleihen.
Wie wird sich die Zukunft des CCR gestalten?
Gegenwärtig haben sich starke Tendenzen ent­wickelt,
die die Zukunft der Wissenschaft vornehmlich in
öffentlich geförderten Forschungsverbünden sehen,
wie z.B. den klassischen Sonderforschungsbereichen
der DFG oder den neuerdings in Deutschland geförderten sog. Exzellenzinitiativen, auf die man grosse
Hoffnungen setzt. Kritisch ist dazu anzumerken, dass
12
Einführung
schaftliche Netzwerke bergen die Gefahr,
diese Entwicklung zu verpassen; auch sie
müssen sich weiterentwickeln, um den wissenschaftlichen Fortschritt nicht zu behindern.
im Rahmen dieser Entwicklung viele Wissenschaftler
ihre Forschungsthemen häufig nicht mehr nach individueller Neigung und Eignung, sondern eher nach
ihrer Netzwerktauglichkeit aussuchen. Das muss nicht
unbedingt mit einer Qualitätssteigerung einhergehen.
Während weiterhin nicht von der Hand zu weisen ist,
dass durch derartige Netzwerke Synergien ausgehoben und die oft knappen Ressourcen gemeinsam
besser genützt werden können, bleibt doch festzuhalten, dass bis zum heutigen Tag die wirkliche Exzellenz
wissenschaftlicher Leistung immer individuell erbracht
worden ist, oft ohne Einbindung in Forschungsverbünde, ja, dass diese aus verschiedenen Gründen
der individuellen Exzellenzentfaltung durchaus auch
hinderlich sein können. Darüber hinaus gilt es zu
bedenken, dass auch die wissenschaftlichen Strukturen selbst einem kontinuierlichen Wandel unterworfen sind. Ein Beispiel dafür ist Entwicklung des
Zusammenspiels von akademischer Forschung und
industrieller Entwicklung im Hinblick auf Arzneimittelentwicklung, auf die in diesem Report ausführlich
eingegangen wird. Traditionelle akademisch-wissen-
Das CCR bewegt sich nach wie vor im
Spannungsfeld zwischen gewollter interdisziplinärer Kooperation und individueller
Forschungsleistung. Es wird sich, um sein
Bestehen zu sichern, intensiv um Forschungsnetzwerke aus seinen eigenen
Reihen und im Verbund mit anderen Institutionen kümmern, gleichzeitig jedoch eine
noch stärkere wissenschaftliche Fokussierung anstreben und vor allem individuelle Forscher von
hoher Qualität anziehen und integrieren müssen. Dies
um so mehr, als die einzelnen Forschungsschwerpunkte der Charité gegenwärtig in einem intensiven
Wettbewerb um Wissenschaftler und Ressourcen
stehen, in dem sich die kardiovaskuläre Forschung
gegenüber anderen Disziplinen wie etwa den Neurowissenschaften nur behaupten kann, wenn sie innerlich geeint auf exzellente wissenschaftliche Qualität
setzt und dies auch nach aussen hin glaubhaft vermitteln kann.
Unter diesen Voraussetzungen sollte das CCR
als Kristallisationspunkt und Integrationszentrum
kardiovaskulärer Forschung an der Charité auch
über deren 300-jährigen und seinen eigenen siebten Geburtstag hinaus in eine erfolgreiche Zukunft
blicken können.
Prof. Dr. med. Thomas Unger
Direktor des CCR
13
Introduction
INTRODUCTION
T
he Center for Cardiovascular Research
can look back on seven years of successful research work. The CCR was founded in
2003 as an interdisciplinary cardiovascular research
center at the Charité, a renowned, traditional site of
medical research that this year celebrates its 300th
anniversary.
At the time the CCR was founded, the “old” Charité,
i.e. the hospital in Berlin-Mitte, which served as the
medical faculty of the Humboldt University Berlin and
was the medical flagship of the German Democratic
Republic (GDR), had already undergone a merger
with the Virchow Hospital in West Berlin. Another
merger was undertaken in the following decade with
the university hospital “Benjamin Franklin” of the Free
University in (West) Berlin. The Charité as an independent corporation with nearly 15,000 employees
thus became the largest university hospital in Europe.
Similar developments have taken place in other European capitals, e.g. in London and Paris. These mergers had a primarily economic background and were
not necessarily scientific or patient-oriented.
Basic and clinical cardiovascular research was carried
out at all Charité sites on a very high level; however,
it was not coordinated to the desired extent. Different
backgrounds and traditions of the individual faculties
and hospitals and insufficient networking with regard
14
to content and structure within Berlin’s cardiovascular
research institutions, between cardiologists, nephrologists and cardiovascular-oriented physiologists and
pharmacologists, between basic and clinical researchers at different Charité sites can be viewed as a reason
why in Berlin – except for one “Transregio” network of
the German Research Foundation (DFG) on the topic
of myocarditis – large-scale research networks like a
DFG collaborative research center on cardiovascular
diseases have not yet been established.
The founding of the CCR was connected with the hope
of bridging this gap and of reorganizing the sometimes
divergent cardiovascular research activities into joint
project endeavors. So far this has succeeded in part.
An initiative for a collaborative research center (SFB)
on Vascular Stress has been launched, mainly through
the efforts of members of the CCR. Much restructuring
with respect to utilization of space and facilities as well
as time-consuming preparations for a new medical
curriculum at the Charité, both of which have had a
great impact on the preclinical program, have delayed
this project. Parallel to this, a number of CCR scientists
are participating in a project proposal of the Charité
for a Germany-wide network project “German Cardiovascular Center (DZHK)” initiated by the German
Federal Ministry of Education and Research (BMBF) in
collaboration with the Helmholtz Association. The first
proposals for this project have been submitted; selection of applicants will take place at the end of the year.
In May 2010 the CCR made an important gain through
the incorporation of the Institute of Vegetative Physiology, directed by Professor Pontus Persson, into the
Charité. The resettlement of this institute became
Introduction
necessary because the Charité was in the process
of selling many of its properties outside of its primary
locations. The scientific advantages of the incorporation and the willingness of both sides to accept space
limitations have facilitated the integration of physiology
into our already fully occupied research center.
Another addition to the CCR is Dr. Michael Schupp, a
former doctoral student at the Institute of Pharmacology in the CCR, who returned to the research center
in August 2010 after a five-year research stay in the
U.S as fellow of the Emmy Noether Foundation of the
German Research Foundation (see specific text).
These positive developments stand in contrast to
a great loss: the research group of Professor Patricia Ruiz Noppinger left the CCR at the end of 2009
because Professor Ruiz Noppinger accepted a position with other scientific-administrative tasks.
The individual research
areas of the CCR have
developed differently in the
past years:
Cardiovascular gender
research has continued its work. The DFG research
group headed by Prof. Regitz-Zagrosek on the topic of
gender-specific mechanisms of myocardial hypertrophy, which is primarily comprised of CCR scientists, is
looking forward to a second grant period (see separate
article). The DFG Graduate School 754 “Myocardial
Gene Expression and Function” is
being phased out. This graduate
school with its many scholarship
holders has enriched the scientific
work at CCR since its inception.
The relatively new research focus Drug Development
has developed especially fast. The research group led
by PD Heiko Funke-Kaiser has received funds amounting to millions of euros provided by the BMBF within
the scope of the Go Bio scheme and by the Investitionsbank Berlin (IBB) as part of the Profit program.
These funds have been successfully used for
CCR ORGANIZATION :
a project to develop an inhibitor of the (pro)Executive Board
renin receptor discovered some years ago and
• Prof. Thomas Unger (Director)
specifically to find molecules with the desired
• Prof. Axel R. Pries (Deputy Director)
characteristics. The requirement of the BMBF
• Prof. Ulrich Kintscher
• Prof. Harm Peters (co-opted in January 2010
to found a start-up company as spin-off from
after the resignation of Professor Ruiz Noppinger)
• Dean of the Charité
the Charité with the aid of the Go Bio scheme
According to the statute, there is still another important body, the Users’ Council,
has been met: The CCR-Pharma GmbH is
which is made up of project leaders and technical staff.
currently being established by its founders
Recently, yet another body has been established, the Conference of Research
Group and Project Leaders, which is not provided for in the CCR statute, but which
PD Heiko Funke-Kaiser, Dr. Jan Schefe, Prof.
has developed into an important information exchange and which meets once a month.
Thomas Unger and Dr. Frank Zollmann (see
Since 2010 the CCR also has a scientific coordinator, Dr. Karin Effertz. She is in
specific text on this subject).
charge of public relations and provides support for cooperative applications and
The organizational structure of the CCR has not
changed significantly in this reporting period; it is
prescribed by the new statute of the CCR which was
adopted by the Faculty Council of the Charité in 2008.
The CCR Executive Board is comprised of the following members:
projects at the CCR.
15
Introduction
Another project has made great progress in this area:
the development of an angiotensin AT(2) receptor agonist into a drug. The research group led by Dr. Ulrike
Steckelings has successfully tested the first representative of this substance group, Compound 21, in a
number of in vitro and in vivo experiments. A number
of collaborations related to this project have evolved
both in Germany and internationally. Following completion of the current toxocological tests, Compound
21 is due to enter Phase I clinical trials at the beginning
of 2011.A project carried out in collaboration with the
company Vicore Pharma in Gothenburg/Sweden and
Uppsala University/Sweden has just recently received
an
EU
grant
within the scope
of the Eurostars
program
(see
corresponding
text about this
project).
The research group led by Professor Ulrich Kintscher,
within the scope of its investigations into cellular transcription factors, is studying modulators of the transcription factor PPARγ in order to develop drugs with
a metabolic indication, based on already existing drug
structures.
Drug development, in the past almost exclusively a
domain of the pharmaceutical industry, as can be seen
in the example of the CCR, has now been taken over
in part by the academic community, especially with
respect to the early phases of development. Keywords
here are the so-called innovation gap in industry but
also the translational gap in the implementation of
new substances originating from academic research
in further industrial drug development. In the face of
16
the great importance of this current change for the
further development of the CCR, this article contains
a report by Prof. Garret A. FitzGerald, director of the
Institute for Translational Medicine and Therapeutics at
the University of Pennsylvania, on the topic of “Drugs,
Industry and Academia”.
The third research focus Vascular Plasticity has
undergone a modification in recent years, mainly due
to the above-mentioned SFB initiative Vascular Stress.
More than one-third of all staff members of the CCR are
involved in this initiative, which is headed by Prof. Axel
Pries and Prof. Thomas Unger. The original vascular
projects of the CCR on arteriogenesis, aneurysma and
transplantation vasculopathy have now been enriched
by new research projects from the area of genetics and
genetically modified animal models as well as microvascular function, including new approaches from
systems biology. Significantly, many cardiovascular
research groups in Berlin are involved in this initiative,
both from the different Charité locations and from the
Max Delbrück Center for Molecular Medicine (MDC)
in Berlin-Buch.
In the field of vascular research at the CCR, the Arteriogenesis Network (Art.Net) initiated and headed by
PD Ivo Buschmann has since become well established
internationally. This research group, which runs a joint
laboratory with the
research group of Ferdinand LeNoble at the
MDC, has successfully solicited funding
from the public sector
and from industry and
is a good example of
translational research
from the theoretical
Introduction
foundations to successful clinical application (see
specific text).
The research focus Metabolism has been further
strengthened. With the integration of the Department
of Endocrinology, Diabetes and Nutrition Medicine
(Prof. J. Spranger), the CCR has now succeeded in
recruiting a DFG Junior Research Group within the
scope of the Emmy Noether Program to the CCR:
Dr. Michael Schupp began his work at the CCR in
August 2010 after his postdoc stay at the University
of Pennsylvania in Philadelphia, USA.
With its SICA-HF study, the area of applied research on
cachexia led by Prof. Stefan Anker has succeeded in
acquiring a grant for a multi-center study funded by the
EU on the prevalence, persistence, and phenotyping
of adipositas, cachexia and type 2 diabetes mellitus
in patients with chronic heart failure, and Professor
Anker will now direct the study. This has strengthened
an important component of metabolic research at the
CCR.
The integration of the research area Metabolism into
thematically related research networks of the Charité
has also been continued. As a result, there is now
close active cooperation with the DFG-funded clinical research groups 218/1 Hormonal Regulation of
Body Weight Maintenance and 192/2 Skeletal Muscle
Growth and Regulation, and this enables cross-disciplinary applied research in metabolism at the CCR.
Researchers from the CCR, especially in the recently
established Vegetative Physiology research group led
by Professor Pontus Persson, are extensively involved
in a nephrophysiological/ clinical-nephrolo­gical
research group of the DFG (FG 1368, Mechanisms
of acute kidney injury). It was recently evaluated in the
CCR and has good chances of funding. One must wait
and see whether a CCR-specific research focus will
develop from this; the CCR Board of Directors and the
researchers involved in the FG 1368 research group
would welcome such a development.
Networks and cooperative research projects
within the CCR and with other Berlin institutions
inside and outside the Charité are meanwhile taken
for granted. Furthermore, the scientists working at the
CCR of course also try to enter collaborations with
researchers of other national and international institutions.
Examples for this
include proposals to
the Fondation Leducq
(Thomas Unger, Ulrike
Steckelings) together
with the U.S. research group led by Irvin Zucker (University of Nebraska) and a number of other scientists in
Germany and abroad; the European research network
Ingenious HyperCare (Heiko Funke-Kaiser), the Berlin
excellence cluster Neurocure (Ivo Buschmann) or the
scientific coordination of the NIH-WARCEF study and
the above-mentioned SICA-HF study by Stefan Anker.
More details are provided in the summary CCR Facts
further below and in the reports of the individual
research groups.
It is planned to again solicit an EU-funded graduate
program for the CCR, headed by Ulrich Kintscher and
PD Kai Kappert as continuation of the very successful
graduate programs (DFG graduate research school
754, and the EU graduate program Cardiovasc / Marie
Curie Early Stage Training Program) and to continue
the joint graduate and training program in Vascular
Medicine of the Universities of Padua, Gdansk and
17
Introduction
the Charité/CCR. The above-mentioned SFB-initiative
Vascular Stress also has a graduate program.
On February 15-16, 2010 the second external scientific evaluation of the CCR was conducted onsite by the CCR Scientific Advisory Board, a body of
internationally renowned scientists from Germany and
other countries (see box).
Prof. Rudolf Tauber, Vice Dean of Research at the
Charité, also took part in this evaluation. The evaluation was prepared on the part of the CCR in a retreat
of all project leaders in Bad Saarow / Brandenburg
in January. An excerpt from the report of the evaluators to the Medical Faculty of the Charité is printed
below:
CCR – EXTERNAL EVALUATORS 2010
•
•
•
•
•
•
•
•
•
•
Prof. Matthias Blüher, Leipzig, Germany
Prof. Thomas Eschenhagen, Hamburg, Germany
Prof. Jürgen Floege, Aachen, Germany
Prof. Xavier Jeunemaitre, Paris, France
Prof. Michael Mulvany, Aarhus, Denmark
Prof. Jeremy Pearson, London, UK
Prof. Thomas Philipp, Essen, Germany
Prof. Eberhard Ritz, Heidelberg, Germany
Prof. Heikki Ruskoaho, Oulu, Finland
Prof. Bernward Schölkens, Frankfurt, Germany
“General
Founded in 2003, the Center for Cardiovascular Research (CCR) is an interdisciplinary center with 12 research
groups and about 130 projects. These impressive numbers are echoed by the scientific output, multiple
graduate and training programs, participation in comprehensive and collaborative scientific projects, the
external funding record, the structural organization representing all research groups and the rich stream of
research proposals. Overall, there was a unanimous feeling of an excellent centre of cardiovascular research
that should be encouraged to continue based on its remarkable performances and possibly reinforce its
activities.
Strengths
The committee was favourably impressed by the high quality and the diversity of research performed
at the CCR, from basic science and experiments to clinical trials and drug development. There was a
consensus that this centre conducts research which is really original, innovative and productive.….The
activities related to education and training of young scientists and students are also one of the strengths
of the centre. Nine students got their Ph.D. through the Marie Curie program in the last 4 years. This
program as well as the Graduate course coordinated by Prof Vera Regitz-Zagrosek are important
instruments not only for the students, but also to stimulate discussions and collaborations between the
groups of the CCR.
18
Introduction
Limitations
Cardiovascular research in Berlin probably suffers from not being sufficiently visible and structured. As a first
step, Prof Unger informed us that he would submit an application to the SFB programme, joining 16 teams of
cardiovascular research, many of them being in the CCR, others being outside, especially at the MDC (Prof
Bader, Prof Hübner). The committee unanimously supported this initiative, hoping that it would get funded
– the project being already well ranked in 2009. Success would provide a platform for a complementary
application in 2011 for a Graduate School in Cardiovascular Medicine.
Another limitation is the very limited amount of financial and structural support given by the Faculty to the
CCR…. The committee believes that it is essential for the viability of the CCR that it has at least some central
funds to allow coordination. Even though the committee understands that the Charité has to face difficult
times with a 25% reduction in its overall budget, it was highlighted that the CCR on its own provided approximately 8% of the external funding of the Faculty. This should be taken into account, and closer support and
connections are strongly requested…
The third limitation that appeared…was the extent of new joint research projects between groups of the
CCR. Links and participation to common projects exist, but there is still room for more formal established
and previously discussed common projects. In that regard, the number of scientific publications involving
several groups at the CCR appeared to be still too limited.
Finally, the advisors also agreed that some very few projects did not really fit into the overall concept. It
would be an option for the CCR to further focus the activities and to redistribute resources and space
to support the more active and collaborating groups. In that regard, the expectation that a young
researcher, Dr Michael Schupp at the moment in the USA, who has obtained funds for the DFG on
a project on metabolism, will return and join the CCR, perfectly fits with the renewal of the structure
and research foci. Clear procedures for reallocating space in the light of scientific development should
however be developed…
Concluding remarks
During the past three years, the CCR has successfully continued to be a unique model of an interdisciplinary
centre for cardiovascular research with very good productivity and performance. The output of the last 3
years is the most appropriate justification for continued support, development and growth. The scientific
direction towards 4 research foci and a greater impact on drug development has been favourably appreciated by the committee.
Since its start in 2003, the CCR has developed into an innovative and productive entity at the university.
The advisors recommend that the university Charité takes actions to further expand the CCR to strengthen
interdisciplinary and translational approaches in cardiovascular research in Berlin. Core functions and infrastructures, in particular, deserve active support from the university. In addition, some improvements in the
19
Introduction
CCR governance (especially increased networking between groups, increased common budget, better
possibilities for space reallocation with the Faculty) should improve the visibility
and the performance of this excellent research centre.
The CCR should under all circumstances be continued based on the excellent track record, the
past performance and future plans”.
This excerpt was taken from the report of the evaluation of the CCR conducted by the external Scientific
Advisory Council in February 2010. On behalf of the
Executive Board of the CCR I would once again like
to take this opportunity to express our appreciation
to the evaluators for their work and for the time they
devoted to our center.
As complement to the evaluation report, a summary of
key statistics is provided in CCR Facts 2010:
Finally, we would once again like to express our appreciation to Boehringer Ingelheim, which for the last
seven years has so generously supported the CCR
lecture series BI Lectures and has thus promoted
international scientific exchange in the CCR (see specific text).
20
Seven years of the CCR: Better than during the initial phase of this center, we can assess today how
many of the original objectives have been achieved
and what still needs improvement. This can also be
determined from the evaluation of the external experts:
They confirm the value of interdisciplinarity, but advo-
Introduction
cate a higher intensity of cooperation within research
groups of the CCR. They commend what has been
achieved under considerable financial constraints, but
call for greater support by the Charité and greater visibility outward for our center. They welcome individual
academic achievement, but also advocate a strengthening of networking in larger research networks and
an expansion of the Center in order to give more weight
in the future to cardiovascular research at the Charité.
What form will the CCR take in the future?
Currently, there is a strong tendency to view the future
of science primarily in publicly funded research networks, such as the classic Collaborative Research
Centers of the DFG and, most recently in Germany, the
so-called Excellence Initiatives, in which one places
great hope. One criticism here is that, due to this development, many scientists do not choose their research
topics based on their own interests and abilities, but
rather according to how well it would be suitable for a
network. This must not necessarily be accompanied
by an increase in quality. While it must be admitted
that such networks enable synergies and can often
make better use of the often scarce resources, the
fact remains that even today real excellence in scientific performance is always accomplished individually,
often without any connection to research networks
which, for various reasons, may actually turn out
to be a hindrance to the individual development of
excellence. In addition, it is important to remember
that the scientific structures themselves are subject
to continuous change. One example is the develop-
ment of the interplay between academic research and
industrial development for drug development which is
discussed in detail in this report. Traditional academic
and scientific networks pose a risk of missing out on
this development; they too must evolve in order not to
hinder scientific progress.
The CCR is still finding its way between the conflicting priorities of intentional interdisciplinary cooperation
and individual research performance. It will, in order to
secure its existence, have to maintain intensive research
networks among its members and increase networking
with other institutions. However, it must also strive to
achieve a stronger academic focus. First and foremost,
it must attract top-level researchers and integrate them
into the overall mission of the CCR. All the more so,
because the individual research foci of the Charité are
currently in an intense competition for scientists and
resources in which cardiovascular research can only
compete with other disciplines such as neuroscience if
it is internally united in its pursuit of excellent scientific
quality and builds on its outward credibility.
Under these conditions, the Center for Cardiovascular
Research, as crystallization and integrative focal point
of cardiovascular research at the Charité, can look
forward far beyond the Charité’s 300th anniversary
and its own seven-year birthday to a successful future.
Thomas Unger MD, PhD, FESC, FAHA
CCR Director
21
Stefan Anker – Applied Cachexia Research
Head of the group
Prof. Dr. Dr. med. Stefan D. Anker
Curriculum Vitae: Stefan D. Anker is Professor of Cardiology and Cachexia
Research at the Department of Cardiology of Charité Berlin, Campus Virchow-Klinikum, in Berlin / Germany (since 2002).
Dr. Anker studied medicine and obtained his M.D. (1993) from Charité Medical School
in Berlin, Germany. He went on to earn a Ph.D. (1998) at the National Heart & Lung
Institute, Imperial College London, UK.
Dr. Anker has authored more than 350 original papers, reviews, and editorials. His published work on the one
hand addresses many issues of the pathophysiology and treatment of acute and chronic heart failure and on
the other hand focuses on treatment development to prevent and reverse muscle wasting, sarcopenia and
cachexia at the animal and human level.
Additionally, Dr. Anker is very active in biomarker, anaemia, metabolic, immunological and nutrition research.
Since 2004, Dr. Anker is extending his work on cachexia research beyond heart failure to other chronic illnesses,
including cancer, COPD, CKD and aging. For his work, Dr. Anker has received numerous grants and several
clinical research awards.
Dr. Anker serves on the editorial boards of 5 scientific journals. Dr. Anker is founding Editor-in-Chief of the
Journal of Cachexia, Sarcopenia and Muscle (JCSM).
Dr. Anker is member of a number of international steering committees (chairing or co-chairing 3), several
DSMB’s (chairing 2) as well as several end-point committees of clinical trials in heart failure and in the field of
cachexia treatment and prevention.
Dr. Anker serves in the board of the Heart Failure Association (HFA) of the European Society of Cardiology
(since 2006) and now is President Elect of HFA (since 2010). He was the chairman of the scientific committee
of the 2010 Annual Meeting of HFA (Berlin, 29.5.–1.6.2010).
In 2010, Dr. Anker became Fellow of the European Society of Cardiology (FESC).
Dr. Anker is the founding president of the International Society on Cachexia and Wasting Disorders (SCWD) –
for more information see www.cachexia.org.
23
Stefan Anker – Applied Cachexia Research
Members of the group
Scientists
Springer, Jochen
Dr. rer nat., lab leader
Barkhudaryan, AnushMD, (guest scientist, Erebouni
Medical Center, Yerevan, Armenia)
Yulia Elkina
Dipl. Biochem.
Rokutan, HirofumiMD, (guest scientist, Tokyo Metropolitan Geriatric
Hospital, Japan)
Palus, Sandra
Veterinarian
von Heahling, Stephan Dr. Dr. med
Technicians
Kiauka, Sabine
24
Apprentice
Students
Baumgarten, Anna
Fetzer, Katharina
Hilman, Arne
Peske, Katrin
Pötsch, Mareike
Schust, Susanne
Suckow, Christian
Tomcakova, Margareta
Tschirner, Anika
PhD-student
DVM-student
PhD-student
DVM-student
Diploma-student
DVM-student
MD-student
MD-student
PhD-student
Stefan Anker – Applied Cachexia Research
Summary
The group is focused on the crippling syndrome of
cachexia. An underlying hypothesis of our work is
that all forms of cachexia as seen in COPD, cancer,
infections and old age lead to pathophysiologically
relevant cardiovascular alterations, namely chronic
heart failure, which is clinically relevant for prognosis.
Disturbance of the endothelial function and the regulation of peripheral perfusion result in ischemia and
hypoxia of the peripheral tissue as well as the induction of reactive oxygen species and inflammation.
We have some evidence that all forms of cachexia,
independently of their underlying chronic disease,
show neurohumoral activation, which is commonly
associated with chronic heart failure. Furthermore,
cachectic patients almost with no exception are short
of breath, fatigue easily and very often show edema,
all of which are hallmarks of chronic heart failure. We
have shown that several therapies of chronic heart
failure display anti-cachectic properties, e.g. ACEinhibitors and beta-blockers. Hence, we are testing
whether these cardiovascular therapies are effective
in other therapeutical areas than cardiac cachexia.
Currently, several projects are focused on cardiovascular drugs and novel compounds on survival,
heart function and body composition in a rat model
of cancer cachexia. Additionally, we are interested
in the effects of chronic use of frusemide on kidney
function and morphology in a rat model of chronic
heart failure on heart function and body composition
in a rat myocardial infarction model.
Methods utilized in these projects include physiological (e.g. monitoring of body weight, body composition by NMR, high resolution echocardiography,
invasive hemodynamics and non-invasive ECG),
immunological (e.g. ELISA, Luminex-assays), biochemical (e.g. Western blotting, enzyme kinetics,
electron paramagnetic resonance spectroscopy
and qRT-PCR) and histopathological (e.g. differential stains, immunohistochemistry and morphometric analysis) techniques.
25
Stefan Anker – Applied Cachexia Research
Zusammenfassung
Der Forschungsschwerpunkt unserer Arbeitsgruppe
liegt im Bereich der Herzinsuffizienz -induzierten
Kachexie.
Eine unserer Arbeit zugrunde liegende Hypothese
ist, dass bei allen Kachexieformen kardiovaskuläre
Veränderungen von besonderer pathogenetischer
Bedeutung sind. Wir denken, dass sich bei allen
Kachexien (z.B. auch bei COPD, Tumoren oder
Infektionen bzw. im hohen Alter) Herzinsuffizienz
entwickelt und klinisch und prognostisch bedeutsam ist.
Störungen der Endothelfunktion und Durchblutungsregulation verursachen Gewebeischämie und
-hypoxie und es kommt zu Radikalproduktion und
lokaler Inflammation. Wir haben erste Hinweise
darauf, dass alle Kachexien (unabhängig von der
zugrunde liegenden chronischen Erkrankung) eine
besonders für die Herzinsuffizienz bekannte neurohormonale Aktivierung aufweisen. Außerdem sind
Patienten mit Kachexie fast ausnahmslos kurzatmig,
leicht ermüdbar, und weisen sehr häufig Ödeme auf
- das sind Kardinalsymptome der Herzinsuffizienz.
26
Wir konnten zeigen, dass viele Herzinsuffizienz-Therapien anti-kachektisch wirken können (z.B. ACEHemmer). Wir wollen testen, ob diese kardiovaskulären Therapien auch außerhalb der (nur scheinbar)
engen Grenzen der kardiovaskulären Medizin antikachektisch wirksam sein können.
Zur Zeit beschäftigt sich ein Projekt mit den Effekten
von Appetit-Stimulanzien auf das Überleben und die
kardiale Funktion im Herzinfarkt-Modell der Ratte, ein
weiteres, klinisches Projekt mit den Mechanismen
des Muskelabbaus und der durch Translokation von
Endotoxin im Darm ausgelösten chronischen Inflammation bei Herzinsuffizienz. Die genannten Projekte
werden mit physiologischen Methoden (Dokumentation der Gewichtsverläufe, Messung der Körperzusammensetzung per MRT, Echokardio­graphie und
invasive Hämodynamik), immunologischen (ELISA),
biochemischen (Western Blotting, Enzymkinetik und
Elektronenspin-Resonanz Spektroskopie) und histo­
pathologischen Methoden (Differentialfärbungen,
Immunhistochemie und morphometrische Untersuchungen) bearbeitet.
Stefan Anker – Applied Cachexia Research
Research projects
1. D
iuretics use and kidney function in a rat model
of chronic heart failure
Project leader
Coworkers
Funding
External cooperations
Jochen Springer
Christian Suckow
self funded
Prof. Dr. med. Berthold Hocher
The use of loop diuretics like frusemide for the treatment of fluid overload in chronic heart failure (CHF)
has been recommended by the American College of
Cardiology guidelines, as patients immediately benefit by the reduction of fluid overload. On the other
hand the use of frusemide has been associated with
an increased risk of hospitalization for the progression of CHF and subsequently increased risk of death
from progressive CHF, as well as an increased cardiovascular and all-cause mortality as compared to
patients receiving no diuretic or a combination therapy
(Domanski et al, J Am Coll Cardiol, 2003). Frusemide
has been shown to induce the neurohumoral activity such as the renin-angiotensin-aldosterone system
(RAAS) in both patients and animals. The RAAS-activation is associated with poorer prognosis and progression of CHF. Inhibition of angiotensin-converting
enzyme reduces the formation and the activity of
RAAS effector hormones, thereby counteracting the
frusemide-induced neurohumoral activation. The
effect on survival of frusemide vs untreated animals
was investigated in a rat model of chronic heart failure
over a period of 6 months. A high mortality was seen
in the frusemid group, which was counteracted by the
use of the ACE-inhibitor ramipril on top of frusemide.
Here we assessed the effect of chronic diuretic-use on
kideney function and morphology (fibrosis, glomerulosclerosis and vascular remodeling).
27
Stefan Anker – Applied Cachexia Research
2. Drug screening in a rat model of cancer cachexia
Project leaders
Stefan Anker, Jochen Springer
CoworkersAnna Baumgarten, Katharina Fetzer, Arne Hilman, Katrin Peske,
Mareike Pötsch, Sandra Palus, Hirofumi Rokutan, Susanne Schust,
Anika Tschirner, Margareta Tomcakova
Funding
self funded, Myotec Therapeutics, ARAIM, Solatarium Dietetics
The rat ascites hepatoma Yoshida AH-130 is a suitable
model system to study the mechanisms involved in the
establishment of cachexia. Its growth causes rapid
and progressive loss of body weight and tissue waste,
particularly in skeletal muscle. Acceleration of tissue
protein breakdown accounts for most of the wasting
in the AH-130 bearers and in particular, skeletal muscle hypercatabolism is mainly due to hyperactivation
of the ATP–ubiquitin-dependent proteolytic system.
High plasma levels of tumour necrosis factor-α (TNF)
and perturbations in the hormonal homeostasis may
play an important role in forcing the metabolic balance
towards the catabolic side. In addition to increased
muscle protein degradation during cancer growth, the
presence of the tumour also induces an increased
rate of apoptosis in skeletal muscle in rats, which also
seems to be activated by cytokines, TNF in particular.
28
The ascites hepatoma Yoshida AH-130 cells (108)
are inoculated into 180-200g male Wistar rats. Alternatively animals receive saline injection only (sham).
The animals are housed in groups of three. The day
after inoculation animals are randomized into various
groups for drug treatment. The rats receive treatment with either placebo or compounds in several
doses over a period maximal 16 days. The primary
endpoints of the study include assessment of body
weight, body composition (via NMR-scanning) and
survival. Secondary endpoints are locomotor activity
and food intake, which serve as indicators for quality
of life, cardiac function (high resolution echocardiography) and organ weights are assessed at the end of
the study (or after death). The model is used to screen
preclinical compounds as well as registered drugs for
second label use.
Stefan Anker – Applied Cachexia Research
3. Progressive loss of cardiac function in cancer cachexia
Project leaders
Jochen Springer, Stefan Anker
CoworkersKatrin Peske, Mareike Pötsch, Sandra Palus, Susanne Schust, Anika Tschirner
Funding
self funded
External cooperationsDr. med. Elena Kaschina, Prof. Dr. med. Thomas Force,
Prof. Dr. Dr. med. Thomas Thum, Prof. Dr. rer.nat. Denise Hilfiker-Kleiner,
Prof. Dr. med. Frederic Jaisser
Cachexia is a very common co-morbidity in cancer
patients which drastically reduces quality of life and
survival. The exact cause of death in cancer is mostly
unknown, yet 22% of the patients are thought to die
from the cachexia itself. However cancer patients
also develop shortness of breath of unknown reason,
which is the hallmark symptom of heart failure. In this
project we assessed the cardiac function in terminally
ill rats with cancer. Tumour-bearing animals shown a
dramatic impairment of cardiac function (see figure
below). Cardiac function was not only significantly
worse than sham at day 11, but also significantly worse
compared to the echocardiographic results obtained
pre-inoculation of the tumor. The heart shows a reduction, i.e. wasting, in weight compared to sham at the
end of the study, which is also reflected in the progressive loss of left ventricular mass and wall thickness
assessed by echocardiography.
4. Heart failure signs in patients with cancer
Project leader
Coworkers
Funding
External cooperations
Jochen Springer, Prof. Dr. Dr. med .Wolfram Döhner
Anush Barkhudaryan
self funded, DAAD
Prof. Dr. med. Manfred Dietel, PD Dr. med. Lars Morawietz
This projects aims to find human correlations for the
heart failure data that we have gathered in the rat cancer cachexia model. Cardiac dimensions, weight and
histology of patients, who died of cancer, is assessed
and correlated to cachexia vs non-cachexia.
29
Stefan Anker – Applied Cachexia Research
5. Cachexia in stroke
Project leader
Coworkers
Funding
External cooperations
Jochen Springer, Prof. Dr. Dr. med .Wolfram Döhner
Katrin Peske, Susanne Schust, Anika Tschirner
BMBF
Prof. Dr. med. Ulrich Dirnagl, PD Dr. med. Andre Rex
The central hypothesis of this project states that body
weight and the loss of body weight contributes to the
morbidity and mortality of patients with a stroke. The
loss of body weight is not considered to be an epiphenomenon or related to co-morbidities, but a result
of stroke-related metabolic changes.
The project addresses this question in a MACO mouse
model using 60 min of ischemia to induce large cerebral infarctions. The aim of the study is to better
characterize the metabolic changes and to assess the
effects of a neuroendocrine activation, which may be
accompanied by inflammatory processes.
6. Effects of xanthine oxidase inhibition in cancer cachexia
Project leader
Coworkers
Funding
External cooperations
Jochen Springer, Stefan D. Anker
Dr. rer. nat. Nadine Möller, Dr. vet med. Kai Hartmann, Anika Tschirner
self funded
Prof. Dr. rer. nat. Josep M. Argiles
The xanthine oxidase (XO) has been implicated in
several major diseases, such as ischemia-reperfusion
injury like stroke, chronic heart failure, vascular diseases like hypertension and chronic inflammatory disorders like gout. Uric acid produced by XO has been
shown to have predictive value for mortality in cardiac
cachexia. The by-product of uric acid production by
XO are reactive oxygen species (ROS) that not only
alter peripheral blood flow and hence further promote
oxidative stress, but also induce the expression of proteins involved in the ubiquitin-proteasome pathway,
which is considered the most important pathway in
cachexia-associated loss of protein. Uric acid itself is
30
considered a “danger signal” for the immune system
leading to an enhanced production of pro-inflammatory cytokines, which in turn contribute to the wasting
process. Tumor-bearing animals showed a 52-fold
increase in XO-generated ROS, assessed by electron
spin resonance spectroscopy. However, the protein
expression of the enzyme was unchanged. Inhibition of
XO with allopüurinol or oxypurinol did not only reduce
the amount of ROS generated, but also improved survival as well as cardiac function and reduced wasting
of fat and lean mass. Furthermore a down-regulation
of mRNA expression of key proteins of the ubiquitin
proteasome pathway was seen in muscle.
Stefan Anker – Applied Cachexia Research
Lainscak M, Keber I, Anker SD (2006): Body composition
changes in patients with systolic heart failure treated with beta
blockers: a pilot study. Int J Cardiol 106:319-322
Jankowska EA, Biel B, Majda J, Szklarska A, Lopuszanska
M, Medras M, Anker SD, Banasiak W, Poole-Wilson PA,
Ponikowski P. (2006): Anabolic deficiency in men with chronic
heart failure: prevalence and detrimental impact on survival.
Circulation 114:1829-37
Genth-Zotz S, von Haehling S, Bolger AP, Kalra PR, Wensel R, Coats AJ, Volk HD, Anker SD. (2006): The anti-CD14
antibody IC14 suppresses ex vivo endotoxin stimulated tumor
necrosis factor-alpha in patients with chronic heart failure.
Eur J Heart Fail 8:366-372
Köhler F, Nettlau H, Schweizer T, Waller T, Anker SD (2006):
The research project of the german federal ministry of economics and technology: ‘partnership for the heart’ – a new
approach in telemedicine. Dis Manage Health Outcomes
14:1-4
Kalantar-Zadeh K, Kuwae N, Wu DY, Shantouf RS, Fouque D,
Anker SD, Block G, Kopple JD (2006): Associations of body
fat and its changes over time with quality of life and prospective mortality in hemodialysis patients. Am J Clin Nutr
83:202-210
John M, Lange A, Hoernig S, Witt C, Anker SD (2006):The
prevalence of anemia in chronic obstructive pulmonary
disease: comparison to other chronic diseases. Int J Cardiol
111:365-370
Publications 2006 - 2010
Levy WC, Mozaffarian D, Linker DT, Sutradhar SC, Anker SD,
Cropp AB, Anand I, Maggioni A, Burton P, Sullivan MD, Pitt
B, Poole-Wilson PA, Mann DL, Packer M (2006): The Seattle
Heart Failure Model: prediction of survival in heart failure.
Circulation 113:1424-1433
Komajda M, Anker SD, Charlesworth A, Okonko D, Metra M,
Di Lenarda A, Remme W, Moullet C, Swedberg K, Cleland
JG, Poole-Wilson PA (2006): The impact of new onset anaemia on morbidity and mortality in chronic heart failure: results
from COMET. Eur Heart J 27:1440-1446
Piepoli MF, Kaczmarek A, Francis DP, Davies LC, Rauchhaus M, Jankowska EA, Anker SD, Capucci A, Banasiak W,
Ponikowski P (2006): Reduced peripheral skeletal muscle
mass and abnormal reflex physiology in chronic heart failure.
Circulation 114:126-134
Anker SD, Clark AL, Winkler R, Zugck C, Cicoira M,
Ponikowski P, Davos CH, Banasiak W, Zardini P, Haass M,
Senges J, Coats AJ, Poole-Wilson PA, Pitt B (2006): Statin
use and survival in patients with chronic heart failure - results
from two observational studies with 5200 patients. Int J
Cardiol 112:234-42
Kennedy LM, Dickstein K, Anker SD, James M, Cook TJ,
Kristianson K, Willenheimer R (2006): Weight-change as a
prognostic marker in 12550 patients following acute myocardial infarction or with stable coronary artery disease. Eur
Heart J 27:2755-62
Krum H, Bailey M, Meyer W, Verkenne P, Dargie H, Lechat P,
Anker S (2007): Impact of statin therapy on clinical outcomes
in chronic heart failure patients according to beta-blocker
use: results of CIBIS II. Cardiology 108:28-34
Koehler F, Doehner W, Hoernig S, Witt C, Anker SD, John M
(2007): Anorexia in chronic obstructive pulmonary disease Association to cachexia and hormonal derangement. Int J
Cardiol 119:83-9
Akashi YJ, Kida K, Suzuki K, Inoue K, Kawasaki K, Yamauchi
M, Musha H, Anker SD (2007): The significance of (123)
I-BMIPP delayed scintigraphic imaging in cardiac patients. Int
J Cardiol 117:145-51
Habedank D, Ewert R, Hummel M, Wensel R, Hetzer R, Anker
SD (2007): Changes in exercise capacity, ventilation, and
body weight following heart transplantation. Eur J Heart Fail
9:310-6
Kennedy LM, Anker SD, Dickstein K, Willenheimer R (2007):
Impact of neurohormonal blockade on association between
body mass index and mortality. Int J Cardiol 119:33-40
von Haehling S, Doehner W, Anker SD (2007): Nutrition,
metabolism, and the complex pathophysiology of cachexia in
chronic heart failure. Cardiovasc Res 73:298-309
31
Stefan Anker – Applied Cachexia Research
Ponikowski P*, Anker SD*, Szachniewicz J, Okonko D,
Ledwidge M, Zymlinski R, Ryan E, Wasserman SM, Baker N,
Rosser D, Rosen SD, Poole-Wilson PA, Banasiak W, Coats
AJ, McDonald K (2007): Effect of darbepoetin alfa on exercise
tolerance in anemic patients with symptomatic chronic heart
failure: a randomized, double-blind, placebo-controlled trial. J
Am Coll Cardiol 49:753-62
Kohler F, Schieber M, Lucke S, Heinze P, Henke S, Matthesius G, Pferdt T, Wegertseder D, Stoll M, Anker SD (2007):
[“Partnership for the Heart”--development and testing of
a new remote patient monitoring system]. Dtsch Med
Wochenschr 32:458-60
Doehner W, Bunck AC, Rauchhaus M, von Haehling S,
Brunkhorst FM, Cicoira M, Tschope C, Ponikowski P, Claus
RA, Anker SD (2007): Secretory sphingomyelinase is upregulated in chronic heart failure: a second messenger system
of immune activation relates to body composition, muscular
functional capacity, and peripheral blood flow. Eur Heart J
28:821-8
Földes G, von Haehling S, Okonko DO, Jankowska EA,
Poole-Wilson PA, Anker SD (2007): Fluvastatin reduces
increased blood monocyte Toll-like receptor 4 expression in
whole blood from patients with chronic heart failure. Int J
Cardiol 124:80-5
Westermann D, Rutschow S, Jager S, Linderer A, Anker S,
Riad A, Unger T, Schultheiss HP, Pauschinger M, Tschope
C (2007): Contributions of inflammation and cardiac matrix
metalloproteinase activity to cardiac failure in diabetic cardiomyopathy: the role of Angiotensin type 1 receptor antagonism.
Diabetes 56:641-6
Thum T, Hoeber S, Froese S, Klink I, Stichtenoth DO, Galuppo
P, Jakob M, Tsikas D, Anker SD, Poole-Wilson PA, Borlak J,
Ertl G, Bauersachs J (2007): Age-dependent impairment of
endothelial progenitor cells is corrected by growth-hormonemediated increase of insulin-like growth-factor-1. Circ Res
100:434-43
Clark AL, Knosalla C, Birks E, Loebe M, Davos CH, Tsang S,
Negassa A, Yacoub M, Hetzer R, Coats AJ, Anker SD (2007):
Heart transplantation in heart failure: The prognostic importance of body mass index at time of surgery and subsequent
weight changes. Eur J Heart Fail 9:839-44
32
Mozaffarian D, Anker SD, Anand I, Linker DT, Sullivan MD,
Cleland JG, Carson PE, Maggioni AP, Mann DL, Pitt B, PooleWilson PA, Levy WC (2007): Prediction of mode of death in
heart failure: the Seattle Heart Failure Model. Circulation
116:392-8
Niethammer M, Sieber M, von Haehling S, Anker SD, Munzel
T, Horstick G, Genth-Zotz S (2007): Inflammatory pathways in
patients with heart failure and preserved ejection fraction. Int
J Cardiol Jul 19; [Epub ahead of print]
Jankowska EA, Witkowski T, Ponikowska B, Reczuch K,
Borodulin-Nadzieja L, Anker SD, Piepoli MF, Banasiak W,
Ponikowski P (2007): Excessive ventilation during early phase
of exercise: A new predictor of poor long-term outcome in
patients with chronic heart failure. Eur J Heart Fail 9:102431
Kempf T, von Haehling S, Peter T, Allhoff T, Cicoira M,
Doehner W, Ponikowski P, Filippatos GS, Rozentryt P, Drexler
H, Anker SD, Wollert KC (2007): Prognostic utility of growth
differentiation factor-15 in patients with chronic heart failure. J
Am Coll Cardiol 50:1054-60
Sandek S, Bauditz J, Swidsinski A, Buhner S, Weber-Eibel
J, von Haehling S, Schroedl W, Karhausen T, Doehner W,
Rauchhaus M, Poole-Wilson P, Volk HD, Lochs H, Anker SD
(2007): Altered intestinal function in patients with chronic
heart failure. J Am Coll Cardiol 50:1561-9
von Haehling S, Jankowska EA, Morgenthaler NG, Vassanelli
C, Zanolla L, Rozentryt P, Filippatos GS, Doehner W, Koehler
F, Papassotiriou J, Kremastinos DT, Banasiak W, Struck J,
Ponikowski P, Bergmann A, Anker SD (2007): Comparison
of mid-regional proANP with N-terminal proBNP in predicting
survival in patients with chronic heart failure. J Am Coll Cardiol
50:1973-80
Springer J, Groneberg DA, Dinh QT, Quarcoo D, Hamelmann
E, Braun-Dullaeus RC, Geppetti P, Anker SD, Fischer A
(2007): Neurokinin-1 receptor activation induces reactive oxygen species and epithelial damage in allergic airway inflammation. Clin Exp Allergy 37:1788-97
Koehler F, Doehner W, Anker SD, John M (2007): Anorexia
in chronic obstructive pulmonary disease - Association to
cachexia and hormonal derangement. Int J Cardiol. [Epub
ahead of print]
Stefan Anker – Applied Cachexia Research
Metra M, Ponikowski P, Dickstein K, McMurray JJ, Gavazzi A,
Bergh CH, Fraser AG, Jaarsma T, Pitsis A, Mohacsi P, Böhm
M, Anker S, Dargie H, Brutsaert D, Komajda M; Heart Failure
Association of the European Society of Cardiology. (2007):
Advanced chronic heart failure: A position statement from the
Study Group on Advanced Heart Failure of the Heart Failure
Association of the European Society of Cardiology. Eur J
Heart Fail 9:684-94
Gazzieri D, Trevisani M, Springer J, Harrison S, Cottrell GS,
Andre E, Nicoletti P, Massi D, Zecchi S, Nosi D, Santucci M,
Gerard NP, Lucattelli M, Lungarella G, Fischer A, Grady EF,
Bunnett NW, Geppetti P (2007): Substance P released by
TRPV1-expressing neurons produces reactive oxygen species that mediate ethanol-induced gastric injury. Free Radic
Biol Med 15;43(4):581-9
Okonko DO, Grzeslo A, Witkowski T, Mandal AK, Slater
RM, Roughton M, Foldes G, Thum T, Majda J, Banasiak W,
Missouris CG, Poole-Wilson PA, Anker SD*, Ponikowski P*
(2008): Effect of intravenous iron sucrose on exercise tolerance in anemic and nonanemic patients with symptomatic
chronic heart failure and iron deficiency FERRIC-HF: a randomized, controlled, observer-blinded trial. J Am Coll Cardiol
51:103-12
Torre-Amione G, Anker SD, Bourge RC, Colucci WS,
Greenberg BH, Hildebrandt P, Keren A, Motro M, Moyé LA,
Otterstad JE, Pratt CM, Ponikowski P, Rouleau JL, Sestier F,
Winkelmann BR, Young JB (2008): Advanced Chronic Heart
Failure CLinical Assessment of Immune Modulation Therapy
Investigators. Results of a non-specific immunomodulation
therapy in chronic heart failure (ACCLAIM trial): a placebocontrolled randomised trial. Lancet 371:228-36
Habedank D, Ewert R, Hetzer R, Anker SD (2008): Reversibility of cachexia after bilateral lung transplantation. Int J
Cardiol. [Epub ahead of print]
Schumann S, Saggu M, Möller N, Anker SD, Lendzian F,
Hildebrandt P, Leimkühler S (2008): The Mechanism of
Assembly and Cofactor Insertion into Rhodobacter capsulatus Xanthine Dehydrogenase. J Biol Chem 283:16602-11
von Haehling S, Schefold JC, Springer J, Anker SD (2008):
The cholesterol paradox revisited: heart failure, systemic
inflammation, and beyond. Heart Fail Clin 4:141-51
Belonje AM, Voors AA, van Gilst WH, Anker SD, Slart RH,
Tio RA, Zijlstra F, van Veldhuisen DJ; HEBE III investigators.
(2008): Effects of erythropoietin after an acute myocardial
infarction: rationale and study design of a prospective, randomized, clinical trial (HEBE III). Am Heart J 155:817-22
Strassburg S, Anker SD, Castaneda TR, Burget L, Perez-Tilve
D, Pfluger PT, Nogueiras R, Halem H, Dong JZ, Culler MD,
Datta R, Tschop MH (2008): Long-term Effects of Ghrelin and
Ghrelin Receptor Agonists on Energy Balance in Rats. Am J
Physiol Endocrinol Metab 295:E78-E84
Filippatos GS, Adamopoulos C, Sui X, Love TE, Pullicino PM,
Lubsen J, Bakris G, Anker SD, Howard G, Kremastinos DT,
Ahmed A (2008): A propensity-matched study of hypertension and increased stroke-related hospitalization in chronic
heart failure. Am J Cardiol 101:1772-6
Sandek A, Rauchhaus M, Anker SD, von Haehling S (2008):
The emerging role of the gut in chronic heart failure. Curr
Opin Clin Nutr Metab Care 11:632-9
Alper AB, Campbell RC, Anker SD, Bakris G, Wahle C, Love
TE, Hamm LL, Mujib M, Ahmed A (2008): A propensitymatched study of low serum potassium and mortality in older
adults with chronic heart failure. Int J Cardiol Aug 6. [Epub
ahead of print]
Evans WJ, Morley JE, Argilés J, Bales C, Baracos V, Guttridge
D, Jatoi A, Kalantar-Zadeh K, Lochs H, Mantovani G, Marks
D, Mitch WE, Muscaritoli M, Najand A, Ponikowski P, Rossi
Fanelli F, Schambelan M, Schols A, Schuster M, Thomas D,
Wolfe R, Anker SD (2008): Cachexia: a new definition. Clin
Nutr 27:793-9
Akashi YJ, Palus S, Datta R, Halem H, Taylor JE, ThoeneReineke C, Dong J, Thum T, Culler MD, Anker SD, Springer
J (2008): No effects of human ghrelin on cardiac function
despite profound effects on body composition in a rat model
of heart failure. Int J Cardiol Aug 22. [Epub ahead of print]
Göhler A, Conrads-Frank A, Worrell SS, Geisler BP, Halpern EF, Dietz R, Anker SD, Gazelle GS, Siebert U (2008):
Decision-analytic evaluation of the clinical effectiveness and
cost-effectiveness of management programmes in chronic
heart failure. Eur J Heart Fail 10:1026-32
33
Stefan Anker – Applied Cachexia Research
Doehner W, Gathercole D, Cicoira M, Krack A, Coats AJ,
Camici PG, Anker SD (2008): Reduced glucose transporter
GLUT4 in skeletal muscle predicts insulin resistance in nondiabetic chronic heart failure patients independently of body
composition. Int J Cardiol Sep 6. [Epub ahead of print]
Reinke S, Karhausen T, Doehner W, Taylor W, Hottenrott K,
Duda GN, Reinke P, Volk HD, Anker SD (2009): The influence
of recovery and training phases on body composition, peripheral vascular function and immune system of professional
soccer players. PLoS One 4:e4910
Pocock SJ, McMurray JJ, Dobson J, Yusuf S, Granger CB,
Michelson EL, Ostergren J, Pfeffer MA, Solomon SD, Anker
SD, Swedberg KB (2008): Weight loss and mortality risk in
patients with chronic heart failure in the candesartan in heart
failure: assessment of reduction in mortality and morbidity
(CHARM) programme. Eur Heart J 29:2641-50
Voors AA, von Haehling S, Anker SD, Hillege HL, Struck J,
Hartmann O, Bergmann A, Squire I, van Veldhuisen DJ, Dickstein K; OPTIMAAL Investigators (2009): C-terminal provasopressin (copeptin) is a strong prognostic marker in patients
with heart failure after an acute myocardial infarction: results
from the OPTIMAAL study. Eur Heart J 30:1187-94
Belonje AM, Westenbrink BD, Voors AA, von Haehling S,
Ponikowski P, Anker SD, van Veldhuisen DJ, Dickstein K
(2009): Erythropoietin levels in heart failure after an acute
myocardial infarction: determinants, prognostic value, and the
effects of captopril versus losartan. Am Heart J 157:91-6.
Anker SD, Voors A, Okonko D, Clark AL, James MK, von
Haehling S, Kjekshus J, Ponikowski P, Dickstein K; OPTIMAAL
Investigators. P (2009): Revalence, incidence, and prognostic
value of anaemia in patients after an acute myocardial infarction: data from the OPTIMAAL trial. Eur Heart J 30:1331-9
Lainscak M, von Haehling S, Anker SD (2009): Natriuretic
peptides and other biomarkers in chronic heart failure: From
BNP, NT-proBNP, and MR-proANP to routine biochemical
markers. Int J Cardiol 32:303-11
von Haehling S, von Bardeleben RS, Kramm T, Thiermann Y,
Niethammer M, Doehner W, Anker SD, Munzel T, Mayer E,
Genth-Zotz S (2009): Inflammation in right ventricular dysfunction due to thromboembolic pulmonary hypertension. Int
J Cardiol May 1. [Epub ahead of print]
Morley JE, Anker SD, Evans WJ (2009): Cachexia and aging:
an update based on the fourth international cachexia meeting.
J Nutr Health Aging 13:47-55
Vaz Pérez A, Doehner W, von Haehling S, Schmidt H, Zimmermann AV, Volk HD, Anker SD, Rauchhaus M (2009):
The relationship between tumor necrosis factor-alpha, brain
natriuretic peptide and atrial natriuretic peptide in patients
with chronic heart failure. Int J Cardiol Jan 18. [Epub ahead
of print]
Ekundayo OJ, Dell’italia LJ, Sanders PW, Arnett D, Aban I,
Love TE, Filippatos G, Anker SD, Lloyd-Jones DM, Bakris
G, Mujib M, Ahmed A (2009): Association between hyperuricemia and incident heart failure among older adults: A
propensity-matched study. Int J Cardiol Feb 5. [Epub ahead
of print]
Palus S, Akashi Y, von Haehling S, Anker SD, Springer J
(2009): The influence of age and sex on disease development
in a novel animal model of cardiac cachexia. Int J Cardiol
133:388-93
34
Jankowska EA, Rozentryt P, Ponikowska B, Hartmann O,
Kustrzycka-Kratochwil D, Reczuch K, Nowak J, BorodulinNadzieja L, Polonski L, Banasiak W, Poole-Wilson PA, Anker
SD, Ponikowski P (2009): Circulating estradiol and mortality in
men with systolic chronic heart failure. JAMA 9;301:1892-901
Kalra PR, Clague JR, Coats AJ, Anker SD, Poole-Wilson PA,
Struthers AD (2009): C type natruiretic peptide production by
the human kidney is blunted in chronic heart failure. Clin Sci
(Lond) May 18.
Jankowska EA, Filippatos G, Ponikowska B, Borodulin-Nadzieja
L, Anker SD, Banasiak W, Poole-Wilson PA, Ponikowski P
(2009): Reduction in circulating testosterone relates to exercise
capacity in men with chronic heart failure. J Card Fail 15:442-50
van Veldhuisen DJ, Cohen-Solal A, Böhm M, Anker SD, Babalis
D, Roughton M, Coats AJ, Poole-Wilson PA, Flather MD;
SENIORS Investigators (2009): Beta-blockade with nebivolol in
elderly heart failure patients with impaired and preserved left ventricular ejection fraction: Data From SENIORS (Study of Effects
of Nebivolol Intervention on Outcomes and Rehospitalization in
Seniors With Heart Failure). J Am Coll Cardiol 53:2150-8
Stefan Anker – Applied Cachexia Research
Anker SD, John M, Laviano A, Filippatos G, Paccagnella
A, Ponikowski P, Schols AW (2009): ESPEN Guidelines on
Parenteral Nutrition: Cardiology. Clin Nutr Jun 8. [Epub
ahead of print]
von Haehling S, Hopkinson NS, Polkey MI, Niethammer M,
Anker SD, Genth-Zotz S. (2009): Elevated TNFalpha production in whole blood in patients with severe COPD: the
potential link to disease severity. Wien Klin Wochenschr
121:303-8
Lainscak M, Hodoscek LM, Düngen HD, Rauchhaus M,
Doehner W, Anker SD, von Haehling S (2009): The burden of
chronic obstructive pulmonary disease in patients hospitalized with heart failure. Wien Klin Wochenschr 121:309-13
Ogino K, Kato M, Furuse Y, Kinugasa Y, Ishida K, Osaki
S, Kinugawa T, Igawa O, Hisatome I, Shigemasa C, Anker
SD, Doehner W (2009): Uric Acid Lowering Treatment with
Benzbromarone in Patients with Heart Failure: a Double-blind
Placebo-controlled Cross-over Preliminary Study. Circ Heart
Fail Nov 20 [Epub ahead of print]
von Haehling S, Schefold JC, Majc Hodoscek L, Doehner
W, Mannaa M, Anker SD, Lainscak M (2009): Anaemia is an
independent predictor of death in patients hospitalized for
acute heart failure. Clin Res Cardiol Nov 17 [Epub ahead of
print]
Anker SD, Comin Colet J, Filippatos G, Willenheimer R,
Dickstein K, Drexler H, Lüscher TF, Bart B, Banasiak W,
Niegowska J, Kirwan BA, Mori C, von Eisenhart Rothe B,
Pocock SJ, Poole-Wilson PA, Ponikowski P; the FAIR-HF
Trial Investigators. (2009): Ferric Carboxymaltose in Patients
with Heart Failure and Iron Deficiency. N Engl J Med
361(25):2436-2448
Anker SD, Colet JC, Filippatos G, Willenheimer R, Dickstein K,
Drexler H, Lüscher TF, Mori C, von Eisenhart Rothe B, Pocock S,
Poole-Wilson PA, Ponikowski P; FAIR-HF committees and investigators. (2009) Rationale and design of Ferinject assessment in
patients with IRon deficiency and chronic Heart Failure (FAIR-HF)
study: a randomized, placebo-controlled study of intravenous
iron supplementation in patients with and without anaemia. Eur J
Heart Fail 11(11):10
Baid-Agrawal S, Frei U, Reinke P, Schindler R, Kopp MA, Martus P, Berg T, Juergensen JS, Anker SD, Doehner W. (2009):
Impaired Insulin Sensitivity as an Underlying Mechanism Linking Hepatitis C and Posttransplant Diabetes Mellitus in Kidney
Recipients. Am J Transplant Nov 20;9(12):2777-2784
Klapholz M, Abraham WT, Ghali JK, Ponikowski P, Anker SD,
Knusel B, Sun Y, Wasserman SM, van Veldhuisen DJ (2009):
The safety and tolerability of darbepoetin alfa in patients with
anaemia and symptomatic heart failure. Eur J Heart Fail
11(11):1071-7
Habedank D, Kung T, Karhausen T, von Haehling S, Doehner
W, Schefold JC, Hasper D, Reinke S, Anker SD, Reinke P
(2009): Exercise capacity and body composition in livingdonor renal transplant recipients over time. Nephrol Dial
Transplant 24(12):3854-60
Szabo T, von Haehling S, Habedank D, Rauchhaus M, Lainscak M, Sandek A, Schefold J, Anker SD, Doehner W (2009):
Usefulness of minimal modelling to assess impaired insulin
sensitivity in patients with chronic heart failure. Int J Cardiol
4. [Epub ahead of print]
Mehrhof F, Löffler M, Gelbrich G, Ozcelik C, Posch M,
Hense HW, Keil U, Scheffold T, Schunkert H, Angermann C,
Ertl G, Jahns R, Pieske B, Wachter R, Edelmann F, Wollert
KC, Maisch B, Pankuweit S, Erbel R, Neumann T, Herzog
W, Katus H, Müller-Tasch T, Zugck C, Düngen HD, RegitzZagrosek V, Lehmkuhl E, Störk S, Siebert U, Wasem J, Neumann A, Göhler A, Anker SD, Köhler F, Möckel M, Osterziel
KJ, Dietz R, Rauchhaus M; on Behalf of the Competence Network Heart Failure (2009): A network against failing heartsIntroducing the German “Competence Network Heart Failure”
Int J Cardiol Aug 11. [Epub ahead of print]
von Haehling S, Schefold JC, Jankowska E, Doehner W, Springer
J, Strohschein K, Genth-Zotz S, Volk HD, Poole-Wilson P, Anker
SD (2009): Leukocyte redistribution: effects of beta blockers in
patients with chronic heart failure. PLoS One 4(7):e6411
McMurray JJ, Anand IS, Diaz R, Maggioni AP, O’Connor
C, Pfeffer MA, Polu KR, Solomon SD, Sun Y, Swedberg K,
Tendera M, van Veldhuisen DJ, Wasserman SM, Young JB;
RED-HF Committees and Investigators. (2009): Design of the
Reduction of Events with Darbepoetin alfa in Heart Failure
(RED-HF): a Phase III, anaemia correction, morbidity-mortality
trial. Eur J Heart Fail 11(8):795-801
35
Stefan Anker – Applied Cachexia Research
Spintzik J, Springer J, Westermann B (2009): Morphological
and histological organization of the pyriform appendage of the
tetrabranchiate Nautilus pompilius (Cephalopoda, Mollusca).
J Morphol 270(4):459-68
Gratze P, Boschmann M, Dechend R, Qadri F, Malchow J,
Graeske S, Engeli S, Janke J, Springer J, Contrepas A, Plehm
R, Klaus S, Nguyen G, Luft FC, Muller DN (2009): Energy
metabolism in human renin-gene transgenic rats: does renin
contribute to obesity? Hypertension 53(3):516-23
von Haehling S, Schefold JC, Hodoscek LM, Doehner W,
Mannaa M, Anker SD, Lainscak M (2010) Anaemia is an independent predictor of death in patients hospitalized for acute
heart failure. Clin Res Cardiol 99(2):107-13
Ronco C, McCullough P, Anker SD, Anand I, Aspromonte N,
Bagshaw SM, Bellomo R, Berl T, Bobek I, Cruz DN, Daliento
L, Davenport A, Haapio M, Hillege H, House AA, Katz N, Maisel A, Mankad S, Zanco P, Mebazaa A, Palazzuoli A, Ronco
F, Shaw A, Sheinfeld G, Soni S, Vescovo G, Zamperetti N,
Ponikowski P; (2010) Acute Dialysis Quality Initiative (ADQI)
consensus group. Cardio-renal syndromes: report from the
consensus conference of the acute dialysis quality initiative.
Eur Heart J 31(6):703-11
Muscaritoli M, Anker SD, Argilés J, Aversa Z, Bauer JM, Biolo
G, Boirie Y, Bosaeus I, Cederholm T, Costelli P, Fearon KC,
Laviano A, Maggio M, Rossi Fanelli F, Schneider SM, Schols
A, Sieber CC. (2010): Consensus definition of sarcopenia,
cachexia and pre-cachexia: joint document elaborated by
Special Interest Groups (SIG) “cachexia-anorexia in chronic
wasting diseases” and “nutrition in geriatrics”. Clin Nutr
29(2):154-9
Poldermans D, Bax JJ, Boersma E, De Hert S, Eeckhout E,
Fowkes G, Gorenek B, Hennerici MG, Iung B, Kelm M, Kjeldsen KP, Kristensen SD, Lopez-Sendon J, Pelosi P, Philippe F,
Pierard L, Ponikowski P, Schmid JP, Sellevold OF, Sicari R,
Van den Berghe G, Vermassen F, Hoeks SE, Vanhorebeek I,
Vahanian A, Auricchio A, Bax JJ, Ceconi C, Dean V, Filippatos G, Funck-Brentano C, Hobbs R, Kearn P, McDonag T,
McGregor K, Popescu BA, Reiner Z, Sechtem U, Sirnes PA,
Tendera M, Vardas P, Widimsky P, De Caterina R, Agewall
S, Al Attar N, Andreotti F, Anker SD, Baron-Esquivias G,
Berkenboom G, Chapoutot L, Cifkova R, Faggiano P, Gibbs
S, Hansen HS, Iserin L, Israel CW, Kornowski R, Eizagaechevarria NM, Pepi M, Piepoli M, Priebe HJ, Scherer M, Stepinska
J, Taggart D, Tubaro M; Task Force for Preoperative Cardiac
Risk Assessment and Perioperative Cardiac Management
in Non-cardiac Surgery of European Society of Cardiology
(ESC); European Society of Anaesthesiology (ESA). (2010):
Guidelines for pre-operative cardiac risk assessment and
perioperative cardiac management in non-cardiac surgery:
the Task Force for Preoperative Cardiac Risk Assessment and
Perioperative Cardiac Management in Non-cardiac Surgery
of the European Society of Cardiology (ESC) and endorsed
by the European Society of Anaesthesiology (ESA). Eur J
Anaesthesiol. 27(2):92-137.
Bagshaw SM, Cruz DN, Aspromonte N, Daliento L, Ronco F,
Sheinfeld G, Anker SD, Anand I, Bellomo R, Berl T, Bobek I,
Davenport A, Haapio M, Hillege H, House A, Katz N, Maisel
A, Mankad S, McCullough P, Mebazaa A, Palazzuoli A,
Ponikowski P, Shaw A, Soni S, Vescovo G, Zamperetti N,
Zanco P, Ronco C (2010): Acute Dialysis Quality Initiative
Consensus Group.Epidemiology of cardio-renal syndromes:
workgroup statements from the 7th ADQI Consensus Conference. Nephrol Dial Transplant 25(5):1406-16. Epub 2010 Feb
25
von Haehling S, Filippatos GS, Papassotiriou J, Cicoira M,
Jankowska EA, Doehner W, Rozentryt P, Vassanelli C, Struck
J, Banasiak W, Ponikowski P, Kremastinos D, Bergmann
A, Morgenthaler NG, Anker SD (2010): Mid-regional proadrenomedullin as a novel predictor of mortality in patients
with chronic heart failure. Eur J Heart Fail 12(5):484-91
36
Stefan Anker – Applied Cachexia Research
House AA, Anand I, Bellomo R, Cruz D, Bobek I, Anker SD,
Aspromonte N, Bagshaw S, Berl T, Daliento L, Davenport
A, Haapio M, Hillege H, McCullough P, Katz N, Maisel A,
Mankad S, Zanco P, Mebazaa A, Palazzuoli A, Ronco F, Shaw
A, Sheinfeld G, Soni S, Vescovo G, Zamperetti N, Ponikowski
P, Ronco C; Acute Dialysis Quality Initiative Consensus
Group. (2010): Definition and classification of Cardio-Renal
Syndromes: workgroup statements from the 7th ADQI Consensus Conference. Nephrol Dial Transplant 25(5):1416-20
Ronco C, McCullough PA, Anker SD, Anand I, Aspromonte N,
Bagshaw SM, Bellomo R, Berl T, Bobek I, Cruz DN, Daliento
L, Davenport A, Haapio M, Hillege H, House A, Katz NM, Maisel A, Mankad S, Zanco P, Mebazaa A, Palazzuoli A, Ronco
F, Shaw A, Sheinfeld G, Soni S, Vescovo G, Zamperetti N,
Ponikowski P; Acute Dialysis Quality Initiative (ADQI) consensus group. (2010): Cardiorenal syndromes: an executive summary from the consensus conference of the Acute Dialysis
Quality Initiative (ADQI). Contrib Nephrol 65:54-67.
Strassburg S, Pfluger PT, Chaudhary N, Tso P, Tschöp MH,
Anker SD, Nogueiras R, Perez-Tilve D (2010): Action Profile
of the Antiobesity Drug Candidate Oleoyl-Estrone in Rats.
Obesity (Silver Spring). Mar 25.
Maisel A, Mueller C, Nowak R, Peacock WF, Landsberg
JW, Ponikowski P, Mockel M, Hogan C, Wu AH, Richards
M, Clopton P, Filippatos GS, Di Somma S, Anand I, Ng L,
Daniels LB, Neath SX, Christenson R, Potocki M, McCord J,
Terracciano G, Kremastinos D, Hartmann O, von Haehling S,
Bergmann A, Morgenthaler NG, Anker SD (2010): Mid-region
pro-hormone markers for diagnosis and prognosis in acute
dyspnea: results from the BACH (Biomarkers in Acute Heart
Failure) trial. J Am Coll Cardiol 11;55(19):2062-76
Gheorghiade M, Follath F, Ponikowski P, Barsuk JH, Blair JE,
Cleland JG, Dickstein K, Drazner MH, Fonarow GC, Jaarsma
T, Jondeau G, Sendon JL, Mebazaa A, Metra M, Nieminen
M, Pang PS, Seferovic P, Stevenson LW, van Veldhuisen DJ,
Zannad F, Anker SD, Rhodes A, McMurray JJ, Filippatos G
(2010): Assessing and grading congestion in acute heart
failure: a scientific statement from the acute heart failure committee of the heart failure association of the European society
of cardiology and endorsed by the European society of intensive care medicine. Eur J Heart Fail 12(5):423-33.
Wu AH, Pitt B, Anker SD, Vincent J, Mujib M, Ahmed A
(2010): Association of obesity and survival in systolic heart
failure after acute myocardial infarction: potential confounding
by age. Eur J Heart Fail 12(6):566-73
McCullough PA, Haapio M, Mankad S, Zamperetti N, Massie
B, Bellomo R, Berl T, Anker SD, Anand I, Aspromonte N, Bagshaw SM, Bobek I, Cruz DN, Daliento L, Davenport A, Hillege
H, House AA, Katz N, Maisel A, Mebazaa A, Palazzuoli A,
Ponikowski P, Ronco F, Shaw A, Sheinfeld G, Soni S, Vescovo
G, Zanco P, Ronco C, Berl T; Acute Dialysis Quality Initiative
(ADQI) Consensus Group. (2010): Prevention of cardio-renal
syndromes: workgroup statements from the 7th ADQI Consensus Conference. Nephrol Dial Transplant 25(6):1777-84
Jankowska EA, Rozentryt P, Witkowska A, Nowak J, Hartmann O, Ponikowska B, Borodulin-Nadzieja L, Banasiak W,
Polonski L, Filippatos G, McMurray JJ, Anker SD, Ponikowski
P (2010): Iron deficiency: an ominous sign in patients with
systolic chronic heart failure. Eur Heart J 31(15):1872-80
de Boer RA, Doehner W, van der Horst IC, Anker SD, Babalis
D, Roughton M, Coats AJ, Flather MD, van Veldhuisen DJ;
SENIORS Investigators. (2010): Influence of diabetes mellitus
and hyperglycemia on prognosis in patients > or =70 years
old with heart failure and effects of nebivolol (data from the
Study of Effects of Nebivolol Intervention on Outcomes and
Rehospitalization in seniors with heart failure [SENIORS]). Am
J Cardiol 1;106(1):78-86.e1
Morley JE, Argiles JM, Evans WJ, Bhasin S, Cella D, Deutz
NE, Doehner W, Fearon KC, Ferrucci L, Hellerstein MK,
Kalantar-Zadeh K, Lochs H, MacDonald N, Mulligan K,
Muscaritoli M, Ponikowski P, Posthauer ME, Rossi Fanelli F,
Schambelan M, Schols AM, Schuster MW, Anker SD; Society
for Sarcopenia, Cachexia, and Wasting Disease. (2010): Nutritional recommendations for the management of sarcopenia. J
Am Med Dir Assoc 11(6):391-6
37
Stefan Anker – Applied Cachexia Research
Habedank D, Ewert R, Hummel M, Dandel M, Habedank F,
Knosalla C, Lehmkuhl HB, Anker SD, Hetzer R (2010): The
effects of bilateral lung transplantation on ventilatory efficiency, oxygen uptake and the right heart: a two-yr follow-up.
Clin Transplant 18 [Epub ahead of print]
Reviews & book chapters ( 2006-2010 )
Strassburg S, Anker SD (2006): Metabolic and immunologic
derangements in cardiac cachexia: where to from here?
Heart Fail Rev 11:57-64
Foldes G, von Haehling S, Anker SD (2006): Toll-like receptor
modulation in cardiovascular disease: a target for intervention? Expert Opin Investig Drugs 15:857-871
Lainscak M, Podbregar M, Anker SD (2007): How does
cachexia influence survival in cancer, heart failure and
other chronic diseases? Curr Opin Support Palliat Care
2007;1:299-305
Kalantar-Zadeh K, Horwich TB, Oreopoulos A, Kovesdy CP,
Younessi H, Anker SD, Morley JE (2007): Risk factor paradox in wasting diseases. Curr Opin Clin Nutr Metab Care
10:433-42
Földes G, von Haehling S, Jankowska EA, Anker SD (2007):
Targeting the toll-system in cardiovascular sciences. Recent
Pat Inflamm Allergy Drug Discov 1(1):57-67
von Haehling S, Doehner W, Anker SD (2007): Nutrition,
metabolism, and the complex pathophysiology of cachexia in
chronic heart failure. Cardiovasc Res 73:298-309
Springer J, von Haehling S, Anker SD. (2006): The need for
a standardized definition for cachexia in chronic illness. Nat
Clin Pract Endocrinol Metab 2:416-7
Lainscak M, von Haehling S, Springer J, Anker SD (2007):
Biomarkers for chronic heart failure. Heart Fail Monit 5:77-82
von Haehling S, Doehner W, Anker SD (2006): Obesity and
the heart a weighty issue. J Am Coll Cardiol 6;47(11):2274-6
Akashi YJ, Springer J, Lainscak M, Anker SD (2007): Atrial natriuretic
peptide and related peptides. Clin Chem Lab Med 45:1259-67
Anker SD, John M, Pedersen PU, Raguso C, Cicoira M,
Dardai E, Laviano A, Ponikowski P, Schols AM; DGEM (German Society for Nutritional Medicine); Becker HF, Bohm M,
Brunkhorst FM, Vogelmeier C (2006): ESPEN guidelines on
enteral nutrition: cardiology and pulmonology. Clin Nutr
25:311-318
Schünemann M, Anker SD, Rauchhaus M (2008): Cancer fatigue
syndrome reflects clinically non-overt heart failure: an approach
towards onco-cardiology. Nat Clin Pract Oncol 5(11):632-3
Springer J, Filippatos G, Akashi YJ, Anker SD (2006): Prognosis and therapy approaches of cardiac cachexia. Curr Opin
Cardiol 21:229-233
Kalantar-Zadeh K, Abbott KC, Kronenberg F, Anker SD,
Horwich TB, Fonarow GC (2006): Epidemiology of dialysis
patients and heart failure patients. Semin Nephrol 26:118133
Jankowska EA, Ponikowski P, Piepoli MF, Banasiak W, Anker
SD, Poole-Wilson PA (2006): Autonomic imbalance and
immune activation in chronic heart failure - pathophysiological
links. Cardiovasc Res 70:434-445
von Haehling S, Doehner W, Anker SD (2006): Ernst Scher­
ing Res Found Workshop 55:155-168
38
Maisel A, Mueller C, Adams K Jr, Anker SD, Aspromonte
N, Cleland JG, Cohen-Solal A, Dahlstrom U, DeMaria A, Di
Somma S, Filippatos GS, Fonarow GC, Jourdain P, Komajda
M, Liu PP, McDonagh T, McDonald K, Mebazaa A, Nieminen
MS, Peacock WF, Tubaro M, Valle R, Vanderhyden M, Yancy
CW, Zannad F, Braunwald E (2008): State of the art: using
natriuretic peptide levels in clinical practice. Eur J Heart Fail
10:824-39
Sandek A, Rauchhaus M, Anker SD, von Haehling S (2008):
The emerging role of the gut in chronic heart failure. Curr
Opin Clin Nutr Metab Care 11:632-9.
Kalantar-Zadeh K, Anker SD, Horwich TB, Fonarow GC.
(2008): Nutritional and anti-inflammatory interventions in
chronic heart failure. Am J Cardiol 101:89E-103E
Lainscak M, Dagres N, Filippatos GS, Anker SD, Kremastinos
DT (2008): Atrial fibrillation in chronic non-cardiac disease:
Where do we stand? Int J Cardiol 128:311-5
Stefan Anker – Applied Cachexia Research
Farmakis D, Filippatos G, Lainscak M, Parissis JT, Anker SD,
Kremastinos DT (2008): Anticoagulants, antiplatelets, and
statins in heart failure. Cardiol Clin 26:49-58, vi
von Haehling S, Lainscak M, Springer J, Anker SD (2008): Cardiac
cachexia: A systematic overview. Pharmacol Ther 121:227-52
Doehner W, von Haehling S, Anker SD, Lainscak M (2009):
Neurohormonal activation and inflammation in chronic cardiopulmonary disease: a brief systematic review. Wien Klin
Wochenschr 121:293-6
Jaarsma T, Beattie JM, Ryder M, Rutten FH, McDonagh T, Mohacsi
P, Murray SA, Grodzicki T, Bergh I, Metra M, Ekman I, Angermann
C, Leventhal M, Pitsis A, Anker SD, Gavazzi A, Ponikowski P, Dickstein K, Delacretaz E, Blue L, Strasser F, McMurray J;Advanced
Heart Failure Study Group of the HFA of the ESC. (2009): Palliative
care in heart failure: a position statement from the palliative care
workshop of the Heart Failure Association of the European Society
of Cardiology. Eur J Heart Fail 11:433-43
Lainscak M, Anker SD (2009): Prognostic factors in chronic
heart failure. A review of serum biomarkers, metabolic
changes, symptoms, and scoring systems. Herz 34:141-7
Heymans S, Hirsch E, Anker SD, Aukrust P, Balligand JL,
Cohen-Tervaert JW, Drexler H, Filippatos G, Felix SB, Gullestad L, Hilfiker-Kleiner D, Janssens S, Latini R, Neubauer G,
Paulus WJ, Pieske B, Ponikowski P, Schroen B, Schultheiss
HP, Tschöpe C, Van Bilsen M, Zannad F, McMurray J, Shah
AM (2009): Inflammation as a therapeutic target in heart
failure? A scientific statement from the Translational Research
Committee of the Heart Failure Association of the European
Society of Cardiology. Eur J Heart Fail 11:119-29
Sandek A, Doehner W, Anker SD, von Haehling S (2009):
Nutrition in heart failure: an update. Curr Opin Clin Nutr
Metab Care 12:384-91
Szabó T, Felger D, von Haehling S, Lainscak M, Anker SD,
Doehner W (2009): Overview of emerging pharmacotherapy
in chronic heart failure. Expert Opin Pharmacother
10(13):2055-74
von Haehling S, Schefold JC, Lainscak M, Doehner W, Anker
SD (2009): Inflammatory biomarkers in heart failure revisited: much more than innocent bystanders. Heart Fail Clin
5(4):549-60
Soukoulis V, Dihu JB, Sole M, Anker SD, Cleland J, Fonarow
GC, Metra M, Pasini E, Strzelczyk T, Taegtmeyer H, Gheorghiade M (2009): Micronutrient deficiencies an unmet need in
heart failure. J Am Coll Cardiol 54(18):1660-73
Riegel B, Moser DK, Anker SD, Appel LJ, Dunbar SB, Grady
KL, Gurvitz MZ, Havranek EP, Lee CS, Lindenfeld J, Peterson
PN, Pressler SJ, Schocken DD, Whellan DJ; American Heart
Association Council on Cardiovascular Nursing; American
Heart Association Council on Cardiovascular Nursing; American Heart Association Council on Clinical Cardiology; American Heart Association Council on Nutrition, Physical Activity,
and Metabolism; American Heart Association Interdisciplinary
Council on Quality of Care and Outcomes Research. (2009):
State of the science: promoting self-care in persons with
heart failure: a scientific statement from the American Heart
Association. Circulation 120(12):1141-63
Kung T, Springer J, Doehner W, Anker SD, von Haehling S
(2010): Novel treatment approaches to cachexia and sarcopenia: highlights from the 5th Cachexia Conference. Expert
Opin Investig Drugs 19(4):579-85
von Haehling S, Stepney R, Anker SD (2010): Advances in
understanding and treating cardiac cachexia: Highlights from
the 5th Cachexia Conference. Int J Cardiol Jun 18. [Epub
ahead of print]
Patents ( 2006-2010 )
DE102004047955A1 (2006)
WO002006058748A1 (2006)
EP000001669074A1 (2006)
EP000001749531A1 (2006)
WO002007014586A1 (2007)
DE102005046539A1 (2007)
WO002007039263A2 (2007)
EP000001818061A1 (2007)
US020070197485A1 (2007)
EP000001863492A1 (2007)
EP000001915159A1 (2008)
US020080139521A1 (2008)
WO002008067833A1 (2008)
WO002008067831A2 (2008)
EP000001937360A1 (2008)
US000007417038B1 (2008)
DE102007010834A1 (2008)
WO002008106938A2 (2008)
US020080269179A1 (2008)
WO002008135571A1 (2008)
DE102007022367A1 (2008)
WO002008106938A3 (2008)
US020090023639A1 (2009)
WO002009056256A1 (2009)
WO002009071405A1 (2009)
US020090197851A1 (2009)
EP000002099474A1 (2009)
US020090248101A1 (2009)
EP000002122363A2 (2009)
39
Stefan Anker – Applied Cachexia Research
General information
Third party funding ( 2006-2009 )
Project leader
Anker S.D.
Anker S.D.
Springer J.
Anker S.D.
Anker S.D.
Springer J.
Anker S.D.
Springer J.
Springer J.
Döhner W.
Anker S.D.
Anker S.D.
Springer J.
Anker S.D.
Döhner W.
Anker S.D.
Anker S.D.
von Haehling S.
Döhner W.
Anker S.D.
von Haehling S.
Döhner W.
Springer J.
Döhner W.
40
Project title
WARCEF study coordination – Germany
& Poland
Warcef study coordination – Netherlands
Heart failure cachexia therapy
Sponsor
NIH USA
Period
2004-2011
NIH USA
2005-2011
Ipsen Pharma, France
2005-2007
Scholarship for biomarker research
Developing a heart failure in old animals
BRAHMS AG
Sonnenfeld-Stiftung Berlin
2006-2007
2006-2007
Development of an in-vitro myocardial
in faction model based on vital heart
sections
To study the effects of a GLP-1 agonist
on heart in rats with chronic heart failure
Investigations on insulin sensitivity in
patients with heart failure and with
COPD with/without cachexia
Scholarship for biomarker research
Cancer cachexia therapy
Erna-Graff- Stiftung
2006-2007
Curatis Pharma GmbH
2007
DFG
2007-2010
BRAHMS AG
Myotec Therapeutics
2008-2009
2008-2009
DFG – BMBF
2008-2013
EU FP 7
2009-2013
EU FP 7
2009-2014
BMBF
2009-2010
Metabolic research on mechanisms of
cachexia in stroke and CHF – Substudy
of the IFB Stroke Center Berlin
Studies investigating comorbidities
aggravating heart failure (SICA-HF):
EU co-ordination (SDA) and 4 subprojects
Biomarker research in the Biostat-CHF
project
Investigations on weight loss and metabolic changes in a stroke animal model
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Head of the group
Priv.-Doz. Dr. med. Ivo Buschmann, MD, F.E.S.C.
Curriculum Vitae: Ivo Buschmann studied medicine at the University of Hamburg.
After 2 years of clinical medicine in cardiology he received a stipend by the MaxPlanck-Society and joined the group of Wolfgang Schaper at the Max-Planck-Institute
for Physiological & Clinical Research Bad Nauheim, which focused on the biology
of the collateral circulation.
In the following years Ivo Buschmann held the position of group leader and published several original papers in
the field of therapeutic Arteriogenesis. One of the experimental trials was the first to show that Arteriogenesis
may be stimulated therapeutically in the brain (PNAS 2004).
In 2000, Ivo Buschmann was granted a VolkswagenStiftung Excellency Programme-Funding for altogether
6 years and had to change – due to the program - location to the University of Freiburg, Dept. of Internal
Medicine (Cardiology). In 2004, Ivo Buschmann and his research group moved to the Charité Berlin, where
they are now located at the Center for Cardiovascular Research at the Campus Mitte, the Campus Virchow
and the Vascular Center Berlin. Ivo Buschmann currently holds the position of a clinical consultant in vascular
medicine (Angiology).
In 2004, Ivo Buschmann received his habilitation in the field of Vascular Medicine. The main focus of the Richard Thoma Laboratories are the experimental and clinical basics of Arteriogenesis. A special emphasis is the
translational approach, i.e. on promoting the transfer of experimental data into the clinic scenario.
By the end of 2010, CardioAccel will be a first spin-off company from the Richard Thoma Laboratories. This
project is initially funded by the German Federal Ministry of Economy and Technology through the EXIST-Forschungstransfer Programme. The Company develops personalized training programmes for vascular diseases
(Herzhose).
Moreover, Ivo Buschmann is a Member of the Board of the Deutsches Angiologie
Zentrum Berlin –DAZB- (German Angiology Center Berlin), a foundation to promote
research and patient care in the field of peripheral vascular medicine.
41
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Members of the group
Spin-Off Company:
Office
Wilke, Thorsten
Experimental Arteriogenesis (CCR):
Coworkers
Dülsner, André
DVM, Dr. med. vet.
Krackhardt, Florian
MD, Dr. med.
Hillmeister, Philipp Dr. rer. nat.
Pagonas, Nikolaos
MD
Carrão, Catarina
Dr. rer. nat.
Gatzke, Nora
DVM
Harnoß, Jonathan
med. & PhD student
Lee, Eun-Ji
med. & PhD student
Li, Meijing MD, PhD student
Ma, Chenming
MD, PhD student
Nagorka, Stephanie DVM, PhD student
Collaboration with
Prof. K.-L. Schulte
Langhoff, Ralf
Lindhorst, Ruhdja 42
Vascular Center Berlin
Director
Deputy-Director
Dr. med., MD
(sponsored by EXIST-Forschungstransfer Programme of
German Federal Ministry of Economy and Technology)
Coworkers
Buschmann, Eva-Elina Dr. med., MD
Funke, Katharina
Dipl.-Kffr., MBA
Paeschke, Robert
Dipl.-Ing.
ECRC collaboration (Berlin-Buch):
Coworkers
Le Noble, Ferdinand Prof. Dr. rer. nat. (Co-Director
Richard Thoma Laboratories)
Shi, Yu
PhD
Wang, Haitao
MD, PhD student
Scholz, Katja
Technician
Zimmer, Anja
Technician
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Summary
Stimulation of biological bypass formation (Arteriogenesis) is an attractive therapeutic alternative
for patients with diffuse occlusive vessel disease.
Extensive in vivo and in vitro studies have revealed
the basic mechanisms of Arteriogenesis during the
past decades. Early non-invasive induction of collateral proliferation in patients at risk is a major aim
of current studies. Cytokine-induced Arteriogenesis
is a potential novel conservative strategy. Clinical
studies have so far been limited by their unifactorial
approach and non-specific end points. An additional
induction of the natural trigger of Arteriogenesis,
i.e. an enhanced fluid shear stress acting on the
endothelium, can be achieved through active and
“passive” physical training. Clinical studies demonstrate therapeutic safety and promising preliminary
results. Arteriogenesis is presumably the most effective endogenous mechanism of compensation for
arterial stenosis or occlusion. Therapeutic strategies
should therefore support preexistent physiological
mechanisms (fluid shear stress, application of paracrine factors), taking into account pathophysiological principles (potential pro-arteriogenic effects of
atherosclerosis through undifferentiated stem cells,
acceleration of mitogenous processes etc.). Current
data indicate that a single pharmacon acting on a
specific receptor might not be available to sufficiently
induce arteriogensis. Especially in patients suffering
from chronic stenoses, an additive induction of fluid
shear stress may be essential to (re-)activate collateral growth and, thus, to allow for an additional therapeutic benefit of any pro-artiogenic strategy. This
concept ist supported by current experimental data
proving that collateral growth without enhanced fluid
shear stress does not lead to a functional improvement oft tissue perfusion. The Richard Thoma Laboratories explore the field of Arteriogenesis both with
respect to its experimental basics of as well as with
respect to transforming the resulting findings into
everyday clinical and ambulant processes.
43
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Zusammenfassung :
Die Stimulation des biologischen Bypasses (Arteriogenese) stellt eine attraktive alternative Behandlungsmethode v. a. für Patienten mit hochgradigen
diffusen stenosierenden Gefäßerkrankungen dar.
Basismechanismen der Arteriogenese konnten in
ausgedehnten Untersuchungen der letzten Jahrzehnte grundlegend verstanden werden. Eine
rechtzeitige und möglichst wenig invasive Induktion
kollateraler Proliferation bei Risikopatienten ist das
Ziel aktueller Studien. Zytokininduzierte Arteriogenese ist eine potenzielle medikamentös-konservative
Strategie. Klinische Studien waren bislang durch
den unifaktoriellen Ansatz sowie z. T. unspezifische
Endpunkte limitiert. Eine zusätzliche Induktion des
natürlichen Auslösers der Arteriogenese, der endothelialen Schubspannung, kann durch aktives und
passives körperliches Training erfolgen. Klinische
Anwendungen zeigen Therapiesicherheit und erste
vielversprechende Ergebnisse. Zusammenfassend
gilt, dass die Arteriogenese vermutlich den wichtigsten Mechanismus darstellt, um Stenosen und
Okklusionen im Körper endogen zu kompensieren.
Therapeutische Strategien sollten daher eine Unterstützung bereits vorhandener nebenwirkungsarmer
biologischer Mechanismen anstreben (Induktion
von Schubspannung, Benefit spezifischer parakriner
Faktoren), ohne dabei pathophysiologische Prinzipien aus dem Auge zu verlieren (mögliche Induk-
44
tion der Atherosklerose z. B. durch undifferenzierte
Stammzellen, Beschleunigung mitogener Prozesse
im Körper etc.). Ein weiterer wesentlicher Punkt
betrifft die bisher erfolgreich angewandte Strategie
der singulären Pharmakonentwicklung und -therapie
mittels eines funktionell wirksamen pharmakologischen Wirkmoleküls (z. B. Angiotensinrezeptorblocker mit spezifischem Effekt auf selektiven Rezeptor
und respektive Blutdruck): Die vorliegenden Daten
deuten bei der Arteriogenese darauf hin, dass
womöglich dieses singuläre pharmakologische Prinzip bei der Arteriogenese nicht wirksam sein könnte.
Gerade bei Patienten, bei denen Gefäßstenosen
schon länger bestehen, kann eine additive Erhöhung
der Schubspannung (durch aktives oder passives
Training) notwendig sein, um kollaterales Wachstum
erneut „anzuschalten“. Erst diese Aktivierung des
Endothels ermöglicht dann additive therapeutische
Verbesserungen durch proarteriogene Therapien.
Diese konzeptio­nelle Überlegung wird von den derzeitigen experimentellen Befunden unterstrichen,
die eindeutig belegen, dass kollaterales Wachstum
ohne „Shear-Stress-Erhöhung“ zu keinen funktionellen Verbesserungen der Gewebsperfusion führt. Die
Richard Thoma Laboratorien forschen im Bereich
der experimentellen Grundlagen der Arteriogenese
sowie der Umsetzung in den ambulanten als auch
klinischen Alltag.
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Research projects
1. Effects of Aspirine on Adaptive Arteriogenesis
Project leader André Dülsner
CoworkersNora Gatzke, Dr. med. vet. Johanna Glaser, Philipp Hillmeister, Meijing Li, Eun-Ji Lee,
Dr. rer. nat. Kerstin Lehmann, Stephanie Nagorka DVM, Vesna Furundzija MD,
Heike Meyborg BSc, Philipp Stawowy MD, Dr. Andreas Busjahn, Ivo Buschmann
Funding
Deutsche Forschungsgemeinschaft (Grant DFG BU1141/4-2),
VolkswagenStiftung
Background and Purpose – Aspirin and CClopidogrel
are well-used in the prevention of vascu­lar diseases.
In virtue of its anti-inflammatory effects aspirin inhibits
peripheral Arteriogenesis. In this study we investigated
the effects of acetylsalicylic acid (ASA) and Clopidogrel
on cere­bral Arteriogenesis.
Methods – Rats underwent the 3-VO method and were
treated with either drinking water as control, ASA or
Clopidogrel for seven or 21 days. They also received
G-CSF every other day for one week to evaluate their
effects on therapeutically induced Arteriogenesis.
Cerebrovas­cular reactivity was examined as well as
post mortem latex angiography.
but not Clopidogrel. Furthermore, it is still significantly
decreased after treat­ment with ASA combined with
G-CSF compared to G-CSF only. The PCA diameters,
which are not influenced by Clopidogrel, do not differ after ASA combined with G-CSF-treated rats from
those, which are treated with ASA alone, and they
are still smaller compared to the G-CSF single treatment. The enhanced monocyte migration after treatment with G-CSF, GM-CSF and M-CSF is significantly
reduced by pretreatment with ASA.
Conclusions – In this study it is shown that ASA, but
not Clopidogrel inhibits G-CSF im­proved cerebral
Arteriogenesis.
Results – Reserve capacity is completely abolished
after 3-VO and is still abrogated after 21 days by ASA,
45
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
2. S
timulation of coronary collateral growth by granulocyte
stimulating factor : role of reactive ox ygen species.
Project leader Ana Catarina R. Carrão
CoworkersWilliam M. Chilian, June Yun, Christopher Kolz, Petra Rocic, Lehmann Kerstin,
Jeroen P.H.M. van den Wijngaard, Pepijn van Horssen, Jos A. E. Spaan,
Vahagn Ohanyan, Yuh Fen Pung, Ivo Buschmann
Funding EU-Grant
CollaborationsDepartment of Integrative Medical Sciences, NEOUCOM, Northeastern Ohio University
College of Medicine, Rootstown, Ohio; Department of Biochemistry and Molecular Biology,
University of South Alabama, Mo­bile; Department of Biomedical Engineering and Physics,
Academic Medical Centre, Amsterdam, Netherlands.
Objective - The purpose of this study was to determine
whether G-CSF promotes coronary collateral growth
(CCG) and decipher the mechanism for this stimulation.
Methods and results - In a rat model of repetitive episodic myocardial ischemia (RI, 40 se­conds LAD occlusion every 20 minutes for 2 hours and 20 minutes, 3
times/d for 5 days) CCG was deduced from collateraldependent flow (flow to LAD re­gion during occlusion).
After RI, G-CSF (100 microg/kg/d) increased CCG
(P<0.01) (0.47+/-0.15) versus vehicle (0.14+/-0.06).
Surprisingly, G-CSF treatment without RI increased
CCG (0.57+/-0.18) equal to G-CSF+RI. We evaluated
ROS by dihydroethi­dine (DHE) fluorescence (LV injec-
46
tion, 60 microg/kg, during two episodes of ische­mia).
DHE fluorescence was double in G-CSF+RI versus
vehicle+RI (P<0.01), and even higher in G-CSF without RI (P<0.01). Interestingly, the DHE signal did not
colo­calize with myeloperoxidase (immunostaining,
neutrophil marker) but appeared in cardiac myocytes.
The study of isolated cardiac myocytes revealed the
cytokine sti­mulates ROS which elicit production of
angiogenic factors. Apocynin inhibited G-CSF effects
both in vivo and in vitro.
Conclusions - G-CSF stimulates ROS production
directly in cardiomyocytes, which plays a pivotal role in
triggering adaptations of the heart to ischemia including growth of the coronary collaterals.
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
3. M
olecular Mechanisms of the BradykininPathway for Arteriogenesis
Project leader Philipp Hillmeister
CoworkersKerstin E Lehmann, Anja Bondke, Henning Witt, André Duelsner, Cle­mens Gruber,
Hans-Jörg Busch, Joachim Jankowski, Patricia Ruiz-Noppinger,
Konstantin-Alexander Hossmann, Ivo Buschmann
Funding Deutsche Forschungsgemeinschaft, (Grant DFG BU 1141/4-2),
Volkswagen Foundation, Germany
CollaborationsCharité University Medicine Berlin: Center for Preclinical Studies (CC2), Institute of Physiology,
Max-Planck-Institute for Molecular Ge­netics, Department of Anatomy,
Institute of Integrative Neuroana­tomy; Medical Clinic IV,; Department of Internal Medicine
(Cardiology), University of Freiburg i. Br.; Max-Planck-Institute for Neurological Re­search,
Cologne;
Cerebral Arteriogenesis constitutes a promising therapeutic concept for cerebrovascular is­chaemia; however, transcriptional profiles important for therapeutic
target identification have not yet been investigated.
This study aims at a comprehensive characterization
of transcrip­tional and morphologic activation during
early-phase collateral vessel growth in a rat model
of adaptive cerebral Arteriogenesis. Arteriogenesis
was induced using a three-vessel occlu­sion (3-VO)
rat model of nonischaemic cerebral hypoperfusion.
Collateral tissue from grow­ing posterior cerebral artery
(PCA) and posterior communicating artery (Pcom) was
selec­tively isolated avoiding contamination with adjacent tissue. We detected differential gene expression
24 h after 3-VO with 164 genes significantly deregulated. Expression patterns contained gene transcripts
predominantly involved in proliferation, inflammation,
and migra­tion. By using scanning electron microscopy, morphologic activation of the PCA endothelium
was detected. Furthermore, the PCA showed induced
proliferation (PCNA staining) and CD68+ macrophage
staining 24 h after 3-VO, resulting in a significant
increase in diameter within 7 days after 3-VO, confirming the arteriogenic phenotype. Analysis of molecular
an­notations and networks associated with differentially
expressed genes revealed that early-phase cerebral
Arteriogenesis is characterised by the expression of
protease inhibitors. These results were confirmed by
quantitative real-time reverse transcription-PCR, and
in situ hybridisation localised the expression of tissue
inhibitor of metalloproteinase-1 (TIMP-1) and kininogen to collateral arteries, showing that TIMP-1 and
kininogen might be molecular markers for early-phase
cerebral Arteriogenesis.
47
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
4. I mprovement of fractional flow reserve and collateral flow
by treatment with external counterpulsation (Art.Net.-2 Trial)
Project leader Eva-Elina Buschmann
Coworkers Utz W, Pagonas N, Schulz-Menger J, Busjahn A, Monti J, Maerz W, le Noble F,
Thierfelder L, Dietz R, Klauss V, Gross M, Buschmann IR
Funding
Principal Investigator funded
CollaborationsFranz-Volhard-Klinik, Department for Cardiology, Helios-Klinikum Ber­lin-Buch,
HealthTwiSt GmbH, Berlin – Buch, Synlab laboratory ser­vices, Eppelheim,
Max-Delbrueck-Center for Molecular Medicine, An­giogenesis and Cardiovascular
Pathology, Berlin, Medizinische Poli­klinik der Universität München; Campus Innenstadt,
Dept. for Cardio­logy, München, Department for Internal Medicine (Cardiology),
Univer­sity Clinic Freiburg/ Germany.
Background - Arteriogenesis (collateral artery growth)
is nature’s most efficient rescue mechanism to overcome the fatal consequences of arterial occlusion or
ste­nosis. The goal of this trial was to investigate the
effect of external counterpulsation (ECP) on coronary
collat­eral artery growth.
Materials and Methods - A total of 23 patients (age 61
+/- 2.5 years) with stable coronary artery disease and
at least one haemodynamic significant stenosis eligible for percutaneous coronary intervention were prospectively recruited into the two study groups in a 2 : 1
man­ner (ECP : control). One group (ECP group, n = 16)
underwent 35 1-h sessions of ECP in 7 weeks. In the
control group (n = 7), the natural course of collateral
circulation over 7 weeks was evaluated. All patients
underwent a cardiac catheterization at baseline and
after 7 weeks, with invasive measurements of the
48
pressure-derived collateral flow index (CFIp, pri­mary
endpoint) and fractional flow re­serve (FFR).
Results - In the ECP group, the CFIp (from 0.08 +/0.01 to 0.15 +/- 0.02; P < 0.001) and FFR (from 0.68
+/- 0.03 to 0.79 +/- 0.03; P = 0.001) improved significantly, while in the control group no change was
observed. Only the ECP group showed a reduc­tion
of the Canadian Cardiovascular Society (CCS, P =
0.008) and New York Heart Association (NYHA, P <
0.001) classification.
Conclusion - In this study, we provide direct functional
evidence for the stimulation of coro­nary Arteriogenesis
via ECP in patients with stable coronary artery disease.
These data might open a novel noninvasive and preventive treatment avenue for pa­tients with non-acute
vascular stenotic disease.
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Publications 2006-2010
Jungehuelsing GJ, Liman TG, Brunecker P, Ebel A, Endres
M, Buschmann I, Pagonas N, Buschmann EE. (2010): Does
External Counterpulsation Augment Mean Cerebral Blood
Flow in the Healthy Brain? Effects of External Counterpulsation on Middle Cerebral Artery Flow Velocity and Cerebrovascular Regulatory Response in Healthy Subjects. Cerebrov­
asc Dis Oct 15;30(6):612-617
Buschmann I, Pries A, Styp-Rekowska B, Hillmeister P,
Loufrani L, Henrion D, Shi Y, Duelsner A, Hoefer I, Gatzke N,
Wang H, Lehmann K, Ulm L, Ritter Z, Hauff P, Hlushchuk R,
Djonov V, van Veen T, le Noble F. (2010): Pulsatile shear and
Gja5 modulate arterial identity and remodeling events during
flow-driven Arteriogenesis. Development Jul;137(13):2187-96
von Knobelsdorff-Brenkenhoff F, Buschmann EE, Pilz B,
Schulz-Menger J (2010): Persistent Cabrol shunt causing
severe right heart failure. Ann Thorac Surg. Jul;90(1):312
Carrão AC, Chilian WM, Yun J, Kolz C, Rocic P, Lehmann K,
van den Wijngaard JP, van Horssen P, Spaan JA, Ohanyan
V, Pung YF, Buschmann I (2009): Stimulation of coronary
collateral growth by granulo­cyte stimulating factor: role of
reactive oxygen species. Arterioscler Thromb Vasc Biol
Nov;29(11):1817-22
Buschmann EE, Utz W, Pagonas N, Schulz-Menger J,
Busjahn A, Monti J, Maerz W, le Noble F, Thierfelder L, Dietz
R, Klauss V, Gross M, Buschmann IR (2009): Arteriogenesis
Network (Art. Net.). Improvement of fractional flow reserve
and collateral flow by treatment with external counterpulsation
(Art.Net.-2 Trial). Eur J Clin Invest Oct;39(10):866-75
von Knobelsdorff-Brenkenhoff F, Rudolph A, Wassmuth R,
Bohl S, Buschmann EE, Abdel-Aty H, Dietz R, Schulz-Menger
J (2009): Feasibility of cardiovascular magnetic resonance to
assess the orifice area of aortic bioprostheses. Circ Cardio­
vasc Imaging Sep;2(5):397-404, 2 p following 404
Pagonas N, Utz W, Schulz-Menger J, Busjahn A, Monti J,
Thierfelder L, Dietz R, Klauss V, Gross M, Buschmann IR,
Buschmann EE; on behalf of the Arteriogenesis Network (Art.
Net.). (2009): Assessment of the effect of external counterpulsation on myocardial adaptive Arteriogenesis by invasive
functional mea­surements - design of the Arteriogenesis
network trial 2. Int J Cardiol Jun 16
Hillmeister P, Lehmann KE, Bondke A, Witt H, Duelsner
A, Gruber C, Busch HJ, Jankowski J, Ruiz-Noppinger P,
Hossmann KA, Buschmann IR (2008): Induction of cerebral
Arteriogenesis leads to early-phase expression of protease
inhibitors in growing collaterals of the brain. J Cereb Blood
Flow Metab Nov;28(11):1811-23
Bondke A, Buschmann IR (2007):Visualization of therapeutic
Arteriogenesis by MR angiography: functional measurements
are superior to surrogate parameters. J Magn Reson Imag­
ing Jul;26(1):215; author reply 216
Buschmann IR, Bondke A, Elgeti T, Kühnle Y, Dietz R, Möckel
M (2007): Positive cardiac troponin I and T and chest pain in a
patient with iatrogenic hypothyroidism and no coronary artery
disease. Int J Car­diol Feb 7;115(2):e83-5
Schefe JH, Lehmann KE, Buschmann IR, Unger T, FunkeKaiser H (2006): Quantitative real-time RT-PCR data analysis:
current concepts and the novel “gene expression’s CT difference” formula. J Mol Med Nov;84(11):901-10
Bergmann CE, Hoefer IE, Meder B, Roth H, van Royen N,
Breit SM, Jost MM, Aharinejad S, Hartmann S, Buschmann IR
(2006): Arteriogenesis depends on circulating monocytes and
macrophage accumulation and is severely depressed in op/
op mice. J Leukoc Biol Jul;80(1):59-65
Reviews & Editorials 2006-2010
le Noble F, Klein C, Tintu A, Pries A, Buschmann I (2008):
Neural guidance molecules, tip cells, and me­chanical factors
in vascular development. Cardiovasc Res May 1;78(2):23241
Buschmann IR, Lehmann K, Le Noble F; Art.Net. (2008):
Physics meets molecules: is modulation of shear stress the
link to vascular prevention? Circ Res Mar 14;102(5):510-2
Bondke A, Buschmann IR, Bode C, Buschmann EE (2007):
[Inducing collaterals in due time. Arteriogenesis as a preventive principle] Haemostaseologie Dec;27(5):363-72
Bondke A, Hillmeister P, Buschmann IR (2007): Exact assessment of perfusion and collateral vessel prolifera­tion in small
animal models. Circ Res Apr 27;100(8):e82-3
49
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Visualization of therapeutic Arteriogenesis by MR angiography: Functional measurements are superior to surrogate
parameters. J Magn Reson Imaging 2007 Jul;26(1):215
Erdo F, Buschmann IR (2007): [Arteriogenesis: a new strategy
of therapeutic intervention in chronic arterial disorders. Cellular mechanism and experimental models] Orv Hetil. Apr
8;148(14):633-42
Donner S: Spaziergänge lassen neue Blutgefäße sprießen,
Die Welt – Online, 07.09.2009
Donner Susanne, Herzhose sorgt über natürlichen Bypass für
bessere Durchblutung, VDI-Nachrich­ten, 25.09.2009
N.N.: Blutgefäße können wieder wachsen, Das Goldene
Blatt, 12.10.2009, 30
Buschmann IR, Bondke A, Elgeti T, Kuhnle Y, Dietz R, Mockel
M (2007): Positive cardiac troponin I and T and chest pain in a
patient with iatrogenic hypothyroidism and no coronary artery
disease. Int J Car­diol Feb 7;115(2)
Blech J: Das Blut in Wallung bringen, Der Spiegel
18.01.2010,102-104.
Book Chapters 2006-2010
Krause Claudia: Mein Herz schlägt wieder kräftig, Mach Mal
Pause, BauerMediaGroup, Nr.33, 1.08.2010, 54
Chapter:
Buschmann I, Pagonas N, “Stimulation of Arteriogenesis via
External Counterpulsation – State of the Art and Clinical Perspective” in Schaper, W; Deindl, E, “The Collateral Circulation
– Molecular Regulation, Pathophysiology and Therapeutics”;
to be published Autumn 2010
Boengler K, Buschmann I, Deindl E, Gottwik M, Hoffmeister
HM, Ito W, Klein H, Mauser M, Nienaber C, Regitz-Zagrosek
V, Sack S (2009): On the occasion of Wolfgang Schaper’s
75th birthday. Basic Res Car­diol Jan;104(1):2-4
Chapter:
Pries A, Buschmann I, Habazettl H, “Key Issues in Vascular
Pathology” in
Lanzer P. Mastering endovascular techniques: a guide to
excellence. Lippincott Williams & Wilkins, 2006
Popular Press Publications / Target
Customer Publications
related to the Group’s research :
Von Lutterotti N: Kreislaufmassage oder wenn kühne
Träume platzen, Frankfurter Allgemeine Zeitung, Nr. 179,
05.08.2009, N5
Abel S: Biologische Bypässe, Gesund – Beilage des Springer
Verlags zum Hamburger Abendblatt etc., 04.09.2009, 4
50
Aumiller J: Herzhose bringt das Blut in Wallung, CardioNews,
Nr. 4, 2010, 34
TV- and Radio-broadcasts related to
the Group’s research :
Engler K: Schaufensterkrankheit und „Herzhose“, MDR
1, Radio Sachsen-Anhalt, Vormittags­programm vom
21.10.2009
Brummerloh D: Biologische Bypässe: Herzhose lässt neue
Umgehungsarterien wachsen –BR5, 11.07.2010
Engler K: Beitrag zu Schaufensterkrankheit und „Herzhose“
im Magazin „Hauptsache Gesund“, MDR Fernsehen,
21.01.2010
Brettschneider E: Training statt Herz-Bypass: Bewegung lässt
neue Blutgefäße am Herzen sprie­ßen, Gesundheitsmagazin
„QuiVive“, RBB-Fernsehen ,03.03.2010
Rosbach J: Mit der Herzhose zu neuen Arterien, Beitrag im
Magazin „Visite“, NDR-Fernsehen, 30.03.2010 (wiederholt
im WDR-Fernsehen, Ratgeber Servicezeit Gesundheit,
19.07. & 20.07.2010)
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
General Information
Third party funding ( 2006-2010 )
Project leader
Buschmann I.
Project title
VolkswagenStiftung Arteriogenese
Sponsor
VolkswagenStiftung
Period
2003-2006
Buschmann I.
Arteriogenese und Risikofaktoren in der
Zucker Fatty Rat
Kompetenznetz Zerebrale Arterioge­nese
Deutsche For­schungsgemein­
schaft, BU 1151/4-1
MBWK
2003-2007
Buschmann I.
Buschmann I.
Buschmann I.
Le Noble F.
EST Teilprojekt
Porcine Arteriogenesis
Flow driven Arteriogenesis
EU
2004-2006
Medtronic (US)
2006-2007
Charité - Experi­mental and Clini- 2008-2011
cal Research Center, Berlin-Buch
Lehmann K.
Lehmann K.
Buschmann I.
Kappert K.
Künstliche Arterie
NGFN Verbundprojekt
Modulation of natu­ral Inhibition as a
therapeutic principle: Protein-TyrosinePhosphatases as interventional target
structures of Arteriogenesis
BMBF
NGFN
DFG – German Research
Commu­nity
2007-2010
2008-2010
2008-2011
Buschmann I.
CardioAccel
2010-2011
Hillmeister P.
Cerebral Arteriogenesis – A Concept for
Prevention
Federal Ministry for Economy
and Tech­nology / EXIST For­
schungstransfer
Creutzfeld Jacob Stipend
from Charité-Center for Stroke
Research Berlin (CSB)
Buschmann I.
2002-2007
2008-2011
51
IVO BUSCHMANN – EXPERIMENTAL AND CLINICAL ANGIOLOGY / ARTERIOGENESIS
Awards
201015. Dreiländertagung der Schweizerischen, Deutschen und Österreichischen Gesellschaft
für Angiolo­gie und 39. Jahrestagung der Deutschen Gesellschaft für Angiologie und
Gefäßmedizin e.V., Cong­ress Center Basel, Basel, Switzerland , 12.-15.September
1st Poster Prize:
“Pioglitazone inhibits Induction of the Biological Bypass“
André Dülsner
1st Poster Prize:,
„Induction of the Biological Bypass in the Circle of Willis“ Kongress
of the German Society for Vascular Surgery, Berlin, 9.9.10: Nora Gatzke
2010For the best Presentation held in scope of the Campaign
“Young Vascular Medical Scientists”
granted a travel grant Jonathan Harnoß
2010
Stephanie Nagorka
Charité Promotion Stipend granted to 2009-2010 Charité Promotion Stipend granted to 52
Nora Gatzke
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Head of the group
Prof. Dr. med. Duska Dragun
Curriculum Vitae:
1988-1993 Medical School: University of Zagreb, Croatia
1993-1995 Intern at university hospital "Rebro", Zagreb, Croatia
1995
License to practise medicine from Ministry of Health Croatia
1995-1997Doctoral thesis at "Max Delbrück Centrum für molekulare Medizin
Berlin-Buch", Prof. Dr. Hermann Haller, Berlin, Germany
1998Degree as a "Dr. med." from the "Humboldt Universität", Berlin, Germany,
Grade: "summa cum laude"
1998Research fellow at "Dept. for Immunology and Organ Transplantation"
Prof. Dr. Barry Kahan, University of Texas, Houston, USA
1999-2000Clinical fellow at "Franz Volhard Klinik", Charité Campus Buch,
Prof. Dr. Friedrich C. Luft, Berlin, Germany
2002-2007 Assistant Professor at the Medical faculty of the Charité, Berlin
2000-2002 Clinical fellow at Department of Nephrology, Charité Campus Mitte,
2005
Specialist in Internal Medicine, Berlin
2005-2006 Attendant at Department of Nephrology, Charité Campus Mitte,
2006-Attendant at Clinic for Nephrology and Intensive Care Medicien, Charité
Campus Virchow Clinic
2008
Specialist in Nephrology, Berlin
2008-
Full Professor at the Medical faculty of the Charité, Berlin
Current administrative functions
2002-
Member of the Faculty board for career development
2005
Member of executive board of "Junge Charité"
2006-
Advisory board of the Charité Mentoring Program for female scientists
2009-
Deputy member of the Board of the Medical faculty of Charité
2009-
Co-chair of the Faculty Board for career development
2009-
Scientific Advisory Board of the German Society of Transplantation
2009-
Task force "Kidney transplantation" German Society of Nephrology
53
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Members of the group
Scientists
Catar, Rusan Ali
Chaykovska, Lyubov
Fielitz-Haase, Anja
Fuller, Florian
Haase, Michael
Hegner, Björn
Hoff, Uwe
Kusch, Angelika
Näther, Melanie
Philippe, Aurélie
Technicians
Eigen, Marc
Gruber, Gertrud
Kämper, Barbara
54
Dr. rer. nat.
Dr. med.
Dr. rer. medic.
PD Dr. med.
PD Dr. med.
Dr. med.
Dr. med.
Dr. med.
Dr. rer. nat.
Dr. rer. nat.
Graduate and undergraduate students
Baris, Christina
Medical student
Böhm, Susanne
Bachelor student;
07/2008 – 02/2009
Eimers, Meike
PhD student; until 05/2008
Gürgen, Dennis
PhD student
Janke, Daniel
Medical student
Karatas, Aysun
PhD student; until 01/2010
Krause, Sebastian
Medical student
Kretzschmar, Tobias
Medical student
Marinez, Julian
Medical student
Pützer, Jennifer
Medical student
Schaub, Theres
PhD student
Schmidt, Maria
Medical student
Urban, Claudia
Diploma student;
12/2007 – 07/2008
Wagner, Philine
Medical student
Wischnewski, Oskar
Medical student
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Summary
Our group studies the pathogenesis and therapy of
alloantigen independent renal transplant injuries and
the development of new therapeutic modalities in
an experimental and clinical setting. The focus of
our research is to increase the long-term survival of
the graft and to minimize cardiovascular morbidity
in transplanted patients. Moreover, we are involved
in development of novel diagnostic tests and their
validation in various contexts of renal and cardiovascular pathologies.
Research areas of interest:
• Role of agonistic antibodies targeting G-protein
coupled vascular receptors in cardiovascular,
autoimmune, and transplant pathologies
• Vascular mechanisms and interventions aimed
at modulation of vascular injury during ischemia
and reperfusion injury in native kidneys and kidney transplants
• Development of biomarkers to monitor vascular
injury during acute renal failure in native kidneys
and organ transplants
•
•
Sex-specific mechanisms of cardiovascular comoribidity in renal patients
Disease and stimulus specific mechanisms of
differentiation of MSCs
We rely on ‘bedside to bench’ approach that allows
us to translate newly identified disease entities or
results of association studies in disease relevant
animal models for dissection of pathophysiologic
mechanisms. Animal experiments include highly
sophisticated age and gender specific kidney transplant models in rats as well as different models to
study the progression of kidney disease and resulting cardiac hyperthrophy and fibrosis (DOCA-salt
HTN, Transgenic animals). Tissue analysis is done
via histology, immunohistology and molecular biology. Different types of cell cultures are used for the
elucidation of signaltransduction pathways.
55
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Zusammenfassung
Unsere Arbeitsgruppe untersucht die Pathogenese
und Therapiemöglichkeiten der alloantigenunabhängigen Schädigung von Nierentransplantaten
und entwickelt neuartige Behandlungsstrategien
auf experimenteller und klinischer Ebene. Unser
wichtigstes Ziel ist eine Verbesserung des LangzeitTransplantatüberlebens bei gleichzeitiger Minimierung der kardiovaskulären Morbidität transplantierter Patienten. Darüberhinaus beschäftigen wir uns
mit der Entwicklung neuer Biomarker in der Diagnostik renaler und kardiovaskulärer Erkrankungen.
Unsere Forschungsgebiete sind:
• Rolle agonistischer Antikörper gegen G-Protein
gekoppelte vaskuläre Rezeptoren in kardiovaskulären, autoimmunen und transplantationsassoziierten Pathologien
• Vaskuläre Mechanismen und Interventionen zur
Modulation des Gefäßschadens im Rahmen
des Ischämie-Reperfusionsschadens in Eigennieren und Nierentransplantaten
• Entwicklung von Biomarkern zur Überwachung
von Gefäßschäden bei akutem Nierenversagen
in Eingennieren und Organtransplantaten
56
•
•
Geschlechtsspezifische Mechanismen kardiovaskulärer Co-Morbidität bei Nierenpatienten
Krankheits- und stimulusspezifische Mechanismen der Differenzierung von MSCs
Unsere Forschung basiert auf dem "bedside to
bench"-Ansatz, der uns gestattet, neu identifizierte
Krankheitsentitäten und Ergebnisse von Assoziationsstudien in krankheitsrelevanten Tiermodellen
in Hinblick auf die zugrunde liegenden pathophysiologischen Mechanismen aufzuschlüsseln. Die
Tierversuche umfassen neben spezifischen altersund geschlechtsbezogenen Rattenmodellen auch
unterschiedliche Modelle zur Untersuchung der
Progression von Nierenerkrankungen und der daraus resultierenden Herzhypertrophie und Fibrose
(DOCA-Salz induzierte Hypertonie, transgene Tiere),
sowie experimentelle Nieren- und Gefäßtransplantation. Die Gewebsanalyse erfolgt mittels Histologie,
Immunhistologie und molekularbiologischen Methoden. Zur Aufklärung von Signaltransduktionsmechanismen werden verschiedene Zellkultursysteme
eingesetzt.
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Research projects
1. A
ngiotensin II type 1 receptor (AT1R-Abs ) and Endothelin-1 type
A receptor antibodies ( ETAR-Abs ) in transplant and autoimmune
pathologies
Project leader
Coworkers Cooperations
Funding Duska Dragun
Aurélie Philippe, Rusan Catar, Melanie Näther, Tobias Kretzschmar,
Oskar Wischnewski, Barbara Kämper
G. Riemekasten, H. Heidecke, C. Schönemann
U. Querfeld, N. Hiemann, R. Hetzer, P. Reinke, HD Volk, J. van Laar (Newcastle),
A. Ghofrani (Giessen), W. Seeger (Giessen), A. Melk (Hannover), C. Schmidt,
B. Tönshoff (Heidelberg), M. Konrad (Münster), B. Nashan (Hamburg),
A. Benigni (Bergamo), G. Remuzzi (Bergamo), S. Schaub (Basel), J. Steiger (Basel),
J. P. Soulillou (Nantes), H. U. Meier-Kriesche (Gainesville), N. Reinsmoen (Los Angeles),
A. Zeevi (Pittsburgh), T. Coates (Adelaide)
Bundesministerium für Wirtschaft und Technologie, Deutsche Stiftung
Sklerodermie,
Actelion Pharmaceuticals Deutschland GmbH,
Institutional funding (Universitäre Forschungsförderung Charité)
Our group has identified functional autoantibodies
against the Ang II type 1 receptor as the first nonHLA target responsible for vascular rejection, providing a link between cardiovascular and immune diseases. Obliterative vascular lesions in SSc resemble
those in transplant vasculopathy. We reasoned that
agonistic autoantibodies to vascular receptors might
also be present in SSc and might serve as a diseasespecific biomarker. We identified functional autoimmunity directed not only against AT1, but also directed
against and ETA receptors in SSc patients. Increased
levels of autoantibodies against these receptors were
associated with more severe disease manifestations. Harboring anti-AT1R and anti-ETAR antibodies
revealed patients with higher likelihood for development of pulmonary hypertension and mortality from
SSc related complications. Both autoantibodies were
biologically active as they enhanced pulmonary vascular reactivity to endogenous receptor agonists in
a dose-dependent manner that could be blocked
by respective pharmacologic antagonists. Anti-AT1R
and anti-ETAR autoantibodies may directly contribute
to the pathogenesis of SSc and could represent a
link between the increased vascular responsiveness
and tissue damage. This interpretation would support
the "vascular hypothesis" in the pathophysiology of
SSc. To better understand the link between the autoantibodies we detected, and the two receptors, we
are currently working on protein immunoprecipitation
using rat vascular smooth muscle cell primary cultures
which express endogenously both receptors. Moreover, we are investigating the implication of the auto57
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
antibodies in the cardiovascular disease by studying
the signaling pathways depending on the AT1 and ETA
receptors.
Angiotensin II (Ang II) and Endothelin-1 (ET-1) mediate
functional alterations in resistance arteries in terms
of enhanced contractility or impaired relaxation. An
increased peripheral resistance in renal, pulmonary,
mesenteric and coronary arteries may contribute to
severe vascular pathologies observed in organ transplant recipients and patients with systemic obliterative vasculopathy due to systemic sclerosis. We study
vascular-bed specific effects of autoantibodies targeting vascular G-protein coupled receptors and their role
as an enhancer of vasoactive responses initiated by
natural ligands, Ang II and ET-1. We also study influence of permissive phenomena such is ischemia on
autoantibody-receptor target interaction.
Agonistic autoantibodies as allosteric receptor activators
58
Clinical screening studies for agonistic
autoantibodies
In cooperation with the DHZB a prospective study on
the role of AT1R-Abs in cardiac transplant recipients
is currently performed. Data submitted for publication show that presence of AT1R-Abs correlated with
earlier onset and more frequent development of cardiac transplant microvasculopathy as determined in
protocol biopsies. In cooperation with Departments
of Pediatric Nephrology at the Charité and at the
Medical School Hannover, we prospectively recruit
pediatric renal transplant recipients. Children with
history of transplant losses due to vascular rejection
and presence of AT1R-Abs are enrolled in preemptive
therapy study employing immunoadsorption, intensified immunosuppression and high-dose sartanes. In
cooperation with the Department of Visceral Surgery
and Organ Transplantation at the Charité Campus Virchow Clinic, we prospectively evaluate liver-, smallbowel and multivisceral transplant recipients for the
presence of AT1R-Abs and initiate rescue protocols in
case of antibody related transplant injury. Preliminary
results implicate association with bile-duct vanishing
lesion in liver transplants and vascular rejection in multivisceral transplants. In cooperation with the Department of Nephrology and Transplant Center Nantes
in France we prospectively study cohort of several
hundreds of renal transplant patients for AT1R-Abs
in conjuction with immune monitoring procedures for
multiple T-cell and B-cell markers, as well as regular
surveillance of allograft histology by protocol biopsies.
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
2. E
ndothelial mechanisms in thrombotic microangiopathy induced
acute renal failure
Project leader Coworkers Cooperations
Funding Duska Dragun
Melanie Näther, Aurélie Philippe, Rusan A. Catar
U. Rauch
Dr. Werner Jackstädt Stiftung
Institutional funding (Universitäre Forschungsförderung Charité)
Thrombotic microangiopathy (TMA) is the morphological substrate in a variety of diseases associated with
acute renal failure. Typical histological features of TMA
are vessel wall thickening with endothelial swelling and
formation of platelet–fibrin hyaline microthrombi that
occlude arterioles and capillaries. TMA occurs in a
wide range of diseases including haemolytic uraemic syndrome (HUS), thrombotic thrombocytopenic
purpura (TTP), autoimmune disorders, malignant
hypertension, and vascular transplant rejection. All
of these diseases have a severe and rapid progression in common with very limited therapeutic options
because the underlying mechanisms are unknown.
In previous studies we identified activating autoan-
tibodies directed against AT1 and ETA receptors in
patients with systemic sclerosis (SSc) and renal crisis.
We could show in cultured HMEC-1 cells that stimulation with IgG from SSc patients resulted in activation
of stress signalling kinase ERK1/2 and upregulation
of prothrombotic Tissue Factor (TF). The observed
effects could be inhibited by specific pharmacologic
blockade of AT1, and ETA receptors. Based on these
results we study transcription factor Ets-1 dependent
activation of TF in HMEC-1 cells and expression of
soluble TF-isoforms in blood samples and renal biopsies of patients with acute transplant vasculopathy,
SSc with renal crisis, and atypic HUS / TTP.
IgG from patients with systemic sclerosis upregulates the proteinexpression of prothrombotic Tissue
Factor in the cytosolic compartment (B) of cultured HMEC-1 cells but not in membrane extracts (A).
59
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
3. S
ystems Biology towards Novel Chronic Kidney Disease - Diagnosis
and Treatment: SysKid
Project leader
Coworkers
Cooperations
Funding
Duska Dragun
Björn Hegner, Aurélie Philippe, Theres Schaub
The SysKid consortium members
European Commission FP7
Chronic kidney disease affects up to 10% of the population, and besides eventual progression towards end
stage renal disease massively impacting patient’s
quality of life, comorbidities including cardiovascular
complications and bone metabolism disorders are
serious consequences. But still, early stage diagnosis
and tailored treatment is not adequate on the everyday
clinical level. On the one hand chronic kidney disease
might not yet have reached its appropriate emplacement in an epidemiological and healthcare perspective, but also the pathophysiology of the disease on a
molecular and cellular level is not well enough understood. The sysKID consortium was installed for precisely addressing these issues: Unravel the molecular
and cellular mechanisms of chronic kidney disease
development, combine this information with clinical
risk factors, and on this basis delineate chronic kidney disease biomarkers. These markers will allow us
preclinical studies of novel therapy approaches for
halting disease progression, and will provide us with
60
the materials for development and clinical evaluation
of tools for early stage diagnosis as well as prognosis
and treatment monitoring.
The main goal of our SysKid project is not only to
identify biomarkers for fast progression and worse
disease outcome but also to elucidate novel pathomechanisms involved in renal disease and related cardiovascular conditions on the molecular, cellular, and
organism level. Here we focus on auto-antibodies that
activate vascular receptors and thereby exacerbate
renal and vascular damage and circulating progenitor cells that might contribute to disease progression
when malfunctioning in pathologic contexts. Within
the SysKid consortium we screen large well defined
patient cohorts for association of auto-antibodies with
specific disease and progression characteristics. Our
biologic models allow for evaluation of a huge variety
of disease relevant processes to test candidate molecules identified by us and other SysKid groups for
their impact on renal and vascular damage.
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
4. D
isease specific mechanisms of smooth muscle and fibroblastic
differentiation of mesenchymal stem cells ( MSC )
Project leader
Coworkers
Cooperations
Funding
Björn Hegner, Duska Dragun
Melanie Näther, Theres Schaub
G. Riemekasten, C. Lange (Hamburg)
Deutsche Stiftung Sklerodermie
Institutional funding (Universitäre Forschungsförderung Charité)
Bone marrow derived MSC home to injured vessels
and contribute to neointima formation during instent
restenosis and transplant arteriosclerosis. Systemic
sclerosis (SSc) is an autoimmune form of obliterative
vasculopathy that eventually leads to systemic fibrosis
affecting skin and various organs. We propose that
MSC from patients with SSc differ from healthy controls in terms of aberrant smooth muscle and fibroblastic differentiation leading to vasculopathy, matrix
deposition and tissue calcification. We focus on signal
transduction and transcriptional control of mobilisation, homing, proliferation, apoptosis, smooth muscle
differentiation, matrix and calcium deposition. Influence on Angiotensin II and Endothelin-1 and putative
amplification of aberrant cellular processes via agonistic autoantibodies against AT1 and ETA receptors
is an additional focus.
L-type Ca channel dependent
calcium influx into MSC upon
stimulation with 60 mM KCl
61
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
5. S
exual dimorphisms in myocardial hypertrophy and cardiorenal
interaction – signal transduction and intervention strategies
Project leader
Coworkers Cooperations
Funding
Angelika Kusch, Björn Hegner, Duska Dragun
Dennis Gürgen, Lyubov Chaykovska, Aysun Karatas, Claudia Urban, Christina Baris
L. de Windt (Utrecht), F. Luft, U. Kintscher, V. Regitz-Zagrosek, O. Huber
DFG, DR 498/1-1 – FOR 1054, TP 2 und GK 752-III, TP 3
We have described sex-specific regulation of the calcineurin pathway in response to largely blood pressure-independent mineralocorticoid action in DOCAsalt induced left ventricular myocardial hypertrophy
(MH) and renal hypertrophy in mice. We now study
the contribution of individual estrogen receptors (ERα
and ERβ) in this setting and expand our research on
diverse signalling pathways implicated in the development of adaptive and maladaptive MH. A major focus
is the Akt/mTOR pathway as a putative master switch
between adaptive and maladaptive MH. We use a
pharmacologic approach of mTOR blockade in our
DOCA-salt model of mineralocorticoid excess. In addition, transverse aortic constriction and voluntary cage
wheel running are employed as models for pressure
and exercise induced MH, respectively.
Sex differences in elements upstream and downstream from mTOR are studied in vitro in female murine
cardiomyocytes (HL-1) using genomic overexpression
and depletion approaches. Presence or absence of
estradiol resembles pre- and postmenopausal hormonal status to determine the influence of estrogen on
hypertrophic signal transduction focusing on a linear
versus non-linear pathway involvement.
Uni-Nx+salt controls compared with DOCA-salt
mice that were treated with hydralazine to remove
confounding effects of blood pressure elevation A:
RT-PCR for calcineurin Aβ in the heart B: RT-PCR
for modulating calcineurin and inhibiting protein 1
(MCIP1) C: Immunoprecipitation and subsequent
Western blot for phosphorylated nuclear factor of
activated T cells c2 (pNFATc2)
62
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
6. R
elevance of auto-antibodies directed against protease activated
receptors 1 and 2 ( PAR-1 and PAR-2 ) receptors in immune-mediated
renal disease and after renal transplantation
Project leader
Coworkers Cooperations
Funding Duska Dragun
Rusan Catar, Uwe Hoff, Björn Hegner, Barbara Kämper
G. Riemekasten, H. Heidecke, R. Schindler, M. Noris (Bergamo), G. Remuzzi
(Bergamo), C. Licht (Toronto), S. Coughlin (San Diego)
Bundesministerium für Wirtschaft und Technologie
Initiation of thrombin dependent pathways mediated
through activation of PAR-1 and PAR-2 is increasingly
recognized as an important trigger of microthrombosis in crescentic glomerulonephritis and thrombotic
microangiopathies. We propose that agonistic antibodies directed against PAR-1 and PAR-2 mediate
sustained receptor activation and thereby actively
contribute to various renal pathologies. Based on our
experience in the development of assays for screening
of autoantibodies against G-protein coupled receptors we have currently developed in collaboration with
Celltrend an assay for detection of PAR-1 and PAR-2
autoantibodies. We screen our cohorts of transplant
recipients, patients with thrombotic microangipathies
in native kidneys, patients with glomerulonephritis
associated thrombotic complications, and patients
with renal involvement of systemic autoimmune diseases PAR1 and 2 auto-antibodies and correlate
autoantibody concentrations with outcome parameters. Isolated patient-IgG is used to study activation
of endothelial cells as well as for the characterization
of target epitopes.
63
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
7. T
he use of antiinflammatory agents for the improvement of early
graft function in marginal renal transplants
Project leader
Coworkers Cooperations Funding T. Florian Fuller, Duska Dragun
Lyubov Chaykovska, Angelika Kusch, Rusan Catar, Jennifer Pützer, Uwe Hoff
J. Troppmair (Innsbruck)
Novartis Pharma
Institutional funding (Universitäre Forschungsförderung Charité)
Due to severe organ shortage, marginal kidneys
from older age donors or with severe preservationreperfusion injury are increasingly used in human
renal transplantation. These organs typically have a
high incidence of delayed graft function (DGF) associated with impaired long-term graft survival. Inflammatory reactions accompanying preservation-reperfusion injury in kidney transplants cause significant
endothelial and tubular damage. Protein-kinase-C
(PKC)- inhibitors have an antiinflammatory effect via
reduction of leukocyte-derived superoxide release in
ischemically damaged organs. We employ our syngeneic and allogeneic rat kidney transplant models
to determine protective capacity of antiinflammatory
(AEB-protein-kinase-C inhibitor). Functional and morphological parameters as well as cellular and molecular markers of inflammation, proliferation and tubular
fibrosis serve as endpoints.
Sotrastaurin has renoprotective
capacity by local modulation of
Erk and p66Shc-activities
64
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
8. C
ytochrome P-450- ( CYP) -derived eicosanoids in acute kidney injury
and acute transplant dysfunction – genetic susceptibilities,
mechanisms and interventions
Project leader
Uwe Hoff, Duska Dragun, Anja Haase-Fielitz, Michael Haase
Coworkers Florian Fuller, Lyubov Chaykovska, Björn Hegner, Sebastian Krause,
Jennifer Pützer, Barbara Kämper, Julian Marinez, Katja Klemz, Melanie Näther
Cooperations
W. H. Schunck, F. C. Luft, M. Gollasch, M. Rother, JR Falck (Dallas), D. N. Müller,
R. Bellomo, M. Oppert and SEPNET
Funding Bundesministerium für Wirtschaft und Technologie,
Alexander von Humboldt-Stiftung,
Deutsche Herzstiftung, Else-Kröner Stiftung, Dr. Werner Jackstädt Stiftung,
Institutional funding (Universitäre Forschungsförderung Charité)
Recent studies indicate that an impaired balance
between production of nitric oxide (NO) and CYPdependent eicosanoids plays a crucial role in the
development of endothelial dysfunction and subsequent tubular injury. In particular, 20-HETE (20-hydroxyeicosatetraenoic acid), which is excessively released
and overproduced during ischemia/reperfusion (I/R)
may be responsible for sustained vasoconstriction
and aggravation of inflammation. Our studies demonstrate that renal injury can be significantly ameliorated by inhibiting the synthesis (HET 0016) and even
more favorably by blocking the action of 20-HETE
(20-HEDE) during the acute phase of ischemia and
reperfusion. The protective action is attributed to antiapoptic, anti-vasoconstrictive and anti-inflammatory
effects due to 20-HETE inhibition. We now extend our
studies on our syngeneic and allogeneic rat kidney
transplant models employing marginal donor kidneys
by means of extended cold preservation times and
increased donor age.
In cooperation with the Max-Delbrück-Center for
Molecular Medicine (MDC), Lipidomix GmbH and the
Deutsche Herzzentrum Berlin (DHZB) we are develop-
ing a system for the measurement of CYP-dependent
eicosanoids in serum and urine. This novel diagnostic tool is intended to be employed as a biomarker
of endothelial dysfunction in recipients of solid organ
Apoptotic tubular cells after 45 minutes of warm ischemia
with and without inhibition of 20-HETE production or 20-HETE
blockade
65
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
transplants (heart, liver, kidney, simultaneous kidneypancreas) to detect sequelae of calcineurin inhibitors
(CNI)-nephrotoxicity. We currently screen serum and
urine of organ transplant recipients for differences in
concentrations of 20-HETE and EETs and correlate
their levels with structural and functional determinants
of CNI mediated toxicity.
Vasodilatory shock and acute kidney injury (AKI) are
associated with an increased mortality after cardiac
surgery and/or during sepsis, the two most common
contributing factors to AKI. We are focused on prediction of the risk for AKI and improvement in individualized patient care by the identification of genetic risk
factors and biomarkers involved in the regulation of the
vascular tone. Circulating and local catecholamines
are primarily catabolized through enzymatic pathways
involving the enzyme catechol-O-methyltransferase
(COMT). We found that cardiac surgery patients who
were homozygous for the low activity COMT L allele
(LL patients) more commonly developed prolonged
vasodilatory shock, AKI, more severe AKI requiring
renal replacement therapy, and prolonged hospital
stay. These patients also had increased concentrations of plasma catecholamine and MAO dependent
catecholamine degradation products that correspond
well in terms of genotype-phenotype correlations. Our
findings provide a conceptual frame for further studies
in vasodilatory shock and AKI in sepsis.
Publications 2006 - 2010
Fuller TF, Hoff U, Rose F, Linde Y, Freise CE, Dragun D, Feng
S (2006): Effect of mycophenolate mofetil on rat kidney grafts
with prolonged cold preservation. Kidney Int 70:570-577
Haase M, Morgera S, Bamberg C, Halle H, Martini S, Dragun
D, Neumayer HH, Budde K (2006): Successful pregnancies in
dialysis patients including those suffering from cystinosis and
familial Mediterranean fever. J Nephrol 19:677-681
Muller G, Catar RA, Niemann B, Barton M, Knels L, Wendel
M, Morawietz H (2006): Upregulation of endothelin receptor
B in human endothelial cells by low-density lipoproteins. Exp
Biol Med (Maywood); 231:766-71
Stielow C, Catar RA, Muller G, Wingler K, Scheurer P,
Schmidt HH, Morawietz H (2006): Novel Nox inhibitor
of oxLDL-induced reactive oxygen species formation in
human endothelial cells. Biochem Biophys Res Commun
26;344:200-5
66
Fuller TF, Rose F, Singleton KD, Linde Y, Hoff U, Freise CE,
Dragun D, Niemann CU (2007): Glutamine donor pretreatment
in rat kidney transplants with severe preservation reperfusion
injury. J Surg Res 140:77-83
Scornik JC, Guerra G, Schold JD, Srinivas TR, Dragun D,
Meier-Kriesche HU (2007): Value of posttransplant antibody
tests in the evaluation of patients with renal graft dysfunction.
Am J Transplant 7:1808-1814
Patecki M, von Schaewen M, Tkachuk S, Jerke U, Dietz R,
Dumler I, Kusch A (2007): Tyk2 mediates effects of urokinase
on human vascular smooth muscle cell growth. Biochem
Biophys Res Commun 359:679-84
Kiyan J, Kusch A*, Tkachuk S, Krämer J, Haller H, Dietz R,
Smith G, Dumler I (2007): Rosuvastatin regulates vascular smooth muscle cell phenotypic modulation in vascular
remodeling: role for the urokinase receptor. Atherosclerosis
195:254-61 (* first co-author)
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Catar RA, Müller G, Heidler J, Schmitz G, Bornstein SR,
Morawietz H (2007): Low-density lipoproteins induce the
renin-angiotensin system and their receptors in human
endothelial cells. Horm Metab Res 39:801-5
Krug AW, Kopprasch S, Ziegler CG, Dippong S, Catar RA,
Bornstein SR, Morawietz H, Gekle M (2007): Aldosterone
rapidly induces leukocyte adhesion to endothelial cells: a new
link between aldosterone and arteriosclerosis? Hyperten­
sion 50:e156-7
Haase M, Haase-Fielitz A, Ratnaike S, Reade MC, Bagshaw
SM, Morgera S, Dragun D, Bellomo R (2008): N-Acetylcysteine does not artifactually lower plasma creatinine concentration. Nephrol Dial Transplant 23:1581-1587
Karatas A, Hegner B, de Windt LJ, Luft FC, Schubert C,
Gross V, Akashi YJ, Gürgen D, Kintscher U, da Costa Goncalves AC, Regitz-Zagrosek V, Dragun D (2008): Deoxycorticosterone acetate-salt mice exhibit blood pressure-independent sexual dimorphism. Hypertension 51:1177-1183
Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz
M, Wabitsch M, Fischer-Posovszky P, Barth TF, Dragun D,
Skurk T, Hauner H, Blüher M, Unger T, Wolf AM, Knippschild
U, Hombach V, Marx N (2008): T-lymphocyte infiltration in
visceral adipose tissue: a primary event in adipose tissue
inflammation and the development of obesity-mediated insulin
resistance. Arterioscler Thromb Vasc Biol 28:1304-1310
Riad A, Jäger S, Sobirey M, Escher F, Yaulema-Riss A, Westermann D, Karatas A, Heimesaat MM, Bereswill S, Dragun D,
Pauschinger M, Schultheiss HP, Tschöpe C (2008): Toll-like
receptor-4 modulates survival by induction of left ventricular
remodeling after myocardial infarction in mice. J Immunol
180:6954-6961
Amico P, Hönger G, Bielmann D, Lutz D, Garzoni D, Steiger
J, Mihatsch MJ, Dragun D, Schaub S (2008): Incidence
and prediction of early antibody-mediated rejection due to
non-human leukocyte antigen-antibodies. Transplantation
85:1557-1563
Krämer J, Ruf RG, Schmidt S, Axthelm C, Strasser R, Janssen G, Thieme T, Kusch A, Waigand J, Dietz R, Gross CM
(2008): The CAIRP (CAndesartan for In-stent Restenosis
Prevention) Trial--a multicenter study of AT1-receptor blocker
therapy in coronary stenting. J Invasive Cardiol 20:205-10
Chaykovska L, Deger S, Wille A, Friedersdorff F, Kasper A,
Dragun D, Liefeldt L, Miller K, Giessing M, Fuller TF (2009):
Kidney transplantation into urinary conduits with ureteroureterostomy between transplant and native ureter: singlecenter experience. Urology 73:380-5
Hegner B, Lange M, Kusch A, Essin K, Sezer O, SchulzeLohoff E, Luft FC, Gollasch M, Dragun D (2009): mTOR
regulates vascular smooth muscle cell differentiation from
human bone marrow-derived mesenchymal progenitors.
Arterioscler Thromb Vasc Biol 29:232-8
Haase M, Haase-Fielitz A, Bellomo R, Devarajan P, Story D,
Matalanis G, Reade MC, Bagshaw SM, Seevanayagam N,
Seevanayagam S, Doolan L, Buxton B, Dragun D (2009):
Sodium bicarbonate to prevent increases in serum creatinine
after cardiac surgery: a pilot double-blind, randomized controlled trial. Crit Care Med 37:39-47
Haase-Fielitz A, Bellomo R, Devarajan P, Story D, Matalanis
G, Dragun D, Haase M (2009): Novel and conventional serum
biomarkers predicting acute kidney injury in adult cardiac surgery -a prospective cohort study. Crit Care Med 37:553-60
Haase-Fielitz A, Haase M, Bellomo R, Lambert G, Matalanis G, Story D, Doolan L, Buxton B, Gutteridge G, Luft FC,
Schunck WH, Dragun D (2009): Decreased catecholamine
degradation associates with shock and kidney injury after
cardiac surgery. J Am Soc Nephrol 20:1393-403
Haase-Fielitz A, Bellomo R, Devarajan P, Bennett M, Story D,
Matalanis G, Frei U, Dragun D, Haase M (2009): The predictive performance of plasma neutrophil gelatinase-associated
lipocalin (NGAL) increases with grade of acute kidney injury.
Nephrol Dial Transplant 24:3349-54
Haase M, Bellomo R, Devarajan P, Ma Q, Bennett MR,
Möckel M, Matalanis G, Dragun D, Haase-Fielitz A (2009):
Novel biomarkers early predict the severity of acute kidney
injury after cardiac surgery in adults. Ann Thorac Surg
88:124-30
Kinkley S, Staege H, Mohrmann G, Rohaly G, Schaub T,
Kremmer E, Winterpacht A, Will H (2009): SPOC1: a novel
PHD-containing protein modulating chromatin structure and
mitotic chromosome condensation. J Cell Sci 122:2946-56
67
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Jerke U, Tkachuk S, Kiyan J, Stepanova V, Kusch A, Hinz M,
Dietz R, Haller H, Fuhrman B, Dumler I. (2009): Stat1 nuclear
translocation by nucleolin upon monocyte differentiation.
PLoS One 14;4:e8302
Kusch A, Tkachuk S, Tkachuk N, Patecki M, Park JK, Dietz
R, Haller H, Dumler I (2009): The tight junction protein ZO-2
mediates proliferation of vascular smooth muscle cells via
regulation of Stat1. Cardiovasc Res 1;83:115-22
Haase-Fielitz A, Bellomo R, Devarajan P, Dragun D, Haase M
(2010): The predictive performance of neutrophil gelatinaseassociated lipocalin (NGAL) increases with severity of acute
kidney injury. Am J Kidney Dis 24:3349-54
Ksi K, Mikusa-Pietrasik J, Catar R, Dworacki G, Winckiewicz M, Frydrychowicz M, Dragun D, Ryszard S, Jörres A,
Witowski J (2010): Oxidative stress-dependent increase in
ICAM-1 expression promotes adhesion of colorectal and
pancreatic cancers to the senescent peritoneal mesothelium.
International Journal of Cancer 127:293-303
Zebger-Gong H, Kampmann J, Kong L, Roigas J, Sommer K,
Hoff U, Krämer S, Peters H, Müller D, Dragun D, Querfeld U
(2010): Decreased transplant arteriosclerosis in endothelial nitric
oxide synthase-deficient mice. Transplantation 89:518-26
Duerr M, Glander P, Diekmann F, Dragun D, Neumayer HH,
Budde K (2010): Increased incidence of angioedema with
ACE inhibitors in combination with mTOR inhibitors in kidney
transplant recipients. Clin J Am Soc Nephrol 5:703-8
Fuller TF, Hoff U, Kong L, Naether M, Wagner P, NieminenKelhä M, Nolting J, Luft FC, Hegner B, Dragun D (2010):
Cytoprotective actions of FTY720 modulate severe preservation reperfusion injury in rat renal transplants. Transplanta­
tion 89:402-8
Hoff U, Lukitsch I, Chaykovska L, Ladwig M, Arnold C, Manthati VL, Fuller TF, Schneider W, Gollasch M, Muller DN, Flemming B, Seeliger E, Luft FC, Falck JR, Dragun D, Schunck WH
(2010): Inhibition of 20-HETE synthesis and action protects
from renal ischemia/reperfusion injury. Kidney Int in press
68
Reinsmoen NL, Lai CH, Heidecke H, Haas M, Cao K, Ong G,
Naim M, Wang Q, Mirocha J, Kahwaji J, Vo AA, Jordan SC,
Dragun D (2010): Anti-Angiotensin Type 1 Receptor Antibodies Associated with Antibody Mediated Rejection in Donor
HLA Antibody Negative Patients. Transplantation in press
Riemekasten G, Philippe A, Näther M, Slowinski T, Müller DN,
Heidecke H, Matucci-Cerinic M, Czirják L, Lukitsch I, Becker
M, Kill A, van Laar JM, Catar R, Luft FC, R. Burmester GR,
Hegner B, Dragun D (2010): Involvement of functional autoantibodies against vascular receptors in systemic sclerosis. Ann
Rheum Dis in press
Reviews & book chapters (2006-20010)
Fuller TF, Liefeldt L, Dragun D, Tüllmann M, Loening SA,
Giessing M (2006): Urological evaluation and follow-up of the
kidney transplant patient. Urologe 45:53-59
Dragun D (2006): Complementary matching. A definite
maybe. Nephrol Dial Transplant 21:3371-3373. Editorial
Dragun D, Hegner B (2006): "FTY720 - Mechanism of
Action of a Novel Immune Modulator: Potential Applica­
tion for Autoimmune Diseases", 1. Auflage, Herausgeber:
T. Böhler,UNI-MED Verlag AG Bremen, Kapitel: FTY protection against ischemia-reperfusion damage
Dragun D (2007): The role of angiotensin II type 1 receptoractivating antibodies in renal allograft vascular rejection.
Pediatr Nephrol 22:911-914
Haase-Fielitz A, Haase M, Bellomo R, Dragun D (2007): Genetic
polymorphisms in sepsis- and cardiopulmonary bypass-associated acute kidney injury. Contrib Nephrol 156:56-91
Dragun D (2007): Agonistic antibody-triggered stimulation
of Angiotensin II type 1 receptor and renal allograft vascular
pathology. Nephrol Dial Transplant 22:1819-22
Riemekasten G, Dragun D (2007): Clinical risk-adapted
Therapies in systemic sclerosis. Z Rheumatol 66:672-674,
676-678
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Dragun D, Rudolph B (2007): Novelties in diagnostics and
therapy of antibody mediated rejection. Nephrol Dial Trans­
plant 22Suppl 8:viii50-viii53
Dragun D, Haase-Fielitz A (2009): Low catechol-O-methyltransferase and 2-methoxyestradiol in preeclampsia: more
than a unifying hypothesis. Nephrol Dial Transplant 24:31-3
Dragun D (2008): Humoral responses directed against nonHLA antigens in solid-organ transplantation. Transplantation
86:1019-25
Schewior L, Dragun D, Schaeffner E (2009): The challenge
of Wegener’s granulomatosis after kidney transplantation.
Transpl Int 22:503-5
Dragun D, Scornik J, Meier-Kriesche HU (2008): Kidneytransplant rejection and anti-MICA antibodies. N Engl J Med
358:196. Letter
Dragun D, Distler JH, Riemekasten G, Distler O 2009): Stimulatory autoantibodies to platelet-derived growth factor receptors in systemic sclerosis: what functional autoimmunity could
learn from receptor biology. Arthritis Rheum 60:907-11
Dragun D (2008): Cellular responses involved in pathogenesis
of chronic allograft nephropathy. International Transplanta­
tion Updates, Herausgeber: J. Grinyo, Permanyer Publications Barcelona, Kapitel: Late renal allograft dysfunction
Dragun D, Hegner B (2008): "Neue Medikamente in der
Transplantationsmedizin", 2. Auflage,Herausgeber: H.-H.
Neumayer, K. Budde, J. Waiser, UNI-MED Verlag AG Bremen,
2008 Kapitel: Antikörper vermittelte Allograftabstoßung Humorale Rejektion
Kusch A. (2008): Linking proteolysis to lipids. Thromb Res
123:191-3
Dragun D, Hegner B (2009): Non-HLA antibodies posttransplantation: clinical relevance and treatment in solid organ
transplantation. Contrib Nephrol 162:129-39
Dragun D, Philippe A, Catar R, Hegner B (2009): Autoimmune
mediated G-protein receptor activation in cardiovascular and
renal pathologies. Thromb Haemost101:643-8
Dragun D, Fielitz-Haase A (2009): Low catechol-O-methytransferase and 2-methoxyestradiol in preeclampsia: More
than a unifying hypothesis. Nephrol Dial Transplant 24:313. Editorial
Dragun D (2009): "Management of acute kidney prob­
lems", Herausgeber: A. Jörres, Springer Verlag Berlin-Heidelberg, Kapitel: Acute kidney failure during pregnancy and
postpartum.
Dragun D, Philippe A (2010): From mother to child--transplacental effect of AT1R-AA in preeclampsia. Nephrol Dial
Transplant 25:1774-6.
69
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Näther M.
Dragun D.
Dragun D.
Catar R.
Philippe A.
Hegner B.
Dragun D.
Hegner B.
Philippe A.
Dragun D.
Catar R.
Näther M.
Chaykovska L.
Dragun D.
Hegner B.
Kusch A.
Haase M.
Haase-Fielitz A.
Dragun D.
Haase M.
Haase-Fielitz A.
Dragun D.
Dragun D.
Fuller F.
Chaykovska L.
Kusch A.
Hoff U.
Hegner B.
Dragun D.
Dragun D.
Kämper B.
Hegner B.
Hoff U.
70
Project title
Endothelial mecha-nisms in thrombotic
microangiopathy induced acute renal
failure
Autoimmune mediated endothelin
receptor activation in renal crisis
Sponsor
Dr. Werner Jackstädt Foundation
Period
2010–2011
Actelion Pharmaceuticals Deutschland GmbH
2009–2010
SysKID (Systems biology towards novel
chronic kidney disease diagnosis and
treatment)
Functional characterization of CXCR3
and CXCR4 auto-antibodies
EU: Large-scale integrating project 2010–2012
(EU)-FP7
BMWi (Federal Ministry of Economy and Technology)
2009–2011
Sex differences in adaptive and maladaptive myocardial hypertrophy – role
of the Akt/mTOR pathway
Multi center trial of sodium bicarbonate
to prevent acute renal failure in patients
undergoing cardiopulmonary surgery
Interventions to prevent acute renal
failure in patients undergoing cardiopulmonary surgery
Effect of the protein-kinase-C-inhibitor
AEB on rat kidney transplants with
severe preservation-reperfusion injury
DFG (German Research Foundation) DR 498/1-1 – FG1054
(Research group 1054)
Else Kröner Fresenius Foundation
2008–2011
German Heart Foundation
2008–2010
Novartis Pharma GmbH
2008–2009
Mesenchymal stem cells in systemic
sclerosis
CYP-Eicosanoid status after organ
transplantation
Deutsche Stiftung Sklerodermie
German Scleroderma Foundation
BMWi (Federal Ministry of Economy and Technology)
2008–2009
2008–2010
2008–2009
Duska Dragun – NEPHROLOGY AND CARDIOVASCULAR IMMUNOLOGY
Dragun D.
Catar R.
Hoff U.
Näther M.
Dragun D.
Hegner B.
Gürgen D.
Dragun D.
Hegner B.
Dragun D.
Philippe A.
Hegner B.
Catar R.
Hoff U.
Dragun D.
Relevance of autoantibodies against
BMWi (Federal Ministry of EconPAR-1 and PAR-2 receptors in renal
omy and Technology)
transplant patients and kidney diseases
2007–2009
Sex and gender specific mechanisms
of myocardial hypertrophy
DFG (German Research Foundation) –Graduiertenkolleg 754-3
(Graduate course 754-3)
German Scleroderma Foundation
2006–2010
EFRE (European Foundation for
Regional Development)
2006–2007
Dr. Werner Jackstädt Foundation
2006–2007
Kidney involvement in systemic sclerosis
Agonistic Autoantibodies
Gender aspects in acute kidney injury
2006–2008
Awards
2010 – 2011
2009 – 2010
2009
2009
2009
2009
2008
2006 – 2008
2006 – 2007
2007
2006 – 2007
2006
2006
2006
2005
2005
2005
2005
Dr. Werner Jackstädt Fellowship
Dr. Werner Jackstädt Fellowship
Bernd-Schönberger-Preis –Stiftung Urologische Forschung Poster award International Society of Gender in Medicine
Nils-Alwall-Preis – DAGKN
Poster award – DGRh
Young investigator award – ERA-EDTA
Rachel Hirsch Fellowship
Alexander-von-Humboldt Fellowship
"New key opinion leader" - WTC
Dr. Werner Jackstädt Fellowship
Theodor-Frerichs-Preis – DGIM
Charité Allianz Research Award European Students Conference
Poster of distinction – World transplant congress
Young Investigator Award – ATC
Poster of distinction – ATC
Apherese Innovationspreis – DAfKN
Biotest Award Clinical Science – ESOT
Melanie Näther
Anja Fielitz-Haase
Lyubov Chaykovska
Angelika Kusch
Duska Dragun, Michael Haase
Björn Hegner
Michael Haase
Angelika Kusch
Michael Haase
Duska Dragun
Uwe Hoff
Duska Dragun
Lyubov Chaykovska
Björn Hegner
Duska Dragun
Uwe Hoff
Duska Dragun
Björn Hegner
71
Berthold Hocher – Pharmacology/Nephrology
Head of the Group
Prof. Dr. med. Berthold Hocher
Curriculum Vitae: Prof. Dr. med. Berthold Hocher studied veterinary medicine
and medicine at the Free University of Berlin and at the University of Heidelberg.He
started his research career at the Department of Molecular Biology and Biochemistry
of the Free University of Berlin where he also accomplished his doctoral thesis. In the
following years he worked clinically in the field of Internal Medicine with the focus on
Nephrology. After clinical posts at the Benjamin Franklin University Hospital of the
Free University of Berlin and at the Charité, he became a Consultant Nephrologist and Associated Professor of
Internal Medicine at the University Hospital Bern/Switzerland. He is currently Translational Medicine Leader at
Roche in Basel/Switzerland, holds a full professorship at the University in Potsdam and is guest scientist at the
CCR.His research group in Berlin is associated to the Department of Pharmacology/CCR of the Charité. The
main topics of his research group are the nitric oxide/endothelin system in renal and cardiovascular diseases
and fetal programming of renal and cardiovascular diseases.
Members of the group
Scientists
Alter, Markus
Chen, Youpeng
Godes, Michael
Kalk, Philipp
Krause-Relle, Katharina
Ott, Ina
Dr. med.
Dr. med.
Dr. med.
Dr. med.
Dr. rer. medic.
Nutritionist
Pfab, Thiemo
Rahnenführer, Jan
Sharkovska, Yuliya
Simon, Alexandra
Websky, Karoline von
Priv-Doz. Dr. med.
Biotechnologist
Dr. med.
Nutritionist
Veterinarian
73
Berthold Hocher – Pharmacology/Nephrology
Students
Andermann, Tim
Blancke Jan-Arne
Dowuona-Hammond, Ekua
Einem, Gina Franziska von
Gorodetski, Jenny
Haumann, Hannah
Heunisch, Fabian
Hohmeier, Sophie
Hübner, Sarah
Kempiners, Nina
Köder, Ina
Krause, Christian
Kyeyamwa, Sarah
Leonhardt, Claudia
74
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Medical student
Gynaecologist
Medical student
Mengeringhausen, Eva
Mersmann, Astrid
Pursche, Lars
Reichetzeder, Christoph
Runge, Silke
Stange, Dörte
Sceplik, Ewelina
Schlemm, Ludwig
Schwietzer, Marcel
Sievers, Gerlind
Tsuprykov, Oleg
Volochko, Evgeny
Weist, Andreas
Ziegner, Maja
Medical student
Medical student
Medical student
Physician
Medical student
Master student
Medical student
Medical student
Medical student
Medical student
PhD student, Physician
Medical student
Physician
Medical student
Berthold Hocher – Pharmacology/Nephrology
Summary
Our research is focused on four major topics:
1. The impact of the endothelin (ET) system on
the pathogenesis of cardiac and renal disease
We established and characterized ET-1 transgenic
mice and analyzed the impact of the ET system in
cardiovascular disease models. Current projects
are focusing on the interaction of the ET – and nitric
oxide system. We thus generated ET-1 transgenic
mice lacking the endothelial NOS in order to further
characterize the complex physiology of the ET-NOSystem in vivo. Other projects are focussing on the
impact of the ETB receptor on tubular anion transporters as well as the impact of the ETB receptor for
the pathogenesis of diabetic nephropathy.
2. Fetal programming of cardiovascular disease
There is meanwhile clear evidence indicating that early
life events play an important role in the pathogenesis of cardiovascular diseases in later life. We have
introduced the concept of maternal genes exerting
unfavorable effects on the offspring into this currently
rapidly growing research field. We were furthermore
able to demonstrate for the first time that insulin resistance may also be present in otherwise healthy human
newborns already at birth. Our animal studies demonstrated that also over-nutrition may exert harmful
effects on the offspring in a gender dependent manner.
Current clinical and preclinical studies are designed
to detect molecular pathways of fetal programming
including epigenetic DNA modifications.
3. Diabetes mellitus and diabetic nephropathy
As the prevalence of diabetes mellitus and its complications, such as diabetic nephropathy, rises in the western
industrial nations, the severe consequences become
more and more present: Diabetic nephropathy is
accountable for one third of all patients who require reg-
ular dialysis – with upward trend. Another important risk
factor is hypertension. Thus, we test new drug classes
in experiments with rats and mice. Mainly, we use the
hyperglycaemia model induced by streptozotocin and
hypertension models induced either by renal arterial
stenosis by surgery or by genetical knock out of the
endothelial NO synthase (eNOS knock out). Of course,
we combine the models as well to simulate the pathogenesis of diabetic nephropathy as close as possible.
So we can analyse all common renal and cardiovascular biomarkers in plasma and urine and round research
off by histological and immunohistological evaluation
of kidneys, hearts and further vital organs. Results are
compared with already established treatment regimes
such as AT2 receptor blockers or ACE inhibitors.
4. Detection of biomarkers – German
Radiocontrast Media Study
The increasing incidence of CIN is one of the major
challenges which nephrologists are facing worldwide.
Mortality is over one third of the in-hospital acute renal
failure after contrast media (CM) administration and
affects especially patients with diabetes mellitus and
impaired renal function. Therefore the prevention of
CM nephropathy is highly relevant to clinical outcomes. In collaboration with the Clinic for Cardiology at the Charité Campus Mitte we investigate the
course and outcome of a huge number of patients
who undergo coronary angiography and receive CM
application. In addition, we collect blood and urine
samples during the course. This material is to be analysed by surface-enhanced laser desorption ionization
time-of-flight mass spectrometry (SELDI-TOF MS) and
results are to be compared with clinical data. The goal
with this open, non-hypothesis driven approach is to
detect new biomarkers in blood and urine which are
able to predict the incidence, course and outcome of
contrast media induced nephropathy.
75
Berthold Hocher – Pharmacology/Nephrology
Zusammenfassung
Unsere Arbeitsgruppe beschäftigt sich
schwerpunktmäßig mit vier Hauptthemen:
1. Der Einfluss des Endothelinsystems auf die
Pathogenese kardialer und renaler Erkrankungen
Wir untersuchen den Einfluss des Endothelinsystems
auf kardiovaskuläre Erkrankungen anhand der von uns
etablierten ET-1 transgenen Mauslinie. Um die Interaktion zwischen Endothelin- und NO-System zu erforschen, wurde die ET-1 transgene Linie mit einer eNOS
(endotheliale NO-Synthase) Knockout Linie gekreuzt.
Ein weiterer Schwerpunkt liegt auf der Untersuchung
der tubulären ETB-Rezeptoren in der Niere und ihrer
Wirkung auf Anionen-Transporter bzw. ihre Rolle in der
Pathogenese der diabetischen Nephropathie.
2. Fetale Programmierung kardiovaskulärer
Erkrankungen
Es gibt klare Hinweise, die belegen, dass bestimmte
Einflüsse im frühen Leben prädispositionierend sind für
kardiovaskuläre Erkrankungen im weiteren Lebensverlauf. Wir konnten als Erste zeigen, dass Insulinresistenz
schon bei der Geburt ansonsten gesunder Neugeborener nachweisbar sein kann. Anhand unserer Tierversuche konnten wir darlegen, dass eine Überernährung
der Mutter geschlechtsabhängige Auswirkungen auf die
Nachkommen hat. Ein Teil unserer laufenden Projekte
beschäftigt sich damit, die molekularen Signalwege der
fetale Programmierung aufzudecken.
3. Diabetes mellitus und diabetische Nephropathie
Der Diabetes mellitus und damit auch seine Spätkomplikationen wie die diabetische Nephropathie zeigen eine zunehmende Prävalenz in den westlichen
Industrienationen mit ernstzunehmenden Folgen:
Inzwischen ist bei gut einem Drittel aller Dialysepatienten der Diabetes mellitus ursächlich für die terminale Niereninsuffizienz – mit steigender Tendenz. Als
weiterer entscheidender Risikofaktor soll die Hyperto76
nie genannt werden. Wir erproben neue Medikamentenklassen im Ratten- und Mäuse-Tierversuch. Dabei
arbeiten wir hauptsächlich, einzeln oder in Kombination, mit dem Streptozotocin-induzierten Hyperglykämiemodell und Hypertoniemodellen, die durch
operatives Setzen einer Nierenarterienstenose (2K1C)
oder genetischem Knockout der endothelialen NOSythase (eNOS) induziert werden. Neben Untersuchungen der gängigen renalen und kardiovaskulären
Biomarker erfolgt die histologische und immunhistologische Aufarbeitung von Herz, Nieren und weiteren
lebenswichtigen Organen. Die Ergebnisse werden mit
den aktuell gängigen Therapiemöglichkeiten (zumeist
Sartane und ACE-Hemmer) verglichen.
4. Ermittlung von Biomarkern – Deutsche
Radiokontrastmittel-Studie
Die Inzidenz des kontrastmittelinduzierten Nierenversagens steigt stetig und ist mittlerweile die dritthäufigste
Ursache des akuten Nierenversagens. Das kontrastmittelinduzierte Nierenversagen ist insbesondere bei
Patienten mit Diabetes mellitus und vorbestehender
Einschränkung der Nierenfunktion mit einer hohen
Mortalität verbunden. Vor diesem Hintergrund hat die
Prävention eines kontrastmittelinduzierten Nierenversagens einen hohen Stellenwert für die Prognose der Patienten. Wir führen in Zusammenarbeit mit der Klinik für
Kardiologie am Campus Mitte der Charité einen Studie
durch, in der wir den klinischen Verlauf von einer hohen
Zahl von Patienten, die sich zur Koronarangiographie
vorstellen und dabei Radiokontrastmittel appliziert
bekommen, verfolgen und dabei Blut- und Urinproben
sammeln. Dieses Material soll einer Proteomanalyse
mittels SELDI-TOF-Massenspektrometrie zugeführt
und mit den klinischen Daten verglichen werden. Ziel
ist es, neue Biomarker zu ermitteln, die imstande sind,
das Auftreten und die Prognose eines kontrastmittelinduzierten Nierenversagens vorherzusagen.
Berthold Hocher – Pharmacology/Nephrology
Research Projects
1. N
itric oxide-independent stimulation of soluble guanylate cyclase
reduces organ damage in experimental low-renin and high-renin models
Project leader
Coworkers
Collaborations Berthold Hocher
Yuliya Sharkovska, Philipp Kalk, Wellkisch K, Michael Godes, Katharina Krause-Relle
Prof. Dr. Johannes-Peter Stasch, Dr. Bettina Lawrenz, Linda Sarah Hoffmann,
Sandra Geschka, Bayer Schering Pharma AG, Cardiovascular Research,
Martin-Luther-University, Halle, Germany
The nitric oxide-soluble guanylate cyclase (sGC)-cGMP
signal transduction pathway is impaired in different cardiovascular diseases, including pulmonary hypertension,
heart failure and arterial hypertension. Riociguat is a novel
stimulator of soluble guanylate cyclase (sGC). However,
little is known about the effects of sGC stimulators in
experimental models of hypertension. We thus investigated the cardio-renal protective effects of riociguat in
low-renin and high-renin rat models of hypertension.
The vasorelaxant effect of riociguat was tested in vitro on
isolated saphenous artery rings of normal and nitrate tolerant rabbits. The cardiovascular in-vivo effects of sGC stimulation were evaluated in hypertensive renin-transgenic
rats treated with the nitric oxide-synthase inhibitor N-nitroL-arginine methyl ester (L-NAME) (high-renin model) and
in rats with 5/6 nephrectomy (low-renin model).
In both animal models, riociguat treatment improved
survival and normalized blood pressure. Moreover,
in the L-NAME study part, riociguat reduced cardiac
target organ damage as indicated by lower plasma
ANP, lower relative left ventricular weight and lower
cardiac interstitial fibrosis, and reduced renal target
organ damage as indicated by lower plasma creatinine and urea, less glomerulosclerosis (Fig. 1) and less
renal interstitial fibrosis. In the 5/6 nephrectomy study
part, riociguat reduced cardiac target organ damage
as indicated by lower plasma ANP, lower relative left
ventricular weight, lower myocyte diameter and lower
arterial media/lumen ratio, and reduced renal target
organ damage as indicated by improved creatinine
clearance (Fig. 2) and less renal interstitial fibrosis.
Our study demonstrate for the first time that the novel
sGC stimulator riociguat shows in two independent
models of hypertension a potent protection against
cardiac and renal target organ damage.
Glomerulosclerosis in renin-transgenic rats under L-NAME
administration (controls) with riociguat treatment at 3 mg/kg
(riociguat low dose) or 10 mg/kg (riociguat high dose). Values
are given as means ± SEM; *: p< 0.01 vs. control.
Creatinine clearance during
the 5/6 nephrectomy (NX)
study was calculated from
blood and urine creatinine.
Values are given as means ±
SEM; ** : p< 0.001 vs. ShamOP, † : p< 0.05 vs. 5/6 NX.
77
Berthold Hocher – Pharmacology/Nephrology
2. N
ew evidence for the fetal insulin hypothesis : fetal
angiotensinogen M235T polymophism is aasociated with birth
weight and elevated fetal total glycated hemoglobin at birth
Project leader
Berthold Hocher
CoworkersLudwig Schlemm, Hannah M. Haumann, Maja Ziegner, Bulza Stirnberg,
Andreas Sohn, Markus Alter, Thiemo Pfab
Collaboration
PD Dr. med. Florian Guthmann, Clinic for Neonatology, Charité, Berlin, Germany
PD Dr. med. Karim Kalache, Department of Obstetrics and Gynecology, Charité, Berlin
Low birth weight is associated with an increased risk
of cardiovascular events in later life. Insulin resistance
is a key finding in adult patients with cardiovascular
diseases. The neonatal phenotype of an individual with
insulin resistance might be low birth weight, as insulin
influences fetal growth. The renin–angiotensin–aldosterone system has been associated with cardiovascular
disease and insulin resistance. We analyzed whether
fetal polymorphisms of the angiotensinogen (AGT)
and angiotensin-converting enzyme genes influence
birth weight and/or fetal total glycated hemoglobin
(fTGH), a surrogate parameter of fetal insulin resistance
at birth. In 1132 white women delivering singletons,
neonatal umbilical blood samples and clinical data of
the mothers and newborns were obtained. Newborns
were genotyped with respect to the AGT M235T and
angiotensin-converting enzyme insertion/deletion
polymorphism. The AGT M235T TT polymorphism
is associated with reduced birth weight (TT: 3288 g
versus TM+MM: 3435 g, P<0.05). Furthermore, newborns with a high percentage of fTGH (>6.5%) are more
likely to have the TT genotype than those with normal
fTGH (<=6.5%, P<0.05). With higher cutoffs for fTGH,
the significance increases to P less than 0.005. No
association was seen between these parameters and
the fetal angiotensin-converting enzyme insertion/deletion phenotype. The fetal AGT M235T polymorphism
is associated with low birth weight and elevated fetal
fTGH at birth. Previous findings show that elevated fetal
fTGH correlates with low birth weight and that higher
activity of the renin–angiotensin–aldosterone system is
an independent risk factor for the development of diabetes mellitus and coronary artery disease. Therefore,
our data are supportive of the fetal insulin hypothesis.
Newborns carrying the TT genotype are more
likely to be SGA and less likely to be LGA.
TT-homozygosity is most often found in babies
SGA, and least likely in those who are LGA
(Padj =0.01). AGA, appropriate for gestational
age; AGT, angiotensinogen; LGA, large for gestational age; SGA, small for gestational age.
78
Berthold Hocher – Pharmacology/Nephrology
3. F
etal sex determines the impact of maternal progins
and PPARgamma2 Pro12Ala polymorphism on maternal
physiology during pregnancy
Project leader
Berthold Hocher, Thiemo Pfab
Coworkers
Ludwig Schlemm, Hannah Haumann, C. Poralla, Aline Burdack, Michael Godes
CollaborationDr. Youpeng Chen, Univeristy of Goughzou, China, Prof. Dr. med. Horst Halle,
Dr. med. Christian Bamberg, Prof. PD Dr. med. Karim Kalache, Department of
Obstetrics and Gynecology, Charité, Berlin; PD Dr. med. Florian Guthmann,
Clinic for Neonatology, Charité, Berlin, Germany
It was suggested that fetal sex may substantially
affect maternal glycemic control during pregnancy
in genetically susceptible mothers. We tested the
hypothesis that fetal sex as a major genetic variant
of the fetal genome may affect maternal physiology
during pregnancy in genetically susceptible pregnant women.
The peroxisome proliferator-activated receptor
gamma2 (PPARgamma2) Pro12Ala polymorphism
is known to affect glycemic control and may act in
a sex-specific manner. We analyzed the impact of
fetal sex on maternal glycemic control during pregnancy in relation to the maternal PPARgamma2
Pro12Ala polymorphism. Over 2000 Caucasian
women without preexisting diabetes and preexisting
hypertension with singleton pregnancies delivering
consecutively at the Charité obstetrics department
were genotyped. Glycemic control was analyzed by
measuring total glycated hemoglobin at birth. The
maternal PPARgamma2 Pro12Ala polymorphism
without consideration of fetal sex had no effect on
blood pressure, new onset of proteinuria and total
glycated hemoglobin at delivery. Mothers carrying
both G alleles (GG genotype) delivering a girl had a
higher total glycated hemoglobin (6.8+/-0.5%) ver-
sus mothers carrying the same alleles but delivering
boys (5.8+/-0.6%; p<0.05; see Figure). Comparing
mothers with the GG genotype delivering girls with
mothers with CC or CG genotypes also delivering
girls (6.3+/-0.7%) revealed a significantly higher
maternal total glycated hemoglobin at delivery in
the former group (p<0.01, see Figure).
We also analyzed the impact of fetal sex on maternal physiology during pregnancy in relationship with
the maternal PROGINS progesterone receptor gene
polymorphism. The maternal PROGINS progesterone receptor polymorphism on its own had no effect
on blood pressure, new onset of proteinuria, and
total glycated hemoglobin at delivery. However, by
considering the offspring’s sex, the AA variant of
the PROGINS progesterone receptor polymorphism
was associated with profound cardiovascular/metabolic effects; mothers carrying both A alleles (AA
genotype) delivering a boy had significantly lower
systolic blood pressure during the first trimester
of pregnancy versus AA mothers delivering girls
(107.9+/-10.2 vs. 116.6+/-15.1 mmHg; p<0.05).
Diastolic blood pressure was similarly lower during
the first trimester of pregnant AA women delivering
boys in comparison with AA women delivering girls
79
Berthold Hocher – Pharmacology/Nephrology
(63.4+/-5.7 vs. 68.2+/-10.9 mmHg; p<0.05). Total
glycated hemoglobin at delivery was significantly
higher in AA mothers delivering boys (6.6+/-0.7%)
versus AA mothers delivering girls (5.9+/-0.6%;
p<0.01).
Our studies indicate that fetal sex may substantially
affect maternal blood pressure as well as glycemic
control during pregnancy in genetically susceptible
mothers.
Low birth weight, a risk factor for cardiovascular diseases in later life, is already associated with elevated
fetal glycosylated hemoglobin at birth.
80
Berthold Hocher – Pharmacology/Nephrology
4. Impact of maternal body mass index on neonatal outcome
Project leader
Berthold Hocher, Philipp Kalk
Coworkers
Katharina Relle, Kathrin Krause, Michael Godes
CollaborationGabriele Gossing, Horst Halle, Department of Gynaecology and Obstetrics,
Charité, Campus Mitte, Florian Guthmann, Roland Wauer,
Department of Neonatology, Charité, Campus Mitte
Maternal body mass index (BMI) has an impact on
maternal and fetal pregnancy outcome. An increased
maternal BMI is known to be associated with more
frequent admission of the newborn to a neonatal care
unit. The reasons and impact of this admission on fetal
outcome, however, remain to be elucidated.
Thus, the aim of our study was to investigate the
impact of maternal BMI on maternal and fetal pregnancy outcome with special focus on the subgroup of
children admitted to a specialized neonatal care unit.
A cohort of 2049 non-diabetic mothers giving birth in
the Charite university hospital was prospectively studied. Mothers and children were grouped according to
maternal BMI. The impact of maternal BMI on maternal and fetal outcome parameters was tested using
multivariate regression analysis. Outcome of children
admitted to a specialized neonatal ward (n = 505) was
analysed using chi² and t-test while the children were
grouped according to maternal BMI.
We found that increased maternal BMI was associated with an increased risk for hypertensive complications, peripheral edema, caesarean section, fetal
macrosomia and increased admission of the newborn
to a neonatal care unit, whereas decreased BMI was
associated with preterm birth and lower birthweight
in the complete study population. In the subgroup of
children admitted to a neonatal ward children from
obese mothers were characterized by hypoglycaemia.
They need less oxygen, and exhibit a shorter stay on
the neonatal ward compared to children from normal
weight mothers, whereas children from underweight
mothers are characterized by lower umbilical blood
pH and increased incidence of death corresponding
to increased prevalence of preterm birth.
Taken our results together we conclude that pregnancy outcome is worst in babies from mothers with
low body mass index as compared to healthy weight
mothers with respect to increased incidence of preterm birth, lower birth weight and increased neonate
mortality on the neonatal ward. We demonstrate that
the increased risk for neonatal admission in children
from obese mothers does not necessarily indicate
severe fetal impairment as in our population it is mostly
due to surveillance because of hypoglycaemia.
81
Berthold Hocher – Pharmacology/Nephrology
5. C
ell-type specific interaction of endothelin and the nitric oxide
system : pattern of prepro-ET-1 expression in kidneys of L-NAME
treated prepro-ET-1 promoter-lacZ-transgenic mice.
Project leaders
Coworkers
Collaboration
Berthold Hocher, Philipp Kalk, Franz Theuring
Maren Christian. Katharina Relle, Michael Godes
Heiko Funke-Kaiser, AG Unger, CCR
Torsten Slowinski, Hans-H. Neumayer, Department of Nephrology, Charité,
Campus Mitte; Christian Bauer, Department of Biochemistry, Free University, Berlin
Fred Schmager, SCHERING AG, now BAYER
Nitric oxide (NO) and endothelin-1 (ET-1) are known to
play a major role in renal and vascular pathophysiology
and exhibit a close interaction with ET-1 stimulating NO
production and NO in turn inhibiting ET-1 expression.
Our objectives were (1) to establish a novel transgenic
mouse model facilitating ET-1 expression assessment
in vivo, (2) to validate this model by assessing preproET-1 promoter activity in mice embryos by means of
our novel model and comparing expression sites to
well-established data on ET-1 in fetal development and
(3) to investigate renal ET-NO interaction by assessing
prepro-ET-1 promoter activity in different structures of
the renal cortex in the setting of blocked NO synthases
via L-NAME administration.
We established transgenic mice carrying a lacZ
reporter gene under control of the human prepro-ET-1
gene promoter sequence (8 kb of 5’ sequences). BluoGal staining of tissue sections revealed intracellular
blue particles as indicators of prepro-ET-1 promoter
activity. In mouse embryos, we detected high preproET-1 promoter activity in the craniofacial region, as well
as in bone and cartilage consistent with the literature.
82
In order to investigate the interaction of ET-1 and NO
in the kidney in vivo, transgenic mice at the age of 3-4
months were treated with a single dose of the NO
synthase inhibitor L-NAME (25 mg (kg bw)(-1) i.p.) 12 h
before kidney removal. Bluo-Gal staining of kidney sections revealed intracellular blue particles as indicators
of prepro-ET-1 promoter activity in tubular and vascular endothelium and glomerular cells. Particle count
was closely correlated to kidney tissue ET-1 content
(R=0.918, P<0.001). Comparison of counts revealed
an increase by 135+/-53% in L-NAME treated (n=12)
compared to non-treated mice (n=10, P=0.001). Celltype specific evaluation revealed an increase of 136+/51% in tubular (P=0.001) and 105+/-41% in glomerular
cells (P=0.046), but no significant increase in vascular
endothelium.
We thus conclude that we indeed a close interaction
of renal endothelin and renal NO system is present,
but the interaction is modulated in a cell-type specific
manner. Furthermore, our new transgenic model provides a unique opportunity to analyse regulation of the
ET system on a cellular level in vivo.
Berthold Hocher – Pharmacology/Nephrology
6. L
ow birth weight – a risk for cardiovascular diseases in
later life – is associated with elevated fetal glycated
hemoglobin already at birth
Project leader
Coworkers
Collaboration
Berthold Hocher, Thiemo Pfab
Torsten Slowinski, Michael Godes
Friedrich Priem, Institute of Laboratory Medicine, Charité, Berlin
Horst Halle, Department of Obstetrics and Gynecology, Charité, Berlin
Several epidemiological studies have confirmed the
intriguing finding that low birth weight is an independent risk factor for glucose intolerance and type 2
diabetes mellitus, as well as cardiovascular disease.
Insulin resistance probably plays an important role;
because it is one of the major underlying causes of glucose intolerance, and it is multifaceted consequences
include dyslipidemia, hypertension, hypercoagulation,
and impaired fibrinolysis.
We quantified total glycosylated hemoglobin (TGH) at
delivery in 1295 mother/child pairs serving as a surrogate of maternal and fetal glycemia. Multivariable
regression analyses considering gestational age al
delivery, the child´s sex, maternal body mass index,
and smoking during pregnancy revealed that an
increase in TGH by 1% in the child was significantly
associated with a mean birth weight reduction of 135g
(p<0.0001), whereas the same increase in the mother
was associated with a mean birth weight increase of
88g (p<0.0001). The ratio of fetal/maternal TGH suggests that lighter newborns have a higher percentage
of TGH than would be expected from maternal TGH.
The study demonstrates for the first time in a large
population that there is an inverse association between
TGH of a newborn and its birth weight. This might be
due to increased insulin resistance in newborns with
lower birth weight. Our data suggest that the pathophysiological mechanism linking prenatal growth and
postnatal sensitivity to insulin are present as early as
before birth.
83
Berthold Hocher – Pharmacology/Nephrology
Birth weight (in kilograms) according
to TGH of child, mother, and its ratio. A
through C, Scatterplots with regression line. D through F, Six groups of
equal size (≈200 cases per group)
were formed for TGH of child, mother,
and its ratio with low to high values (1–
6) from left to right. Data are given as
mean ± SEM. Correlation coefficients
and probability values are given.
84
Berthold Hocher – Pharmacology/Nephrology
7. N
ovel Approches for the treatment of diabetic end-organ damage
( diabetic nephropathy/ diabetic heart disease )
Project leader
Coworkers
Collaboration
Berthold Hocher
Markus Alter, Ina Ott, Karoline von Websky, Alexandra Simon, Katharina Krause-Relle
Prof. Dr. Florian Schweigert, PD Dr. med. vet. Jens Raila,
Dr. rer. nat. Alexandra Henze, Institute of Nutritional Science, University of Potsdam
As the prevalence of diabetes mellitus and hypertension
rises in the western industrial nations, their complications
such as diabetic nephropathy become more and more
present. The model of diabetic and eNOS deficient mice
comes very close to the situation in humans and established treatments such as medication with AT2 receptor
blockers still lack satisfactory efficacy. Dipeptidylpeptidase-IV inhibition (BI1356) is reported to have beneficial
effects on glucose tolerance and to act antifibrotically. The
aim of the study was to investigate the renal and cardiovascular effects of treatment with BI1356 on diabetic and
eNOS deficient mice and to compare the results with the
established treatment with Telmisartan.
65 male eNOS knock out C57BL/6J mice were randomly divided into four groups 1,5 week after they had
intraperitonally received streptozotocin (each 100 mg/
kg body weight on 2 consecutive days): Telmisartan (1
mg/ kg), BI1356 (3 mg/ kg), BI 1356+ Telmisartan (3+1
mg/ kg), and vehicle (n=14-18 per group). Another 14
mice received vehicle after they had been administered
citrate puffer instead of streptozotocin (non-diabetic
controls). All substances were given once daily by oral
gavage with equal volumes per body weight. After 11
weeks of treatment, experiments with metabolic cages
were performed, urine and blood were obtained and
blood pressure was measured.
None of the substances had an influence on survival.
Unlike BI1356, Telmisartan reduced systolic blood pressure compared to vehicle treated, diabetic and non-dia-
betic mice. BI1356 in combination with Telmisartan was
able to significantly reduce albuminuria compared to diabetic, vehicle treated animals (3.07±0.67 µg/[g*24h] vs.
7.27±1.50 µg/[g*24h]; mean±SEM; n= 12-13; p<0.05).
No significant differences were measured between diabetic animals treated solely with BI1356 (4.92±1.60
µg/[g*24h]) or Telmisartan (4.53±1.19 µg/[g*24h]; n=
12-14), respectively, and vehicle (7.27±1.50 µg/[g*24h]).
Plasma Cystatin C levels were not significantly altered.
This is due to the short hyperglycaemic and treatment
period resulting in hyperfiltrating, stage 1 renal disease.
We demonstrate for the first time, that treatment with the
novel dipeptidylpeptidase-IV inhibitor BI1356 in combination with Telmisartan reduces albuminuria in an early
stage hypertensive diabetic nephropathy model and
might enhance the effects of established treatments.
85
Berthold Hocher – Pharmacology/Nephrology
8. T he combined endothelin-converting enzyme / neutral
endopeptidase inhibitor SLV338 prevents myocardial
remodelling in rats with renovascular hypertension in a
blood pressure independent manner
Project leader
Coworkers
Collaboration
Berthold Hocher
Philipp Kalk, Yuliya Sharkovska, Katharina Krause-Relle, Thiemo Pfab
Prof. Dr. Yvan Fischer, Dr. Katrin Hoffmann, Abbott Products GmbH,
Hannover, Germany; Philippe Guillaume, Daniel Provost, Porsolt and
partners Pharmacology, Le Genest-Saint-Isle, France; Prof. Dr. Sebastian Bachmann,
Dr. Kerim Mutig, Dr. Alexander Paliege, Institut für Vegetative Anatomie, Charité,
Campus Mitte: Dr. Elena Kaschina, Center for Cardiovascular Research/
Institut für Pharmakologie, Charité, Campus Mitte
Hypertensive heart disease is a major contributor to
cardiovascular morbidity and mortality. Endothelin is a
potent vasoconstrictive, pro-inflammatory, profibrotic
and proliferative mediator produced by the endothelin
converting enzyme(s) (ECE), whereas natriuretic peptides – degraded by the neutral endopeptidase (NEP)
— have diuretic, vasodilatory, anti-inflammatory, antifibrotic, and anti-proliferative properties. Thus combined ECE/NEP inhibition may represent an option
to halt hypertensive cardiac remodelling. The present
study examined the effects of SLV338, a novel ECE/
NEP inhibitor, on cardiac tissue protection in a rat
model of renovascular hypertension (2-kidney-1-clip;
2K1C).
Male rats were allocated to 5 groups: Sham-operated rats, untreated animals with 2K1C, 2K1C animals
treated with oral SLV338 (30 and 100 mg/kg/d), and
2K1C animals treated with oral losartan (20mg/kg/d).
86
Treatment duration was 12 weeks (starting 2 weeks
after clipping). Blood pressure was assessed every
four weeks. At study end hearts were taken for histology/computer-aided histomorphometry. Basic pharmacological properties of SLV338 are also described.
SLV338 is a dual ECE/NEP inhibitor as demonstrated
both in vitro and in vivo in functional assays. In the
2K1C study losartan lowered blood pressure by up
to 46 mmHg, whereas both dosages of SLV338 had
no effect. However, SLV338 (both dosages) completely normalized cardiac interstitial fibrosis (Figure
1), perivascular fibrosis, myocyte diameter and media/
lumen-ratio of cardiac arteries, as did losartan.
These observations show that the dual ECE/NEP
inhibitor SLV338 prevents cardiac remodelling to
the same extent as losartan, but in a blood pressure
independent manner, in a rat model of renovascular
hypertension.
Berthold Hocher – Pharmacology/Nephrology
Cardiac interstitial fibrosis in the 2K1C study. Results were calculated as percentage of Sirius-Redpositive area in randomly chosen pictures from cardiac sections; all values are given as mean ± SEM;
; *: p < 0.05 vs Sham-group; †††/† - p < 0.001/0.05 vs 2K1C.
Publications 2006-2010
Quaschning T, Hocher B, Ruhl S, Kraemer-Guth A, Tilgner J,
Wanner C, Galle J (2006): Vasopeptidase inhibition normalizes blood pressure and restores endothelial function in
renovascular hypertension. Kidney Blood Press Res 29 (6):
351-9
Pfab T, Thöne-Reineke C, Theilig F, Lange I, Witt H, MaserGluth C, Bader M, Stasch JP, Ruiz P, Bachmann S, Yanagisawa M, Hocher B (2006): Diabetic endothelin B receptordeficient rats develop severe hypertension and progressive
renal failure. J Am Soc Nephrol 17 (4): 1082-9
Langnickel D, Enghard P, Klein C, Undeutsch R, Hocher B,
Manz R, Burmester GR, Riemekasten G (2006): Induction of
pathogenic anti-dsDNA antibodies is controlled on the level of
B cells in a non-lupus prone mouse strain. J Clin Immunol
26 (1): 86-95
Chen YP, Pfab T, Slowinski T, Richter CM, Godes M, Hocher
B (2006): Impact of genetic variation of tumor necrosis factoralpha on gestational hypertension. Chin Med J (Engl) 119 (9):
719-24
Pfab T, Franz U, Herfeld F, Lun A, Armbruster F, Hocher B
(2006): Rapid immunochromatographic strip test for the
detection of albuminuria and brief literature review on albuminuria screening. Eur J Med Res 11 (1): 3-6
Regitz-Zagrosek V, Hocher B, Bettmann M, Brede M, Hadamek K, Gerstner C, Lehmkuhl HB, Hetzer R, Hein L (2006):
Alpha2C-adrenoceptor polymorphism is associated with
improved event-free survival in patients with dilated cardiomyopathy. Eur Heart J 27 (4): 454-9
Wagner FD, Buz S, Zais H, Stasch JP, Hetzer R, Hocher B
(2006): Humoral and hemodynamic responses after left ventricular assist device implantation and heart transplantation.
Exp Biol Med (Maywood) 231 (6): 861-4
Kalk P, Godes M, Relle K, Rothkegel C, Hucke A, Stasch
JP, Hocher B (2006): NO-independent activation of soluble
guanylate cyclase prevents disease progression in rats with
5/6 nephrectomy. Br J Pharmacol 148 (6): 853-9
87
Berthold Hocher – Pharmacology/Nephrology
Thone-Reineke C, Kalk P, Dorn M, Klaus S, Simon K, Pfab T,
Godes M, Persson P, Unger T, Hocher B (2006): High-protein
nutrition during pregnancy and lactation programs blood
pressure, food efficiency, and body weight of the offspring in
a sex-dependent manner. Am J Physiol Regul Integr Comp
Physiol 291 (4): R1025-30
Pfab T, Slowinski T, Godes M, Halle H, Priem F, Hocher B
(2006): Low birth weight, a risk factor for cardiovascular
diseases in later life, is already associated with elevated fetal
glycosylated hemoglobin at birth. Circulation 114 (16): 168792
Pfab T, Poralla C, Richter CM, Godes M, Slowinski T, Priem
F, Halle H, Hocher B (2006): Fetal and maternal peroxisome
proliferator-activated receptor gamma2 Pro12Ala does not
influence birth weight. Obesity 14 (11): 1880-5
Bamberg C, Diekmann F, Haase M, Budde K, Hocher B, Halle
H, Hartung J (2007): Pregnancy on intensified hemodialysis:
fetal surveillance and perinatal outcome. Fetal Diagn Ther
22 (4): 289-93
Hocher B, Liefeldt L, Quaschning T, Kalk P, Ziebig R, Godes
M, Relle K, Asmus G, Stasch JP (2007): Soluble CD154 is a
unique predictor of nonfatal and fatal atherothrombotic events
in patients who have end-stage renal disease and are on
hemodialysis. J Am Soc Nephrol 18 (4): 1323-30
Seeliger E, Flemming B, Wronski T, Ladwig M, Arakelyan K,
Godes M, Möckel M, Persson PB (2007): Viscosity of contrast
media perturbs renal hemodynamics. J Am Soc Nephrol 18
(11) :2912-20
Pfab T, Stirnberg B, Sohn A, Krause K, Slowinski T, Godes
M, Guthmann F, Wauer R, Halle H, Hocher B (2007): Impact
of maternal angiotensinogen M235T polymorphism and
angiotensin-converting enzyme insertion/deletion polymorphism on blood pressure, protein excretion and fetal outcome
in pregnancy. J Hypertens 25 (6): 1255-61
Slowinski T, Kalk P, Christian M, Schmager F, Relle K, Godes
M, Funke-Kaiser H, Neumayer HH, Bauer C, Theuring F,
Hocher B (2007): Cell-type specific interaction of endothelin
and the nitric oxide system: pattern of prepro-ET-1 expression
in kidneys of L-NAME treated prepro-ET-1 promoter-lacZtransgenic mice. J Physiol 581 (Pt 3): 1173-81
88
Kalk P, Eggert B, Relle K, Godes M, Heiden S, Sharkovska
Y, Fischer Y, Ziegler D, Bielenberg GW, Hocher B (2007): The
adenosine A1 receptor antagonist SLV320 reduces myocardial fibrosis in rats with 5/6 nephrectomy without affecting
blood pressure. Br J Pharmacol 151 (7): 1025-32
Quaschning T, Voss F, Relle K, Kalk P, Vignon-Zellweger N,
Pfab T, Bauer C, Theilig F, Bachmann S, Kraemer-Guth A,
Wanner C, Theuring F, Galle J, Hocher B (2007): Lack of
endothelial nitric oxide synthase promotes endothelin-induced
hypertension: lessons from endothelin-1 transgenic/endothelial nitric oxide synthase knockout mice. J Am Soc Nephrol
18 (3): 730-40.
Wiessner R, Hannemann-Pohl K, Ziebig R, Grubitzsch H,
Hocher B, Vargas-Hein O, Lun A, Schimke I, Liefeldt L (2008):
Impact of kidney function on plasma troponin concentrations
after coronary artery bypass grafting. Nephrol Dial Trans­
plant 23 (1): 231-8
Quaschning T, Voss F, Herzfeld S, Relle K, Kalk P, Godes
M, Pfab T, Kraemer-Guth A, Bonz AW, Theuring F, Galle J,
Hocher B (2008): Lack of iNOS impairs endothelial function in
endothelin-1 transgenic mice. Kidney Blood Press Res 31
(2): 127-34
Hocher B, Kalk P, Godes M, Liefeldt L, Ziebig R, Stasch JP,
Quaschning T, Pfab T (2008): Gender-dependent impact of
risk factors for cardiovascular and non-cardiovascular mortality in end-stage renal disease patients on haemodialysis.
Kidney Blood Press Res 31 (5): 360-6
Kalk P, Westermann D, Herzfeld S, Relle K, Pfab T, Bauer C,
Tschöpe C, Stasch JP, Hocher B (2008): Additional lack of
iNOS attenuates diastolic dysfunction in aged ET-1 transgenic
mice. Can J Physiol Pharmacol 86 (6): 353-7
Heiden S, Pfab T, von Websky K, Vignon-Zellweger N, Godes
M, Relle K, Kalk P, Theuring F, Zidek W, Hocher B (2008):
Tissue specific activation of the endothelin system in severe
acute liver failure. Eur J Med Res 13 (7): 327-9
Kalk P, Senf P, Deja M, Petersen B, Busch T, Bauer C,
Boemke W, Kaisers U, Hocher B (2008): Inhalation of an
endothelin receptor A antagonist attenuates pulmonary
inflammation in experimental acute lung injury. Can J Physiol
Pharmacol 86 (8): 511-5
Berthold Hocher – Pharmacology/Nephrology
Kalk P, Mach A, Thone-Reineke C, Godes M, Heiden S,
Sharkovska Y, von Websky K, Relle K, Hocher B (2008):
Pulmonary fibrosis in L-NAME-treated mice is dependent on
an activated endothelin system. Can J Physiol Pharmacol
86 (8): 541-5
Seeliger E, Wronski T, Ladwig M, Dobrowolski L, Vogel
T, Godes M, Persson PB, Flemming B (2009): The reninangiotensin system and the third mechanism of renal blood
flow autoregulation. Am J Physiol Renal Physiol 296 (6):
F1334-45
Raila J, Kalk P, Pfab T, Thöne-Reineke C, Godes M, Yanagisawa M, Schweigert FJ, Hocher B (2008): Urinary protein
profiling with surface-enhanced laser desorption/ionization
time-of-flight mass sxpectrometry in ETB receptor-deficient
rats. Can J Physiol Pharmacol 86 (8): 566-70
Mitrovic V, Seferovic P, Dodic S, Krotin M, Neskovic A, Dickstein K, de Voogd H, Böcker C, Ziegler D, Godes M, Nakov R,
Essers H, Verboom C, Hocher B (2009): Cardio-renal effects
of the A1 adenosine receptor antagonist SLV320 in patients
with heart failure. Circ Heart Fail 6: 523-31
Hocher B, Chen YP, Hügle S, Repey J, Krause K, Slowinski
T, Godes M, Schaeffeler E, Guthmann F, Wauer R, Halle H,
Gossing G, Pfab T (2008): Impact of maternal endothelial
nitric oxide synthase gene polymorphisms on blood pressure,
protein excretion and fetal outcome in pregnancy. J Hum
Hypertens 22 (9): 641-7
Kalk P, Rückert M, Godes M, von Websky K, Relle K, Neumayer HH, Hocher B, Morgera S (2009): Does endothelin
B receptor deficiency ameliorate the induction of peritoneal
fibrosis in experimental peritoneal dialysis? Nephrol Dial
Transplant 25:1474-1478
Aufdenblatten M, Baumann M, Raio L, Dick B, Frey BM,
Schneider H, Surbek D, Hocher B, Mohaupt MG (2009): Prematurity is related to high placental cortisol in preeclampsia.
Pediatr Res 65 (2): 198-202
Kleeberg L, Morgera S, Jakob C, Hocher B, Schneider M,
Peters H, Rötzer S, Müller C, Kaiser M, Fleissner C, Heider
U, Neumayer HH, Sezer O (2009): Novel renal replacement
strategies for the elimination of serum free light chains in
patients with kappa light chain nephropathy. Eur J Med Res
14 (2): 47-54
Kalk P, Thöne-Reineke C, Schwarz A, Godes M, Bauer C,
Pfab T, Hocher B (2009): Renal Phenotype of ET-1 Transgenic
Mice is Modulated by Androgens. Eur J Med Res 14 (2):
55-8
Kalk P, Guthmann F, Krause K, Relle K, Godes M, Gossing G,
Halle H, Wauer R, Hocher B (2009): Impact of maternal body
mass index on neonatal outcome. Eur J Med Res 14 (5):
216-22.
Hocher B, Chen YP, Schlemm L, Burdack A, Li J, Halle H,
Pfab T, Kalk P, Lang F, Godes M (2009): Fetal sex determines
the impact of maternal PROGINS progesterone receptor
polymorphism on maternal physiology during pregnancy.
Pharmacogenet Genomics 9: 710
Hocher B, Schlemm L, Haumann H, Poralla C, Chen YP, Li J,
Guthmann F, Bamberg C, Kalache KD, Pfab T (2009): Interaction of maternal peroxisome proliferator-activated receptor
gamma2 Pro12Ala polymorphism with fetal sex affects maternal glycemic control during pregnancy. Pharmacogenet
Genomics 20:139-142
Sharkovska Y, Kalk P, Lawrenz B, Godes M, Hoffmann LS,
Wellkisch K, Geschka S, Relle K, Hocher B, Stasch JP (2010):
NO-independent Stimulation of Soluble Guanylate Cyclase
Reduces Target Organ Damage in Low- and High-Renin
Models of Hypertension. J Hypertens 28 (8):1666-1675
Schlemm L, Haumann HM, Ziegner M, Stirnberg B, Sohn A,
Alter M, Pfab T, Kalache KD, Guthmann F, Hocher B (2010):
New evidence for the fetal insulin hypothesis: fetal angiotensinogen M235T polymorphism is associated with birth
weight and elevated fetal total glycated hemoglobin at birth. J
Hypertens 28 (4) :732-739
Vinon-Zellweger N, Relle K, Kienlen E, Rahnenführer J,
Sharkovska Y, Heiden S, Kalk P, Schwab K, Neuman B,
Scheler C, Albrecht-Küpper B, Stasch JP, Theuring F, Hocher
B (2010): Endothelin-1 overexpression restores cardiac function in eNOS knockout mice. submitted
89
Berthold Hocher – Pharmacology/Nephrology
Wengenmayer C, Krikov M, Müller S, Lucht K, Villringer A,
Unger T, Hocher B, Thöne-Reineke C (2010): Novel therapy
approach in primary stroke prevention: Simultaneous inhibition of endothelin converting enzyme and neutral endopeptidase in spontaneously hypertensive, stroke–prone rats
improves survival. Neurological Research in press
Zebger-Gong H, Müller D, Diercke M, Haffner D, Hocher B,
Verberckmoes S, Schmidt S, D’Haese PC, and Querfeld U
(2010): 1,25-Dihydroxyvitamin D3 - induced Aortic Calcifications in Experimental Uremia: Upregulation of Osteoblast
Markers, Calcium-transporting Proteins and Osterix. J
Hypertens in press
Föller M, Mahmud H, Qadri SM, Gu S, Braun M, Bobbala D,
Hocher B, Lang F (2010): Endothelin B receptor stimulation
inhibits suicidal erythrocyte death. FASEB J 24 (9):3351-9
Hocher B, Heimerl D, Slowinski T, Godes M, Halle H, Priem F,
Pfab T (2010): Birthweight and fetal glycosylated hemoglobin
at birth in newborns carrying the GLUT1 XbaI gene polymorphism. Clin Lab accepted
Alter M, Pfab T, Guthmann F, Burdack A, Kempiners N, Kalk
P, Hocher B (2010): Maternal and Fetal PROGINS Progesterone Receptor Polymorphism reduces the Risk for Transient
Tachypnea of the Newborn. Clin Lab in press
Hocher B, Schlemm L, Haumann H, Rahnenführer J, Li J,
Guthmann F, Bamberg C, Kalk P, Pfab T, Chen YP (2010):
Offspring sex determines the impact of the maternal ACE I/D
polymorphism on maternal glycemic control during the last
weeks of pregnancy. J Renin Angiotensin Aldosterone
Syst in press.
Schmerbach K, Kalk P, Wengenmayer C, Lucht K, Unger T,
Hocher B, Thoene-Reineke C (2010): Renal Outcome in Equipotent Antihypertensive Treatment with Telmisartan, Ramipril
and Combination in SHR-SP Rats. submitted
Kalk P, Sharkovska Y, Relle K, Pfab T, Guillaume P, Provost
D, Hoffmann K, Fischer Y, Hocher B (2010): The Combined
Endothelin-Converting Enzyme / Neutral Endopeptidase
Inhibitor SLV338 Prevents Myocardial Remodelling in Rats
with Renovascular Hypertension in a Blood Pressure Independent Manner. Hypertension under review
Fischer SS, Kempe DS, Leibrock CB, Rexhepaj R, Siraskar B,
Boini KM, Ackermann TF, Föller M, Hocher B, Rosenblatt KP,
Kuro-O M, Lang F (2010): Hyperaldosteronism in Klotho-deficient mice. Am J Physiol Renal Physiol Epub ahead print
Schildroth J, Rettig-Zimmermann J, Kalk P, Steege A, Fähling
M, Sendeski M, Paliege A, Lai EY, Bachmann S, Persson PB,
Hocher B, Patzak A (2010): Endothelin type A and B receptors in the control of afferent and efferent arterioles in mice.
Nephrol Dial Transplant Epub ahead print
90
Reviews & book chapters (2006-2010)
Hocher B (2006): The cost-effectiveness analysis (CEA) is
dependent on the quality of the NEPHRIC study. Am Heart
J 151 (1)
Hocher B (2007): Fetal programming of cardiovascular diseases in later life - mechanisms beyond maternal undernutrition. J Physiol 579 (Pt 2): 287-8
Li J, Doerffel Y, Hocher B, Unger T (2007): Inflammation in
the genesis of hypertension and its complications--the role of
angiotensin II. Nephrol Dial Transplant 22 (11): 3107-9
Hocher B (2009): Neonatal handling: a potential new underlying reason for programming of renal diseases in later life.
Kidney Blood Press Res 32 (4): 284-5
von Websky K, Heiden S, Pfab T, Hocher B (2009): Pathophysiology of the endothelin system - lessons from genetically
manipulated animal models. Eur J Med Res 14 (1): 1-6
Hocher B (2010): Adenosine A1 Receptor Antagonists in Clinical Research and Development. Kidney Int 78 (5): 438-45
Berthold Hocher – Pharmacology/Nephrology
General information
Third party funding
Project leader
Hocher B.
Hocher B.
Hocher B.
Hocher,B.
Hocher B.
Hocher B.
Kalk P.
Hocher B.
Project title
Generation of cross-bred mice of ET
transgenic and eNOS knockout mice
Impact of NO-independent activation of
sGC on cardiac and renal target organ
damage in rats with 5/6 nephrectomy
Impact of adenosine A1 receptor
antagonist SLV320 on cardiac and
renal target organ damage in rats with
5/6 nephrectomy
Role of endothelin B receptor in diabetic cardiomyopathy
Role of ETB receptor in acute renal
failure
Therapeutic potential of selective
adenosine A1 receptor blockade in
hepatorenal syndrome in rats
sGC Activation as potential therapeutic
approach in diabetic nephropathy
Cathepsin S in uremic animal models
Sponsor
DFG
Period
2005-2007
BayerHealthcare
2006
Solvay Pharmaceuticals
2007
EU-Comission-Marie-Curie early
stage training programme
Dr. Werner-Jackstädt-Stiftung
2006-2009
Solvay Pharmaceuticals
2006-2008
Bayer Schering Pharma AG
2010-2011
Roche
2010-2013
2005-2006
91
Ulrich Kintscher – Pharmacology/Obesity Research
Head of the group
Prof. Dr. med. Ulrich Kintscher
Curriculum Vitae: Prof. Dr. Ulrich Kintscher studied medicine in Giessen and Hamburg, where he completed his MD thesis in 1997. He then worked as a clinical fellow
at the German Heart Centre/Humboldt University in the Dept. of Cardiology in Berlin
till 2002, interrupted by a three year postdoctoral research fellowship at the University of California in Los Angeles in the of Division of Endocrinology, Diabetes and
Hypertension. In 2002 he joined the CCR/ Institute for Pharmacology at the Charite
Universitary Medicine in Berlin, where he passed his habilitation for Experimental Pharmacology in 2004. In
2006 he became a W2-Professor for Pharmacology/ Obesity Research at the Charité, where he obtained his
board certification in Internal Medicine in 2007.
Members of the group
Scientists
Anna Foryst-Ludwig
Mandy Bloch
Technicians
Sprang, Christiane Thalke, Beata
Dr. rer. nat. (Laboratory Head)
Dipl. Biol.
Students
Bähr, Ilse Nirmala
Benz, Verena
Böhm, Christian
Giersch, Katja
Herbst, Lena
Wardat, Sami
Winkler, Robin
PhD student
PhD student
PhD student
PhD student
PhD student
PhD student
PhD student
93
Ulrich Kintscher – Pharmacology/Obesity Research
Summary
The focus of the Kintscher group is the characterization of mechanisms which mediate cardiovascular
disease in obesity, insulin resistance and diabetes.
Based on these findings we are aiming on the identification of new pharmacological targets for cardiovascular disease prevention.
A centrepiece of our research is a group of nuclear
hormone receptors named peroxisome proliferator-activated receptors (PPAR). The PPAR-family includes the isoforms PPARalpha, delta, and
gamma. Activated by specific ligands these receptors function as transcriptional regulators of genes
involved in lipid- and glucose metabolism. In addition
to their role in metabolic diseases, these receptors
exert direct cardio-vasculo protective effects. Ligand-mediated activation of PPARs requires a coordinated binding and release of nuclear cofactors, a
process also called PPAR-modulation.
Our work is focussing on the characterization of
mechanism involved in selective PPARgamma-modulation during the development of insulin resistance,
diabetes mellitus and associated cardiovascular disease by using a translational approach ranging from
in-vitro molecular studies in cell culture to animals to
molecular studies in patients. Hereby, the expression
levels of the different components of PPARgammatranscriptional complexes, in-vivo binding of PPARgamma-cofactor complexes on target gene promoters and their functional analysis play an important
role. In our preclinical studies we use a broad array
of molecular techniques, and genetically-modified
in-vivo mouse models. In clinical studies we investigate the expression levels of defined PPARgamma94
cofactors (qPCR) and their binding on target gene
promoters (ChIP) in human samples from fat tissue
biopsies and monocyte/ macrophages to characterize mechanisms of selective PPARgamma modulation during disease progression.
This work has been recently expanded to the characterization of metabolic functions of nuclear coactivator- and corepressor complexes which have been
identified as dysregulated cofactors in metabolic tissues (e.g. adipose, skeletal muscle, liver) during the
development of diet-induced obesity/ insulin resistance. We have identified candidate cofactors which
are prominently dysregulated during disease progression. Functional characterization of candidate
genes are currently ongoing.
PPARgamma is activated by synthetic ligands, glitazones, which are used as insulin-sensitizers in antidiabetic therapy. However, the clinical usage of glitazones is limited by its propensity to induce weight
gain, fluid retention and edema. Therefore, we are
currently in the process to investigate substances
which are called selective PPARgamma modulators
(SPPARMs), which regulate a distinct set of PPARgamma-target genes, thereby improving insulin sensitivity without the induction of weight gain. We have
recently identified a subgroup of angiotensin type
1 receptor blockers (ARB) as SPPARMs. Following
this line, we are currently analyzing new bi-modal
ARB/ SPPARM compounds (synthesized in collaboration with the Institute of Pharmacy, Free University,
Berlin, Germany) as potential new treatment options
for the therapy of patients suffering from both high
blood pressure and impaired insulin- and glucose
metabolism.
Ulrich Kintscher – Pharmacology/Obesity Research
Along the line of nuclear hormone receptors as
major pharmacological targets for cardiovascular
prevention, we are actively investigating the role of
estrogen receptors (ER) in sex differences in myocardial hypertrophy and their role in body weight loss
and maintenance of reduced body weight. These
projects are focussed on tissue crosstalk, in particular adipose tissue – heart, during the development of
myocardial hypertrophy. We want to understand the
impact of ER-regulated adipose tissue metabolism
during exercise, and its interaction with the myocardium via defined systemic substrate disposal. Furthermore, the impact of ER-pathways on adipose
tissue lipolysis and lipogenesis is investigated during weight reduction and weight regain in mice. For
this adipose-tissue specific ER-deficient mice will
be utilized.
Zusammenfassung
Der Schwerpunkt der Arbeitsgruppe Kintscher liegt
in der Charakterisierung von molekularen Mechanismen kardiovaskulärer Erkrankungen bei Adipositas,
Insulinresistenz und Diabetes mellitus. Das Ziel ist
die Identfizierung neuer pharmakologischer Zielmoleküle für die Prävention kardiovaskulärer Erkrankungen.
Im Mittelpunkt steht eine Gruppe nukleärer Hormonrezeptoren, die Peroxisome Proliferator-Activated Receptors (PPARs) welche aus 3 Isoformen
besteht PPARalpha, PPARdelta und PPARgamma.
PPARs werden durch Liganden aktiviert und regulieren als Transkritionsfaktoren wichtige Gene im
Lipid- und Glukosestoffwechsel. Zusätzlich zu ihrer
metabolischen Funktion vermitteln diese Rezeptoren
kardiovaskulär-protektive Wirkungen. Die ligandenvermittelte Aktivierung von PPARs setzt die koordinierte Bindung und Abspaltung sog. nukleärer
Kofaktoren voraus, ein Prozess der als selektive
PPAR-Modulation bezeichnet wird.
Unsere Arbeit ist auf die molekulare Charakterisierung der selektiven PPARgamma-Modulation
während der Pathogenese von Insulinresistenz/
Diabetes Mellitus und begleitender kardiovaskulärer Erkrankungen konzentriert. Hierbei folgen wir
einem strikt translationalen Ansatz und versuchen
molekularbiologische in-vitro Studien mit Tierstudien und klinischen Studien zu kombinieren. Wir
charakterisieren die Expression und Funktion von
beteiligten Komponenten des PPARgamma-Transkriptionskomplexes in verschiedenen Geweben in
unterschiedlichen Stadien der Krankheitsentwicklung. In unseren präklinischen Studien kommen eine
Vielzahl an molekularbiologischen Techniken zur
Anwendung, begeleitet durch Studien in Knock-out
Mausmodellen. Im Rahmen klinischer Studien versuchen wir unsere molekularbiologische Expertise
auf humane Proben anzuwenden und charakterisieren PPARgamma-Transkriptionskomplexe mittels
Geweb-ChIP und weiterführenden Methoden in isolierten Monozyten und Fettgewebsbiopsien.
Diese Arbeiten wurden kürzlich ergänzt durch die
Analyse der metabolischen und kardiovaskulären
Funktionen von nukleären Koaktivatoren und –
repressoren in unterschiedlichen Geweben (z.B.
Skeletmuskel, Fett, Monozyten/ Makrophagen),
95
Ulrich Kintscher – Pharmacology/Obesity Research
welche während der Entstehung der diät-induzierten Insulinresistennz als dysreguliert charakterisiert wurden. In noch laufenden Studien konnten
wir Kandidatengene aus dieser Gruppe identifizieren, welcher einer ausgeprägten diät- und/ oder
gewichtsabhängigen Regulation unterliegen.
PPARgamma wird durch synthetische Liganden aus
der Gruppe der Glitazone aktiviert, welche als Insulinsensitizer in der oralen antidiabetischen Therapie
etabliert sind. Der klinische Einsatz von Glitazonen
ist wesentlich durch das Nebenwirkungsspektrum
dieser Substanzen limitiert einschließlich Körpergewichtszunahme, Flüssigkeitsretention und Ödemen.
Aus diesem Grunde entwickeln wir in einem weiteren Projektbereich derzeitig sogenannte selektive
PPARgamma Modulatoren (SPPARMs), die bei
verbesserter Wirksamkeit (anti-diabetisch, antiatheroskerlotisch) weniger Nebenwirkungen aufweisen sollen. In diesem Zusammenhang konnten
wir eine Subgruppe von Angiotensin Typ1 – Rezeptorblockern (ARB) als SPPARMs charakterisieren.
In Kollaboration mit dem Institut für Pharmazie der
Freien Universität Berlin (Prof. R. Gust) analysieren
wir derzeitig neue bi-modale ARB/ SPPARM Substanzen als eine zukünftige Therapieoption für Patienten, welche bei bestehender arterieller Hypertonie
96
gleichzeitig an einer Störung des Insulin- und Glukosestoffwechsel leiden.
Ein weiterer Schwerpunkt des Bereiches: Nukleäre
Hormonrezeptoren als Zielmoleküle für neue pharmakologische Ansätze zur kardiovaskulären Prävention, liegt auf der Analyse von Estrogenrezeptoren (ERs) als zentrale Mediatoren von Geschlechterunterschieden bei Myokardhypertrophie, sowie
auf Untersuchungen zur Bedeutung von ERs bei
der Körpergewichtsreduktion und dem Gewichtserhalt nach Reduktion. Diese Studien untersuchen
die Interaktionen zwischen Fettgewebe und Myokard im Rahmen der trainings-induzierten kardialen Hypertrophieentstehung. Die Bedeutung der
Estrogenrezeptor-vermittelten Regulation der Fettgewebslipolyse als myokardiale Energiesubstratquelle während Training und deren Einfluss auf die
kardiomyozytäre Hypertrophie werden untersucht.
Im Weiteren wird die Bedeutung der FettgewebsERs im Rahmen der geschlechtsspezifischen Körpergewichtsregulation untersucht. Hierbei wird die
Relevanz der ERs bei der Fettgewebslipolyse und
–lipogenese im Verlauf einer Körpergewichtsreduktion und erneuter Zunahme („Weight Cycling“)
analysiert.
Ulrich Kintscher – Pharmacology/Obesity Research
Research projects
1. M
olecular Characterisation of New Selective Modulators for the
Peroxisome Proliferator-Activated Receptor gamma
Project leader
Coworkers
Funding
External cooperations
Ulrich Kintscher
Anna Foryst-Ludwig, Lena Herbst, Beata Thalke
- DFG (KI 712/ 3-2)
Prof. Ronald Gust, Institute of Pharmacy, University Innsbruck, Austria
Aim of this project is the development of detailed
structure-activity relationships for the design of new
selective PPARgamma modulators (SPPARMs) based
on our knowledge of the PPARgamma-modulating
properties of the angiotensin type 1 receptor blocker
telmisartan. In our previous work, we characterized
major telmisartan components regarding their relevance for PPARgamma activation. (Goebel M et al.
ChemMedChem 2009/1+2, and BioorgMedChem
2010). To further characterize these compounds, we
are currently performing cofactor-PPARgamma LBD
binding studies using FRET analysis. These studies
will be complemented by Microarray analysis for gene
expression profiling under distinct SPPARM stimulation. Finally, we want to connect cofactor binding with
the regulation of gene expression, and will perform
ChIP on identified target gene promoters.
Results of FRET (Fluorescence-Resonance Energy
Transfer) analysis for binding
of DRIP205 to PPARg-LBD.
A specific signal is generated
when ligand-induced cofactor - PPARg-LBD interaction
occurs. The number represent
newly synthesized SPPARMs;
Pio: Pioglitazon.
97
Ulrich Kintscher – Pharmacology/Obesity Research
2. T
he role of sex specific lipolysis in exercise-induced cardiac
hypertrophy
Project leader
Coworkers
Funding
External cooperations
Anna Foryst-Ludwig
Beata Thalke, Christiane Sprang, Christian Böhm
- DFG-FG 1054 (TP6)
Dr. med. Michael Kreissl, Universität Würzburg
Erin Kershaw, PhD, University of Pittsburgh, USA
Adipose tissue undergoes profound molecular
changes during exercise such as increased lipolysis
as a result of an activation of hormone-sensitive lipase
(HSL) and possibly additional adipose tissue lipases,
such as adipose tissue triglyceride lipase (ATGL).
Women show higher adipose tissue lipolysis than men
during exercise. Importantly, HSL is regulated in a sexdependent manner that might be mediated through
direct estrogen actions. Sex-specific modulation of
adipose fatty-acid metabolism during exercise may
result in alteration of circulating fatty acid levels leading
to changes in cardiac substrate utilization. Dysregulation of cardiac fatty oxidation plays an important role
in the development of cardiac hypertrophy, and may
98
mediate sexual dimorphisms in the development of
physiological cardiac hypertrophy (see figure)
The aim of the present project is the identification and
characterization of molecular and biochemical lipolytic
targets such as HSL/ ATGL in adipose tissue, which
mediate sexual dimorphism in exercise-induced cardiac hypertrophy with a focus on estrogen receptordependent gene regulation in adipose tissue. Male
and female mice will be challenged with active treadmill running and comprehensive metabolic, cardiac,
and molecular phenotyping will be performed. Finally,
HSL and ATGL gene regulation by estrogen receptors
will be studied in-vitro.
Ulrich Kintscher – Pharmacology/Obesity Research
3. A
dipose tissue estrogen receptors and body weight loss/
maintenance of reduced weight
Project leader
Coworkers
Funding
External cooperations
Anna Foryst-Ludwig and Mandy Bloch
Verena Benz, Beata Thalke, Christiane Sprang
- DFG-KFO 218 (TP1)
Dr. rer. nat. Petra Wiedmer, DIFE, Potsdam
Jan-Åke Gustafsson, Karolinska Institute, Stockholm, Sweden
Adipose tissue undergoes profound molecular
changes during exercise such as increased lipolysis
as a result of an activation of hormone-sensitive lipase
(HSL) and possibly additional adipose tissue lipases,
such as adipose tissue triglyceride lipase (ATGL).
Women show higher adipose tissue lipolysis than men
during exercise. Importantly, HSL is regulated in a sexdependent manner that might be mediated through
direct estrogen actions. Sex-specific modulation of
adipose fatty-acid metabolism during exercise may
result in alteration of circulating fatty acid levels leading
to changes in cardiac substrate utilization. Dysregulation of cardiac fatty oxidation plays an important role
in the developmemt of cardiac hypertrophy, and may
mediate sexual dimorphisms in the development of
physiological cardiac hypertrophy (see figure)
The aim of the present project is the identification and
characterization of molecular and biochemical lipolytic
targets such as HSL/ ATGL in adipose tissue, which
mediate sexual dimorphism in exercise-induced cardiac hypertrophy with a focus on estrogen receptordependent gene regulation in adipose tissue. Male
and female mice will be challenged with active treadmill running and comprehensive metabolic, cardiac,
and molecular phenotyping will be performed. Finally,
HSL and ATGL gene regulation by estrogen receptors
will be studied in-vitro.
99
Ulrich Kintscher – Pharmacology/Obesity Research
4. F
unction of HDAC6 for glucocorticoid receptor-mediated
impairment of glucose metabolism
Project leader
Coworkers
Funding
External cooperations
Ulrich Kintscher
Robin Winkler
- German Diabetes Foundation
Prof. Patrick Matthias, Friedrich Miescher Institute, Basel, Switzerland
The glucocorticoid receptor (GR) is an important regulator of insulin sensitivity and glucose tolerance. Since
the histone deacetylase 6 (HDAC6) deacetylates heat
shock protein 90, a GR chaperone, we investigated
the effect of HDAC6 on GR function in a metabolic
context in vivo and in vitro.
Wildtype (wt) and HDAC6-deficient (HDAC6ko) mice
were subjected to dexamethasone (dex) treatment. In
wt-mice dex injection resulted in a marked glucose
intolerance. In HDACko mice dex-induced glucose
intolerance was completely abolished. Furthermore,
100
dex-mediated hyperinsulinemia in wt mice did not
occur in HDACko mice and GR-mediated insulin
resistance in these mice was ameliorated ascertained
by ITT. After dex stimulation glucose production was
augmented by in wt hepatocytes whereas the increase
in HDAC6ko hepatocytes was diminished.
The present study identifies HDAC6 as an important
regulator of metabolic GR-function preventing liganddependent induction of gluconeogenesis. Modulation
of GR-function by HDAC6 appears to be gene-specific which may provide an opportunity for therapeutic
intervention.
Ulrich Kintscher – Pharmacology/Obesity Research
Publications 2006 - 2010
Schupp M, Kintscher U, Fielitz J, Thomas J, Pregla R, Hetzer
R, Unger T, Regitz-Zagrosek V. 2006 Cardiac PPARalpha
expression in patients with dilated cardiomyopathy. Eur J
Heart Fail 8:290-4
Schupp M, Lee LD, Frost N, Umbreen S, Schmidt B, Unger
T, Kintscher U. 2006 Regulation of peroxisome proliferatoractivated receptor gamma activity by losartan metabolites.
Hypertension 47:586-9
Kintscher U, Bramlage P, Paar WD, Thoenes M, Unger T.
Irbesartan for the treatment of hypertension in patients
with the metabolic syndrome: A sub analysis of the Treat to
Target post authorization survey. Prospective observational,
two armed study in 14,200 patients. Cardiovasc Diabetol
2007;6(1):12
Scholze J, Grimm E, Herrmann D, Unger T, Kintscher U.
2007 Optimal treatment of obesity-related hypertension: the
Hypertension-Obesity-Sibutramine (HOS) study. Circulation
115(15):1991-1998
Kappert K, Meyborg H, Clemenz M, Graf K, Fleck E, Kintscher U, Stawowy P. 2007 Insulin facilitates monocyte
migration: A possible link to tissue inflammation in insulinresistance. Biochem Biophys Res Commun 19:19
Walcher D, Hess K, Heinz P, Petscher K, Vasic D, Kintscher
U, Clemenz M, Hartge M, Raps K, Hombach V, Marx N.
2008 Telmisartan inhibits CD4-positive lymphocyte migration
independent of the angiotensin type 1 receptor via peroxisome proliferator-activated receptor-gamma. Hypertension
51:259-266
Clemenz M, Frost N, Schupp M, Caron S, Foryst-Ludwig A,
Bohm C, Hartge M, Gust R, Staels B, Unger T, Kintscher U.
2008 Liver-specific peroxisome proliferator-activated receptor
alpha target gene regulation by the angiotensin type 1 receptor blocker telmisartan. Diabetes 57(5):1405-1413
Karatas A, Hegner B, de Windt LJ, Luft FC, Schubert C,
Gross V, Akashi YJ, Gurgen D, Kintscher U, da Costa Goncalves AC, Regitz-Zagrosek V, Dragun D. 2008 Deoxycorticosterone acetate-salt mice exhibit blood pressure-independent sexual dimorphism. Hypertension 51(4):1177-1183
Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M,
Wabitsch M, Fischer-Posovszky P, Barth TF, Dragun D, Skurk
T, Hauner H, Bluher M, Unger T, Wolf AM, Knippschild U,
Hombach V, Marx N. 2008 T-lymphocyte infiltration in visceral
adipose tissue: a primary event in adipose tissue inflammation
and the development of obesity-mediated insulin resistance.
Arterioscler Thromb Vasc Biol 28(7):1304-1310
Schmerbach K, Schefe JH, Krikov M, Muller S, Villringer A,
Kintscher U, Unger T, Thoene-Reineke C. 2008 Comparison
between single and combined treatment with candesartan
and pioglitazone following transient focal ischemia in rat brain.
Brain Res 1208:225-233
Foryst-Ludwig A, Clemenz M, Hohmann S, Hartge M, Sprang
C, Frost N, Krikov M, Bhanot S, Barros R, Morani A, Gustafsson JA, Unger T, Kintscher U. 2008 Metabolic actions of
estrogen receptor beta (ERbeta) are mediated by a negative
cross-talk with PPARgamma. PLoS Genet 4(6):e1000108
Kaschina E, Grzesiak A, Li J, Foryst-Ludwig A, Timm M,
Rompe F, Sommerfeld M, Kemnitz UR, Curato C, Namsolleck
P, Tschope C, Hallberg A, Alterman M, Hucko T, Paetsch I,
Dietrich T, Schnackenburg B, Graf K, Dahlof B, Kintscher U,
Unger T, Steckelings UM. 2008 Angiotensin II Type 2 Receptor Stimulation. A Novel Option of Therapeutic Interference
With the Renin-Angiotensin System in Myocardial Infarction?
Circulation 24:24
Goebel M, Clemenz M, Staels B, Unger T, Kintscher U, Gust
R. 2009 Characterization of new PPARgamma agonists:
analysis of telmisartan‘s structural components. ChemMed­
Chem 4(3):445-456
Brinckmann M, Kaschina E, Altarche-Xifro W, Curato C, Timm
M, Grzesiak A, Dong J, Kappert K, Kintscher U, Unger T, Li
J. 2009 Estrogen receptor alpha supports cardiomyocytes
indirectly through post-infarct cardiac c-kit+ cells. J Mol Cell
Cardiol 47(1):66-75
Mai K, Andres J, Biedasek K, Weicht J, Bobbert T, Sabath M,
Meinus S, Reinecke F, Mohlig M, Weickert MO, Clemenz M,
Pfeiffer AF, Kintscher U, Spuler S, Spranger J. 2009 Free fatty
acids link metabolism and regulation of the insulin-sensitizing
fibroblast growth factor-21. Diabetes 58(7):1532-1538
101
Ulrich Kintscher – Pharmacology/Obesity Research
Goebel M, Staels B, Unger T, Kintscher U, Gust R. 2009
Characterization of new PPARgamma agonists: benzimidazole derivatives - the importance of position 2. ChemMed­
Chem 4(7):1136-1142
Kappert K, Tsuprykov O, Kaufmann J, Fritzsche J, Ott I,
Goebel M, Bahr IN, Hassle PL, Gust R, Fleck E, Unger T, Stawowy P, Kintscher U. 2009 Chronic Treatment With Losartan
Results in Sufficient Serum Levels of the Metabolite EXP3179
for PPAR{gamma} Activation. Hypertension 54(4):738-743
Kilter H, Werner M, Roggia C, Reil JC, Schafers HJ, Kintscher
U, Böhm M. 2009 The PPAR-gamma agonist rosiglitazone
facilitates Akt rephosphorylation and inhibits apoptosis in cardiomyocytes during hypoxia/reoxygenation. Diabetes Obes
Metab 11:1060-1067
Van Linthout S, Foryst-Ludwig A, Spillmann F, Peng J, Feng Y,
Meloni M, Van Craeyveld E, Kintscher U, Schultheiss HP, De
Geest B, Tschope C. 2010 Impact of HDL on adipose tissue
metabolism and adiponectin expression. Atherosclerosis
210(2):438-444
Fliegner D, Schubert C, Penkalla A, Witt H, Kararigas G,
Dworatzek E, Staub E, Martus P, Ruiz Noppinger P, Kintscher
U, Gustafsson JA, Regitz-Zagrosek V. 2010 Female sex and
estrogen receptor-beta attenuate cardiac remodeling and
apoptosis in pressure overload. Am J Physiol Regul Integr
Comp Physiol 298(6):R1597-1606
Kintscher U, Marx N, Martus P, Stoppelhaar M, Schimkus J,
Schneider A, Walcher D, Kummel A, Winkler R, Kappert K,
Dorffel Y, Scholze J, Unger T. Effect of high-dose valsartan
on inflammatory and lipid parameters in patients with Type 2
diabetes and hypertension. 2010 Diabetes Res Clin Pract
89(3):209-215
Goebel M, Wolber G, Markt P, Staels B, Unger T, Kintscher U,
Gust R. Characterization of new PPARgamma agonists: benzimidazole derivatives-importance of positions 5 and 6, and
computational studies on the binding mode. 2010 Bioorg
Med Chem 18(16):5885-5895
102
Reviews & book chapters (2006-2010)
Clemenz M, Kintscher U, Unger T (2006): The metabolic
syndrome: cluster with a self-fulfilling loop? J Hypertens
24:257-8
Hartge MM, Kintscher U, Unger T (2006): Endothelial dysfunction and its role in diabetic vascular disease. Endocrinol
Metab Clin North Am 35:551-60, viii-ix
Kintscher U (2007): Does adiponectin resistance exist in
chronic heart failure? 2007 Eur Heart J 28(14):1676-1677
Kappert K, Unger T, Kintscher U (2008): [Aliskiren hemifumarat]. Dtsch Med Wochenschr 133(24):1308-1312
Kintscher U (2008): The cardiometabolic drug rimonabant:
after 2 years of RIO-Europe and STRADIVARIUS. Eur Heart
J 13:13
Kintscher U (2008): Pharmacological Differences of Glitazones - Does PPARa Activation Make the Difference? J Am
Coll Cardiol 52(10):882-884
Kintscher U, Foryst-Ludwig A, Unger T (2008): Inhibiting Angiotensin Type 1 Receptors as a Target for Diabetes. Expert
Opinion on Therapeutic Targets 12(10):1257-1263
Kintscher U, Goebel M (2009): INT-131, a PPARgamma agonist for the treatment of type 2 diabetes. Curr Opin Investig
Drugs 10(4):381-387
Kintscher U (2009): ONTARGET, TRANSCEND, and PRoFESS: new-onset diabetes, atrial fibrillation, and left ventricular hypertrophy. J Hypertens 27 Suppl 2(2):S36-39.
Foryst-Ludwig A, Kintscher U (2010): Metabolic impact of
estrogen signalling through ERalpha and ERbeta. J Steroid
Biochem Mol Biol 122(1-3):74-81.
Kintscher U (Herausgeber) und Marx N (Ko-Herausgeber)
2008 Antihypertensiva bei Metabolischem Syndrom und
Diabetes Mellitus. Unimed Buch-Verlag
Ulrich Kintscher – Pharmacology/Obesity Research
General information
Third party funding ( 2006-2010 )
Project leader
Kintscher U.
Kintscher U.
Kintscher U.
Kintscher U.
Project title
Der Einfluß von Fettgewebe auf die
Myokardhypertrophie: Bedeutung von
PPARs und Estrogenrezeptoren
Molekulare Charakterisierung neuer
selektiver Modulatoren des Peroxisome
Proliferator-Activated Receptor (PPAR)
Molekulare Charakterisierung neuer
selektiver Modulatoren des Peroxisome
Proliferator-Activated Receptor (PPAR)
The role of sex-specific lipolysis in
exercise-induced cardiac hypertrophy
Kintscher U.
Foryst-Ludwig, A.
The role of adipose tissue estrogen
receptors during body weight loss and
the maintenance of reduced weight
Kintscher U.
The role of HDAC6 in GR-mediated
impairment of glucose metabolism
Caveolopathies - insulin resistance,
lipid metabolism
Boschmann M.
Kintscher U.
Kintscher U.
Unger T.
Kintscher U.
Fleck E.
Gust R.
Sponsor
DFG – GRK 754-III Geschlechtsspezifische Mechanismen bei
Myokardhypertrophie
DFG-KI 712/ 3-1
Period
2006-2010
DFG-KI 712/ 3-2
2010-2012
DFG-FG 1054/1
TP6 – KI 712/ 5-1
Sex-specific mechanisms in myocardial hypertrophy
DFG-KFG 218/1
TP6 – KI 712/ 6-1
Hormonal regulation of body
weight maintenance
Deutsche Diabetes Stiftung
2008-2011
DFG-KFG 192/2
TP9 – Skeletal muscle growth
regulation and dysregulation
Regulation of tissue-specific endothelial Nippon Boehringer
dysfunction by telmisartan: The role of
PPAR activation
PPAR activation by Losartan in Hyper- Merck, Sharp & Dohme
tensive Patients: The Importance of
Losartan-Metabolites
2006-2008
2009-2012
2010-2011
2010-2013
2006-2008
2006-2008
103
Ulrich Kintscher – Pharmacology/Obesity Research
Kintscher U.
Kintscher U.
Unger T.
Kintscher U.
Kintscher U.
Einfluss von Telmisartan auf Parameter
der Inflammation bei hypertensiven
Patienten (METATEL-STUDIE)
Regulation of Muscular and Hepatic
Insulin Signaling by Telmisartan
Regulation of histone deacetylases by
telmisartan
Metabolic actions of telmisartan in
adipose-specific PPAR-deficient mice
Bayer Vital
2007-2009
Boehringer Ingelheim
2005-2007
Boehringer Ingelheim
2008-2009
Boehringer Ingelheim
2009-2010
Awards
2008
104
Best Oral Abstract Presentation;
13th Annual Meeting of the European Council for
Cardiovascular Research
Mandy Bloch
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Head of the group
Prof. Dr. Harm Peters
Curriculum Vitae: Prof. Peters studied medicine at the Free University Berlin and
started his scientific career as a MD candidate at the Department for Endocrinology at
the Benjamin Franklin Hospital in Steglitz, today Charité Campus Benjamin Franklin.
In 1992 he earned his degree with the thesis entitled “Impact of maternal TSH receptor-blocking antibodies in cases of genetic hypothyroidism”. In 1993 he changed to
the Department of Nephrology at the Benjamin Franklin Hospital, followed by a 2-year
postdoctoral research period in the lab of Prof. Nancy Noble and Prof. Wayne Border, University of Utah, Salt
Lake City, USA. Since 1999 Prof. Peters works as a assistant medical director at the Medical Clinic / Focus
Nephrology, Charité Campus Mitte. In 2000 he passed his habilitation on the topic: ”Impact of the L-Arginin-/
NO metabolism in cases of acute and chronic glomerulosclerosis”.
Members of the group
Scientists
Rosenberger, Christian
Brand, Daniel
Gaedeke, Jens
Grosz, Bianca
Halleck, Fabian
Khadzhynov, Dmytro
Krämer, Stephanie
Loof, Tanja
Martini, Sebastian
Wang-Rosenke, Yingrui
PD Dr. med.
Veterinarian
Dr. med.
Physician
Physician
Physician
Dr. vet. med.
Dipl. Biochem.
Dr. med.
Dr. med.
Technicians
Mika, Alice
Students
Grüger, Jens
Horoszynska, Lillianna
Scheidl, Julia
Medical student
Medical student
Medical student
105
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Summary
The main research of the group „molecular fibrosis and hypoxia research“ concentrates on the
one hand on new aspects in the pathogenesis and
treatment of acute and chronic fibrotic diseases
of the kidney and their consequences for the cardiovascular system. With regard to the kidney, the
research group compares immune-mediated (acute
anti-Thy1 glomerulonephritis, lupus nephritis) with
not primarily inflammatory disease models (5/6
nephrectomy, diabetic nephropathy). With regard
to the vascular consequences of impaired renal
function, the research focus is on changes of the
myocard, coronary arteries and large vessels. In
order to unravel the underlying mechanism of pathological changes, the group concentrates on cellular
mediators such as mast cells, lymphocytes (S1P
modulation by FTY720) und platelets (inhibition by
clopidogrel) and several extra- and intracellular pathways. These include transforming growth factor beta
(small molecule inhibition), PDGF (receptor blockade with imatinib), mammalian target of rapamycin
(mTOR-inhibitor sirolimus), NO/cGMP signal transduction (stimulator of soluble guanylate cyclase BAY
41-2272) and the renin-angiotensin-systems (maximizing RAS inhibition and AT2 receptor stimulation).
On the other hand, the research focus Hypoxia
includes detection of hypoxia and hypoxia adapta-
106
tion, as well as potential therapeutic use of hypoxia
adaptation in renal and cardiac diseases. The
emphasis lies on acute kidney injury, chronic renal
fibrosis, diabetic nephropathy, renal transplantation and myocardial infarction. Particular attention
is given to the clinical relevance of injury models,
as well to subtle analysis of morphological tissue
injury. Our group is dedicated to in vivo transcriptional response to hypoxia, which is complex and
still poorly understood. It is widely accepted that
hypoxia-inducible transcription factors (HIFs) play a
pivotal role in this setting. Theoretically, both beneficial and detrimental effects can arise from HIF activation depending on the HIF target genes involved.
Our group investigates the impact of HIF up-regulation achieved with different hypoxic stimuli,
cell types and co-transcription factors. HIF target
genes like erythropoietin, heme oxygenase-1, glucose transporter-1 are located within tissues with
help of high amplification immunohistochemistry,
RT-PCR, or in situ hybridisation. Therapeutical HIF
activation is achieved by pharmaceutical blockade
of HIF degradation or using inducible transgenic
technology. Human biopsies help to test for the
clinical relevance of our findings in animal models. In the following we provide an overview of our
projects from 2008 until present.
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Zusammenfassung
Die Forschungsschwerpunkte der Arbeitsgruppe
„Molekulare Fibrose- und Hypoxieforschung“ konzentrieren sich auf neue Aspekte in der Pathogenese und Therapie von akuten und chronischen
fibrotischen Nierenerkrankungen sowie ihrer Auswirkungen auf das Herzkreislaufsystem. In Bezug
auf Erkrankungen der Nieren werden insbesondere
immun-vermittelte (akute Anti-Thy1-Glomerulonephritis, Lupusnephritis) mit nicht primär entzündlichen
Krankheitsmodellen (5/6 Nephrektomie, diabetische
Nephropathie) verglichen. Bei den kardiovaskulären
Auswirkungen renaler Funktionseinschränkung stehen Veränderungen des Myokards, der Koronarien
und der großen Gefäße im Vordergrund. Bei der
Aufarbeitung der den pathologischen Veränderungen zugrunde liegenden Mechanismen fokussieren
sich die Untersuchungen der Arbeitsgruppe auf
zelluläre Mediatoren, wie Mastzellen, Lymphozyten
(S1P-Modulation mit FTY720) und Thrombozyten
(Inhibition mit Clopidogrel) sowie auf verschiedene
extra- und intrazellulare Pathways. Hierzu gehören
Transforming Growth Factor-beta (Hemmung mittels small molecules), PDGF (Rezeptorinaktivierung
mitles Imatinib), mammalian Target of Rapamycine
(mTOR-lnhibitor Sirolimus), NO/cGMP Signaltransduktion (Stimulator der löslichen Guanylatcyclase
BAY 41-2272) und des Renin-Angiotensin-Systems
(Optimierung der RAS-Hemmung, Stimulation des
AT2-Rezeptors). Auf der anderen Seite umfasst der
Forschungsschwerpunkt „Hypoxieforschung“ den
Nachweis von Hypoxie bzw. Hypoxieanpassung,
sowie die mögliche therapeutische Nutzung der
Hypoxieanpassung bei Nieren- und Herzerkrankungen. Ein besonderes Augenmerk liegt auf akutes Nierenversagen, chronische Nierenfibrose, diabetische Nephropathie, Nierentransplantation und
Herzinfarkt. Zum Leitbild der Arbeitsgruppe gehört
eine sorgfältige Bewertung experimenteller Modelle
auf ihre klinische Übertragbarkeit, sowie eine sorgfältige anatomisch-pathologische Aufarbeitung von
Organschäden. Die komplexe transkriptionelle Antwort auf Hypoxie wird in vivo untersucht. Dabei spielen sogenannte Hypoxie-induzierbare Faktoren (HIF)
eine zentrale Bedeutung. Deren Aktivierung kann
theoretisch positive oder negative Auswirkungen
auf den Krankheitsverlauf haben. Unsere Gruppe
erforscht die HIF-Wirkung in Abhängigkeit verschiedener Schädigungsreize, Zelltypen und Ko-Transkriptionsfaktoren. HIF-Zielgene wie Erythropoietin,
Hämoxygenase-1, Glukose-Transporter-1 werden
innerhalb von Organen mittels Immunhistochemie,
RT-PCR und/oder In-situ-Hybridisierung nachgewiesen. HIF-Aktivierung erfolgt über verschiedene
Hypoxietypen, medikamentös, oder durch den Einsatz transgener Tiere. Die klinische Relevanz des
Systems wird anhand humaner Biopsien überprüft.
Im folgenden finden Sie einen Überblick über die
2008 bis aktuell bearbeiteten Projekte und Fragestellungen.
107
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Research projects
1. D
ysfunction in epithelial sodium transport under proteinuric
conditions
Project leader
Coworkers
Funding
Harm Peters and Franziska Theilig, Institute of Anatomy
Henriette J. Kaminski, medical student, Institute of Anatomy
Tanja Loof
Christian Kastner, doctoral fellow, Institute of Anatomy
Sebastian Bachmann, Institute of Anatomy
DFG FOR 667, Epithelial mechanisms in renal volume regulation, TP8
Changes of tubular sodium reabsorption play an
important role in the maintenance of volume homeostasis under physiological conditions. In proteinuric
states however, defective sodium balance likely contributes to accompanied clinically evident sequelaes
like edema and hypertension. The underlying mechanisms, as well as the nephron segments and transport mechanism involved need to be clarified. We
test the hypothesis that proteinuria leads to changes
in epithelial transporters and channels which favour
volume retention. We aspire to identify possible variations in sorting and expression of tubular membrane
proteins. Therefore, proteinuric rat models of antiThy1-glomerulonephritis (Thy1-GN), and puromycininduced nephritis, and a mouse model of anti-glomerular basement membrane nephritis (GBM-GN) will be
evaluated with biochemical and histochemical methods. In Thy1-GN, nephron segment specific changes
108
in expression and localisation of epithelial transporters
and channels, concentration of systemic parameters
of the renin-angiotensin-system and vasopressin will
be evaluated. GBM-GN induced in megalin transgenic
mice will clarify the role of increased proximal tubular protein endocytosis on expression and activity of
proximal tubular transporters and channels. Mechanisms of activation and deactivation of transporters
and channels, participating cell compartment specific
proteins, and the role of lipid composition under proteinuria will be characterized. The anticipated results
will provide a comprehensive analysis of the cell biological basis underlying the clinically important consequences of nephron epithelial shuttling and transport
in proteinuric diseases.
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
2. O
pen European Nephrology Science Center / a Charité renal biopsy
metadata base repository for system biology
Project leader
Coworkers
Funding
External cooperations
Harm Peters, Christian Rosenberger
Fabian Halleck, Jens Grüger, Lilianna Horoszynska, Sebastian Martini,
Birgit Rudolph, Depart. of Pathology
Thomas Schrader, Depart. of Pathology
- DFG, 10692/1-1 und 1-2, TP GN
- Astellas Pharma, München
- Berlin-Brandenburgische Gesellschaft für Nephrologie,
- Deutsche Gesellschaft für Nephrologie
The typical problems of research in biomedical disciplines are apparent: a limitation of the amount of
cases of more or less rare diseases and complications, heterogeneous data types with varying quality
of automatically and „by hand“ generated data, and
decentralized data retention inhibiting the scientific
work. The main Task of Open European Nephrology
Science Center (OpEN.SC) is to create a large renal
biopsy data base of patients with transplanted and
native kidneys with implementation of a metadata
repository for clinical and laboratory data, including
a focus on vascular consequences of impaired renal
function. The metadata repository will be stored and
processed by an intelligent data management tool –
the intelligent Catalogue. Actual projects focus on the
epidemiology of primary glomerular diseases in BerlinBrandenburg and the identification of unusual cases.
Another recent focus has been the HIF activation in
human kidney transplants. In these studies we have
shown that in long-term renal transplants HIF is activated in clinical rejection, but not in subclinical rejection or borderline changes [Rosenberger et al, JASN].
We are currently investigating the potential diagnostic
gain provided by additional HIF stainings in transplant
biopsies. In a pilot project we have demonstrated that
renal biopsies stained for HIF can be displayed by virtual microscopy.
HIF-1a immunohistochemistry in a human
renal transplant biopsy
109
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
3. S
timulation of soluble guanylate cyclase improves renal
recovery following relief of unilateral ureteral obstruction
Project leader
Coworkers
Funding
External cooperations
Yingrui Wang-Rosenke
Alice Mika, Tanja Loof, Harm Peters
Charité-Forschungsstipendium
Bayer Schering Pharma GmbH.
Antifibrotic effects of soluble guanylate cyclase (sGC)
activation and cGMP production have been shown
in anti-thy1 renal disease. In this study, the specific
sGC stimulator Bay 41-8543 was administered to animals with UUO after relief of ureteral obstruction in
order to further observe the renoprotective effects of
sGC/cGMP in restoring and preserving progressive
renal disease. Untreated UUO rats showed elevation
of systolic blood pressure, marked tubular atrophy,
tubulointerstitial apoptosis, macrophage infiltration
and fibrosis, α-smooth muscle actin (SMA) expression and mRNA expression of TGF-β. Administration
of Bay 41-8543 significantly increased plasma cGMP.
This was paralleled by significant reductions in systolic
blood pressure, tubular diameter, tubular apoptosis,
tubulointerstitial macrophage infiltration and cell proliferation, and tubulointerstitial fibrosis. Thus, stimulation
of sGC through Bay 41-8543 increased cGMP production and further prevented the progression of UUO,
via reductions in renal atrophy, tubular apoptosis and
tubulointerstitial fibrosis and macrophage infiltration.
The findings suggest that sGC stimulator may be an
effective treatment to restore or preserve renal histology and renal function in this experimental model of
renal disease.
Typical alpha-smooth muscle actin staining
in renal tubular tissue of rats subjected
unilateral urethral obstruction.
110
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
4. S
ignal agonistic AT2 receptor stimulation prevents renal
alterations in the experimental model of subtotal nephrectomy
Project leader
Coworkers
External cooperations
Stephanie Krämer, Harm Peters
Tanja Loof, Alica Mika, Dmytro Khazhyno, v
Ulrike Steckelings, Institute of Pharmacology
Thomas Unger, Instititute of Pharmacology
Vicore Pharma AB, Göteborg, Sweden
A major contributor to progression in chronic renal
disease (CKD) is the renin angiotensin system with its
main effector peptide angiotensin II, exerting various
pleiotropic actions via binding at Angiotensin type 1
(AT1) and type 2 (AT2) receptors. AT2 mediated actions
were analyzed by applying the novel non-peptide AT2
agonist Compound 21 (C21) in the model of subtotal nephrectomy (SNX) in male Wistar rats (250-280
g BW). After 12 weeks SNX was characterized by
increased systolic blood pressure and protein excretion. Administration of C21 had no significant effects
blood pressure, but on proteinuria. On the molecular level, TGF-ß and fibronectin mRNA-expression
was up-regulated in SNX-animals and significantly
reduced by C21. Western blot analysis corresponded
with reductions of TGF-ß immunohistological protein
detection of fibronectin. Histological collagen I protein expression was lowered as well. Furthermore C21
reduced renal macrophage and lymphocyte infiltration as well as renal cell proliferation. The investigation
show that C21 significantly reduces the expression of
renal pro-fibrotic and pro-inflammatory markers at a
molecular and histological level in the model of SNX.
The results underline previous findings pointing at
direct AT2 mediated anti-fibrotic and anti-inflammatory
properties. Therefore, pharmacological AT2 stimulation with C21 might represent a novel therapeutic tool
for the treatment of CKD.
Course of proteinuria in rats subjected
to subtotal nephrectomy (SNX) over 12 weeks
without of with administration of
Compound 21 (C21).
111
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
5. Cardiovascular NO/cGMP signalling in mild renal insufficiency
Project leader
Coworkers
Funding
External cooperations
Bianca Grosz, Harm Peters
Stephanie Krämer. Tanja Loof, Yingrui Wang-Rosenke, Dmytro Khadzhynov
European Union, Marie Curie program
EST-CT-2005-020268
Prof. Dr. Kerstin Amann, Inst. of Pathology, Erlangen
This project aims at characterizing cardiovascular and
renal morphological and molecular changes in a rat
model of mild renal insufficiency, with a special focus
on the role played by NO-cGMP signalling pathway
and the modulator effect of gender. The second experimental study was also directed towards investigating
the therapeutic potential of BAY 41-8543, a compound
which enhances NO-cGMP signal transduction, in
the development of cardiovascular and renal disease
in the same animal model. Summarizing the results
obtained, in a rat model of mild renal insufficiency
prominent gender effects on the degree of hypertension, proteinuria, renal hypertrophy, and cardiac and
aortic remodelling were found. Compared to uremic
male rats, females showed less hypertension, left
ventricular and aortic hypertrophy but contrastingly
higher proteinuria. These discordant results suggest
that progression of cardiovascular and renal disease is
differently influenced by sex hormones. Furthermore,
this is the first study demonstrating that enhancing
NO/GMP signalling by Bay 41-8543 ameliorates aortic
wall hypertrophy and stiffening significantly and in a
blood pressure-independent manner, thus proposing
it as a future therapeutic strategy in the treatment of
vascular remodelling.
Aortic wall elastin staining of 18 weeks
after induction of mild renal insufficiency
in rats.
112
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
6. Inducible transgenic HIF activation in acute kidney injury
Project leader
Coworkers
Funding
External cooperations
Christian Rosenberger
Harm Peters
Sebastian Bachmann, Institute of Anatomy
Alexander Paliege, Institute of Anatomy
Alice Mika, BTA
- DFG FOR 1368
Prof. Robert Koester, Nephrology, Paris
Rhabdomyolysis is an important cause of acute kidney injury (AKI) in the human. We have shown that
in experimental rhabdomyolysis induced by glycerol
injection into the hind limbs renal tubular hypoxia is
deep and protracted, whereas hypoxia adaptation is
short-lived and limited to a minority of hypoxic cells.
We hypothesize that therapeutic activation of HIF may
improve renal outcome in experimental rhabdomyolysis. To activate HIF we employ a transgenic mouse
model, based on tetracycline-inducible knockout of
the von Hippel Lindau (VHL) protein, which is crucial
for physiological HIF degradation. Transgenic animals
feature strong and persistent HIF activation in all renal
tubules. To test for the potential of a prophylactic or
therapeutic HIF activation, the transgene is induced at
different time points before or after AKI. Renal function,
fine morphology, and HIF target genes are assessed
at different time points after AKI.
Immunohistochemistry for HIF and the cell
protective HIF target genes glucose transporter-1
(GLUT-1) and heme oxygenase-1 (HO-1) 24 h after
rhabdomyolysis-induced acute kidney injury:
Asterixes mark proximal convoluted tubules, which
are the main sites of hypoxia and injury in this
model. Control animals exhibit neither HIF (A), nor
GLUT-1 (C) or HO-1 (E) in proximal convoluted
tubules. By contrast, VHL knockout animals feature robust activation of all three markers (B, D, F).
113
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
7. M
echanisms of hypoxia tolerance in acute on chronic kidney
injury
Project leader
Coworkers
External cooperations
Christian Rosenberger
Harm Peters, Alice Mika
Prablheen Singh, La Jolla, CA, USA
Statistically, chronic renal insufficiency is a well known
predisposing factor to acute kidney injury (AKI) in
humans. However, in individual patients with moderate
chronic renal insufficiency the impact of acute hypoxic
insults on renal function is hardly predictable. Most
likely, at least in some of these patients, increased
hypoxia adaptation may exists at baseline. This is probably due to sub-lethal hypoxia and consequent adaptive responses of remaining hypertrophied tubules.
We hypothesize that hypoxia-inducible transcription
factors (HIFs), which are master regulators of hypoxia
adaptation, are activated at baseline in animals with
renal mass reduction with and without additional renal
interstitial fibrosis, and that such HIF activation can
ameliorate super-imposed ischemic AKI. To this end
either normal control rats, rats with unilateral nephrectomy, or rats with subtotal nephrectomy are subjected
to renal warm ischemia and reperfusion. Renal function, morphology, pimonidazole adducts (which indicate profound hypoxia with tissue pO2 below 10 mm
Hg), HIF, and HIF target genes are measured. We are
particularly interested in evidences for hypoxia and
hypoxia adaptation in S3 proximal tubules, which are
the major site of injury in this model of AKI.
HIF-1a immunohistochemistry in the remnant kidney
114
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
8. The role of HIF in renal cyclosporine toxicity
Project leader
Coworkers
External cooperations
Christian Rosenberger
Harm Peters, Julia Scheidl, Daniel Brand, Alice Mika
Sebastian Bachmann, Inst. of Anatomy
Robert Koesters, Nephrology, Paris
Cyclosporine toxicity is a major course of renal graft
failure, leading to tubular injury, arteriolar hyalinosis
and interstitial fibrosis. The responsible mechanisms
are incompletely understood, but, most likely, involve
vasoconstriction and renal hypoxia. Hypoxia-inducible
transcription factors (HIFs) are master regulators of
hypoxia adaptation. Recent data suggest that HIF
may aggravate chronic renal fibrosis. The role of HIF
in cyclosporine toxicity has not been investigated, so
far. We employ a mouse model of cyclosporine toxic-
ity to study the occurrence of hypoxia (pimonidazole
adducts), HIF activation, and up-regulation of HIF
target genes during disease progression. In addition,
we test the impact of an inducible transgenic HIF activation in all renal tubular segments on the course of
cyclosporine-induced renal fibrosis. Micro array technique will reveal the contribution of acknowledged cell
protective vs pro-fibrotic HIF target genes. Candidate
genes will be located within the kidney with help of
immunohistochemistry and in situ hybridization.
HIF-1a after
cyclosporine
treatment
115
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Publications 2006 – 2010
Bernhardt WM, Schmitt R, Rosenberger C, Münchenhagen
PM, Gröne HJ, Frei U, Warnecke C, Bachmann S, Wiesener
MS, Willam C, Eckardt KU (2006): Expression of hypoxiainducible transcription factors in developing human and rat
kidneys. Kidney Int 69(1):114-22
Wang Y, Krämer S, Loof T, Martini S, Kron S, Shimizu F,
Kawachi H, Neumayer HH, Peters H (2006): Enhancing cGMP
in experimental progressive renal fibrosis: soluble guanylate
cyclase stimulation versus phosphodiesterase inhibition. Am
J Physiol Renal Physiol 290: F167-76
Goldfarb M, Rosenberger C, Abassi Z, Shina A, Zilbersat F,
Eckardt KU, Rosen S, Heyman SN (2006): Acute-on-chronic
renal failure in the rat: Functional compensation and hypoxia
tolerance. Am J Nephrol 26:22-33
Rosenberger C, Shina A, Rosen S, Goldfarb M, Eckardt K,
Heyman SN (2006): Hypoxia inducible factors and tubular cell
survival in isolated perfused kidneys. Kidney Int 70:60-70
Hammer MH, Brestrich G, Andree H, Engelmann E, Rosenberger C, Tillmann H, Zwinger S, Babel N, Nickel P, Volk HD,
Reinke P (2006): HLA type-independent method to monitor
polyoma BK virus-specific CD4 and CD8 T-cell immunity. Am
J Transplant 6(3): 625-31
Waiser J, Dell K, Kreutzkamp J, Bohler T, Budde K, Peters H,
Neumayer HH (2006): FK506, transforming growth factorbeta1 and mesangial matrix synthesis: Parallels and differences compared with cyclosporine A. Cytokine 21:59-65
Haase M, Morgera S, Bamberg C, Halle H, Martini S, Hocher
B, Diekmann F, Dragun D, Peters H, Neumayer HH, Budde K
(2006): A systematic approach to managing pregnant dialysis
patients--the importance of an intensified haemodiafiltration
protocol. Nephrol Dial Transplant 20: 2537-42
Rosenberger C, Pratschke J, Rudolph B, Heyman SN, Schindler R, Babel N, Eckardt KU, Frei U, Rosen S, and Reinke P
(2007): Immunohistochemical detection of hypoxia-inducible
factor-1alpha in human renal allograft biopsies. J Am Soc
Nephrol 18(1): 343-51
116
Rosenberger C, Solovan C, Rosenberger AD, Li J, Treudler
R, Frei U, Eckardt KU, and Brown LF (2007): Upregulation
of hypoxia-inducible factors in normal and psoriatic skin. J
Invest Dermatol 127(10): 2445-52
Seiler M, Brabcova I, Viklicky O, Hribova P, Rosenberger
C, Pratschke J, Lodererova A, Matz M, Schoenemann C,
Reinke P, Volk HD, Kotsch K (2007): Heightened expression
of the cytotoxicity receptor NKG2D correlates with acute
and chronic nephropathy after kidney transplantation. Am J
Transplant 7(2): 423-33
Martini S, Krämer S, Loof T, Wang-Rosenke Y, Daig U, Budde
K, Neumayer HH, Peters H (2007): The S1P modulator
FTY720 limits matrix expansion in acute anti-thy1 mesangioproliferative glomerulonephritis. Am J Physiol Renal Physiol
292:F1761-70
Schneider M, Thomas K, Liefeldt L, Kindgen-Milles D, Peters
H, Neumayer HH, Morgera S (2007). Efficacy and safety of
intermittent hemodialysis using citrate as anticoagulant: a
prospective study. Clin Nephrol 68:302-7
Rosenberger C, Rosen S, Heyman SN (2007): Normotensive
ischemic acute renal failure. N Engl J Med 357(21): 2204-5
Rosenberger C, Khamaisi M, Abassi Z, Shilo V, WekslerZangen S, Goldfarb M, Shina A, Zibertrest F, Eckardt K-U,
Rosen S and Heyman SN (2008): Adaptation to hypoxia in the
diabetic rat kidney. Kidney Int 73(1): 34-42
Rosenberger C, Goldfarb M, Shina A, Bachmann S, Frei
U, Eckardt KU, Schrader T, Rosen S, Heyman SN (2008):
Evidence for Sustained Renal Hypoxia and Transient Hypoxia
Adaptation in Experimental Rhabdomyolysis-Induced Acute
Kidney Injury. Nephrol Dial Transplant 23(4): 1135-43
May D, Gilon D, Djonov V, Itin A, Lazarus A, Rosenberger
C and Keshet E (2008): A conditional transgenic system
for induction and rescue of chronic Myocardial hibernation
provides insights into genomic programs of hibernation and
reversible heart remodeling. Proc Nat Acad Sci USA 105(1):
282-7
Khamaisi M, Raz I, Shilo V, Shina A, Rosenberger C, Dahan
R, Abassi Z, Meidan R, Lecht S, Heyman SN (2008): Diabetes
and radiocontrast media increase endothelin converting
enzyme-1 in the kidney. Kidney Int 74(1):91-100
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Rosenberger C, Rosen S, Shina A, Frei U, Eckardt KU, Flippin LA, Arend M, Klaus SJ, Heyman SN (2008): Activation
of hypoxia inducible factors (HIF) ameliorates hypoxic distal
tubular injury in the isolated perfused rat kidney. Nephrol
Dial Transplant 23(11): 3472-8
Krämer S, Wang-Rosenke Y, Scholl V, Loof T, Binder E,
Khadzhynov D, Kawachi H, Shimizu F, Diekmann F, Neumayer
HH, Peters H (2008): Low-dose mTOR inhibition by rapamycin
attenuates progression in anti-thy1-induced chronic glomerulosclerosis of the rat. Am J Physiol Renal Physiol 294:F440-9
Krämer S, Kron S, Wang-Rosenke Y, Loof T, Khadzhynov D,
Morgera S, Kawachi H, Shimizu F, Martini S, Neumayer HH,
Peters H (2008): Rosuvastatin is additive to high-dose candesartan
in slowing progression of experimental mesangioproliferative
glomerulosclerosis. Am J Physiol Renal Physiol 294:F801-11
Câmpean V, Karpe B, Haas C, Atalla A, Peters H, Rupprecht
H, Liebner S, Acker T, Plate K, Amann K (2008): Angiopoietin
1 and 2 gene and protein expression is differentially regulated
in acute anti-Thy1.1 glomerulonephritis. Am J Physiol Renal
Physiol 294:F1174-1184
Schneider M, Liefeldt L, Slowinski T, Peters H, Neumayer HH,
Morgera S (2008). Citrate anticoagulation protocol for slow
extended hemodialysis with the Genius dialysis system in
acute renal failure. Int J Artif Organs 31:43-8
Rosenberger C, Khamaisi M, Goldfarb M, Shina A, Shilo V,
Zilbertrest F, Rosen S, Heyman SN (2008): Acute kidney injury
in the diabetic rat: studies in the isolated perfused and intact
kidney. Am J Nephrol 28(5): 831-9
Babel N, Eibl N, Ulrich C, Bold G, Sefrin A, Hammer MH, Rosenberger C, Reinke P (2008): Development of Kaposi’s sarcoma
under sirolimus-based immunosuppression and successful treatment with imiquimod. Transpl Infect Dis 10(1): 59-62
Welker P,Krämer S, Groneberg D, Neumayer HH, Bachmann
S, Amann K, Peters H (2008): Increased mast cell number
in human hypertensive nephropathy. Am J Physiol Renal
Physiol 295:F1103-9
Thumfart J,Jung S, Amasheh S, Kraemer S, Peters H, Sommer K, Biber J, Murer H, Meij I, Querfeld U, Wagner CA,
Muller DN (2008): Magnesium stimulates renal phosphate
reabsorption. Am J Physiol Renal Physiol 295:F1126-33
Rosenberger C, Rosen S, Paliege A, Heyman S (2009):
Pimonidazole adduct immunohistochemistry in the rat kidney:
detection of tissue hypoxia. Methods Mol Biol 466:161-74
Paliege A, Rosenberger C, Bondke A, Sciesielski L, Shina
A, Heyman SN, Flippin LA, Arend M, Klaus SJ, Bachmann S
(2009): Hypoxia-inducible factor-2alpha-expressing interstitial
fibroblasts are the only renal cells that express erythropoietin under hypoxia-inducible factor stabilization. Kidney Int.
77:312-8
Gadau J*, Peters H* (* equal contribution), Kasner C, Kühn
H, Krämer C, Castrop H, Bachmann S, Theilig F (2009):
Tubular involvement of volume retention in acute experimental
glomerulonephritis Kidney Int 75: 699-710
Krämer S, Binder E, Loof T, Wang-Rosenke Y, Khadzhynov
D, Budde K, Neumayer HH, Peters H (2009): The lymphocyte
migration inhibitor FTY720 attenuates experimental hypertensive nephropathy. Am J Physiol Renal Physiol 297:F218-27
Morgera S, Schneider M, Slowinski T, Vargas-Hein O, Zuckermann-Becker H, Peters H, Kindgen-Milles D, Neumayer HH
(2009). A safe citrate anticoagulation protocol with variable
treatment efficacy and excellent control of the acid-base
status. Crit Care Med 37:2018-24
Kleeberg L, Morgera S, Jakob C, Hocher B, Schneider M,
Peters H, Rötzer S, Müller C, Kaiser M, Fleissner C, Heider U,
Neumayer HH, Sezer O (2009): Novel renal replacement strategies for the elimination of serum free light chains in patients
with kappa light chain nephropathy. Eur J Med Res 18: 47-54
Knebel F, Schimke I, Schroeckh S, Peters H, Eddicks S,
Schattke S, Brechtel L, Lock J, Wernecke KD, Dreger H, Grubitz S, Schmidt J, Baumann G, Borges AC (2009). Myocardial
function in older male amateur marathon runners: assessment by tissue Doppler echocardiography, speckle tracking,
and cardiac biomarkers. J Am Soc Echocardiogr 22:803-9
Riad A,Van Linthout S, Mohra Z, Rütten H, Peters H,
Schultheiss HP, Tschöpe C (2008). Role of pharmacological enhancement of eNOS in experimental diabetes mellitus.
Diabetologia 51:2325-32
117
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Liefeldt L, Rylski B, Walcher F, Manhart J, Kron S, WangRosenke Y, Paul M, Neumayer HH, Amann K, Peters H
(2009). Transgenic overexpression of endothelin-2 aggravates
moderately diabetic myocardiopathy in rats. Eur J Clin Invest
40:203-10
Zebger-Gong H, Kampmann J, Kong L, Sommer K, Akashi Y,
Janke M, Krämer S, Peters H, Roigas J, Müller DN, Dragun
D, Querfeld U (2009): Decreased Transplant Arteriosclerosis
in Endothelial Nitric Oxide Synthase-deficient Mice. Trans­
plantation 89:518-26
37.Werth N, Beerlage C, Rosenberger C, Yazdi AS, Edelmann M, Amr A, Bernhardt W, von Eiff C, Becker K, Schäfer
A, Peschel A, Kempf VA (2010): Activation of hypoxia inducible factor 1 is a general phenomenon in infections with
human pathogens. PLoS One Jul 14;5(7):e11576
Reviews & book chapters (2006-20010)
Gaedeke J, Peters H (2006): Pharmacological management of
fibrotic renal disease. Expert Opin Pharmacother 7:377-86
Budde K, Schütz M, Glander P, Peters H, Waiser J, Liefeldt L,
Neumayer HH, Böhler T (2006): FTY720 (fingolimod) in renal
transplantation. Clin Transplant 20 Suppl 17:17-24
Goldfarb M, Rosenberger C, Shina A, Rosen S, Heyman SN
(2006): A Role for Erythropoietin in the Attenuation of Contrast Medium-Induced Acute Renal Failure in Rats? Renal
Failure 28: 345-350
Rosenberger C, Rosen S, Heyman SN (2006): Renal Parenchymal Oxygenation and Hypoxia Adaptation in Acute Kidney
Injury. Clin Exp Pharmacol Physiol 33: 980-988
Hampl H, Hennig L, Rosenberger C, Gogoll L, Riedel E,
Scherhag A (2006): Proven strategies to reduce cardiovascular
mortality in hemodialysis patients. Blood Purif 24(1): 100-6
Peters H, Neumayer HH, Gaedeke J (2006): Hochdrucktherapie und frühe Nephroprotektion: eine hausärztliche Herausforderung. Hausarzt- und Notfallmedizin 32:138-42
Peters H, Unger T (2007): Mast cells and the power of local
RAS activation. Nephrol Dial Transplant 22:40-2
118
Martini S, Peters H, Böhler T, Budde K (2007): Current perspectives on FTY720. Expert Opin Investig Drugs 16:505518
Martini S, Peters H (2007): Medical treatment in retroperitoneal fibrosis: The difficulty to provide good evidence in rare
diseases. J Urol 178, 143-144
Heyman SN, Rosen S, Rosenberger C (2008): Renal parenchymal hypoxia, hypoxia adaptation, and the pathogenesis
of radiocontrast nephropathy. Clin J Am Soc Nephrol 3(1):
288-96
Heyman SN, Khamaisi M, Rosen S, Rosenberger C (2008):
Renal Parenchymal Hypoxia, Hypoxia Response and the
Progression of Chronic Kidney Disease. Am J Nephrol 28(6):
998-1006
Unger JK and Peters H (2008): Hepatitis B in chronic kidney
disease: moving toward effective prevention. Kidney Int
2008;73:799-801
Wang-Rosenke Y, Neumayer HH, Peters H (2008): NO
signaling through cGMP in renal tissue fibrosis and beyond:
key pathway and novel therapeutic target. Curr Med Chem
15:1396-406
Heyman, SN, Rosen S, Rosenberger C (2009): Critical
assessment of experimental models of acute renal failure. In:
Critical Care Nephrology, 2nd Ed; Ronco C, Bellomo R &
Kellum J (Eds), Springer/Kluwer Acad Pub
Slowinski T, Schneider H, Peters H (2009): Akutes Nierenversagen: Fragen und Probleme im intensivmedizinischen Alltag.
In: Akutes Nierenversagen bei Intensivpatienten, 1st Ed:
Jörres A. Deutscher Ärzteverlag
Peters H, Martini S (2008): Glomeruläre Erkrankungen (Übersetzung Lewis JB, Neilson EG)
In: Harrison Innere Medizin, 17 Ed. Zeitz M, Dietel M, Suttorp
N; ABW-Wissenschaftsverlag
Martin DR, Semelka RC, Chapman A, Peters H, Finn PJ, Kalb
B, Thomsen H (2009): Nephrogenic systemic fibrosis versus
contrast-induced nephropathy: risks and benefits of contrastenhanced MR and CT in renally impaired patients. J Magn
Reson Imaging 30:1350-6
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Heyman SN, Rosenberger C, Rosen S (2010): Experimental
ischemia-reperfusion: biases and myths - the proximal vs.
distal hypoxic tubular injury debate revisited. Kidney Int 77(1):
9-16
Heyman SN, Rosen S, Khamaisi M, Idée JM, Rosenberger
C (2010): Reactive oxygen species and the pathogenesis of
radiocontrast-induced nephropathy. Invest Radiol 45(4):
188-95
General information
Third party funding ( 2006-2010 )
Project leader
Theilig F.
Peters H.
Peters H.
Peters H.
Budde K.
Peters H.
Peters H.
Rosenberger C.
Peters H.
Theilig F.
Peters H.
Peters, H.
Budde K.
Peters H.
Peters H.
Project title
Zelluläre Translokation tubulärer Membrantransporter bei Proteinurie“ Forschergruppe
667, Teilprojekt 10
„Wachstumsfaktor-Rezeptor-Inhibition und
renale Progression“ (Dmytro Khadzhynov,
A/06/09454)
Teilprojekt: „Glomerulonephritis-Register“
Forscherverbundes „Open
Nephrology Sciences Center“ (OpEN.SC).
(AZ Inst/10692/1-1)
“Cardiovascular NO/cGMP signaling in
renal insufficiency“ CARDIOVASC
(EU-Nr. 020268)
Nephrologische Forschung
Teilprojekt: “HIF in renal biopsies” Forscherverbund „Open Nephrology Sciences
Center“ (OpEN.SC). (AZ Inst/10692/1-2)
„AT2 Agonismus und Progression von chronischer Anti-Thy1-induzierter Nierenfibrose“
(Anna Abadyjan, A/08/77466)
„Glomerulonephritis, tubuläre Endozytose
und Proteinurie“ Forschergruppe 667,
Teilprojekt 8:
Teilprojekt: „Glomerulonephritis-Register“,
Forscherverbund „Open Nephrology Sciences Center“ (OpEN.SC). (AZ
Inst/10692/1-2)
Experimentelle nephrologische Forschung
Klinische nephrologische Forschung
Sponsor
Period
Deutsche Forschungsgemein- 2006-2008
schaft, Bonn
Deutscher Akademischen
Austauschdienst, Bonn
2006
Deutsche Forschungsgemein- 2006-2008
schaft, Bonn
Marie Curie, Early Stage Train- 2006-2010
ing, Brüssel, EU
Sonnenfeld-Stiftung Berlin
2006-2012
Deutsche Forschungsgemein- 2007-2009
schaft, Bonn
Deutscher Akademischen
Austauschdienst, Bonn
2009-2011
Deutsche Forschungsgemein- 2009-2011
schaft, Bonn
Deutsche Forschungsgemein- 2009-2011
schaft, Bonn
Industrie, verschiedene
Industrie, verschiedene
2006-2010
2006-2010
119
Harm Peters – NEPHROLOGY / MOLECULAR FIBROSIS AND HYPOXIA RESEARCH
Awards
2006 Best Poster Award. ISN Forefronts Conference
“Endothelial Biology and Renal Disease”,
New York; USA.
Harm Peters
2006
Rainer Greger - Promotionspreis,
Gesellschaft für Nephrologie
Yingrui Wang-Rosenke
2006
Georg Haas-Doktorandenpreis des Verbandes
Deutscher Nierenzentren, DDnÄ e.V. Yingrui Wang-Rosenke
2007
Hans-U. Zollinger-Preis der Gesellschaft
für Nephrologie Harm Peters
2007
„Lehrbär“ für beste Unterrichtsveranstaltung,
Reformstudiengang Charité Harm Peters
2008
Best Poster Presentation.
Annual Meeting of the European Council for
Cardiovascular Research; Nice, France Bianca Grosz
2010
Best Poster Presentation.
Annual Meeting of the European Council for
Cardiovascular Research; Nice, France Tanja Loof
120
Axel Pries – PHYSIOLOGY
Head of the group
Prof. Dr. med. A xel R. Pries
Curriculum Vitae: From 1973 to 1980 Axel Pries studied medicine at the medical
school University of Cologne, and also worked on his doctoral thesis on ‘Model
studies on phase separation at a capillary orifice’. He finished his studies with the
approbation as medical doctor in 1980 and his doctoral thesis was ranked ‘summa
cum laude’. In the following years he worked as postdoctoral fellow at the Institute
for Physiology, University of Cologne and Free University of Berlin. After a positions
as lecturer and associate professor, he had an appointment in 1997-1998 at the Institute of Anesthesiology of
German Heart Center Berlin. In 1998, hebecame full professor at the Department of Physiology of the Free
University Berlin.
Since 2001, he is Professor of Physiology and head of the Institute for Physiology (one of three physiological
institutes) of the Charité, Berlin. His main tasks involve organization, teaching and research. He is also involved in
work for national and international scientific organisations, including the ESC (European Society for Cardiology)
and the ESM (European Society for Mirocirculation). In the ESC, he is chairing the council for basic cardiovascular science (CBCS) while in the ESM, he is serving as general secretary. Scientifically, his key areas of interest
include microcirculation, and tumor vasculature, blood rheology, vascular adaptation, angiogenesis, ischemia/
reperfusion and the endothelial surface. Recently, he worked on the mechanisms preventing maldistribution
and shunting in microvascular networks related to the differences in normal and tumor vascular beds (Pries et.
al Plos Comp Biol, 2009 and Nat Rev Cancr 2010).
121
Axel Pries – PHYSIOLOGY
Members of the group
Office
Bünsch, Brigitte
Marruhn, Cornelia
Hofmann, Eveline
Wisniewski, Susanne
Research group leaders
Gunga, Hanns-Christian
Habazettl, Helmut
Höpfner, Michael
Kübler, Wolfgang
Munz, Barbara
Preissner, Robert Siegel, Günther
Zakrzewicz, Andreas Prof. Dr. med.
Prof. Dr. med.
PD Dr. rer. nat.
Prof. Dr. med.
Prof. Dr. rer. nat.
PD Dr. rer. nat.
Prof. Dr. med
Dr. med
Scientists
Berger, Felicitas
Ermilov, Eugeny
Hoffmann, Julia
Klippel, Nina
Márki, Alex
Neye, Nils
Nickles, Hannah
Nitzsche, Bianca
Opatz, Oliver
Samapati, Rudi
Reglin, Bettina
Schlabs, Thomas
Stahn, Alexander
Supe, Steffi
122
Dr. rer. nat.
Dr. rer. nat.
Dr. med. Dr. rer. nat.
Dr. med.
Dr. med. vet.
Dr. rer. medic.
Technicians
Becker, Angela
Himmelsbach, Bärbel
Hoffmann, Björn
Noske-Reimers, Renate
Plog, Sylvia
Students
Phillip Zöller
Christian Hoffmann
Bernd Vorderwülbecke
Julian Maroski
Margret Hohberg
Sven Chlench
Christoph Glösenkamp
Felix Landmann
Julian Lenk
Bianca Nitzsche
Kera Westphal
Florian Thilo
Axel Pries – PHYSIOLOGY
Summary
Integrative Physiology and
Organ Perfusion
The research profile of the section ‘Integrative Physiology and Organ Perfusion’ including the groups
Pries, Kübler, Gunga, Kunz, Preissner und Höpfner
integrates physiological research from the molecular
level to human physiology according to the motive of
the American Physiological Society „Integrating Life
Sciences from Molecule to Organism“. The development of integrative concepts is a prerequisite
for the fast and efficient translation of mechanistic
knowledge back into the complex environment of
human physiology and pathophysiology. For this
kind of research, there is also a high demand by
extra-university institutions.
cular networks was established (Pries et al. Nature
Reviews Cancer 2010). The group of W. Kübler uses
intravital microscopy with advanced imaging techniques together with molecular biology to investigate relevant pathophysiological mechanisms in the
alveolo-capillary units of the lung.
The exponents for investigation of human physiology
in the spectrum,are the groups Gunga and Kunz.
H.-C Gunga and his coworkers are longstanding
players in the field of integrative regulatory phenomena during space flight (space medicine) and
in extreme environments. The concept „from Molecule to Organism“. is completed by the integrative
research of circadian phenomena and sleep disorders in healthy people and in patients by the group
Kunz.
Within the section, the group Preissner is located
at the molecular end of the spectrum, investigating molecular interactions with bioinformatic
approaches. Some of the studies are performed
together with the group of M. Hoepfner which is
interested in the molecular and functional events in
Tumor cells and the influence of growth factors and
growth factor inhibitors. In the investigation of tumor
microvasculature, the cooperation of the groups
Hoepfner and Pries has added the techniques of
intravital microscopy and computer simulation.
With this approach, the importance of conducted
responses for the characteristics of tumor microvas-
123
Axel Pries – PHYSIOLOGY
Zusammenfassung
Integrative Physiologie
und Organperfusion
Das übergreifende Forschungsprofil der Abteilung
„Integrative Kreislauf-Physiologie und Organperfusion’“ mit den Arbeitsgruppen Pries, Kübler, Gunga,
Kunz, Preissner und Höpfner ist die Integration physiologischer und pathophysioloigscher Grundlagenforschung von der molekularen Ebene bis hin zum
Menschen in Anlehnung an das Leitmotiv der Amerikanischen Gesellschaft für Physiologie „Integrating
Life Sciences from Molecule to Organism“.
Die Entwicklung leistungsfähiger molekularbiologischer Methoden und die Erfolge der biomedizinischen Forschung in molekularen und zellulären
Bereichen haben zu einem enormen Erkenntniszuwachs über molekulare Mechanismen geführt. Dies
macht die Entwicklung integrativer Konzepte zum
Zusammenwirken der molekularen Einzelmechanismen im gesunden oder kranken Individuum oder
Organ dringend erforderlich. Integrative grundlagenmedizinische Forschung ist eine Voraussetzung für
einen schnellen und erfolgreichen Wissenstransfer
von der molekularen/zellulären Ebene zurück in
das komplexe Gesamtsystem und die Lösung der
ursprünglichen Fragestellungen. Die Bedeutung
integrativer, anwendungsorientierter Forschungsansätze wird auch durch die erhebliche Nachfrage
gesellschaftlicher Institutionen nach konkreten Problemlösungen und nach langfristiger wissenschaftlicher Kooperation deutlich.
Selbstverständlich kann mit einer begrenzten Zahl
von Arbeitsgruppen ein derartiger Ansatz nicht
124
flächendeckend bearbeitet werden. Daher ist der
inhaltliche Zusammenhang der beteiligten Arbeitsgruppen von besonderer Bedeutung. Am molekularen und submolekularen Ende des Integrationsbogens angesiedelt ist die Arbeitsgruppe Preissner, die
mit bioinformatischen Methoden molekulare Interaktionen untersucht. Basierend auf entsprechenden
Datenbanken und Suchalgorithmen erlaubt dieser
Ansatz die z.B. Identifikation geeigneter Kandiatenmoleküle für gezielte pharmakologische Beeinflussung relevanter zellulärer Abläufe. Ein Schwerpunkt
liegt hierbei in der Suche nach kleinen molekularen
Inhibitoren von zellulären Wachstumsfaktor-Rezeptoren und von nachgeschalteten Signalwegen, z.B.
für die Tumortherapie. Diese Studien werden in enger
Zusammenarbeit mit der AG Höpfner durchgeführt,
die primär an zellulären Modellen die Wirksamkeit
unterschiedlicher antioangionetischer TumorzellInhibitoren untersucht.
Eine wesentliche Erweiterung der methodischen
Möglichkeiten für die AG Höpfner ergibt sich wiederum aus der Nutzung der intravitalmikroskopischen
Ansätze der AG’s Pries und Kübler. So kann z.B. das
Wachstum und die Vaskularisierung unterschiedlicher Tumorzelllinien mit und ohne Applikation der
zu testenden Substanzen in situ in der Rückenhautkammer der Maus untersucht werden. In der
Arbeitsgruppe Pries werden zur Analyse derartiger
Daten und zur Entwicklung weitergehender Konzepte mathematische Simulationsverfahren eingesetzt. Mit Hilfe dieser Ansätze können Hypothesen
generiert werden, die dann experimentell untersucht
werden können. So wurde in den letzten Jahren die
Bedeutung intravaskulärer Kommunikation über
Axel Pries – PHYSIOLOGY
Gap-Junctions für Funktionsfähigkeit mikrovaskulärer Grefäßnetzwerke untersucht. In einem Artikel
in Nature Reviews Cancer (Pries et al. 2010) wird
die Hypothese ausgestellt, dass eine Fehlfunktion
dieser Kommunikation wesentlich an der Maldistribution der Perfusion in Tumoren beteiligt ist. Eine
Verbesserung dieser Funktion wäre demnach ein
Ansatz um den Effekt pharmakologischer Therapien
zu erhöhen.
Die quantitative Analyse der funktionellen Integration relevanter Mechanismen auf vaskulärer Ebene
im Organismus mit Hilfe der quantitativen Intravitalmikroskopie ist auch für andere Fragestellungen
von zunehmender Bedeutung und stellt eine zentrale Kompetenz der Gruppen Pries, Kübler und
Habazettl dar. Durch eine deutliche Verbesserung
der Meßmöglichkeiten dieses Ansatzes haben sich
neue Anwendungsfelder in der Untersuchung funktioneller, zellulärer und molekularer Mechanismen
erschlossen. Hierzu haben die genannten Arbeitsgruppen durch Entwicklung eigener Tiermodelle,
Meßverfahren und Bildanalysesoftware beigetragen.
Der Schwerpunkt der Arbeitsgruppe Gunga am
Physiologischen Institut der Charité in Dahlem liegt
auf dem Gebiet integrative Humanphysiologie, im
speziellen die Anpassungen des Herz-Kreislauf­
systems, der Thermoregulation sowie der Chronobiologie unter klinischen, labor- und feldphysiologischen extremen Bedingungen. Die Projekte
werden finanziell bzw. logistisch gefördert durch
das Bundesministerium für Wirtschaft (BMWi)/Deutsches Zentrum für Luft- und Raumfahrt (DLR), das
Alfred-Wegener-Institut für Polarforschung in Bremerhaven, die European Space Agency (ESA), die
National Space Administration (NASA), das Karlsruher Institute of Technology (KIT), das österreichische Bundesheer und die deutsche Bundeswehr,
den Deutschen Schwimmverband, die Deutsche
Lebensrettungsgesellschaft (DLRG) sowie die Humboldt- und Nathan-Zuntz-Stiftung. Mit integrativer
Forschung auf der Ebene der Humanphysiologie
auch an Patienten durch die AG Kunz, die sich mit
circadianen Rhythmen und ihre Beeinflussung durch
Lichteposition beschäftigt, vervollständigt sich der
Bogen „from Molecule to Organism“.
Der Arbeitsschwerpunkt der Gruppen Pries,
Kübler und Habazettl liegt im Bereich mikrovaskulärer Adaptationsvorgänge, der pulmonalen Endstrombahn und Untersuchungen der Mikrozirkulation am Menschen.
125
Axel Pries – PHYSIOLOGY
Research Projects
1. T
he shunt problem : control of functional shunting in normal and
tumour vasculature
Project leader Axel R Pries
Coworkers
Micheal Höpfner, Ferdiand Le Noble
Researchers know that the blood vessels in tumors
are poor in distriubuting oxygen and drugs, and comparing vascular networks in tumors with normal tissues may suggest new approaches to improving drug
delivery in tumors. Axel Pries, Michael Höpfner, and
Ferdinand le Noble of the CCR, Mark Dewhirst of Duke
University Medical Center and Timothy Secomb of the
University of Arizona have offered a new hypothesis.
Their argument, published in Nature Reviews Cancer1,
is that impaired communication along vessels in tumor
microvascular networks can lead to shunting of blood
away from some regions of the tumor and the generation of hypoxic regions (blue and purple in the graph)
There are always both long pathways and shortcuts from the
arteries to the veins. These shortcuts in tumors tend to grow
in diameter, shunting the flow away from the long pathways.
Anti-angiogenic treatments (treatments that inhibit the growth
of new vessels) may help to restore communication and
improve flow distribution in tumor vessels.
In detail, the communication along blood vessel walls, which
is needed to coordinate the distribution of blood flow was
investigated. Gap junctions connect the endothelial cells of
the vessels.It is theorized that the gap junctions do not exist or
don’t work well in tumors, which leads to loss of communication. The vessel structure in the tumor reflects this breakdown
of communication.
126
One line of attack would be to give a VEGF antagonist, to improve communication and reduce shunting,
which enables better delivery of anti-tumor therapies. It
is assumed that approaches targeted at improving gap
junction communication may be able to take advantage
of this concept in a more predictable manner.
(1) Pries AR, Hopfner M, Le Noble F., Dewhirst MW, Secomb
TW. The shunt problem: control of functional shunting in
normal and tumour vasculature. Nat Rev Cancer 2010 August
1;10(8):587-93.
Axel Pries – PHYSIOLOGY
2. Metabolic control of microvascular structure :
Where are the ox ygen sensors?
Project leader
Coworkers
Bettina Reglin
Axel R Pries, Tim Secomb
Generation and maintenance of functional vascular networks requires structural adaptation of vessel
diameters in response to the metabolic state of tissue.
The exact mechanism of this response is not known
but may involve the release of vasoactive substances
in response to low oxygen by tissue (“tissue signaling”,
e.g. CO2, adenosine), by vessel walls (“wall signaling”,
e.g. prostaglandins, adenosine) and/or by red blood
cells, RBCs (“RBC signaling”, e.g. ATP and NO).
sel walls or in RBC. Resulting network structures were
analyzed with regard to tissue oxygenation and to the
difference between estimated blood flow velocities
and corresponding experimental data (velocity error).
Wall signaling led to the highest hemodynamic similarity and low oxygen deficit. Tissue signaling also led
to low oxygen deficit, but resulted in higher velocity
error and less realistic diameters. RBC signaling led to
widespread hypoxia, unrealistic velocity distributions
and shrinkage of small vessels. The results suggest
that the metabolic signal for structural adaptation of
vessel diameters originates mainly in vessel walls.
Such data are used to develop an integrated view
of control of vascular structure and function, including short term changes of vessel diameter effected
by vascular smooth muscle and tone (right), long
term changes of vessel diameter and wall mass by
structural adaptation (middle), and changes in vessel number by elimination of unnecessary vessels by
pruning or generation of new vessels by the sprouting
and splitting modes of angiogenesis left).
In this project, the goal was to test the capability of
these three modes of oxygen sensing to adequately
control vascular diameters and tissue oxygenation. A
theoretical model of structural diameter adaptation
based on experimental data on microvascular network
structure and hemodynamics was used considering
oxygen-dependent metabolic signals in tissue, in ves127
Axel Pries – PHYSIOLOGY
3. Regulation of microvascular Permeability by Sphingolipids
Project leader
Coworkers
Funding
Cooperations
Wolfgang Kübler
Yang Yin
DFG- Focus “Sphingolipids“
Institut für Pharmakologie und Toxikologie, RWTH Aachen
Platelet-activating factor (PAF) increases pulmonary
micrivascular permeability within minutes. This effect
equally depnds on activation of cyclooxygenase and
acicd sphingomyelinase (ASM). Correspondingly,
pharmacological inhibition of ASM by imipramin
reduces acute lung injury in a number of preclinical
animal models. However, the mechanisms underlying
this new signal transduction pathway are still unresolved.
Caveolae are plasma domains which are speciffically
rich in sphingomyelin (the substrate of ASM), and
they contain Caveolin-1 (cav1) which
binds and blocks
endothelial NO synthase (eNOS). Therefore we analyzed the
relationship between
ASM, cav1 und eNOS
in the context of PAFinduced lung oedema
in the isolatd perfused
mouse and rat-lung.
Caveolae were isolated from pulmonary
endothelial cells after
marking with silica
beads preparing the
detergent-resistant
128
membrane fraction. Endthelial NOS was quantitatively
assessed by in situ fluoreszenz-microscopy. Caveolae, isolated 10 min after PAF stimulation showed an
increase in Caveolin-1, eNOS und ASM activity. PAF
in parralel reduced endothelial NO synthesis. These
effects could be reproduced by direct lung-perfusion
with ASM,while inhibition of the ASM signal pathway
by imipramin, D609 or dexamethason blocked PAFinduced increase in Caveolin-1 and eNOS in caveolae,
as well as the simultaneous decline in NO production,
and the generation of a PAF-induced lung oedma..
Restitution of NO levels by application of exogeneous NO donors reduce PAF-induced microvascular
barrier defect.
Thus PAF perturbates the microvascular barrier resulting in lug oedema. The mechanism includes activation
of ASM leading to ioncreased levels of Caveolin-1 and
eNOS in caveoli. This results in reduced endothelial
NO synthesis, which contributes to microvascular barrier defect.
Yang Y, Yin J, Baumgartner W, Samapati R, Solymosi EA,
Reppien E, Kuebler WM*, Uhlig S*: Platelet-activating factor
reduces endothelial NO production - role of acid sphingomyelinase. Eur Respir J 2010;36:417-27 (*shared senior authorship) sowie in Kuebler WM, Yang Y, Samapati R, Uhlig S:
Vascular barrier regulation by PAF, ceramide, caveolae, and
NO - an intricate signaling network with discrepant effects
in the pulmonary and systemic vasculature. Cell Physiol Biochem 2010;26:29-40.
Axel Pries – PHYSIOLOGY
4. N
ovel antiangiogenic compounds with antitumor activity for innovative
treatment approaches in cisplatin resistant urologic tumors
Project leader
Coworkers
Funding
Collaborators
Michael Höpfner
Bianca Nitzsche, Christoph Gloesenkamp, Björn Hoffmann
Stiftung Urologische Forschung Berlin
Prof. Dr. Mark Schrader, Department of Urology, University of Ulm
Prof. Dr, Matthias Ocker, Institute of Surgical Research, Philipps University Marburg
Testicular germ cell tumor (TGCT) is the most common
cause of death from solid tumors in young men and
especially for platinum-refractory TGCT patients novel
treatment approaches are urgently needed. As angiogenesis is essential for the development, growth and
metastases of tumors we hypothesised that targeting
angiogenic growth factor receptor signalling pathways
may be a promising approach for novel treatment
approaches of platinum resistant testicular germ cell
tumors (TGCT). Two VEGFR-blocking antiangiogenic
compounds, HP-2 and HP-14, that were recently identified by our group, were evaluated for their suitability to
inhibit the formation of tumor microvasculature as well
as the growth of normal and platinum-resistant TGCTs in
vitro and in vivo. Proliferation assays revealed the antineoplastic effects of HP-2 and HP-14 alone or in combina-
HP-2 and HP-14 induced vasodegeneration of
the developing CAM. Upper panel depicts CAMs
before treatment. In the lower panel effects of 48
h of treatment with PBS, HP-2 (10µM) or HP-14
(10µM) are shown. (a = artery and v = vein).
tion with platinum compounds in both platinum sensitive
and –resistant TGCT cell lines. For in vivo evaluation of
the antiangiogenic effects of the new compounds the so
called CAM assay (chicken chorioallantoic membrane
assay) was employed. 48 h incubation of the CAM of
fertilized chicken eggs with HP-2 or HP14 resulted in
a pronounced arrest in microvessel formation (Fig. 1).
Moreover TGCT cells inoculated onto the CAM were
treated with the HP-14. As depicted in Figure 2 a marked
inhibition of TGCT growth was observed upon treatment.
The novel compounds effectively suppressed the growth
of both platinum- sensitive as well as –resistant TGCTs.
Together, these data suggest that HP-2 and HP-14 may
be interesting new drugs for targeted therapy of urologic
cancers, particularly for those being resistant to the usually successful platinum-based interventions.
HP-14 induced inhibition of tumor growth of
TGCT cells on the chicken chorioallantoic
membrane (CAM). Cells were transplanted onto the CAM and treated
with HP-14 for 96h. Left side: Grown tumors were photographed (x6
magnification, representative images) and tumor volumes were calculated. Right side: Growth inhibition by HP-14 is depicted as the percentage of growth compared to those of untreated controls (means of n=3
independent experiments)
129
Axel Pries – PHYSIOLOGY
5. C
onnecting the regulation of angiogenesis to shear stress –
a new role for ADAMTS1
Project leader
Coworkers
External cooperations
Andreas Zakrzewicz
Margret Hohberg, medical student
Christian Hoffmann, medical student
Sven Chlench, physician
Luis Da Silva-Azevedo, PhD
Axel R. Pries
Robert Lehmann
ADAMTS1 inhibits capillary sprouting, and since
capillary sprouts do not experience the shear stress
caused by blood flow, this study undertook to clarify
the relationship between shear stress and ADAMTS1.
It was found that endothelial cells exposed to shear
stress displayed a strong upregulation of ADAMTS1,
dependent upon both the magnitude and duration of
their exposure. Investigation of the underlying pathways demonstrated involvement of phospholipase C,
phosphoinositid 3-kinase and nitric oxide. Forkhead
box protein O1 was identified as a likely inhibitor of
the system, as its knockdown was followed by a slight
increase in ADAMTS1 expression. In silico prediction
displayed a transcriptional binding site for forkhead
box protein O1 in the promotor region of the ADAMTS1
gene, as well as sites for nuclear factor 1, SP1 and
AP 1. The anti-angiogenic effects of ADAMTS1 were
attributed to its cleavage of thrombospondin 1 into
a 70-kDa fragment, and a significant enhancement
of this fragment was indeed demonstrated by immunoblotting shear stress-treated cells. Accordingly,
scratch wound closure displayed a slowdown in conditioned medium from shear stress-treated endothelial cells, an effect that could be completely blocked
by a knockdown of thrombospondin 1 and partially
130
blocked by a knockdown of ADAMTS1. Nonperfused
capillary sprouts in rat mesenteries stained negative
for ADAMTS1, while vessels in the microcirculation that
had already experienced blood flow yielded the opposite results. The shear stress-dependent expression
of ADAMTS1 in vitro was therefore also demonstrated
in vivo and thereby confirmed as a mechanism connecting blood flow with the regulation of angiogenesis.
Sprouting capillaries (visualized by staining with
GS-IB4) in rat mesenteries stain positive for Angiopoietin-2 but negative for ADAMTS1.
Axel Pries – PHYSIOLOGY
6. Possible atheroprotective effects of hyaluronic acid from the
endothelial surface layer and its modulation by shear stress
Project leader
Coworkers
External cooperations
Andreas Zakrzewicz, Günter Siegel
Julian Maroski, medical student
Bernd Vorderwülbecke, medical student
Eugeny Ermilov, PhD
Axel R. Pries
M. Malmsten, Uppsala University Biomedical Center, Uppsala
The endothelium is covered with a relatively thick (0.5 to oscillating (“atheroprone”) flow did not induce any dif1 µm) surface layer (ESL) which contains hyaluronic acid. ferences towards static control cultures. Nanoplaque
In arteries, there is a correlation between disturbed blood formation was measured by in situ ellipsometry. The
flow (reduced wall shear stress), reduction of the ESL and atherogenic lipoprotein fraction VLDL/IDL/LDL forms
a predilection for arteriosclerosis. This research project is ternary complexes (nanoplaques) on proteoheparan
designed to investigate if endothelial hyaluronan synthases sulfate-coated surfaces in a calcium dependant man(HAS) are regulated by shear stress and if hyaluronic acid ner. Hyaluronic acid proved to be similarly effective in
inhibiting this ternary complex formation as HDL.
has a role to prevent nanoplaque formation.
So far, human umbilical vein endothelial cells, were These data support the hypothesis that the synthesis
exposed to distinct flow conditions in a cone-and- of hyaluronic acid by endothelial cells contributes to
plate system, and analysed for hyaluronan synthase 2 shear stress-dependent athero-protective mechaexpression by real-time RT-PCR and immunoblotting, nisms especially during the initiation of arteriosclerosis
as well as for hyaluronan by ELISA, both in cell cul- by blocking ternary complex formation and do thus
ture supernatant and in a cell surface derived fraction. encourage further investigation.
Thereby
hyaluronan synthase 2 and
hyaluronan
were
found to be shear
stress-dependently
increased via the
PI3K-Akt-pathway.
Especially
atheroprotective flow was
found to induce
both – enzyme and Monomolecular coating of a methylated silica surface with proteoheparan sulfate molecules and their interaction both with Ca2+ and Na+ ions and lipoprotein particles in blood substitute solution (not to scale).
hyaluronan – effectively, while low and Siegel et al.: Biosens Bioelectron.18 (2003) 635-647; Siegel et al.: Biosens Bioelectron.24 (2009) 1512-1517
131
Axel Pries – PHYSIOLOGY
7. T
hermoregulation and cardiovascular adaptation in extreme
environments
Project leader
Coworkers
Hanns-Christian Gunga, PhD, MD, Dipl. Geol.
Oliver Opatz, Alexander Stahn, Mathias Steinach, MD, Andreas Werner, MD, LtCol MC
Space (Micro-g)
The current research projects of the group are structured
in studies under simulated (immersion, isolation, bed
rest, head-down-tilt) and real (parabolic flights, International Space Station ISS) micro-g conditions. The
immersion-, isolation-, bed rest- and head-down-tilt
studies under principal- and co-investigator guidance of
the group take place in close cooperation with the German Federal Armed Forces, the IMBP, ESA and NASA in
Manching (Flight-Medical Research Institute of the German Air Force), in Moscow (MARS 500), Berlin (Berlin
Bed Rest Study) and in Galveston (Mood Bed Study,
Texas). With the aid of the double sensor for non-invasive
determination of the body core temperature at the head
(Gunga et al., J Thermal Biology 33: 297-307, 2008),
developed in close cooperation with the company
Drägerwerke in Lübeck in the course of seven years of
intensive research work (Figure 1), the effectiveness of
protective suits as well as the effect of long-time isolation, confinement and immobilization on the temperature
regulation and chronobiology of the human organism
are investigated. Similar problems are addressed on
the International Space Station ISS by current studies
on astronauts as well as in parabolic flights with shorttime micro-g expositions. The latter studies are complemented by infrared photographs and Laser Doppler
method for the recording of changes in microcirculation.
The projects mentioned are carried out in close cooperation with the groups of Pries/Habazettl and Kunz. Joint
continuative studies are planned for 2011 in the human
centrifuge of the DLR in Cologne-Porz.
132
Data recorder (Flashmaster), temperature satellites
and two double sensors. To compare the size of the
technical systems, a 1 Euro coin is depicted.
Cold climate and hypothermia
The studies in this field of research are divided into two
main center points: On the one hand field physiological
studies in the Georg Neumayer and Concordia Stations
in the Antarctic (cold climate), on the other hand clinical
investigations on thermoregulation under moderate and
deep hypothermia during heart transplantations in the
German Heart Institute Berlin, the latter again in cooperation with the Pries group (Opatz et al. Resuscitation,
2010). In the Antarctic, changes in body composition
Axel Pries – PHYSIOLOGY
(muscle mass, fat mass, body water), various hormones
and circadian rhythm of the core temperatures under
long-time isolation and confinement are analyzed at sea
level (Georg Neumayer Station) and in comparison also
at an elevation of 3,800 m (Concordia Station).
Barophysiology
This research area as well is structured in two parts: hypoand hyperbaric physiology. Hypobaric studies are carried
out under laboratory conditions in the height chamber
of the German Federal Armed Forces at Königsbrück,
in order to elucidate the effects of hypobaric hypoxia
on disagreeable thermic sensations at the head. These
investigations are done in cooperation with the Flight
Medical Service of the German Federal Armed Forces.
Hyperbaric studies are carried out in cooperation with
the German Life Saving Association (DLRG) to enable a
continuous measurement of the body core temperature
of divers. The development of this sensor is based upon
the further development of the double sensor.
Publications ( 2006-2010 )
Akram A, Han B, Masoom H, Peng C, Lam E, Litvack L, Bai
X, Shan Y, Hai T, Batt J, Slutsky AS, Zhang H, Kuebler WM,
Haitsma JJ, Liu M, dos Santos CC (2010): Activating transcription factor 3 confers protection against ventilator-induced
lung injury. Am J Respir Crit Care Med 182(4):489-500
Boning D, Maassen N, Pries A (2010): The Hematocrit Paradox - How Does Blood Doping Really Work? Int J Sports
Med Epub ahead of print
Buschmann I, Pries A, Styp-Rekowska B, Hillmeister P,
Loufrani L, Henrion D, Shi Y, Duelsner A, Hoefer I, Gatzke
N, Wang H, Lehmann K, Ulm L, Ritter Z, Hauff P, Hlushchuk R, Djonov V, van Veen T, Le Noble F. (2010): Pulsatile
shear and Gja5 modulate arterial identity and remodeling
events during flow-driven arteriogenesis. Development
137(13):2187-96
Fung YL, Kim M, Tabuchi A, Aslam R, Speck ER, Chow L,
Kuebler WM, Freedman J, Semple JW (2010): Recipient T
lymphocytes modulate the severity of antibody-mediated
transfusion-related acute lung injury (TRALI). Blood Epub
ahead of print
Habazettl H, Athanasopoulos D, Kuebler WM, Wagner H,
Roussos C, Wagner PD, Ungruhe J, Zakynthinos S, Vogiatzis
I (2010): Near-infrared spectroscopy and indocyanine green
derived blood flow index for noninvasive measurement of muscle perfusion during exercise. J Appl Physiol 108(4):962-7
Hohberg M, Knochel J, Hoffmann CJ, Chlench S, Wunderlich
W, Alter A, Maroski J, Vorderwulbecke BJ, Silva-Azevedo
LD, Knudsen R, Lehmann R, Fiedorowicz K, Bongrazio M,
Nitsche B, Hoepfner M, Styp-Rekowska B, Pries AR, Zakrzewicz A (2010): Expression of ADAMTS1 in endothelial cells is
induced by shear stress and suppressed in sprouting capillaries. J Cell Physiol
Kerem A, Yin J, Kaestle SM, Hoffmann J, Schoene AM, Singh
B, Kuppe H, Borst MM, Kuebler WM (2010): Lung endothelial dysfunction in congestive heart failure: role of impaired
Ca2+ signaling and cytoskeletal reorganization. Circ Res
106(6):1103-16
Kuebler WM, Yang Y, Samapati R, Uhlig S (2010): Vascular
barrier regulation by PAF, ceramide, caveolae, and NO - an
intricate signaling network with discrepant effects in the
pulmonary and systemic vasculature. Cell Physiol Biochem
26(1):29-40
Meissner S, Tabuchi A, Mertens M, Kuebler WM, Koch E
(2010): Virtual four-dimensional imaging of lung parenchyma
by optical coherence tomography in mice. J Biomed Opt
15(3):036016
Nickles HT, Kuebler WM (2010): Take my breath away:
perivascular fluid cuffs impair lung mechanics. Crit Care
Med 38(6):1494-6
133
Axel Pries – PHYSIOLOGY
Nitzsche B, Gloesenkamp C, Schrader M, Ocker M, Preissner
R, Lein M, Zakrzewicz A, Hoffmann B, Hopfner M (2010):
Novel compounds with antiangiogenic and antiproliferative
potency for growth control of testicular germ cell tumours. Br
J Cancer 103(1):18-28
Zhou C, Chen H, King JA, Sellak H, Kuebler WM, Yin J,
Townsley MI, Shin HS, Wu S (2010): Alpha1G T-type calcium
channel selectively regulates P-selectin surface expression in
pulmonary capillary endothelium. Am J Physiol Lung Cell
Mol Physiol 299(1):L86-L97
Preissner S, Kroll K, Dunkel M, Senger C, Goldsobel G,
Kuzman D, Guenther S, Winnenburg R, Schroeder M,
Preissner R (2010): SuperCYP: a comprehensive database
on Cytochrome P450 enzymes including a tool for analysis
of CYP-drug interactions. Nucleic Acids Res 38(Database
issue):D237-D243
Alter A, Schmiedeck D, Fussnegger MR, Pries AR, Freesmeyer WB, Zakrzewicz A (2009): Angiopoietin-1, but not
platelet-derived growth factor-AB, is a cooperative stimulator
of vascular endothelial growth factor A-accelerated endothelial cell scratch closure. Ann Vasc Surg 23(2):239-45
Pries AR, Hopfner M, Le Noble F., Dewhirst MW, Secomb TW
(2010): The shunt problem: control of functional shunting in normal and tumour vasculature. Nat Rev Cancer 10(8):587-93
Sander PM, Christian A, Clauss M, Fechner R, Gee CT,
Griebeler EM, Gunga HC, Hummel J, Mallison H, Perry SF,
Preuschoft H, Rauhut OW, Remes K, Tutken T, Wings O, Witzel U (2010): Biology of the sauropod dinosaurs: the evolution
of gigantism. Biol Rev Camb Philos Soc
Sodian R, Schaefermeier P, begg-Zips S, Kuebler WM, Shakibaei M, Daebritz S, Ziegelmueller J, Schmitz C, Reichart B
(2010): Use of human umbilical cord blood-derived progenitor
cells for tissue-engineered heart valves. Ann Thorac Surg
89(3):819-28
Bauer RA, Gunther S, Jansen D, Heeger C, Thaben PF, Preissner R (2009): SuperSite: dictionary of metabolite and drug
binding sites in proteins. Nucleic Acids Res 37(Database
issue):D195-D200
Bergmann F, Breinig M, Hopfner M, Rieker RJ, Fischer L,
Kohler C, Esposito I, Kleeff J, Herpel E, Ehemann V, Friess
H, Schirmacher P, Kern MA (2009): Expression pattern and
functional relevance of epidermal growth factor receptor and
cyclooxygenase-2: novel chemotherapeutic targets in pancreatic endocrine tumors? Am J Gastroenterol 104(1):171-81
Bickenbach J, Dembinski R, Czaplik M, Meissner S, Tabuchi
A, Mertens M, Knels L, Schroeder W, Pelosi P, Koch E, Kuebler WM, Rossaint R, Kuhlen R (2009): Comparison of two in
vivo microscopy techniques to visualize alveolar mechanics. J
Clin Monit Comput 23(5):323-32
Valois CR, Braz JM, Nunes ES, Vinolo MA, Lima EC, Curi R,
Kuebler WM, Azevedo RB (2010): The effect of DMSA-functionalized magnetic nanoparticles on transendothelial migration of monocytes in the murine lung via a beta2 integrindependent pathway. Biomaterials 31(2):366-74
Bueltmann M, Kong X, Mertens M, Yin N, Yin J, Liu Z, Koster
A, Kuppe H, Kuebler WM (2009): Inhaled milrinone attenuates
experimental acute lung injury. Intensive Care Med 35(1):171-8
Von EJ, Gunther S, Preissner R (2010): Structural features
and evolution of protein-protein interactions. Genome Inform
22:1-10
Da Silva-Azevedo L, Jahne S, Hoffmann C, Stalder D, Heller
M, Pries AR, Zakrzewicz A, Baum O (2009): Up-regulation
of the peroxiredoxin-6 related metabolism of reactive oxygen
species in skeletal muscle of mice lacking neuronal nitric
oxide synthase. J Physiol 587(Pt 3):655-68
Yang Y, Yin J, Baumgartner W, Samapati R, Solymosi EA,
Reppien E, Kuebler WM, Uhlig S (2010): Platelet-activating
factor reduces endothelial nitric oxide production: role of acid
sphingomyelinase. Eur Respir J 36(2):417-27
Yin J, Kuebler WM (2010): Mechanotransduction by TRP
channels: general concepts and specific role in the vasculature. Cell Biochem Biophys 56(1):1-18
134
Dunkel M, Schmidt U, Struck S, Berger L, Gruening B,
Hossbach J, Jaeger IS, Effmert U, Piechulla B, Eriksson R,
Knudsen J, Preissner R (2009): SuperScent--a database
of flavors and scents. Nucleic Acids Res 37(Database
issue):D291-D294
Axel Pries – PHYSIOLOGY
Eisenreich A, Bogdanov VY, Zakrzewicz A, Pries A, Antoniak S, Poller W, Schultheiss HP, Rauch U (2009): Cdc2like kinases and DNA topoisomerase I regulate alternative
splicing of tissue factor in human endothelial cells. Circ Res
104(5):589-99
Franz R, Hummel J, Kienzle E, Kolle P, Gunga HC, Clauss
M (2009): Allometry of visceral organs in living amniotes
and its implications for sauropod dinosaurs. Proc Biol Sci
276(1662):1731-6
Fullbeck M, Gebhardt N, Hossbach J, Daniel PT, Preissner
R (2009): Computer-assisted identification of small-molecule
Bcl-2 modulators. Comput Biol Chem 33(6):451-6
Fullbeck M, Dunkel M, Hossbach J, Daniel PT, Preissner R
(2009): Cellular fingerprints: a novel approach using largescale cancer cell line data for the identification of potential
anticancer agents. Chem Biol Drug Des 74(5):439-48
Gunga HC, Werner A, Stahn A, Steinach M, Schlabs T,
Koralewski E, Kunz D, Belavy DL, Felsenberg D, Sattler F,
Koch J (2009): The Double Sensor-A non-invasive device to
continuously monitor core temperature in humans on earth
and in space. Respir Physiol Neurobiol 169 Suppl 1:S63S68
Gunther S, Von EJ, May P, Preissner R (2009): JAIL: a
structure-based interface library for macromolecules. Nucleic
Acids Res 37(Database issue):D338-D341
Hildebrand PW, Goede A, Bauer RA, Gruening B, Ismer J,
Michalsky E, Preissner R (2009): SuperLooper--a prediction
server for the modeling of loops in globular and membrane
proteins. Nucleic Acids Res 37(Web Server issue):W571W574
Hossbach J, Michalsky E, Henklein P, Jaeger M, Daniel PT,
Preissner R (2009): Inhibiting the inhibitors: retro-inverso
Smac peptides. Peptides 30(12):2374-9
Lipnicki DM, Gunga HC, Belavy DL, Felsenberg D (2009): Bed
rest and cognition: effects on executive functioning and reaction time. Aviat Space Environ Med 80(12):1018-24
Lipnicki DM, Gunga HC, Belavy DL, Felsenberg D (2009):
Decision making after 50 days of simulated weightlessness.
Brain Res 1280:84-9
Lipnicki DM, Gunga HC (2009): Physical inactivity and cognitive functioning: results from bed rest studies. Eur J Appl
Physiol 105(1):27-35
Mecha DN, Styp-Rekowska B, Hinz B, Da Silva-Azevedo
L, Pries AR, Zakrzewicz A (2009): Differential expression of
VEGFA, TIE2, and ANG2 but not ADAMTS1 in rat mesenteric
microvascular arteries and veins. Physiol Res 58(2):193-202
Mertens M, Tabuchi A, Meissner S, Krueger A, Schirrmann
K, Kertzscher U, Pries AR, Slutsky AS, Koch E, Kuebler WM
(2009): Alveolar dynamics in acute lung injury: heterogeneous
distension rather than cyclic opening and collapse. Crit Care
Med 37(9):2604-11
Paris S, Wolgin M, Kielbassa AM, Pries A, Zakrzewicz A
(2009): Gene expression of human beta-defensins in healthy
and inflamed human dental pulps. J Endod 35(4):520-3
Pries AR, Cornelissen AJ, Sloot AA, Hinkeldey M, Dreher MR,
Hopfner M, Dewhirst MW, Secomb TW (2009): Structural
adaptation and heterogeneity of normal and tumor microvascular networks. PLoS Comput Biol 5(5):e1000394
Pries AR, Secomb TW (2009): Origins of heterogeneity in tissue perfusion and metabolism. Cardiovasc Res 81(2):32835
Reglin B, Secomb TW, Pries AR (2009): Structural adaptation
of microvessel diameters in response to metabolic stimuli:
where are the oxygen sensors? Am J Physiol Heart Circ
Physiol 297(6):H2206-H2219
Rother K, Hildebrand PW, Goede A, Gruening B, Preissner
R (2009): Voronoia: analyzing packing in protein structures.
Nucleic Acids Res 37(Database issue):D393-D395
Schmidt U, Struck S, Gruening B, Hossbach J, Jaeger
IS, Parol R, Lindequist U, Teuscher E, Preissner R (2009):
SuperToxic: a comprehensive database of toxic compounds.
Nucleic Acids Res 37(Database issue):D295-D299
Verdant CL, De BD, Bruhn A, Clausi CM, Su F, Wang Z, Rodriguez H, Pries AR, Vincent JL (2009): Evaluation of sublingual
and gut mucosal microcirculation in sepsis: a quantitative
analysis. Crit Care Med 37(11):2875-81
135
Axel Pries – PHYSIOLOGY
Vogiatzis I, Athanasopoulos D, Habazettl H, Kuebler WM,
Wagner H, Roussos C, Wagner PD, Zakynthinos S (2009):
Intercostal muscle blood flow limitation in athletes during
maximal exercise. J Physiol 587(Pt 14):3665-77
Hildebrand PW, Gunther S, Goede A, Forrest L, Frommel C,
Preissner R (2008): Hydrogen-bonding and packing features
of membrane proteins: functional implications. Biophys J
94(6):1945-53
Yin J, Kuebler WM (2009): New targets in pulmonary
hypertension--another ACE up the sleeve. Am J Respir Crit
Care Med 180(5):481-2
Hopfner M, Schuppan D, Scherubl H (2008): Treatment of
gastrointestinal neuroendocrine tumors with inhibitors of
growth factor receptors and their signaling pathways: recent
advances and future perspectives. World J Gastroenterol
14(16):2461-73
Yin N, Kaestle S, Yin J, Hentschel T, Pries AR, Kuppe H,
Kuebler WM (2009): Inhaled nitric oxide versus aerosolized
iloprost for the treatment of pulmonary hypertension with left
heart disease. Crit Care Med 37(3):980-6
Bauer RA, Rother K, Bujnicki JM, Preissner R (2008): Suffix
techniques as a rapid method for RNA substructure search.
Genome Inform 20:183-98
Bauer RA, Bourne PE, Formella A, Frommel C, Gille C, Goede
A, Guerler A, Hoppe A, Knapp EW, Poschel T, Wittig B,
Ziegler V, Preissner R (2008): Superimpose: a 3D structural
superposition server. Nucleic Acids Res 36(Web Server
issue):W47-W54
Boldt LH, Fraszl W, Rocker L, Schefold JC, Steinach M,
Noack T, Gunga HC (2008): Changes in the haemostatic system after thermoneutral and hyperthermic water immersion.
Eur J Appl Physiol 102(5):547-54
Boning D, Maassen N, Pries A (2008): No proof for augmented
arterial oxygen content as only factor influencing exercise
capacity after Epo doping. J Appl Physiol 105(6):1988
Dunkel M, Gunther S, Ahmed J, Wittig B, Preissner R (2008):
SuperPred: drug classification and target prediction. Nucleic
Acids Res 36(Web Server issue):W55-W59
Frassl W, Kowoll R, Katz N, Speth M, Stangl A, Brechtel L,
Joscht B, Boldt LH, Meier-Buttermilch R, Schlemmer M,
Roecker L, Gunga HC (2008): Cardiac markers (BNP, NT-proBNP, Troponin I, Troponin T, in female amateur runners before
and up until three days after a marathon. Clin Lab 54(3-4):81-7
Gunther S, Kuhn M, Dunkel M, Campillos M, Senger C, Petsalaki
E, Ahmed J, Urdiales EG, Gewiess A, Jensen LJ, Schneider R,
Skoblo R, Russell RB, Bourne PE, Bork P, Preissner R (2008):
SuperTarget and Matador: resources for exploring drug-target
relationships. Nucleic Acids Res 36(Database issue):D919-D922
136
Hopfner M, Schuppan D, Scherubl H (2008): Targeted medical therapy of biliary tract cancer: recent advances and future
perspectives. World J Gastroenterol 14(46):7021-32
Hopfner M, Schuppan D, Scherubl H (2008): Growth factor
receptors and related signalling pathways as targets for novel
treatment strategies of hepatocellular cancer. World J Gas­
troenterol 14(1):1-14
Huebener N, Fest S, Strandsby A, Michalsky E, Preissner R,
Zeng Y, Gaedicke G, Lode HN (2008): A rationally designed
tyrosine hydroxylase DNA vaccine induces specific antineuroblastoma immunity. Mol Cancer Ther 7(7):2241-51
Kuebler WM (2008): Hitting new barriers in ventilator-induced
lung injury. Intensive Care Med 34(4):592-4
Kuebler WM (2008): How NIR is the future in blood flow monitoring? J Appl Physiol 104(4):905-6
Le NF, Klein C, Tintu A, Pries A, Buschmann I (2008): Neural
guidance molecules, tip cells, and mechanical factors in vascular development. Cardiovasc Res 78(2):232-41
Pries AR, Habazettl H, Ambrosio G, Hansen PR, Kaski JC,
Schachinger V, Tillmanns H, Vassalli G, Tritto I, Weis M, de
WC, Bugiardini R (2008): A review of methods for assessment
of coronary microvascular disease in both clinical and experimental settings. Cardiovasc Res 80(2):165-74
Pries AR, Secomb TW (2008): Modeling structural adaptation
of microcirculation. Microcirculation 15(8):753-64
Pries AR, Mulvany MJ, Bakker EN (2008): MBEC special
issue on microcirculation “engineering principles of vascular
networks”. Med Biol Eng Comput
Axel Pries – PHYSIOLOGY
Redlin M, Koster A, Huebler M, Boettcher W, Nagdyman N,
Hetzer R, Kuppe H, Kuebler WM (2008): Regional differences
in tissue oxygenation during cardiopulmonary bypass for
correction of congenital heart disease in neonates and small
infants: relevance of near-infrared spectroscopy. J Thorac
Cardiovasc Surg 136(4):962-7
Baradari V, Hopfner M, Huether A, Schuppan D, Scherubl H
(2007): Histone deacetylase inhibitor MS-275 alone or combined with bortezomib or sorafenib exhibits strong antiproliferative action in human cholangiocarcinoma cells. World J
Gastroenterol 13(33):4458-66
Schmidt U, Ahmed J, Michalsky E, Hoepfner M, Preissner R
(2008): Comparative VEGF receptor tyrosine kinase modeling
for the development of highly specific inhibitors of tumor
angiogenesis. Genome Inform 20:243-51
Chlench S, Mecha DN, Hohberg M, Hoffmann C, Pohlkamp
T, Beyer G, Bongrazio M, Da Silva-Azevedo L, Baum O,
Pries AR, Zakrzewicz A (2007): Regulation of Foxo-1 and
the angiopoietin-2/Tie2 system by shear stress. FEBS Lett
581(4):673-80
Schobersberger W, Toff WD, Eklof B, Fraedrich G, Gunga
HC, Haas S, Landgraf H, Lapostolle F, Partsch H, Perschler F,
Schnapka J, Schobersberger B, Scurr JH, Watzke H (2008):
Traveller’s thrombosis: international consensus statement.
VASA 37(4):311-7
Gunga HC, Kirsch KA, Roecker L, Kohlberg E, Tiedemann
J, Steinach M, Schobersberger W (2007): Erythropoietin
regulations in humans under different environmental and
experimental conditions. Respir Physiol Neurobiol 158(23):287-97
Schwarzer R, Kaiser M, Acikgoez O, Heider U, Mathas S, Preissner R, Sezer O, Doerken B, Jundt F (2008): Notch inhibition
blocks multiple myeloma cell-induced osteoclast activation.
Leukemia 22(12):2273-7
Gunga HC, Suthau T, Bellmann A, Friedrich A, Schwanebeck
T, Stoinski S, Trippel T, Kirsch K, Hellwich O (2007): Body
mass estimations for Plateosaurus engelhardti using laser
scanning and 3D reconstruction methods. Naturwissen­
schaften 94(8):623-30
Secomb TW, Beard DA, Frisbee JC, Smith NP, Pries AR
(2008): The role of theoretical modeling in microcirculation
research. Microcirculation 15(8):693-8
Struck S, Schmidt U, Gruening B, Jaeger IS, Hossbach J,
Preissner R (2008): Toxicity versus potency: elucidation of
toxicity properties discriminating between toxins, drugs, and
natural compounds. Genome Inform 20:231-42
Tabuchi A, Kuebler WM (2008): Endothelium-platelet interactions in inflammatory lung disease. Vascul Pharmacol
Tabuchi A, Mertens M, Kuppe H, Pries AR, Kuebler WM
(2008): Intravital microscopy of the murine pulmonary microcirculation. J Appl Physiol 104(2):338-46
Yin J, Hoffmann J, Kaestle SM, Neye N, Wang L, Baeurle J,
Liedtke W, Wu S, Kuppe H, Pries AR, Kuebler WM (2008):
Negative-feedback loop attenuates hydrostatic lung edema
via a cGMP-dependent regulation of transient receptor potential vanilloid 4. Circ Res 102(8):966-74
Ahmed J, Gunther S, Moller F, Preissner R (2007): A structural
genomics approach to the regulation of apoptosis: chimp vs.
human. Genome Inform 18:22-34.
Gunther S, May P, Hoppe A, Frommel C, Preissner R (2007):
Docking without docking: ISEARCH--prediction of interactions using known interfaces. Proteins 69(4):839-44
Gunther S, Hempel D, Dunkel M, Rother K, Preissner R
(2007): SuperHapten: a comprehensive database for small
immunogenic compounds. Nucleic Acids Res 35(Database
issue):D906-D910
Hentschel T, Yin N, Riad A, Habbazettl H, Weimann J, Koster
A, Tschope C, Kuppe H, Kuebler WM (2007): Inhalation of the
phosphodiesterase-3 inhibitor milrinone attenuates pulmonary hypertension in a rat model of congestive heart failure.
Anesthesiology 106(1):124-31
Huether A, Hopfner M, Baradari V, Schuppan D, Scherubl
H (2007): Sorafenib alone or as combination therapy for
growth control of cholangiocarcinoma. Biochem Pharmacol
73(9):1308-17
Kaestle SM, Reich CA, Yin N, Habazettl H, Weimann J, Kuebler WM (2007): Nitric oxide-dependent inhibition of alveolar
fluid clearance in hydrostatic lung edema. Am J Physiol
Lung Cell Mol Physiol 293(4):L859-69
137
Axel Pries – PHYSIOLOGY
Kuebler WM, Parthasarathi K, Lindert J, Bhattacharya
J (2007): Real-time lung microscopy. J Appl Physiol
102(3):1255-64
Meinke M, Schroder M, Schutz R, Netz U, Helfmann J, Dorschel K, Pries A, Muller G (2007): Frequency weighted laser
Doppler perfusion measurements in skin. Laser Physics
Letters 4(1):66-71
Mittermayr M, Fries D, Gruber H, Peer S, Klingler A, Fischbach U, Gunga HC, Koralewski E, Faulhaber M, Simmer
M, Schobersberger W (2007): Leg edema formation and
venous blood flow velocity during a simulated long-haul flight.
Thromb Res 120(4):497-504
Mulder ER, Kuebler WM, Gerrits KH, Rittweger J, Felsenberg
D, Stegeman DF, De HA (2007): Knee extensor fatigability
after bedrest for 8 weeks with and without countermeasure.
Muscle Nerve 36(6):798-806
Schobersberger W, Mittermayr M, Fries D, Innerhofer P,
Klingler A, Faulhaber M, Gunga HC, Streif W (2007): Changes
in blood coagulation of arm and leg veins during a simulated
long-haul flight. Thromb Res 119(3):293-300
Secomb TW, Styp-Rekowska B, Pries AR (2007): Two-dimensional simulation of red blood cell deformation and lateral
migration in microvessels. Ann Biomed Eng 35(5):755-65
Spohr F, Busch CJ, Reich C, Motsch J, Gebhard MM, Kuebler
WM, Bloch KD, Weimann J (2007): 4-Aminopyridine restores
impaired hypoxic pulmonary vasoconstriction in endotoxemic
mice. Anesthesiology 107(4):597-604
Bongrazio M, Da Silva-Azevedo L, Bergmann EC, Baum
O, Hinz B, Pries AR, Zakrzewicz A (2006): Shear stress
modulates the expression of thrombospondin-1 and CD36
in endothelial cells in vitro and during shear stress-induced
angiogenesis in vivo. Int J Immunopathol Pharmacol
19(1):35-48
Brueckl C, Kaestle S, Kerem A, Habazettl H, Krombach F,
Kuppe H, Kuebler WM (2006): Hyperoxia-induced reactive
oxygen species formation in pulmonary capillary endothelial
cells in situ. Am J Respir Cell Mol Biol 34(4):453-63
Dunkel M, Fullbeck M, Neumann S, Preissner R (2006):
SuperNatural: a searchable database of available natural
compounds. Nucleic Acids Res 34(Database issue):D678D683
Fest S, Huebener N, Weixler S, Bleeke M, Zeng Y, Strandsby
A, Volkmer-Engert R, Landgraf C, Gaedicke G, Riemer AB,
Michalsky E, Jaeger IS, Preissner R, Forster-Wald E, JensenJarolim E, Lode HN (2006): Characterization of GD2 peptide
mimotope DNA vaccines effective against spontaneous neuroblastoma metastases. Cancer Res 66(21):10567-75
Fullbeck M, Michalsky E, Jaeger IS, Henklein P, Kuhn H,
Ruck-Braun K, Preissner R (2006): Design and biological
evaluation of photo-switchable inhibitors. Genome Inform
17(1):141-51
Goede A, Michalsky E, Schmidt U, Preissner R (2006):
SuperMimic--fitting peptide mimetics into protein structures.
BMC Bioinformatics 7:11
Struijker-Boudier HA, Rosei AE, Bruneval P, Camici PG, Christ
F, Henrion D, Levy BI, Pries A, Vanoverschelde JL (2007):
Evaluation of the microcirculation in hypertension and cardiovascular disease. Eur Heart J 28(23):2834-40
Greie S, Humpeler E, Gunga HC, Koralewski E, Klingler A,
Mittermayr M, Fries D, Lechleitner M, Hoertnagl H, Hoffmann
G, Strauss-Blasche G, Schobersberger W (2006): Improvement of metabolic syndrome markers through altitude specific
hiking vacations. J Endocrinol Invest 29(6):497-504
Styp-Rekowska B, Disassa NM, Reglin B, Ulm L, Kuppe
H, Secomb TW, Pries AR (2007): An imaging spectroscopy
approach for measurement of oxygen saturation and hematocrit
during intravital microscopy. Microcirculation 14(3):207-21
Gunther S, Senger C, Michalsky E, Goede A, Preissner R
(2006): Representation of target-bound drugs by computed
conformers: implications for conformational libraries. BMC
Bioinformatics 7:293
Baradari V, Huether A, Hopfner M, Schuppan D, Scherubl H
(2006): Antiproliferative and proapoptotic effects of histone
deacetylase inhibitors on gastrointestinal neuroendocrine
tumor cells. Endocr Relat Cancer 13(4):1237-50
Habazettl H, Kukucka M, Weng YG, Kuebler WM, Hetzer R,
Kuppe H, Pries AR (2006): Arteriolar blood flow pulsatility in
a patient before and after implantation of an axial flow pump.
Ann Thorac Surg 81(3):1109-11
138
Axel Pries – PHYSIOLOGY
Herre S, Schadendorf T, Ivanov I, Herrberger C, Steinle W,
Ruck-Braun K, Preissner R, Kuhn H (2006): Photoactivation
of an inhibitor of the 12/15-lipoxygenase pathway. Chembio­
chem 7(7):1089-95
Hildebrand PW, Lorenzen S, Goede A, Preissner R (2006):
Analysis and prediction of helix-helix interactions in membrane channels and transporters. Proteins 64(1):253-62
Hopfner M, Baradari V, Huether A, Schofl C, Scherubl H
(2006): The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine
gastrointestinal tumours. Endocr Relat Cancer 13(1):135-49
Hopfner M, Huether A, Sutter AP, Baradari V, Schuppan
D, Scherubl H (2006): Blockade of IGF-1 receptor tyrosine
kinase has antineoplastic effects in hepatocellular carcinoma
cells. Biochem Pharmacol 71(10):1435-48
Hopfner M, Sutter AP, Huether A, Baradari V, Scherubl H
(2006): Tyrosine kinase of insulin-like growth factor receptor as target for novel treatment and prevention strategies of
colorectal cancer. World J Gastroenterol 12(35):5635-43
Roecker L, Kowoll R, Fraszl W, Battal K, Brechtel L, Brachmann S, Meier-Buttermilch R, Gunga HC, Stangl A, Kiesewetter H (2006): Observation of serum erythropoietin concentrations in female athletes for up to eight days after a marathon
run. Clin Lab 52(9-10):511-3
Secomb TW, Hsu R, Pries AR (2006): Tribology of capillary
blood flow. Proceedings of the Institution of Mechani­
cal Engineers Part J-Journal of Engineering Tribology
220(J8):767-74
Sodian R, Lueders C, Kraemer L, Kuebler W, Shakibaei M,
Reichart B, Daebritz S, Hetzer R (2006): Tissue engineering of
autologous human heart valves using cryopreserved vascular
umbilical cord cells. Ann Thorac Surg 81(6):2207-16
Steinbrink J, Fischer T, Kuppe H, Hetzer R, Uludag K, Obrig
H, Kuebler WM (2006): Relevance of depth resolution for
cerebral blood flow monitoring by near-infrared spectroscopic
bolus tracking during cardiopulmonary bypass. J Thorac
Cardiovasc Surg 132(5):1172-8
Hudlicka O, Brown MD, May S, Zakrzewicz A, Pries AR
(2006): Changes in capillary shear stress in skeletal muscles
exposed to long-term activity: role of nitric oxide. Microcir­
culation 13(3):249-59
Valenti G, Fraszl W, Addabbo F, Tamma G, Procino G, Satta
E, Cirillo M, De Santo NG, Drummer C, Bellini L, Kowoll R,
Schlemmer M, Vogler S, Kirsch KA, Svelto M, Gunga HC
(2006): Water immersion is associated with an increase in
aquaporin-2 excretion in healthy volunteers. Biochim Bio­
phys Acta 1758(8):1111-6
Huether A, Hopfner M, Sutter AP, Baradari V, Schuppan D,
Scherubl H (2006): Signaling pathways involved in the inhibition of epidermal growth factor receptor by erlotinib in hepatocellular cancer. World J Gastroenterol 12(32):5160-7
Williams JL, Weichert A, Zakrzewicz A, Da Silva-Azevedo L,
Pries AR, Baum O, Egginton S (2006): Differential gene and
protein expression in abluminal sprouting and intraluminal
splitting forms of angiogenesis. Clin Sci (Lond) 110(5):587-95
Kuebler WM (2006): Selectins revisited: the emerging role
of platelets in inflammatory lung disease. J Clin Invest
116(12):3106-8
Book chapters
Opitz B, Vinzing M, van L, V, Schmeck B, Heine G, Gunther S,
Preissner R, Slevogt H, N’Guessan PD, Eitel J, Goldmann T,
Flieger A, Suttorp N, Hippenstiel S (2006): Legionella pneumophila
induces IFNbeta in lung epithelial cells via IPS-1 and IRF3, which
also control bacterial replication. J Biol Chem 281(47):36173-9
Redlin M, Boettcher W, Huebler M, Berger F, Hetzer R, Koster A,
Kuebler WM (2006): Detection of lower torso ischemia by near-infrared spectroscopy during cardiopulmonary bypass in a 6.8-kg infant
with complex aortic anatomy. Ann Thorac Surg 82(1):323-5
Pries AR, Secomb TW. Blood flow in microvascular networks.
In: Tuma RF, Durán WN, Ley K, editors. Handbook of Physi­
ology: Microcirculation. 2 ed. San Diego: Academic Press,
Elsevier; 2008. p. 3-36
Secomb TW, Pries AR. Basic Principles of Hemodynamics.
In: Baskurt OK, hardeman MW, Rampling MW, Meiselman
HJ, editors. Handbook of Hemorheology and Hemody­
namics. Amsterdam: IOS Press; 2007
Pries AR, Kuebler WM (2006): Normal endothelium. Handb
Exp Pharmacol (176 Pt 1):1-40
139
Axel Pries – PHYSIOLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Kübler W.
Blottner D.
Blottner D.
Siegel G.
Kunz D.
Kunz D.
Kunz D.
Kunz D.
Gunga H.C.
Gunga H.C.
Gunga H.C.
Gunga H.C.
Gunga H.C.
Gunga H.C.
Munz B.
Munz B.
Munz B.
Munz B.
140
Project title
SFB Transregio 19
Protektive Beatmung TP1
Protektive Beatmung TP2
Strömungsmechanik
Sphingolipide FP 1
Sphingolipide FP 2
Angiotensin(1-7)
Totraumventilation
Vscular regulation
Pulmotension
Vasodilatory effect of RO50241
Therapeutic potential of oral LF1166910
Sildenafil citrate
Connexin 40
Pulmonary hypertension
Molekulare Zellstrukturen
Bed Rest Study
Fluvastatin
PLACAR
Energieoptimiertes Bauen: Beleuchtung
Nikotin und Schlaf
Fit for School
Langzeitaufenthalte unter Schwerelosigkeit
MILIEU
Spacelife
Brain function and physical exercise
Fluid Shift - Thermolab
Thermolab
Bedeutung von sKNac
Rhabdomyosarkomzellen
Rolle von Rip-Protein_2
ZFP 36 Proteine
Sponsor
DFG
DFG
DFG
DFG
DFG
DFG
DFG
DFG
DFG (GRK865)
EU
Hoffmann–La Roche Inc
Solvay Pharmaceuticals
Pfizer Inc
CIHR
HSFC
BMBF
EU
Industrie
BMBF
BMWi
DFG
BMBF
BMBF
Exzellenzcluster FU
Graduiertenkolleg DLR
Karlsruhe Institute of Technology (KIT)
BMBF
BMBF
DHSF
Kutzner Stiftung
DFG
DFG
Period
2008-2012
2005-2007
2007-2009
2009-2011
2007-2010
2010-2013
2009-2011
2009-2011
2003-2008
2006-2009
2009-2011
2009-2011
2004-2011
2009-2012
2010-2012
2005-2008
2007-2010
2001-2005
2006-2010
2007-2010
2006-2011
2007-2010
2002-2007
2009-2011
2008-2010
2010
2007-2010
2010-2013
2007-2010
2009-2011
2006-2009
2010-2014
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Head of the group
Prof. Dr. med. Uwe Querfeld
Curriculum Vitae:
1979‑1984 Training in Pediatrics at the Children’s Hospital Heidelberg
University. Specialization in Pediatric Nephrology
1984‑1986 Fellow, Division for Pediatric Nephrology, UCLA Medical School,
Los Angeles,
Bench training in lipid research at Los Angeles Veterans
Administration Medical Center.
1987‑1988 Advanced Research Fellowship grant by the American Heart Association
1987-1988Fellow, Division for Pediatric Nephrology, Cedars‑Sinai Medical Center, Los Angeles
1991-1999Attending physician in Pediatrics, University Children’s Hospital, Cologne, Germany
Since 10/99 Director, Department of Pediatric Nephrology, Charité Berlin, Germany.
2003European Soecity for Pediatric Nephrology (ESPN) Congress President,
World Congress of Nephrology
Since 2004
Head of a research group at the Center for Cardiovascular Research, Berlin
Main scientific interest:Cardiovascular disease in children and adolescents with chronic kidney disease,
prevention of atherosclerosis, non-invasive imaging methods of arterial structure and
function, vascular calcification, vitamin D metabolism, lipid metabolism
Members of the group
Scientists
Müller, Dominik
Stuiver, Marchel
Thumfart, Julia
PD Dr. med.
Dipl. – Molbiol.
Dr. med.
Technicians
Sommer, Kerstin MTA
Students
Diercke, Michaela
Jung, Susanne
Kopplin, Kathrin
Hecht, Eva
Will, Constanze
Medical Student
PhD student
PhD student
PhD student
PhD student
141
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Summary
Our group conducts translational research in cardiovascular aspects of clinical pediatric nephrology
and vascular and renal physiology.
metalloproteinases (MMP-2 and MMP-9) on highdose calcitriol induced vascular calcifications in
uremic animals.
Vitamin D- induced vascular calcification in
chronic kidney disease
Whether treatment with vitamin D receptor activators
(VDRA) contributes to cardiovascular disease (CVD) in
patients with chronic kidney disease (CKD) is a matter
of debate. Observational studies in adult CKD patients
show that VDRA provide a survival benefit associated
with a decrease in CVD-related mortality. These data
are in contrast to studies in children and adolescents
and in young adults with childhood-onset CKD, where
treatment with 1-alpha or 1,25(OH)2D3 is associated
with the occurrence of arterial calcifications. As a rule,
children and adolescents require comparatively high
doses of VDRA to prevent the development of rickets
in the growing skeleton.
We are therefore interested in studying the mechanisms leading to vitamin D-induced calcifications by
using an animal model and cell culture experiments.
The Cardiovascular Comorbidity in Children
with Chronic Kidney Disease Study (4 C Study)
In this prospective observational study of children
and adolescents with CKD, a comprehensive study
of arterial biopsy specimens, usually taken at the
time of AV fistula surgery or pre-emptive renal
transplantation, is performed for histopathological
assessment in a biopsy sub-study. Data will be analyzed for associations with the clinical progression
of renal and cardiovascular disease as monitored
prospectively by non-invasive imaging methods.
We are currently studying the relative contribution of
calcitriol treatment and elevated PTH levels to vascular calcifications. In animal experiments, we have
investigated the effect of cinacalcet, a modulator
of the calcium-sensing receptor in the parathyroid
gland, and of parathyroidectomy (PTX), on highdose calcitriol induced vascular calcifications in
uremic rats. In addition, we are studying the effect
of high-dose calcitriol, Cinacalcet and PTX on uremic bone disease In cooperation with Laboratory
of Pathophysiology, University of Antwerp, Belgium.
We have also initiated studies of the role of matrix
142
Claudin tight junction proteins in renal ion
homeostasis
Tight junction proteins of the Claudin family are tetraspanning transmembrane proteins which serve as
a semipermeable barrier in epithelia. They regulate
the paracellular diffusion of solutes through epithelial and endothelial cell layers. The different family
members render the epithelia leaky or tight and
contribute to the specifity of ions and molecules
that are absorbed. In the kidney, Claudins show a
distinct expression pattern along the tubule and are
involved in paracellular ion resorption and contribution to urine formation, thus they contribute to volume regulation and blood pressure control. We are
evaluating the physiological and pathophysiological
role Claudin-3, -10 and -16 which are all expressed
in the thick ascending limb (TAL) of Henle´s loop a
crucial region of volume regulation and blood pressure control.
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Zusammenfassung
Unsere Arbeitsgruppe befasst sich mit kardiovaskulären Aspekten der klinischen pädiatrischen Nephrologie und der Gefäß- und Nierenphysiologie.
Vitamin D-induzierte vaskuläre Verkalkungen
bei chronischer Niereninsuffizienz
Ob Vitamin D-Präparate zur hohen kardiovaskulären Morbidität und Mortalität bei chronischer Niereninsuffizienz beitragen, wird gegenwärtig kontrovers diskutiert. Einerseits fanden epidemiologischen Studien bei erwachsenen Patienten einen
günstigen Effekt in Hinblick auf kardiovaskuläre
Komplikationen und Überlebensstatistiken; anderseits haben Studien bei Kindern gezeigt, dass das
Auftreten von koronaren Verkalkungen (unabhängig
von anderen Risikofaktoren) mit der Dosierung von
Vitamin D-Präparaten verbunden war. Im Prinzip
müssen Kinder und Jugendliche dauerhaft und mit
höheren Dosen als Erwachsene behandelt werden,
um das Auftreten von rachitischen Erkrankungen im
wachsenden Skelett zu verhindern. Wir sind deswegen daran interessiert, die zugrunde liegenden
Mechanismen der Vitamin D-induzierten Gefäßverkalkung besser zu charakterisieren.
Gegenwärtig untersuchen wir die Frage, in welchem
Ausmaß erhöhte PTH-Spiegel bei der Entstehung
von Vitamin D-induzierten Verkalkungen beteiligt
sind. Zu diesem Zweck werden in Experimenten an
niereninsuffizienten Ratten, die unter einer hochdosierten Calcitriol Gabe ausgeprägte Verkalkungen
entwickeln, parathyreoidektomierte und (mit dem
Calcimimetikum) Cinacalcet- behandelte Tiere mit
urämischen Kontrolltieren verglichen.
Außerdem gehen wir der Frage nach, ob Metallproteinasen (MMP-2, MMP-9) eine maßgebliche Rolle
in der Pathogenese von Vitamin D-induzierten Verkalkungen spielen.
Studie zur kardiovaskulären Komorbidität bei
Kindern mit chronischen Nieren-erkrankungen
(The Cardiovascular Comorbidity in Children
with Chronic Kidney Disease, 4 C Study)
Die 4C-Studie ist eine prospektive Beobachtungsstudie an Kindern und Jugendlichen mit chronischer Niereninsuffizienz, in der die kardiovaskuläre
Komorbidität mit nicht-invasiven vaskulären Untersuchungsmethoden charakterisiert wird. In einer
Sub-Studie wird bei Patienten, die das Terminalstadium der Niereninsuffizienz erreichen, zum Zeitpunkt
der Anlage einer AV-Fistel für die Hämodialyse oder
einer präemptiven Nierentransplantation eine Arterienbiopsie durchgeführt. Im CCR werden Biopsien
mit färberischen Methoden, der Histochemie, der
Elektronenmikroskopie, der Mineralzusammensetzung von Verkalkungen sowie durch Vergleich der
Genexpression untersucht. Die Daten werden hinsichtlich einer Assoziation mit klinischen Variabeln,
d.h. der Progression der Niereninsuffizienz und der
kardiovaskulären Komorbilität ausgewertet.
Claudine: Ionerenabsorption, Volumenregula­
tion und Blutdruckkontrolle
Claudine sind membranständige Tight Junction Proteine mit 4 transmembranen Domänen, die wichtig
für epitheliale und endotheliale Zell-Zell Kontakte
sind. Sie begünstigen oder verhindern parazelluläre
Transportvorgänge indem sie das Epithel abdichten
143
Uwe Querfeld – PEDIATRIC NEPHROLOGY
oder durchgängig machen. Dabei weisen die einzelnen Mitglieder der Claudin-Familie eine Substrat-,
Ladungs- und Größenspezifität für Substrate auf.
In der Niere sind die Claudine an der Rückresorption glomerulär filtrierter Substanzen beteiligt, ihre
Expression variiert mit dem reginalen Ionenresorptionsprofil und der Entwicklung. Durch diese Fähigkeiten tragen Claudine maßgeblich zur Volumenregulation und damit zur Blutdruckkontrolle und der
144
gesamten Ionenhomöostase bei. Unsere Arbeitsgruppe beschäftigt sich mit der Funktion der renal
exprimierten Claudine -3, -10 und-16, die im aufsteigenden Ast der Henleschen Schleife exprimiert sind
– einem Bereich des Tubulussystems, der besonders eng mit der Blutdruckregulation korreliert ist.
Um Funktion und Fehlfunktion besser zu verstehen,
entwickeln wir für diese Claudine murine Knock-out
Modelle, bei denen das jeweilige Gen inaktiviert ist.
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Research projects
1. V
itamin D-induced vascular calcification in uremic rats
-Effect of cinacalcet ( CINA) and parathyroidectomy ( PTX)
Project leader
Coworkers
Cooperations
Uwe Querfeld
Dominik Müller, Susanne Jung, Kerstin Sommer
AG Peters, CCR: Harm Peters, Stephanie Krämer
Pediatric patients with CKD need treatment with
active vitamin-D preparations to prevent the development of rickets and to suppress hyperparathyroidism.
The therapeutic window, however, is small, and clinical and experimental data show that both calcitriol
and PTH are involved in the pathogenesis of vascular
calcifications. We have studied the question to what
extent calcitriol-induced vascular calcifications can be
prevented by parathyroidectomy (PTX) and cinacalcet
(CINA) in a rat model of uremia (5/6 nephrectomy).
Five groups of animals were studied:
shamoperated (CON), uremic (U), uremic + calcitriol
(U+vitD),
uremic+calcitriol+PTX
(U+vitD+PTX),
uremic+calcitriol+CINA (U+vitD+CINA).
We found that vascular calcifications (%area of aortic
wall) were not seen in CON and U animals, present
(>20%) in U+vitD animals, and reduced to a similar degree (<10%) in both, the U+vitD+PTX and the
U+vitD+CINA group. PTH was significantly lower in
U+D rats compared to U rats, and further suppressed
to a similar degree in animals treated with PTX or CINA.
The serum calcium-phosphate product was elevated
in all groups receiving calcitriol, without a significant
difference between groups. The creatinine clearance
was significantly decreased only in U+vitD animals
compared to uremic controls (U). Systolic blood pressure was elevated in all experimental groups com-
pared to controls (CON). Growth and weight gain
was not suppressed in the U+vitD+CINA group. We
conclude that in uremic rats, CINA attenuates vitamin D- induced calcification.Neither CINA nor PTX
can completely prevent vitamin-D induced calcification. Calcification occurs by direct vascular effects of
calcitriol, independent of serum calcium and serum
phosphorus concentrations.
We could show that calcitriol treatment induces endochondral differentiation in vascular smooth muscle
cells (Figure below).
145
Uwe Querfeld – PEDIATRIC NEPHROLOGY
The experimental model used in these studies (highdose vitamin D-induced vascular calcifications in
uremic animals) is potentially suitable for identifying
molecular mechanisms for vascular calcification,
which may provide therapeutic targets for intervention
studies. Thus, by further modifications of the experi-
146
mental design, it may be possible to identify the therapeutic window for active vitamin d preparations, which
is essential for patients with chronic kidney disease,
especially children and adolescents. Furthermore,
current reasearch is aimed at characterizing the role
of matrix metalloproteinases (MMP-2 and MMP-9) on
high-dose calcitriol induced vascular calcifications in
uremic animals.
In addition, we are studying the effect of high-dose
calcitriol, Cinacalcet and PTX on uremic bone disease
in cooperation with Laboratory of Pathophysiology,
University of Antwerp, Belgium.
Uwe Querfeld – PEDIATRIC NEPHROLOGY
2. T
he Cardiovascular Comorbidity in Children with Chronic
Kidney Disease Study (4 C Study )
Project leader
Coworkers
Uwe Querfeld
Franz Schaefer (Heidelberg)
Dominik Müller, Kerstin Sommer, The 4 C Study group
The pediatric population appears uniquely suited to
study the effects of CKD on the cardiovascular system
due to the virtual absence of vascular morbidity related
to ageing, diabetes and smoking.
The 4 C Study is a 3+ year prospective observational
study in up to 625 children and adolescents with CKD.
Enrollment has started in 2010 and thus far includes
> 500 children aged 6 to 17 years with a glomerular
filtration rate of 10 to 45 ml/min/1.73 m² in 51 pediatric
nephrology units in 14 European countries. Patients
are studied by annual non-invasive assessment of
arterial and cardiac morphology and function, Monitoring
of putative anthropometric and
biochemical predictor variables,
and by whole-genome study of
common genetic variants for
association with progression of
cardiovascular comorbidity and
renal failure progression.
“Functional“ endpoints are
the increase of local (Carotid,
M-mode) and systemic arterial
stiffness (pulse wave velocity)
and myocardial dysfunction.
„Morphology“ endpoints are the
increase of carotid IMT and left
ventricular mass index .
In the CCR, a comprehensive study of arterial
biopsy specimens, usually taken at the time of AV
fistula surgery or pre-emptive renal transplantation,
is performed for histopathological assessment in
a biopsy sub-study. Biopsy samples will be analyzed by conventional staining, histochemistry and
by electron microscopy, physicochemical characterisation of mineral precipitates, and comparative
gene expression profiling. Data will be analyzed for
associations with the clinical progression of renal
and cardiovascular disease obtained from the 4 C
Study database.
147
Uwe Querfeld – PEDIATRIC NEPHROLOGY
3. Claudin-3 tight junction protein in renal ion homeostasis
Project leader
Dominik Müller
Coworkers
Kathrin Kopplin,Marchel Stuiver, Kerstin Sommer, Uwe Querfeld
CollaborationsThomas Willnow (MDC Berlin), Markus Bleich (University Kiel),
Dorothee Günzel, Michael Fromm (Charite CBF)
Funding
DFG (FOR 721)
Eunefron (EU, FP 7)
Claudin-3 is ubiquitously expressed in various epithelia. The functional and physiological role of claudin-3
is still unclear, however, its distinct expression profile
and widespread distribution in the body makes it likely
to be important for the integrity of epithelia. Here we
investigate the function of claudin-3 with a special
focus on its role in the kidney. It is known that claudin-3 is expressed in the tighter segments of the neph-
ron, where the resorption processes of ions like Na+,
Mg2+, Ca2+ occur. We are interested in the impact
of claudin-3 for ion and water homeostasis and thus
blood pressure regulation. A claudin KO strain has
been generated successfully and is currently being
investigated. Additionally a double KO (cldn16 / 3 KO)
has been obtained.
Immunohistochemistry of Claudin-3 in mouse kidney
demonstrating its expression along the nephron
148
Uwe Querfeld – PEDIATRIC NEPHROLOGY
4. Claudin-10 tight junction protein in renal ion homeostasis
Project leader
Coworkers
Collaborations
Funding:
Dominik Müller
Constanze Will, Marchel Stuiver, Kerstin Sommer, Uwe Querfeld
Thomas Willnow (MDC Berlin), Dorothee Günzel, Michael Fromm (Charite CBF)
DFG (FOR 721)
Eunefron (EU, FP 7)
The Cldn10 gene consists of two alternative first exons,
which give rise to the expression of two Claudin-10
isoforms. Of these isoforms, Claudin-10a shows exclusive expression in the kidney and has previously been
reported to influence anion permeability in epithelia.
Claudin 10b, however, is ubiquitously expressed in
various epithelia and has been functionally characterized as pore former. The expression of both isoforms
along the nephron makes Claudin-10 an attractive
candidate for distinct renal ion resorption processes
and thereby the regulation of sodium in body fluids,
water homeostasis and blood pressure.
The goal of our study is to evaluate the contribution
of Claudin-10 to epithelial transport processes and
the effect of Claudin-10 deficiency on the organisms´
physiology. To obtain a suitable in vivo model, we have
successfully generated a murine conditional knockout
strain which offers the possibility to analyze total or
tissue specific deficiency as well as gene deletion at
certain timepoints of pre- or postnatal development.
Our first data from Cldn10 total KO animals suggests
that Claudin-10 deficiency leads to vital impairment
within the first days after birth. Subsequent expression studies, investigation of organ morphology and
body fluid composition analysis will provide us with
information about the physiological defects in this
mouse model. Furthermore, conditional gene deletions will offer the possibility to establish vital mouse
models which will allow investigate special salt and
water stress conditions to further characterize the role
of Claudin-10 in vivo. Due to the widespread tissue
distribution of Claudin-10, this mouse model approach
is an excellent basis for interdisciplinary analysis with
experts on the fields of cardiology, pulmonology and
nephrology.
149
Uwe Querfeld – PEDIATRIC NEPHROLOGY
5. Claudin-16 tight junction protein in renal ion homeostasis
Project leader
Coworkers
Collaborations
Funding:
Dominik Müller
Constanze Will, Marchel Stuiver, Kerstin Sommer, Uwe Querfeld
Thomas Willnow (MDC Berlin), Markus Bleich (Universitiy of Kiel)
Michael Fromm (Charite CBF)
Eunefron (EU, FP 7)
DFG (FOR 721)
Claudin-16 is exclusively expressed in the kidney in the
thick ascending limb of Henle´s loop, where it serves as
a cation channel during the resorption of mono- and
bivalent ions. Mutations in the human CLDN16 gene
have been reported to come out with the salt loss syndrome FHHNC (Familial hypomagnesemia with hypercalciuria and nephrocalcinosis), which cause a severe
loss of Ca and Mg and an imbalance of hormone levels
regulating the homeostasis of bivalent ions. To further analyse ion handling in vivo and to obtain a suitable mouse model for FHHNC, we have targeted the
Cldn16 gene to set up a conditional murine Claudin-16
deficiency strain, which allows to study total loss of
Claudin-16 as well as temporo-spatial deficiency.
Metabolic analysis of Claudin-16 null mice reveals
hypomagnesemia and hypercalciuria in these animals,
as well as elevated levels of parathyroid hormone and
Calcitriol to overcome salt loss. These data confirm
the human phenotype and point out the impact of
Claudin-mediated paracellular transport mechanisms
on ion homeostasis and the composition of body fluids. Subsequent investigations will focus on morphologic and physiologic analysis of the kidneys, as well
as counteracting processes, e.g. renal transcellular
ion transport mechanisms. Furthermore, we plan to
150
perform pharmacological and salt stress models on
Cldn16 KO mice, where we hope to have insights into
compensative/regulative mechanisms in metabolism
which might be applicable for patients with FHHNC.
We already identified differences of acid-base handling in these animals. Another goal of our studies is to
study Cldn16 deficiency at different timepoints of preand postnatal development, to further understand the
role of Claudin-16 in organogenesis and ontogenesis.
For this purpose a Tamoxifen inducible cldn16 cre line
has been successfully established.
Cldn-16 KO mice exhibit severe renal Ca2+- and Mg2+ waste
and serve as a robust model for the human disorder FHHNC.
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Publications 2006 - 2010
Milatz S, Krug SM, Rosenthal R, Günzel D, Müller D, Schulzke
JD, Amasheh SS, Fromm M (2010): Claudin-3 acts as a sealing component of the tight junction for ions of either charge
and uncharged solutes.Biochem Biophys Acta – Biomem­
branes: July 22 [Epub ahead of print]
Querfeld U, Anarat A, Bayazit AK, Bakkaloglu AS, Bilginer
Y, Caliskan S, Civilibal M, Doyon A, Duzova A, Kracht D,
Litwin M, Melk A, Mir S, Sözeri B, Shroff R, Zeller R, Wühl E,
Schaefer F; the 4C Study Group (2010): The Cardiovascular
Comorbidity in Children with Chronic Kidney Disease (4C)
Study: Objectives, Design, and Methodology. Clin J Am Soc
Nephrol Jun 24. [Epub ahead of print]
Querfeld U, Mak RH. (2010): Vitamin D deficiency and toxicity
in chronic kidney disease: in search of the therapeutic window. Pediatr Nephrol Jun 22. [Epub ahead of print]
Höcker B, Weber LT, Feneberg R, Drube J, John U, Fehrenbach H, Pohl M, Zimmering M, Fründ S, Klaus G, Wühl E,
Tönshoff B. (2010): Improved growth and cardiovascular risk
after late steroid withdrawal: 2-year results of a prospective,
randomised trial in paediatric renal transplantation. Nephrol
Dial Transplant 25(2):617-24. Epub 2009 Sep 30
Will C, Breiderhoff T, Thumfart J, Stuiver M, Kopplin K, Sommer K, Günzel D, Querfeld U, Meij IC, Shan Q, Bleich M, Willnow TE, Müller D (2010): Targeted deletion of murine Cldn16
identifies extra- and intrarenal compensatory mechanisms of
Ca2+ and Mg2+ wasting. Am J Physiol Renal Physiol Feb
10. [Epub ahead of print]
Schaefer F, van de Walle J, Zurowska A, Gimpel C, van Hoeck
K, Drozdz D, Montini G, Bagdasorova IV, Sorof J, Sugg J,
Teng R, Hainer JW (2010): Candesartan in Children with
Hypertension Investigators. Efficacy, safety and pharmacokinetics of candesartan cilexetil in hypertensive children from 1
to less than 6 years of age. J Hypertens 28(5):1083-90.
Franke D, Völker S, Haase S, Pavicic L, Querfeld U, Ehrich JH,
Zivicnjak M (2010) Prematurity, small for gestational age and
perinatal parameters in children with congenital, hereditary
and acquired chronic kidney disease. May 31. [Epub ahead
of print]
Kempf C, Winkelmann B, Roigas J, Querfeld U, Müller D
(2010): Severe complications after endoscopic injection of
polydimethylsiloxane for the treatment of vesicoureteral reflux
in early childhood. Scand J Urol Nephrol Jun 14. [Epub
ahead of print]
Zipfel PF, Mache C, Müller D, Licht C, Wigger M, Skerka C; for
the European DEAP-HUS Study Group (2010): DEAP-HUS:
Deficiency of CFHR plasma proteins and autoantibody-positive form of hemolytic uremic syndrome. Pediatr Nephrol
Feb 16. [Epub ahead of print]
Hoppe A, von Puttkamer C, Linke U, Kahler C, Booß M, Braunauer-Kolberg R, Hofmann K, Joachimsky P, Hirte I, Schley S,
Utsch B, Thumfart J, Briese S, Gellermann J, Zimmering M,
Querfeld U, Müller D. (2010): A Hospital-Based Intermit­
tent Nocturnal Hemodialysis Program for Children and
Adolescents. J Pediatr Aug 5. [Epub ahead of print]
Zebger-Gong H, Kampmann J, Kong L, Roigas J, Sommer K, Krämer S, Peters H, Müller D, Dragun D, Querfeld U.
Decreased Transplant Arteriosclerosis in Endothelial Nitric
Oxide Synthase-Deficient Mice. Transplantation 2009 Dec
16. [Epub ahead of print]
Günzel D, Stuiver M, Kausalya PJ, Haisch L, Hunziker W,
Krug S, Meij IC, Fromm M, Müller D (2009): Identification and
characterization of six novel Claudin-10 splice variants J Cell
Sci 15: 1507-15017
ESCAPE Trial Group, Wühl E, Trivelli A, Picca S, Litwin M,
Peco-Antic A, Zurowska A, Testa S, Jankauskiene A, Emre
S, Caldas-Afonso A, Anarat A, Niaudet P, Mir S, Bakkaloglu
A, Enke B, Montini G, Wingen AM, Sallay P, Jeck N, Berg
U, Caliskan S, Wygoda S, Hohbach-Hohenfellner K, Dusek
J, Urasinski T, Arbeiter K, Neuhaus T, Gellermann J, Drozdz
D, Fischbach M, Möller K, Wigger M, Peruzzi L, Mehls O,
Schaefer F. (2009): Strict blood-pressure control and progression of renal failure in children. N Engl J Med 361:1639-50
Gimpel C, Wühl E, Arbeiter K, Drozdz D, Trivelli A, Charbit M,
Gellermann J, Dusek J, Jankauskiene A, Emre S, Schaefer F;
ESCAPE Trial Group (2009): Superior consistency of ambulatory blood pressure monitoring in children: implications for
clinical trials. J Hypertens 27:1568-74
151
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Raile K, Klopocki E, Holder M, Wessel T, Galler A, Deiss D,
Müller D, Riebel T, Horn D, Maringa M, Weber J, Ullmann R,
Grüters A (2009): Expanded clinical spectrum in HNF1BMODY. J Clin Endocrinol Metab 94: 2658-2664
Offner G, Toenshoff B, Höcker B, Krauss M, Bulla M, Cochat
P, Fehrenbach H, Fischer W, Foulard M, Hoppe B, Hoyer PF,
Jungraithmayr TC, Klaus G, Latta K, Leichter H, Mihatsch MJ,
Misselwitz J, Montoya C, Müller-Wiefel DE, Neuhaus TJ, Pape
L, Querfeld U, Plank C, Schwarke D, Wygoda S, Zimmerhackl
LB (2008): Efficacy and safety of basiliximab in pediatric renal
transplant patients receiving cyclosporine, mycophenolate
mofetil, and steroids. Transplantation 86:1241-8
Thumfart J, Jung S, Amasheh S, Krämer S, Peters H, Sommer K, Biber J, Murer H, Meij I, Querfeld U, Wagner CA,
Müller D (2008): Magnesium stimulates renal phosphate
reabsorption. Am J Physiol Renal 295: F126-133
Raile K, Klopocki E, Wessel T, Deiss D, Horn D, Müller D,
Ullmann R, Grüters A (2008): HNF1B abnormality (MODY5) in
children and adolescents: high prevalence in autoantibodynegative type 1 diabetes with kidney defects. Diabetes Care
31: e83
Briese S, Claus M, Querfeld U (2008): Arterial stiffness in
children after renal transplantation. Pediatr Nephrol Jun 27.
[Epub ahead of print]
Müller D, Zimmering M, Chan CT, McFarlane PA, Pierratos
A, Querfeld U (2008):.Intensified hemodialysis regimens:
neglected treatment options for children and adolescents.
Pediatr Nephrol Mar 12. [Epub ahead of print]
Nissel R, Faraj S, Sommer K, Henning L, van der Giet M,
Querfeld U (2008): Oxidative stress markers in young hemodialysis patients – a pilot study. Clin Nephrol 70, 2:135-143
Robben J, Sze M, Eggert P, Knoers NV, Deen PM, Müller D
(2007): Relief of nocturnal enuresis by desmopressin is kidney
and Vasopressin type-2 receptor independent. J Am Soc
Nephrol 18:1534-1539
Verberckmoes S, Persy V, Behets GJ, Hufkens A, Müller D,
Haffner D, Querfeld U, Bohic S, De Broe ME, D’Haese
PC (2007): Uremia-related Vascular Calcification: More than
Apatite Deposition. Kidney Int 71: 298-303
152
König S, Ringe H, Dorner BG, Diers A, Uhlenberg B, Müller D,
Varnholt V, Gaedicke G (2007): Atypical tetanus in a completely
immunized 14-year old boy. Pediatrics 120: e1355-1358
Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Seelow
D, Nurnberg G, Garg P, Verma R, Chaib H, Hoskins BE,
Ashraf S, Becker C, Hennies HC, Goyal M, Wharram B, Schachter AD, Drummond I, Kerjaschki D, Waldherr R, Dietrich A,
Ozaltin F, Bakkaloglu A, Cleper R, Basel-Vanagaite L, Pohl M,
Griebel M, Tsygin AN, Soylu, Muller D, Katan M, Liu J, Attanasio M, O’Toole JF, Hasselbacher K, Mucha M, Otto EA, Airik
R, Kispert A, Kelley GG, Smrcka AV, Gudermann T, Holzman
LB, Nurnberg P, Hildebrandt F (2006): Positional cloning of
PLCE1 mutations as the first cause of a nephrotic syndrome
variant which may be reversible. Nat Genet 38: 1397-1404
Müller D, Kausalya PJ, Meij IC, Hunziker W (2006): Familial
hypomagnesemia with hypercalciuria and nephrocalcinosis:
blocking endocytosis restores surface expression of a novel
Claudin-16 mutant that lacks the entire C-terminal cytosolic
tail. Hum Mol Genet 15: 1049-1058
Kausalya JP, Amasheh S, Günzel D, Wurps H, Müller D,
Fromm M, Hunziker W (2006): Disease-associated mutations
affect intracellular traffic and paracellular Mg2+ transport
function of Claudin-16 J Clin Invest 116: 878-891
Müller D, Kausalya PJ, Bockenhauer D, Thumfart J, Meij IC,
Dillon M, van’t Hoff W, Hunziker W (2006): Unusual clinical
presentation and possible rescue of a novel Claudin-16 mutation. J Clin Endocrinol Metab 91: 3076-3079
Müller D, Klopocki E, Taupitz M, Mundlos S, Schulze I, Ropers
HH, Querfeld U, Ullmann R (2006): A complex phenotype with
cystic renal disease. Kidney Int 70: 1656-1660
Fliegauf M, Horvath J, von Schnackenburg C, Olbrich H,
Müller D, Thumfart J, Schermer B, Pazour GJ, Neumann
HPH, Zentgraf H, Benzing T, Omran H (2006): Nephrocystin
specifically localizes to the transition zone of renal and respiratory cilia and photoreceptor connecting cilia. J Am Soc
Nephrol 17: 2424-2433
Pieper AK, Haffner D, Hoppe B, Dittrich K, Offner G, Bonzel
KE, John U, Frund S, Klaus G, Stubinger A, Duker G,
Querfeld U (2006): A randomized crossover trial comparing
sevelamer with calcium acetate in children with CKD. Am J
Kidney Dis 47:625-35
Uwe Querfeld – PEDIATRIC NEPHROLOGY
Briese S, Wiesner S, Will JC, Lembcke A, Opgen-Rhein B, Nissel R, Wernecke KD, Andreae J, Haffner D, Querfeld U (2006):
Arterial and cardiac disease in young adults with childhoodonset end-stage renal disease-impact of calcium and vitamin D
therapy. Nephrol Dial Transplant 21:1906-14 (2006)
Günzel D, Haisch L, Pfaffenbach S, Krug S, Milatz S,
Amasheh S, Hunziker W, Müller D (2008): Claudin function in
the thick ascending limb of Henle’s loop. Ann NY Acad Sci
1165: 152-162
Reviews & book chapters (2006-20010)
Thumfart J, Müller D (2008): Hämolytisch-urämisches Syndrom im Kindesalter. Nephrologe 3: 297-307
Querfeld U: Dialyse im Kindesalter
In: F. Keller, M. Ketteler, R. Schindler, U. Heemann, G. Wolf
(Hrsgb): Manuale Nephrologicum, 2nd edition, Dustri-Verlag Dr. Karl Feistle, München-Orlando, 2009, p. 427-443
Müller D, Zimmering M, Chan CT, McFarlane PA, Pierratos
A, Querfeld U (2008): Intensified hemodialysis regimens:
Neglected treatment options for children and adolescents.
Pediatr Nephrol 23: 1729-1736
Querfeld U: Urämische Vaskulopathie im Kindesalter
In: Pourhassan S, Sandmann W (Hrsgb): Gefäßerkrankun­
gen im Kindes- und Jugendalter, p. 250-256, SpringerVerlag Berlin Heidelberg New York 2009
Müller D (2007): Das hämolytisch-urämische Syndrom im
Kindes- und Jugendalter. Päd 13: 403-406
Zipfel PF, Mache C, Müller D, Licht C, Wigger M, Skerka C
How Deficiency of CFHR1 and CFHR3 Plasma Proteins and
Autoantibodies to Factor H Contribute Hemolytic Uremic
Syndrome: THE DEAP HUS Subtype- Form Pediatr Nephrol
in press
Giessing D, Müller D, Winkelmann B, Roigas J, Loening SA
(2007): Kidney Transplantation in Children and Adolescents.
Transplant Proceedings 39: 2197-2201
Müller D (2007): Rapid progressive Glomerulonephritis:
Schnittstelle zwischen pädiatrischer Nephrologie und
pädiatrischer Rheumatologie. Aktuelle Rheumatologie 32:
149-153
Devuyst O, Meij I, Jeunemaitre X, Ronco P, Antignac C, Christensen EI, Knoers NV, Levtchenko EN, Deen PM, Müller D,
Wagner CA, Rampoldi L, Van’t Hoff WG (2009): EUNEFRON,
the European Network for the Study of Orphan Nephropathies Nephrol Dial Transplant 24: 2011-2015
Winkelmann B, Thumfart J, Müller D, Giessing M, Wille A,
Deger S, Schnorr D, Querfeld U, Loening S, Roigas J.(2006):
Nierentransplantation im Kindes- und Jugendalter. Urologe
A. 45: 18-24
Müller D, Müller DN (2009): Battle against the renin-agiotensin
system: Help from an unexpected party. Nephrol Dial
Transplant 24: 1110-1112
Müller D (2007): Nierenersatztherapie bei terminaler Niereninsuffizienz. In: S. Wirth (Hrsg.): Leitlinien Kinder- und
Jugendmedizin P11: 36-39, Urban und Fischer, München
Will C, Fromm M, Müller D (2008): Claudin Tight Junction
Proteins – Novel Aspects in paracellular transport. Peritoneal
Dial Int 28: 577-584
Querfeld U (2006): Adipocyte signaling: at the crossroads
of metabolism, inflammation, and vascular function. Pediatr
Transplant Mar; 10(2) 136-9
Cardiovascular Disease in Pediatric Chronic Kidney Disease
Querfeld U, Mitsnefes M (2008): In: Geary D, Schaefer FS
(eds): Clinical Pediatric Nephrology, Mosby-Elsevier, Phila­
delphia p. 793-810
153
Uwe Querfeld – PEDIATRIC NEPHROLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Querfeld U
Querfeld U
Müller D
Müller D
Müller D
Müller D
Project title
Taurolidine-citrate vs heparin as catheter
lock solutions in paediatric patients
The Cardiovascular Comorbidity in
Children with Chronic Kidney Disease
(„4C“) Study
Spatio-temporal aspects in renal paracellular transport
Evaluation of in-center nocturnal hemodialysis
Genetically modified mouse modelst o
study paracellular ion transport
Rolle in der Ionenhomöostase und Blutdruckregulation
Sponsor
Tauropharm
Period
2006-2010
Kuratorium für Heimdialyse (KfH)
Stiftung Präventivmedizin
2009-2014
EU: The European Network of Orphan 2008-2012
Nephropathies (EUNEFRON, FP 7)
Else Kröner-Fresenius Stitung
2006-2009
EU: European Renal Genome Project
(EUREGENE, FP 6)
DFG: Molekulare Struktur und Funktion der Tight Junction (FOR 721)
2005-2009
2006-2010
Awards ( 2006-2009 )
2010
Else Kröner Fresenius-Preis
Zentrumsbasierte intermittierende nächtliche Hämodialyse für
Kinder und Jugendliche Gesellschaft für
Pädiatrische Nephrologie Hamburg Anne Hoppe
2009
Arbeitsgemeinschaft Pflegekräfte in der Dialyse, 3. Preis
Etablierung eine nächtlichen Hämodialyseprograms für
Kinder und Jugendliche. Fulda
Christina von Puttkamer
2009
Shortterm Fellowship
European Network of Orphan Nephropathies
(EUNEFRON) Brüssel
Kathrin Kopplin
2009
Else Kröner Fresenius-Preis
Gesellschaft für Pädiatrische Nephrologie Functional characterization
of six alternative CLDN10 splice variants found in renal epithelia;
40. Jahrestagung der Gesellschaft für Pädiatrische Nephrologie
Amsterdam Lea Haisch
154
Uwe Querfeld – PEDIATRIC NEPHROLOGY
2009
Reisestipendium der Gesellschaft für
Pädiatrische Nephrologie
An in-center based intermittent nocturnal hemodialysis
program for children and adolescents, 40. Jahrestagung
der Gesellschaft für Pädiatrische Nephrologie, Amsterdam 2009
Reisestipendium der Gesellschaft für
Pädiatrische Nephrologie
Generation and characterization of a CLDN-16 KO model
40. Jahrestagung der Gesellschaft für Pädiatrische Nephrologie
Amsterdam
Constanze Will
2008
Short term Fellowship
European Renal Genome Project (EUREGENE)
Paris
Constanze Will
2007
Forschungsstipendium der Deutschen Nierenstiftung
Ca2+- und Mg2+ Regulation am transgenen Tiermodell
Jahrestagung der deutschen Gesellschaft für
Nephrologie, München Julia Thumfart
2007
Else Kröner Fresenius-Preis
Arbeitsgemeinschaft für pädiatrische Nephrologie
Magnesium stimuliert die renale Phosphatexkretion
38. Jahrestagung der Arbeitsgemeinschaft für
Pädiatrische Nephrologie Stuttgart
Julia Thumfart
2006
Poster Preis
Mouse models to study renal paracellular solute transport
2nd Summerschool of the European Renal Genome
Project Oxford
Constanze Will
Anne Hoppe
155
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Head of the group
Prof. Dr. med. Vera Regitz-Zagrosek
Curriculum Vitae: She received her medical degree in 1979 from Saarland University and completed her fellowship in clinical cardiology at Dept. of Biochemistry,
University of Madison, Wisconsin. She also completed her residency at the German
Heart Institute Munich, where she served as Chief Medical Resident.
1985 she became senior fellow at the German Heart Institute Berlin with responsibility
for a large cardiovascular outpatient department. In 1995 she became professor in
Internal Medicine and in 2002 she became the first and only German professor of Cardiovascular Disease in
Women at the Charite Berlin in cooperation with the German Heart Institute Berlin. In 2003, she founded the
Berlin Institute for Gender in Medicine (GiM) and is President of the International Society for Gender in Medicine
(IGM). The aim of the GiM is the systematic analysis of sex and gender differences in medicine and its introduction into medical education. The research interests of Vera Regitz-Zagrosek include gender differences in
cardiovascular disease, estrogen receptors and molecular and genetic mechanisms in heart failure. She leads
and participates in major projects funded by European Union, Federal Ministry of Education and Research
(BMBF), German Research Foundation (DFG) and in industry sponsored projects.
She is the founder, project leader and speaker of the Graduate Course GK 754 “Sex-specific mechanisms of
myocardial hypertrophy” (2001-2010) and project leader and speaker of the Research Group FOR 1054 on the
same subject. She is project leader of EUGene Heart Task Gender and the pilot project “Gender Medicine” in
cooperation with German Heart Institute Berlin. Since 2009 she is task force leader of the ESC’s Committee
to develop the new version of the Guidelines for the Management of Cardiovascular Diseases during Pregnancy (New Version 2011). She is reviewer for the German Research Foundation (DFG), the European Union
and several journals. Her scientific publication record consists of over 150 scientific publications, more than
200 abstracts, and some book chapters. Her work has been published in numerous peer-reviewed journals
including Nature Reviews.
157
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Members of the group
Office
Schmidt, Stefanie
Scientists
Becher, Eva
Brožová, Eva
Dworatzek, Elke
Fliegner, Daniela
Kararigas, Georgios
Kendel, Friederike
Lehmkuhl, Elke
Mahmoodzadeh, Shokoufeh
Sanchez Ruderisch, Hugo
Schubert, Carola
Technicians
Angelov, Anja
Brincker, Sven
Kühne, Arne
Fielitz, Britta
Niehage, Sylke
Riese, Vanessa
Thomas, Jenny
158
Dr. rer. nat.
Dr. rer. nat.
Dr. rer. medic.
Dr. rer. nat.
Dr. rer. nat.
Psychologist
Dr. med.
Dr. rer. nat.
Dr. rer. nat.
Dr. rer. nat.
Students
Brunhuber, Claudia
Brzank, Maria
Eschen, Claudia
Fischer, Susanne
Fritschka, Stephan
Hampl, Hannah
Leber, Joachim
Penkalla, Adam
Petrov, Georgi
Pham, Thi Hang
Queiros, Ana
Schanz, Miriam
Urbschat, Annika
Westphal, Christina
Zhang, Xiang
Medical student
Medical student
PhD student
Medical student
PhD student
Medical student
PhD student
Medical student
Medical student
PhD student
PhD student
Medical student
Medical student
PhD student
Guest scientist,
University of Bejing
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Summary
The group of Prof. Regitz-Zagrosek investigates the
gender specific aspects of cardiovascular disease,
focussing on regulation of myocardial hypertrophy,
heart failure and vascular mechanisms. Our projects
stretch from basic research to clinical research and
clinical trials.
- Research Group Myocardial Hypertrophy
(DFG, FOR 1054)
The main aim of the research group “Myocardial
Hypertrophy” (FOR 1054) is the analysis of gender
differences in the adaptation of the heart to mechanical stress. In this research group, we perform a
series of mechanical analysis using cell cultures and
animal models. This includes models of physiological and pathological myocardial stress, e.g. strength
training and hypertension. The role of estrogen and
androgen receptors is selectively analyzed. Moreover, the effect of sex hormones on the lipid turnover
is investigated. Several collaborations with clinical
projects exist; in this area we mainly focus on gender
differences in myocardial reaction to overload due to
aortic valve disease.
- Graduate Group 754
Graduate group GK 754 „Gender-specific mechanisms of myocardial hypertrophy“ (DFG) originates
from the collaboration of 12 principal investigators
within Charité, 8 at the Center for Cardiovascular
Research (CCR), and the German Heart Institute in
Berlin. It comprises four interconnected modules: 1)
Animal models of hypertrophy, 2) functional genomics and proteomics, 3) gender-specific molecular
mechanisms and 4) gender aspects in the clinics of
cardiovascular diseases. In this project, the research
group of Prof. Regitz-Zagrosek is analyzing the sexspecific regulation of the energy metabolism and
its influence on heart function. Furthermore, matrix
synthesis and its control by estrogen is analyzed.
Another goal is the identification of transcriptional
factors and their potential binding sites within the
promoter region of matrixmetalloproteinases (MMP2), as well as the characterization of other matrixrelated genes.
- EuGeneHeart European Excellence Project
Heart Research
The team of Prof. Regitz-Zagrosek participates in
the EU-funded EUGeneHeart project. The project
aims at the elucidation of the mechanisms leading
from myocardial hypertrophy to heart failure. Task
number 4 “gender” is coordinated by Prof. RegitzZagrosek and includes collaboration with research
groups at the INSERM (Institut national de la santé et
de la recherche médicale) and the Karolinska Institute. Aim is the investigation of gender differences in
myocardial hypertrophy induced by stress, training,
infarction and pressure. Experiments are performed
using cell cultures and animal models and focus on
the role of sex hormones.
- Gender differences in human aortic stenosis
In 2006 a single project has been designed to link
basic and clinical research in analysing gender differences in human aortic stenosis. The project was
funded by the DFG, German Research Foundation
(DFG-Re 662/6-1). Gender-specific differences
after aortic valve transplantation are investigated in
patients and in experimental animal models.
- CARDIOVASC
Several projects at the CCR/GiM are conducted by
investigators funded through the Marie Curie Early
Stage Research Training Program CARDIOVASC.
Cardiovascular diseases display significant sex and
159
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
gender differences. Sexual hormones, especially
estrogens, are believed to play a major role in these
differences. To define the effects of estrogen on transcription regulation in the human heart, systematic
comparison between microarray gene expression
in estrogen-stimulated and non-stimulated cardiac
tissue was performed. This project should identify
and elucidate molecular mechanisms influencing the
incidence and progression of cardiac disease.
- Clinical Research
In the area of clinical research we conduct studies
with primarily cardiologic focus and maintain intense
collaboration with the German Heart Institute in Berlin.
We also collaborate with the German Competence
Network “Heart Failure” and were able to introduce
our sex/gender-specific point of view, especially
through project TP13. We also work at the definition
of the young discipline of gender medicine and its current state-of-the-art through the Pilot project “Gender
Medicine” funded by The BMBF. Within this project
we test scientific publications for gender relevance
and systematically classify the results.
Zusammenfassung
Die Gruppe Prof. Regitz-Zagrosek untersucht
Geschlechterunterschiede in der Entstehung von
Herz-Kreislauferkrankungen mit besonderer Beachtung von Herzhypertrophie, Herzinsuffizienz und
vaskulären Mechanismen. Unsere Projekte reichen
von der Grundlagenforschung bis hin zur klinischen
Forschung sowie klinischen Studien.
- Forschergruppe Myokardhypertrophie (FOR
1054)
Das vordringliche Ziel der Forschergruppe „Myokardhypertrophie“ (FOR 1054) ist es, die Anpassung
des Herzens an mechanische Belastung in beiden
Geschlechtern zu untersuchen. Dazu werden eine
Reihe mechanistischer Untersuchungen an Zellkultur- und Tiermodellen durchgeführt. Dies beinhaltet Modelle für physiologische und pathologische
Belastungen des Myokards, zum Beispiel Ausdauertraining und Bluthochdruck. Die Rolle von Östrogen- und Androgenrezeptoren sowie Sexualhormonen wird gezielt analysiert. Darüber hinaus untersuchen wir die Effekte von Sexualhormonen auf den
160
Lipidstoffwechsel. In Kooperation mit verschiedenen
klinischen Projekten untersuchen wir insbesondere
die Reaktion des männlichen und weiblichen Myokards auf Druckbelastungen bei Erkrankungen der
Aortenklappe.
- Graduiertenkolleg 754
Im GK 754 „Geschlechtsspezifische Mechanismen
bei Myokard-Hypertrophie“ (DFG) kooperieren
Arbeitsgruppen der Charité, davon 8 des Center
for Cardiovascular Research (CCR), und des Deutschen Herzzentrums Berlin. Es besteht aus vier
eng miteinander verbundenen Modulen: 1) Tiermodelle bei Hypertrophie, 2) funktionelle Genomik und
Proteomik, 3) geschlechtsspezifische molekulare
Mechanismen und 4) klinische Geschlechtsaspekte
bei Herz-Kreislauferkrankungen. In diesem Projekt
analysiert die Arbeitsgruppe von Prof. Regitz-Zagrosek die geschlechtsspezifische Regulierung des
Energiemetabolismus und seinen Einfluss auf die
Herzfunktion. Darüber hinaus werden die Matrixsynthese und ihre Kontrolle durch Östrogen analy-
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
siert. Ein weiteres Ziel ist es, Transkriptionsfaktoren
zu identifizieren und ihre mutmaßlichen Bindungsstellen innerhalb der Promoterregion von MatrixMetallproteinasen (MMP-2) und anderen matrixbezogenen Genen zu charakterisieren.
- EUGeneHeart - Europäisches Exzellenzprojekt Herzforschung
Die Arbeitsgruppe Regitz-Zagrosek nimmt an dem
durch die EU geförderten Projekt EUGeneHeart teil,
das sich mit dem Übergang der Herzhypertrophie zur
Herzinsuffizienz befasst. Die Koordination des Tasks
Nr. 4 “Geschlecht“ hat Prof. Regitz-Zagrosek inne.
Es beinhaltet die Kooperation mit Forschergruppen
des INSERM (Institut national de la santé et de la
recherche médicale) und des Karolinska Instituts bei
der Untersuchung von Geschlechterunterschieden
bei Myokardhypertrophie, die durch Stress, Training, Infarkt oder Druckbelastung ausgelöst wird.
Dies geschieht in Zellkultur- und Tiermodellen und
fokussiert auf die Wirkung von Sexualhormonen.
- Geschlechterunterschiede bei menschlicher
Aortenstenose
Im Jahre 2006 wurde in der Schnittstelle zwischen
Grundlagen- und klinischer Forschung ein DFGEinzelprojekt (DFG-Re 662/6-1) konzipiert und eingeworben, in dem es um Geschlechterunterschiede
bei menschlicher Aortenstenose geht. Hier werden
sowohl geschlechtsspezifische Verläufe nach Aortenklappenersatz am Patienten untersucht als auch
experimentelle Untersuchungen am Tiermodell
durchgeführt.
- CARDIOVASC
Im Rahmen der Marie Curie Early Stage Research
Training Programme CARDIOVASC sind mehrere
Projekte im CCR/GiM angesiedelt. Herzkreislauferkrankungen weisen deutliche Geschlechterunterschiede auf. Es wird angenommen, dass Sexualhormone, speziell Östrogen hierbei eine wesentliche
Rolle spielen. Um die Funktion von Östrogen für die
Regulation der Transkription im humanen Herzen zu
untersuchen, wird in diesem Projekt ein systematischer Vergleich der Genexpression von mit Östrogen stimuliertem mit unstimuliertem Herzgewebe
ex vivo mittels DNA Microarrays vorgenommen.
Dieses Projekt soll molekulare Mechanismen, die
zu geschlechtsspezifischen Unterschieden in Entstehung und Verlauf von Herzerkrankungen führen,
identifizieren und aufklären.
- Klinische Forschung
Im Rahmen der klinischen Forschung führen wir Studien
mit primär kardiologischem Fokus durch und pflegen
dabei sehr enge Kollaborationen mit dem Deutschen
Herzzentrum Berlin. Wir kooperieren weiterhin mit dem
Kompetenznetz „Herzinsuffizienz“ und bringen dabei
unseren geschlechtsspezifischen Gesichtspunkt ein,
besonders durch das Teilprojekt 13.
Weiterhin befassen wir uns in einem vom BMBF
geförderten Projekt, mit der Definition des jungen
Fachbereiches Gendermedizin und dessen aktuellen
Standes durch das Pilotprojekt „Gender Medicine“,
bei dem Fachliteratur auf ihre Geschlechtsspezifik hin
überprüft und systematisch analysiert wird.
161
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Research projects
1. 1
7β-oestradiol ( E2 ) effects and mechanisms of sex-specific gene
regulation in the heart
Project leader
Coworkers
Funding
External cooperations
Georgios Kararigas
Eva Becher, Daniela Fliegner, Shokoufeh Mahmoodzadeh, Karina Nawrath,
Vanessa Riese
EC: MEST-CT-2005-020268 CARDIOVASC (2006-2009)
EC: EUGeneHeart EC-018833 (WP18; currently)
Boehringer Ingelheim Fonds (2007)
Dr. Ba Tiep Nguyen & Prof. Hubertus Jarry, Goettingen University
Dr. Virginie Bito & Prof. Karin Sipido, Leuven University
Prof. Roland Hetzer, German Heart Institute Berlin
Cardiovascular disease (CVD) manifests differently in
men and women. Sex steroid hormones are generally believed to play a major role in the occurrence
of sex-related differences observed in the development of CVD. In particular, a large body of evidence
from several observational and experimental studies
indicates that E2 might be involved in reducing risk
for cardiovascular disease. However, harmful effects
of E2 have also been reported leading to the current
controversy regarding the actions of E2. In addition,
so far there has been no report of E2-dependent gene
regulation in a sex-specific manner in the heart, while
such effects have been described in other organs.
The main goal here is to investigate the effect(s) of
E2 in the heart. To achieve this, we are comparing
the transcriptomes of human cardiac tissues and of
hearts from mice treated with E2. We combine these
analyses with data collected from in vivo studies and
assays on cellular effects generated in vitro.
Sex-specific PGR (progesterone receptor) regulation by E2 in
human cardiac tissues. (HBC: control) From Kararigas et al.
Biology of Sex Differences 2010 Accepted
162
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
2. R
ole of sex and sex hormones on extracellular matrix remodelling in pressure overload induced myocardial hypertrophy
Project leader
Coworkers
Funding
Elke Dworatzek
George Petrov, Carola Schubert, Shokoufeh Mahmoodzadeh, Elke Lehmkuhl,
Irina Wenzel, Anja Angelov
DFG: GK II/III 754 TP4
DFG: Re662/6-1
Patients with pressure overload induced left ventricular
(LV) hypertrophy show sex-differences in cardiac function and extracellular matrix (ECM) remodeling, with
more pronounced fibrosis in male hearts. Goal of the
study was to identify clinical parameters that determine
LV hypertrophy, and to correlate underlying mechanisms of ECM remodeling with clinical endpoints under
gender specific aspects. In order to investigate the role
of 17β-Estradiol (E2) in ECM remodeling, we performed
in-vitro studies in isolated adult rat cardiac fibroblasts.
We found that women with aortic (AS) adapt differently
to pressure overload compared to men. Male patients
had a significant higher gene expression of ECMassociated genes, e.g. collagen I and III compared to
females. We could show a sex-specific regulation of
collagen I and III expression in isolated cardiac fibroblast. Furthermore, we showed for the first time the
inhibitory effect of E2 on MMP-2 expression in cardiac
fibroblast via the activation of estrogen receptors (ER)
and MAP kinase-ERK1/2 signalling (Figure 1). Based
on these data, we speculate that E2 regulates ECM
turnover and may be, at least partially, responsible for
sex-differences in cardiac remodelling. Further work
will analyze the sex-differences in ECM remodeling in
LV hypertrophy more detailed, and identify underlying
molecular mechanisms by which E2 regulates ECM
components in a sex-specific manner. This could contribute to a better understanding of sex-differences in
cardiac remodeling and help to design sex-specific
pharmacological interventions.
Proposed model of the E2 regulation on human MMP-2 promoter activity by ER alpha and MAP kinase-ERK1/2 signaling
pathway (Mahmoodzadeh S. & Dworatzek E. et al., Cardiovascular Research 2010).
163
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
3. E
strogen receptor modulation of myocardial energy metabolism
contributes to sex differences in myocardial hypertrophy
Project leader
Coworkers
Funding
Vera Regitz-Zagrosek, Shokoufeh Mahmoodzadeh
Elke Dworatzek, Eva Brozova, Georgios Kararigas, Karina Nawrath, Carola Schubert,
Sylke Niehage
DFG: FOR 1054 / TP1
Females develop a more physiological myocardial
hypertrophy (MH) than males. Male hearts have a
higher incidence of cardiac dysfunction and heart
failure in response to stress. Exercise leads to more
physiological MH in female than in male mice. Female
sex or estrogens may lead to an improved cardiac
response to stress. Based on our previous findings, we
hypothesize that estrogen (E2) and its receptors ERa
and ERb improve the cardiac response to mechanical
load by regulating mitochondrial function and energy
metabolism. AKT signalling and key regulators of mito-
chondrial function and metabolic genes are probably
involved in these processes. Therefore in this study,
we investigate:
1) sex differences in cardiac function in physiological
(exercise) MH model. 2) Effect of E2 and pharmacological/mechanical load on cellular growth and metabolic
genes. 3) Effect of E2-activated ERa and ERb on gene
expression and biological activity of PGC1a and MEF2
transcription factors that control mitochondrial activity.
The project will identify sex-specific MH mechanisms
with therapeutic potential relevant to both genders.
A: Experimental setup for voluntary cage running, B: Female mice show higher exercise performance and C: a significant
increase in left ventricular mass / Tibila length.
164
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
4. Analysis of the role of estrogen receptor a in the heart in a
transgenic mouse model
Project leader
Coworkers
Funding
External cooperations
Shokoufeh Mahmoodzadeh
Joachim Leber, Britta Fielitz, Arne Kühne,
DFG: GK III 754 TP4
EC: EUGeneHeart EC-018833 (WP17)
Dr. F. Jaisser, INSERM, France
Estrogen exerts its beneficial effects on the myocardium during stress via estrogen receptors (ER) a and
b. During and after myocardial infarct (MI), administration of ERa-selective agonist has protective effects in
the heart. ERa-KO animals have a poorer outcome
and abnormal mitochondrial morphology after MI.
We hypothesize that ERa contributes to protection
of ischemic myocardium and this differs in male and
female. we therefore generate and evaluate the phe-
notype of a transgenic mouse model with an inducible,
myocardial ERa-overexpression (ERa-OE). Furthermore, we perform functional and biochemical analysis
of the transgenic mice in unstressed condition and
after induction of MI, in a sex-specific context. We
expect major deviations from WT at base line conditions and after induction of MI, possibly leading to the
discovery of major cellular pathways influenced by ERa
and thereby to new therapeutic targets.
A
Transactivator mouse
MHC
tTA
Responder mouse
tetO
MHC
tTA
tetO
ERα
ERα
Conditional mouse model
(generated by Frederic Jaisser, INSERM, Paris)
A: Mouse model with inducible heart specific
expression of the ERα gene using the Tet-Off
system (collaboration with Dr. F. Jaisser,
INSERM, Paris). B: Westernblot analysis
of heart tissue from double transgenic
(ERα-OE) and wild type mice using antibody
against ERα. C: Representative micrographs
of a left ventricular cross section of Masson
trichrome staining at 14 days post-MI.
C
B
myocardial infarction
Western-Blot ERa
M
WT
ERα-overexpresser
ER α,
66 kD
165
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
5. Sex differences and the influence of estrogen receptor b
Project leader
Daniela Fliegner, Carola Schubert
Coworkers Adam Penkalla
Funding
DFG: GK 754 II/III TP2
EC:
EuGeneHeart EC-018833 (WP19)
Charité: Post-doctoral fellowship
External Collaborations Prof. Dr. Ulrich Kintscher (CCR), Prof. Jan-Ake Gustafsson,
Karolinska Institute Stockholm
Myocardial hyper- tribute to the maintenance of energy homeostasis and
trophy (MH) rep- attenuate the development of fibrosis and apoptosis,
resents the car- thus slowing the progression to heart failure.
diac response to
different stimuli, Current ongoing studies are focused now on the
such as pressure effects of sex and sexual hormones on the cardiac
Development of myocardial
overload, hyper- mitochondrial function in physiological and pathologihypertrophy.
tension, diet or cal conditions.
aging and can result in heart failure (HF). The
development of MH is associated with alterations in cardiac geometry (size and shape),
which is characteristically referred to as ventricular remodeling and appears to be different
between the sexes. Sex differences in the cardiovascular system have largely been attributed to the effects of sex steroid hormones
such as estrogen, which are mainly mediated
by their nuclear receptors: ERα, ERβ. The goal
of this study was to investigate the molecular
mechanisms underlying these sex differences.
We used a mouse model of pressure overload
induced myocardial hypertrophy in both sexes
with or without deletion of ERb. In this project
our findings indicate that female sex offers protection against ventricular chamber dilation in Genes involved in oxidative phosphorylation were regulated in a sex-specific
the TAC model. Both female sex and ERb con- manner. (Fliegner et al. Am J Physiol Regul Integr Comp Physiol (June 2010).
166
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
6. I nfluence of different estrogen receptor modulators on
pressure overload-induced myocardial hypertrophy
Project leader
Coworkers
Funding
External Collaborations
Carola Schubert
Christina Westphal
DFG: GK 754 II/III TP2
EC:
EuGeneHeart EC-018833 (WP19)
Bayer Schering AG
Dr. Katja Prelle (Bayer Schering AG)
Estrogens influence many physiological processes
in mammals, including cardiovascular health. It is
also implicated in the development or progression
of cardiovascular diseases. In many of the diseases,
17b-estradiol (E2) mediates its effects through its estrogen receptors a and b (ERa and ERb), which serves
as the basis for many therapeutic interventions. Selective estrogen receptor modulators (SERMs) such as
tamoxifen and raloxifen are examples of compounds
that exhibit tissue-specific estrogenic activity. They
may be useful for the reduction of cardiovascular risk.
In the current project we wanted to identify the estrogen receptor, which is responsible for the protection of
the heart against pressure-overload. Ovariectomiced
(OVX) female mice were pressure loaded by transverse aortic constriction (TAC) and treated with E2
and specific estrogen receptor agonists (ERaA and
ERbA) or raloxifen to prevent cardiac hypertrophy and
heart failure.
Time flow of the animal experiment (OVX/ovariectomy; PI/pellet implantation with treatment substances; TAC/transverse aortic
constriction; Echo/echocardiographic measurements; OW/organ withdrawal)
167
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
7. S
ex specific differences in microRNA expression and their role
in heart diseases
Project leader
Coworkers
Funding
Hugo Sanchez-Ruderisch
Claudia Eschen, Ana Queiros, Daniela Fliegner
DFG: GK 754 II/III
MicroRNAs (miRs) are small non-coding RNAs that partially rescued by estrogen administration. Based
control gene expression by duplex formation with on these data and on our previous results showing
3´ untranslated regions of target mRNAs. Dysregu- gender differences after TAC we hypothesise that an
lation of miRs has been reported in various cardiac estradiol-dependent gender different expression of
diseases. Most of the miRNAs expressed in mouse miRs is in part responsible for the observed gender
cardiac tissue are regulated during cardiomyocytes different development of cardiac hypertrophy after
hypertrophy. However, nothing is known about the pressure overload. The aims of this project are the
regulation of expression of miRs considering the gen- identification of those miRNAs expressed in a genderder aspect. Gender differences in the development specific manner in the mice heart and elucidate their
and progression of cardiovascular diseases such as role in heart hypertrophy comparing male and female
miR-1
inhibits
the expression
of miR1-targeted
myocardial infarction, hypertension, heart failure and Endogenous
mice after TAC
treatment.
Preliminary
results indicated
HL-1
cells
sudden death have been described. In mouse model sex different expression of various
miRs.
The influence
of cardiomyopathies, a more severe cardiovascular of altered miRs expression on putative target genes
phenotype is observed in male animals and can be involved in fibrosis is under current investigation.
RLU (Renilla/Luc)
2,5
2,0
1,5
1,0
0,5
0,0
psiCHECK
168
psiCheckmiR1
psiCheckmiR1 +
mimic1
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
8. M
elusin protects against cardiac rupture and maladaptive
remodeling after myocardial infarction sex dependently
Project leader
Coworkers
Funding
External cooperations
Vera Regitz-Zagrosek, Carola Schubert
Susette Bernert, Jenny Thomas
EC: EUGeneHeart EC-018833 (WP3.1)
MD Bernhard Unsöld (Göttingen), Prof. Karin Sipido (Leuven),
Prof. Guido Tarone (Turin)
The muscle specific ß1 integrin interacting protein
Melusin has been found to be upregulated during
cardiac hypertrophy and protects against the transition into heart failure after pressure overload. Attenuation of cardiac hypertrophy was accompanied by
lower tissue deposition and apoptosis of cardiomyocytes. A careful analysis of the influence of melusin on
remodeling processes after myocardial infarction (MI)
and sex differences therein is needed. We wanted to
understand the protective effects of melusin against
cardiac remodeling and early death after MI and sex
differences therein. For that reason an animal model
for MI was performed in male and female WT mice
and in mice overexpressing the melusin gene (TG).
Animals were killed 3 or 14 days after induction of MI.
The experiments showed that melusin protects male
animals against early cardiac rupture by decreasing
MMP9 activity. In later maladaptive processes following MI the TG affects mainly females and protects them
against heart failure, while in males it does not.
Survival after MI in wildtype (WT) and melusin transgene (TG) animals
169
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Publications 2006 - 2010
Mahmoodzadeh S, Eder S, Nordmeyer J, Ehler E, Huber O,
Martus P, Weiske J, Pregla R, Hetzer R, Regitz-Zagrosek V
(2006): Estrogen receptor alpha up-regulation and redistribution in human heart failure. Faseb J 20(7) 926-34
Philipp S, Jürgensen JS, Fielitz J, Bernhardt WM, Weidemann A,
Schiche A, Pilz B, Dietz R, Regitz-Zagrosek V, Eckardt KU, Willenbrock R (2006): Stabilization of hypoxia inducible factor rather than
modulation of collagen metabolism improves cardiac function after
acute myocardial infarction in rats. Eur J Heart Fail 8(4) 347-54
Regitz-Zagrosek V, Hocher B, Bettmann M, Brede M, Hadamek K, Gerstner C, Lehmkuhl HB, Hetzer R, Hein L (2006):
{alpha}2C-Adrenoceptor polymorphism is associated with
improved event-free survival in patients with dilated cardiomyopathy. Eur Heart J 27(4) 454-459
Schupp M, Kintscher U, Fielitz J, Thomas J, Pregla R, Hetzer R,
Unger T, Regitz-Zagrosek V (2006): Cardiac PPARalpha expression
in patients with dilated cardiomyopathy. Eur J Heart Fail 8(3) 290-4
Brokat S, Thomas J, Herda LR, Knosalla C, Pregla R, Brancaccio M, Accornero F, Tarone G, Hetzer R, Regitz-Zagrosek
V (2007): Altered melusin expression in the hearts of aortic
stenosis patients. Eur J Heart Fail 9(6-7) 568-73
Diedrich M, Tadic J, Mao L, Wacker MA, Nebrich G, Hetzer R,
Regitz-Zagrosek V, Klose J (2007): Heart protein expression
related to age and sex in mice and humans. Int J Mol Med
20(6) 865-74
Fielitz J, Philipp S, Herda LR, Schuch E, Pilz B, Schubert C,
Günzler V, Willenbrock R, Regitz-Zagrosek V. (2007): Inhibition
of prolyl 4-hydroxylase prevents left ventricular remodelling in
rats with thoracic aortic banding. Eur J Heart Fail 9(4):336-42
Fliegner D, Westermann D, Riad A, Schubert C, Becher E, Fielitz
J, Tschöpe C, Regitz-Zagrosek V (2008): Up-regulation of PPARgamma in myocardial infarction. Eur J Heart Fail 10(1) 30-8
Isensee J, Witt H, Pregla R, Hetzer R, Regitz-Zagrosek V, Noppinger PR (2008): Sexually dimorphic gene expression in the
heart of mice and men. J Mol Med 86(1) 61-74
Karatas A, Hegner B, de Windt LJ, Luft FC, Schubert C, Gross
V, Akashi YJ, Gürgen D, Kintscher U, da Costa Goncalves AC,
Regitz-Zagrosek V, Dragun D (2008): Deoxycorticosterone
acetate-salt mice exhibit blood pressure-independent sexual
dimorphism. Hypertension 51(4) 1177-83
170
Kendel F, Dunkel A, Lehmkuhl E, Hetzer R, Regitz-Zagrosek V
(2008): Does time spent on household activities or housework
stress complicate recovery following coronary artery bypass
surgery? Women Health 48(3) 325-38
Witt H, Schubert C, Jaekel J, Fliegner D, Penkalla A, Tiemann K,
Stypmann J, Roepcke S, Brokat S, Mahmoodzadeh S, Brozova
E, Davidson MM, Ruiz Noppinger P, Grohé C, Regitz-Zagrosek V
(2008): Sex-specific pathways in early cardiac response to pressure overload in mice. J Mol Med 86(9) 1013-24
Boengler K, Buschmann I, Deindl E, Gottwik M, Hoffmeister
HM, Ito W, Klein H, Mauser M, Nienaber C, Regitz-Zagrosek V,
Sack S (2009): On the occasion of Wolfgang Schaper’s 75th
birthday. Basic Res Cardiol 104(1) 2-4
Dunkel A, Kendel F, Lehmkuhl E, Babitsch B, Oertelt-Prigione
S, Hetzer R, Regitz-Zagrosek V (2009) Predictors of preoperative depressive risk in patients undergoing coronary artery
bypass graft surgery. Clin Res Cardiol 98 643-650
Friedrichs F, Zugck C, Rauch GJ, Ivandic B, Weichenhan D,
Müller-Bardorff M, Meder B, El Mokhtari NE, Regitz-Zagrosek
V, Hetzer R, Schäfer A, Schreiber S, Chen J, Neuhaus I, Ji R,
Siemers NO, Frey N, Rottbauer W, Katus HA, Stoll M (2009):
HBEGF, SRA1, and IK: Three cosegregating genes as determinants of cardiomyopathy. Genome Res 19(3) 395-403
Hassel D, Dahme T, Erdmann J, Meder B, Huge A, Stoll M,
Just S, Hess A, Ehlermann P, Weichenhan D, Grimmler M,
Liptau H, Hetzer R, Regitz-Zagrosek V, Fischer C, Nürnberg P,
Schunkert H, Katus HA, Rottbauer W (2009): Nexilin mutations
destabilize cardiac Z-disks and lead to dilated cardiomyopathy.
Nat Med 15(11) 1281-8
Mahmoodzadeh S, Fritschka S, Dworatzek E, Pham TH, Becher
E, Kuehne A, Davidson MM, Regitz-Zagrosek V (2009): Nuclear
factor-Kappa B regulates estrogen receptor-alpha transcription
in the human heart. J Biol Chem 284(37) 24705-24714
Fliegner D, Schubert C, Penkalla A, Witt H, Kararigas G,
Dworatzek E, Staub E, Martus P, Ruiz Noppinger P, Kintscher
U, Gustafsson JA, Regitz-Zagrosek V (2010): Female sex and
estrogen receptor-beta attenuate cardiac remodeling and
apoptosis in pressure overload. Am J Physiol-Reg I 298(6)
R1597-606
Kendel F, Wirtz M, Dunkel A, Lehmkuhl E, Hetzer R, RegitzZagrosek V (2010): Screening for depression: Rasch analysis
of the dimensional structure of the PHQ‑9 and the HADS-D. J
Affect Disorders 122 241-246
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
Mahmoodzadeh S, Dworatzek E, Fritschka S, Pham TH,
Regitz-Zagrosek V (2010): 17{beta}-estradiol inhibits matrix
metalloproteinase-2 transcription via MAP kinase in fibroblasts.
Cardiovasc Res 85(4) 719-728
Regitz-Zagrosek V, Petrov G, Lehmkuhl E, Smits JM, Babitsch
B, Brunhuber C, Jurmann B, Stein J, Schubert C, Merz NB,
Lehmkuhl HB, Hetzer R (2010): Heart transplantation in women
with dilated cardiomyopathy. Transplantation 89(2) 236-44
Ruiz-Holst C, Bölck B, Ghanem A, Tiemann K, Brokat S,
Regitz-Zagrosek V, Bloch W, Schwinger RHG, Brixius K (2010):
eNOS phosphorylation and translocation are altered in male
but not female mice by increased activation of the Galphaq
protein. Can J Physiol Pharm 88(2) 121-9
Toischer K, Rokita AG, Unsöld B, Zhu W, Kararigas G, Sossalla
S, Reuter SP, Becker A, Teucher N, Seidler T, Grebe C, Preuß
L, Gupta SN, Schmidt K, Lehnart SE, Krüger M, Linke WA,
Backs J, Regitz-Zagrosek V, Schäfer K, Field LJ, Maier LS,
Hasenfuss G (2010): Differential Cardiac Remodeling in Preload
Versus Afterload. Circulation 122 993-1003
Petrov G, Regitz-Zagrosek V, Lehmkuhl E, Krabatsch T, Dunkel
A, Dandel M, Dworatzek E, Mahmoodzadeh S, Schubert C,
Becher E, Hampl H, Hetzer R (2010): Regression of Myocardial Hypertrophy After Aortic Valve Replacement. Circulation
122(suppl 1) 23-28
Reviews & book chapters (2006-20010)
Regitz-Zagrosek V (2006): Therapeutic implications of the
gender-specific aspects of cardiovascular disease. Nat Rev
Drug Discov 5(5) 425-38
Regitz-Zagrosek V, Lehmkuhl E, Weickert MO (2006): Gender
differences in the metabolic syndrome and their role for cardiovascular disease. Clin Res Cardiol 95(3) 136-47
Regitz-Zagrosek V, Brokat S, Tschöpe C (2007)
: Role of
gender in heart failure with normal left ventricular ejection fraction. Prog Cardiovasc Dis 49(4) 241-51
Regitz-Zagrosek V, Wintermantel TM, Schubert C (2007):
Estrogens and SERMs in coronary heart disease. Curr Opin
Pharmacol 7(2) 130-9
Gohlke-Bärwolf C, Pildner von Steinburg S, Kaemmerer H,
Regitz-Zagrosek V (2008): Anticoagulation and thrombophilia in
pregnancy. Internist 49(7) 779-87
Regitz-Zagrosek V, Gohlke-Bärwolf C, Geibel-Zehender A,
Haass M, Kaemmerer H, Kruck I, Nienaber C (2008): Heart
diseases in pregnancy Clin Res Cardiol 97(9) 630-65
Regitz-Zagrosek V, Lehmkuhl E, Lehmkuhl HB (2008): Herzinsuffizienz: geschlechts-spezifische Aspekte? Internist 49(4)
422-8
Regitz-Zagrosek V, Schubert C, Krüger S (2008): Gender differences in psychopharmacology Internist 49(12) 1516-23
Regitz-Zagrosek V, Schubert C, Krüger S (2008): Sex differences in cardiovascular drug targeting Internist 49(11) 13836, 1388-90
Mehrhof F, Löffler M, Gelbrich G, Özcelik C, Posch M, Hense
HW, Keil U, Scheffold T, Schunkert H, Angermann C, Ertl
G, Jahns R, Pieske B, Wachter R, Edelmann F, Wollert KC,
Maisch B, Pankuweit S, Erbel R, Neumann T, Herzog W, Katus
H, Müller-Tasch T, Zugck C, Düngen HD, Regitz-Zagrosek V,
Lehmkuhl E, Störk S, Siebert U, Wasem J, Neumann A, Göhler
A, Anker S, Köhler F, Möckel M, Osterziel KJ, Dietz R, Rauchhaus M, on B.o.t.C.N.H.F (2009): A network against failing
hearts-Introducing the German “Competence Network Heart
Failure” Int J Cardiol
Oertelt-Prigione S, Regitz-Zagrosek V (2009): Women’s cardiovascular health: prevention is key. Arch Intern Med 169(19)
1740-1
Regitz-Zagrosek V, Oertelt-Prigione S, Seeland U, Hetzer R
(2010): Sex and gender differences in myocardial hypertrophy
and heart failure. Circ J 74(7) 1265-73
Sliwa K, Hilfiker-Kleiner D, Petrie MC, Mebazaa A, Pieske B,
Buchmann E, Regitz-Zagrosek V, Schaufelberger M, Tavazzi
L, van Veldhuisen DJ, Watkins H, Shah AJ, Seferovic PM,
Elkayam U, Pankuweit S, Papp Z, Mouquet F, McMurray JJV
(2010): Current state of knowledge on aetiology, diagnosis,
management, and therapy of peripartum cardiomyopathy: a
position statement from the Heart Failure Association of the
European Society of Cardiology Working Group on peripartum
cardiomyopathy. Eur J Heart Fail 12(8) 767-78
171
VERA REGITZ-ZAGROSEK – GENDER in CARDIOVASCULAR MEDICINE
General information
Third party funding ( 2006-2010 )
Project leader
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Mahmoodzadeh S.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Mahmoodzadeh S.
Schubert C.
Regitz-Zagrosek V.
Schubert C.
Mahmoodzadeh S.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Mahmoodzadeh S.
Regitz-Zagrosek V.
Mahmoodzadeh S.
Regitz-Zagrosek V.
Schubert C.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Regitz-Zagrosek V.
Becher E.
Project title
Kompetenznetzwerk Herzinsuffizienz (KNHI TP4)
Research Training Programm GK 754/2: Geschlechtsspezifische Mechanismen bei Myokardhypertrophie
Genstudie
IP EUGeneHeart, Task Gender
Research Training Programm GK 754/3: Geschlechtsspezifische Mechanismen bei Myokardhypertrophie
Klinische Studie von herzoperierten Frauen
Kompetenznetzwerk Herzinsuffizienz (KNHI
TP13)
Satellitensymposium
GiM Symposium
Einfluss des Geschlechts bei myokardialer
Druckbelastung
Research Group: Sex-specific mechanisms in
myocardial hypertrophy
Crataegus
Research Training Programm GK 754/4: Geschlechtsspezifische Mechanismen bei Myokardhypertrophie
Pilotprojekt „Gender Medicine“ (PPGM)
EUGIM
Sponsor
BMBF
German Research Foundation
Period
2003-2008
2004-2006
Institut für Herz-Kreislaufforschung an
der Universität Witten/Herdecke
European Commission
EC: 018833
2005-2008
German Research Foundation
2006-2010
Margarete-Ammon-Stiftung
BMBF
2007-2010
2006-2008
Bundesministerium für Familie,
Senioren & Frauen
German Research Foundation DFG Re
662/6-1
German Research Foundation 1054
TP1 + Z2
Industry: Dr. Willmar Schwabe
GmbH & Co. KG, Karlsruhe
German Research Foundation
BMBF
FP7
2006-2010
2007
2008
2007-2010
2008-2010
2009-2010
2010-2011
2008-2011
2009-2011
Awards
2008
Margherita-von-Brentano-Preis der FU Berlin
172
Joachim Dudenhausen,
Marianne Kriszio, Mechthild Koreuber,
Vera Regitz-Zagrosek, Martina Dören,
Martin Paul
MICHAEL SCHUPP – ENDOCRINOLOGY
Head of the group
Michael Schupp, PhD.
Emmy-Noether group leader
Dept. Endocrinology, Diabetes and Nutrition
Curriculum Vitae: Michael Schupp studied pharmacy in Regensburg, Cardiff, London, Madrid and Munich. He earned his PhD in 2005 under the supervision of Prof.
U. Kintscher and Prof. T. Unger at the Institute of Pharmacology, Charité University
Medicine Berlin. Parts of his doctoral thesis were carried out in Prof. B. Staels group
at the Pasteur Institute in Lille. He then spent 5 years as a postdoctoral fellow at the University of Pennsylvania,
Philadelphia in the laboratory of Prof. M. Lazar in the Division of Endocrinology, Diabetes and Metabolism. He
returned August 2010 to Germany to install an Emmy-Noether group at the Charité, Division of Endocrinology,
Diabetes and Nutritional Medicine, located in the CCR.
Members of the group
Witte, Nicole
Münzner, Matthias
Pharmacist, PhD student
Technichal Assistant
173
MICHAEL SCHUPP – ENDOCRINOLOGY
Summary
After my return from a 5-year postdoctoral fellowship at
the University of Pennsylvania (Prof. Mitchell A. Lazar
laboratory) in August 2010, I am establishing my own
research lab here at the CCR. As part of the Dept.
of Endocrinology (Prof. Andreas Pfeiffer and Prof.
Jochen Spranger), our lab is aimed at understanding
the function of the enzyme Retinol Saturase (RetSat) in
the development of obesity and type 2 diabetes. The
enzyme catalyzes the reduction of retinol to 13,14-dihydroretinol and possibly functions in other yet unknown
reactions. RetSat is expressed at high levels in metabolically active tissues like liver and fat. Liver expression
of RetSat is strongly regulated by fasting and feeding,
suggesting that it is involved in hepatic glucose and
fatty acid metabolism. Furthermore, RetSat expression
is induced during adipocyte differentiation. Strategies
that modify the activity of this enzyme could be used
for the treatment of metabolic diseases.
We also study the mechanisms by which certain transcription factors determine the fate of mesenchymal
stem cells. These cells can give rise to a variety of lineages such as adipocytes, osteoblasts, chondrocytes or
myogenic precursors. We are particularly interested in
the interaction of these transcriptional regulators with
cofactors, which mediate their activity in the modification
of histones. We are trying to understand how these transcriptional networks commit cells to a defined lineage by
the induction of the determining factors PPARg, Runx2,
MyoD, Sox proteins, and others. Reprogramming of
these pathways may allow us to control disease related
maladaptation in mesenchymal differentiation programs.
Zusammenfassung
Nach der Rückkehr von einem 5-jährigen Aufenthalt an
der University of Pennsylvania (Prof. Mitchell A. Lazar
laboratory) im Sommer 2010, etabliere ich meine EmmyNoether Nachwuchsgruppe hier am CCR. Zugehörig
zur Abteilung für Endokrinologie, Diabetes und Ernährungsmedizin (Prof. Andreas Pfeiffer und Prof. Jochen
Spranger), untersucht unser Labor die Rolle des Enzymes Retinol Saturase (RetSat) in der Entwicklung von
Typ-2 Diabetes und der Adipositas. RetSat reduziert in
einer NADH-abhängigen Reaktion Retinol zu 13,14-Dihydroretinol. Außerdem scheint das Enzym eine weitere,
bisher unbekannte enzymatische Reaktion zu katalysieren. RetSat wird in metabolisch aktiven Organen, wie
z.B. der Leber oder im Fettgewebe, stark exprimiert. Die
hepatische Expression der RetSat wird zusätzlich durch
Nahrungsaufnahme reguliert. Wir erforschen, ob eine
Modulation der enzymatischen Aktivität der RetSat eine
174
therapeutische Anwendung bei metabolischen Krankheiten haben könnte.
Weiterhin untersuchen wir die Mechanismen, durch
die bestimme Transkriptionsfaktoren die Differenzierung mesenchymaler Stammzellen beeinflussen. Diese
pluripotenten Zellen können zu verschiedensten Zelltypen, wie Chondrozyten, Adipozyten, Osteoblasten
oder Myoblasten, differenzieren. Wir sind besonders
an den Wechselwirkungen dieser Transkriptionsfaktoren mit Kofaktoren interessiert, die deren Aktivität in die
Modifizierung von Histonen vermitteln. Wir versuchen
zu verstehen, wie diese transkriptionellen Netzwerke zur
Festlegung auf ein spezielles Differenzierungsprogramm
durch die Expression von PPARg, Runx2, MyoD oder Sox
Proteinen führen. Eine Manipulation dieser Signalwege
könnte uns ermöglichen, krankheitsabhängige Störungen in der Zelldifferenzierung zu kontrollieren.
MICHAEL SCHUPP – ENDOCRINOLOGY
Research project
Role of Retinol Saturase in Liver metabolism
Project leader
Michael Schupp
Coworkers
Nicole Witte, Matthias Münzner
Collaborators
Prof. Ulrich Kintscher, MD (CCR), Prof. Jochen Spranger, MD (Endocrinology)
Funding
- DFG SCHU 2546/1-1
External cooperations- Prof. Lorraine J. Gudas, PhD. and Prof. Stephen L. Gross. PhD.,
Department of Pharmacology, Weill Cornell Medical College New York,
United States (Retinoid chemistry, MassSpec)
- Dr. Andreas Birkenfeld MD, DIfE Potsdam, Germany
(euglycemic-hyperinsulinemic clamp studies)
We initially identified the oxidoreductase RetSat as
an upregulated gene during adipocyte differentiation.
Depletion of RetSat by siRNA in preadipocytes inhibited adipocyte conversion. 13,14-dihydroretinol, the
putative product of RetSat’s enzymatic activity, did
not rescue this defect in adipocyte differentiation, suggesting that RetSat catalyzes an additional enzymatic
reaction. Furthermore, we found that adipose RetSat
expression is under the control of the Peroxisome
Proliferator-activated Receptor g (PPARg), the molecular target of insulin-sensitizing glitazones, through an
intronic binding site in the RetSat gene.
In addition to adipose tissue, RetSat is strongly
expressed in liver. Hepatic RetSat expression is
regulated by PPARa, the key transcription factor in
the hepatic fasting response via binding to the same
intronic binding site. Accordingly, it was shown that the
fasting induced increase in hepatic RetSat expression
was blunted in PPARa (-/-) mice.
The molecular function of RetSat in liver is unknown.
Preliminary studies suggest that RetSat is involved in
the induction of gluconeogenic gene expression and
intermediary metabolism during fasting. We will use
molecular-, cell-, and animal experiments to understand the physiology of hepatic RetSat in the context
of metabolic diseases.
175
MICHAEL SCHUPP – ENDOCRINOLOGY
Publications 2006 - 2010
Steger DJ, Grant GR, Schupp M, Tomaru T, Lefterova MI,
Schug J, Manduchi E, Stoeckert CJ, Jr., Lazar MA (2010):
Propagation of adipogenic signals through an epigenomic
transition state. Genes Dev 24:1035-1044
Schupp M, Lefterova MI, Janke J, Leitner K, Cristancho AG,
Mullican SE, Qatanani M, Szwergold N, Steger DJ, Curtin
JC, Kim RJ, Suh M, Albert MR, Engeli S, Gudas LJ, Lazar
MA (2009a): Retinol saturase promotes adipogenesis and
is downregulated in obesity. Proc Natl Acad Sci U S A
106:1105-1110
Schupp M, Cristancho AG, Lefterova MI, Hanniman EA,
Briggs ER, Steger DJ, Qatanani M, Curtin JC, Schug J,
Ochsner SA, McKenna NJ, Lazar MA (2009b): Re-expression
of GATA2 Cooperates with Peroxisome Proliferator-activated
Receptor-{gamma} Depletion to Revert the Adipocyte Phenotype. J Biol Chem 284:9458-9464
Lefterova MI, Mullican SE, Tomaru T, Qatanani M, Schupp
M, Lazar MA (2009): Endoplasmic reticulum stress regulates
adipocyte resistin expression. Diabetes 58:1879-1886
Tomaru T, Steger DJ, Lefterova MI, Schupp M, Lazar MA
(2009): Adipocyte-specific Expression of Murine Resistin Is
Mediated by Synergism between Peroxisome Proliferatoractivated Receptor {gamma} and CCAAT/Enhancer-binding
Proteins. J Biol Chem 284:6116-6125
Clemenz M, Frost N, Schupp M, Caron S, Foryst-Ludwig A,
Bohm C, Hartge M, Gust R, Staels B, Unger T, Kintscher U
(2008): Liver-specific peroxisome proliferator-activated receptor alpha target gene regulation by the angiotensin type 1
receptor blocker telmisartan. Diabetes 57:1405-1413
Steger DJ, Lefterova MI, Ying L, Stonestrom AJ, Schupp M,
Zhuo D, Vakoc AL, Kim JE, Chen J, Lazar MA, Blobel GA,
Vakoc CR (2008): DOT1L/KMT4 recruitment and H3K79
methylation are ubiquitously coupled with gene transcription
in mammalian cells. Mol Cell Biol 28:2825-2839
Schupp M, Curtin JC, Kim RJ, Billin AN, Lazar MA (2007): A
widely used retinoic acid receptor antagonist induces peroxisome proliferator-activated receptor-gamma activity. Mol
Pharmacol 71:1251-1257
Kershaw EE, Schupp M, Guan HP, Gardner NP, Lazar MA,
Flier JS (2007): PPARgamma regulates adipose triglyceride
lipase in adipocytes in vitro and in vivo. Am J Physiol Endo­
crinol Metab 293:E1736-1745
Schambach F, Schupp M, Lazar MA, Reiner SL (2007):
Activation of retinoic acid receptor-alpha favours regulatory
T cell induction at the expense of IL-17-secreting T helper cell
differentiation. Eur J Immunol 37:2396-2399
Schupp M, Lee LD, Frost N, Umbreen S, Schmidt B, Unger
T, Kintscher U (2006a): Regulation of peroxisome proliferatoractivated receptor gamma activity by losartan metabolites.
Hypertension 47:586-589
Moise AR, Dominguez M, Alvarez S, Alvarez R, Schupp M,
Cristancho AG, Kiser PD, de Lera AR, Lazar MA, Palczewski
K (2008): Stereospecificity of retinol saturase: absolute configuration, synthesis, and biological evaluation of dihydroretinoids. J Am Chem Soc 130:1154-1155
Schupp M, Kintscher U, Fielitz J, Thomas J, Pregla R, Hetzer
R, Unger T, Regitz-Zagrosek V (2006b): Cardiac PPARalpha
expression in patients with dilated cardiomyopathy. Eur J
Heart Fail 8:290-294
Lefterova MI, Zhang Y, Steger DJ, Schupp M, Schug J, Cristancho A, Feng D, Zhuo D, Stoeckert CJ, Jr., Liu XS, Lazar
MA (2008): PPAR{gamma} and C/EBP factors orchestrate
adipocyte biology via adjacent binding on a genome-wide
scale. Genes Dev 22:2941-2952
Janke J, Schupp M, Engeli S, Gorzelniak K, Boschmann M,
Sauma L, Nystrom FH, Jordan J, Luft FC, Sharma AM (2006):
Angiotensin type 1 receptor antagonists induce human invitro adipogenesis through peroxisome proliferator-activated
receptor-gamma activation. J Hypertens 24:1809-1816
Johnstone CN, Mongroo PS, Rich AS, Schupp M, Bowser
MJ, Delemos AS, Tobias JW, Liu Y, Hannigan GE, Rustgi AK
(2008): Parvin-beta inhibits breast cancer tumorigenicity and
promotes CDK9-mediated peroxisome proliferator-activated
receptor gamma 1 phosphorylation. Mol Cell Biol 28:687-704,
Research Highlights (2006-2010)
176
Schupp M, Lazar MA (2010): Fingered for a fat fate. Cell
Metab 11:244-245.
MICHAEL SCHUPP – ENDOCRINOLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Michael Schupp
Project title
Regulierung des hepatischen Glukoseund Fettstoffwechsels durch die Retinol
Saturase
Sponsor
DFG (German Research Society)
Emmy-Noether program
Period
2010-2013
Awards
2004
New Investigator Award, Council on High Blood Pressure Research, Chicago
2004
German Research Award Obesity and Hypertension (group award), Hannover
2005
Forssmann Award for young scientists, University of Bochum
2008 Scholarship Keystone Conference Diabetes
2009 Scholarship Keystone Conference Diabetes
2010 Emmy-Noether group leader DFG
177
Franz Theuring – Molecular Pharmacology
Head of the group
Prof. Dr. rer. nat. Franz Theuring
Curriculum Vitae: Professor Franz Theuring, PhD, studied electronics and biology
at the Universities of Braunschweig and Göttingen.
He gained his PhD in Mechanisms of meiotic nondisjunction in mammalian oocytes
in 1986 in the Institute of Human Genetics, University of Gottingen, and was postdoctoral research fellow at the Max Planck Institute for Biophysical Chemistry under
Professor Peter Gruss, world leader in mouse genetics.
Professor Theuring was responsible for the establishment and introduction of transgenic technologies both
in the Gruss laboratory, and then from 1988 in the Institute of Cellular and Molecular Biology of Schering AG,
Berlin, where he had responsibility for implementation of transgenic technologies in areas of pharmacological
research for drug target validation and model development at Schering. He developed transgenic mouse and
rat models in eurodegeneration, experimental tumour biology and cardiovascular function.
Professor Theuring received his Habilitation from the Free University of Berlin in Molecular Biology and Biochemistry in 1996, and after 8 years at Schering AG, was appointed in 1996 to a Professorship in Molecular
Pharmacology at the Charite Hospital, which is the Medical Faculty of the Humboldt University Berlin.
He is an acknowledged expert in employing transgenic technologies to study gene regulation, gene function
and establishment of animal models for human disease. He is consultant to the Berlin State in Biotechnology,
to the European Union in Molecular Medicine and to Pharmaceutical Industry,and was elected as Member of
the Berlin Scientific Society.
179
Franz Theuring – Molecular Pharmacology
Members of the group
Scientists
Dietze, Silke
Fischer, Andreas Marschall, Peter
Zabke, Claudia
Schwab, Karima
Dr.vet.med.
Dr. med.
Dr. rer. nat., until May 2010
Dr. rer.nat., until May 2009
Dipl. Ing.
Technicians
Magbagbeolu, Mandy Seelhorst, Bettina
Tanneberger, Cornelia Kemnitz, Rudi
04.2010 – 09.2010
PhD, Diploma and Master Students
Gluth, Markus PhD student
Gül, Nadir
PhD student
Neumann, Boris PhD student
Schwab, Karima PhD student
Vignon-Zellweger, Nicolas PhD student, until March 2010
Seider, Patrick Diploma student
Melis, Valeria PhD student, guest,
03.2008 – 04.2008 and
04.2009 – 05.2009,
University of Aberdeen
Ilnaz, Jallayier Bachelor Student,
01.2010 – 07.2010
Terzi Menderes, Yusuf Student Apprentice, Molecular
Medicine, 11.2007 – 01.2008
180
Animal Care Taker
Thoma, Anna
Trainee (Auszubildende)
Lange, Sebastian
Osterberg, Stephanie
11.2007 – 05.2008,
from Bayer Schering Pharma, Berlin
05.2008 – 10.2008,
from Bayer Schering Pharma, Berlin
Franz Theuring – Molecular Pharmacology
Summary
The group is using methods and state-of-the-art
techniques in biomedical research to identify new
potential drug targets, to validate drug targets and
already known key molecules for their involvement
in pathophysiological processes, and, in the frame
of a R&D program, to characterise and test new
pharmacological active compounds for their abilities
to act as therapeutic drugs in clinical dementia, i.e.
Alzheimer’s Disease. These studies represented the
final phase in preclinical drug testing and paved the
way for the future drugs to enter into clinical trials.
Therefore, transgenic techniques in mice are being
employed to study gene regulation and function
and to generate animal models relevant for human
diseases. Such animal models have given essential
insights into the regulation and function of genes in the
context of the whole organism. They further provide
a basis for transcriptomics and/or proteomics as well
as for the identification and characterization of genes
and proteins involved in human disease processes.
In the last CCR report we had reported on a major
breakthrough in the treatment of Alzheimer’s disease (AD) in the line of our successful phase II clinical trial. The product – rember®, a novel form of
methylthioninium chloride (MTC) – is the first drug
to act on the tau tangles discovered by Alois Alzheimer. The teams of the University of Aberdeen and
the Charite, working with Tau Rx Therapeutics – a
Singapore-based company spun out of these two
Universities – developed a novel treatment based
on an entirely new approach which targets the tangles, aggregates of abnormal fibres of Tau protein
forming inside nerve cells in the brain. By employing
our transgenic mice a group of second-generation
rember® derivatives had now been discovered and
successfully tested. TauRx has now initiated preparations for Phase 3 studies in mild and moderate
AD. A very similar approach is used to identify new
drugs acting on Parkinson’as Disease (PD). Transgenic mice modelling PD have been generated and
a detailed analysis of these model systems identified
the relevant transgenic lines which will be used to
test newly synthesized substances for their activity
to fight PD.
In parallel, we use different inbred strains of mice to
analyse for gender and age related effects in cardiovascular function. By employing modern 2-dimensional gel electrophoresis, mass spectrometry, and
phosphoproteomics we hope to identify key proteins
being responsible for and mediating these changes.
These candidates could then provide an entry point
into defining a more specific pharmacotherapy for
these alterations.
In addition to their importance as model systems for
human disorders, transgenic animals furthermore
represent valuable tools for the characterization of
general cell biological processes, providing the possibility to analyze the role of distinct molecules in an
in vivo context. Insights gained in these model systems will improve our understanding of fundamental
molecular mechanisms governing e.g. the regulation
of tight junction permeability, thereby facilitating the
identification of new pharmacological approaches
for the modulation of these processes. Transgenic
animals expressing various components of the RhoA
signalling pathway as well as in vitro systems mimicking the intestinal barrier furthermore provide an
insight into the effects of established and innovative
pharmacological compounds on tight junction permeability and intestinal barrier function.
181
Franz Theuring – Molecular Pharmacology
Zusammenfassung
Mittels modernster Methoden und Techniken in der biomedizinischen Forschung versuchen wir neue potenzielle
drug targets zu identifizieren und zu validieren. Zusätzlich
beziehen wir bereits bekannte, zentral agierende Moleküle in unsere Analysen mit ein, um Aussagen über ihre
Beteiligung an pathophysiologischen Prozessen machen
zu können. Letztendlich, im Rahmen eines Wirkstofffindungs- und Entwicklungsprogramms zur Entwicklung
eines therapeutisch wirksamen Alzheimer Medikamentes,
charakterisieren und testen wir pharmakologisch aktive
Substanzen im Rahmen von präklinischen Studien in
unseren entsprechenden tierexperimentellen Modellen.
Zur Realisierung dieser Vorhaben verwenden wir sowohl
in vivo (transgene und knockout Mäuse als Modellsysteme für menschliche Erkrankungen) als auch in vitro
(Zellinien epithelialen Ursprungs) Modellsysteme. In
Zusammenarbeit mit TauRx Therapeutics - einer Ausgründung der Universität Aberdeen und der Charite –
konnten wir im letzten CCR-Report von einem bahnbrechenden Erfolg unserer klinischen Studie der Phase
II in der Alzheimerforschung berichten. Die von TauRx
Therapeutics entwickelte neue Form des Wirkstoffes
Methylthioniniumchlorid (MTC)- Rember® - scheint die
erste Substanz zu sein, die in der Lage ist, ein Fortschreiten der Demenz aufzuhalten. Die Testung weitere Substanzen in unseren transgenen Mausmodellen führte nun
zur Entwicklung von verbesserten second generation
rember® Derivaten, deren Testung in einer Phase III Studie vorbereitet wird. Ein ähnlicher Forschungsansatz
bezieht sich auf die Entwicklung neuartiger Therapeutika zur Behandlung der Parkinsonschen Erkrankung.
Auch hier wurden zunächst relevante Tiermodelle etabliert und anschließend umfassend charakterisiert, um
in diesen dann unsere Substanzen auf ihre Wirksamkeit
in vivo testen zu können.
182
Weiterhin werden Endothelin 1 transgene und eNOS
knockout Mäuse als Modellsysteme für kardiovaskuläre Erkrankungen eingesetzt. Diese Tiermodelle
stellen die Ausgangsbasis zur detaillierten Analyse
zugrunde liegender molekularer Pathomechanismen
dar. Mittels Transkriptom- und Proteom-Analysen und
nach entsprechender Validierung werden anschließend Moleküle identifiziert, die eine zentrale Rolle im
Krankheitsgeschehen spielen. Im Rahmen eines Wirkstofffindungs- und Entwicklungsprogramms könnten
diese Moleküle als potenzielle drug targets fungieren.
Unter Verwendung verschiedener Maus-Inzuchtstämme erfolgt darüber hinaus die Analyse von
geschlechts- und altersspezifischen Veränderungen
im kardiovaskulären System. Hierzu werden modernste
Proteomtechniken wie 2-Dimensionale Gelelektrophorese, massenspektrometrische Analysen und Phosphoproteomics eingesetzt, um Schlüsselproteine, die
an der Vermittlung dieser Veränderungen beteiligt sind,
zu identifizieren und damit potenzielle Ansatzpunkte für
eine spezifischere Pharmakotherapie aufzuzeigen.
Neben ihrer Bedeutung als Modellsysteme für spezifische menschliche Erkrankungen stellen transgene
Tiere ebenfalls wertvolle Werkzeuge zur Charakterisierung zellbiologischer Prozesse dar und erlauben es
so, die Funktion spezifischer Moleküle in einem in vivo
Kontext zu studieren. Ziel ist es hier, durch ein verbessertes Verständnis der molekularen Mechanismen,
die zum Beispiel die tight junction Permeabilität regulieren, neue Ansatzpunkte zur pharmakologischen
Modulation dieser Prozesse zu identifizieren. Hierzu
existieren in unserer Arbeitsgruppe sowohl transgene
Tiermodelle zur Analyse verschiedener Komponenten
des RhoA-Signaltransduktionsweges als auch in vitro
Modellsysteme der gastrointestinalen Barriere, die
ebenfalls die Analyse der Effekte etablierter und innovativer Pharmaka auf die Barrierefunktion gestatten.
Franz Theuring – Molecular Pharmacology
Research projects
1. A
daptation of proteomic techniques for the identification and
characterization of protein species from murine heart
Project leader
Coworkers
Funding
Cooperation
Franz Theuring
Karima Schwab, Boris Neumann, Ilnaz Jallayier
Hypatia-Funding, Projektverbund Chancengleichheit für Frauen
Charite, PhD-grant
Dr. C. Scheler, Proteome Factory, Berlin; Dr. P.R. Jungblut,
MPI for Infection Biology, Berlin
Cardiovascular diseases (CVD) are the number one
cause of morbidity and mortality with an age and
sexual divergence. Premenopausal women are at a
lower risk for cardiovascular disease as compared to
age-matched men, but this risk increases dramatically after menopause, indicating that estrogens may
play a protective role. However hormone replacement therapy in humans yielded conflicting results
and phytoestrogens such as genistein, widely used
in traditional asian medicine, could represent alternative compounds as they are known to exert estrogenic
activity and to have beneficial effect on a wide range
of cardiovascular parameters.
In the cardiovascular context a disturbed energy
metabolism with impaired fatty acid oxidation, ATP
synthesis and changing levels of contractile proteins
have been observed during diseased conditions.
Whereas numerous studies focussed on gene expression analyses at the messenger RNA level, other holistic and undirected techniques, such as proteomics,
have been applied to the analysis of CVD. Multiple
identifications of a single protein from various spots
on 2-DE gels revealed that the suggestion of a single gene or transcript, encoding for a single protein,
is obsolete. The diversity in different forms of a pro-
tein emerging from one single protein-coding gene
promoted the new term of protein species. While
genomic and transcriptomic data lack information on
protein species in a given tissue, advances in proteome analysis and mass spectrometry enabled the
identification and characterization of post-translational
modifications (PTMs) in peptides derived from a protein species, which lead to an increasing number of
protein species in databases. Furthermore, advances
in mass spectrometry allowed the localization of cleavage sites for protein processing, maturation, truncation
and degradation. Protein species resulting from such
cleavage events play an important role in inflammation,
cell degeneration, apoptosis and oncogenesis. In the
cardiovascular context, protein species derived from
modifications such as acetylation, phosphorylation
and cleavage are involved in various processes and
disease development.
In this project we analyze the effects of a dietary supplement with the phytoestrogen genistein on the cardiac
proteome pattern of young, adult and castrated male
and female mice. Our analysis demonstrates considerable changes of the heart proteome with dietary genistein administration for both male and female animals.
A changing abundance, of among others metabolic,
183
Franz Theuring – Molecular Pharmacology
energetic and contractile proteins, was observed. Initially, we focussed on the identification of PTMs of four
selected proteins, using a multiple digestion protocol
to enhance sequence coverage (see figure). PTMs
were identified by standard NanoLC electrospray ionization ion trap mass spectrometry (nanoLC-ESI-MS/
MS) and linear ion trap fourier transform ion cyclotron
resonance mass spectrometry (LTQ-FT-ICR-MS/MS)
and revealed several modified and truncated species.
Protein species resulting from all protein modifications, post-translational chemical modifications and
protein truncation are different products of one single
gene. These modifications influence subcellular location, degradation, subunit assembly, tertiary structure
or enzyme activity and thus protein function.
Therefore, prime importance should be rather given
to systematically identify and specify proteins at their
species level than to quantify total protein amount.
The newly detected protein species were regulated in
the myocardium of mice related to age, sex and oral
genistein treatment. Therefore those species could
be relevant in cardiac disease and should
be taken into consideration for the molecular
understanding of pathological processes.
Workflow of 2-DE and MS strategy for identification
of proteins and protein species. 2-DE two-dimensional gel electrophoresis, ESI–MS/MS electrospray
ionization mass spectrometry, LTQ-FT-ICR-MS/
MS linear ion trap Fourier transform ion cyclotron
resonance mass spectrometry, db database
184
Franz Theuring – Molecular Pharmacology
2.Phosphorylation analysis of the mouse kidney proteome
Project leader
Coworkers
Funding
Cooperation
Franz Theuring
Boris Neumann, Karima Schwab
University Research Funding
Dr. C. Scheler, Proteome Factory, Berlin
Phosphorylation is one of the most prominent posttranslational modifications of proteins. It regulates
a wide range of metabolisms in living cells. Modern
methodology utilises mass spectrometry (MS) for the
detection and localisation of phosphorylation sites.
However the detection and site-specific characterization is a challenging task in life science. While immunologic techniques are highly specific (often limited to a
single site and protein) and have a low detection limit
the undirected analysis via mass spectrometry is often
hindered by the low abundance and poor ionization of
the phosphorylated analyte in common methodology.
Essential for the detection of phosphorylation sites in
“real-life” samples via common mass spectrometers
is an antecedent enrichment of the phospho-proteins
or –peptides. While mass spectrometers for the analysis of proteins or peptides (i.e.: MALDI-MS, ESI-MS/
MS) permit an indirect detection of phosphorus due to
mass shift and neutral loss, the usage of element mass
spectrometers (ICP-MS) allows the direct detection on
atomic level. ICP-MS has unrivalled detection limits but
discards structural information of the analyte.
Current focus of the project is the evaluation of
common phosphopeptide or –protein enrichment
schemes for the mouse kidney proteome by ICP-MS.
Further the project focusses the promising combination of nanoLC-ESI-MS/MS with ICP-MS in a parallel
fashion to produce merged data of structural data,
proteins phosphorylation and its absolute abundance.
2D-PAGE of 120 mouse
kidney proteins. Left:
initial sample, mid:
flow-through, right:
MOAC-eluate. Horizontal: isoelectric focussing (pH 4-10), vertical:
SDS-PAGE (approx. 250
kDa - 8 kDa)
185
Franz Theuring – Molecular Pharmacology
3. E
ndothelin-1 transgenic/eNOS Knockout Mice : A new genetic mouse
model to study gender and age effects in cardiovascular diseases
Project leader
Co-workers
Funding
Cooperation
Franz Theuring
Nicolas Vignon-Zellweger, Karima Schwab
Marie Curie Host Fellowship for ES-Training (CARDIOVASC,
to N. V-Z and F.T), and DFG (TH 466/7-1)
Prof. B. Hocher, CCR; Dr. J.P. Stasch, Bayer Schering Pharma, Wuppertal
The intact endothelium produces a variety of vasoactive substances. Important among those is endothelin-1 (ET-1), a potent vasoconstrictor and mitogen in
vivo and in vitro. The nitric oxide (NO) system is considered as the natural functional counterpart of the
ET system, thus contributing to the subtle balance of
vascular tone. The clinical relevance of this delicate
interplay has been acknowledged because of its implication in many cardiovascular diseases such as pulmonary arterial hypertension, systemic hypertension,
and coronary artery disease. However, the underlying
molecular mechanisms remain to be fully clarified.
ET-1 overexpressing transgenic mice develop a sclerotic and/or fibrotic renal, pulmonary and myocardial
phenotype. Surprisingly the ET+/+ mice remain normotensive. This lack of hypertension is believed to be
the consequence of a compensatory effect of the nitric
oxide system. To disrupt this compensatory effect we
decided to crossbreed ET+/+ mice and eNOS knockout mice to generate the four different genotypes:
ET+/+, eNOS-/-, ET+/+eNOS-/-, and WT.
The crossbred animals (ET+/+eNOS-/-) develop significantly high systolic blood pressure compared to WT
mice and eNOS-/- mice. Furthermore, at the age of
nine month , the eNOS-/-, but not the ET+/+eNOS-/mice, are characterized by diastolic dysfunction. These
findings suggested that transgenic overexpression of
186
ET‐1 on an eNOS‐/‐ background could be beneficial
for diastolic functions.
In order to identify the underlying mechanisms leading
to this phenotype, molecular, histological, physiological and protein chemical methods were employed.
In particular to analyze for the complete cardiac proteome of three months old animals high resolution
two Dimensional Gel Electrophoresis coupled to mass
spectrometry was performed. We could demonstrate
that the cardiac proteome of the three different genotypes compared to WT animals resulted in prominent
changes of the cardiac protein abundance.
The proteomics study revealed that transgenic overexpression of ET‐1, with or without eNOS, led to a
higher abundance of proteins regulating oxidative
stress indicating that, in contrast to eNOS‐/‐ animals,
ET+/+ and ET+/+eNOS‐/‐ mice developed molecular mechanisms limiting oxidative damages. Moreover,
diastolic dysfunction observed in eNOS‐/‐ mice may
be explained by the differential abundance of proteins
involved in the contractile machinery. Overexpression
of ET‐1 in eNOS‐/‐ mice restored these changes
and may have thereby benefited the cardiac functions.
Finally, this study indicated that a shift from fatty acid to
glucose metabolism, considered as cardioprotective,
may have occurred to a greater extent in crossbred
animals than in eNOS‐/‐ mice.
Franz Theuring – Molecular Pharmacology
4. C
haracterization of signalling properties of the Endothelin
system in mouse mammary gland physiology
Project leader
Coworkers
Funding
Franz Theuring, Andreas Fischer
Nadir Gül
Charite-Stipend
Endothelin 1 (ET-1) is a small vasoactive peptide having
wide physiological effects on vascular homeostasis
and a variety of physiological and pathophysiological
processes unrelated to cardiovascular physiology. It
exerts its effect by binding to two distinct G Protein
Coupled Receptors (GPCR). In addition to the classical GPCR signaling pathways, these receptors are also
able to activate structurally unrelated receptors such
as those belonging to the receptor tyrosine kinase
(RTK) family in what is termed receptor transactivation.
Deregulation of this transactivation as induced by
overexpression, amplification or mutation of critical
pathway elements and autocrine stimulation through
aberrant growth factor loops is frequently linked to
hyperproliferative diseases.
The aim of this project is to further characterize the
crosstalk between these two systems in the mammary epithelium in order to elucidate its contribution
to mammary gland physiology and pathophysiological
processes. This is achieved by the detailed molecular
and histological characterization of female transgenic
mice overexpressing ET-1. These mice were found
to exhibit a lactational incompetence while lactating
newborn animals. In parallel, an in vitro system was
established employing mammary epithelial cell lines to
further study relevant cell biological processes in these
specific cell types. First results suggest that endothelin
is able to activate various signaling pathways in vivo
and in vitro and that this activation is accompanied by
impaired lactational competence in ET-1 transgenic
animals. To further characterize the molecular events
involved in this project, functional experiments employing various inhibitor compounds will be performed in
order to identify the signaling pathways mediating the
observed effects.
Whole mount preparations
of wild type and ET-1 transgenic mammary glands from
lactation day 3 demonstrating
lactational incompetence in
the ET-1 transgenic mammary
glands.
187
Franz Theuring – Molecular Pharmacology
5. Pharmacological Modulation of the Intestinal Barrier
Project leader
Coworkers
Funding
External cooperations
Andreas Fischer, Franz Theuring
Markus Gluth, Cornelia Tanneberger
University Research Funding, Industry funding
Prof. D.C. Baumgart, Dr. U.F. Pape – Division of Hepatology
and Gastroenterology CVK
The intestinal barrier constitutes the largest mucosal
surface of the human body, separating the highly
antigenic environment of the intestinal lumen from
the milieu intérieur. Perturbations of this barrier have
long been recognized as key features in inflammatory
bowel diseases such as Crohn’s disease and ulcerative colitis, but have also been found in celiac disease, graft-versus-host disease and food allergies. In
addition, structural components of the epithelial tight
junctions have been identified as primary targets for
various pathogenic bacteria-derived toxins. Pharmacological strategies to modulate the permeability of
intestinal tight junctions therefore represent an attractive approach to improve the management of these
disorders.
Using Caco-2 and T-84 monolayers cultivated on
semipermeable filter supports as a model system of
the intestinal epithelial barrier, our work focuses on the
analysis of the impact of proinflammatory cytokines
such as TNFα or IFNγ on barrier function. Using specific inhibitor compounds, we aim at characterizing
signaling pathways responsible for the deleterious
effects of these cytokines in order to identify potential
targets for the treatment of inflammatory bowel diseases. In addition, compounds well established in the
treatment of these disorders such as glucocorticoid
hormones and anti-TNFα antibodies are evaluated
in terms of their effect on barrier function. By further
188
characterizing the mechanisms regulating paracellular permeability in the intestine, these studies might
provide a basis for developing new pharmacological
approaches to modulate barrier function.
Adalimumab prevents barrier disruption by the proinflammatory cytokines TNFα and IFNγ. T-84 intestinal epithelial cells
were treated with TNFα and IFNγ or a combination of both
cytokines and the therapeutic anti-TNFα antibody adalimumab. The tight junction components occludin and ZO-1 were
visualized by immuno-staining.
Franz Theuring – Molecular Pharmacology
6. C
haracterization of the Rho Signaling Pathway and its Role in the
Regulation of Tight Junction Permeability
Project leader
Coworkers
Funding
External cooperations
Andreas Fischer, Franz Theuring
Markus Gluth, Cornelia Tanneberger
University Research Funding
Prof. J.R. Turner, University of Chicago
Among signaling pathways associated with the tight
junction, the small GTPase RhoA has been shown to
be of pivotal importance to the control of cell proliferation, migration and differentiation. Previous studies have demonstrated that expression of either constitutively active or dominant negative RhoA results
in reduced tight junction barrier function in cultured
MDCK monolayers. We have shown that modulation
of signaling pathways downstream of RhoA regulates
tight junction function in the mammary epithelium,
both in vivo and in vitro. However, the precise mechanisms by which RhoA controls barrier function have
not been defined.
Therefore, our work focuses on the characterization
of RhoA and one of its downstream targets, the protein kinase PKN in order to elucidate molecular events
involved in the RhoA-mediated regulation of epithelial
tight junction permeability.
By employing an inducible expression system as an in
vitro barrier model, the effects of RhoA and PKN can be
studied at various developmental stages, enabling us
to delineate their contribution to the regulation of tight
junction function. Functional observations are complemented by the analysis of changes in the expression and localization of key tight junction components
and the characterization of the molecular cross-talk to
other signaling pathways. As Rho GTPases are pivotal
in the pathogenesis of a variety of diseases including arterial and pulmonary hypertension, myocardial
hypertrophy and vasospastic angina as well as tumorigenesis and metastasis, our work might contribute
to a better understanding of the signaling pathways
downstream of these molecules.
A) Dose- and time-dependend
induction of RhoA expression. B)
Immunofluorescent staining of RhoA
expression (red) and MLC phosphorylation (green) after doxycycline
withdrawal in Caco-2 intestinal
epithelial cells.
189
Franz Theuring – Molecular Pharmacology
7. Alzheimer’s Disease: Testing of new drug candidates in tau-transgenic mice
Project leader
Coworkers
Funding
Cooperation
Franz Theuring
Silke Dietze, Claudia Zabke, Mandy Magbagbeolu, Bettina Seelhorst,
Valeria Melis, Karima Schwab, Anna Thoma
TauRx Therapeutics, WisTa Laboratories, Singapore
Prof. C.M. Wischik, University of Aberdeen, Scotland
Tau transgenic mouse lines had been established to in the memory-critical brain regions where the density
further analyse the functional role this protein and its of Alzheimer tangles is greatest. In the control group,
aggregates, the so-called tau tangles play in clinical there was a significant decline from the starting score
dementia, i.e. Alzheimer’s Disease and to test puta- in cognitive testing and on brain scans.
tive new drug candidates in a preclinical setting to fight By employing our transgenic mice a group of secondthis neurodegenerative and terminal brain disease. In a generation rember® derivatives had been discovered
most fruitful scientific collaboration with Prof. Wischik’s and successfully tested. These compounds have the
group from the University of Aberdeen for the last 12 same mechanism of action as rember® acting as Tau
years these mice had been pivotal in establishing a rel- Aggregation Inhibitors, with potential utility in the treatevant transgenic tau-based Alzheimer mouse model. ment of Alzheimer’s disease and other neurodegeneraSeveral dozens of newly discovered and synthesized tive disorders. TauRx has now initiated preparations for
drug candidates had been tested in vivo for their activ- Phase 3 studies in mild and moderate AD, and also in
ity in reducing tau pathology and to enhance cognitive orphan indications such as Fronto-Temporal Dementia
behaviour and motor skills in the drug-treated animals. and provisionally Progressive Supranuclear Palsy.
By teaming up with TauRx Pharma - a Singapore-based These data underline the importance of generating
company spun out of the University of Aberdeen and relevant transgenic mouse models and their use in
the Charité - it is now our great pleasure to reveal a identification and validation of new drug candidates
major breakthrough in the treatment of Alzheimer’s dis- for human diseases, which then are planned to enter
ease. A total of 321 patients with mild and moderate into clinical testing.
Alzheimer’s Disease were treated in a phase II
clinical trial with Rember® a novel form of methylthioninium chloride (MTC). Patients receiving the study treatment experienced an 81%
reduction in cognitive decline over one year,
and did not experience a significant decline in
their mental function over 24 months. In addition patients had repeated brain scans at the
start of the study and after 25 weeks. These An increase of tau aggregates in cortical sections of transgenic mice is
showed that the treatment effect was greatest observed with increasing age (left: 6 mo old, right 12 mo old).
190
Franz Theuring – Molecular Pharmacology
8. Parkinson’s Disease : Testing of new drug candidates
in α-synuclein transgenic mice
Project leader
Coworkers
Funding
Cooperation
Franz Theuring
Silke Dietze, Mandy Magbagbeolu, Bettina Seelhorst, Karima Schwab,
Anna Thoma, Claudia Zabke
WisTa Laboratories, Singapore
Prof. C.M. Wischik, University of Aberdeen, Scotland
Parkinson’s disease (PD) is a common human neuro- tion or reversal of synuclein aggregation is believed to
degenerative movement disorder and affects 1% of the be of therapeutic benefit.
elderly population. PD is neuropathologically charac- The development of drugs that prevent this aggreterized by a marked and progressive degeneration of gation form the basis for the scientific rationale
dopaminergic neurons and by the presence of fibrillar for this project. The aim is to model the molecular
cytoplasmic inclusions (Lewy bodies [LBs]) and dys- processes of α-synuclein aggregation in an animal
trophic neurites (Lewy neurites [LNs]) in the substantia model to test and evaluate new therapeutic drug
nigra and other regions of the brain. Although the loss of candidates for PD.
dopamine neurons is certainly related to the major clini- Consequently α-synuclein transgenic mice had been
cal symptoms of PD, the causes and the pathogenesis generated carrying different transgenic constructs.
of this multifactorial disease as well as that of related These mice are currently being characterized in more
“synucleinopathies” are still largely unknown.
detail and will be used to study basic mechanisms
The major components of both LBs and LNs are of the pathogenesis underlying PD. Most importantly,
fibrillar aggregates of α-synuclein. α Synuclein is a these mice will then be used to screen for α-synuclein
widely expressed, neuronal presynaptic protein that aggregation inhibitors to facilitate the development of a
appears to play a role in membrane-associated proc- new therapeutic intervention for this disease.
esses and synaptic plasticity
and has been linked to learning
and development processes.
While the mechanism(s) of formation of LBs and LNs and their
association with PD are yet not
understood, several lines of evidence suggest that α synuclein
fibrillization is associated with PD
and that α-synuclein fibrillization Hippocampal sections of wildtype (left) and transgenic mouse (right) brains exhibiting
causes toxicity. Thus the inhibi- prominent α-synuclein staining
191
Franz Theuring – Molecular Pharmacology
Publications ( 2006-2010 )
Bert B, Fink H, Hörtnagl H, Veh RW, Davies B, Theuring F,
Kusserow H (2006): Mice over-expressing the 5-HT1A receptor in cortex and dentate gyrus display exaggerated locomotor and hypothermic response to 8-OH-DPAT. Behav Brain
Res 167: 328-41.
Davies B, Behnen M, Cappallo-Obermann H, Spiess AN,
Theuring F, KirchhoffC.(2006): Novel epididymis-specific
mRNAs down-regulated by HE6/Gpr64 receptor gene disruption. Mol Reprod Dev 74: 539-553
Quaschning T, Voss F, Relle K, Kalk P, Vignon-Zellweger
N, Pfab T, Bauer C, Theilig F, Bachmann S, Kraemer-Guth
A, Wanner C, Theuring F, Galle J, Hocher B (2007): Lack
of Endothelial Nitric Oxide Synthase Promotes EndothelinInduced Hypertension: Lessons from Endothelin-1 Transgenic/Endothelial Nitric Oxide Synthase Knockout Mice. J
Am Soc Nephrol 18: 730-740
Slowinski T*, Kalk P*, Christian M, Schmager F, Relle K,
Godes M, Funke-Kaiser H, Neumayer NN, Bauer C, Theuring
F*, Hocher B* (2007): *authors contributed equally. Cell-type
specific interaction of Endothelin- and Nitric Oxide System
- Pattern of prepro-ET-1 expression in kidneys of L-NAME
treated prepro ET-1 promoter-LacZ-transgenic mice. J.
Physiol 581: 1173-1181 Epub 2007 Mar 29
Fischer A, Stuckas H, Gluth M, Russell TD, Rudolph MC,
Beeman NE, Bachmann S, Umemura S, Ohashi Y, Neville
MC,* Theuring F* (2007): *both authors contributed equally.
Impaired Tight Junction Sealing and Precocious Involution in
Mammary Glands of PKN Transgenic Mice. J Cell Sci 120:
272-2283
Quaschning T, Voss F, Herzfeld S, Relle K, Kalk P, Godes
M, Pfab T, Kraemer-Guth A, Bonz AW, Theuring F, Galle J,
Hocher B (2008): Lack of iNOS impairs endothelial function in
endothelin-1 transgenic mice. Kidney Blood Press Res 31:
127-134 Epub 2008 Apr 7
Heiden S, Pfab T, von Websky K, Vignon-Zellweger N, Godes
M, Relle K, Kalk P, Theuring F, Zidek W, Hocher B (2008):
Tissue specific activation of the endothelin system in severe
acute liver failure. Eur J Med Res 13: 327-329
Bert B, Voigt JP, Kusserow H, Theuring F, Rex A, Fink H
(2009): Increasing the number of 5-HT1A-receptors in cortex
and hippocampus does not induce mnemonic deficits in
mice. Pharmacol Biochem Behav 92: 76-81. Epub 2008
Oct 31
Küster K, Grötzinger C, Koschel A, Fischer A, Wiedenmann
B, Anders M (2010): Sodium butyrate increases expression of
the Coxsackie and Adenovirus Receptor in colon cancer cells.
Cancer Invest 28: 268-274
Russell TD, Palmer CA, Orlicky DJ et al. (2007): Cytoplasmic
lipid droplet accumulation in developing mammary epithelial
cells: roles of adipophilin and lipid metabolism. J Lipid Res
48:1463-75
Küster K, Koschel A, Rohwer N, Fischer A, Wiedenmann
B, Anders M (2010): Downregulation of the coxsackie and
adenovirus receptor in cancer cells by hypoxia depends on
HIF-1alpha. Cancer Gene Ther 17: 141-146
Baumgart DC, Fischer A (2007): Virchow’s node. Lancet
370:1568
Schwab K, Neumann B, Scheler C, Jungblut PR, Theuring
F (2010). Adaptation of proteomic techniques for the identification and characterization of protein species from murine
heart. Amino Acids: Jul 6. [Epub ahead of print].
Baumann C, Davies B, Peters-Kottig M, Kaufmann U, Lessl
M, Theuring F (2007): AKR 1B7 (Mouse Vas Deferens Protein)
is dispensible for mouse development and reproductive success. Reproduction 134:97-109
Deng DR, Djalali S, Ahnert-Hilger G, Große G, Stroh T, Voigt,
I, Kusserow H, Theuring F, Hen, R, Hörtnagl H (2007): Embryonic and postnatal development of the serotonergic raphe
system and its target regions in the brains of 5-HT1A receptor
knockout and transgenic mice. Neuroscience 147: 388-402.
Epub 2007 Jun 1
192
Franz Theuring – Molecular Pharmacology
General information
Third party funding ( 2006-2010 )
Project leader
Schwab K.
Theuring F.
Gül N.
Theuring F.
Theuring F.
Project titel
Sex, age and diet influences on the
cardiac proteome
Characterisation of the mouse mammary gland
Tau and a-synuclein transgenic mice
Theuring F.
Endothelin and NO interaction in cardiac remodelling
Cardiac proteome analysis
Analysis of PKN1
Theuring F.
Fischer A.
Theuring F.
Sponsor
Hypathia and Charite PhD programs
Charite PhD program
Period
2006-2009
TauRxTherapeutics/WisTa Laboratories
EU-EST Program CARDIOVASC
2006-2010
DFG, TH 466/7-1
DFG FI 1718/1-1
Applied for: 01.06.2010
2009-2010
2010-2012
2006-2008
2006-2008
Awards
2008
United European Gastroenterology Federation
Travel Grant Award: (Adalimumab Prevents TNFα Induced
Barrier Disruption in an In vitro Model)
Andreas Fischer
193
Thomas Unger – Pharmacology
Head of the Group Pharmacology
Prof. Dr. med. Thomas Unger
Curriculum Vitae: Professor Thomas Unger holds the Chair of Pharmacology and
is Director of the Institute of Pharmacology at the Charité – Universitätsmedizin Berlin. He is also the Director of the Center for Cardiovascular Research (CCR) at the
Charité, Berlin and the Chairman of the German Institute for High Blood Pressure
Research in Berlin (DIB). Between 1994 and 2001, he was Director of the Institute of
Pharmacology at the University of Kiel, Germany. Professor Unger studied medicine
in Germany and the UK, and gained his MD from the University of Heidelberg, Germany. He then carried out
postdoctoral research at the Clinical Research Institute of Montreal, Canada, and the Department of Pharmacology in Heidelberg, where he received his PhD in Pharmacology. Until 1994, Professor Unger held professorships
in pharmacology and hypertension research at the University of Heidelberg. In recognition of his work, Professor Unger has received the German Hypertension Society´s Franz Gross Award for Hypertension Research,
the Meilahti Lecture Award of the Medical Faculty, University of Helsinki, Finland, the Björn Folkow Award of
the European Society of Hypertension, and the Robert Tigerstedt Award of the Finnish Hypertension Society.
He is a member of the German Societies of Pharmacology, Cardiology and Hypertension (Council Member
1995–2001), the International Society of Hypertension, the European Society of Hypertension (Council Member
1989–97), the European Council for Blood Pressure and Cardiovascular Research (President, 2000–2), the
Inter-American Society of Hypertension, He is Honorary Member of the British, Finnish and Italian Hypertension
Society and a Fellow of the European Society of Cardiology and the American Heart Association. Professor
Unger has authored more than 700 scientific publications. He is or has been a member of the Editorial Boards
of the American Journal of Physiology, American Journal of Hypertension, Biochemical Pharmacology, Blood
Pressure, Cardiovascular Drugs and Therapy, Clinical and Experimental Hypertension, Hypertension, Hypertension Research, Journal of Hypertension, Fundamental and Clinical Pharmacology, Physiological Genomics,
Regulatory Peptides, High Blood Pressure & Cardiovascular Prevention.
195
Thomas Unger – Pharmacology
Members of the group
Administrative Assistant
Schröder, Miranda
Groups of the Institute of Pharmacology in the CCR
196
Workshop
Röder , Bettina
HEIKO FUNKE-KAISER – Pharmacology
Head of the group
PD Dr. med. Heiko Funke-Kaiser
Curriculum Vitae: He studied medicine at the universities of Düsseldorf and Berlin.
He obtained his medical approbation (3rd state examination) in 1998 and received
his MD degree - based on an experimental work at the Institute of Clinical Pharmacology and Toxicology of the Freie Universität Berlin in 1999. After a specialization
(“Facharzt”) in pharmacology and toxicology in 2004, he built up his research group
in the Institute of Pharmacology at the Center for Cardiovascular Research (CCR).
In 2008 he passed his habilitation in pharmacology and toxicology. His research focus is on eukaryotic gene
regulation and drug development
Members of the group
Scientists
Zollmann, Frank
Goldin-Lang, Petra
Schmitz, Jennifer
Li, Yaosi
Schmerbach, Kristin
Seidel, Kerstin
Technicians
Klare, Sabrina
Kroh, Melanie
Students
Kirsch, Sebastian
Zaade, Daniela
Bernhard, Sarah
Hope, Antonia
Schrezenmeier, Eva Dr. med.
Dr. rer. nat.
Dr. rer. nat.
Physician
Dipl.-Biol.
Dr. rer. nat.
Dipl.-Biol.
Dipl.-Biochem., PhD student
M.Sc., PhD student
MD student
MD student
MD student
197
HEIKO FUNKE-KAISER – Pharmacology
Summary
Drug development
Cardiac, renal and ophthalmological end-organ
damage due to hypertension and/ or diabetes is
currently one of the major medical challenges. They
contribute to approximately 60-80% of all heart
failure and 70% of all renal failure cases. In addition, up to 30% of blindness in western countries
is caused by these diseases. Current therapeutic
strategies, such as ACE inhibitors, angiotensin AT1
receptor blockers (ARBs), b-adrenergic receptor
antagonists or antidiabetic agents only ameliorate
but do not abolish cardiovascular end-organ damages. Therefore, cardiovascular end-organ damage represents an unmet medical need.
Worsening this situation, the pharmaceutical
industry faces a productivity crisis. While research
and development (R&D) spend has risen annually
~7% over the last decade, investigational new
drug applications (IND) and new molecular entity
(NME) output largely remained unaltered. This
insufficient transfer of scientific concepts into
approved drugs opens interesting opportunities
for small and flexible biotech companies based
on an academic backbone.
Supported by the BMBF GO-Bio initiative [http://
www.bmbf.de/de/6868.php] the aim - as described
below in greater detail - of an interdisciplinary project
team is to develop a novel drug class, which inhibits
the renin/ prorenin receptor, for the indication renal
and cardiac end-organ damage. Within this GO-Bio
program a start-up company called CCR Pharma
has been founded as a spin-off from the Charité to
develop scientific ideas up to clinical phase I and to
license compounds in return for payments ensuring
the growth of CCR Pharma´s own pipeline.
198
Eukaryotic gene regulation
Eukaryotic gene regulation - especially on the promoter level - is a further major focus of our group.
Promoters are key regulatory elements in our
genome that transform genomic information into
structure. They determine when (e.g. regarding
developmental biology), where (i.e, cell and tissue
specificity) and under which (patho)physiological
conditions a gene is transcribed. Furthermore, they
can increase the complexity of the transcriptome
relative to the genome, since, e.g., multiple mRNA
isoforms can be generated from one gene through
the use of alternative promoters. This mechanism is
of importance in the “postgenomic era” which has
to explain the physiology and pathophysiology of the
human species with its relative low gene number.
The (patho)physiological importance of promoters is
demonstrated by effects of genetic polymorphisms
within promoter regions which can be associated
with psychological traits, physiological traits and the
susceptibility for certain diseases. Furthermore, promoter polymorphisms are involved in the frequently
discussed gene-environment interactions as well as
pharmacokinetic and pharmacodynamic aspects.
Our past and current projects center around the following topics:
(1) Basic mechanisms of transcriptional regulation
including epigenetics,
(2) functional characterisation of polymorphisms in
regulatory regions,
(3) altered gene regulation in cardiovascular and
neurodegenerative diseases,
(4) and transcriptional regulation in developmental
biology.
HEIKO FUNKE-KAISER – Pharmacology
Methods used by our group to explore these topics include e.g. yeast-two-hybrid screening and
coimmunoprecipitation (CoIP) for protein-protein
interactions, electromobility shift assay (EMSA) and
chromatin-immunoprecipitation (ChIP) for proteinDNA interactions as well as promoter reporter gene
assays, 5´-RACE and RNase protection assay (RPA)
for promoter analysis. The binding of unknown tran-
scription factors to regulatory regions is analysed
by DNA affinity chromatography and mass spectrometry.
Furthermore, we consider promoters as therapeutic
targets which can be influenced by classical drugs (e.g.
steroids and glitazones) and gene therapy (e.g. decoy
oligonucleotides and triple helix forming substances),
driving synergy with our drug development focus.
Zusammenfassung
Medikamentenentwicklung
Kardiale, renale und ophthalmologische Endorganschäden bedingt durch arterielle Hypertonie und/ oder
Diabetes mellitus stellen derzeit eines der relevantesten
medizinischen Probleme dar. Sie sind u.a. verantwortlich für ca. 60-80% aller Herzinsuffizienzen, ca. 70%
aller dialysepflichtigen Niereninsuffizienzen und bis zu
30% der Erblindungen in den Industrienationen. Die
bisherigen Therapiestrategien (u.a. ACE-Hemmer,
Angiotensin-AT1-Rezeptorblocker (ARBs), b-Blocker
und orale Antidiabetika) können allesamt die Entwicklung von Endorganschäden nur verlangsamen, jedoch
nicht verhindern. Endorganschäden stellen somit einen
sogenannten "unmet medical need" dar.
Erschwerend kommt hinzu, dass die pharmazeutische Industrie derzeit eine Produktivitätskrise erlebt,
da die Ausgaben für Forschung und Entwicklung
in der letzten Dekade um ca. 7% jährlich gestiegen sind, während die Anzahl an Neuzulassungen
nahezu konstant geblieben ist.
Dies spiegelt unter anderem einen insuffizienten
Transfer von wissenschaftlichen Ideen in die Entwicklung von Pharmaka wider, ermöglicht jedoch
kleinen und flexiblen Biotech-Unternehmen mit akademischem Hintergrund Erfolgsmöglichkeiten.
Wie weiter unten ausführlich beschrieben besteht
das Ziel eines durch die GO-Bio-Initiative des BMBF
[http://www.bmbf.de/de/6868.php] geförderten,
interdisziplinären Projektteams darin, eine neue
Medikamtenklasse, welche den Renin-/ ProreninRezeptor inhibiert, für die Indikation renaler und
kardialer Endorganschäden zu entwickeln. Innerhalb
dieses GO-Bio-Programms wurde ein Unternehmen
mit dem Namen "CCR Pharma" aus der Charité ausgegründet, welches präklinische Medikamentenentwicklungen bis hin zur klinischen Phase I durchführen soll. Dabei sollen u.a. im Rahmen der derzeitigen
Substanzentwicklung generierte Lizenzeinnahmen
dazu genutzt werden, eine solide, finanzielle Basis
für "CCR Pharma" bereitzustellen, um die Entwicklung von innovativen Pharmaka zu gewährleisten.
Eukaryontische Genregulation
Eukaryontische Genregulation, insbesondere auf
Promotorebene, stellt einen weiteren Fokus unserer
Forschungsarbeit dar. Promotoren sind regulative
Schlüsselelemente in unserem Genom und bedingen die Transformation von genetischer Information
in Struktur. Sie bestimmen, wann (z.B. im Rahmen
der Embryonalentwicklung), wo (d.h. Gewebe- bzw.
199
HEIKO FUNKE-KAISER – Pharmacology
Zellspezifität) und unter welchen physiologischen
und pathophysiologischen Bedingungen ein Gen
transkribiert wird. Sie tragen des Weiteren zur Erhöhung der Komplexität des Transkriptoms relativ zum
Genom bei, da mittels alternativer Promotoren eines
Gens multiple mRNA-Isoformen aus diesem generiert werden können. Promotoren sind daher in der
Postgenom-Ära von besonderem Interesse, welche
die Physiologie und Pathophysiologie der humanen
Spezies mit seiner relativ geringen Genanzahl erklären muss.
Die (patho)physiologische Bedeutung von Promotoren zeigt sich anhand des Effektes von genetischen
Polymorphismen in Promotorbereichen, die mit psychologischen Merkmalen, physiologischen Merkmalen und der Anfälligkeit für bestimmte Erkrankungen assoziiert sein können. Des Weiteren spielen
Promotorpolymorphismen bei den vieldiskutierten
Gen-Umwelt-Interaktionen, sowie bei pharmakokinetischen und pharmakodynamischen Fragestellungen eine Rolle.
Unsere Forschungsprojekte weisen folgende
Schwerpunkte auf:
(1) Basale Mechanismen der transkriptionellen
Regulation inklusive Epigenetik,
200
(2) funktionelle Charakterisierung von Promotorpolymorphismen,
(3) Genregulation im Rahmen von kardiovaskulären
und neurodegenerativen Erkrankungen,
(4) sowie transkriptionelle Regulation bei entwicklungsbiologischen Prozessen.
Dabei werden u.a. „yeast-two-hybrid screening“
und Coimmunopräzipitation (CoIP) zur Analyse
von Protein-Protein-Interaktionen, „electromobility
shift assay“ (EMSA) und Chromatin-Immunopräzipitation (ChIP) zur Untersuchung von Protein-DNAInteraktionen sowie Promotor- Reportergen-Assays,
5´-RACE und „RNase protection assay“ (RPA) zur
Promotoranalyse verwendet. Die Bindung von molekular nicht identifizierten Transkriptionsfaktoren an
Promotorregionen wird mittels DNA-Affinitätschromatographie und Massenspektrometrie analysiert.
Schließlich betrachten wir die Promotorebene als
therapeutische Zielstruktur, welche durch klassische
Pharmaka (z.B. Steroide oder Glitazone) und gentherapeutische Ansätze (z.B. “decoy”-Oligonukleotide oder “triple helix”-Bildner) beeinflusst werden
kann, wodurch sich Synergien mit unserem “Drug
development”-Schwerpunkt ergeben.
HEIKO FUNKE-KAISER – Pharmacology
1. D
rug development - the renin/ prorenin receptor as a
pharmacological target
Project leader
Heiko Funke-Kaiser
CoworkersJennifer Schmitz, Petra Goldin-Lang, Sebastian Kirsch, Kristin Schmerbach,
Daniela Zaade, Sabrina Klare, Kerstin Seidel, Melanie Kroh, Sarah Bernhard,
Eva Schrezenmeier, Yaosi Li, Jan H. Schefe, Frank Zollmann
Funding
BMBF GO-Bio
IBB ProFIT (EU/ EFRE)
Stiftung Charité
External CooperationsDr. Jens Peter von Kries (Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin),
Evotec AG, Hamburg, Prof. Dr. Achim Kramer, Dr. Rudolf Volkmer-Engert
(Department of Immunology, Charité)
Cardiac, renal and ophthalmological end-organ damage due to hypertension and/ or diabetes is currently
one of the major medical challenges. They contribute
to approximately 60-80% of all heart failure and 70%
of all renal failure cases. In addition, up to 30% of blindness in western countries is caused by these diseases.
Current therapeutic strategies, such as ACE inhibitors,
angiotensin AT1 receptor blockers (ARBs), b-adrenergic receptor antagonists or antidiabetic agents only
ameliorate but do not abolish cardiovascular endorgan damages. Therefore, cardiovascular end-organ
damage represents an unmet medical need.
A human renin/ prorenin receptor (RER, also called (P)
RR), which can specifically bind prorenin and renin,
has been cloned in 2002. Binding of renin to this receptor increases renin´s catalytic activity about 4- to 5-fold.
Furthermore, the receptor is able to unmask the enzymatic activity of prorenin.
The RER plays a crucial role in cardiac and renal endorgan damage. Various animal models from independent research groups demonstrated that inhibition of
prorenin binding to the RER by parenteral delivery
of a decoy peptide can prevent or even abolish the
development of cardiac fibrosis and diabetic nephropathy via angiotensin II-independent mechanisms.
Therefore, the RER is a promising therapeutic drug
target for cardiovascular disease. In addition, recent
publications indicate that the RER might also be a drug
target in oncology, e. g. based on the direct physical
interaction between this protein and Wnt receptors
which is crucial for Wnt signaling.
Our group revealed a novel signal transduction
pathway involving direct protein–protein interaction
between the RER and the transcription factor promyelocytic zinc finger protein (PLZF) as well as the
nuclear translocation of PLZF upon renin and prorenin
stimulation. Downstream effects of a RER activation by
renin and prorenin include repression of the RER gene
itself, induction of the p85alpha subunit of the phosphatidylinositol-3 kinase (PI3K-p85alpha) and - consistently - an increase in proliferation and a decrease
in apoptosis.
This (pro)renin-RER-PLZF-RER/ PI3K pathway was
filed as a patent (disclosure EP 1 890 152 A1; PCT
201
HEIKO FUNKE-KAISER – Pharmacology
filing PCT/EP2007/006100), since it can be used as
a read-out for RER activity within high-throughput
screening (HTS) assays.
The aim of an interdisciplinary project team is to
develop a novel drug class called renin/ prorenin
receptor blockers (RERBs). RERBs will represent
small molecules with oral bioavailability and the ability
to inhibit the renin/ prorenin receptor for the indication
hypertension- and diabetes-related renal and cardiac
end-organ damage.
During a complex assay development phase, two
HTS assays based on double stably transfected cell
lines and a luciferase read-out were established.
Subsequently, two independent HTS campaigns of
about 100,000 and about 20,000 compounds (cpds),
respectively, have been successfully performed. Afterwards, the primary hits were subjected to hit confirmation and hit profiling phases, with the latter analysing
dose-response relationships.
To validate the confirmed compounds generated by
these primary screenings, multiple secondary screening assays e. g. based on nuclear translocation of
PLZF, mRNA quantification or protein-protein/ proteinDNA interactions were set up to yield so-called hits.
In addition, a hit-to-lead program to filter down these
multiple hits according to pharmacodynamic, pharmacokinetic and toxicologic parameters has been
prepared. Accordant methods including Ames test,
micronucleus test, PAMPA assay, cytochrome P450
induction as well as the cloning/ expression of multiple recombinant proteins of the (pro)renin-RER-PLZF
pathway for subsequent analysis by e.g. the Alpha
Screen technology or Biacore have been built up.
Finally, after complex spin-off negotiations with the
Charité, which were finalised in Summer 2010 by
notarial signing, a start-up company called CCR
Pharma GmbH [http://www.ccr-pharma.de] will be
founded in late 2010. Furthermore, senior advisers
regarding e.g. medicinal chemistry and a putative CFO
have been recruited. CCR Pharma´s aim is to develop
RERBs up to clinical phase I/ II and to license compounds in return for payments ensuring the growth of
CCR Pharma´s own pipeline.
,
Function and signal transduction of the
renin/ prorenin receptor (RER).
RERBs (Renin/ Prorenin Receptor
Blockers) designate a putative novel
drug class which inhibits the RER (also
abbreviated (P)RR).
202
HEIKO FUNKE-KAISER – Pharmacology
2. Role of PLZF in neuroprotection
Project leader
Coworkers
Funding
External Cooperations
Heiko Funke-Kaiser
Kerstin Seidel, Sebastian Kirsch, Kristin Lucht, Daniela Zaade, Jana Reinemund,
Jennifer Schmitz, Sabrina Klare, Yaosi Li, Jan H. Schefe, Kristin Schmerbach,
Petra Goldin-Lang, Frank Zollmann
IBB ProFIT (EU/ EFRE)
EU Network of Excellence “InGenious HyperCare”
Christa Thöne-Reineke (Charité)
Stroke is one of the major medical burdens in industrialised countries. Animal experiments indicate that blockade of the angiotensin AT1 receptor (AT1R) improves
neurological outcome after cerebral ischemia. These
protective effects are partially mediated by the angiotensin AT2 receptor (AT2R). The transcription factor
promyelocytic zinc finger protein (PLZF) was identified
as a direct adapter protein of the AT2R. Furthermore,
our group was able to demonstrate that PLZF also
directly binds and mediates the effects of the human
(pro)renin receptor ((P)RR, also called RER) which is
involved in brain development. Therefore, we hypothesised that PLZF is involved in neuroprotection.
Here we show that PLZF and its receptors (P)RR and
AT2R exhibited an
ubiquitous expression pattern in different brain regions.
Furthermore, stable
PLZF
overexpression in human neuronal cells was able
to mediate neuroprotection in a glutamate
toxicity model in vitro.
Consistently, PLZF
mRNA and protein
were downregulated on the ipsilateral side in a stroke
model in vivo, whereas the neurodetrimental PLZF target genes cyclin A2 and BID were upregulated under
this condition. Further analyses indicated that the neuroprotective AT2R is upregulated upon stable PLZF
overexpression in cultured neuronal cells.
Finally, reporter gene assays demonstrated the functionality of (P)RR promoter polymorphisms regarding
basal and PLZF-induced activity.
Our data indicate that the transcription factor PLZF
could be a novel regulator in the pathophysiology of
ischemic brain injury based on its neuroprotective
role in vitro and its downregulation on the stroke side
in vivo.
Mechanisms of PLZF-induced neuroprotection.
The effects of PLZF on the neuroprotective factor AT2R, the proproliferative protein cyclin A2 and the
pro-apoptotic protein BID are shown.
The link between pro-proliferative
signals and neuronal loss is based on
the fact that activation of the cell cycle
causes cellular death in terminally differentiated neurons.
203
HEIKO FUNKE-KAISER – Pharmacology
3. R
ole of promoter polymorphisms within cardiovascular genes
in Alzheimer´s disease
Project leader
Coworkers
Cooperations
Funding
Heiko Funke-Kaiser
Yaosi Li, Michael Klein, Antonia Hope, Peter Marschall, Jens Schacherl,
Sylvia Scheele, Kristin Schmerbach, Kerstin Seidel, Petra Goldin-Lang,
Jennifer Schmitz, Mario Menk, Jan H. Schefe, Jana Reinemund
Kompetenznetz Demenzen [PD Dr. Heike Kölsch, Prof. Dr. Wolfgang Maier
(Department of Psychiatry, University Bonn), Dr. Oliver Peters (Department of
Psychiatry, Charité), Dr. Christian Roos (Leibniz-Institut für Primatenforschung,
Göttingen), Ulrich Kintscher (CCR, Charité), Prof. Dr. Achim Kramer,
Dr. Rudolf Volkmer-Engert (Department of Immunology, Charité),
Prof. Dr. Peter Walden, Dipl.-Biochem. Rebecca Hugel
(Department of Dermatology, Charité)
Dr. Werner Jackstädt-Stiftung
Alzheimer’s disease (AD) is the most common form
of dementia affecting approximately 5 million patients
within the European Union. AD can be grouped into
seldom familial (monogenetic) and frequent sporadic
forms. Regarding the pathophysiology of AD extracellular deposition of amyloid b (Ab) plaques, composed of
Ab40-42, as well as intraneuronal neurofibrillary tangles,
consisting of hyperphosphorylated tau protein, seem
crucial. Cardiovascular risk factors such as hypertension,
diabetes and hypercholesterolemia not only contribute
to cardiovascular morbidity but also increase the risk
for Alzheimer´s disease. Research in recent years suggests that cholesterol modulates synthesis of amyloid ß
which itself is a potent vasoconstrictor and can cause
endothelial dysfunction. Clinical data indicate that cerebral hypoperfusion precedes cognitive decline in AD.
In addition, cerebral vessels in AD can be affected not
only by the amyloid angiopathy but also by alterations of
endothelial cells, VSMCs and the basement membrane.
In sporadic forms of AD the decreased degradation/
clearance of amyloid b is an important pathophysi204
ological factor. Important cardiovascular gene products such as angiotensin-converting enzyme (ACE),
neprilysin (NEP) and endothelin-converting enzyme-1
(ECE-1) are capable of degrading amyloid b. ECE-1
can degrade Ab40 and Ab42 in vitro. Furthermore,
cerebral Ab40/42 content is significantly increased in
ECE-1 knockout mice compared to wild type animals.
Since sporadic Alzheimer´s disease has a strong genetic
component of about 80%, this project examines the
role of promoter polymorphisms within endothelin system genes. Especially, we functionally analyse regulatory sequences which bind unidentified transcription
factors (i.e., a positive signal in EMSA but with unknown
matrix (consensus seqeunce) according to bioinformatics) using DNA affinity chromatography, mass spectrometry and reporter gene assay. In addition we have
genotyped promoter polymorphisms of several hundreds of patients with AD and respective controls in
cooperation with the German Kompetenznetz Demenzen, a nationwide network which includes 14 university
centers in the field of dementia research.
HEIKO FUNKE-KAISER – Pharmacology
Role of ECE-1 in Alzheimer´s disease.
Objectives: As shown by other groups, cerebral Ab40/ 42 content is significantly increased in ECE-1 knockout mice compared
to wild type animals. Consistently, induction of ECE-1 by transgenic overexpression of PKC strongly reduces plaque and Ab
deposit area. In humans, single nucleotide polymorphisms (SNPs) in the ECE-1b isoform-specific promoter, which were initially
described by our group in the context of arterial hypertension (Funke-Kaiser, H. et al. (2003) Hum. Mol. Genet.), affect prefrontal
ECE-1 mRNA expression and are associated with the likelihood of developing AD to a similar degree as the APOE2 allele.
205
Kai Kappert – Pharmacology
Head of the group
Priv.-Doz. Dr. med. Kai Kappert
Curriculum Vitae: After a three-year post-doctoral position at the Karolinska
In­stitute, Stockholm, Sweden, Kai Kappert, MD, Group Leader Molecular Pharmacology, joined the Center for Cardiovascular Research (CCR), Institute of Pharmacology, in 2006. He received the medical approbation (3rd state examination) and
an MD degree in 2003, based on experimental work at the German Heart Center
Berlin (DHZB). Educated as a medical doctor (Georg-August-University Göttingen;
Humboldt-University Berlin; University of California Los Angeles, USA; Inselspital Bern, Switzerland) with clinical
experience in the field of cardiology (University Clinic Cologne), his major focus of his research has been growth
factor signalling, including the impact of endogenous inhibitory protein tyrosine phosphatases, in vascular
cells and tissue remodelling. In 2009 he passed his habilitation in experimental pharmacology at the CharitéUniversity Medicine Berlin.
Members of the group
Students
Hackbusch, Daniel
Häßle, Paul
Krüger, Janine
Daniel Hackbusch
PhD student
Medical student
PhD student
Paul Häßle
Janine Krüger
207
Kai Kappert – Pharmacology
Summary
Protein phosphorylation on tyrosine residues represents an important cell-signaling mechanism,
controlled by the combined balanced actions of
tyrosine kinases and protein tyrosine phosphatases
(PTPs). Regulatory mechanisms of classical PTPs,
a cysteine-based subclass of the PTP superfamily
that exclusively dephosphorylates phospho-tyrosine
in proteins, include changes in expression, phosphorylation, and subcellular localization. Furthermore, a novel negative regulatory mechanism is the
reversible oxidation of the conserved catalytic-site
cysteine.
Differential regulation of PTPs has recently been
demonstrated in vascular tissue remodelling, diabetes, cell differentiation, and cancer development.
Thus, targeting PTPs might serve as a novel thera-
208
peutic approach altering tyrosine-signaling. Furthermore, accumulating data on the structural basis and
protein interaction of PTPs should pave the way for
considering PTPs as strategic therapeutic targets in
human diseases.
The research focus of our group is the unravelling of
the significance of PTPs on distinct regulatory levels,
such as post-transcriptional oxidation, expression,
and activity in various experimental organ-based
and animal models. This includes analyses of transient negative regulation of PTPs by oxidation under
conditions of ischemia/reperfusion, and the characterization of PTPs during cerebro-vascular positive
outward remodelling. An additional research focus is
to investigate the impact of PTPs in models of insulin
resistance. Thus, we expect to further identify and
establish PTPs as targets for treatment of cerebral
occlusive vascular disease and insulin resistance.
Kai Kappert – Pharmacology
Zusammenfassung
Die Phosphorylierung von Tyrosinresten in Proteinen
stellt einen wichtigen Mechanismus in der zellulären
Signaltransduktion dar, der durch die koordinierte
Aktivität von Tyrosinkinasen und Protein-TyrosinPhosphatasen (PTPs) reguliert wird. Zu den Regulationsmechanismen der klassischen PTPs, eine
Cystein-basierte Untergruppe der PTP-Superfamilie, die ausschließlich Phosphotyrosin in Proteinen dephosphoryliert, gehören Expressionsunterschiede, Veränderung der Phosphorylierung von
PTPs selbst, sowie die subzelluläre Lokalisation von
PTPs. Ein neuartiger negativer Regulationsmechanismus ist die reversible Oxidation des konservierten
Cysteinrestes im aktiven katalytischen Zentrum von
PTPs.
Die differentielle Regulierung von PTPs konnte kürzlich in Umbauprozessen von Gefäßen, beim Diabetes mellitus, in der Zelldifferenzierung und auch bei
der Tumorigenese nachgewiesen werden, so dass
PTPs als molekulare Strukturen für die gezielte Ver-
änderung der Tyrosin-vermittelten Signaltransduktion in Frage kommen. Darüber hinaus sollten die
zunehmenden Kenntnisse der strukturellen Basis
der Proteininteraktionen von PTPs den Weg für die
Nutzung von PTPs als Zielmoleküle in der humanen
Therapieforschung ebnen.
Der Forschungsschwerpunkt unserer Gruppe liegt
in der Aufklärung der Bedeutung von PTPs auf
unterschiedlichen regulatorischen Ebenen, wie die
der post-transskriptionellen Oxidation, der Expression und der Aktivität in unterschiedlichen Organund Tiermodellen. Dies beinhaltet die Analyse von
transienter negativer Regulation von PTPs unter
den Bedingungen von Ischämie/ Reperfusion und
die Charakterisierung von PTPs während positivem
zerebrovaskulären Outward-Remodelling. Ein weiterer Forschungsschwerpunkt ist die Untersuchung
der Bedeutung von PTPs in Insulinresistenzmodellen. Insgesamt verfolgen wir den Ansatz, PTPs als
Zielstrukturen im Rahmen der Behandlung von zerebralen Gefäßerkrankungen und der Insulinresistenz
zu etablieren.
209
Kai Kappert – Pharmacology
Research projects
1. Protein tyrosine phosphatases as negative regulators and
novel targets in collateral growth
Project leader
Coworkers
Funding
External Cooperations
Kai Kappert
Daniel Hackbusch
- DFG KA 1820/4-1
- Deutsche Stiftung für Herzforschung F/04/08
Ivo Buschmann, Nora Gatzke, André Dülsner (Department of Cardiology/ ) CCR
Markus Dagnell, Arne Östman (Department of Oncology-Pathology,
Cancer Centrum Karolinska, Karolinska Institute, Stockholm, Sweden)
Protein tyrosine phosphatases (PTPs) are dephosphorylating enzymes with primarily negative impact
on tyrosine signal transduction. This project will characterize the dynamic regulation and function of PTPs
during cerebral collateral growth/ arteriogenesis in a
model of 3-vessel occlusion. Furthermore, in a proofof-concept approach the role of PTPs on arteriogenesis
is being tested by pharmacological intervention using
PTP-antagonists. We hypothesize that PTP-inhibition
and thus disinhibition of tyrosine-phosphorylation in
receptor tyrosine kinases will lead to an increase of
vascular proliferation and consecutively to enhanced
cerebral blood flow. Taken together, the project aims
at demonstrating that PTPs represent novel targets
for treatment of cerebral occlusive vascular disease.
Schematic diagram of the potential
regulation of arteriogenesis by receptor
tyrosine kinase (RTK)-antagonizing
PTPs, and the therapeutic approach
of enhanced arteriogenesis through
inhibition of inhibitory PTPs.
210
Kai Kappert – Pharmacology
2. R
ole of protein tyrosine phosphatases in high-fat diet
induced insulin resistance
Project leader
Coworkers
Funding
External cooperations
Kai Kappert
Janine Krüger
Personal funding of Charité-University Medicine Berlin
Dissertation scholarship of Charité-University Medicine Berlin
German Diabetes Society
Christian Böhm, Christiane Sprang, Ulrich Kintscher (Institute of Pharmacology/ CCR)
Heike Meyborg, Philipp Stawowy (German Heart Center Berlin)
Diabetes and the metabolic syndrome are major risk
factors for the development and progression of cardiovascular disease, including atherosclerosis. On a
molecular level, obesity-linked insulin resistance has
been attributed to a post-receptor deficiency. Recent
data suggest protein tyrosine phosphatases (PTPs)
significantly impacting on insulin receptor signal transduction (see Figure). In this research project the tissuespecific regulation and function of PTPs will be ana-
lyzed in an animal model of high-fat diet induced insulin
resistance in vivo. Mice are being fed either low-fat
(10% kcal from fat) or high-fat (60% kcal from fat) diet
for 16 weeks with or without additional treatment with
PTP-inhibitors. In parallel, the precise characterization
of PTPs will be performed in insulin-sensitive cell in
vitro. Aim of the project is to establish PTPs as novel
targets in the treatment of insulin resistance.
Recent evidence suggests that protein
tyrosine phosphatases (PTPs) are impacting on the tyrosine status of the insulin
receptor, and thus downstream signaling.
211
Elena Kaschina – Pharmacology
Head of the group
Dr. med. Elena Kaschina
Curriculum Vitae: Elena Kaschina studied medicine at the St.-Petersburg State
I.P.Pavlov Medical University. After medical approbation she worked as an internist
at the hospital. 1995 she obtained MD degree based on the experimental work at
the Institute of Pharmacology, Medical Academy St. Petersburg. 1999 she received
a Grant from Otto Benecke Stiftung in order to continue research at the University
of Kiel. 2005 she built up her research group at the Institute of Pharmacology, the
Center for Cardiovascular Research (CCR).
From 2010 she is passing her habilitation in experimental pharmacology. Her research is focused on vascular
and cardiac remodelling and drug development.
Members of the group
Technicians
Sommerfeld, Manuela
Kemnitz, Ulrich Rudolf
BTA
BTA
Students
Akohov, Aleksej
Lauer, Dilyara
Slavic, Svetlana
Schrader, Felix
Medical student
PhD student
Medical student
Medical student
SvetlanaSlavic
ManuelaSommerfeld
ManuelaSommerfeld
SvetlanaSlavic
UlrichRudolfKemnitz
UlrichRudolfKemnitz
DilyaraLauer
DilyaraLauer
AleksejAkohov
AleksejAkohov
213
Elena Kaschina – Pharmacology
Summary
Pathophysiological mechanisms of aortic aneurysms and possible therapeutic implications
Abdominal aortic aneurysm (AAA) is a common
vascular disorder which causes significant mortality. The treatment of AAA is restricted to surgical
interventions. Determination of specific genes and
pathomechanisms relevant for the development of
aneurysms may identify target sites for potential pharmacologic interventions.
Aortic aneurysms are characterised by vascular
inflammation, apoptosis and the degradation of extracellular matrix. Our aneurysm research focuses on
the kallikrein-kinin- and renin-angiotensin systems,
which are known players in hemodynamic stress, a
Zusammenfassung
Pathophysiologische Mechanismen von Aor­
tenaneurysmen und mögliche therapeutische
Implikationen
Das Aneurysma der Aorta abdominalis (AAA) ist
eine weitverbreitete vaskuläre Erkrankung mit hoher
Sterblichkeitsrate. Bis heute existiert keine therapeutische Alternative zur chirurgischen Intervention. Die
genaue Bestimmung von spezifischen Genen und
Pathomechanismen, die für die Aneurysma-Entstehung bedeutsam sind, könnte Angriffsmöglichkeiten
für eine pharmakologische Therapie dieser Erkrankung identifizieren.
Die Entstehung von AAA ist durch Inflammation,
Apoptose und einen massiven Abbau der extrazellulären Matrix gekennzeichnet. Unsere Untersuchungen konzentrieren sich auf das Renin-Angiotensin-System (RAS), sowie auf das Kallikrein-KininSystem (KKS), die bereits von uns und anderen
214
causative factor for aneurysm formation, and also in
vascular wall homeostasis. Well established animal
models and primary cell cultures from aorta allow us
to evaluate important signalling cascades and investigate new therapeutic targets. Human abdominal
aortic tissue from patients undergoing surgery is also
studied, and candidate genes for aneurysm disease
are analysed. Research is focused on the role of different components of the renin-angiotensin system as
well as kininogen in vascular inflammation, proteolysis
and apoptosis as part of the aneurysmal disease.
The link of AAA with known risk factors for the
disease could help us in the understanding of the
pathomechanisms. Therefore, we are also interested
in vascular remodelling by obesity, insulin resistance
and uremia.
Gruppen als pathogenetisch äußerst wichtig für die
Aneurysma-Entstehung erkannt wurden. Diese beiden endokrinen Systeme betreffend weiß man inzwischen, dass Angiotensin II über den AT1-Rezeptor
eine fördernde, das KKS über Kininogen anderseits
eine hemmende Wirkung auf Krankheitsgeschehen
ausübt. Gut etablierte Tiermodelle und Primärzellkulturen aus Gewebe der Aorta sowie humanes abdominelles Aortengewebe erlauben uns, wichtige Signalkaskaden zu evaluieren und neue therapeutische
Strategien zu untersuchen.
Die aktuelle Forschung wird sich auf die Rolle des Kininogens sowie verschiedener Komponenten des RAS
für Inflammation, Proteolyse und Apoptose im Rahmen
der Pathogenesis von Aneurysmen konzentrieren.
Die Bedeutung des Adipositas und der Insulinresistenz sowie der Einfluss von Uremia auf Gefäßremodeling werden auch in verschiedenen experimentellen Modellen untersucht.
Elena Kaschina – Pharmacology
Research projects
1. H
igh molecular weight kininogen, a novel factor in the
regulation of matrix proteinases
Project leader
Coworkers
Funding
External cooperations
Elena Kaschina
Dilyara Lauer
Manuela Sommerfeld, Svetlana Slavic, Ulrich Rudolf Kemnitz
Bayer Schering Pharma
Christa Thöne-Reineke, CCR
Prof. Marjan Boerma, University of Arkansas, USA
Increased activity of matrix metalloproteinases
(MMPs), leading to degradation of extracellular matrix
components, is considered to play a crucial role in
the pathogenesis of cardiovascular diseases such as
heart failure and aneurysms. Thereby, a pivotal role
has been attributed to MMPs belonging to the subgroup of gelatinases, including MMP-2 (gelatinase A)
and MMP-9 (gelatinase B).
We previously reported that
kininogen-deficient rats are
predisposed
to
develop
abdominal aortic aneurysms.
Thereby, aneurysm formation
was associated with enhanced
elastolysis and increased expression of MMPs, thus
indicating a role for kininogens in the regulation of
MMPs. We investigated whether cleaved high molecular weight kininogen (cleaved HK) affects the regulation of MMPs in primary vascular smooth muscle cells
(VSMCs), cultured from the rat aorta. We found that
cleaved HK reduced in a concentration dependent
manner cytokine-induced release of both MMP-9 and
MMP-2 by VSMCs. Furthermore, cytokine-induced
MMP-9 mRNA expression was negatively regulated
by cleaved HK to almost the same extent. Determina-
tion of expression levels of the endogenous inhibitors
of MMPs, the TIMPs, revealed that TIMP-1 mRNA
expression, already increased as a result of cytokinestimulation, was further enhanced by cleaved HK.
Altogether, these findings indicate that the balance
between MMPs and TIMPs was shifted towards less
net MMP activity by cleaved HK.
Our current investigations are focused on the antioproteolytic effect of kininogen and its domains in primary
cardiomyocytes. We found that cleaved HK prevented
cytokine-induced release of MMP-9 and reduced
gelatinase/collagenase activity of cardiomyocytes.
In summary, high molecular weight kininogen is a
potential drug target for diseases with excessive
extracellular matrix turnover. Further research will be
focused on therapeutic effects of kininogen in the
models of heart failure and aneurysm formation.
(published in Biochemical Pharmacology, 2010)
215
Elena Kaschina – Pharmacology
2. Aneurysm formation in mild uremia
Project leader
Coworkers
Funding
External cooperations
Elena Kaschina
Aleksej Akohov, Manuela Sommerfeld, Ulrich Rudolf Kemnitz
internal resources
Harm Peters, CCR, Department of Nephrology, Charité Universitätsmedizin Berlin,
Dr. Stephanie Kraemer, Deutsche Institut für Ernährungsforschung, Berlin
Mild renal failure is known to be an independent risk renal functional parameters and serum pro-inflammafactor for cardiovascular disease. Mild uremia is fol- tory cytokines were investigated.
lowed by fluid overload, inflammation and production Uremia induced aortic dilatation. Histological analyof uremic toxins. All these factors may contribute to sis revealed an outward aortic remodelling, increased
vascular remodelling.
elastin fragmentation, cystic medial degradation, calThe aim of this study was to investigate the effects of cification of tunica media and inflammatory infiltrates
mild uremia on aneurysm size, as well as on matrix in the adventitia. After aneurysm induction, aortic
remodelling, apoptosis, proteolysis and inflamma- diameter was further increased in the NE group as
tion and their related gene- and protein expression compared to AAA rats without NE. Hydralazine treatpatterns in the rat. Investigation was performed in a ment significantly reduced blood pressure but did not
combined model of experimental uremia followed by influence aortic diameter.
elastase-induced aneurysm
formation. Mild uremia was
induced in rats by right uninephrectomy and removing
2/3 of the left kidney surgically (5/6 nephrectomy)
(NE). 4 weeks later aneurysm (AAA) was induced
via continuous infusion of
an isolated aortic segment
with elastase. One group of
NE animals was additionally treated with hydralazine.
Aortic diameter and heart
function were measured by
A: Ultrasound image of rat aorta. Arrows represent aortic diameter. B: Histological crossultrasound. Aortic tissues, sections of the rat abdominal aorta: Weigert’s stain, Sirius red-picric acid stain, x20, Lum
indicates luminal side of aorta; adv indicates adventitial side.
216
Elena Kaschina – Pharmacology
Protein expression of NF-kB was strongly (3-fold)
up-regulated in aortic tissues from NE rats. MMP2,
MMP9, cathepsin D and TGF-beta1 were significantly
increased after AAA and further up-regulated in the
NE/AAA group. Heart function was also negatively
affected by uremia: Ejection fraction and fractional
shortening were significantly decreased whereas left
ventricular volume was increased in the systole and
in the diastole.
Alltogether, we could demonstrate that mild uremia
induces aortic outward remodeling independently of
blood pressure elevation. Activation of NFkB by uremic
toxins may contribute to remodelling via inhibition of
elastin- and collagen gene transcription.
3. R
egulation of cardiac and vascular functions by selective
cannabinoid 1 receptor blocker rimonabant
Project leader
Coworkers
Funding
External cooperations
Elena Kaschina
Svetlana Slavic, Manuela Sommerfeld, Ulrich Rudolf Kemnitz
Sanofi-Aventis GmbH Deutschland
Ulrich Kintscher, CCR
PD Dr. med. Johannes Baulmann, University of Lübeck
The biological effects of endocannabinoids such as
anandamide, are mediated by specific G protein-coupled cannabinoid (CB) receptors. The CB1 receptor
is present in heart and vascular tissues, fat tissue and
liver. CB1 receptor blockade by a selective antagonist
rimonabant is known to be beneficial by obesity and
metabolic syndrome in humans.
Our study aimed to investigate whether cannabinoid-1
receptor blockade may result in heart protection by
metabolic syndrome and whether this protection is primary and not dependent from metabolic corrections.
We could show that rimonabant improves heart function in Obese Spontaneously Hypertensive Koletsky
Rats. In non-obese rats, the effects of CB1-blockade
were studied in the model of myocardial infarction
(MI), which was induced by permanent ligation of the
left coronary artery. Treatment with rimonabant was
started one week before MI and continued for 7 days or
6 weeks. Hemodynamic parameters were measured
via transthoracic Doppler echocardiography and intracardiac Samba catheter. Arterial function was studied using pulse wave analysis. Cytokines, apoptotic
and fibrotic markers were determined in heart tissue.
Rimonabant improved systolic left ventricular function, lowered left ventricular end-diastolic pressure
and increased heart contractility in the early and late
phase after myocardial infarction. Arterial function
parameters, such as pulse pressure, augmentation
217
Elena Kaschina – Pharmacology
pressure- and index, were decreased in the rimonabant group mostly 6 weeks after MI compared to vehicle. Analysis of locomotor behaviour demonstrated
continuously increasing activity of treated rats. Upregulated IL-1beta-expression and TGF-beta expression in postinfarcted myocardium was ameliorated by
rimonabant treatment at both time points, whereas
caspase 3 was down-regulated only 7 days after MI.
Thus, the cannabinoid-1 receptor antagonist rimonabant continually improves heart function in rats after
myocardial infarction. Reduced inflammation in the
heart and reduced pulse wave reflection may contribute to cardiac protection by rimonabant.
Since rimonabant improved vascular function by
decreasing arterial stiffness as well as by diminishing
concentration of hydroxyprolin in aorta, our research
will be further focused on the role of CB1 receptor in
vascular diseases.
Representative M-mode
echocardiograms (short
axis parasternal view) of
the LV from rats 6 weeks
after myocardial infarction treated with rimonabant (A) and vehicle (B).
218
Jun Li – Pharmacology
Head of the group
Dr. med. Jun Li
Curriculum Vitae: Jun Li initially studied medicine in China and subsequently at
the University of Kiel, where he earned his MD. He then pursued research in cellular immunology and molecular cardiology at the University of Mainz and the Free
University of Berlin. Since July 2002, he has been working as a group leader dealing
with adaptive protective mechanisms against ischemic injury in the brain and heart
at the Center for Cardiovascular Research and Institute of Pharmacology at the
Charité – Universitary Medicine in Berlin. In 2005, he won the Dieter-Klaus-Award for Hypertension Research
of the German Hypertension Society for his innovative work exploring the interactions of angiotensin AT2
receptor and T lymphocytes in the heart. A main focus of his current research is the elucidation of the adaptive protective mechanisms of cell biology and the impact of these cellular mechanisms on the outcomes of
cardiovascular disease.
Members of the group
Technicians
Timm, Melanie
Students
Adjemian, Sandy
Altarche-Xifró, Wassim Brinckmann, Marie Curato, Caterina Miteva, Kapka
Skorska, Anna
Slavic, Svetlana
Master student
PhD student
Medical student
PhD student
Master student
PhD student
Medical student (physician)
219
Jun Li – Pharmacology
Summary
Acute myocardial infarction (MI) leads to loss of
cardiomyocytes and impaired pump function of the
heart. When the remaining cardiomyocytes are unable to reconstitute with time, heart failure results. A
central question in the cardiac cell biology is what are
the cellular mechanisms that mediate the generation
and maintenance of cardiac performance. The focus
of our research is the adaptive interaction of cardiac
various cell populations including c-kit+ precursor
cells, T lymphocytes, and cardiomyocytes. A wealth
of information indicates that the renin-angiotensin
system and endogenous estrogen can interfere with
the cardiac injury/repair process after MI. Given the
recent evidence that both angiotensin AT2 receptor
and estrogen receptor a are involved in cardioprotective actions, we are particularly interested in the
elucidation of cellular mechanisms by which cells
of the periinfarct myocardium exert cardioprotection via the activation of angiotensin AT2 receptor or
estrogen receptor a.
Zusammenfassung
Angiotensin II, das Haupteffektorprotein des ReninAngiotensin-System (RAS), ist an der Entwicklung
von kardiovaskulären Erkrankungen beteiligt. Der
AT2-Rezeptor ist im fetalen Leben der dominierende
ATR-Subtyp. Später wird er nach Verletzungen bzw.
im Verlauf pathosphysiologischer Prozesse, wie zum
Beispiel Schämie im Herz bzw. Gehirn, exprimiert.
Dieses spezifische Expressionsmuster weist darauf
hin, dass der AT2-Rezeptor bei der Gewebsadaptation und -regeneration eine wichtige Rolle spielt.
In Vorarbeiten konnten wir zeigen, dass der AT2-
220
Rezeptor Antiinflammation, Differenzierung, sowie
Regeneration vermittelt. Weitere Forschungen
befassten sich mit der Rolle des AT2-Rezeptors
beim Entzündungs- und Regenerationsprozess
nach einem Herzinfarkt. Ein anderer Schwerpunkt
lag auf Untersuchungen zum Einfluss von Östrogen
auf das kardiale Remodeling nach Myokardinfarkt.
Insbesondere wird die Rolle von Östrogenrezeptoren, wie ERa und ERb auf die Selbsterneuerung und
Differenzierung von Stammzellen im Tiermodel mit
experimentellem Myokardinfarkt untersucht.
Jun Li – Pharmacology
Research projects
1. Angiotensin AT2 receptor in cardiac regeneration
Project leader
Coworkers
Funding
External cooperations
Jun Li
Wassim Altarche-Xifró, Caterina Curato, Svetlana Slavic, Melanie Timm,
Elena Kaschina, Aleksandra Grzesiak
- BMBF (BCRT-kick off grant and 1st. BCRT grant)
Institute of Cell Biology, University of Bern, Switzerland (Prof. H. Imboden);
Reference and Translation Center for Cardiac Stem Cell Therapy,
University of Rostock (Prof. Dr. G. Steinhoff)
The expression pattern of AT2 receptors with predominance during fetal life and upregulation under
pathological conditions during tissue injury/repair
points towards the fact that AT2 receptors may exert
an important action in injury/repair adaptive mechanisms, which may be entirely different from the known
effects of angiotensin II mediated through the AT1
receptor. Indeed, recent experimental evidence from
our group supports that angiotensin II, through its AT2
receptor subtype, promotes cellular differentiation and
tissue regeneration. In this project, we aimed to understand the adaptive cellular mechanisms underlying
angiotensin AT2 receptor-mediated cardioprotection.
First of all, we examined the
cellular regulation of cardiac
AT2 receptor in response to
acute myocardial infarction in
rats. Further, we isolated and
characterized
post-infarct
cardiac c-kit+AT2+ cell population. To explore a potential
role of post-infarct cardiac
c-kit+AT2+ cell population in
cardioprotection, we examined the in vitro effect of AT2
receptor activation on the performance of adult cardiomyocytes co-cultured with post-infarct cardiac
c-kit+AT2+ cells. To extrapolate the in vivo role of
post-infarct cardiac c-kit+AT2+ cell population, we
studied whether selective AT2 receptor activation may
enhance adaptive cellular regeneration, especially in
post-infarct cardiac c-kit+AT2+ cell population and
cardiomyocytes following acute ischemic injury in rats.
Thus far we have shown that cardiac c-kit+AT2+ cell
population exists and increases after acute ischemic
injury. AT2 receptor activation supports cardiomyocyte
performance, hence contributing to cardioprotection
via cardiac c-kit+AT2+ cell population.
Representative images of
stained cardiac free-floating
sections showed that specific
signals for AT2 receptor were
detected in accumulating
c-kit+ cells distributing in
the peri-infarct myocardium.
Arrows are pointing at the
c-kit+AT2+ cell clusters under
a Leica LSM confocal microscope. Scale bar, 10 mm
221
Jun Li – Pharmacology
2. Angiotensin AT2 receptor in cardiac inflammation
Project leader
Coworkers
Funding
External cooperations
Jun Li
Caterina Curato, Anna Skorska, Svetlana Slavic, Melanie Timm,
Kapka Miteva, Elena Kaschina
- EU EST- CARDIOVASC-(TP 3.2)
German Rheumatism Research Centre and Berlin-Brandenburg
Center for Regenerative Therapies (Dr. J. Dong, Prof. Dr. A. Thiel);
2nd Affliated Hospital of Zhejiang University, China (Prof. Dr. J. Wang);
Institute of Cell Biology, University of Bern, Switzerland (Prof. H. Imboden);
Reference and Translation Center for Cardiac Stem Cell Therapy, University of
Rostock (Prof. Dr. G. Steinhoff); Department of Cardiology,
Charité-Universitätsmedizin Berlin, Campus Virchow – Klinikum (Dr. S. von Haehling)
Emerging evidence suggests a cardioprotective role
of the angiotensin AT2R, albeit the underlying cellular
mechanisms are poorly understood. Here, we defined
the CD8+AT2R+ T cell population, which increased
following acute myocardial infarction (MI). Further,
we developed a method to selectively isolate these
cardiac CD8+AT2R+ T cells, which exhibited downregulated expression of pro-inflammatory cytokines
and noncytotoxicity to cardiomyocytes. AT2R activation engendered a significant increment in cardiac
222
CD8+AT2R+ T cells and IL-10 production, likely contributing to reduced infarct size and improved cardiac
function as shown recently by our group. In addition,
intramyocardial transplantation of CD8+AT2R+ T cells
(vs. CD8+AT2R-) led to reduced ischemic heart injury.
These findings advance our understanding of the functional role of AT2R in the heart and provide a novel
cellular mechanism via cardioprotective CD8+AT2R+
T cells.
Jun Li – Pharmacology
3. C
ardioprotective role of estrogen receptor a via c-kit+
precursor cells
Project leader
Coworkers
Funding
External cooperations
Jun Li
Marie Brinckmann, Svetlana Slavic, Anna Skorska, Wassim Altarche-Xifró,
Caterina Curato, Melanie Timm, Elena Kaschina, Aleksandra Grzesiak
- DFG (GK 754-III, TP7b)
- FERMENTATION – BIOTEC GmbH
Institute of Gender in Medicine and Center for Cardiovascular Research (CCR),
Charité University Medicine Berlin (Prof. Dr. V. Regitz-Zagrosek); Reference
and Translation Center for Cardiac Stem Cell Therapy, University of Rostock
(Prof. Dr. G. Steinhoff); Department of Cardiology, Charité-Universitätsmedizin Berlin,
Campus Virchow – Klinikum (Dr. S. von Haehling)
A growing body of clinical evidence demonstrates
that endogenous estrogen may protect cardiovascular health in both female and male patients. Given
the recent observations that estrogen exerts cardioprotective effects via augmented mobilization of
bone marrow-derived endothelial progenitor cells
and enhanced angiogenesis, it is also possible that
estrogen receptors may be involvoed in regulating
cardiac performance via c-kit+ precursor cells. To
understand the potential cellular mechanisms underlying estrogen-mediated cardioprotection, we examined
the regulation of estrogen receptor (ER) in post-infarct
cardiac c-kit+ cells and their functional relevance in
response to acute ischemic injury in male rats. The
current results have demonstrated that ERa is mainly
induced in post-infarct cardiac c-kit+ cells in response
to acute myocardial infarction in rats and that ERa
stimulation promotes the proliferation of undifferentiated myoblast cells. In addition, ERa activation supports survival of adult cardiomyocytes via post-infarct
cardiac c-kit+ cells which affords a direct explanation
for protective actions of ERa against cardiac injury.
223
Jun Li – Pharmacology
Awards
2009
224
PhD Student Award of the Berlin-Brandenburg
School for Regenerative Therapies Wassim Altarche-Xifró
Ulrike Steckelings – Pharmacology
Head of the group
Dr. med. Ulrike Steckelings
Curriculum Vitae: Ulrike Steckelings is a dermatologist and has longterm and
profound experience in pharmacological teaching and research.
She gained her MD at the University of Heidelberg and did her specialisation in dermatology at the Charité Universitary Medicine in Berlin. She worked as a postdoc at
the Institute for Pharmacology in Heidelberg, in the Department for Internal Medicine
at the University of Melbourne, Australia and in the Department for Cardiovascular
Research at Ciba Geigy in Basel, Switzerland. Her research foci are the AT2 receptor as well as the renin
angiotensin system of the skin. At the CCR she coordinates preclinical research of compound 21, an oral, non
peptidogenic AT2 agonist.
Members of the group
Scientists
Paulis, Ludovit
Santi, Francecsca
MD, PhD (EU Marie Curie Fellow)
MD (visiting scientist from
Bologna, Italy)
Villela, Daniel
Wardat, Sami
Wieland, Anja
PhD student (guest scientist,
University of Belo Horizonte, Brazil)
PhD student (student on joint
project with Prof. Kintscher’s group)
MD student
Technicians
Lucht, Kristin
Students
Becker, Sophie
Hosenfeld, Ann-K.
Horlbeck, Marie
Kummert, Maxi
Leonhardt, Julia
Namsolleck, Pawel
Reichenbach, Anne
Rompe, Franziska
Sadegh pour Saleh, H.
Schwengel, Katja
Ströder, Katja
Valero, Veronica
MD student
Diploma student
MD student
MD student
MD student
PhD student
MD student
PhD student
Diploma student
PhD student
PhD student
PhD student
225
Ulrike Steckelings – Pharmacology
Summary
The peptide hormone angiotensin II exerts its actions
mainly via two receptor subtypes, the AT1- and the
AT2-receptor. Actions mediated via the AT1-receptor
are well defined and mainly associated with cardiovascular physiology and disease. In contrast, AT2R
research has been hampered for almost 2 decades
by the lack of an in vivo active and stable AT2Ragonist. In November 2004, design and synthesis
of the first specific and selective non-peptide AT2Ragonist, Compound 21 (C21), have been published
by the group around Anders Hallberg from Uppsala
University. Our group started working with C21 in
mid 2005 first confirming in vitro the agonistic properties of this molecule and the specificity of its effects
Zusammenfassung
Das Peptidhormon Angiotensin II wirkt im wesentlichen über zwei Rezeptor-Subtypen, den AT1- und
den AT2-Rezeptor. Die über den AT1-Rezeptor vermittelten Wirkungen sind gut charakterisiert und
betreffen im Wesentlichen physiologische und
patho-physiologische Wirkungen im Bereich des
kardiovaskulären Systems. Die Erforschung des
AT2-Rezeptors dagegen wurde jahrelang dadurch
erschwert, dass kein in vivo stabiler und wirksamer
Agonist verfügbar war. Im November 2004 publizierte die Gruppe um Anders Hallberg von der Universität Uppsala Design und Synthese des ersten
spezifischen und selektiven, nicht peptidischen
AT2R-Agonisten, Compound 21. Unsere Gruppe
arbeitet seit Mitte 2005 mit dieser neuen Substanz,
zunächst mit in vitro Arbeiten, die die agonistische
und AT2R-spezifische Wirkweise von Compound 21
226
through the AT2R (Rompe et al., Hypertension 2010,
55: 924-931; see project 1 below for more details).
A first in vivo study in rats in cooperation with Elena
Kaschina (AG Unger, CCR) showed the in vivo activity of the compound in general and specifically a
favourable therapeutic effect of AT2R stimulation
after myocardial infarction resulting in improved scar
formation and cardiac function (Kaschina et al., Circulation 2008, 118: 2523-2532). At present, approx.
30 studies are going on – many with cooperations
inside and outside of CCR - testing AT2R function
and the therapeutic potential of AT2R-stimulation in
a broad spectrum of experimental models mainly
covering cardiovascular, chronic inflammatory, neurological and dermatological diseases.
bestätigten (Rompe et al., Hypertension 2010, 55:
924-931; für weitere Details siehe Projekt 1 unten).
Eine erste in vivo Studie in Kooperation mit Elena
Kaschina (AG Unger, CCR) belegte nicht nur prinzipiell die in vivo Wirksamkeit von C21, sondern zeigte
auch im Speziellen einen günstigen, therapeutischen
Effekt auf die Narbenbildung und die Herzfunktion
im infarzierten Herzen in Ratten. (Kaschina et al.,
Circulation 2008, 118: 2523-2532). Zur Zeit werden
ca. 30 Projekte unter Verwendung von Compound
21 durchgeführt - viele davon mit Kooperationen
innerhalb und außerhalb des CCR – in denen die
Funktion des AT2R sowie das therapeutische Potential einer AT2R-Stimulation in einem breiten Spektrum experimenteller Modelle getestet werden, wobei
der Schwerpunkt auf kardiovaskulären, chronisch
inflammatorischen, neurologischen und dermatologischen Erkrankungen liegt.
Ulrike Steckelings – Pharmacology
Research projects
This group performs a whole series of projects – either at CCR or in cooperation with external partners - studying
direct AT2-receptor stimulation with the novel AT2-receptor agonist Compound 21 (C21) in various in vivo and
in vitro models and testing AT2-receptor stimulation as therapeutic approach for a range of potential indications
focusing on cardiovascular, chronic-inflammatory and neurological diseases.
In the following, three of these studies are described in more detail:
1. A
T2-receptor stimulation as anti-inflammatory
therapeutic approach
Project leader
Coworkers
Funding
External cooperations
Ulrike Steckelings
Franziska Rompe, Pawel Namsolleck, Kristin Lucht
internal resources
Prof. Wolf-Hagen Schunck, MDC Berlin, Prof. Michael Bader, MDC Berlin,
Dr. Metin Artuc, Dept. of Dermatology, Charité, Prof. Anders Hallberg, Uppsala
University, Sweden, Prof. Björn Dahlöf, Göteborg University, Sweden
Angiotensin AT2-receptors can be regarded as an
endogenous repair system because the AT2-receptor
is upregulated in tissue damage and mediates tissueprotection. A potential therapeutic utilisation of this
system has only recently come within reach through
synthesis of the first selective, orally active, non-peptide AT2-receptor agonist, Compound 21. This study
tested AT2-receptor stimulation by Compound 21
(C21) as a potential future therapeutic approach for the
inhibition of pro-inflammatory cytokines and of NF-kB.
Using primary human and murine dermal fibroblasts,
we found that C21 dose-dependently (1nM-1µM)
reduced tumor-necrosis-factor (TNF)a-induced interleukin-6 levels. AT2-receptor specificity was controlled for by inhibition with the AT2-receptor antagonist,
PD123319, and by absence of effects in AT2-receptor
deficient cells. AT2-receptor coupled signalling leading to reduced interleukin-6 levels involved inhibition
of NF-kB, activation of protein-phosphatases, and
synthesis of epoxyeicosatrienoic acid (EET) – the latter being a signalling pathway described for the first
time for AT2R-mediated anti-inflammation. Compound
21 also reduced monocyte-chemoattractant-protein
(MCP)-1 and TNFa levels in vitro and in bleomycininduced toxic cutaneous inflammation in vivo.
Since inhibition of NF-kB activity is a mechanisms
of action shared by the AT2R and glucocorticoids,
we compared the efficiency of equal doses of C21
and hydrocortisone with respect to Inhibition of interleukin-6 promoter activity. We found that inhibition of
promoter activity by C21 was comparable in strength
to inhibition by hydrocortisone.
227
Ulrike Steckelings – Pharmacology
Data of this study suggest that pharmacological AT2-receptor stimulation may be an orally applicable future therapeutic approach
in pathological settings requiring the reduction of interleukin-6 or inhibition of NF-kB.
(published 2010: Hypertension 55: 924-931)
228
Ulrike Steckelings – Pharmacology
2. AT2-receptor stimulation in spinal cord injury
Project leader
Coworkers
Funding
External cooperations
Ulrike Steckelings
Pawel Namsolleck, Katja Schwengel, Christa Thöne-Reineke, Kristin Lucht
- Vicore Pharma
Prof. Sven Hendrix, Hasselt University, Belgium
Francesco Boato, PhD student, Dept. of Neuroanatomy, Charité
Prof. Anders Hallberg, Uppsala University, Sweden
Prof. Björn Dahlöf, Göteborg University, Sweden
One of the first physiological effects ever described for
the AT2R was neuroprotection. In this study we test
pharmacological AT2R-stimulation as a therapeutic
approach in spinal cord injury in mice using the novel
non-peptide AT2R-agonist, Compound 21 (C21).
Complementary experiments in primary neurons and
organotypic cultures serve to identify underlying proregenerative mechanisms. Functional recovery and
plasticity of corticospinal tract (CST) fibers following
spinal cord injury (SCI) by compression in mice is monitored after application of C21 (0.3 mg/kg/day i.p.) or
vehicle for 4 weeks. Organotypic co-culture of GFPpositive entorhinal cortices with hippocampal target
tissue serves to evaluate the impact of C21 (1µM) on
reinnervation. Neuronal differentiation, apoptosis and
expression of neurotrophins are investigated in primary murine neuronal cells.
Preliminary data indeed indicate that AT2-receptor
stimulation improves functional recovery in experimental spinal cord injury through promotion of axonal
outgrowth and through neuroprotective and antiapoptotic mechanisms.
229
Ulrike Steckelings – Pharmacology
3. AT2-receptor stimulation in experimental stroke
Project leader
Coworkers
Funding
External cooperations
Ulrike Steckelings
Katja Schwengel, Pawel Namsolleck, Christa Thöne-Reineke
Kristin Lucht
- Vicore Pharma
Prof. Masatsugu Horiuchi, Ehime University, Japan
Francesco Boato, PhD student, Dept. of Neuroanatomy, Charité
Prof. Anders Hallberg, Uppsala University, Sweden
Prof. Björn Dahlöf, Göteborg University, Sweden
The aim of this study is to investigate the effect of direct
AT2-receptor (AT2R) stimulation with the novel specific
and selective non-peptide AT2R agonist Compound
21 (C21) on infarct size, survival and neurological outcome after middle cerebral artery occlusion (MCAO)
in mice. For this purpose, C57/BL-6 or AT2R knockout mice (on C57/BL-6 background) are forwarded to
MCAO for 30 minutes followed by reperfusion. Starting
45 minutes after MCAO, mice are treated daily with
either vehicle (0,9% NaCl i.p.) or C21 (0,03mg/kg i.p.)
for a period of 4 days. Garcia neurological score is
230
performed daily to assess the severity of neurological
deficits. Infarct volumes are measured in vivo 96 hours
post-stroke by MRI. To get a first impression about
potential molecular mechanisms of AT2R stimulation
in stroke, mRNA-levels of neurotrophins and markers
for neuronal sprouting are measured in brain samples
by quantitative RT-PCR.
Preliminary results indicate that C21 improves neurological outcome and survival without affecting blood
pressure.
Ludovit Paulis – Pharmacology
Head of the group
Dr. med. Ludovit Paulis, PhD.
Curriculum Vitae: Ludovit Paulis, MD, PhD is currently holder of an EU Fellowship (Marie Curie) at the CCR under the supervision of Ulrike Steckelings and Prof.
Thomas Unger.
Dr. Paulis graduated in biomedical physics in 2004 and in general medicine 2006
at the University in Bratislava. He undertook his postgraduate studies in Prague,
Berlin and Montpellier and again in Bratislava, where he received his PhD in 2008.
He continued his post-doctoral research in Montpellier and later in Berlin.
Members of the group
Students
Becker, Sophie
Medical student
231
Ludovit Paulis – Pharmacology
Summary
The group represents a young researcher team in
the frame of the Steckelings workgroup. The main
interest of the group is directed towards the prevention and modulation of pathological cardiovascular
conditions, in particular arterial hypertension, myocardial fibrosis and arterial remodeling. The targetorgan damage is investigated in several animal
models, including L-NAME-induced hypertension
or experimental autoimmune myocarditis.
The main project of this group will investigate the
effects of the AT2 receptor agonist, compound 21,
and the pineal hormone, melatonin, on target-organ
damage in experimental hypertension induced by
NO-synthase inhibition in rats. This project is supported by the 7th EU FP project “COME-in-CARE”.
The aim is to observe, whether and to what extent
the investigated substances are able to prevent the
development of target-organ damage. Furthermore,
the aim is to asses, whether their effect can be additive and what mechanisms might contribute to this
expected protection. Special attention is devoted
to the regulation of collagen turn-over and immune
modulating effects of the AT2 receptor agonist.
Zusammenfassung
Die Arbeitsgruppe ist eine Nachwuchswissenschaftlergruppe innerhalb der AG Steckelings, mit
der sie eng zusammenarbeitet. Das Hauptinteresse
der Gruppe richtet sich auf die Prävention und das
Modulieren von pathologischen Veränderungen
im Herzkreislaufsystem wie arterieller Hypertonie,
Herzfibrose und arteriellem Remodeling. Die Organschäden werden an verschiedenen Tiermodellen
untersucht wie L-NAME induzierte Hypertonie,oder
experimentelle Autoimmunmyokarditis.
Im Hauptprojekt der Gruppe werden die Effekte des
AT2 Rezeptor Agonisten Compound 21 und des
Pinealhormons Melatonin auf Endorganschaden bei
experimenteller Hypertonie untersucht, wobei die
232
Hypertonie durch eine Inhibierung der NO-Synthase
in der Ratte induziert wird. Dieses Projekt wird durch
das EU 7th FP Projekt „COME-in-CARE“ gefördert.
Ziel dieses Projektes ist es, zu beobachten, ob und
im welchem Ausmaß die genannten Substanzen
die Entstehung der Endorganschäden verhindern
können. Darüber hinaus soll festgestellt werden, ob
die Effekte dieser zwei Substanzen additiv sind und
welche zellulären Mechanismen den Wirkungen der
Substanzen zugrunde liegen. Besondere Aufmerksamkeit wird der Regulation des Kollagenmetabolismus und den immunmodulierenden Eigenschaften
des AT2 Agonisten gewidmet.
Ludovit Paulis – Pharmacology
Research project
COmpound 21 and MElatonin in CArdiovascular REmodeling
( COME IN CARE )
Project leader
Coworkers
Funding
External cooperations
Ludovit Paulis, Thomas Unger, Ulrike M. Steckelings
Sophie Becker
EU: 7. FP-PIEF-237834
Dr. Johannes Baulmann, Medizinische Klinik und Poliklinik I,
Universitätsklinikum Würzburg, Würzburg, Prof. Fedor Simko,
Faculty of Medicine, Comenius University, Bratislava
The most effective current therapeutic strategies for
the treatment of hypertension including the use of
ACE inhibitors, AT1 receptor blockers (and partially
also β-blockers and renin inhibitors) are aimed to
reduce the deleterious stimulation of AT1 receptors.
However, only the recent discovery of the non-peptide
AT2 receptor agonist, compound 21, unleashed the
possibility of the investigation of the role of the AT2
receptor and whether its stimulation could counteract
the undesired effects by the AT1 receptor. Melatonin
is a pineal hormone with antioxidant, blood pressure
lowering and cardioprotective effects, but the mechanisms of these protective actions are not completely
elucidated.
In this project, adult male Wistar rats are made
hypertensive and NO deficient by administration of
an NO-synthase inhibitor, L-NAME. The subsequent
hypertension is associated with target-organ damage
including cardiac hypertrophy, fibrosis, renal damage
and arterial remodelling. These alterations are followed
by echocardiographic, hemodynamic and biochemical means. The aim is to observe, whether and to what
extent the investigated substances are able to prevent
the development of target-organ damage. Furthermore, the aim is to asses, whether their effect can be
additive and what mechanisms might contribute to this
expected protection.
The preliminary results suggest that while melatonin shows antifibrotic effects in the left ventricle,
compound 21 prevents arterial remodelling and the
increase in arterial stiffness, which is an independent cardiovascular risk factor. Further experiments are
performed to tackle the anti-stiffening mechanisms of
compound 21 with special attention to the regulation
of collagen turn-over and immune modulating effects
of the AT2 receptor agonist.
233
Pharmacology
Thomas Unger’s Group
Publications 2006-2010
Sandmann S, Li J, Fritzenkoetter C, Spormann J, Tiede K,
Fischer JW, Unger T (2006): Differential effects of olmesartan
and ramipril on inflammatory response after myocardial infarction in rats. Blood Press 15: 116-128
Schefe JH, Lehmann KE, Buschmann IR, Unger T, FunkeKaiser H (2006): Quantitative real-time RT-PCR data analysis: current concepts and the novel “gene expression’s C (T)
difference” formula. J Mol Med 84: 901-910
Schefe JH, Menk M, Reinemund J, Effertz K, Hobbs RM,
Pandolfi PP, Ruiz P, Unger T, Funke-Kaiser H. (2006): A
Novel Signal Transduction Cascade Involving Direct Physical
Interaction of the Renin/Prorenin Receptor With the Transcription Factor Promyelocytic Zinc Finger Protein.
Circ Res 99: 1355-1366
Schupp M, Lee LD, Frost N, Umbreen S, Schmidt B, Unger
T, Kintscher U (2006): Regulation of peroxisome proliferatoractivated receptor gamma activity by losartan metabolites.
Hypertension 47: 586-589
Thoene-Reineke C, Kalk P, Dorn M, Klaus S, Simon K, Pfab T,
Godes M, Persson P, Unger T, Hocher B (2006): High-protein
nutrition during pregnancy and lactation programs blood
pressure, food efficiency, and body weight of the offspring in
a sex-dependent manner. Am J Physiol Regul Integr Comp
Physiol 291: R1025-1030
Rutschow S, Li J, Schultheiss HP, Pauschinger M (2006):
Myocardial proteases and matrix remodeling in inflammatory
heart disease. Cardiovasc Res 69: 646-656
Clemenz M, Kintscher U, Unger T (2006): The metabolic
syndrome: cluster with a self-fulfilling loop? J Hypertens 24:
257-258
Kappert K, Caglayan E, Huntgeburth M, Bäumer AT, Sparwel
J, Uebel M, Rosenkranz S (2006): 17-beta estradiol attenuates PDGF signaling in vascular smooth muscle cells at the
post-receptor level. Am J Physiol Heart Circ Physiol 290:
H538-46
Xia QG, Reinecke A, Dorenkamp M, Daemen MJ, Simon R,
Unger T (2006): Effects of endothelin ET(A) receptor blocker
LU 135252 on cardiac remodeling and survival in a hypertensive rat model of chronic heart failure. Acta Pharmacol Sin
27: 1417-1422
Kappert K*, Leppänen O*, Paulsson J, Furuhashi M, Carlsson MA, Heldin CH, Fätkenheuer G, Rosenkranz S, Östman
A (2006): Highly Active Antiretroviral Therapy Attenuates ReEndothelialization and Alters Neointima Formation in the Rat
Carotid Artery After Balloon Injury. J Acquir Immune Defic
Syndr 43: 383-921 (*: contributed equally)
Hoheisel U, Unger T, Mense S (2007): Sensitization of rat
dorsal horn neurons by NGF-induced subthreshold potentials
and low-frequency activation. A study employing intracellular
recordings in vivo. Brain Res 1169: 34-43
Kappert K, Sparwel J, Sandin Å, Seiler A, Siebolts U, Leppänen O, Rosenkranz S, Östman A (2006):
Antioxidants relieve phosphatase inhibition and reduce PDGF
signaling in cultured VSMCs and in restenosis Arterioscler
Thromb Vasc Biol 26: 2644-51
Schupp M, Kintscher U, Fielitz J, Thomas J, Pregla R, Hetzer
R, Unger T, Regitz-Zagrosek V (2006): Cardiac PPARalpha
expression in patients with dilated cardiomyopathy. Eur J
Heart Fail 8: 290-294
Pechanova O, Matuskova J, Capikova D, Jendekova L, Paulis
L, Simko F (2006): Effect of spironolactone and captopril on
nitric oxide and S-nitrosothiol formation in kidney of L-NAMEtreated rats. Kidney Int 70: 170–176
234
Kappert K, Paulsson J, Sparwel J, Leppänen O, Hellberg C,
Östman A, Micke P (2007): Dynamic changes in the expression of DEP-1 and other PDGF receptor-antagonizing PTPs
during onset and termination of neointima formation
FASEB J 21: 523-34
Weibrecht I, Böhmer SA, Dagnell M, Kappert K, Östman
A, Böhmer FD (2007): Oxidation sensitivity of the catalytic
cysteine of the protein-tyrosine phosphatases SHP-1 and
SHP-2.Free Radic Biol Med 43: 100-10
Micke P*, Kappert K*, Ohshima M, Sundquist C, Scheidl
S, Lindahl P, Heldin CH, Ponten F, Östman A (2007): In situ
identification of genes regulated specifically in fibroblasts of
human basal cell carcinoma. J Invest Dermatol 27: 1516231 (*: contributed equally)
Pharmacology
Pechanova O, Zicha J, Paulis L, Zenebe W, Dobesova Z,
Kojsova S, Jendekova L, Sladkova M, Dovinova I, Simko F,
Kunes J (2007): The effect of N-acetylcysteine and melatonin
in adult spontaneously hypertensive rats with established
hypertension. Eur J Pharmacol 561: 129-136
Paulis L, Liskova S, Pinterova M, Dobesova Z, Kunes J, Zicha
J (2007): Nifedipine-sensitive noradrenergic vasoconstriction
is enhanced in spontaneously hypertensive rats: the influence of chronic captopril treatment. Acta Physiol (Oxf) 191:
255-266
Paulis L, Važan R, Simko F, Pecháňová O, Styk J, Babál P,
Janega P (2007): Morphological Alterations and NO-Synthase
Expression in the Heart after Continuous Light Exposure of
Rats.Physiol Res 56: S71-S76
Simko F, Matuskova J, Luptak I, Pincikova T, Krajcirovicova
K, Stvrtina S, Pomsar J, Pelouch V, Paulis L, Pechanova O
(2007): Spironolactone Differently Influences Remodeling of
the Left Ventricle and Aorta in L-NAME-Induced Hypertension.
Physiol Res 56: S25-S32
Simko F, Potacova A, Pelouch V, Paulis L, Matuskova J,
Krajcirovicova K, Pechanova O, Adamcova M (2007): Spontaneous, L-Arginine-Induced and Spironolactone-Induced
Regression of Protein Remodeling of the Left Ventricle in
L-NAME-Induced Hypertension. Physiol Res 56: S55-S62
Vazan R, Janega P, Hojna S, Zicha J, Simko F, Pechanova O,
Styk J, Paulis L (2007): The Effect of Continuous Light Exposure of Rats on Cardiac Response to Ischemia-Reperfusion
and NO-Synthase Activity. Physiol Res 56: S63-S69
Scholze J, Grimm E, Herrmann D, Unger T, Kintscher U
(2007): Optimal treatment of obesity-related hypertension: the
Hypertension-Obesity-Sibutramine (HOS) study. Circulation
115: 1991-1998
Slowinski T, Kalk, P, Christian, M, Schmager, F, Relle, K.,
Godes M, Funke-Kaiser H, Neumayer H-H, Bauer C,
Theuring F, Hocher B (2007): Cell-type specific interaction
of endothelin- and nitric oxide system - pattern of preproET-1 expression in kidneys of L-NAME treated prepro ET-1
promoter-lacZ-transgenic mice. J Physiol 581: 1173-1181
Unger T, Stoppelhaar M (2007): Rationale for double reninangiotensin-aldosterone system blockade. Am J Cardiol
100: 25J-31J
Westermann D, Rutschow S, Jaeger S, Linderer A, Anker S,
Riad A, Unger T, Schultheiss HP, Pauschinger M, Tschoepe
C (2007): Contributions of inflammation and cardiac matrix
metalloproteinase activity to cardiac failure in diabetic cardiomyopathy: the role of angiotensin type 1 receptor antagonism.
Diabetes 56: 641-646
Li J, Leschka S, Rutschow S, Schwimmbeck PL, Husmann L,
Noutsias M, Westermann D, Poller W, Zeichhardt H, Klingel K,
Tschöpe C, Schultheiss HP, Pauschinger M (2007): Immunomodulation by interleukin-4 suppresses matrix metalloproteinases and improves cardiac function in murine myocarditis.
Eur J Pharmacol 554: 60-68
Bakris G, Boehm M, Dagenais G, Diener HC, Fujita T, Gorelick
P, Kjeldsen SE, Laakso M, Mancia G, Pitt B, Sharma A, Sleight
P, Teo K, Unger T, Weber M, Williams B, Zannad F (2008): Cardiovascular protection for all individuals at high risk: evidencebased best practice. Clin Res Cardiol 97: 713-725
Kappert K, Meyborg H, Clemenz M, Graf K, Fleck E,
Kintscher U, Stawowy P (2008): Insulin facilitates monocyte
migration: a possible link to tissue inflammation in insulinresistance. Biochem Biophys Res Commun 365: 503-8
Bäumer AT, Ten Freyhaus H, Sauer H, Wartenberg M,
Kappert K, Schnabel P, Konkol C, Hescheler J, Vantler M,
Rosenkranz S (2008): Pi3 kinase-dependent membrane
recruitment of rac-1 and p47phox is critical for alpha PDGF
receptor-induced production of reactive oxygen species. J
Biol Chem 283: 7864-76
Tabet F, Schiffrin EL, Callera G, Yao G, Östman A, Kappert
K, Tonks NK, Touyz RM (2008): Redox-sensitive Signaling by
Angiotensin II Involves Oxidative Inactivation and Blunted Phosphorylation of Protein Tyrosine Phosphatase SHP-2 in Vascular
Smooth Muscle Cells from SHR. Circ Res 103: 149-158
Clemenz M, Frost N, Schupp M, Caron S, Foryst-Ludwig A,
Boehm C, Hartge M, Gust R, Staels B, Unger T, Kintscher U
(2008): Liver-specific peroxisome proliferator-activated receptor alpha target gene regulation by the angiotensin type 1
receptor blocker telmisartan. Diabetes 57: 1405-1413
235
Pharmacology
Foryst-Ludwig A, Clemenz M, Hohmann S, Hartge M, Sprang
C, Frost N, Krikov M, Bhanot S, Barros R, Morani A, Gustafsson JA, Unger T, Kintscher U (2008): Metabolic actions of
estrogen receptor beta (ERbeta) are mediated by a negative
cross-talk with PPARgamma. PLoS Genet 4: 1-16
Kaschina E, Schrader F, Sommerfeld M, Kemnitz UR, Grzesiak A, Krikov M, Unger T (2008): Telmisartan prevents aneurysm progression in the rat by inhibiting proteolysis, apoptosis
and inflammation. J Hypertens 26: 2361-2373
Kaschina E, Grzesiak A, Li J, Foryst-Ludwig A, Timm M,
Rompe F, Sommerfeld M, Kemnitz UR, Curato C, Namsolleck
P, Tschoepe C, Hallberg A, Alterman M, Hucko T, Paetsch I,
Dietrich T, Schnackenburg B, Graf K, Dahloef B, Kintscher U,
Unger T, c (2008): Angiotensin II type 2 receptor stimulation:
a novel option of therapeutic interference with the reninangiotensin system in myocardial infarction? Circulation 118:
2523-2532
Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz
M, Wabitsch M, Fischer-Posovszky P, Barth TF, Dragun D,
Skurk T, Hauner H, Blueher M, Unger T, Wolf AM, Knippschild
U, Hombach V, Marx N (2008): T-lymphocyte infiltration in
visceral adipose tissue: a primary event in adipose tissue
inflammation and the development of obesity-mediated insulin
resistance. Arterioscler Thromb Vasc Biol 28: 1304-1310
Krikov M, Thoene-Reineke C, Muller S, Villringer A, Unger T
(2008): Candesartan but not ramipril pretreatment improves
outcome after stroke and stimulates neurotrophin BNDF/TrkB
system in rats. J Hypertens 26: 544-552
Lueders S, Schrader J, Berger J, Unger T, Zidek W, Boehm
M, Middeke M, Motz W, Luebcke C, Gansz A, Brokamp L,
Schmieder RE, Trenkwalder P, Haller H, Dominiak P (2008):
The PHARAO study: prevention of hypertension with the
angiotensin-converting enzyme inhibitor ramipril in patients
with high-normal blood pressure: a prospective, randomized,
controlled prevention trial of the German Hypertension
League. J Hypertens 26: 1487-1496
The ONTARGET Investigators (2008): Telmisartan, ramipril,
or both in patients at high risk for vascular events. N Engl J
Med 358: 1547-1559
236
Van Linthout S, Spillmann F, Riad A, Trimpert C, Lievens J,
Meloni M, Escher F, Filenberg E, Demir O, Li J, Shakibaei M,
Schimke I, Staudt A, Felix SB, Schultheiss HP, De Geest B,
Tschöpe C (2008): Human apolipoprotein A-I gene transfer
reduces the development of experimental diabetic cardiomyopathy. Circulation 117: 1563-1573
Schmerbach K, Schefe JH, Krikov M, Muller S, Villringer A,
Kintscher U, Unger T, Thoene-Reineke C (2008): Comparison
between single and combined treatment with candesartan
and pioglitazone following transient focal ischemia in rat brain.
Brain Res 1208: 225-233
Schefe JH, Neumann C, Goebel M, Danser J, Kirsch S, Gust
R, Kintscher U, Unger T, Funke-Kaiser H (2008): Prorenin
engages the (pro)renin receptor like renin and both ligand
activities are unopposed by aliskiren. J Hypertens 26: 17871794
Paulis L, Zicha J, Kunes J, Hojna S, Behuliak M, Celec P,
Kojsova S, Pechanova O, Simko F (2008): Regression of
L-NAME-induced hypertension: The role of NO and endothelium-derived constricting factor. Hypertens Res 31: 793-804
Paulis L, Matuskova J, Adamcova M, Pelouch V, Simko J,
Krajcirovicova K, Potacova A, Hulin I, Janega P, Pechanova O,
Simko F (2008): Regression of left ventricular hypertrophy and
aortic remodelling in NO-deficient hypertensive rats: effect of
l-arginine and spironolactone. Acta Physiol (Oxf) 194: 45-55
Telmisartan Randomised AssessmeNt Study in ACE iNtolerant subjects with cardiovascular Disease (TRANSCEND)
Investigators (2008): Effects of the angiotensin-receptor
blocker telmisartan on cardiovascular events in high-risk
patients intolerant to angiotensin-converting enzyme inhibitors: a randomised controlled trial. Lancet 372: 1174-1183
Thoene-Reineke C, Neumann C, Namsolleck P, Schmerbach
K, Krikov M, Schefe JH, Lucht K, Hoertnagl H, Godes M,
Muller S, Rumschuessel K, Funke-Kaiser H, Villringer A,
Steckelings UM, Unger T (2008): The beta-lactam antibiotic,
ceftriaxone, dramatically improves survival, increases glutamate uptake and induces neurotrophins in stroke. J Hyper­
tens 26: 2426-2435
Pharmacology
Westermann D, Mersmann J, Melchior A, Freudenberger T,
Petrik C, Schaefer L, Lullmann-Rauch R, Lettau O, Jacoby C,
Schrader J, Brand-Herrmann SM, Young MF, Schultheiss HP,
Levkau B, Baba HA, Unger T, Zacharowski K, Tschoepe C,
Fischer JW (2008): Biglycan is required for adaptive remodeling after myocardial infarction. Circulation 117: 1269-1276
Altarche-Xifro W, Curato C, Kaschina E, Grzesiak A, Slavic
S, Dong J, Kappert K, Steckelings UM, Imboden H,
Unger T, Li J (2009): Cardiac c-kit+AT2+ Cell Population is
Increased in Response to Ischemic Injury and Supports Cardiomyocyte Performance. Stem Cells 27: 2488-2497
Brinckmann M, Kaschina E, Altarche-Xifro W, Curato C,
Timm M, Grzesiak A, Dong J, Kappert K, Kintscher U, Unger
T, Li J (2009): Estrogen receptor alpha supports cardiomyocytes indirectly through post-infarct cardiac c-kit+ cells. J Mol
Cell Cardiol 47: 66-75
Goebel M, Staels B, Unger T, Kintscher U, Gust R (2009):
Characterization of new PPARgamma agonists: benzimidazole derivatives - the importance of position 2. ChemMed­
Chem 4: 1136-42
Paulis L, Pechanova O, Zicha J, Krajcirovicova K, Barta A,
Pelouch V, Adamcova M, Simko F (2009): Melatonin prevents
fibrosis but not hypertrophy development in the left ventricle of NG-nitro-L-arginine-methyl ester hypertensive rats. J
Hypertens 27: S11-S16
Simko F, Pechanova O, Pelouch V, Krajcirovicova K, Mullerova
M, Bednarova K, Adamcova M, Paulis L (2009):Effect of
melatonin, captopril, spironolactone and simvastatin on blood
pressure and left ventricular remodelling in spontaneously
hypertensive rats. J Hypertens 27: S5-S10
Vrankova S, Jendekova L, Paulis L, Sladkova M, Simko F,
Pechanova O, (2009): Comparison of the effects of indapamide and captopril on the development of spontaneous
hypertension. J Hypertens 27: S42-S46
Benova M, Stebelova K, Paulis L, Simko F, Zeman M (2009):
Effect of L-NAME-induced hypertension on melatonin receptors and melatonin levels in the pineal gland and the peripheral organs of rats. Hypertens Res 32: 242-247
Sparwel J, Vantler M, Caglayan E, Kappert K, Fries JW, Dietrich
H, Böhm M, Erdmann E, Rosenkranz S (2009):Differential effects
of red and white wines on inhibition of the PDGF receptor:
Impact of the mash fermentation. Cardiovasc Res 81: 758-70
Kappert K, Tsuprykov O, Kaufmann J, Fritzsche J, Ott I,
Goebel M, Bahr IN, Hassle PL, Gust R, Fleck E, Unger T,
Stawowy P, Kintscher U (2009): Chronic Treatment With
Losartan Results in Sufficient Serum Levels of the Metabolite
EXP3179 for PPAR{gamma} Activation. Hypertension 54:
738-743
Kappert K, Meyborg H, Baumann B, Furundzija V, Kaufmann
J, Graf K, Stibenz D, Fleck E, Stawowy P (2009): Integrin
cleavage facilitates cell surface-associated proteolysis
required for vascular smooth muscle cell invasion. Int J Bio­
chem Cell Biol 41: 1511-7
Kaschina E, Scholz H, Steckelings UM, Sommerfeld M,
Kemnitz UR, Artuc M, Schmidt S, Unger T (2009): Transition
from atherosclerosis to aortic aneurysm in humans coincides
with an increased expression of RAS components. Athero­
sclerosis 205: 396-403
Ohshima M, Yamaguchi Y, Kappert K, Micke P, Otsuka K (2009):
bFGF rescues imatinib/STI571-induced apoptosis of sis-NIH3T3
fibroblasts. Biochem Biophys Res Commun 381: 165-70
Micke P, Hackbusch D, Mercan S, Stawowy P, Tsuprykov O,
Unger T, Ostman A, Kappert K (2009): Regulation of tyrosine
phosphatases in the adventitia during vascular remodelling.
Biochem Biophys Res Commun 382: 678-684
Goebel M, Clemenz M, Staels B, Unger T, Kintscher U, Gust
R (2009): Characterization of new PPARgamma agonists:
analysis of telmisartan’s structural components. ChemMed­
Chem 4: 445-456
Bal MS, Paulis L, Zicha J, Kunes J (2009): Effect of protein
kinase C and protein kinase A inhibitors on contraction of
isolated femoral arteries of SHR and Wistar rats. Physiol Res
58: 793-798
Reinemund J, Seidel K, Steckelings UM, Zaade D, Klare S,
Rompe F, Katerbaum M, Schacherl J, Li Y, Menk M, Schefe
JH, Goldin-Lang P, Szabo C, Olah G, Unger T, Funke-Kaiser
H (2009): Poly(ADP-ribose) polymerase-1 (PARP-1) transcriptionally regulates angiotensin AT2 receptor (AT2R) and AT2R
binding protein (ATBP) genes. Biochem Pharmacol 77:
1795-1805
237
Pharmacology
Vosgerau U, Lauer D, Unger T, Kaschina E (2010): Cleaved
High Molecular Weight Kininogen, a Novel Factor in the Regulation of Matrix Metalloproteinases in Vascular Smooth Muscle
Cells. Biochem Pharmacol 79: 172-179
Furundzija V, Fritzsche J, Kaufmann J, Meyborg H, Fleck E,
Kappert K, Stawowy P (2010): IGF-1 increases macrophage
motility via PKC/p38-dependent alphavbeta3-integrin insideout signaling. Biochem Biophys Res Commun 394(3):786-9
Kappert K, Furundzija V, Fritzsche J, Margeta C, Krüger J,
Meyborg H, Fleck E, Stawowy P (2010): Integrin cleavage
regulates bidirectional signaling in vascular smooth muscle
cells. Thromb Haemost 103(3):556-63
Kintscher U, Marx N, Martus P, Stoppelhaar M, Schimkus
J, Schneider A, Walcher D, Kümmel A, Winkler R, Kappert
K, Dörffel Y, Scholze J, Unger T (2010): Effect of high-dose
valsartan on inflammatory and lipid parameters in patients
with Type 2 diabetes and hypertension. Diabetes Res Clin
Pract 89(3):209-215
Rompe F, Artuc M, Hallberg A, Alterman M, Ströder K, ThöneReineke K, Reichenbach A, Schacherl J, Dahlöf B, Bader M,
Alenina N, Schwaninger M, Zuberbier T, Funke-Kaiser H,
Schmidt C, Schunck W-H, Unger T, Steckelings UM (2010):
Direct angiotensin AT2-receptor stimulation acts antiinflammatory through epoxyeicosatrienoic acid and inhibition of NF-κB.
Hypertension 55: 924-931
Paulis L, Pechanova O, Zicha J, Liskova S, Celec P, Mullerova
M, Kollar J, Behuliak M, Kunes J, Adamcova M, Simko F
(2010. Melatonin improves the restoration of EDCF-signalling
and inner diameter in the rat femoral artery after cessation of
L-NAME treatment. J Hypertens 28: S19-S24
Menk M, von Haefen C, Funke-Kaiser H, Sifringer M, Schefe
JH, Kirsch S, Seidel K, Reinemund J, Steckelings UM,
Unger T, Spies CD (2010): Ethanol-induced downregulation of
the angiotensin AT2 receptor in murine fibroblasts is mediated
by PARP-1. Alcohol 44: 495-506
Seidel K, Kirsch S, Lucht K, Zaade D, Reinemund J, Schmitz
J, Klare S, Li Y, Schefe JH, Schmerbach K, Goldin-Lang P,
Zollmann FS, Thöne-Reineke C, Unger T, Funke-Kaiser H
(2010): The promyelocytic leukemia zinc finger (PLZF) protein
exerts neuroprotective effects in neuronal cells and is dysregulated in experimental stroke. Brain Pathol (in press)
Simko F, Pechanova O, Pelouch V, Krajcirovicova K, Celec
P, Palffy R, Bednarova K, Vrankova S, Adamcova M, Paulis L
(2010): Continuous light and L-NAME-induced left ventricular
remodelling: different protection with melatonin and captopril.
J Hypertens 28: S13-S18
Paulis L, Pechanova O, Zicha J, Barta A, Gardlik R, Celec P,
Kunes J, Simko F (2010): Melatonin interactions with blood
pressure and vascular function during L-NAME-induced
hypertension. J Pineal Res 48:102-108
238
Zimmermann M, Kappert K, Stan ACU373-MG cells express
PepT2 and accumulate the fluorescently tagged dipeptidederivative β-Ala-Lys-N(ε)-AMCA. Neurosci Lett [in press]
Ohshima M, Yamaguchi Y, Matsumoto N, Micke P, Takenouchi Y, Nishida T, Kato M, Komiyama K, Abiko Y, Ito K,
Otsuka K, Kappert K (2010): TGF-β Signaling in Gingival
Fibroblast-Epithelial Interaction. J Dent Res [in press]
Caglayan E, Romeo G, Kappert K, Odenthal M, Südkamp M,
Body S, Sherman S, Hackbusch D, Vantler M, Kazlauskas A,
Rosenkranz (2010): Profilin-1 is expressed in human atherosclerotic plaques and induces atherogenic effects on vascular
smooth muscle cells. PloS ONE [in press]
Curato C, Slavic S, Dong J, Skorska A, Altarche-Xifró W,
Miteva K, Kaschina E, Thiel A, Imboden H, Wang J, Steckelings UM, Steinhoff G, Unger T, Li J (2010): Identification of
non-cytotoxic and IL-10-producing CD8+AT2R+ T cell population in response to ischemic heart injury. J Immunol 185(10)
[Epub ahead of print]
Koon K T, Sleight P, Dagenais G, Probstfield J, Anderson C,
Gao P, Diaz R, Dans A, Levine M, Unger T, Fagard R, Yusuf
S, for the ONTARGET (Ongoing Telmisartan Alone and in
Combination with Ramipril Global Endpoint Trial) and TRANSCEND (Telmisartan Randomized Assessment Study in ACE
Intolerant Subjects with Cardiovascular Disease) Trials (2010):
Effect of Ramipril, Telmisartan or Combination Therapy on the
Risk of Cancer in 31,000 Individuals. under review
Reviews & book chapters (2006-2010)
Hartge MM, Kintscher U, Unger T (2006): Endothelial dysfunction and its role in diabetic vascular disease. Endocrinol
Metab Clin North Am 35: 551-560
Pharmacology
Steckelings UM, Funke-Kaiser H, Unger T (2006): Mtus1.
AfCS-Nature Molecule Pages (doi:101038/mpa00394801)
Kappert K (2007): Folsäure als Sekundärprophylaxe bei
kardiovaskulären Erkrankungen. Der Kardiologe 1: 120–122
Kappert K (2006) Weitere atherosklerotische Risikofaktoren In: Prävention atherosklerotischer Erkrankungen.
Rosenkranz, Schneider, Erdmann (Editors) Georg Thieme
Verlag
Li J, Doerffel Y, Hocher B, Unger T (2007): Inflammation in
the genesis of hypertension and its complications - the role of
angiotensin II. Nephrol Dial Transpl 22: 3107-3109
Kappert K, Fätkenheuer G, Rosenkranz S (2006): Beeinflussung des Atherosklerose-Risikos durch medikamentöse
Therapien In: Prävention atherosklerotischer Erkrankun­
gen Rosenkranz, Schneider, Erdmann (Editors) Georg
Thieme Verlag
Peters H, Unger T (2006): Mast cells and the power of local
RAS activation. Nephrol Dial Transplant 22: 40-42
Clemenz M, Steckelings UM, Unger T (2006): Regulationsmechanismen des Renin-Angiotensin-Systems im kardiovaskulären System. In: Ganten, Detlev; Köhrle, Josef und
Ruckpaul, Klaus (Hrsg.) Molekularmedizinische Grundla­
gen von para- und autokrinen Regulationsstörungen.
Springer-Verlag Berlin Heidelberg 377-407
Östman A, Kappert K (2007) Protein Tyrosine Phosphatases
In: Endothelial Biomedicine William Aird (Editor) Cambridge
University Press
Micke P, Moustakas A, Ohshima M, Kappert K (2007):
Transforming Growth Factor-b in Cancer Therapy: Cancer
associated fibroblast and the role of TGFβ In: Transforming
Growth Factor-ß in Cancer Therapy; Volume 2: Cancer
Treatment and Therapy Sonia B. Jakowlew (Editor) The
Humana Press
Kaschina E, Doerfel N, Unger T (2007): Hypertonie In:
Schwandt P, Parhofer KG, eds. Handbuch der Fettst­
offwechselstörungen Stuttgart: Schattauer 538-550
Funke-Kaiser H, Schefe JH, Unger T (2007): Signaltransduktion des Renin-/ Prorenin-Rezeptors. MedReport 31: 8
Funke Kaiser H, Steckelings UM, Unger T (2006): Stimulation of AT2 receptors: role in the effect of angiotensin II receptor antagonists. In: Mancia G, ed. Angiotensin II Receptor
Antagonists: Informa Healthcare; 31-46
Steckelings UM, Unger T (2007): Angiotensin in the kidney:
a key to understanding hypertension? Cell Metab 5: 7-8
Thoene-Reineke C, Steckelings UM, Unger T (2006): Angiotensin receptor blockers and cerebral protection in stroke. J
Hypertens Suppl 24: S115-121
Schacherl J, Menard J, Unger T (2007): Towards an understanding of the renin-angiotensin-aldosterone system: an
historical review In: Sever P, ed. New Perspectives on
Hypertension. Direct Renin Inhibition. Birmingham, UK:
Sherborne Gibbs Limited 8-19
Unger T (2006): [Innovation or pseudo-innovation: that is the
question Med Klin (Munich) 101: 257-262
Unger T (2006): Laudatio. Zur Verleihung des Robert Koch
Award 2006 an MSD für das Medikament Alendronat (FOSAMAX/ FOSAVANCE) Med Klin (Munich) 101: 25-26
Unger T (2006): Stellungnahme. Antwort des Autors auf den
Leserbrief von M.H. Freitag und W.A. Wohlgemuth Med Klin
(Munich) 101: 590-593
Unger T (2006): Stellungnahme. Antwort des Autors auf den
Leserbrief von Herrn Becker-Brüser Med Klin (Munich) 101:
512-515
Unger T (2008): Comparing the cerebroprotective properties
of antihypertensive drugs in terms of their effects on angiotensin. Nat Clin Pract Nephrol 4: 12-13
Kappert K, Unger T, Kintscher U (2008): [Aliskiren hemifumarate] Dtsch Med Wochenschr 133: 1308-1312
Kappert K (2008): Pharmakologie der antihypertensiven
Therapie bei metabolischem Syndrom In: Antihypertensiva
bei metabolischem Syndrom und Typ 2 Diabetes Ulrich
Kintscher, Nikolaus Marx (Editors) UNI-MED Verlag AG
Schefe JH, Unger T, Funke-Kaiser H (2008): PLZF and the
(pro)renin receptor. J Mol Med 86: 623-627
239
Pharmacology
Steckelings UM, Unger T (2008): The renin-angiotensinaldosterone system. In: Mancia G, Grassi G, Kjeldsen S, eds
Manual of Hypertension of the European Society of
Hypertension London, U.K.: informa healthcare 110-116
Unger T (2008): Gegenwart und Zukunft der Hypertonieforschung. In: Lenz T, eds. Hypertonie in Klinik und Praxis.
Stuttgart, New York: Schattauer Verlag; 461-467
Kintscher U, Foryst-Ludwig A, Unger T (2008): Inhibiting Angiotensin Type 1 Receptors as a Target for Diabetes. Expert
Opinion on Therapeutic Targets 12: 1257-1263
Werner C, Baumhaekel M, Teo KK, Schmieder R, Mann J,
Unger T, Yusuf S, Boehm M (2008): RAS blockade with ARB
and ACE inhibitors: current perspective on rationale and
patient selection. Clin Res Cardiol 97: 418-431
Kappert K, Kusserow H, Unger T (2008): The pharmacological rationale behind polypharmacy in heart failure. Heart
Failure Monitor 6(1):20-7
Kolloch R, Unger T (2009): Aliskiren Stuttgart: Ligatur Verlag
für Klinik und Praxis
Mancia G, Unger T, Zanchetti A (2009): Introduction: Reducing cardiovascular risk: ONTARGET-a new standard in cardiovascular protection. J Hypertens Suppl 27(Suppl 5): S1
Unger T, Kintscher U, Kappert K, Steckelings UM
(2009):The ONTARGET Trial Programme: Facts and Lessons.
Current Hypertension Reviews 5: 202-209
Steckelings UM, Unger T (2009): Angiotensin receptors and
autophagy: live and let die. Hypertension 53: 898-899
Steckelings UM, Rompe F, Kaschina E, Unger T (2009):
The evolving story of RAAS in hypertension, diabetes and
cardiovascular disease- moving from macrovascular to microvascular targets. Fund Clin Pharmacol 23: 693-703
Kaschina E, Steckelings UM, Unger T (2009): RAS activation contributes to transition from atherosclerosis to aneurysm. International Atherosclerosis Society, IAS website,
Commentary., 4 December.
240
Steckelings UM (2009): Direct stimulation of Angiotensin
(AT)2-receptors: a therapeutic option of the future? Diabetes,
Stoffwechsel und Herz 17: 518-519
Unger T, Dahlöf B (2009): Compound 21, the first orally active,
selective agonist of the angiotensin II type 2 (AT2) receptor:
implications for AT2 receptor research and therapeutic potential. J Renin Angio Aldo Syst [Epub ahead of print]
Funke Kaiser H, Reinemund J, Steckelings UM, Unger T
(2009): Adapter proteins and promoter regulation of the angiotensin AT2 receptor – implications for cardiac pathophysiology. J Renin Angio Aldo Syst 11: 7-17
Steckelings UM, Rompe F, Kaschina E, Namsolleck P,
Grzesiak A, Funke-Kaiser H, Bader M, Unger T (2009):
The past, present and future of angiotensin II type 2 receptor
stimulation. J Renin Angio Aldo Syst 11: 67-73
Unger T (2009): Antihypertensives as over-the-counter drugs?
Dtsch Med Wochenschr 134: 2302-2304
Häßle P, Fätkenheuer G, Rosenkranz S, Kappert K (2009):
Direkte und indirekte atherogene Effekte der HIV-Infektion
und antiretroviralen Therapie. Der Kardiologe 3: 24-34
Unger T (2009): The rationale for choosing telmisartan and
ramipril in the ONTARGET programme. Eur Heart J Suppl
11(Suppl F):F3-F8
Funke-Kaiser H, Zollmann FS, Schefe JH, Unger T (2010):
Signal transduction of the (pro)renin receptor as a novel therapeutic target for preventing end-organ damage. Hyperten­
sion Research 33: 98-104
Kaschina E and Unger T (2010). Pathophysiologic link
between atherosclerosis and nephrosclerosis. In: Cardiore­
nal Syndrome (Berbari AE, Mancia G ed.), 396p, Springer
Italia, Milan 245-253
Steckelings UM, Th.Unger (2010): Kurzbewertung Aliskiren.
Internistische Praxis 50: 371-374
Ströder K, Unger Th, Steckelings UM (2010) The renin-angiotensin-system and the eye. Frontiers in Diabetes: Diabetic
retinopathy - an update; Eds.: M.Porta, H.-P. Hammes,
Karger, Basel, Vol.20: 142-157
Pharmacology
Rompe F, Unger T, Steckelings UM (2010): The AT2-receptor in inflammation. Drug News Perspectives 23: 104-111
Steckelings UM, Widdop RE, Paulis L, Unger Th (2010): The
Angiotensin AT2-Receptor in left ventricular hypertrophy. J
Hypertens 28 (suppl 1): S50–S55
Kjeldsen SE, Schmieder RE, Unger T, Mancia G (2010): Combination therapy with telmisartan and hydrochlorothiazide: a
strategy for improved blood pressure control. Curr Med Res
Opin (in press)
Widdop RE, Steckelings UM, McCarthy CA, Callaway JC
(2010): Angiotensin receptors in neuroprotection. In: ‘Neuropeptides in Neuroprotection and Neuroregeneration’. F
Nyberg (Editor); Taylor & Francis (publisher); in press
Patents (2006-2009)
DETERMINATION OF RENIN/ PRORENIN RECEPTOR
ACTIVITY (EP 1890152A International Application No.: PCT/
EP2007/006100, Publication Date: 20.02.2008, International
Filing Date: 14.08.2006) Funke-Kaiser H, Zollmann F, Schefe
JH, Unger T
Thomas Unger’s Group
Third party funding ( 2006-2010 )
Project leader
Unger T.
Funke-Kaiser H.
Steckelings UM.
Funke-Kaiser H.
Unger T.
Thöne-Reineke C.
Funke-Kaiser H.
Unger T.
Funke-Kaiser H.
Funke-Kaiser H.
Unger T.
Funke-Kaiser H.
Schefe J.H.
Zollmann F.
Unger T.
Project title
TP7: Der AT2-Rezeptor bei MyokardHypertrophie.
Effects of renin inhibitors on the renin/
prorenin receptor-PLZF signal
transduction cascade.
Neuroprotective and neuroregenerative
effects of aliskiren in double transgenic
rats expressing human renin and angiotensinogen genes.
Identifizierung und molekulare, epigenetische und klinische Charakterisierung eines CpG-Mikrosatellitenbindenden Transkriptionsfaktors.
JRP B1 (Genetics and genomics of
hypertensive patients with early stroke)
- P15
Der Renin-/ Prorenin-Rezeptor als
neue pharmakologische Zielstruktur.
Sponsor
DFG GK-754-III: Geschlechtsspezifische Mechanismen bei
Myokard-hypertrophie
Actelion Pharmaceuticals
Period
2006-2010
Novartis
2008-2009
DFG-Einzelantrag
(FU 463/2-1)
2006-2007
EU: Network of Excellence
“InGenious HyperCare”
(no. 037093)
Investitionsbank Berlin (IBB)
- ProFIT-Programm
(Antragsnummer
10138510)
2006-2010
2007-2008
2008-2010
241
Pharmacology
Project leader
Funke-Kaiser H.
Funke-Kaiser H.
Schefe J.
Zollmann F.
Unger T.
Funke-Kaiser H
Unger T.
Li J. Unger T.
Li J. Unger T.
Li J.
Unger T.
Bader M.
Li J. Unger T.
Li J.
Unger T.
Unger T.
Thöne-Reineke C.
Unger T.
Thöne-Reineke C.
Unger T.
Kintscher U.
Unger T.
Li J.
Kaschina E.
Unger T.
Kaschina E.
Unger T.
242
Project title
Entwicklung von RERBs, einer neuen
Medikamentenklasse zur Endorganprotektion.
Der Renin/ Prorenin-Rezeptor als
pharmakologische Zielstruktur.
Sponsor
BMBF -GO-Bio-Programm
(GO-Bio 0315092)
Period
2008-2010
Stiftung Charité
2008-2010
Molekulare und klinische Charakterisierung eines Transkriptionsfaktor­
bindenden CpG-Promotor-Mikrosatelliten bei M. Alzheimer.
Angiotensin receptors in apoptosis and
inflammation after myocardial infarction
Angiotensin AT2 receptor in cardiac
regeneration
Functional relevance of AT2+c-kit+
cells in cardiac ischemic injury
Dr. Werner Jackstädt-Stiftung
2009-2010
EU: CardioVasc-3.2 (contract
MEST-CT-2005-020268)
2006-2009
BMBF: BCRT-kick off grant
2007-2008
BMBF: 1st. BCRT grant
2008-2010
Einfluss von Östrogen auf die UmgeDFG: GK-754-III
staltung der myocardialen Matrix nach
Myokardinfarkt
Einfluß von Östrogenrezeptor auf die
FERMENTATION – BIOTEC GmbH
Plastizität und Regenerationsfähigkeit
hämatopoetischer Stammzellen
Aliskiren im Schlaganfall in DTGR
Novartis Pharma GmbH
2006-2010
Schlaganfall Intervention
u.Präventation
Regulation of tissue-specific endothelial dysfunction by telmisartan: The role
of PPARg activation
Stammzellen von Patienten mit Herzinsuffizienz
Genetics of aneurysm
Solvay Pharmaceuticals GmbH
2006-2009
Nippon Boehringer Ingelheim Co.,
Ldt.
2006-2009
Praxis Dr. Pesic
2008-2011
National Genome Research Network (NGFN -2), BMBF
Sanofi-Aventis
2004-2008
Rimonabant in heart failure
2008-2010
2006-2010
2006-2009
Pharmacology
Project leader
Kaschina E.
Unger T.
Kaschina E.
Unger T.
Kaschina E.
Unger T.
Unger T
Paulis L.
Unger T.
Funke-Kaiser H.
Steckelings UM.
Steckelings UM.
Steckelings UM.
Thöne-Reineke C.
Unger T.
Steckelings UM.
Unger T.
Valero V.
Steckelings UM.
Steckelings UM.
Unger T.
Kappert K.
Kappert K.
Kintscher U.
Kappert K.
Kappert K.
Project title
Sponsor
Period
AT2 receptor agonist Compound 21 in EU: Marie Curie Early Stage
2006-2009
myocardial infarction
Research Training Program CARDIOVASC MEST-CT-2005-020268
Telmisartan in aneurysm
Bayer Schering Pharma
2006-2008
Kininogen in heart failure
Bayer Schering Pharma
2010
COME-in-CARE (COmpound 21
and MElatonin in CArdiovascular
REmodeling)
Gender specific mechanisms in myocardial hypertrophy
EU: 7. FP-PIEF-237834
8/20091/2011
DFG GK 754-III
2006-2010
Bedeutung des Renin-AngiotensinSystems bei Pathogenese und Therapie der Sklerodermie
Candesartan in diabetic retinopathy
Deutsche Stiftung Sklerodermie
2006-2007
Takeda Pharma
2006-2009
Preclinical development of an orally
active AT2-receptor agonist
Evaluation of neuroprotective and antiinflammatory effects of direct AT2
receptor stimulation in multiple sclerosis
Preclinical development of an orally
active AT2-receptor agonist
Return Scholarship
Vicore Pharma
2008-2010
Berlin-Padua-Gdansk Graduate
Program/DIB
2009-2011
Eurostars, HEARTSAVE, EU
2010-2011
German Research Foundation
(no. KA1820/2-1)
European Commission
2006-2007
Role of protein tyrosine phosphatases
in metabolic and cardiovascular disease
PPARγ activation by Losartan in Hyper- Merck Sharp & Dohme (MSD)
tensive Patients: The Importance of
Losartan-Metabolites
Oxidation of the catalytic site cysteine Charité-University Medicine Berlin
of protein tyrosine phosphatases in
Research Scholarship
cardiac ischemia/reperfusion
2007-2009
2007-2008
2007-2007
243
Pharmacology
Project leader
Kappert K.
Kappert K.
Kappert K.
Kappert K.
Kappert K.
Krüger J.
Project title
Transfer of the hemodynamic 3-vessel
occlusion model into the genetically
modified mouse – Role of the protein
tyrosine phosphatase SHP-1 in cerebral arteriogenesis
Protein Tyrosine Phosphatases in
Arteriogenesis
SHP-1 in cerebral arteriogenesis in
mice
Targeting protein tyrosine phosphatases in high fat diet induced
insulin resistance
Protein Tyrosine Phosphatase DEP-1
as molecular target in insulin resistance
Protein Tyrosine Phosphatases (PTPs)
as therapeutic targets treating high fat
diet induced insulin resistance
Sponsor
Charité-University Medicine Berlin
Habilitation Scholarship
Period
2007-2008
German Research Foundation
(no. KA1820/4-1)
Deutsche Stiftung für Herzforschung
(F/04/08)
Charité-University Medicine Berlin
Personal funding
2008-2010
2008-2010
2008-2010
German Diabetes Society
2010-2011
Charité-University Medicine Berlin
PhD scholarship
2010-2011
Awards ( 2006-2010 )
2006
Charité Research Scholarship K. Kappert
2006
Travel Award, ESH Investigator’s Initiative Group
F. Rompe
2006 Finalist, Young Investigator Award, Gordon Research
Conference on Angiotensin
F. Rompe
2006 Travel Grant, European Society of Hypertension, Madrid L. Paulis
2006 Education Award of Volksbank AG L. Paulis
2006 Jiri Widimsky Award for promising young scientists
in hypertension research on the 16th European Society
for Hypertension Meeting, Madrid L. Paulis
244
Pharmacology
2006 1st Prize winner in the 1st PhD Student Scientific Conference,
School of Medicine, Bratislava L. Paulis
2007
Prize winner of the GO-Bio-competition of the BMBF
H. Funke-Kaiser
2007
Young Investigator Travel Award,
European Society of Cardiology
K. Kappert
2007
Young Investigator Award, German Hypertension Society K. Kappert
2007
Travel Award, European Society of Hypertension F. Rompe
2007
Participant of the 57th Meeting of Nobel Laureates, Lindau, Germany J. Schefe
2007
Charité Habilitation Scholarship K. Kappert
2007
Travel Award, European Council for Cardiovascular Research P. Namsolleck
2007 1st Prize winner in the Physiology Section and
5th in the Zondek Price competition of the 18th European
Student Conference, Berlin L. Paulis
2007 Travel Grant, European Society of Hypertension, Milan
L. Paulis
2007
Poster award: 12th Annual Meeting of the European
Council for Cardiovascular Research (ECCR)
E. Kaschina
2008 Travel Grant, European Society of Hypertension, Berlin L. Paulis
2008
Young Investigator Travel Award, European Society of Cardiology
K. Kappert
2008
Austin Doyle Award of the International Society of Hypertension (ISH) A. Grzesiak
2008
Poster Prize, European Council for Cardiovascular Research F. Rompe
2008
Research Scholarship, Deutsche German Hypertension Society J. Schefe
245
Pharmacology
2009
Charité-Promotionspreis J. Schefe
2009
Berlin-Brandenburg School for Regenerative Therapies (BSRT)
PhD Student Award Wassim Altarche-Xifró
2009
Travel Award, European Society of Hypertension P. Namsolleck
2009
Young Investigator Award, Deutsche Hypertonie Liga S. Wardat
2009
Travel Award, European Society of Hypertension F. Santi
2009
Travel Award, European Council for Cardiovascular Research F. Rompe
2009
Honorary Member of the British Hypertension Society T. Unger
2009
1st Prize winner in competition for the best publication in
experimental hypertension by Blood Pressure, NGO, Prague L. Paulis
2009 Acknowledgement for Scientist of the Year 2008,
Min. Edu. SR, Bratislava, L. Paulis
2010
Honorary Member of the Italian Hypertension Society
T. Unger
2010
Subsession Poster Prize, Frontiers in Cardiovascular Biology,
European Society of Cardiology
J. Krüger
2010
Young Investigator Travel Award, European Society of Cardiology
D. Hackbusch
2010 Accommodation Grant, European Society of Hypertension, Oslo L. Paulis
2010 Travel Award, Heart Failure of the ESC, Berlin L. Paulis
2010
Travel Grant, European Society for Cardiology, Stockholm L. Paulis
2010 1st prize winner in the Young Investigator Award in
Life Sciences of the Slovak Academy of Sciences, Bratislava L. Paulis
246
Pharmacology
2010
Travelling Award for the 20th European Meeting
of Hypertension, Oslo
S. Slavic
2010
Scholarship for the Promotion from Charité
Universitätsmedizin S. Slavic
Genbank entries
Schefe, J.H., Neumann, C., Funke-Kaiser, H., Thone-Reineke, C., Unger, T. (2006):
Rattus norvegicus glutamate transporter 1 gene, promoter region and partial cds.
Genbank accesion DQ489741, GI:94958152
247
PONTUS B. PERSSON – VEGETATIVE PHYSIOLOGY
Head of the Group
Prof. Dr. Pontus B. Persson
Curriculum Vitae: Pontus B. Persson, born in 1962 in Lund Sweden received his
education in the USA, Germany and Italy. He graduated from the Ruprecht-KarlsUniversity in Heidelberg, Germany in 1987 and subsequently received his training in
physiology at the same University from 1987-1993. In 1991 he finished his Habilitation
(an extensive PhD-like thesis). After his appointment of full professor of Physiology
1994 at the Charité-Universitätsmedizin Berlin, Germany he became Director of the
Institute for Vegetative Physiology.
Pontus B. Persson was awarded with the Ruprecht-Karls-Prize of the University Heidelberg (1990), the Research
Prize of the German Research Foundation (Deutsche Forschungsgemeinschaft: Gerhard-Hess-Förderprogramm) (1991), the Young Investigator Award of the American Physiological Society (1998) and the Henry
Pickering Bowditch Award of the American Physiological Society (2002).
Pontus B. Persson was Editor-in-Chief of the American Journal of Physiology – Regulatory, Integrative and
Comparative Physiology from 2001-2007 and is now Consulting Editor at the same Journal. He is Review Editor
for the Journal Acta Physiologica. For Pflügers Archiv- European Journal of Physiology he works as Executive
Editor. Prof. Persson’s research interests include kidney hemodynamics, the renin-angiotensin system and the
reflex control of circulation.
249
PONTUS B. PERSSON – VEGETATIVE PHYSIOLOGY
Summary
The current focus of our department is kidney function and its importance for cardiovascular control.
The individual research groups act in a concerted
effort to disentangle the complex relationship
between systemic hemodynamics and the kidney.
Moreover, light is shed on to developmental aspects.
These efforts are supported by several federal
grants, mainly by the German Research Foundation.
The various levels at which research takes place is
mirrored by the broad spectrum of Journals where
the results of this department have been published
over the latest decade.
For more information regarding research activities
your are welcome to visit the individual research
group sites.
Zusammenfassung
Der derzeitige Forschungsschwerpunkt der HerzKreislauf Physiologie liegt auf der Nierenfunktion
und deren Bedeutung für die Kontrolle des Kreislaufs. Die einzelnen Forschungsgruppen arbeiten
eng zusammen, um die komplexen Beziehungen
zwischen dem Blutkreislauf und der Niere zu untersuchen. Des weiteren werden Aspekte der Entwicklungsphysiologie erforscht. Diese Versuche werden
durch verschiedene Institutionen gefördert, haupt-
250
sächlich durch die Deutsche Forschungsgemeinschaft. Die Forschung findet auf unterschiedlichen
Ebenen statt. Während der letzten 10 Jahre wurden
die daraus resultierenden Ergebnisse in einem breiten Spektrum von Journalen veröffentlicht.
Weitere Informationen zu den Forschungstätigkeiten
können Sie auf den Seiten der Forschungsgruppen
nachlesen.
PONTUS B. PERSSON – VEGETATIVE PHYSIOLOGY
Publications 2006 – 2010
Lai EY, Patzak A, Steege A, Mrowka R, Brown R, Spielmann
N, Persson PB, Fredholm BB, Persson AEG (2006): Contribution of adenosine receptors in the control of arteriolar tone
and adenosine-angiotensin II interaction. Kidney Intern. 70:
690-698
Bräutigam M, Persson PB (2006): Do iodinated contrast
media interfere with renal tubular creatinine secretion?
Radiol. 240: 615
Persson PB, Liss P, Hansell P (2007): Evaluation and comparison between visipaque (Iodixanol) and Hexabrix (Ioxaglate) in
coronary angiography. J Am Coll Cardiol 49: 1668-1671
Stauss HM, Persson PB (2006): Cardiovascular variability and
the autonomic nervous system. J of Hyperten. 24: 19021905
Persson PB, Liss P, Hansell P, Lagerqvist B (2007): Response
to “Iodixanol vs ioxaglate for preventing contrast nephropathy:
Who is the winner?” Kidney Intern. 71: 828-829
Fähling M, Mrowka R, Steege A, Nebrich G, Perlewitz A,
Persson PB, Thiele BJ (2006): Translational control of collagen prolyl 4-hydroxylase-α (I) gene expression under hypoxia.
J Biol Chem. 281: 26089-26101
Mrowka R, Steege A, Kaps C, Herzel HP, Thiele BJ, Persson
PB, Blüthgen N (2007): Dissecting the action of an evolutionary conserved non-coding region on rennin promoter activity.
Nucleic Acids Res. 35 (15): 5120-5129
Thone-Reineke C, Kalk P, Dorn M, Klaus S, Simon K, Pfab
T, Godes M, Persson PB, Unger T, Hocher B (2006): Highprotein nutrition during pregnancy and lactation programs
blood pressure, food efficiency and body weight of the
offspring in a gender dependent manner. Am J Physiol. 291:
R1025-R1030
Gericke A, Martinka P, Nazarenko I, Persson PB, Patzak A
(2007): Impact of α1 –adrenoreceptor expression on contractile properties of vascular smooth muscle cells. Am J
Physiol. 293: R1215-R1221
Liss P, Persson PB, Hansell P, Lagerqvist B (2006): Renal
failure in 57 925 patients undergoing coronary procedures
using iso-osmolar or low-osmolar CM. Kidney Intern. 70
(10): 1811-1817
Lai EY, Martinka P, Fähling M, Mrowka R, Steege A, Gericke
A, Sendeski M, Persson PB, Persson AEG, Patzak A (2006):
Adenosine restores angiotensin II-induced contractions by
receptor-independent enhancement of calcium sensitivity in
renal arterioles. Circ Res. 99 (10): 1117-1124
Persson PB (2006): Guest editor appreciation. Am. J.
Physiol. 290: R493
Persson PB (2006): From clinical insights to new therapies.
Am. J. Physiol. 290: R124-R125
Persson PB (2006): Where we stand: American Journal of
Physiology – Regulatory, Integrative and Comparative Physiology 2006. Am J Physiol. 291: R489-R490
Persson PB (2007): A last look: American Journal of Physiology-Regulatory, Integrative and Comparative Physiology
2001-2007. Am J Physiol. 292: R665 3
Seeliger E, Flemming B, Wronski T, Ladwig M, Arakelyan K,
Godes M, Möckel M, Persson PB (2007): Viscosity of contrast
media perturbs renal hemodynamics. J Am Soc Nephrol.
18: 2912-2920
Patzak A, Lai EY, Fähling M, Sendeski M, Martinka P, Persson
PB, Persson AEG (2007): Adenosine enhances long-term the
contractile response to angiotensin II in afferent arterioles. Am
J Physiol. 293: R2232-2242
Patzak A, Steege A, Lai EY, Brinkmann JO, Kupsch E, Spielmann N, Gericke A, Skalweit A, Stegbauer J, Persson PB,
Seeliger E (2008): Angiotensin II response in afferent arterioles of mice lacking either the endothelial or neuronal isoform
of nitric oxide synthase. Am J Physiol. 294:R429-R437
Martinka P, Lai EY, Fähling M, Jankowski V, Jankowski J,
Schubert R, Gaestel M, Persson AEG, Persson PB, Patzak A (2008): Adenosine increases calcium sensitivity via
receptor-independent activation of the P38/MK2 pathway in
mesenteric arteries. Acta Physiologica 193:37-46
Steege A, Fähling M, Paliege A, Bondke A, Kirschner KM,
Martinka P, Kaps C, Patzak A, Persson PB, Thiele BJ, Scholz
H, Mrowka R (2008): Wilms’ tumor protein (-KTS) modulates
renin gene transcription. Kidney Intern. 74: 458-466
251
PONTUS B. PERSSON – VEGETATIVE PHYSIOLOGY
Fähling M, Mrowka R, Steege A, Kirschner KM, Benko E,
Förstera B, Persson PB, Thiele BJ, Meier JC, Scholz H
(2009): Translational Regulation of the Human Achaete-scute
Homologue-1 by Fragile X Mental Retardation Protein. J Biol
Chem. 284 (7): 4255-4266
Lüdemann L, Nafz B, Elsner F, Große-Siestrup C, Meissler
M, Kaufels N, Rehbein H, Persson PB, Michaely HJ, Lengsfeld P, Voth M, Gutberlet M (2009): Absolute quantification
of regional renal blood flow in swine by dynamic contrastenhanced magnetic resonance imaging using a blood pool
contrast agent. Invest Radiol. 44(3):125-134
Sendeski M, Patzak A, Pallone TL, Cao C, Persson AEG,
Persson PB (2009): Iodixanol, constriction of medullary
descending vasa recta, and risk for contrast medium-induced
nephropathy. Radiol. 251(3):697-704
Seeliger E, Wronski T, Ladwig M, Dobrowolski L, Vogel
T, Godes M, Persson PB, Flemming B (2009): The reninangiotensin system and the third mechanism of renal blood
flow autoregulation. Am. J. Physiol. Renal Physiol.
296:F1334-F1345
Lai EY, Fähling M, Ma Z, Källskog Ö, Persson PB, Patzak A,
Persson AEG, Hultström M (2009): Norepinephrine increases
calcium sensitivity of mouse afferent arteriole, thereby
enhancing angiotensin II-mediated vasoconstriction. Kidney
Intern. 76:953-959
Sendeski M, Patzak A, Person PB (2010): Constriction of the
vasa recta, the vessels supplying the area at risk for acute
kidney injury, by four different iodinated contrast media, evaluating ionic, nonionic, monomeric and dimeric agents. Invest.
Radiol. 45: 453-457
252
Perlewitz A, Nafz B, Skalweit A, Fähling M, Persson PB, Thiele
BJ (2010): Aldosterone and vasopressin affect α- and γ-ENaC
mRNA translation. Nucleic Acids Res. 38: 5747-5760
Seeliger E, Becker K, Ladwig M, Wronski T, Persson PB,
Flemming B (2010): Up to 50-fold increase in urine viscosity with iso-osmolar contrast media in the rat. Radiol. 256:
406-414
Schildroth J, Rettig-Zimmermann J, Kalk P, Steege A, Fähling
M, Sendeski M, Paliege A, Lai EY, Bachmann S, Persson PB,
Hocher B, Patzak A (2010): Endothelin type A and B receptors in the control of afferent and efferent arterioles in mice.
Nephrol Dial Transplant. E-pub ahead of print Sept 2nd,
2010
Reviews & book chapters (2006-2010)
Persson PB (2006): Pathophysiology of contrast-induced
nephropathy. In: Contrast-Induced Nephropathy edited by A.
L. Bartorelli and G. Marenzi. London: Taylor & Francis, p. 3-17
Persson PB (2006): Temperature control: from molecular
insights, regulation in king penguins and diving seals, to studies in humans. Am. J. Physiol. 291: R512-R514
Persson PB (2007): The Magic Mountain or Death in Venice:
Chronic Hypoxia may alleviate Oxidative Stress in the Kidney.
J Physiol. 582.1: p1
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
Head of the group
Prof. Dr. med. Holger Scholz
Curriculum Vitae: Holger Scholz studied medicine at the University of Heidelberg.
He received his medical degree in 1987 based on experimental studies on the electrophysiology of the renin secreting juxtaglomerular cells in the kidney. After a one-year
practice at the Department of Pathology (University of Heidelberg) he worked from
1988 to 1991 as a post-doctoral fellow in the Institute of Physiology at the University
of Zurich, Switzerland, on the oxygen-regulated synthesis of erythropoietin, the major
humoral growth factor for red blood cell precursors. From 1991-1999 he was in the Institute of Physiology at
the University of Regensburg where he received the Habilitation degree in 1995 for his work on the blood
pressure-dependent control of renin secretion. Supported by fellowships from the Max-Kade-Foundation and
the German Research Foundation (DFG), he joined the Developmental Biology Group at the Department of
Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA (1995-1998). In 1998
Holger Scholz became a Professor in Physiology at the Medical Faculty Charité. The research projects of his
group are at the interface of cancer and development. It is a major goal to elucidate the regulation and function
of the Wilms’ tumor protein, Wt1, which is necessary for normal embryogenesis and additionally functions as
a tumor suppressor molecule. Other scientific interests allude to the role of neurotrophins and their receptors
in the cardiovascular system and the oxygen-dependent control of gene expression.
Members of the group
Scientists
Bondke Persson, Anja
Kirschner, Karin
Dr. med.
Dr. rer. nat.
Technicians
Grätsch, Inge Richter, Angelika
Ing. (FH)
Ing. (FH)
Students
Braun, Julian
Hagen, Patricia
Jacobi, Charlotte
Karadeniz, Serap
Schiebel, Susann Sciesielski, Lina
Trams, Mareike
MD student
MD student
PhD student
Bachelor student (2009)
PhD student (2007-2008)
PhD student (2006-2009)
Scientist (January to July 2010)
Diploma student (2008)
From left to right, front row: H. Scholz, K. Kirschner,
L. Sciesielski, A. Richter, I. Grätsch
Back row: C. Jacobi, S. Karadeniz, J. Braun,
A. Bondke Persson
253
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
Summary
Our research team is interested in the molecular
mechanisms that control the development of the
cardiovascular system. The focus of our work is
on the regulation and function of the Wilms’ tumor
gene, WT1. Another area of interest refers to the role
of hypoxia as a physiological signal during development and a determining factor in many disease
processes. In this field we currently focus on the
oxygen-regulated expression of the gene encoding
the neurotrophin receptor TrkB.
Normal organ formation is established by the temporally and spatially coordinated gene expression,
which is controlled by a variety of transcription factors. Therefore, the identification and characterization of transcriptional regulators with essential functions during embryogenesis may allow one to decipher the key signaling pathways in cardiovascular
development. The Wilms’ tumor transcription factor
WT1 was initially identified as a tumor suppressor due to loss-of-function mutations in malignant
pediatric kidney tumors known as Wilms’ tumors
or nephroblastomas. Targeted inactivation of Wt1
in mice caused intrauterine lethality of homozygous
embryos (Wt1-/-) with a failure of normal formation
of the genitourinary system, mesothelium, sensory
neuropithelia, myocardial blood vessels, and the
hematopoietic system. By the identification of tar-
254
get genes that are regulated by Wt1, we aim at
analyzing important transcriptional pathways in
cardiovascular development. Since tissue repair
in the heart and other organs frequently involves
reactivation of fetal genes, it is our expectation to
discover novel candidate mechanisms for future
regenerative therapies.
Besides acting as an oxidant, molecular oxygen is
also important for the control of gene expression.
A variety of oxygen-regulated genes were identified since the initial discovery of the hematopoietic
growth factor Erythropoietin (Epo). A group of heterodimeric molecules known as hypoxia-inducible
transcription factors (HIFs) play a key role in oxygendependent gene expression. Tissue hypoxia resulting from an imbalance between oxygen supply and
oxygen demand is a frequent condition in rapidly
growing tumors. In many malignancies the hypoxic
microenvironment stimulates the formation of new
blood vessels (angiogenesis) thereby promoting
tumor expansion. Recent studies indicate that local
tissue hypoxia can alter gene expression irrespective
of its proangiogenic effect in certain neuronal tumors
(neuroblastomas) towards an immature, i.e. malignant paradigm. By identification of the gene encoding the neurotrophin receptor TrkB as a functional
HIF target gene, we discovered a novel molecular
switch for the malignant transformation of neuro­
blastoma cells.
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
Zusammenfassung
Das wissenschaftliche Interesse unserer Arbeitsgruppe gilt den molekularen Steuerungsprozessen
bei der Entwicklung des kardiovaskulären Systems.
Der Schwerpunkt der Untersuchungen liegt bei der
Regulation und Funktion des Wilmstumor Transkriptionsfaktors WT1. Ein weiteres Arbeitsgebiet beinhaltet die Rolle von Sauerstoffmangel (Hypoxie) als
physiologisches Signal in der Entwicklung und als
Determinante von Krankheitsprozessen. Gegenwärtig konzentrieren wir uns auf die sauerstoffabhängige
Regulation des Neurotrophinrezeptors TrkB.
Die normale Organentwicklung erfordert eine durch
Transkriptionsfaktoren in ihrer zeitlichen und räumlichen Abfolge koordinierte Genexpression. Durch
Identifizierung transkriptionaler Regulatoren, die für
eine normale Organogenese notwendig sind, können deshalb zentrale Signalwege in der Embryonalentwicklung analysiert werden. Der Wilmstumor
Transkriptionsfaktor WT1 wurde ursprünglich aufgrund von Mutationen in malignen Nierentumoren
bei Kindern (Wilmstumoren, Nephroblastome) als
ein Tumorsuppressor charakterisiert. Die gezielte
Inaktivierung von Wt1 hatte bei Mäusen u. a. eine
Störung der Entwicklung von Nieren, Mesothel,
Sinnesepithelien, Herzmuskelgefäßen und blutbildendem System zur Folge. Durch Ermittlung von
Zielgenen, deren Expression durch den WT1 Transkriptionsfaktor kontrolliert wird, versucht unsere
Arbeitsgruppe, wichtige molekulare Regulationsmechanismen bei der kardiovaskulären Entwicklung aufzudecken. Die Untersuchungen erfolgen
vor dem Hintergrund, daß Organschädigung häufig
zur Reaktivierung eines fötalen Genexpressionsprogramms führt. Detailkenntnisse über die molekularen Entwicklungsmechanismen sind deshalb für das
Verständnis von Regenerationsprozessen und deren
therapeutische Beeinflussung relevant.
Sauerstoff ist nicht nur als Oxidans wirksam, sondern
spielt auch eine wichtige Rolle bei der Genexpressionskontrolle. Neben dem zunächst identifizierten
hämatopoietischen Wachstumsfaktor Erythropoietin
ist inzwischen eine Vielzahl weiterer Gene bekannt,
deren Transkription durch Sauerstoffmangel (Hypoxie) stimuliert wird. Hypoxieinduzierbare Transkriptionsfaktoren (HIFs) gelten als zentrale Mediatoren
einer bei Sauerstoffdefizit veränderten Genexpression. In Tumoren bewirkt ein Mißverhältnis von
Sauerstoffangebot zu zellulärem Sauerstoffbedarf
eine lokale Gewebehypoxie, wodurch die Bildung
von Blutgefäßen (Angiogenese) stimuliert und das
Tumorwachstum gefördert wird. Neueren Untersuchungen zufolge kann Sauerstoffmangel, unabhängig von einer proangiogenen Wirkung, die Genexpression in neuronalen Tumoren (Neuroblastomen)
in Richtung auf ein entdifferenziertes, d.h. malignes
Muster verändern. Mit der Charakterisierung des
Neurotrophinrezeptors TrkB als ein HIF-reguliertes
Zielgen haben wir einen neuen molekularen Schalter
für die maligne Transformation von Neuroblastomzellen entdeckt.
Multiple roles of the WT1 zinc finger protein in development
255
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
Research projects
1. The role of WT1 in blood vessel formation in the heart
Project leader
Coworkers
Funding
External cooperations
Karin Kirschner
Anja Bondke Persson, Inge Grätsch, Angelika Richter
- SCHO 634/4-1
Drs. Kay-Dietrich and Nicole Wagner, INSERM U907, University of Nice, France,
Prof. Dr. Andreas Schedl, INSERM U636, Centre de Biochimie,
University of Nice/Sophia-Antipolis, Nice, France
Recent studies demonstrate that blood vessel formation in the developing heart takes its origin in the
epicardium, a mesothelial tissue covering the outer
heart surface. During cardiogenesis epicardial cells
acquire a mesenchymal phenotype, loose their intercellular contacts and populate the subjacent myocardium. Here, the epicardium-derived cells differentiate to vascular endothelial and smooth muscle cells,
amongst other cell types. Hence, normal formation
of the epicardium is a critical precondition for blood
vessel development in the heart, and important information about myocardial vasculogenesis can be
expected from gene expression analysis in epicardial
cells. We reported for the first time that the Wilms’
tumor transcription factor Wt1, which is normally
expressed in the epicardium, is necessary for normal
vascularization of the heart. Mouse embryos with Wt1
deficiency (Wt1-/-) exhibited a severely reduced blood
vessel density in their hearts compared to wild-type
littermates. Remarkably, left coronary artery ligation
in rats resulted in de novo expression of Wt1 in blood
vessels adjacent to the infarcted tissue. It is therefore
conceivable that re-expression of Wt1 in response to
myocardial ischemia supports neovascularization of
256
the injured tissue through promoting mesenchymal
transition of resident vascular cells.
The aim of this project is to identify Wt1-regulated
genes with a role in blood vessel formation in the heart.
In this regard we identified the gene encoding the TrkB
neurotrophin receptor, Ntrk2, as a transcriptional Wt1
target. A Wt1 binding motif was identified in the TrkB
promoter that was required for tissue-specific expression of a reporter transgene in the developing epicardium of mice. TrkB-deficient embryos, like Wt1-/- fetal
mice, had fewer blood vessels in their hearts. Additional findings indicate that activation of the TrkB neurotrophin receptor by its natural ligand, brain-derived
neurotrophic factor (BDNF) prevents the newly formed
vascular cells in the heart from apoptosis. Another
downstream target of Wt1 with a presumed role in
vascular development is vascular endothelial (VE)
cadherin. It is suggested that enhanced expression of
VE-cadherin in response to Wt1 supports intercellular
contact formation. Overall our results qualify Wt1 as
an important transcriptional regulator for the appearance of the blood vessel system in the heart. Future
studies shall address the role of Wt1 in tissue repair
after myocardial damage.
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
2. Gene transcription in the epicardium
Project leader
Coworkers
Funding
External cooperation
Karin Kirschner
Inge Grätsch, Angelika Richter
- SCHO 634/4-1
Drs. Kay-Dietrich and Nicole Wagner, INSERM U907, University of Nice, France
The epicardium is a mesothelial tissue on the outer sur- in the developing heart. In an effort to analyze transcripface of the heart. It arises during embryogenesis from tional signaling pathways in cardiovascular progenitor
the proepicardial tissue which is formed by a cluster of cell formation we identified the gene encoding alpha-4
cells just dorsal to the developing heart tube. Integrity of integrin as a novel downstream target of Wt1. Alpha-4
the epicardium is a critical precondition for normal heart integrin is a heterodimeric cell adhesion molecule
development. Thus, epicardial defects are frequently which is expressed on epidardial and hematopoietic
associated with hypoplasia of the myocardium sug- progenitor cells. It binds to the extracellular matrix progesting that normal heart formation requires the transfer teins fibronectin and vascular cell adhesion molecule-1
of growth promoting signals from the epicardium to the (VCAM-1) that are present on cardiomyocytes. Lack of
cardiomyocytes. Recent findings indicate that epicar- alpha-4 integrin in mice resulted in a failure of proepidial cells contribute to the cardiomyocyte lineage and cardial cells to attach to the surface of the heart and,
also provide the source for different vascular cell types consequently, myocardial growth defects. Other Wt1
in the embryonic heart. During cardiogenesis epicardial candidate targets are currently investigated by analycells undergo a process of epithelial-to-mesenchymal sis of differentially expressed genes in the hearts of
transition (EMT), which allows them to become cardio- wild-type (Wt1+/+) vs. Wt1-deficient (Wt1-/-) mouse
vascular progenitor cells. Epicardium-derived mesen- embryos. Hopefully, analysis of Wt1-regulated genes
chymal cells migrate towards the myocardium, where may advance our understanding of the mechanisms of
they form vascular endothelial and smooth muscle cells myocardial tissue repair.
in addition to perivascular fibroblasts and cardiomyocytes.
Important insights into the molecular mechanisms of heart development can be obtained
from studies on the regulation and function
of epicardial genes. The Wilms’ tumor transcription factor Wt1 is among the molecules
that are necessary for normal development of
the epicardium. Mice with homozygous Wt1
deletion (Wt1-/-) lack an intact epicardium
Co-localization of Wt1 (red) and alpha-4 integrin (green) in the develand exhibit severe myocardial hypoplasia. oping epicardium of a mouse embryo by double immunofluorescence
Wt1 is necessary for EMT of epicardial cells labeling. Nuclei were counterstained with Dapi.
257
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
3. I dentification of WT1 target genes in the developing
kidney and other tissues
Project leader
Coworkers
External cooperation
Anja Bondke Persson
Karin Kirschner, Julian Braun, Charlotte Jacobi, Inge Grätsch, Angelika Richter
Dr. Kai Schmidt-Ott, MDC Berlin, Prof. Dr. Christof Englert, Fritz-Lipmann-Institut, Jena
Loss-of-function mutations in the WT1 gene are
responsible for approximately 10% of pediatric renal
tumors (Wilms’ tumors, nephroblastomas). Wilms’
tumors can arise when pluripotent progenitor cells
in the developing kidney rather continue to proliferate than differentiating to glomeruli and tubules. The
WT1 gene product is a zinc finger protein, whose
alternative splice variants predominantly function as
transcription factors. The most striking characteristic
of mice with homozygous Wt1 deletion (Wt1-/-) consists in a failure of normal formation of the kidneys and
gonads. Mammalian kidney development depends on
the reciprocal interaction of the metanephric mesenchyme and the invading ureteric bud. Epithelial differentiation and subsequent nephron formation does
not occur in Wt1-deficient metanephric mesenchyme,
which undergoes programmed cell death (apoptosis)
instead. It is the goal of this project to discover molecular Wt1 downstream target genes in the developing
kidney and other tissues. For this purpose we made
use of the M15 cell line, which is derived from the
murine mesonephros and expresses Wt1 at a robust
level. Differentially expressed genes were identified
by DNA microarray analysis of M15 cells with normal
and low Wt1 expression. Wt1 levels in M15 cells were
adjusted by transfection with gene-specific and nontargeting siRNAs, respectively. More than one dozen
differentially expressed genes, encoding cell adhesion
molecules, metalloproteinases, interleukins and other
molecules, could be verified by real time RT-PCR. The
258
regulatory sequences of these genes are currently
cloned and tested for Wt1 responsiveness in reporter
assays. The respective cis-elements are mapped, and
binding of Wt1 protein is assessed by electrophoretic
mobility shift assay and chromatin immunoprecipitation (ChIP). Additional experiments include expression
analysis of candidate target genes by immunohistochemistry and/or in situ mRNA hybridization of normal
and Wt1-deficient murine embryos. Functional studies
will be performed on explant cultures of embryonic
mouse kidneys. Eventually this project will allow for the
discovery of novel candidate genes in kidney formation
and renal tissue repair.
Organ culture of a mouse embryonic kidney explant. Developing glomeruli were stained with anit-WT1 antibody (red).
Labeling of cyterkeratin was used for visualization of the
branching ureteric bud (green).
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
4. Nuclear import pathways for WT1
Project leader
Coworkers
External cooperation
Holger Scholz
Anja Bondke Persson, Karin Kirschner, Inge Grätsch, Angelika Richter
Dr. Reinhard Depping, Institut für Physiologie, Universität Lübeck
More than two dozens protein variants which are cellular GFP is easy to detect by its emission of a green
generated by alternative mRNA splicing, the use of fluorescence upon excitation at approximately 490 nm
additional transcription start sites, RNA editing and wavelength. The basic construct was engineered such
post-translational modification, contribute to the mul- that GFP is retained outside the nuclei in the cytoplasm
tiple functions of the WT1 protein. A further regulatory of the transfected cells. Nuclear localization of GFP
paradigm is provided by the recently discovered abil- molecules fused to different amino acid sequences
ity of WT1 to shuttle between the nucleus and cyto- of the WT1 protein therefore suggests inclusion of
plasm. Consistent with its established function as a potential NLS. Using this approach we could map a
transcription factor WT1 is contained predominantly in sequence of 10 amino acids in the zinc finger domain
the cell nuclei of embryonic tissues. However, a vari- of WT1 that was necessary for nuclear import. Coety of tumors express WT1 mainly in the cytoplasm immunoprecipitation experiments revealed that WT1
suggesting that cytoplasmic WT1 has a role in tum- protein interacts with importin alpha-1 and importin
origenesis. Prompted by this possibility we under- beta in embryonic kidney cells. We intend to genertook efforts to analyze the nuclear import pathways ate mutant WT1 proteins, which lack functional NLS
for Wt1 protein. Bidirectional transport between the and are therefore expressed exclusively in the cell
cell nucleus and cytoplasm occurs through nuclear cytoplasm. These proteins will be used to address
pore complexes and involves a group of transport the question whether cytoplasmic WT1 can promote
proteins known as karyopherins. Translocation of pro- malignant growth characteristics.
teins into the nucleus is mediated by importins which bind to
nuclear localization sequences
(NLS) in their cytoplasmic cargos. NLS frequently consist of
clusters of basic amino acids
that are sometimes separated
by a spacer of approximately
10 amino acids (bipartite NLS).
Intracellular localization of green fluorescent protein (GFP) GFP in human embryonic
In order to pinpoint relevant NLS
kidney cells. Transfection of empty expression vector causes GFP expression excluin the Wt1 protein we made use
sively in the cytoplasm sparing the nuclei (A). Inclusion of a nuclear localization signal
of plasmids expressing a green
(NLS) from the zinc finger domain of the WT1 protein directs GFP expression into the
cell nuclei (B).
fluorescent protein (GFP). Intra259
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
5. R
egulation of the genes encoding Eryrthopoietin ( Epo ) and
its receptor ( EpoR ) during hematopoiesis
Project leader
Coworkers
Funding
External cooperation
Karin Kirschner
Patricia Hagen, Inge Grätsch, Angelika Richter
- DFG DA 484/2-1
Prof. Dr. Christof Dame, Klinik für Pädiatrie m. S. Molekulare Neonatologie,
Charité, Berlin, Dr. Matthias Ballmeier, Klinik für Pädiatrische Hämatologie
und Onkologie, Medizinische Hochschule Hannover
The Wilms’ tumor transcription factor WT1 is expressed
in CD34-positive hematopoietic progenitor cells, and
high WT1 levels are detected in acute leukemias with
unfavorable outcome. Previous studies showed that
murine hematopoietic progenitor cells with Wt1 deficiency (Wt1-/-) have a reduced colony forming capacity compared to wild-type (Wt1+/+) and heterozygous
(Wt1+/-) cells. It is the aim of this project to study the
role of Wt1 in normal hematopoiesis and to analyze the
molecular signaling pathways involved. In particular,
we investigate whether the expression of the major
hematopoietic growth factor, Erythropoietin (Epo), and
its cognate receptor (EpoR) are regulated by Wt1.
Electrophoretic mobility shift assays (EMSAs)
revealed that Wt1 protein can bind to several consensus motifs in the promoters of the Epo and EpoR
genes. Reporter constructs carrying Epo and EpoR
promoter sequences were stimulated 20- and 7-fold,
respectively, by co-transfection of blood-derived
cell lines with Wt1 expression constructs. Promoter
activation by Wt1 was abrogated upon site-directed
mutagenesis of the identified cis-elements. Chromatin
260
immunoprecipitation (ChIP) confirmed the interaction
of Wt1 protein with the Epo and EpoR promoters in
their normal chromosomal configuration. Cellular coexpression of Wt1 and Epo/EpoR was detected by
immunohistochemistry in fetal mouse liver, which is
the major site of prenatal hematopoiesis. Consistently,
Wt1-deficient fetal liver cells contained significantly
lower Epo and EpoR mRNA levels than cells of wildtype and Wt1-heterozygous (Wt1+/-) embryos. The
proliferative response of CD117-positive hematopoietic
progenitor cells to recombinant human (rh) EPO was
approximately 10-fold lower in the absence than in
the presence of Wt1. Further on, significantly fewer
hemoglobin-positive cells could be isolated from the
livers of Wt1-/- embryos than of their wild-type littermates. These results demonstrate that transcription
of the genes encoding Epo and its receptor is regulated by Wt1. Our findings establish Wt1 as a novel
transcriptional regulator during mouse hematopoiesis. Future investigations shall address the question
whether abnormal EpoR expression contributes to the
malignancy of certain forms of human leukemia.
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
6. M
echanisms and ( patho ) physiological consequences of an
ox ygen-dependent expression of the TrkB neurotrophin receptor
Project leader
Coworkers
Funding
External cooperation
Lina Scisielski
Anja Bondke Persson, Karin Kirschner, Inge Grätsch, Angelika Richter
- SCHO 634/6-1
Dr. Christina Warnecke, Med. Klinik IV, Universität Erlangen-Nürnberg
Neurotrophins are a family of secreted nerve growth
factors with critical roles in the development and function of the nervous system and other tissues. The cellular effects of neurotrophins are mediated by three
high-affinity tyrosine kinase receptors (Trks). Nerve
growth factor binds to TrkA, brain-derived neurotrophic factor (BDNF) and neurotrophin-4 interact
with TrkB, whereas neurotrophin-3 has high affinity
for TrkC. The p75NTR membrane receptor binds to
all neurotrophins with low affinity.
High level expression of TrkB in certain types of
neuronal neoplasias (neuroblastomas) correlates
with aggressive tumor growth and poor prognosis.
Regional hypoxia due to insufficient supply with oxygen changed the gene expression pattern in neuroblastomas towards an immature, more malignant pattern. We therefore reasoned that hypoxia favors tumor
expansion by stimulating TrkB expression in neuroblastoma cells. Exposure of neuroblastoma-derived
Kelly cells in a 1% oxygen atmosphere increased TrkB
mRNA levels more than 20-fold compared to incubation at 21% O2. The rise in TrkB transcripts was associated with increased receptor protein expression. With
the combinatorial use of various cell and molecular
biology techniques we identified the hypoxia-inducible
factor-1 (HIF-1) as a transcriptional activator of the TrkB
encoding gene, NTRK2, in hypoxia. Using transwell
assays we could show that low oxygen stimulates the
migration of cultured neuroblastoma cells. This effect
of hypoxia was abrogated by blockade of the BDNFTrkB signaling pathway.
Interestingly, highest TrkB expression under hypoxic
conditions in vivo was found in the airway epithelium.
Exposure of rats to 8% O2 increased TrkB transcripts
and protein in airway epithelial cells approximately
15-fold. Consistently, the TrkB ligand, BDNF, significantly enhanced the contractile response to acetylcholine (ACh) of isolated tracheal segments from
hypoxic (8% O2) but not from normoxic (21% O2) rats.
This effect of BDNF was prevented by pre-incubation of the tissue specimens with the tyrosine kinase
inhibitor K252a and by mechanical removal of the TrkB
expressing airway epithelium. Likewise, the nitric oxide
(NO) synthase inhibitor L-NAME abrogated the influence of BDNF on ACh-induced airway contractions.
These findings indicate that activation of TrkB signaling by BDNF in hypoxia enhances mechanical airway
contractility to ACh through a mechanism that requires
NO. Our results are in agreement with clinical observations that neurotrophins contribute to airway hyperresponsiveness in patients with bronchial asthma.
261
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
Publications 2006 - 2010
Dame C, Kirschner KM, Bartz KV, Wallach T, Hussels CS,
Scholz H (2006): Wilms tumor suppressor, Wt1, is a transcriptional activator of the erythropoietin gene. Blood 107 (11):
4282-4290
Wagner N, Wagner KD, Scholz H, Kirschner KM, Schedl A
(2006): Intermediate filament protein nestin is expressed in
developing kidney and heart and might be regulated by the
Wilms’ tumor suppressor Wt1. Am J Physiol Regul Integr
Comp Physiol 291(3): R779-787
Kirschner KM, Wagner N, Wagner KD, Wellmann S, Scholz H
(2006): The Wilms tumor suppressor Wt1 promotes cell adhesion through transcriptional activation of the alpha4integrin
gene. J Biol Chem 281(42): 31930-31939
Martens LK, Kirschner KM, Warnecke C, Scholz H (2007):
Hypoxia-inducible factor-1 (HIF-1) is a transcriptional activator of the TrkB neurotrophin receptor gene. J Biol Chem
282(19): 14379-14388
Steege A, Fähling M, Paliege A, Bondke A, Kirschner KM,
Martinka P, Kaps C, Patzak A, Persson PB, Thiele BJ, Scholz
H, Mrowka R (2008): Wilms’ tumor protein WT1(-KTS) modulates renin gene transcription. Kidney Int 74(4): 458-466
Kirschner KM, Hagen P, Hussels CS, Ballmaier M, Scholz H,
Dame C (2008): The Wilms’ tumor suppressor Wt1 activates
transcription of the erythropoietin receptor in hematopoietic
progenitor cells. FASEB J 22(8): 2690-2701
Fähling M, Mrowka R, Steege A, Kirschner KM, Benko E,
Förstera B, Persson PB, Thiele BJ, Meier JC, Scholz H
(2009): Translational regulation of the human achaete-scute
homologue-1 by fragile X mental retardation protein. J Biol
Chem 284(7): 4255-4266
262
Sciesielski LK, Paliege A, Martinka P, Scholz H (2009):
Enhanced pulmonary expression of the TrkB neurotrophin
receptor in hypoxic rats is associated with increased acetylcholine-induced airway contractility. Acta Physiol 197(3):
253-264
Paliege A, Rosenberger C, Bondke A, Sciesielski L, Shina
A, Heyman SN, Flippin LA, Arend M, Klaus SJ, Bachmann S
(2010): Hypoxia-inducible factor-2alpha-expressing interstitial
fibroblasts are the only renal cells that express erythropoietin
under hypoxia-inducible factor stabilization. Kidney Int 77(4):
312-318
Kirschner KM, Sciesielski LK, Scholz H (2010): Wilms’ tumour
protein Wt1 stimulates transcription of the gene encoding vascular endothelial cadherin. Pflügers Arch PMID: 20811903,
Sep 02
Reviews & book chapters (2006-20010)
Scholz H, Wagner KD, Wagner N (2009): Role of the Wilms’
tumour transcription factor, Wt1, in blood vessel formation.
Pflügers Arch 458(2): 315-323
Kamkin A, Scholz H, Kiseleva I (2010): Cardiac fibroblasts
in normal and diseased heart: Single mechanically-gated
ion channels, mechanosensitive currents and mechanically
induced potentials in isolated cells and tissue. In: The car­
diac fibroblast, Neil Turner (ed.), Research Signpost
HOLGER SCHOLZ – DEVELOPMENTAL PHYSIOLOGY AND CELLULAR PATHOPHYSIOLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Scholz H.
Nafz B.
Nafz B.
Scholz H.
Scholz H.
Dame C.
Scholz H.
Fähling M.
Scholz H.
Sciesielski, L.
Jacobi C.
Scholz H.
Project title
Characterizing the oxygen-dependent
expression of the Wilms’ tumor gene, WT1,
in vivo and in vitro
Subproject “Functional genomics of cardiovascular damage in hypertension”
Mechanisms and (patho)physiological
consequences of an oxygen-dependent
regulation of the gene encoding the neurotrophin receptor, TrkB
Regulation of the genes encoding erythropoietin and its receptor by the Wilms’
tumor transcription factor, Wt1
Post-transcriptional control of the basic
helixloop-helix transcription factor Mash1
Oxygen-dependent expression of the neurotrophin receptor, TrkB.
Regulation and function of the metalloproteinase ADAMTS16
Sponsor
Deutsche Forschungsgemeinschaft (DFG SCHO 634/5-1)
Period
2004-2007
BMBF, Medical genome Research
(NGFN), KBCV 1.2, 01GS0416
Deutsche Forschungsgemeinschaft (DFG SCHO 634/6-1)
2004-2007
2008-2011
Deutsche Forschungsgemeinschaft (DFG DA 484/2-1)
2005-2006
Deutsche Forschungsgemeinschaft (DFG FA 845/2-1)
Studienstiftung des Deutschen
Volkes
Sonnenfeld-Stiftung
2008-2011
2006-2008
2010-2011
Awards
2007
Young Investigator Award:
International Congress „Hypoxia, from Integrative Biology
to Human Disease“, Monte Verità/ Ascona, Switzerland
L. Martens
2008
Poster award:
„The Wilms’ tumor transcription factor Wt1 regulates
Expression of the VE-cadherin gene.” Annual Meeting
of the German Physiological Society, Cologne
Karin Kirschner
263
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
Head of the group
Prof. Dr. med. Ralf Mrowka
Curriculum Vitae: Prof. Dr. med. Ralf Mrowka, received the ICID (Imperial College
International Diploma) in “Engineering and Physical Science in Medicine” at Imperial College London (UK) in 1993 and finished Humboldt University medical school
in 1996. He received his doctorate degree 1998 from Humboldt-University and his
habilitation degree in Physiology in 2006 from Charité – Universitätsmedizin Berlin.
In 2010 he became professor for Experimental Nephrology at Friedrich-SchillerUniversität Jena, Germany. His research interests are in the field of computational and experimental analysis
of: Gene Regulation, Protein-Protein Interactions, Gene-Expression Data, DNA-sequences, Transcription Factor
Binding Sites, miRNA function, drug based iPS. He took part in funded national collaborative networks such as
the national genome network NGFN Herz-Kreislauf-Netzwerk. Currently, he coordinates a funded collaborative project within the BMBF initiative Medical Systems Biology dealing with drug induced generation of iPS.
Members of the group
Scientist
Andreas Steege
Stephan Lorenzen
Axel Göhring
Frank Wenke Dr. rer. nat. (Guest)
Dr. rer. nat.
Dipl. Biol.
Dipl. Biochem.
Technicians
Christine Reinhold
Students
Thomas Brinkmeier
MD Student
AG Systems Biology: From left to right: S. Lorenzen,
F. Wenke, Ch. Reinhold, Th. Brinkmeier, A. Göhring,
R. Mrowka, A. Steege
265
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
Summary
We investigate questions of the physiology of the
cardiovascular system with theoretical approaches
as well as by means of tools of molecular biology.
One research focus is the renin-angiotensin-aldosterone-system (RAAS) which plays a key role in the
blood pressure regulation and water and electrolyte balance in humans. We investigate mechanisms
of gene regulation on the transcriptional as well as
posttranscriptional level such as action of proteins
and miRNAs on gene regulation. Our work contributes to the elucidation of pathophysiological mechanisms that lead to cardiovascular mortality which
is the main cause of death in Europe. Furthermore
we are interested in drug induced pluripotent stem
cells (iPS).
Zusammenfassung
Wir untersuchen mit theoretischen und molekularbiologischen Ansätzen Fragestellungen auf dem
Gebiet der Herz-Kreislauf-Physiologie. Der Fokus
liegt dabei auf dem Renin-Angiotensin-Aldosteron
System (RAAS), das eine entscheidende Rolle für
die Regulation des arteriellen Blutdrucks und den
Salz-Wasserhaushalt spielt. Eines unserer Schwerpunktprojekte ist die Regulation von Genen im
RAAS. Von Interesse sind dabei Mechanismen der
Genregulation auf transkriptioneller Ebene wie auch
fein modulierende Einflüsse, die z.B. über miRNA
vermittelt werden. Darüber hinaus sind wir an substanzinduzierter Reprogrammierung von Zellen inte-
266
ressiert (drug induced iPS). Methodisch integrieren
wir dabei verschiedene biologische Datenentitäten
wie z. B. Gen-Expressionsdaten, DNA-Sequenzinformation, Protein-Protein-Wechselwirkungen und
Ontologie-Information. Diese sind geeignet, um mit
mathematischen Modellen Voraussagen zum Beispiel über die Wirkung von Transkriptionsfaktoren
und deren Interaktion zu treffen, die wir dann mit
zellbiologischen Experimenten überprüfen. Die enge
Kombination von mathematischer Modellierung
und experimentellen Ansätzen unterstützt damit die
Erforschung von Herz-Kreislauf-Erkrankungen, die
die Haupttodesursache in Europa darstellen.
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
Research projects
1. D
rug based induction of pluripotent human stem cells
derived from human somatic cells
Project leader
Coworkers
Funding
External cooperations
Ralf Mrowka
Stephan Lorenzen, Frank Wenke, Axel Göhring, Christine Reinhold
BMBF
Max-Delbrueck-Centre for Molecular Medicine Berlin, MDC
Computational Systems Biology Group, Dr. Miguel Andrade
Max-Planck-Institute for Molecular Genetics, Molecular Embryology Group,
MPI-MEG, Dr. James Adjaye Humboldt-University Berlin, Prof. Edda Klipp
University of Heidelberg, Institute for Pharmacy and Molecular Biotechnology,
IPMB, Prof. Stefan Wölfl Genomatix Software GmbH, Munich, GENOMATIX,
Dr. Alexander Hahn European Screening Port GmbH, Hamburg, ESP, Dr. Phil Gribbon
The topic relates to the field of gene regulation and
stem cell research. The aim of the project is to exploit
large scale post genomic data entities by employing
state of the art tools of systems biology to identify
optimize and commercialise small molecule drugs that
are capable to generate pluripotent human stem cells
from adult tissue. The project addresses a highly relevant issue of modern Biology and Medicine which
has a high potential for innovation. Pluripotent human
stem cells are useful for multiple applications in transplantation medicine and tissue engineering as well
as for the generation of new disease models and in
drug development. Induced pluripotent human stem
cells that are derived from adult tissue may serve as
a complementary alternative to inner cell mass (ICM)derived embryonic pluripotent stem cells that are
highly debated regarding ethical issues. Moreover,
generation of pluripotent stem cells from an adult will
open completely new possibilities since auto transplantation of engineered tissue will not be subjected
to tissue rejection due to immune responses.
This multidisciplinary project employs methods of
systems biology, such as mathematical modelling of
biological processes in an iterative manner with wet
lab experiments. The project includes the exploitation
of high throughput data that has been obtained previously by projects that have been funded by the BMBF
to reach a high synergism of the funding schemes
of the BMBF. A main goal of the project is to obtain
chemical compounds that have the potential for commercialization.
Because of its high economic, therapeutic and scientific impact, the production of induced pluripotent
stem cell lines (iPS) is a hot topic in current biomedical
research. It has been shown recently that it is possible to induce pluripotency in somatic mouse cells by
the activation of four genes. Human somatic fibroblasts have been converted to pluripotent cell lines by
the activation of the genes Sox2, Nanog, Oct4 and
Lin28. Although it has been shown that it is in principle possible to induce pluripotency, current methods however are of very limited use, since the genes
267
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
are activated by viral transfection which is the main
hurdle for any therapeutic application and commercial exploitation. The insertion of viral genetic material
into the human genome is highly problematic for a
number of reasons. First, the viral insertion of material is not directed to a specific locus in the genome.
This is a major drawback, since it is unknown what
genes and consequently what cellular functions are
affected by this insertion. Second, the viral insertion
is irreversible, which is another major limitation. Therefore it is highly desirable to have an alternative way to
induce pluripotency. We suggest using chemical small
molecule compounds that would allow activating the
mentioned human genes that convey pluripotency in
a reversible manner. This approach makes it possible to use pluripotent human stem cells in therapeutic
contexts and would allow translating that knowledge
into products.
2. Molecular basis of the regulation of the renin gene
Project leader
Coworkers
Funding
External cooperations
Ralf Mrowka
Andreas Steege, Andreas Patzak, Christine Reinhold
Sonnenfeld Stiftung
Humboldt-University Berlin, Prof. Edda Klipp
Institute for Pathology, Dr. Nils Blüthgen
Cardiovascular diseases account for approximately 40%
of all deaths in European countries ranging from about
30.6% in Spain up to 46.8% in Greece [OECD, 2007].
The renin-angiotensin-aldosterone-system (RAAS) is
the most important system for long term regulation
of blood pressure and plays a key role in the water
and electrolyte balance of the human body. Consequently, Renin and other components of the RAAS
have been the focus of cardiovascular and related
clinical studies. Current agents targeting the RAAS
such as angiotensin-converting-enzyme (ACE) inhibitors and angiotensin II receptor antagonists and renin
antagonists are not free of side effects, however exact
numbers are sparse.
Control of RAAS gene expression is determined simultaneously at several levels, which are integrated and
inter-connected inducing complexity on both in the
individual control of gene expression (such as the
268
Renin gene itself) as well as in its interactions, due
to numerous positive and negative feedback loops at
multiple levels. How precisely RAAS gene expression
is regulated in healthy individuals, and dys-regulated in
cases of hypertension has remained an enigma, for at
least two reasons: First, studies have tended to focus
on singular causes of the disease and there has been
little emphasis on studying the systems properties of
RAAS in order to deal with the multifactorial essence
of the disease. Second, the global importance of posttranslational gene regulation by micro-RNAs has only
been appreciated in the recent years and its impact on
RAAS remains essentially unexplored, although there
is emerging evidence that this type of gene regulation impacts the RAAS. For example, interactions of
proteins with the renin 3’ untranslated region (3’ UTR)
of messenger RNA (mRNA) influences stability and
translational efficiency.
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
Integrative Systems Biology has developed as a discipline that can deal with mutifactorial processes,
through an integration of quantitative experimentation
and modelling. And, even more recently, quite a few
methodologies have become available to assess the
role of miRNAs in the regulation of gene expression.
Ultimately the progress in the understanding of RAAS
that will be generated by the project is likely to impact
our means to predict and prevent RAAS-related dysfunctions at the clinical level: Identification of novel
regulatory elements or their targets could provide new
focal points for genetic analysis in order to estimate
genetic risk factors for cardio-vascular, or even facilitate the identification of potential new drug targets.
3. Posttranskriptional Control of the HIF-1α Gen
Project leader
Coworker
Funding
Ralf Mrowka
Michael Fähling, Anja Bondke Persson, Andreas Patzak, Christine Reinhold
Intramural, funding application planned
HIF-1α is a key regulator in oxygen dependent gene
regulation. It is important in many cell functions as well
as in pathological situations such as in cancer.
Posttranscriptional processes of gene regulation in
the context of this factor are largely unexplored. This
project is dedicated to fill this gap.
Figure: Oxygen dependent signalling of HIF1-alpha. Modified
from Schofield et al.
Figure: Genomic structure of HIF-1α. There is a high evolutionary conservation in a potential regulatory region. The
figure was generated with Genome-Browser at UCSC.
269
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
4. Systematic discovery of transcription factor target genes
Project leader
Coworker
Funding
External cooperations
Ralf Mrowka
Michael Fähling, Stephan Lorenzen, Christine Reinhold
BMBF
Max-Planck-Institute for Molecular Genetics, Dr. Szymon Kiełbasa
Institute for Pathology, Dr. Nils Blüthgen
Reliable prediction of specific transcription factor target genes is a major challenge in systems biology and
functional genomics. Current sequence-based methods yield many false predictions, due to the short and
degenerated DNA-binding motifs. Here, we describe
a new systematic genome-wide approach, the seeddistribution-distance method that searches largescale genome-wide expression data for genes that
are similarly expressed as known targets. This method
is used to identify genes that are likely targets, allowing sequence-based methods to focus on a subset of
genes, giving rise to fewer false-positive predictions.
We show by cross-validation that this method is robust
in recovering specific target genes. Furthermore, this
method identifies genes with typical functions and
binding motifs of the seed.
Targetfinder.org (http://targetfinder.org/) provides a
web-based resource for finding genes that show a
similar expression pattern to a group of user-selected
genes. It is based on a large-scale gene expression
compendium (>1200 experiments, >13 000 genes).
The primary application of Targetfinder.org is to
expand a list of known transcription factor targets
by new candidate target genes. The user submits a
group of genes (the ‘seed’), and as a result the web
site provides a list of other genes ranked by similarity of their expression to the expression of the seed
270
genes. Additionally, the web site provides information on a recovery/cross-validation test to check for
consistency of the provided seed and the quality of
the ranking. Furthermore, the web site allows to analyse affinities of a selected transcription factor to the
promoter regions of the top-ranked genes in order to
select the best new candidate target genes for further
experimental analysis.
Figure: Free website of our transcription factor target finding
algorithm.
RALF MROWKA – SYSTEMS BIOLOGY – COMPUTATIONAL PHYSIOLOGY
Publications 2006 - 2010
Kiełbasa SM, Blüthgen N, Fähling M and Mrowka R (2010):
Targetfinder.org: a resource for systematic discovery of
transcription factor target genes. Nucleic Acids Res 38,
W233-238
Mrowka R, Steege A, Kaps C, Herzel H, Thiele BJ., Persson PB and Blüthgen N (2007): Dissecting the action of an
evolutionary conserved non-coding region on renin promoter
activity. Nucleic Acids Res 35, 5120-9
Fähling M, Mrowka R, Steege A, Kirschner KM, Benko E,
Förstera B, Persson PB, Thiele BJ, Meier JC and Scholz H
(2009): Translational regulation of the human achaete-scute
homologue-1 by fragile X mental retardation protein. J Biol
Chem 284, 4255-66
Fähling M, Mrowka R, Steege A, Martinka P, Persson PB and
Thiele BJ (2006): Heterogeneous nuclear ribonucleoproteinA2/B1 modulate collagen prolyl 4-hydroxylase, alpha (I)
mRNA stability. J Biol Chem 281, 9279-86
Mrowka R, Blüthgen N and Fähling M (2008): Seed-based
systematic discovery of specific transcription factor target
genes. FEBS J 275, 3178-92
Steege A, Fähling M, Paliege A, Bondke A, Kirschner KM,
Martinka P, Kaps C, Patzak A, Persson PB, Thiele BJ et al.
(2008). Wilms’ tumor protein (-KTS) modulates renin gene
transcription. Kidney Int 74, 458-66
Kralemann B, Cimponeriu L, Rosenblum M, Pikovsky A and
Mrowka R (2008): Phase dynamics of coupled oscillators
reconstructed from data. Phys Rev E 77, 066205
Kralemann B, Cimponeriu L, Rosenblum M, Pikovsky A and
Mrowka R (2007): Uncovering interaction of coupled oscillators from data. Phys Rev E 76, 055201
Lai EY, Patzak A, Steege A, Mrowka R, Brown R, Spielmann
N, Persson PB, Fredholm BB and Persson AEG (2006):
Contribution of adenosine receptors in the control of arteriolar
tone and adenosine-angiotensin II interaction. Kidney Int 70,
690-8
Fähling M, Mrowka R, Steege A, Nebrich G, Perlewitz A,
Persson PB and Thiele BJ. (2006): Translational control of
collagen prolyl 4-hydroxylase-alpha(I) gene expression under
hypoxia. J Biol Chem 281, 26089-101
Lai EY, Martinka P, Fähling M, Mrowka R, Steege A, Gericke
A, Sendeski M, Persson PB, Persson AEG and Patzak A.
(2006): Adenosine restores angiotensin II-induced contractions by receptor-independent enhancement of calcium
sensitivity in renal arterioles. Circ Res 99, 1117-24
General information
Third party funding ( 2006-2010 )
Project leader
Mrowka R.
Mrowka R.
Project title
Drug-iPS
Regulation of the Renin Gene
Sponsor
BMBF PTJ
Sonnenfeld-Stiftung
Period
2009-2011
2008-2012
271
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
Head of the group
Prof. Dr. A. Patzak
Curriculum Vitae: He studied medicine at the university in Leipzig and at the ErnstMoritz-Arndt-University in Greifswald, where he received the Diploma (Master) in
Medicine in 1982. He was authorized as a physician in 1983 and was promoted to
Dr. med. at the University of Greifswald in 1985. From 1983 he worked as a research
fellow, and after the specialization in Physiology, as a senior scientist in the Institute of
Physiology, Charité, Humboldt-University of Berlin. In 1998 he received the Habilitation in Physiology and the authorization in teaching. Andreas Patzak was guest scientist in the Department of
Physiology, University Odense, Denmark in 1998 and at the Department of Medical Cell Biology, Biomedical
Center, University Uppsala, Sweden in 2002, 2003, and 2004. He is professor in physiology since 2008. The
main fields of his interest are vessel physiology and dynamics of respiratory and cardiovascular control.
Members of the group
Scientists
Sendeski, Mauricio
Dr.
Technicians
Amoneit, Yvonne
Gerhardt, Andrea
Neumann, Ulrike
MTA (until 2009)
MTA (Ing., FH)
BTA
Students
Brinkmann, Ole
Kaufmann, Jana
Liu, Zhizhao
Moede, Olivia
Rettig-Zimmermann, Juliane
Schildroth, Janice
Schmidt, Sebastian
Da Silva, Ivana
Viegas, Vinicius MD student (degree 2008)
MD student
PhD student
MD student
MD student (degree 2009)
MD student
MD student (degree 2010)
PhD student (2009)
PhD student
273
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
Summary
Our group works about the control of renal vessels
at the organ and cell level. The kidney significantly
contributes to water and electrolyte balance of the
organism. Pre- and postglomerular arterioles are
effectors in control systems. They regulate renal
perfusion and pressure. Sympathetic nerve activity, myogenic response, tubuloglomerular feedback,
and autocrine as well as paracrine substances contribute to the adjustment of the arteriolar tone. Norepinephrine, angiotensin II, nitric oxide, adenosine
and endothelin are important players in this context.
The projects of our group focus on interactions of
angiotensin II and adenosine in the control of the
arteriolar tone. Further, we investigate the function
of endothelin receptors and the role of p38 MAPK
and reactive oxygen species in the pathogenesis
acute renal injury in models of hypoxia/reperfusion
and contrast media induced nephropathy. Results
of these studies improve our knowledge about the
function of small renal resistance vessel in renal
physiology and pathophysiology.
Zusammenfassung
Unsere Arbeitsgruppe interessiert sich vor allem für
die Regulation der Nierengefäße auf Organ- und
Zellebene. Die Niere spielt eine zentrale Rolle für
den Wasser- und Salzhaushalt des Organismus.
Wichtige Stellglieder renaler Regulationsmechanismen sind die prä- und postglomerulären Arteriolen,
welche die Nierendurchblutung und die glomeruläre
Filtrationsrate über Änderungen ihres Durchmessers
bzw. Widerstandes beeinflussen. Der Tonus dieser
Arteriolen wird durch die Aktivität sympathischer
Nerven, durch die myogene Antwort der Gefäße,
den tubuloglomerulären Rückkopplungsmechanismus sowie eine Reihe autokriner und parakriner
Substanzen bestimmt. Noradrenalin, Angiotensin II,
Stickstoffmonoxid, Adenosin und Endothelin gehören zu den wichtigen vasoaktiven Stoffen. In den
274
einzelnen Projekten unserer Gruppe beschäftigen
wir uns mit den Interaktionen von Angiotensin II
und Adenosin, mit Endothelinwirkungen und der
Funktion der Endothelinrezeptoren in glomerulären
Arteriolen sowie pathophysiologischen Aspekten.
In Modellen der Nierenhypoxie/-reoxygenierung
und der Wirkung von Röntgenkontrastmitteln auf
die Nierenfunktion wird die Rolle von p38 MAPK,
Sauerstoffradikalen und der Stickstoffmonoxidverfügbarkeit bei der Pathogenese akuter Niereninsuffizienz untersucht. Diese Arbeiten erweitern unser
Verständnis von der Regulation der Nierendurchblutung und können weitere Einblicke in die Rolle großer
und kleiner Nierengefäße z.B. bei der Entstehung
von Bluthochdruck oder der kontrastmittelinduzierten Nephropathie geben.
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
Research projects
1. Endothelin receptors in the control of glomerular arterioles
Project leader
Coworkers
Funding
A. Patzak
Janice Schildroth, Juliane Rettig-Zimmermann, Andrea Gerhardt,
Ulrike Neumann, Mauricio Sendeski
- Charité
Endothelin is produced and released by endothelial
cells. Increased endothelin concentrations induce renal
damage via different signalling pathways. Endothelin
also induce renal vascular smooth muscle cell contraction. This leads to reduction of renal perfusion and
glomerular filtration, and to volume retention, which
may contribute to the development of hypertension.
We investigate the function and signalling of endothelin
receptors in glomerular arterioles using mice lacking
the endothelin type receptor („rescued ETB-knock
out“). The study shows that endothelin type A receptors (ETA-R) mediates vasoconstriction in afferent
arterioles and contribute to vasoconstriction in efferent arterioles. In contrast, ETB-R have no effect in
afferent but mediates basal nitric oxide release and
constriction in efferent arterioles. The stronger effect of
endothelin-1 on afferent arterioles compared to efferent arterioles indicates an influence on glomerular filtration rate and a contribution to the pathogenesis of
kidney diseases.
Fig.1 The selective ETB-receptor agonist
ALA-ET-1 constricts efferent arterioles
of wild type mice. There is no effect in
arterioles of ETB-deficient mice. A) Absolute diameter in µm. B) Relative changes in
relation to the basal diameter, * indicates
significant differences between both
groups.
275
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
2. R
eactive ox ygen species and vascular smooth muscle
contraction – intracellular pathways
Project leader
Coworkers
Funding
External cooperations
A. Patzak
Sebastian Schmidt, Mauricio Sendeski Olga Zavaritskaya, Ulrike Neumann,
Ivana da Silva
- DAAD
Dr. Olaf Grisk, Dr. Torsten Günther, Ernst Moritz-Arndt-Universität Greifswald,
Dr. Vera and Prof. Dr. Joachim Jankowski, Nephrology, Charité,
Campus Benjamin Franklin
Oxidative stress plays a role in the development and
maintenance of cardiovascular dysfunction such as in
hyperlipidemia, ischemic heart disease, heart failure,
and probably hypertension. Reactive oxygen species
(ROS) affect vessel function by several mechanisms,
for example by decreasing the bioavailability of nitric
oxygen („scavenger effect“). Oxidative stress may
also contribute to the process of aging. We hypothesize that ROS increase the contractility of mesenterial arteries and that this effect is dependent on age.
In this project, we investigate the mechanisms and
intracellular pathways of such an effect. Pilot studies
demonstrate an increase of the contraction to angiotensin II in adult mice after stimulation of the NADPHoxidase (Fig. 2). This effect was completely prevented
by treatment with the p38 MAPK inhibitor SB 220025,
suggesting an important role of this enzyme in the
mediation of the NADPH-induced increase of the angiotensin II response.
Fig. 2: Stimulation with NADPH, the substrate for the NADPH
oxidase increases the contraction to angiotensin II (Ang II)
in mesenteric vessels of adult mice (old). The selective p38
MAPK inhibitor SB 220025 prevents this effect. ** indicates
significant differences in the course of the concentrationsresponse-curve of NADPH-treated compared to non-treated
(NADPH) and SB 220025 treated vessels, respectively.
276
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
3. Contrast media and function of descending vasa recta
Project leader
Coworkers
Funding
External cooperations
A. Patzak
Mauricio Sendeski, Zhizhao Liu, P.B. Persson, Yvonne Amoneit, Andreas Gerhardt
- Dr. Werner Jackstädt-Stiftung (to M. Sendeski)
- Else Kröner-Fresenius-Stiftung (to A. Patzak)
- Government of People’s Republic of China
Prof. Dr. Tom Pallone, University of Maryland, Baltimore, USA
Prof. Dr. A.E.G. Persson, University Uppsala, Sweden
Dr. Vera and Prof. Dr. Joachim Jankowski, Nephrology, Charité,
Campus Benjamin Franklin
The contrast media induced nephropathy (CIN) is
the third most common reason for hospital-acquired
acute kidney injury. The renal damage goes along with
a reduction in renal medullary blood flow and oxygen tension. We hypothesize that iodinated contrast
media (CM) affect small renal vessels supplying the
renal medulla, namely the arterioles of juxtamedullary
nephrons and the descending vasa recta. Contrast
media may also damage tubular epithelial cells. These
cells then release vasoactive substances, which in turn
constrict arterioles and vasa recta and reduce renal
medullary perfusion. Our project focuses on the effect
of CM on the tone and vasoreactivity of small renal
vessels. CM probably induce oxidative stress in the
wall of the vessels and in tubular cells thereby reducing the bioavailability of the vessel dilator nitric oxygen
(Fig. 3). We assume that signalling pathways involving
the p38 MAPK also mediate the increase in vascular
tone and reactivity.
Fig. 3 Increase of the fluorescence of DAF‑FM, an indicator of nitric oxygen, after application of angiotensin II in
an isolated perfused vas rectum of the rat (pseudo color
presentation of fluorescence intensity).
277
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
4. I nfluence of sex hormones on the reactivity of interlobar
arteries in the mouse - role of nitric oxide
Project leader
Coworkers
Funding
A. Patzak
Pontus B. Persson, Vinicius Viegas, Mauricio Sendeski, Ulrike Neumann,
Andrea Gerhardt
- DFG GK754-III
Women have lower blood pressure until menopause
in comparison with men. This phenomenon is probably due to differences in the hormone status between
women and men. It has been shown in animal experiments that estrogens have protective effects on blood
vessels. Also, mesenterial arteries of female mice
express lower vascular tone compared to male animals. The differences in vascular tone disappear in
superoxid dismutase deficient mice, suggesting an
important role for reactive oxygen species and nitric
oxide in this context.
Nitrite from food and from other sources may act as
substrate fort the generation of nitric oxide. Studies
show a reduction of blood pressure after uptake of
nitrate in form of vegetables. The mechanisms of
this effect are not completely known. In the present
project, we ask whether nitric oxide produced from
nitrite plays a role fort the differences of vascular tone
seen in female compared to male mice. We also test
a possible protective effect of nitrite in vessels of male
animals.
Fig. 4: Original traces of vessel
tension (renal interlobar artery) in
response to increasing doses of
estrogen (upper trace) and testosterone (lower trace).
278
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
Publications 2006 - 2010
Sendeski MM, Consolim-Colombo FM, Leite CC, Rubira MC,
Lessa P, Krieger EM (2006): Increased sympathetic nerve
activity correlates with neurovascular compression at the
rostral ventrolateral medulla. Hypertension 47:988-995
Lai EY, Martinka P, Fahling M, Mrowka R, Steege A, Gericke
A, Sendeski M, Persson PB, Persson AE, Patzak A (2006):
Adenosine restores angiotensin II-induced contractions by
receptor-independent enhancement of calcium sensitivity in
renal arterioles. Circ Res 99: 1117-1124
Lai EY, Patzak A, Steege A, Mrowka R, Brown R, Spielmann
N, Persson PB, Fredholm BB, Persson AE (2006): Contribution of adenosine receptors in the control of arteriolar tone
and adenosine-angiotensin II interaction. Kidney Int 70:
690-698
Gericke A, Martinka P, Nazarenko I, Persson PB, Patzak
A (2007): Impact of alpha1-adrenoceptor expression on
contractile properties of vascular smooth muscle cells. Am J
Physiol Regul Integr Comp Physiol 293: R1215-R1221
Hultstrom M, Lai EY, Ma Z, Kallskog O, Patzak A, Persson AE
(2007): Adenosine triphosphate increases the reactivity of the
afferent arteriole to low concentrations of norepinephrine. Am
J Physiol Regul Integr Comp Physiol 293: R2225-R2231
Lai EY, Jansson L, Patzak A, Persson AE (2007): Vascular
reactivity in arterioles from normal and alloxan-diabetic mice:
studies on single perfused islets. Diabetes 56: 107-112
Patzak A, Lai EY, Fahling M, Sendeski M, Martinka P, Persson
PB, Persson AE (2007): Adenosine enhances long term the
contractile response to angiotensin II in afferent arterioles. Am
J Physiol Regul Integr Comp Physiol 293: R2232-R2242
Petersson J, Schreiber O, Steege A, Patzak A, Hellsten A,
Phillipson M, Holm L (2007): eNOS involved in colitis-induced
mucosal blood flow increase. Am J Physiol Gastrointest
Liver Physiol 293: G1281-G1287
Petersson J, Phillipson M, Jansson EA, Patzak A, Lundberg
JO, Holm L (2007): Dietary nitrate increases gastric mucosal
blood flow and mucosal defense. Am J Physiol Gastroin­
test Liver Physiol 292: G718-G724
Carlstrom M, Lai EY, Steege A, Sendeski M, Ma Z, Zabihi
S, Eriksson UJ, Patzak A, Persson AE (2008): Nitric oxide
deficiency and increased adenosine response of afferent
arterioles in hydronephrotic mice with hypertension. Hyper­
tension 51: 1386-1392
Jankowski V, Patzak A, Herget-Rosenthal S, Tran TN, Lai EY,
Gunthner T, Buschmann I, Zidek W, Jankowski J (2008): Uridine adenosine tetraphosphate acts as an autocrine hormone
affecting glomerular filtration rate. J Mol Med 86: 333-340
Martinka P, Lai EY, Fahling M, Jankowski V, Jankowski J,
Schubert R, Gaestel M, Persson AE, Persson PB, Patzak A (2008): Adenosine increases calcium sensitivity via
receptor-independent activation of the p38/MK2 pathway in
mesenteric arteries. Acta Physiol (Oxf) 193: 37-46
Nordquist L, Lai EY, Sjoquist M, Patzak A, Persson AE (2008):
Proinsulin C-peptide constricts glomerular afferent arterioles
in diabetic mice. A potential renoprotective mechanism. Am J
Physiol Regul Integr Comp Physiol 294: R836-R841
Patzak A, Steege A, Lai EY, Brinkmann JO, Kupsch E, Spielmann N, Gericke A, Skalweit A, Stegbauer J, Persson PB,
Seeliger E (2008): Angiotensin II response in afferent arterioles of mice lacking either the endothelial or neuronal isoform
of nitric oxide synthase. Am J Physiol Regul Integr Comp
Physiol 294: R429-R437
Steege A, Fahling M, Paliege A, Bondke A, Kirschner KM,
Martinka P, Kaps C, Patzak A, Persson PB, Thiele BJ, Scholz
H, Mrowka R (2008): Wilms’ tumor protein (-KTS) modulates
renin gene transcription. Kidney Int 74: 458-466
Stegbauer J, Kuczka Y, Vonend O, Quack I, Sellin L, Patzak
A, Steege A, Langnaese K, Rump LC (2008): Endothelial
nitric oxide synthase is predominantly involved in angiotensin
II modulation of renal vascular resistance and norepinephrine
release. Am J Physiol Regul Integr Comp Physiol 294:
R421-R428
Carlstrom M, Lai EY, Ma Z, Patzak A, Brown RD, Persson AE
(2009): Role of NOX2 in the regulation of afferent arteriole
responsiveness. Am J Physiol Regul Integr Comp Physiol
296: R72-R79
279
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
Gericke A, Mayer VG, Steege A, Patzak A, Neumann U,
Grus FH, Joachim SC, Choritz L, Wess J, Pfeiffer N (2009):
Cholinergic responses of ophthalmic arteries in M3 and M5
muscarinic acetylcholine receptor knockout mice. Invest
Ophthalmol Vis Sci 0: 4822-4827
Kastner C, Pohl M, Sendeski M, Stange G, Wagner CA,
Jensen B, Patzak A, Bachmann S, Theilig F (2009): Effects of
receptor-mediated endocytosis and tubular protein composition on volume retention in experimental glomerulonephritis.
Am J Physiol Renal Physiol 296: F902-F911
Kovacs R, Rabanus A, Otahal J, Patzak A, Kardos J, Albus
K, Heinemann U, Kann O (2009): Endogenous nitric oxide is
a key promoting factor for initiation of seizure-like events in
hippocampal and entorhinal cortex slices. J Neurosci 29:
8565-8577
Lai EY, Fahling M, Ma Z, Kallskog O, Persson PB, Patzak A,
Persson AE, Hultstrom M (2009): Norepinephrine increases
calcium sensitivity of mouse afferent arteriole, thereby
enhancing angiotensin II-mediated vasoconstriction. Kidney
Int 76: 953-959
Lai EY, Patzak A, Persson AE, Carlstrom M (2009): Angiotensin II enhances the afferent arteriolar response to adenosine through increases in cytosolic calcium. Acta Physiol
(Oxf) 196: 435-445
Sendeski M, Patzak A, Pallone TL, Cao C, Persson AE,
Persson PB (2009): Iodixanol, constriction of medullary
descending vasa recta, and risk for contrast medium-induced
nephropathy. Radiology 251: 697-704
280
Cao C, Edwards A, Sendeski M, Lee-Kwon W, Cui L, Cai CY,
Patzak A, Pallone TL (2010): Intrinsic nitric oxide and superoxide production regulates descending vasa recta contraction.
Am J Physiol Renal Physiol (ahead of print)
Schildroth J, Rettig-Zimmermann J, Kalk P, Steege A, Fahling
M, Sendeski M, Paliege A, Lai EY, Bachmann S, Persson PB,
Hocher B, Patzak A (2010): Endothelin type A and B receptors in the control of afferent and efferent arterioles in mice.
Nephrol Dial Transplant (ahead of print)
Sendeski M, Patzak A, Persson PB (2010): Constriction of
the vasa recta, the vessels supplying the area at risk for acute
kidney injury, by four different iodinated contrast media, evaluating ionic, nonionic, monomeric and dimeric agents. Invest
Radiol 45: 453-457
Carlstrom, M, Lai EY, Ma Z, Steege A, Patzak A, Eriksson U J,
Lundberg J O, Wilcox C S, and Persson A E G (2010): SOD1
limits renal microvascular remodeling and attenuates arteriole
and blood pressure responses to Ang II via modulation of NO
bioavailability. Hypertension (accepted)
Reviews & book chapters (2006-20010)
Patzak A, Persson AE (2007): Angiotensin II-nitric oxide
interaction in the kidney. Curr Opin Nephrol Hypertens 16:
46-51
ANDREAS PATZAK – VASCULAR PHYSIOLOGY
General information
Third party funding ( 2006-2010 )
Project leader
Patzak A.
Sendeski M.
Patzak A. Persson
P.B. Jankowski V.
Liu Z.
Project title
Sex- and Gender- Specific Mechanisms
in Myocardial Hypertrophy
Influence of contrast media on the control
of outer medullary descending vasa recta
Contrast media and oxidative stress in the
renal medulla.
Effect of contrast media on the function
of the juxtaglomerular apparatus
Sponsor
DFG GK 754-III, GK 754-II,
Period
2006-2010
Dr. Werner-Jackstädt-Stiftung
2007-2008
Else Kröner Fresenius-Stiftung
2010-2013
Government of People’ Republic 2009-2013
of China
Awards
2008
Poster award of the German Society for
Sleep Medicine and Sleep Research
Patzak A, Gesche H
2009
„Postdoctoral Novel Disease Model Award“,
American Physiological Society
Sendeski M
281
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
Head of the group
Dr. med. Erdmann Seeliger
Curriculum Vitae: Erdmann Seeliger studied medicine at Berlin’s Charité, and
obtained his license to practise medicine (Approbation) in 1988. He joined the Institute of Physiology at the Charité for his postgraduate specialisation and received his
board certification as a specialist in Physiology (Facharzt für Physiologie) in 1993.
In his thesis (Dr. med.), he focussed on cardiovascular control in an animal model
of cardiomyopathy. In 1993, he joined the Experimental Anaesthesia group at the
Virchow Clinics of Berlin’s Free University; the group studied the interrelationship between control of sodium
and water balance and long-term control of arterial blood pressure. Seeliger, who returned to the Institute of
Physiology at the Charité in 1998, continued these studies until 2006. He then joined the Integrative Kidney
Research group at the Institute of Vegetative Physiology; in 2009, he became head of the group. By use of
integrative experimental methods, the group studies renal hemodynamics and oxygenation under physiological conditions, and pathophysiological mechanisms involved in kidney diseases like contrast media induced
nephropathy, diabetic nephropathy, and renal ischemia/reperfusion injury.
Members of the group
Scientists
Cantow, Kathleen
Flemming, Bert
Ladwig, Mechthild
Sargsyan, Lilit
Wronski, Thomas
Dr. med. vet.
OA Dr. med. (guest scientist,
head of the group till 2009)
Veterinarian
Dr. med.
Dr. rer. nat. (guest scientist)
Technicians
Anger, Ariane Gerhardt, Andrea
BTA
MTA, Dipl.-Ing. (FH)
283
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
Summary
From a physiological point of view, the kidney is a
special organ, not just because of the functional
role that it plays for the organism, but because of
the unique and very complex interactions among
perfusion, glomerular filtration, tubular resorption,
and hormonal control. Unfortunately, such interactions also play pivotal roles in the pathophysiological
development of various kidney diseases.
Renal hemodynamics offer striking differences to
hemodynamics of other vascular beds. Whereas
metabolic necessities and oxygen consumptions
determine perfusion in other organs, in the kidney
it is the other way round: Renal oxygen consumption is largely determined by perfusion, because an
increase in renal blood flow (RBF) that is accompanied, as a rule, by an increase in glomerular filtration
rate (GFR) necessitates an increase in energy- and
oxygen-dependent tubular resorption. Although
whole-kidney blood flow is high and the arteriovenous difference in oxygen content is low, there
exist certain regions in the kidney with low partial
pressure of oxygen (pO2). Meagre pO2 in these
kidney areas is due to the particular architecture of
intrarenal vasculature that enables shunt-diffusion of
oxygen both in the medulla and in the cortex.
The control of renal hemodynamics as well as of
tubular resorption involves a number of factors, e.g.
nitric oxide (NO), angiotensin II (Ang II), adenosine (Ado), among others. Furthermore, the kidney
is equipped with highly efficient mechanisms of
autoregulation, i.e., the ability to maintain RBF and
GFR relatively constant in the face of changes in
renal perfusion pressure (RPP). Renal autoregulatory mechanisms include the myogenic mechanism,
the tubulo-glomerular feedback (TGF), and a third
284
mechanism (3M) that has been discovered only
recently. Both the TGF and the 3M seem to serve
the purpose of balancing oxygen delivery and, thus,
RBF with metabolic and oxygen demands arising
from tubular resorption. The outer medulla is particularly prone to imbalance between oxygen delivery
and demand, because, in this region, the oxygen
demand is high while the pO2 is low. Imbalance
between medullary oxygen delivery and oxygen
demand is widely accepted to constitute a pivotal
element in acute kidney injury (AKI) of various origins
(e.g., contrast-medium induced nephropathy [CIN],
ischemia-reperfusion injury [I/R-I]) as well as in some
chronic kidney diseases, e.g., diabetic nephropathy
(DNP).
For a couple of years, our group’s focus has been
on regulation and autoregulation of global (total
renal blood flow) and regional (cortical and medullary) hemodynamics and oxygenation under various
physiological and pathophysiological conditions. In
this context, we developed a number of new methods for integrative in vivo studies on the regulation
of kidney hemodynamics and oxygenation in rats.
This includes the simultaneous measurements of
local perfusion and oxygenation, and a number of
specific reversible tests, that allow us comprehensive quantitative characterisation of regulation and
autoregulation.
By use of these methods, we study 1) the contribution of certain regulatory elements like the NOsystem, Ang II, and Ado to control of hemodynamics
and oxygenation under physiological conditions, 2)
pathophysiological mechanisms involved in kidney
diseases like CIN, I/R-I, and DNP, and 3) pharmacological and procedural measures that may enable
prevention or treatment of these diseases.
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
Zusammenfassung
Was die Niere aus physiologischer Sicht zu einem
besonderen Organ macht, ist nicht allein ihre Funktion für den Organismus, sondern sind vor allem die
einzigartigen und komplexen Wechselwirkungen
zwischen Perfusion, glomerulärer Filtration, tubulärer Resorption und hormoneller Regulation. Leider spielen solche Wechselwirkungen auch in der
pathophysiologischen Entwicklung vieler Nierenerkrankungen eine zentrale Rolle.
Die Durchblutung der Niere weist gegenüber anderen
Organen bedeutende Unterschiede auf. Das Stromzeitvolumen ist gemessen am Sauerstoffverbrauch
sehr hoch. Während bei anderen Organen die aktuelle Stoffwechsellage und damit der O2-Verbrauch
die lokale Durchblutung bestimmt, bestimmt bei
der Niere umgekehrt die aktuelle Durchblutung den
Stoffwechsel und damit den O2-Verbrauch, da ein
erhöhter renaler Blutfluss (RBF) und eine i. d. R. damit
verbundene erhöhte glomeruläre Filtrationsrate (GFR)
eine vermehrte tubuläre Resorption erfordern. Trotz
hoher Gesamtdurchblutung und niedriger arteriovenöser Differenz des O2-Gehaltes gibt es Nierenregionen mit sehr niedrigem O2-Partialdruck (pO2).
Ursache ist die besondere Gefäßarchitektur der
Niere, die arterio-venöse O2-Shuntdiffusionen in der
Nierenrinde und im Nierenmark zur Folge hat.
An der Regulation der Nierenhämodynamik wie auch
der tubulären Resorption sind eine Reihe von Faktoren beteiligt, u. a. Stickstoffmonoxid (NO), Angiotensin II (Ang II), und Adenosin (Ado). Die Niere verfügt
darüber hinaus über Mechanismen der Autoregulation, d. h. einer relativen Konstanz von RBF und GFR
bei Änderung des renalen Perfusionsdruckes. Dazu
gehört der myogene Mechanismus, der tubuloglomeruläre Feedback (TGF), und ein dritter, erst
neu entdeckter, langsamer Mechanismus (3M), der
vermutlich eine metabolische Komponente darstellt.
Für TGF und 3M wird postuliert, dass sie vor einem
Missverhältnis zwischen Resorptions-Notwendigkeit
und niedrigem O2-Angebot schützen sollen. Prädestiniert für ein solches Missverhältnis ist der dicke
aufsteigende Teil der Henleschen Schleife, der mit
seinem hohen O2-Bedarf in einer Region mit niedrigem pO2 lokalisiert ist. Ein solches Missverhältnis
wird als zentrales Element in der Pathogenese verschiedener Nierenerkrankungen wie der Röntgenkontrastmittel-induzierten Nephropathie (CIN), der
Ischämie/Reperfusions-Schädigung (I/R-I) und der
diabetischen Nephropathie (DNP) angesehen.
Seit längerem befasst sich unsere Arbeitsgruppe mit
der Regulation und Autoregulation der globalen (renaler Blutfluss) und regionalen (kortikalen und medullären)
Hämodynamik und Oxygenierung unter physiologischen wie auch pathophysiologischen Bedingungen. In
diesem Rahmen haben wir auch eine Reihe von neuen
Methoden für Untersuchungen zur Regulation von renaler Hämodynamik und Oxygenierung an Ratten in vivo
etabliert. Dazu gehören u. a. die gekoppelte Messung
von lokalen Perfusions- und Oxygenierungsgrößen und
eine Reihe spezifischer reversibler Testreize, die uns eine
umfassende quantitative Charakterisierung der Regulation und Autoregulation erlauben.
Mittels dieser Methoden untersuchen wir zum einen
den Beitrag bestimmter Regulationselemente, wie
z.B. des NO-Systems, des Ang II und des Ado, zu
Regulation und Autoregulation von renaler Hämodynamik und Oxygenierung unter physiologischen
Bedingungen, zum anderen pathophysiologische
Mechanismen bestimmter Nieren-Erkrankungen wie
der CIN, der I/R-I und der DNP, sowie pharmakologische und prozedurale Möglichkeiten der Prävention
bzw. Therapie dieser Erkrankungen.
285
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
Research projects
1. D
irect effects of Glucagon-like peptide 1 on renal hemodynamics
and ox ygenation – animal studies into potential prevention of
diabetic nephropathy
Project leader
Coworkers
Funding
External cooperations
Erdmann Seeliger
Bert Flemming, Mechthild Ladwig, Kathleen Cantow, Lilit Sargsyan, Andrea Gerhardt,
Ariane Anger
DFG SE 998/2-1
Michael Fähling (AG Molecular Physiology)
Patients who suffer from diabetes mellitus (DM) often
develop renal impairment, i.e., diabetic nephropathy
(DNP). The mechanisms that contribute to the development of DNP are not entirely clear, however, two
factors appear to play a pivotal role: oxygen deficiency in the renal medulla, and deficiency of nitric
oxide (NO). Glucagon-like peptide 1 (GLP-1) facilitates the release of insulin, thereby reducing glucose
concentration. Moreover, GLP-1 has effects that are
independent from glucose homeostasis. In the kidney,
GLP-1 receptors have been found that could mediate a reduction in tubular sodium resorption. We have
established methods for integrative in vivo studies on
286
the regulation of kidney hemodynamics and oxygenation in rats. Previous studies of ours demonstrated that
kidney-specific interactions among perfusion, tubular
resorption, and oxygenation play an outstanding role
in the development of other forms of nephropathy. The
present project aims at studying possible direct effects
of GLP-1 on renal hemodynamics and oxygenation,
both in healthy rats and in a rat model of DM. We
want to assess potential glucose-independent renoprotective effects of GLP-1 in DNP, and clarify the roles
renal oxygen deficiency and NO deficiency play in this
context.
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
2. C
ontrast-media induced nephropathy: role of physico-chemical
properties of contrast media
Project leaders
Coworkers
Funding
External cooperations
Bert Flemming, Erdmann Seeliger, Pontus B. Persson
Mechthild Ladwig, Thomas Wronski, Andrea Gerhardt
Bayer Schering Pharmaceuticals
Klaus Becker (Zürich University)
Interventional procedures such as cardiac catheterization often require large amounts of contrast media
(CM). Although available CM are well tolerated,
they potentially can cause kidney damage (contrast
medium induced nephropathy, CIN). Osmolality and
viscosity are important physico-chemical properties
of CM, that have been suggested to play a role in
CIN. Therefore, we instrumented anaesthetized rats to
assess either renal hemodynamics and oxygenation
and hindquarter hemodynamics, or urine flow rate,
urine viscosity, and glomerular filtration rate (GFR). A
contrast medium with higher osmolality (iopromide)
was compared to a contrast medium with higher viscosity (iodixanol) at the same iodine concentrations
(320 mgI/ml). The effects of mannitol (same osmolality
as iopromide) and dextran (same viscosity as iodixanol) were also studied. Kidney functions are pressure dependent, thus, pressure dependent effects
on renal hemodynamics were investigated. Also the
tubuloglomerular feedback response, a crucial controller of renal hemodynamics, was assessed. Both
contrast media transiently increased renal blood flow,
and, even more, hindquarter blood flow. Iopromide
and mannitol increased urine flow rate much more
than iodixanol. Iodixanol led to a dramatic increase in
urine viscosity and to marked transient decrease in
GFR. Iopromide increased urine viscosity much less
and did not affect GFR. Only iodixanol and dextran (the
high viscous substances) decreased blood flux, erythrocyte concentration and pO2 in the renal medulla.
Reduction in renal medullary blood flux by iodixanol
was most pronounced at renal perfusion pressure
above 60 mmHg. Moreover, iodixanol prolonged the
tubuloglomerular feedback response. In conclusion,
iodixanol, by virtue of its high viscosity, reduced blood
flux, erythrocyte concentration and pO2 of the renal
medulla and decreases GFR, by increasing tubular
fluid viscosity, and, probably also by increasing plasma
viscosity. Iopromide, by virtue of its higher osmolality,
prevented excessive urine viscosity levels, medullary
hypoperfusion and hypoxia, and decline in GFR.
287
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
3. R
enal safety of contrast media : mechanisms
and prevention strategies
Project leaders
Coworkers
Funding
Erdmann Seeliger, Pontus B. Persson
Bert Flemming, Mechthild Ladwig, Kathleen Cantow, Lilit Sargsyan,
Andrea Gerhardt, Ariane Anger
Bayer Schering Pharmaceuticals
Current medical diagnosis and thereby therapeutic
success critically depends on imaging techniques
using radiocontrast media. However, the application of
contrast media (CM) may cause severe complications
such as contrast media induced nephropathy (CIN).
Previous results of ours indicate that CM may cause
renal medullary hypoperfusion and hypoxia, an effect
that is related to rheological properties of CM, in particular to CM viscosity. High CM viscosity may cause
medullary hypoperfusion by two mechanisms. Along
the passage of blood through the vasa recta supplying
the renal medulla, water escapes while CM cannot follow, which will increase blood viscosity, thereby hindering perfusion. Likewise, water is reabsorbed along the
tubules, whereas CM is not, which increases tubular
fluid viscosity, thereby hindering tubular fluid flow. The
latter will hinder glomerular filtration, in addition, it will
prolong contact time of CM, thus, toxic effects, such
as vasoconstriction of vasa recta will be pronounced.
288
We found that the renal tubular concentration process concentrates high-viscous iso-osmolar CM to a
great extent, thus, in non-hydrated rats, tubular fluid
viscosity dramatically increases and GFR declines.
CM of higher osmolality (so called low-osmolar CM),
on the other hand, bear the advantage of preventing
excessive urine viscosity levels. In the present project,
we set out to further underscore the significance of
renal fluid viscosity in CIN. The key question to be
answered is if CM are retained in the kidney for longer
periods of time, particulary iso-osmolar high-viscous
CM. Furthermore, heavy ongoing debate on the pros
and cons of fluids best suited in preventing CIN reflects
the need to understand the basic mechanisms of various fluid expansion regimes. We set out to study different regimen in the rat. Finally, we seek to elucidate
the mechanism(s) behind the flush-phenomenon after
giving CM.
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
4. A
cute kidney injury, renal hemodynamics and potential
benefit of nitrite
Project leaders
Coworkers
Funding
External cooperations
Erdmann Seeliger, Bert Flemming, Pontus B. Persson
Mechthild Ladwig, Kathleen Cantow, Lilit Sargsyan, Andrea Gerhardt, Ariane Anger
Grant application for DFG FOR 1368/1
D. Dragun, U. Hoff (Nephrology, Charité)
Dr. rer. nat. W.-H. Schunck (MDC Berlin)
Renal outer medullary hypoxia plays a pivotal role
in acute kidney injury (AKI). Reduced bioavailability
of vasodilatory nitric oxide (NO) directly promotes
hypoperfusion and hypoxia; in addition, it disinhibits
the synthesis of vasoconstrictive 20-hydroxyeicosatetraenoic acid (20-HETE). According to a new paradigm of hypoxic vasodilation, hemoglobin (Hb) is a
regulated nitrite reductase. Hb reduces nitrite to NO as
it deoxygenates, thus causing vasodilation in hypoxic
regions. We hypothesize that, in the setting of AKI, low
dose nitrite may result in renal medullary vasodilation.
As nitrite is reduced to NO in hypoxic areas only, it may
constitute a reasonably specific measure to prevent
AKI. We have established methods for integrative in
vivo studies on regulation of kidney hemodynamics
and oxygenation in rats, and applied these methods
to study contrast medium induced nephropathy (CIN)
and renal ischemia/reperfusion (I/R) injury. The project
that is part of an application for a DFG Forschergruppe, aims at studying the role of nitrite-derived NO
for hemodynamics and oxygenation in rat models of
CIN and of (I/R) injury. In close cooperation with other
participants of the Forschergruppe, we will also study
the role of 20-HETE in I/R injury.
289
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
Publications 2006 - 2010
Seeliger E, Ladwig M, Reinhardt HW (2006): Are large
amounts of sodium stored in an osmotically inactive form
during sodium retention? Balance studies in freely moving
dogs. Am J Physiol Regul Integr Comp Physiol 290:
R1429-R1435
Seeliger E, Wronski T, Ladwig M, Dobrowolski L, Vogel
T, Godes M, Persson P, Flemming B (2009): The reninangiotensin system and the third mechanism of renal
blood flow autoregulation. Am J Physiol Renal Physiol
296:F1334-F1345
Liss P, Persson PB, Hansell P, Lagerquist B (2006): Renal
failure in 57925 patients undergoing coronary procedures
using iso-osmolar or low-osmolar contrast agents. Kidney
Int 70: 1811-1817
Seeliger E, Becker K, Ladwig M, Wronski T, Persson PB,
Flemming B (2010): Up to 50-fold increase in urine viscosity
by iso-osmolar contrast media in the rat. Radiology 256:406414
Stauss HM, Persson PB (2006): Cardiovascular variability
and the autonomic nervous system. J Hypertension 24:
1902-1905
Seeliger E, Lunenburg T, Ladwig M, Reinhardt HW (2010):
Role of the renin-angiotensin-aldosterone system for control
of arterial blood pressure following moderate deficit in totalbody sodium – balance studies in freely moving dogs. Clin
Exp Pharmacol Physiol 37:e43-e51
Seeliger E, Flemming B, Wronski T, Ladwig M, Arakelyan K,
Godes M, Möckel M, Persson PB (2007): Viscosity of contrast
media perturbs renal hemodynamics. J Am Soc Nephrol
18:2912-2920
Bräutigam M, Persson PB (2006): Do iodinated contrast
media interfere with renal tubular creatinine secretion? Radi­
ology 240: 615
Persson PB, Liss P, Hansell P (2007): Evaluation and comparison between visipaque (Iodixanol) and hexabrix (Ioxaglate)
in coronary angiography. J Am Coll Cardiol 49:1668-1671
Patzak A, Steege A, Lai EY, Brinkmann O, Kupsch E, Spielmann N, Gericke A, Skalweit A, Stegbauer J, Persson PB,
Seeliger E (2008): Angiotensin II response in afferent arterioles of mice lacking either the endothelial or neuronal isoform
of nitric oxide synthase. Am J Physiol Regul Integr Comp
Physiol 294:R429-437
Hoff U, Lukitsch I, Chaykovska L, Ladwig M, Arnold C, Manthati VL, Fuller FT, Schneider W, Gollasch M, Müller DN, Flemming B, Seeliger E, Luft FC, Falck JR, Dragun D, Schunck WH
(2010): Inhibition of 20-HETE synthesis and action protects
from renal ischemia/reperfusion injury. Kidney Int in press
Reviews & book chapters (2006-2010)
Persson PB (2006): Pathophysiology of contrast-induced
nephropathy. In: Contrast-Induced Nephropathy; eds.
Bartorelli AL, Marenzi G; Taylor & Francis Group, London, pp
3-17
Persson PB, Tepel M (2006): Contrast medium-induced
nephropathy. Kindney Int Suppl. 100:S8-S10
Persson PB (2006): Dimeric contrast media: A true leap
ahead? EJHPPractice 12: 73-74
290
ERDMANN SEELIGER – INTEGRATIVE KIDNEY RESEARCH
General information
Third party funding ( 2006-2010 )
Project leader
Seeliger E.
Flemming B.
Persson P.B.
Seeliger E.
Persson P.B.
Flemming B.
Persson P.B.
Seeliger E.
Persson P.B.
Flemming B.
Persson P.B.
Seeliger E.
Patzak A.
Persson P.B.
Seeliger E.
Seeliger E.
Persson P.B.
Project title
Stellenwert von Nierennerven, Blutdruckoszillationen und Drucknatriurese
bei der Regulation des Natriumbestandes und des Blutdruckes – Bilanzuntersuchungen an wachen Hunden
Effects of SLV320 and furosemide on the
autoregulation of renal blood flow and
renal tissue oxygen concentration
Einfluss maternaler Varianten des Gens
der endothe-lialen NO-Synthase (eNOS)
auf die Programmierung von Herz-Kreislauferkrankungen in der F1-Generation
Contrast medium induced nephropathy:
Prevention of acute effects of iodixanol
by SLV320
Contrast medium induced nephropathy:
importance of contrast medium viscosity
Contrast medium induced nephropathy:
Iopromide 370, Iodixanol, Iopromide 370
+ osmodiuresis
Contrast medium induced nephro-pathy:
modulation of acute effects of Iodixanol
by the Adenosine A1-agonist, BR-4461
Renal hemodynamic effects of the contrast media, Iodixanol and Iopromide
Sponsor
DFG SE 998/1-3
Period
2003-2006
Solvay Pharma-ceuticals GmbH
2003-2006
DFG PE 388/20-1
2005-2008
Solvay Pharma-ceuticals GmbH
2005-2008
Schering AG
2005–2009
Bayer-Schering Pharmaceuticals
2007-2009
Bayer Healthcare
2007-2010
Bayer-Schering Pharmaceuticals
2008-2010
DFG SE 998/2-1
Direkte Wirkungen von Glukagon-like
peptide 1 auf renale Hämodynamik und
Oxygenierung – tierexperimentelle Untersuchungen zur potentiellen Prävention
der diabetischen Nephropathie
Renal safety of contrast media: mecha- Bayer-Schering Pharmaceuticals
nisms and prevention strategies
2009-2012
2010-2011
291
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Head of the group
Dr. rer. nat. Michael Fähling
Prof. Dr. rer. nat. Bernd J. Thiele (until Oct. 2009)
PD Dr. med. Benno Nafz (until April 2010)
Curriculum Vitae: Michael Fähling started his carrier 1996-2001 as a student at the Free
University of Berlin, Faculty of Natural Sciences with the special subject Molecular Biology/
Genetics. He obtained his PhD degree in 2004 as a member of the Graduate Course GK
754 “Myocardial Hypertrophy” at the Charité - Universitätsmedizin Berlin/ German Heart
Institute Berlin (DHZB). Following 3 years of post-doctoral experience he became group leader of the research group
“Molecular Physiology” at the Institute of Vegetative Physiology. The main focus of his research is “Posttranscriptional
control of gene expression – regulation at the mRNA layer” under stress conditions such as hypoxia, and during development. This involves regulative aspects that specifically modulate mRNA-stability, efficiency of mRNA translation or cellular
mRNA-localization. Michael Fähling is a registered reviewer for several journals (e.g. Nucleic Acids Research, American
Journal of Physiology, European Journal of Physiology) and Associate Editor of Frontiers in Molecular Neuroscience.
Members of the group
Scientists
Franjicevic, Monika
Skalweit, Angela
Dipl. Biol.
Dr. med.
Technicians
Neumann, Ulrike
Stöbe, Regine Werner, Jeannette
BTA (pro rata basis)
Dipl. Ing. (FH)
MTLA
Scientists and Technicans. From left to right first row:
Regine Stöbe, Ulrike Neumann, Jeannette Werner, back row:
Dr. Benno Nafz, Dr. Michael Fähling, Dr. Angela Skalweit, Prof.
Dr. Bernd-Joachim Thiele
Students
Benko, Edgar
Gardow, Stefanie-Josefin
Perlewitz, Andrea
Schmidt, Irina
Staudacher, Jonas
PhD student
Medical student
PhD student
PhD student
Medical student
Students. From left to right: Andrea Perlewitz, Jonas Staudacher, Stefanie-Josefin Gardow, Edgar Benko.
293
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Summary
The information for specific gene regulation is mainly
localized in the non-coding regions of the genome.
At the DNA level cis-elements that are essential for
gene transcription can be found in the gene promoter
or in enhancer/ silencer regions upstream or downstream of the gene. Further regulatory elements are
localized in the mRNA untranslated regions (UTRs).
The message they carry is crucial to define mRNAhalf life time, mRNA localization and the efficiency of
translation. In our group we investigate these posttranscriptional - mRNA UTR based - mechanisms of
gene expression control in cellular models.
Nearly all eucaryotic mRNAs contain a 5’- and
3’-UTR. Specific mRNA UTR cis-elements are
responsible for the action of trans-factors like
RNA-binding proteins (RNA-BPs) or microRNAs
(miRNAs). In an interspecies comparision mRNA
5’UTRs consist, on average, of ~200 nucleotides
(nt). In contrast, mRNA 3’UTRs are more divergent,
ranging from ~200 nt in plants and fungi to ~440 nt
in mammals. Human mRNA 3’UTRs, for instance,
have an average size of ~1.000 nt, thus, are similar in
length to human mRNA translated regions. The longest known 3’UTR is that of FMR2, containing 9.280
nt. Indicators of functionally (or regulatory) relevance
in mRNA UTRs are repetetive sequences or conserved regions. The best-defined cis-elements are
specific mRNA-secondary structures (e.g. Internal
Ribosome Entry Sites (IRES)), CU-rich elements (e.g.
294
Differentiation Control Elements – DICE) or AU-rich
elements (ARE). The latter motives (with the consensus: [AA]UAAAU[AA]) mainly convey alterations
in mRNA stability. The list of known trans-factors
is constantly growing. Currently more than 370
RNA-binding proteins have been identified (source:
Human Protein Reference Database), as well as lots
of miRNAs. Mechanisms of posttranscriptional gene
expression control seem to be crucial under stress
conditions and during development. Consequently,
several diseases are known which are a result of
loss of function (e.g. by mutations) of cis-elements
or trans-factors.
At present we focus in our studies on the posttranscriptional control of the Epithelial Na2+-Channel
(ENaC) under the influence of hormones and on the
transcription factor human Achaete-Scute Homologue-1 (hASH1, ASCL1) during development and
the hypoxia response. Furthermore, we investigate
the influence of metabolic rate depression on mRNA
translation.
To address these questions we use a couple of
molecular standard methods as well as special
techniques such as Electrophoretic-Mobility Shift
Assays (EMSA), UV-Cross-linking, Affinity Chromatography, (Sucrose-) Polysomal Gradient Analysis,
UTR-dependent reporter gene assays or methods
to estimate mRNA and protein half life times.
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Zusammenfassung
Die Informationen für die Regulation der spezifischen Genexpression sind hauptsächlich in den
nicht-kodierenden Regionen des Genoms lokalisiert.
Ähnlich wie auf DNA-Ebene (Promotoren, Enhancer)
verhält es sich auch auf der mRNA-Ebene, wo die
mRNA untranslatierten Regionen (UTRs) wichtige
regulative cis-Elemente enthalten, die die Eigenschaften der jeweiligen mRNA bezüglich ihrer Stabilität, zellulären Lokalisation und Translatierbarkeit
beeinflussen. Unsere Arbeitsgruppe befasst sich mit
der posttranskriptionellen Genexpressionskontrolle
in zellbiologischen Untersuchungen.
Nahezu alle eukaryotischen mRNAs enthalten eine
5’- und 3’-untranslatierte Region. Die in den mRNA 5’und 3’-UTRs lokalisierten cis-regulatorischen Informationen werden hauptsächlich durch trans‑Faktoren wie RNA-Bindungsproteine (RNA-BPs), oder
micro-RNAs (miRNAs) umgesetzt. In Vergleich
einer Vielzahl untersuchter Spezies wurde ermittelt,
dass mRNA 5’UTRs eine durchschnittliche Länge
von ca. 200 nt aufweisen. Demgegenüber variieren
die Längen der mRNA 3’UTRs stärker (~200 nt bei
Pflanzen und Pilze, ~440 nt bei Säugern und speziell
beim Menschen ~1 000 nt). Damit weisen humane
3’UTRs eine vergleichbare durchschnittliche Länge
wie die der translatierten Bereiche auf. Die längste
bekannte 3’UTR umfasst 9 280 nt und wurde in der
humanen FMR2 mRNA identifiziert. Indikatoren für
funktionell relevante Elemente sind hauptsächlich
repetitive Elemente und konservierte Bereiche in
den UTRs. Die bekanntesten RNA-Bindungsmotive
sind spezifische Sekundärstrukturen (bspw. IRES –
Internal Ribosome Entry Site), CU-reiche Elemente
(bspw. Differentiation Control Elements – DICE)
sowie ARE (AU-Rich Elements). Die Liste von potentiell interagierenden trans-Faktoren wächst stetig
und umfasst z.Z. über 370 RNA-Bindungsproteine
(Quelle: Human Protein Reference Database) und
eine Vielzahl von miRNAs. Die posttranskriptionelle
Expressionskontrolle scheint insbesondere bei der
Anpassung von Zellen an veränderten Umweltbedingungen (Stress) sowie bei Wachstums- und Entwicklungsprozessen von besonderer Bedeutung zu
sein. Eine Veränderung der Bindungsmotive bzw.
der interagierenden trans-Faktoren durch Mutationen oder differentielles Spleißen ist daher auch mit
verschiedenen Krankheiten assoziiert.
Der Fokus unserer Gruppe liegt auf der Untersuchung mRNA-basierter Expressionsveränderungen
zentraler Faktoren wie dem Epithelialen Na2+-Kanal
(ENaC) unter hormoneller Stimulation oder dem
Transkriptionsfaktor human Achaete-Scute Homologue-1 (hASH1, ASCL1) während der Entwicklung
und unter Hypoxie. Darüber hinaus untersuchen wir
zentrale Prozesse wie den der mRNA Translation
unter Sauerstoff-Mangelbedingungen (Hypoxie).
Neben den molekularbiologischen Standardmethoden kommen bei unseren Untersuchungen spezielle Verfahren wie: Electrophoretic-Mobility Shift
Assays (EMSA), UV-Cross-linking, Affinitätschromatographie, Polysomengradientenanalyse, UTRabhängige Reportergen Assays oder mRNA- bzw.
Protein-Halbwertzeitbestimmungen zum Einsatz.
295
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Research projects
1. Posttranscriptional control of epithelial sodium channel
( ENaC ) expression
Project leader
Coworkers
Funding
Bernd-Joachim Thiele, Benno Nafz
Angela Skalweit, Michael Fähling, Andrea Perlewitz, Jeannette Werner,
Ulrike Neumann
German Research Foundation (DFG), Research Unit #667
„Tubular Mechanisms of renal Volume Regulation“ TP8
The sodium channel ENaC consists of three subunits:
α, β, and γ and is found in the apical membrane of saltabsorbing epithelia of organs such as the kidney, colon
or lung, where it constitutes the rate-limiting step in
Na+ and water absorption. In the kidney, vasopressin
and aldosterone play key roles in the minute adjustments of sodium re-absorption in the distal nephron.
Both hormones act via ENaC. The number of functional ENaC channels at the cell surface depends on
basic molecular processes like transcription of the
ENaC genes, synthesis of protein subunits, storage
in vesicles, vesicle trafficking and channel assembly
in the membrane, and finally on removal of channels
from the surface by endocytosis and degradation by
the ubiquitine/proteasome system. Basically, hormonal and environmental regulation of ENaC expression
is exerted through transcriptional control. In addition,
there is evidence of mRNA related posttranscriptional
processes, which affect mRNA stability or translational
296
efficiency, playing an important role in the synthesis
of ENaC subunits.
In this project, we investigate the influence of hormones,
e.g. aldosterone and vasopressin, at the level of posttranscriptional control of ENaC subunit expression. We
identified a wide range of RNA binding proteins (RBP)
as trans-factors and mRNA sequence motifs involved
in translational control of gamma-ENaC synthesis. For
instance, the γ-ENaC mRNA 3’UTR contains an AUrich element (ARE), which was shown by RNA-affinity
chromatography to interact with AU-rich element binding proteins (ARE-BP) like HuR, AUF1 and TTP. Some
RBPs associate with γ-ENaC mRNA in polysomes in
a hormone-dependent manner and are crucial for the
up-regulation of γ-ENaC synthesis. Furthermore, we
could demonstrate that aldosterone and vasopressin,
for instance, affect the synthesis of the α- and γ-, but
not the β-ENaC subunit, utilizing RBPs as effectors of
translational control.
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Figure 1: Sucrose polysome gradient analysis: Aldosterone and
dDAVP increase translational efficiency of alpha- and gamma-ENaC
mRNA.
Cytosolic extracts of aldosterone- or dDAVP-stimulated mpkCCDcells (S10) were separated by ultracentrifugation over 10-50%
sucrose gradients and divided into 12 fractions. Total RNA was
prepared and alpha-, beta- and gamma-ENaC mRNAs were visualized by RT-PCR. Polysome-bound mRNAs (which are translationally
active) sediment in fractions 2-8, monosomes and free mRNPs in
fractions 9-12.
(from: Perlewitz,A., B.Nafz, A.Skalweit, M.Fähling, P.B.Persson, and
B.J.Thiele. “Aldosterone and vasopressin affect alpha- and gammaENaC mRNA translation.” Nucleic Acids Research (2010). accepted.)
2. Posttranscriptional control of human achaete-scute
homologue-1 ( hASH1) synthesis
Project leader
Coworkers
Funding
External cooperations
Michael Fähling, Holger Scholz
Edgar Benko, Angelika Richter, Regine Stöbe,
German Research Foundation (DFG); FA 845/2-1
Prof. Dr. Dr. Joachim Klose, Institute of Human Genetics, Charité, CVK)
Junior-Prof. Dr. Jochen C. Meier, Max-Delbrück-Centrum for Molecular Medicine (MDC)
The human achaete-scute homologue-1 (hASH1;
ASCL1; Mash1) belongs to the bHLH (basic helix-loophelix) family of transcription factors. Mash1/ hASH1 is
expressed in both the central and the peripheral nervous system during development and promotes early
neuronal differentiation as well as the specification
of neuronal subtype identities. Furthermore, misex-
pression of Mash1/ hASH1 is correlated with a broad
variety of tumors, including lung cancer, neuroendocrine tumors, foregut and midgut carcinoid tumors,
pancreatic adenocarcinomas, islet cell tumors, gastrinomas, as well as parathyroid adenomas. Although
the importance of Mash1/ hASH1 in neurogenesis and
cancer has been well documented, the mechanism
297
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
of its regulation and downstream targets are largely
unknown. The aim of this project is the identification
of trans-factors which modulate hASH1 gene expression at the level of posttranscriptional control. For
instance, we identified hASH1 as a novel downstream
target of FMRP (Fragile X-Mental Retardation Protein).
FMRP was found to gate the translation of a large set
of mRNAs in dendrites that are involved in synaptic
plasticity. Dysfunction of the Fragile X-Mental Retardation-1 (FMR1) gene transcription is associated with
neuronal disorders, such as the Fragile X-Syndrome
(FXS) and the Fragile X-Associated Tremor/Ataxia
Syndrome (FXTAS). Thus, improved translational effi-
ciency of hASH1 mRNA by FMRP may represent an
important regulatory switch in neuronal differentiation.
Currently we investigate the role of oxygen in the alteration of hASH1 gene expression. Hypoxic suppression
of hASH1 synthesis may participate in the hypoxiainduced dedifferentiation of tumor cells, especially
of neuroblastoma cells. Although the initial cellular
response to low oxygen concentrations is the suppression of energy consuming processes, including
mRNA translation, our data suggest that inhibition of
hASH1 in hypoxia is actively controlled by cis-regulatory elements, located in the hASH1 mRNA 3’UTR.
Figure 2: Sucrose polysome gradient analysis: Neuroblastoma-derived Kelly cells were incubated either under control or
hypoxic conditions (21% vs. 0.5% O2) for 6 h. Cytosolic extracts were separated by sucrose gradient centrifugation as
described in Figure 1. A: Ribosomal profile recorded by absorbance at 254 nm. B, C: mRNA levels for hASH1 (B) and beta-Actin
(C) were quantified by real-time PCR and visualized by RT-PCR. The data indicate no dramatic alteration, neither in global translational efficiency (A) nor translation rate of beta-Actin mRNA (C). In contrast, the hASH1 mRNA (B) shifts from polysomes to the
translationally inactive non-polysomal fractions, indicating an inhibition of hASH1 mRNA translation under hypoxic conditions.
298
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
3. Rate of protein synthesis under hypometabolic conditions
Project leader
Coworkers
Funding
External cooperations
Michael Fähling
Stefanie-Josefin Gardow, Jonas Staudacher, Regine Stöbe,
Charité - Universitätsmedizin Berlin
Dr. Andreas Steege, Universitätsklinikum Regensburg
Under normoxic conditions, oxidative phosphorylation constitutes the major source of the cellular energy
intermediate adenosine triphosphate (ATP). High-rate
energy production is a prerequisite for cells and tissues to achieve a high metabolic rate. Increased oxygen consumption or disturbance in oxygen supply
result in decreased oxygen levels. Thus, hypoxia is a
consequence of inadequate oxygen availability. As a
consequence, multiple metabolic processes must be
coordinated to achieve a net suppression that balances the rates of ATP production and ATP consuming
processes at a new lower net rate of ATP turnover.
The process of protein synthesis belongs to the most
energy-consuming processes in the cell, accounting
for 25-30% under resting conditions. Consequently,
hypoxia causes metabolic rate depression and overall
protein synthesis drops to ~10% compared to control
level. Interestingly, it is well known that hundreds of
hypoxia-sensitive genes are increasingly expressed,
especially under prolonged hypoxic conditions. Thus,
effective mRNA translation following oxygen deprivation seems to be possible.
The aim of this project is to find out how specific
mRNAs can avoid the global suppression of mRNA
translation. Our hypothesis is that mRNAs of hypoxiasensitive genes are recruited to membrane-bound
polysomes, a process mediated by trans-factors interacting with cis-elements in the mRNA UTRs.
299
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Figure 3: Simplified model of cellular adaptation in moderate hypoxia and its influence on mRNA translation. Upon rapid suppression of global protein synthesis by PERK mediated inhibition of translational initiation, cells respond to hypoxia by activation
of glycolysis. Restored ATP levels ensure the expression of survival factors, which may be released such as signal molecules
(e.g. endothelin-1 (ET1), vascular endothelial growth factor (VEGF), or adenosine). Furthermore, newly synthesized factors can
be involved in glucose uptake, glycolysis, prevention of acidosis, or may represent factors affecting mRNA specific translation.
Under prolonged hypoxic conditions glucose may be consumed to a large extent, which causes a second drop in ATP levels. A
combined action of AMPK and mTOR, as well as unknown factors cause a global translational arrest accompanied by sustained
specific mRNA translation. (from: Fähling,M. “Surviving hypoxia by modulation of mRNA translation rate.” Journal of Cellular and
Molecular Medicine (2009). 13(9A):2770-2779.)
300
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Publications 2006 - 2010
Schildroth J, Rettig-Zimmermann J, Kalk P, Steege A, Fähling
M, Sendeski M, Paliege A, Lai EY, Bachmann S, Persson PB,
Hocher B, Patzak A (2010): Endothelin type A and B receptors in the control of afferent and efferent arterioles in mice.
Nephrol Dial Transplant accepted
Perlewitz A, Nafz B, Skalweit A, Fähling M, Persson PB,
Thiele BJ (2010): Aldosterone and vasopressin affect alphaand gamma-ENaC mRNA translation. Nucleic Acids Res
accepted
Kielbasa SA, Blüthgen N, Fähling M, Mrowka R (2010):
Targetfinder.org: A resource for systematic discovery of
transcription factor target genes. Nucleic Acids Res 38
Suppl:W233-8
Westphal K, Stangl V, Fähling M, Dreger H, Weller A, Baumann G, Stangl K, Meiners S (2009): Human-specific Induction of Glutathione Peroxidase-3 by Proteasome Inhibition in
Cardiovascular Cells. Free Radic Biol Med 47(11):1652-60
Eichler SA, Förstera B, Smolinsky B, Jüttner R, Lehmann TN,
Fähling M, Schwarz G, Legendre P, Meier JC (2009): Splicespecific roles of glycine receptor a3 in the hippocampus. Eur
J Neurosci 30(6):1077-91
Lai EY, Fähling M, Ma Z, Källskog Ö, Persson PB, Patzak A,
Persson AE, Hultström M (2009): Norepinephrine increases
calcium sensitivity of mouse afferent arteriole, thereby
enhancing angiotensin II-mediated vasoconstriction. Kidney
Int 76(9):953-9
Fähling M, Mrowka R, Steege A, Kirschner KM, Benko E,
Förstera B, Persson PB, Thiele BJ, Meier JC, Scholz H
(2009): Translational regulation of the human Achaete-Scute
Homologue-1 (hASH1) by Fragile X Mental Retardation Protein
(FMRP). J Biol Chem 284(7):4255-66
Lüdemann L, Nafz B, Elsner F, Grosse-Siestrup C, Meissler
M, Kaufels N, Rehbein H, Persson PB, Michaely HJ, Lengsfeld P, Voth M, Gutberlet M (2009): Absolute quantification
of regional renal blood flow in swine by dynamic contrastenhanced magnetic resonance imaging using a blood pool
contrast agent. Invest Radiol 44(3):125-34
Ufer C, Wang CC, Fähling M, Schiebel H, Thiele BJ, Billett
EE, Kuhn HH, Borchert A (2008): Translational regulation of
glutathione peroxidase 4 expression through guanine-rich
sequence binding factor 1 is essential for embryonic brain
development. Genes Dev 22(13):1838-50
Steege A, Fähling M, Paliege A, Bondke A, Kaps C, Kirschner
KM, Martinka P, Patzak A, Scholz H, Persson PB, Thiele BJ,
Mrowka R (2008): Wilms’ tumor protein WT1(-KTS) modulates renin gene transcription. Kidney Int 74(4):458-66
Mrowka R, Blüthgen N, Fähling M (2008): Seed based systematic discovery of specific transcription factor target genes.
FEBS J 275(12):3178-92
Martinka P, Lai EY, Fähling M, Jankowski V, Jankowski J,
Schubert R, Gaestel M, Persson AE, Persson PB, Patzak A
(2008): Adenosine increases calcium sensitivity via receptorindependent activation of p38 MAPK/MK2 pathway in isolated mesenteric arteries. Acta Physiol (Oxf.) 193(1):37-46
Patzak A, Steege A, Lai EY, Brinkmann JO, Kupsch E, Spielmann N, Gericke A, Skalweit A, Stegbauer J, Persson PB,
Seeliger E (2008): Angiotensin II response in afferent arterioles of mice lacking either the endothelial or neuronal isoform
of nitric oxide synthase. Am J Physiol Regul Integr Comp
Physiol 294(2):R429-37
Patzak A, Lai EY, Fähling M, Sendeski M, Martinka P, Persson
PB, Persson AE (2007): Adenosine enhances long-term the
contractile response to angiotensin II in afferent arterioles. Am
J Physiol Regul Integr Comp Physiol 293(6):R2232-42
Mrowka R, Steege A, Kaps C, Herzel H, Thiele BJ, Persson
PB, Blüthgen N (2007): Dissecting the action of an evolutionary conserved non-coding region on renin promoter activity.
Nucleic Acids Res 35(15):5120-9
Schmidt I, Fähling M, Nafz B, Skalweit A, Thiele BJ (2007):
Induction of translationally controlled tumour protein (TCTP)
by transcriptional and posttranscriptional mechanisms. FEBS
J 274(20):5416-24
Lai EY, Martinka P, Fähling M, Mrowka R, Steege A, Gericke
A, Sendeski M, Persson PB, Persson AE, Patzak A (2006):
Adenosine Restores Angiotensin II-Induced Contractions by
Receptor-Independent Enhancement of Calcium Sensitivity in
Renal Arterioles. Circulation Res 99:1117-1124
301
MICHAEL FÄHLING – MOLECULAR PHYSIOLOGY
Fähling M, Mrowka R, Steege A, Nebrich G, Perlewitz A,
Persson PB, Thiele BJ (2006): Translational Control of Collagen Prolyl 4-Hydroxylase alpha (I) Gene Expression under
Hypoxia. J Biol Chem 281(36):26089-10
Andree H, Thiele H, Fähling M, Schmidt I, Thiele BJ (2006):
Expression of the human TPT1 gene coding for translationally controlled tumor protein (TCTP) is regulated by CREB
transcription factors. Gene 380(2):95-103
Fähling M, Mrowka R, Steege A, Martinka P, Persson PB,
Thiele BJ (2006): Heterogeneous Nuclear RibonucleoproteinA2/B1 Modulate Collagen Prolyl 4-Hydroxylase alpha (I)
mRNA Stability. J Biol Chem 281(14):9279-86
Theilig F, Debiec H, Nafz B, Ronco P, Nüsing R, Seyberth HW,
Pavenstädt H, Bouby N, Bachmann S (2006): Renal cortical
regulation of COX-1 and functionally related products in early
renovascular hypertension (rat). Am J Physiol Renal Physiol
291(5):F987-94
Reviews & book chapters (2006-2010)
Fähling M (2009): Surviving hypoxia by modulation of mRNA
translation rate. J Cell Mol Med 13(9A):2770-2779
Fähling M (2009): Cellular oxygen sensing, signalling and how
to survive translational arrest in hypoxia. Acta Physiol (Oxf.)
195(2):205-230
General information
Third party funding ( 2006-2010 )
Project leader
Thiele BJ.
Nafz B.
Nafz B.
Fähling M.
Scholz H.
302
Project title
Tubuläre Mechanismen der renalen
Volumenregulation
Sponsor
Deutsche Forschungsgemeinschaft (DFG); FoGr 667, TP8,
TH 459/5-1
Functional genomics of cardiovascular NGFN 2,
damage in hypertension
BMBF KBCV 1.2,
01GS0416
Posttranskriptionelle Kontrolle der
Deutsche ForschungsgemeinGenexpression des bHLH-Transkrip- schaft (DFG)
tionsfaktors Mash1
FA 845/2-1
Period
2005-2010
2004-2007
2008-2011
SCIENTIFIC COORDINATION
As a Scientific Coordinator of the CCR Dr. rer. nat. Karin Effertz is responsible for the webpage (www.ccr.
charite.de), the bi-annual report of the CCR and for general public relations. Moreover she gives strategic and
administrative support for the preparation of cooperative grant proposals and for the realisation of joint research
projects at the CCR.
Frau Dr. rer. nat. Karin Effertz kümmert sich als Wissenschaftliche Koordinatorin um die Belange der Öffentlichkeitsarbeit, wie die CCR Webseite (www.ccr.charite.de), den CCR Jahresbericht, und die allgemeine Außendarstellung. Darüber hinaus unterstützt sie strategisch, inhaltlich und administrativ die Erstellung von Verbundanträgen und die Durchführung von Verbundprojekten am CCR.
303
VASCULAR TRAINING
GefäSStraining : Von der Stange zum molekularen MaSSanzug
Nicht jede Art von Sport ist die richtige Antwort auf eine Gefäßerkrankung
Von Ivo Buschmann
„Treiben Sie mehr Sport!“ So rich­tig dieser ärztliche Rat
auch für Patienten mit Gefäßkrankheiten ist, so komplex und facettenreich stellt er sich im klinischen Alltag
dar. Welchen Sport soll ein Patient mit peripherer arterieller Ver­schlusskrankheit (PAVK) machen? Radfahren oder lieber laufen? Wie lange muss ein Herzpatient
am Tag laufen? Eher morgens oder besser abends?
Was macht der Patient der nicht richtig trainieren
kann? Durch die molekulare Grundla­genforschung
der letzten Jahr­zehnte kommt nun langsam Licht in
dieses Dunkel und erstmals zeichnet sich am Horizont
eine zu­nehmend verbesserte Behand­lungsform von
Gefäßkrankheiten ab: vom unspezifischen Gefäß­sport
zum patientenindividuellen, personalisierten Gefäßtraining. Dabei stehen insbesondere 3 Säu­len der
Therapie im Vordergrund:
- die Nutzung der patientenei­genen Regeneration von
biolo­gischen Bypässen.
- die Kombination von gefäß­therapeutischer Intervention (Ballondilatation und Stent) mit Gefäßtraining und
- die Personalisierung der Ge­fäßtherapie (Maßanzug)
statt unspezifischem Training (von der Stange).
Regelmäßige Belastung tut den Beinen gut
Biologische Bypässe sind kleine Kollateralkreisläufe, die
sich zu vollwertigen Arterien umbilden können. Fließt
das Blut in einem Umgehungskreislauf schneller und
damit mit hoher Pulsatilität, er­weitert sich als biologische Ant­wort der Querschnitt dieser Arte­rie und kompensiert somit das Blutflussdefizit auf der verschlos­
senen Hauptstrecke. Je länger und effizienter diese
304
Pulsatilität beste­hen bleibt bzw. erhöht vorliegt, umso
besser kann sich ein kollate­raler Kreislauf ausbilden.
Und das ist auch der Grund, wa­rum eine regelmäßige
Belastung dem Herzen und den Beinen so gut tut
und zum Beispiel Hobbysport­ler statistisch gesehen
länger le­ben. Zwar treten auch bei ihnen atherosklerotische Veränderungen der Arterien auf, durch deren
bio­logische Regeneration über eigene biologische
Bypässe sind sie je­doch im Falle von Gefäßverengun­
gen geschützt.
Hohe Pulsatilität gibt das Signal zur Bildung
von Kollateralen
Das wichtigste Wachstumssignal für solche Kollateralen ist ein ho­her pulsatiler Blutfluss. Demnach können sich biologische Bypässe (z. B. im Unterschenkel)
besser ver­größern, wenn dieses Blutflusssig­nal auch
in der Peripherie bei ih­nen ankommt. Ist also eine
große Beckenarterie verschlossen, ist es sinnvoll, eine
solche Einengung– entweder mithilfe eines Ballonka­
theters sowie gegebenenfalls auch einer Gefäßstütze
(Stent) – zu­nächst wieder zu öffnen, bevor der Patient
mit einem strukturierten Gefäßtraining beginnen kann.
Durch die Erfindung eines „Gefäß­tachometers“ an der
Charité und im Gefäßzentrum Berlin lassen sich diese
kollateralspezifischen Beschleunigungswerte im Blut­
strom nun mithilfe von Ultra­schallverfahren messen und
visua­lisieren [1]. Möglicherweise erge­ben sich dadurch
auch neue Ein­sichten zu den interessanten Er­gebnissen
der MIMIC1-Studie [2], in der eine optimale Medikation
1 Mild to Intermittent Claudication
VASCULAR TRAINING
plus Gefäßtraining gegen eine op­timale Medikation plus
Gefäßtrai­ning plus eine Angioplastie der Becken- und
der Oberschenkel­etage verglichen wurden.
Wurde auch eine Angioplastie durchgeführt, verbesserten sich sowohl der Knöchel-Arm-Index („ankle
brachial index“; ABI) als auch die Gehstrecke signifikant. Ferner wissen wir, dass Therapie­verfahren additiv wirken können.
Der Schlüssel sind individuell angepasste
Therapieschemata
Ein unspezifisches Training ist nicht immer die richtige
Antwort auf eine Gefäßerkrankung. Bei­spielsweise
ist ein Training mit dem Fahrradergometer bekann­
termaßen bei Weitem nicht so effi­zient wie ein strukturiertes Geh­training. Interessanterweise ver­bessert aber
ein Arm-Ergometer­Training die periphere arterielle Verschlusskrankheit. Dass hierbei die pulsatile Beschleunigung des Blutstromes eine viel wichtigere Rolle spielt
als bisher vermutet [1], eröffnet neue Behandlungspfade
für stenosierende Gefäßkrankhei­ten.
Der bereits erwähnte Gefäßtacho­meter wird es in
Zukunft möglich machen, das Training der Patien­ten
(wie bei einem Tachometer im Auto) zu dosieren und
damit zu spezifizieren. Erstmals ergibt sich hiermit
auch die Möglichkeit, pas­sive Trainingsverfahren wie
die externe Gegenpulsation spezifisch anzuwenden:
Wurde bisher mit zum Teil noch sehr hohen Drü­
cken, die bei einigen Patienten zu deutlichen Nebenwirkungen führ­ten („enhanced counterpulsation“;
EECP), blind behandelt, lässt sich jetzt das schonende Verfahren der Herzhose [3] in Kombination mit
einer Gefäßtachometer-Darstel­lung kombinieren und
spezifizie­ren.
Damit ist es möglich, statt einer Standardtherapie
von 6–7 Wochen ein individuell angepasstes The­
rapieschema zu starten. Derzeit laufen am Gefäßzen-
trum Berlin in Kooperation mit der Charité mehrere
Studien, die diesen neuen individualisierten Gefäßansatz verfolgen. Spannende Zeiten für alle BlutflussPhysiologen also: „Go with the flow!“
Literatur
1 Buschmann I, Pries A, Styp-Rekowska B et al. Pulsatile shear and Gja5 modulate arterial identity and
re­modeling events during flow-driven arteriogenesis. Development 2010; 137: 2187–2196
2 Greenhalgh RM, Belch JJ, Brown LC et al. The adjuvant benefit of an­gioplasty in patients with mild to
moderate intermittent claudication (MIMIC) managed by supervised exercise, smoking cessation
advice and best medical therapy: results from two
randomised trials for ste­notic femoropopliteal and
aortoiliac arterial disease. Eur J Vasc Endovasc Surg
2008; 36: 680–688
3 Buschmann EE, Utz W, Pagonas N et al. Improvement of fractional flow reserve and collateral flow by
treat­ment with external counterpulsa­tion (Art.Net.-2
Trial). Eur J Clin In­vest 2009; 39: 866–87 5
305
Research Foci at the CCR
Drug Development
Principle Investigators
Partners
Heiko Funke-Kaiser, Ulrike Steckelings, Franz Theuring, Thomas Unger,
Berthold Hocher
Prof. Jürgen Scholze , Charité Campus Mitte, Berlin, Klinik für Innere
Medizin/ Kardiologie am Deutschen Herzzentrum Berlin
Drug development is a complex, time- and costintensive process involving different disciplines from
medicine and natural sciences. Classically, drug development can be divided into two sequential modules
called drug discovery and drug development
(subsequent figure). Discovery comprises all steps
from the therapeutic concept, i.e. from the biological
target, to the molecule, whereas development encompasses the operating processes from the molecule to
the registered drug. Discovery often starts with target
identification, e.g. the cloning and (patho)physiological characterisation of a novel gene. A crucial step in
early drug discovery is assay development. This can
be accomplished through the generation of stable
cell lines and a luciferase-based read-out to measure receptor activity. A high-throughput screening
(HTS; primary screening) to explore the chemical space
for pharmacological activities represents the next step
which can be performed in cooperation with contract
research organisations (CROs). So-called confirmed
compounds resulting from the HTS are validated by
a secondary screening to yield hits. The hit-to-lead
programme addresses aspects such as structureactivity-relationships (SAR)/ definition of a pharmacophore, determination of affinity (KD) and potency
(IC50), specificity/ selectivity (panel-screens), pharmacokinetic (i.e., absorption, distribution, metabolism,
excretion (ADME)) and toxicological (T) parameters.
The hit-to-lead programme can be viewed as a filtering
306
process to narrow down the hundreds and thousands
of hits generated by the HTS/ secondary screening,
but also comprises synthetic chemistry (medicinal
chemistry (MC)) steps. Leads - chemical structures
with a potency in the 1-10 microM range, selectivity
and appropriate ADMET parameters - represent the
milestone of the hit-to-lead programme.
The lead-to-candidate programme, also named
lead optimisation phase, aims to further increase
affinity/ potency, specificity/ selectivity and ADMET
properties by using e.g. in vivo ADMET analyses. The
in vivo proof-of-concept (PoC) is a crucial milestone
within the lead-to-candidate programme to analyse
if a certain compound is effective with respect to a
certain indication in an animal model. So-called devel­
opmental candidates have passed the PoC and are
further optimised by MC to attain e.g. a potency in the
1-10 nM range, which is a usual prerequisite for clini­
cal candidates for the first trials in humans.
The clinical phase of drug development comprises
for defined phases: Phase I studies are performed
on a small group of healthy volunteers with the aim to
check for drug safety, tolerability, and pharmacokinetic
properties. Phase II studies are performed on groups
of patients with a defined set of indications to test for
clinical efficacy, and for dose finding analysis. Often
such studies will cover several distinct clinical disorders to identify the possible therapeutic indications for
the new compound. Phase III trials are the definitive
Research Foci at the CCR
double-blind randomised trials, commonly performed
as multicentre trials on 1000-3000 patients, aimed at
comparing the new drug with commonly used alternatives. At the end of a “successful” phase III, the drug
will be submitted to the relevant regulatory authority
for licensing. Phase IV studies comprise the obligatory postmarketing surveillance designed to detect
any long-term adverse effects. More importantly,
these studies are important to identify new potential
indications for the drug in a clinical setting in many
thousands of patients.
Clinical studies are a major cornerstone of the drug
development agenda and the translational research
approach at the CCR. In close collaborations with
clinical departments, in particular the Outpatient
Department at the Charité Campus Mitte under
the guidance of Prof. Jürgen Scholze, and the
Department of Internal Medicine/ Cardiology at
the German Heart Institute, multiple clinical trials are
currently conducted in different clinical phases and cardiovascular indications. In addition, clinical investigators
with a focus on cardiovascular research from various
departments at the Charité have their laboratories at
the CCR, and provide an optimal platform for mutual
research interactions.
I. Drug discovery
target
identification
target
validation
assay
development
HTS
secondary
screening
II. Drug discovery & preclinical drug development
hit-to-lead-programme
lead-to-candidate programme
developmental
candidates
leads
hits
clinical
candidate
III. Clinical drug development
phase I
phase II
phase III
approval
phase IV
CCR´s research teams cover a broad spectrum within
the drug discovery and development process as illustrated in the next table:
target identification
and validation
assay development to
hit-to-lead-programme
lead-to-candidateprogramme, e.g.
preclinical in vivo proofof-concept/
in vivo evaluation of different indications
Phase II
Phase III
Phase IV
Funke-Kaiser/
Unger, Kintscher
Funke-Kaiser/
Unger, Kintscher
Steckelings/
Unger, Hocher. Theuring
Hocher
Theuring
Kintscher/ Unger/ Scholze
To give an example, Prof. Dr Thomas Unger and Dr.
Ulrike Steckelings are in strategic partnership with the
company Vicore Pharma to contribute to the preclinical
and clinical development of AT2 Receptor Agonists.
Moreover two drug developing companies were
founded by members of the CCR as spin offs of the
Charité. CCR Pharma was founded by PD Dr. Heiko
Funke-Kaiser, Prof. Dr. Thomas Unger, Frank Zollmann
and Dr. Jan Schefe and is developing prorenin/ rennin receptor (RER) antagonists as potential future cardiovascular drugs, a project funded by the “Go Bio”
program of the BMBF.
http://www.bmbf.de/de/6868.php,
http://www.ccr-pharma.de
The company of Prof. Dr. Franz Theuring is targeting
neurodegenerative diseases. http://www.taurx.com
For more detailed information on the various projects
please refer to the chapters of the individual research
groups.
307
Research Foci at the CCR
Gender in Cardiovascular Research
Principle Investigators
Partners
Vera Regitz-Zagrosek (Research Focus-, GK-,
RG-coordinator), Duska Dragun (GK-, RG-Member),
Ulrich Kintscher (GK-, RG-member), Andreas Patzak,
Pontus Persson (GK) Thomas Unger/ Ulrike Steckelings/
Heiko Funke-Kaiser (GK-members)
national: Gender in Medicine (GIM) Charité, Members
of the Graduate Course GK 754 and of the Research Group FOR 1054
international: European curriculum gender in medicine (EUGIM) members
(Karin Schenck-Gustafsson, Center on Gender in Medicine at the Karolinska Institute
Stockholm, Ineke Klinge, University of Maastricht, Netherlands; Maria Koop,
Semmelweiss University Budapest, Hungary; Margarete Hochleitner, Womens Health
Center of Insbruck Medical University, Austria; Flavia Franconi, Universita di Sassari,
Italy, Toine Lagro Janssen, University of Njimegen, Netherlands);
Eugeneheart Gender Task members (Fred Jaisser, Paris, H Jarry, Goettingen,
Leon de Windt, Maastricht, Guido Tarone, Turin, Karin Sipido, Leuven),
International Society of Gender in Medicine Board members
Gender differences in widespread diseases like cardiovascular disease, stroke, immunological disease
etc. are well known, but the underlying molecular and
cellular mechanisms are still poorly understood. The
hypothesis of steroid hormones as molecular regulators of gender specific pathological phenotypes
is tempting, but in many cases still to be proven.
Research groups at the CCR, which are interested in
this topic, work together with groups from Charité and
MDC in the Research Focus “Gender in Cardiovascular Disease”. The common aim of the group is to investigate sex and gender differences in cardiovascular
and cardiometabolic and -renal diseases and to define
the role of sex hormones. Eleven groups including 4
from CCR, one from DHZB (Deutsches Herzzentrum
Berlin), other from Charité and MDC are cooperating
since 2001 with similar goals in the Research Train308
ing Program GK 754: “Sex and gender specific
mechanisms in myocardial hypertrophy This GK
was particularly successful. It has been funded from
2001 – 2011 and about 55 MD, veterinarian and PhD
students will obtain their degrees in this graduate
course, most of them already finished with summa
and magna cum laude. Sex differences in myocardial hypertrophy A Research Group (DFG,
FOR 1054) - Forschergruppe – Sex differences in
myocardial hypertrophy is funded from September
2008- 2011 with the contribution of the CCR-groups of
Dragun, Kintscher and Regitz-Zagrosek (coordinator).
Animal models are an appropriate tool for analyzing
molecular and cellular mechanisms of cardiovascular
and cardiorenal diseases and show often sex specific
differences. Three such models with known gender
differences are established at the CCR: a mouse
Research Foci at the CCR
model for pressure induced hypertrophy (transverse
aortic banding), a mouse model for salt induced
hypertrophy (DOCA salt) and a model for physiological hypertrophy after voluntary exercise.. Forced exercise has been established as another physiological
hypertrophy model. In these models, sex differences
and estrogen/androgen mediated signal transduction are investigated. Moreover, in hypothesis-driven
approaches, the interaction of sex differences and sex
hormones on cardiomyocyte signalling will be analyzed and, in a more systemic approach, influence of
sex hormones on gene expression and protein pattern
will be examined.
Karima Schwab and Nicolas Vignon-Zellweger from
the Group of F. Theuring work on effects of estrogens
and phytoestrogenes on protein expression pattern in
systemic approaches.
Furthermore gender aspects in cardiovascular diseases are investigated in international cooperations
with the INSERM Institute Toulouse, France and the
John Hopkins School of Public Health, Baltimore US.
At the European level gender aspects are analyzed
in Task 4 of the EUGENE project (consortium of 21
partners from research institutes and SMEs from 10
different countries).
An additional goal is the implementation of Gender
aspects in the medical education all over Europe with
the Erasmus Project EUGIM, developing a Bologna
compatible module „Gender Medicine“ with 7 European partner universities.
At the national level Gender aspects will be implemented systematically in the novel medical education
at the Charité Universitätsmedizin Berlin („Modellstudiengang Medizin“), which will start in October 2010.
309
Research Foci at the CCR
Metabolism
Principle Investigators Partners Ulrich Kintscher, Michael Schupp, Berthold Hocher, Stefan Anker
Joachim Spranger (Endocrinology, Charité), Jürgen Scholze (Outpatient Clinic,
Charité), Philipp Stawowy (Cardiology, DHZB), Simone Spuler (ECRC, Charité),
M. Boschmann (ECRC, Charité)
Deregulation of body weight control occurs in two
opposite directions: progressive gain of weight which
leads to obesity or progressive loss of weight resulting
in cachexia. Between these two extremes an interfacial area exists with common molecular mediators
of pathogenesis. Based on these joint pathologies,
the present research focus metabolism concentrates
on molecular mechanisms involved in body weight
regulation in a bidirectional manner. Moreover studies
of genetic factors and epigenetic DNA modifications
shed light on their contribution to metabolic control.
Cachexia (Anker)
An underlying hypothesis of our work is that all forms
of cachexia as seen in COPD, cancer, infections and
old age lead to pathophysiologically relevant cardiovascular alterations, namely chronic heart failure,
which is clinically relevant for prognosis. Disturbance
of the endothelial function and the regulation of peripheral perfusion result in ischemia and hypoxia of the
peripheral tissue as well as the induction of reactive
oxygen species and inflammation. We have some
evidence that all forms of cachexia, independently of
their underlying chronic disease, show neurohumoral
activation, which is commonly associated with chronic
heart failure. Furthermore, cachectic patients almost
with no exception are short of breath, fatigue easily
and very often show edema, all of which are hallmarks
of chronic heart failure. We have shown that several
therapies of chronic heart failure display anti-cachectic properties, e.g. ACE inhibitors and beta-blockers.
Hence, we are testing whether these cardiovascular
therapies are effective in other therapeutical areas
than cardiac cachexia. Currently, several projects
are focused on cardiovascular drugs and novel compounds on survival, heart function and body composition in a rat model of cancer cachexia.
Obesity (Kintscher)
Obesity is a major risk factor for cardiovascular disease (CVD). Our work is focussing on mechanisms
involved in obesity-mediated CVD. Hereby we are concentrating on tissue cross-talk between metabolic tissues such as adipose tissue, skeletal muscle and liver
310
Research Foci at the CCR
and CV-organs such as the heart and the vasculature.
Regulation of tissue crosstalk by endocrine mediators such as nuclear hormone receptors is one of the
main topics. We follow a strict translational research
approach from cell culture to animal to proof-of-concept studies in humans.
Genetic Mechanisms and Impact of Epigenetic
Modifications (Hocher)
Fetal programming of cardiovascular disease
There is meanwhile clear evidence indicating that early
life events play an important role in the pathogenesis of cardiovascular diseases in later life. We have
introduced the concept of maternal genes exerting
unfavourable effects on the offspring into this currently rapidly growing research field. We were furthermore able to demonstrate for the first time that insulin
resistance may also be present in otherwise healthy
human newborns already at birth. Our animal studies demonstrated that also over-nutrition may exert
harmful effects on the offspring in a gender dependent manner. Current clinical and preclinical studies
are designed to detect molecular pathways of fetal
programming including epigenetic DNA modifications.
The Research Focus Metabolism is in close collaboration with a series of other clinical and basic research
groups. The schematic presentation below shows
currently funded collaboration projects at the Charité.
311
Research Foci at the CCR
Vascular Stress
Principle Investigators
Partner
Axel Pries, Thomas Unger, Kai Kappert, Ivo Buschmann, Harm Peters,
Elena Kaschina, Ulrike Steckelings, Heiko Funke-Kaiser, Duska Dragun,
Ullrich Kintscher, Andreas Zakrzewicz, Vera Regitz-Zagrosek
Ferdinand le Noble (MDC Berlin-Buch), Gilbert Schönfelder (Charité), Michael Bader
(MDC, Berlin-Buch), Norbert Hübner (MDC, Berlin-Buch),
Heinz-Peter Schultheiss (Charité)
For a major group of vascular diseases (arteriosclerosis, myocardial infarction, stroke, hypertension,
tumours) and clinically relevant adaptations (wound
healing, development, aging, exercise) vascular reactions to hemodynamic, inflammatory, oxidative or metabolic stimuli play an important role. In this context all
functional and pathologic scenarios are seen as vascular stressors, as long as they increase vascular load.
Frontiers between functionally adequate response and
maladaptation are fluent and are determined by the
type of stimuli, genetic predisposition and other protective or harmful factors.
The vascular wall is a flexible and integrating organ
consisting of cellular components (endothelial cells,
smooth muscle cells, cells of the adventitia and fibrob312
lasts) and the extracellular matrix. Within the process
of vascular remodelling number and form of all components of the vascular wall are continuously varied
resulting in a structural reorganisation of the vascular
wall. It responds to physiologic stimuli to guarantee
its stability under normal conditions. In the case of
cardiovascular disease pathological stimuli cause
maladaptation of the vascular wall.
Schematic representation of the CCR focus
“Vascular Stress”. Tissue remodelling of the
vascular wall is based on the capability of the
artery to alter in size and shape, and occurs in
response to numerous stress factors. This CCR
focus addresses aspects of pathological inward
remodelling in terms of atherosclerosis, restenosis
and transplant vasculopathy, as well as outward
remodelling regarding arteriogenesis and aneurysm formation. Integrating both clinicians and
basic scientists we are focussing at unravelling the
biological and molecular basis of these physiological and pathological remodelling processes, which
should pave the way for developing therapeutic
and preventive strategies for the benefit of patients
suffering or not yet suffering from vascular disease.
The involved principle investigators at the CCR are
listed along the sphere.
Research Foci at the CCR
Within the Research Focus „Vascular Stress“ at
the CCR clinicians and basic scientists are working
together to understand physiological and pathophysiological remodelling of the vascular wall.
A number of current research projects address topics
in the field of „Vascular Stress”. Some of the house
intern co-operations were initiated within the European early stage research training programm “Cardiovasc”. Within the CCR “Vascular Stress” focus
an interdisciplinary team of researchers (internists,
cardiologists, nephrologists, cell biologists, biochemists, physiologists and pharmacologists) is studying
vascular changes in different organs (heart, brain,
kidney, aorta and extremities). Diseases like stroke,
myocardial infarction, cardiac hypertrophy, aortic
aneurisms, cardiac, retinal or nephrotic changes due
to adipositas or diabetes, chonic kidney disease,
glomerulosclerosis, transplant vasculopathy but also
beneficial adaptations like arteriogenesis after stenosis are explored. On a molecular level, contributions
of the renin-angiotensin-aldosteron-system. NOcGMP-, TGF beta-, endothelin- estrogen- and PPAR
( Peroxisom-Proliferator-Activated Receptor)- and HIF
(hypoxia-inducible factors)-mediated signal transduction pathways are investigated.
One of the major claims of the research focus “Vascular Stress” is the advancement of internal and
external co-operations in order to increase the quality
of scientific output. This is why the research focus
concentrates on projects which need the expertise of
two or more groups.
313
DFG-RESEARCH GROUP MYOCARDIAL HYPERTROPHY
Research Group Myocardial Hypertrophy ( DFG, FOR 1054 )
Coordinator
Members
Vera Regitz-Zagrosek (CCR, GIM)
Duska Dragun (CCR), Ulrich Kintscher (CCR), Michael Bader (MDC),
Otmar Huber (CBF), Joachim Klose (CVK), Wolf-Hagen Schunck (MDC),
Dominik Müller (MDC), Shokoufeh Mahmoodzadeh (CCR)
The main aim of the research group “Myocardial
Hypertrophy” (FOR 1054) is the analysis of gender
differences in the adaptation of the heart to mechanical stress. Women develop a more advantageous and
physiological adaptation of the myocardium to stress
compared to men. This includes the synergistic activation of numerous cell-based protective pathways at
the molecular and cellular level, which are most often
linked to the activation of estrogen receptors or estrogens themselves. This influences mitochondria and
the myocardial energy metabolism and limits apoptosis and fibrosis. Testosterone, on the other hand,
appears to favour fibrosis and a more pronounced and
unfavourable myocardial proliferation. Our research
group performs a series of mechanical analysis using
cell cultures and animal models. This includes models
of physiological and pathological myocardial stress,
e.g. strength training and hypertension. The role of
estrogen and androgen receptors is selectively analyzed. Moreover, the effect of sex hormones on the
314
lipid turnover is investigated. Several collaborations
with clinical projects exist; in this area we mainly focus
on gender differences in myocardial reaction to overload due to aortic valve disease.
Graduate Courses
Graduate Courses
1. M ARIE CURIE early stage research training programme CARDIOVASC
Project Coordinator Project Manager Steering Committee Funding Patricia Ruiz Noppinger
Dian Michel
Eva Becher, Ivo Buschmann, Karin Effertz, Harm Peters, Catarina Curato,
EST fellow, George Kararigas, EST fellow
European Commission, FP6, Human Resources and Mobility Activity,
MEST-CT-2005-020268
CARDIOVASC is the acronym for cellular and molecular mechanisms of cardiovascular diseases representing a Marie Curie Early Stage Research Training
Programme for PhD students that started in January
2006. The aim of CARDIOVASC is to provide training
for young researchers in the specialized and highly
competitive area of cardiovascular diseases (CVD)
under the tutorial of highly experienced scientists at
the Center for Cardiovascular Research (CCR).
Research projects apply systematic and hypothesisdriven approaches in molecular, cellular and physi­
ological biology focusing on heart, kidney and
the vascular wall. The scientific programme of CARDIOVASC focuses on the following questions: (i)Gender-specific aspects of cardiac function and
dysfunction
(ii) Molecular mechanisms of endothelial dysfunction
in cardiovascular disease, and
(iii)New strategies in the prevention and treatment of
myocardial infarction.
A structured training programme with special emphasis on the integration of basic and clinical research is
provided to bundle and integrate these questions. Our
objective is to train a new generation of scientists and
clinicians with the capacity of working at the interface
of genetics, developmental biology, physiology and
human disease educated by specialists of the different
disciplines with the possibility of interacting across the
traditional boundaries.
The training programme is based upon three columns
• Personalized training guaranteed by a direct supervisor and a mentor, as well as the steering committee
•Instruments for structured training, such as seminars, lectures, workshops, meetings and e-cooperation
• Regular and interactive quality control of the fellows
and the supervisors
Fellows and supervisors of the Marie Curie EST programme
CARDIOVASC at the Harnack-Haus, Berlin-Dahlem at the Day
of-Science Meeting.
315
Graduate Courses
2. G
raduate course 754-III :
Gender specific mechanisms in myocardial hypertrophy
Project leader
Coworkers
Funding
Vera Regitz-Zagrosek
Duska Dragun, Heiko Funke-Kaiser, Roland Hetzer (DHZB) Ulrich Kintscher,
Joachim Klose, C.Knossalla (DHZB), Jun Li, Shokufeh Mahmoodzadeh,
Ingo Morano (MDC Berlin), Andreas Patzak, Martin Paul, Pontus Persson,
Patricia Ruiz Noppinger, Gilbert Schönfelder, Carola Schubert, Ulrike Steckelings,
Thomas Unger
German Research Foundation
GK754 III analyses sex specific mechanisms in myocardial hypertrophy and heart failure. It is based
on the established projects and collaborations in
GK754 II and I. The promotion period was extended
to 2010 because of a very good evaluation in 2006.
An impaired cardiac function heart failure (HF) is one
of the most common health problems in our present
society. It develops often on the basis of myocardial
hypertrophy and manifests differently in women and
men. HF appears in women at a higher age compared
to men and shows specific features in its epidemiology, clinical manifestation, pathophysiological mechanisms, genetic bases and molecular phenotypes in the
myocardium. At the comparable levels of mechanical
stress and neurohumoral stimulation women develop
less cardiac hypertrophy and less systolic dysfunction
compared to men. However, severe cardiac hypertrophy has more adverse consequences for women. The
molecular basis of gender differences may be related
to the effect of sex hormones on the myocardium or to
X-chromosomal gene products. Experimental results
point to an impact of sexual hormones on NO-synthesis, on the gluco- and mineralocorticoidsystem, the
316
renin-angiotensin system, the
endothelin system, lipid and glucose metabolism, expression of
contractile proteins and calcium
handling, neo-aniogenesis, and
aging processes. The scientific
aim of this program (GK) is to analyse the mechanisms
of sex differences in the pathophysiology of myocardial
hypertrophy. This focus is the base for a very close
cooperation of all working groups and finds outstanding partners in the new focus of gender research in
Berlin and in our international cooperation partners. All
scientific approaches are well organized in an interdisciplinary manner. Aims and systematic teaching contents are related to biology, genomics, endocrinology,
cardiology, nephrology, pharmacology, physiology
and veterinary medicine. The involved medical students learn principles and methods of basic research,
while the scientists get across the basics of medical
science. The table shows all projects with project leaders and involved students from 2006 to 2010.
Graduate Courses
TP Project leader
Project title
PhD students
Models of hypertrophy
1
2
3
Patzak /
P. Persson
C. Schubert /
U. Kintscher
Dragun
Influence of sex on mechanisms in the development of cardiac
hypertrophy and fibrosis in NO-deficient mice
Sex differences in aortic stenosis; effects of sex hormones and
modulation of estrogen receptors
Sex specific differences in the development of left ventricular
hypertrophy in hypertensive kidney disease in mice
M. Sendeski,
O. Zavaritskaya
D. Fliegner,
C. Westphal
A. Karatas,
D. Gürgen
Hormonal effects and hormone receptors; sex dependent effects in the myocardium
4a
V. Regitz-Zagrosek
4b
S.Mahmoodzadeh
5
I. Morano
6
G. Schönfelder /
M. Paul
7a
T. Unger /
H. Funke-Kaiser /
U. Steckelings
T. Unger /
J. Li
U. Kintscher
7b
8
Effects of estrogen and testosterone on myocardial hypertrophy M. Schanz,
and matrix synthesis
H. Hampl,
A. Penkalla
Organ specific transcriptional and translational regulation of
E. Dworatzek,
estrogen receptor ER α und ER β
J. Leber
Analysis of sex specific changes in cardiac function by tranL. Schulz,
scriptional regulation of hALC-1
J. Lossie
Sex specific differences in the regulation of VEGF receptor 1
N. Wendt,
(FLT1) J. Langen,
W. Weiss,
M. Martinelli
AT2-receptor and ATBP in cardiac hypertrophy: role of sex
J. Reinemund,
differences
K. Ströder
Role of estrogen in modulation of myocardial collagen
degradation
Influence of adipose tissue on myocardial hypertrophy: relevance of Peroxisome Proliferator-Activated Receptors and
estrogen receptors
M. Brinckmann,
S. Slavic
M. Clemenz,
C. Böhm
Clinics Dependence of age and sex on gene and protein expression profile
9
P. Ruiz
Age and sex specific profiles of gene expression in myocardial
hypertrophy and relevant mechanisms
J. Isensee,
A. Queiros
10
J. Klose
Proteins of cardiac muscles in dependence on sex and age
11
R. Hetzer /
C. Knosalla
Aortic stenosis
M. Diedrich,
S. Forler
H. Pham,
C. Eschen,
G. Petrov
317
Graduate Courses
3. I nternational PHD Programme on arterial hypertension
and vascular biology
The PhD Programme on Arterial Hypertension and
Vascular Biology has been established in cooperation with the Università degli Studi di Padova (Italy)
and the Medical University of Gdansk (Poland) for
graduates wishing to attend a three-year doctoral
course, and started in the academic year 2005/06.
Research emphasises are placed on collaborative and
interdisciplinary research that ranges from molecular
biology and cell culture studies on the pathophysiology of hypertension and hypertensive end-organ disease, obesity and diabetes to defined animal models
318
of human disease processes (e.g. stroke, myocardial
infarction, hypertension, vasculopathies).The candidates fulfil the requirements for admittance to the thesis programme (Dr. rer. Medic) at the Medical Faculty
of the Charite-Universtitätsmedizin Berlin (or adequate
in Italy: laurea/laurea specialistica-magistrale). Each
PhD-student is admitted to the course at only one of
the partner universities (the Home University) but will
attend the other universities involved in the cooperation agreement for at least 9 months.
Graduate Courses
4. Initiative „Young CCR“
Coordinators
Daniela Fliegner, Pawel Namsolleck
Since 2003 we offered a weekly seminar for PhD fellows, which was part of the structured research training programmes “Gender-specific mechanisms in
myocardial hypertrophy” (graduate course 754, DFG)
and “CARDIOVASC” (Marie Curie Early Stage Training
program, EU). As this seminar was very successful
and well accepted, we wanted to keep the tradition
beyond the end of the program funding periods, albeit
in a slightly modified way. As an initiative of the junior
post-doctoral fellows Daniela Fliegner (Prof. RegitzZagrosek´s lab) and Pawel Namsollek (Prof. Unger`s
lab) the main objective here is to have a platform for
scientific exchange of young researchers. To lower
the barrier for free communication among them, the
management and moderation of the seminar will be
performed by D. Fliegner and P. Namsolleck them-
selves. The agenda of the seminar comprises not only
the discussion of scientific projects and the exchange
of knowledge, experience and methods, but also the
training of communication skills. Right in the beginning, it is planned to give an introduction to presentation techniques. In the course of the seminar, feedback
on the quality and the content of each presentation
will be elaborated by the audience. In addition, invited
speakers will give introductions to statistical analysis,
literature databases and soft skills.
The seminar is especially addressed to diploma
students, Master students and Ph.D. fellows of the
CCR. The registration is optional, but once registered
attendance is obligatory. This seminar should provide
a platform for networking and the initiation of future
collaborations.
319
RAS Symposium
Spotlight on the Renin Angiotensin System ( RAS ) –
the period from 1950 to 2010
Symposium at the Tagungszentrum Katholische Akademie in Berlin, 29th October 2010
The spotlight symposium about
the RAS honoured one of the outstanding investigators in the field of
the RAS during the last decades,
Professor Thomas Unger. On the
occasion of Professor Unger´s 60th
birthday, we have invited national
and international experts to discuss
past, recent, and future developments in the RAS and hypertension
research. The topics cover a wide range of RAS pharmacology from preclinical studies to clinical trials. The
renin-angiotensin-system (RAS) is one of the major
peptide hormone systems in our body and an impor-
tant regulator of cardiovascular function. The RAS
has been successfully targeted by various pharmacological approaches to reduce cardiovascular risk
such as ACE-inhibitors, AT1 receptor blockers, and
direct renin inhibitors. Recently, this work has been
expanded and new pharmacological RAS interventions are now being tested inpreclinical development
including AT2 receptor agonists and rennin receptor
blockers. In summary, the RAS has been extensively
studied in preclinical and clinical settings, and more
importantly, drugs acting on the RAS have markedly
improved the outcome of millions of patients suffering from cardiovascular disease such as myocardial
infarction or heart failure.
Programme
Chair:
Axel Pries, Berlin
Detlev Ganten, Berlin
12.00 – 12.30 The renin-angiotensin-system – what didn´t work?
Martin Paul, Maastricht, The Netherlands
9.00 – 09.30 Introduction
12.30 – 14.00 Lunch Break
Chair:
Ulrich Kintscher, Berlin
Thomas Unger, Berlin
9.30 – 10.00Exploiting the potential of the RAS in therapeutic
approaches Bernward Schölkens, Frankfurt
10.00 – 10.30 Arterial hypertension and RAS-blockade
Björn Dahlöf, Göteborg, Sweden
10.30 – 11.00 Coffee Break
Chair:
Reinhold Kreutz, Berlin
Gilbert Schönfelder, Berlin
11.00 – 11.30 Blockade of the RAS in Cardiology
Georg Nickenig, Bonn
11.30 – 12.00 Angiotensin receptors – an experimental view
Lutz Hein, Freiburg
320
Monika Stoll, Münster
Jürgen Scholze, Berlin
14.00 – 14.30Renin – A new therapeutic target for
cardiac hypertrophy
Massimo Volpe, Rome, Italy
14.30 – 15.00 Renal mechanisms of hypertension
Rainer Rettig, Greifswald
15.00 – 15.30 Hypertension therapy in 2010
Peter Sever, London, UK
15.30 – 16.00 New avenues from Berlin targeting the RAS
Heiko Funke-Kaiser, Berlin
Ulrike Steckelings, Berlin
16.00
End of symposium
FCVB Congress
FCVB Congress in Berlin, from 16 th to 19 th July
From July 16th to 19th the first meeting for basic and
translational research ‚Frontiers in Cardio-Vascular
Biology’ (FCVB 2010 – conference president Axel R
Pries) took place at the Institute of Anatomy of the
Charité and the CCR.
FCVB has been brought
into being by the ‚European Society for Cardiology’ (ESC) und its‚ Council for Basic Cardiovascular Research’ (CBCS)
which is constituted by Working Groups of the ESC
and scientific Societies in Europe in the cardiovascular
field (EVBO, ISHR, ESM, EAS, ECCR, AECVP). The
ESC is one of the greatest medical specialist societies
in Europe and organizes annually the world biggest
congress in the field of cardiovascular research and has
founded the ‚Council for Basic Cardiovascular Science’
including relevant external specialist societies with the
aim to support basic and translational research.
abroad. The programme included
keynote speeches and symposia by outstanding experts from
Europe and beyond, excellent
abstracts of researchers from the
whole world in the cardiovascular
field as well as very stimulating
contributions by young colleagues.
The congress was aimed to be the starting step for the
development of the most important congress in the field
of basic cardiovascular research in Europe. According
to the members of the CBCS, this goal was met and
the next FCVB will be held in 2012 at Imperial College,
London, UK under the chairmanship of Sian Harding.
A lot of researchers from the CCR participated (e.g.
Kintscher, Le Noble, Mammoozadeh, Regitz-Zagrozek, Unger) contributed and helped to strengthen the
cardiovascular ‘community’ in Berlin. A special ‘breakfast’ symposium organized at the CCR by Ulrich Kintscher got a lot of positive comments.
The FCVB was designed as a central forum for exchange
with a special emphasis on young researchers. The
congress came out with a great success including an
unexpected high participation (700 registrations, 550
free scientific abstracts) by scientists of Europe and
321
BI Lectures
BI Lectures
Since 2004, a lecture serial – called BI-Lectures– takes place at the CCR with
speakers representing international experts in the field of cardiovascular medicine.
BI-Lectures are supported by Boehringer Ingelheim Pharma GmbH & Co. KG.
Programme 2010
19.05.2010 Sebastian Frische
Regulation of renal acid excretion: how does the
kidney know what to do?
Anatomisk Institut, Åarhus Universitet,
Århus, Denmark
09.06.2010 Stephan Herzig
Transcriptional checkpoints in metabolism
and Molecularmetabolic disorders
German Cancer Research Center (DKFZ), Metabolic Control, Heidelberg, Germany
19.07.2010 Dennis Bruemmer
(Monday)
Telomeres and telomerase in vascular biology
Cardiovascular Research Center, University of
Kentucky, Lexington, USA
15.09.2010 Florian Lang
Physiology and pathophysiology of SGK1
Physiology, University of Tübingen, Tübingen,
Germany
22.09.2010 Daniel Henrion
Flow induced arterial remodeling
Laboratoire de Biologie Neurovasculaire Intégrée,
UMR CNRS 6214 - INSERM 771
Faculté de Médecine, Université d’Angers, Angers,
France
29.09.2010 Detlef Böckenhauer
Go EAST: Walking the frontier of
neuro- and nephrology
Pediatric Nephrology, Great Ormond Street
Hospital, London, UK
322
06.10.2010Rukshana Shroff
Which cellular mechanisms produce progressive
vascular damage in chronic kidney
disease?
Institute of Child Health, Great Ormond Street
Hospital, London, UK
13.10.2010Kurt A. Jäger
Modern vascular therapy - Intervention or no intervention? Novel insights into training physiology
Universitätsspital Basel, Universität Basel,
Basel, Switzerland
20.10.2010Thomas Thum
Development of microRNA-based therapeutic
strategies in cardiovascular disease
Institute for Molecular and Translational Therapeutic
Strategies, MHH, Hannover, Germany
03.11.2010
aria Grazia Modena
M
Metabolic syndrome and gender
Dipartimento ad Attività Integrata di Emergenza,
Urgenza, Università degli Studi di
Modena e Reggio Emilia, Modena, Italy
10.11.2010Michael Ristow
Promoting metabolic health and life expectancy by
increasing oxidative stress
Institute of Nutrition, Friedrich-Schiller-University
Jena, Jena, Germany
24.11.2010Denise Hilfiker-Kleiner
STAT-3 in cardiovascular pathology
Molecular Cardiology, Dept. of Cardiology and
Angiology, MHH, Hannover, Germany
Perspectives
Drug Development in Industry and Academia : the Emergence of TMAT
Garret A. FitzGerald MD, Institute for Translational Medicine and Therapeutics,
University of Pennsylvania, Philadelphia, Pa.
Background
It is now widely appreciated that the current model
of drug discovery and development is unsustainable.
While the number of new drugs approved by the FDA
has remained quite constant since 1950, the estimates
of the cost of having a drug approved have increased in
a log linear fashion (1). While some argue that the “low
hanging fruit” has been snatched, that this is a cyclical
phenomenon or that the segmentation of previous classifications of disease account for this failure, it is worth
pondering two realities. The first is that the revolution in
approaches to target discovery has been remarkably
successful; we have more rationally selected targets,
often with attendant chemistry, than ever. The second
is that the ballooning cost of bringing a drug to market
is mostly accounted for by the ballooning cost of failure
of the current model which is built into these estimates.
One might conclude that what we face is (i) a failure in
drug development rather than discovery and (ii) that
the current cost estimates largely do not apply ab initio
to a novel approach to drug development and should
not deter such innovation.
The Emerging Paradigm of Drug Development
We are presently witnessing the progressive disintegration of the vertically integrated pharmaceutical industry.
Roughly half of the companies that produced drugs
that were approved since 1950 are no longer with us (1)
and the industry’s response to production failure of its
research enterprise has been an ever more aggressive
strategy of merger. The temporary little fix of firing people
is no more likely to rescue the productivity of the industry
than is the cautious perestroika evident at the margins of
the boundaries of intellectual property (IP (2).
Perhaps the most striking hint of the future is in the “not
for profit” sector where initiatives to find new treatment
classes for previously neglected diseases is burgeoning. Here, driven by altruism, the perception that there
is little money to be made (perhaps not so true as one
witnesses the rising middle classes of the semi tropical BRICs) and charismatic leadership, central funds
have been accumulated. These are used to incent participants from both academia and industry along the
chain from target identification to large scale trials to
collapse their IP, adhere to timelines and hand off their
results to the next group along the chain.
Although conversion of this model to the for profit sector will not be instant, a truly adventurous approach to
venture capital, revisions of our outmoded and unrealistic concepts of IP, revived capacity to participate in
the academic sector and perhaps above all, a sense
of crisis within the industry, might afford the necessary
ingredients(3).
So, how are we to move to a modular approach to
drug discovery and development (4), harnessing the
heterogeneous talents necessary across sectors and
geographical boundaries?
The Deficit of Human Capital
Thirty years ago, the leading exemplars of the discipline of clinical pharmacology housed within one
organizational structure experts in cellular biology,
model systems, mechanistic studies of physiology,
disease and drug action in humans, pharmacokinet323
Perspectives
ics and modeling, chemistry, statistics and information
handling, toxicology, poisoning and clinical trials (5).
These heterogeneous talents were blended to foster
both training and the execution of what we now call
interdisciplinary science, specifically “T1” translational
research. Graduates of such programs populated
chairs of medicine and pharmacology, rose to leadership positions in industry and currently lead the exemplar UK organizations in comparative effectiveness.
Gradually, particularly as Departments of Medicine
shifted to a cost center model, they lost interest in this
discipline so that it atrophied to an essentially industry
based activity, often pursuing rather unimaginative and
routine studies of drug availability and disposition. We
have paid a price. Firstly, the most critical and expensive mistakes in drug development occur in phase II –
either failure to see an impact on surrogates of efficacy,
ignoring surrogates of risk or errors in dose selection
for phase III. Secondly, physicians in the US, just like
their patients, get most of their information from direct
to consumer advertising. Thirdly, the regulatory bodies
have minimal intramural capacity to address the mechanistic plausibility of apparent signals of risk detected by
epidemiological methods and finally expertise in basic
or human pharmacology is unapparent in approaches
to comparative effectiveness in the US.
We must rebrand and update a translational discipline
that integrates expertise in pharmacology with contemporary science. The term “Clinical Pharmacology” has
lost its luster and in contemporary perception only covers
some of what we need (6). To attract the best and brightest we need a new brand, backed by funders, academics
and industry, one that transcends geography. Potential
trainees must perceive the field to be “hot”, where the
excitement, the money and the jobs will be. We suggested the term “Translational Medicine and Therapeutics
(TMAT)” as it captures the fashion for translation, places
324
the discipline in the heart of medicine and indicates the
focus on the development of novel therapeutics (5). Initiatives on both sides of the Atlantic, by the Wellcome Trust
and the NIH, are congruent with this term.
The development of TMAT will require the development
of new interdisciplinary training structures. We favor a
Masters as a general introductory degree with subsequent areas of focus within the spectrum of TMAT. The
flexibility of a Masters to combine with lead degrees
in medicine, science, dentistry, veterinary medicine
etc makes it attractive. However, training support and
grant cycles will need to lengthen to support the particular challenges of this type of science.
Revision of Intellectual Property
There was a time when people patented transistors and we
have reached that level of absurdity in drug development.
Access to compound libraries, transparency and access
to “failed” compounds for their potential repurposing,
expansion of the precompetitive space to foster systems
pharmacology and physiology – in cells, model systems
and humans - all offer the promise of releasing value presently locked unproductively in industry. Similarly, academic
systems, patenting everything in the slim hope of finding a
Gatorade, need to get real. The unmeasured cost of these
IP stockades is the inertia that they bring to much potential
collaborative interaction across the sectors (3).
A more contentious proposal relates to the foundation of
IP, configured as it is on composition of matter. Most of
this matter never makes it to become an approved drug
and, if it does, many people of diverse skill sets have
to work effectively together. It is easy, within a vertically
integrated company, to restrict IP reward to composition of matter as the fiscal benefits that derive from this
can be distributed internally in a way that recognizes the
efforts of all the actors. However, it is a different game in
academia. How do we motivate the cardinal efforts of
Perspectives
experts in TMAT if the fiscal reward is restricted to the
chemist? Perhaps we should consider new models of
IP as well as just expanding the domains where it does
not apply. A postponed and more evenly distributed
system of reward might be necessary within the emerging context of trans-national and trans-sectoral modular
drug discovery and development.
The Informational Infrastructure
What are the foundations on which this new model
might be built? Here the challenge is to establish structures that permit the secure and compliant sharing
of highly heterogeneous data, including clinical data,
across not just geographies but also sectors. In some
ways, this is the greatest challenge of all. Partial solutions are emerging however and here, the global
Pharma industry can contribute greatly to establishing best practice.
Adventurous Venture
Finally, what provides the incentive? The Venture Capital (VC) industry has always been rather conventional
in this space and the recent recession has only served
to heighten its conservative instincts, much to the detriment of the Biotech industry in particular. Will there
be a VC investment analogous to the pot of money
contributed to by Governments, industries and individuals to incent novel behavior in the altruistic sector?
Traditionally, the experts in drug development that sit
in the Venture industry are alumni of the unsustainable
current model. When will the VC industry break free
of convention and provoke a disruptive approach to
drug development?
Conclusion
Roughly $100bn (€79bn, £67bn) in sales from medicines will be lost in the next five years as IP protections
expire, while the value of drugs in development in the
industry’s collective pipeline that could be launched
during that period are worth just $30bn. That is the
challenge to the present model of drug discovery and
development.
It seems likely that a potentially abrupt change beckons, such as we have previously observed in the airline, print media, computing and music industries. It
is unlikely that a spot of glasnost and a touch of perestroika will rescue current mores.
References:
1. Munos B. Lessons from 60 years of pharmaceutical innovation. Nat Rev Drug Discov 2009; 8:
959–968.
2. FitzGerald G.A. Perestroika in Pharma; Evolution
or Revolution in Drug Development? Mt. Sinai
Med. J. 2010; 77: 327-332.
3. FitzGerald GA. Drugs, industry and academia.
Science 2008; 320: 1563.
4. Skarke C, FitzGerald GA. Training translators for
smart drug discovery. Sci Transl Med 2010; 2:
26 cm12.
5. FitzGerald GA. Opinion: anticipating change in
drug development: the emerging era of translational medicine and therapeutics. Nat Rev Drug
Discov 2005; 4: 815–818.
6. FitzGerald GA. Clinical pharmacology or translational medicine and therapeutics: reinvent or
rebrand and expand? Clin Pharmacol Ther 2007;
81: 19–20.
Acknowledgements:
Dr. FitzGerald is the McNeill Professor of Translational
Medicine and Therapeutics.
Email: [email protected]
325