the biochemical institute christian-albrechts-university

66
The Biochemical Institute Christian-Albrechts-University Kiel 2018/2019

Upload: others

Post on 24-Jun-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Biochemical Institute Christian-Albrechts-University

The Biochemical Institute

Christian-Albrechts-University Kiel

2018/2019

Page 2: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

Contact

Biochemical Institute

Christian-Albrechts-University Kiel

Olshausenstr. 40

D-24098 Kiel

Germany

phone: +49 (0) 431/880-2211

fax: +49 (0) 431/880-2007

http: www.uni-kiel.de/Biochemie

Page 3: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

Figures from current research at the

Biochemical Institute

Page 4: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

Legends to the cover figures

Top left: Structural models of interleukin-6, CNTF and the designer cytokine IC7 and their

respective receptor assemblies (Findeisen et al, (2019) Nature).

Top right: The lysosomal membrane protein LIMP-2 involved in lysosomal cholesterol efflux

(Heybrock et al. (2019) Nat. Comm.). Depicted is the intraluminal structure of LIMP-2 with the

intramolecular hydrophobic tunnel able to transport lipids.

Bottom: Model of a membrane-bound ADAM10 based on the ectodomain (yellow, PDB:

6BE6) and pro-meprin β (blue). The pro-peptide of meprin β is shown in orange. The red arrow

indicates the shedding site within pro-meprin β (Scharfenberg et al. (2019) Cell Mol Life Sci.)

Page 5: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

1

Editorial

Our Institute is the only Biochemical Institute at the Christian-Albrechts-University in Kiel.

The main task of the Institute is the teaching of students of Human Medicine and Dental

Medicine in Biochemistry. Moreover, our Institute is engaged in a study course of Biochemistry

for natural science students. This study course is organized together with colleagues from the

Faculty of Mathematics and Natural Sciences. Besides teaching, our Institute is very active in

biomedical research as detailed in this Annual Report of the years 2018 and 2019.

In the year 2010, Prof. Stefan Rose-John together with Prof. Paul Saftig from the Institute of

Biochemistry initiated a new collaborative research center (SFB877) entitled 'Proteolysis as a

regulatory event in pathophysiology'. Part of this collaborative research center is an Integrated

Research Training group for natural science and medical graduate students, which is

coordinated by Prof. Becker-Pauly. After a positive review by the Deutsche

Forschungsgemeinschaft (DFG) in 2014 and 2018, funding of the SFB877 was granted until

summer 2022. Prof. Paul Saftig and Prof. Christoph Becker-Pauly are now in the process of

setting up a new collaborative research center for the time after the third and final period of the

SFB877. This new collaborative research center is planned to focus on the posttranslational

regulation of membrane proteins in health and disease.

In the years 2018/2019, members of the Biochemistry Institute were represented in an additional

biomedical collaborative research center (SFB841 'Liver inflammation: infection, immune

regulation und consequences'). The SFB841 was positively evaluated in summer 2017 and will

continue until the end of 2021.

The scientific work of most members of the Institute of Biochemistry is supported by grants

from the European Union and the Deutsche Forschungsgemeinschaft, including a research unit

group together with the University of Hamburg (FOR 2625), priority programs (e.g. SPP1580)

and BMBF funding (e.g. NCL2TREAT).

Since 2008, the Cluster of Excellence ‘Inflammation at Interfaces’ formed by scientists from

the University of Kiel, the University of Lübeck and the Research Center Borstel is funded by

the Deutsche Forschungsgemeinschaft. With the help of members of the Institute of

Biochemistry, the Cluster of Excellence ‘Inflammation at Interfaces’ has been renewed in 2012.

Under the new name 'Precision Medicine in Chronic Inflammation' funding of this Cluster of

Excellence has been approved for a next funding period in the year 2019.

Thus, our Institute continues to represent a spearhead in biomedical research in the Northern

Germany landscape of science. This is underlined by the fact that two junior scientists of our

Institute in 2018 were appointed W2 professors in Dresden and Magdeburg. In this brochure

you find an overview of projects of principal investigators at the Institute of Biochemistry and

a summary of the scientific achievements carried out in the Institute of Biochemistry in 2018

and 2019, along with internet addresses where you can find more detailed information.

Kiel, January 2020

Stefan Rose-John

Page 6: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

2

Contents

Editorial ...................................................................................................................................... 1 Scientific Staff Members 2018/2019 .......................................................................................... 3 1. Research Group Prof. Dr. Stefan Rose-John ....................................................................... 5 2. Research Group Prof. Dr. Joachim Grötzinger ................................................................. 13 3. Research Group Dr. Friederike Zunke .............................................................................. 19 4. Research Group PD Dr. Dirk Schmidt-Arras .................................................................... 23 5. Research Group Dr. Matthias Voss ................................................................................... 29 6. Research Group Prof. Dr. Paul Saftig ............................................................................... 33 7. Research Group PD Dr. Markus Damme .......................................................................... 41 8. Research Group Prof. Dr. Christoph Becker-Pauly .......................................................... 45 9. Research Group Prof. Dr. Hilmar Lemke ......................................................................... 53 Appendix .................................................................................................................................. 55 Biochemisches Kolloquium 2018/2019 ................................................................................... 55 Publications 2018/2019 ............................................................................................................ 56

Page 7: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

3

Scientific Staff Members 2018/2019

Scientists:

Prof. Dr. rer. nat. Stefan Rose-John – Executive Director

Prof. Dr. rer. nat. Joachim Grötzinger

Prof. Dr. rer. nat. Hilmar Lemke

PD Dr. rer. nat. Dirk Schmidt-Arras

Dr. rer. nat. Friederike Zunke

Dr. rer. nat. Matthias Voss

Prof. Dr. rer. nat. Paul Saftig – Director

PD Dr. rer. nat. Markus Damme

Prof. Dr. rer. nat. Christoph Becker-Pauly - Director

Post-Docs

Dr. rer. nat. Bleibaum, Florian

Dr. rer. nat. Di Spiezio, Alessandro

Dr. rer. nat. Kissing, Sandra

Dr. rer. nat. Marques, Andre

Dr. rer. nat. Massa Lopez , David

Dr. rer. nat. Peters, Florian

Dr. rer. nat. Rahn, Sascha

Dr. rer. nat. Scharfenberg, Franka

Dr. rer. nat. Schumacher, Neele

Doctoral students

Agthe, Maria

Armbrust, Fred

Bartels, Anne-Kathrin

Bolik, Julia

Cabrera, Florenica

Cabron, Anne-Sophie

Cappel, Cedric

Colmorgen, Cynthia

Diez Tellez, Maria

Flynn, Charlotte

Gallwitz, Lisa

Gandraß, Monja

Gonzalez, Adriana

Gradtke, Ann-Christin

Grieb, Wolfram

Heybrock, Saskia

Hobohm, Laura

Hofmann, Anna

Hülsebus, Theis

Mentrup, Torben

Pathak, Kriti

Riechmann, Mara

Page 8: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

4

Rudnik, Sönke

Sammel, Martin

Sauer, Louise-Marie

Schäfer, Miriam

Schrader, Markus

Schrell, Friederike

Seipold, Lisa

Thiessen, Niklas

Werny, Ludwig

Wessolowski, Luisa

Winkelmann, Anne

Page 9: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

5

1. Research Group Prof. Dr. Stefan Rose-John

A Group Leader: Prof. Dr. Stefan Rose-John

B Lab Members: Post-Doc:

Dr. Neele Schumacher

Doctoral Students:

Julia Bolik

Charlotte Flynn

Maria Agthe

Monja Gandrass

Anne-Sophie Cabron

Technician:

Christian Bretscher

Alyn Gerneth

Annett Lickert

C Research Report

C1 Interleukin-6 (IL-6)

The interleukin (IL)-6 family cytokines is a group of cytokines consisting of IL-6, IL-11, ciliary

neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), oncostatinM(OSM),

cardiotrophin 1 (CT-1), cardiotrophin-like cytokine (CLC), and IL-27. They are grouped into

one family because the receptor complex of each cytokine contains two (IL-6 and IL-11) or one

molecule (all others cytokines) of the signaling receptor subunit gp130. IL-6 family cytokines

have overlapping but also distinct biologic activities and are involved among others in the

regulation of the hepatic acute phase reaction, in B-cell stimulation, in the regulation of the

balance between regulatory and effector T-cells, in metabolic regulation, and in many neural

functions. IL-6 binds to the IL-6 receptor (IL-6R), forming an IL-6/IL-6R complex. This

complex thereafter associates with the signal-transducing IL-6R subunit β (also known as

gp130), initiating intracellular signaling. The thereby induced signal transduction cascade

within the cell mainly involves activation of the Janus kinase (JAK) and signal transducer and

activator of transcription (STAT) pathway (mainly via STAT1 and STAT3), and the RAS-

Page 10: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

6

dependent mitogen-activated protein kinase (MAPK) signaling cascade. Activation of

intracellular signaling via gp130 also initiates a negative feedback loop via suppressor of

cytokine signaling 3 (SOCS3), eventually leading to termination of signaling. Neither IL-6 nor

IL-6R exhibits measurable affinity for gp130; however, as a complex, IL-6 and IL-6R can bind

to and activate gp130, leading to dimerization of gp130 and intracellular signaling. In the classic

IL-6 signaling pathway, IL-6 acts via membrane-bound IL-6R and subsequently interacts with

membrane-bound gp130. Alternatively, IL-6 can bind to soluble IL-6R (sIL-6R), which is

generated by proteolytic cleavage and to a lesser extent by alternative splicing. The complex of

IL-6 and sIL-6R subsequently binds to the membrane-bound gp130 in what is known as the

trans-signaling pathway. Interestingly, whereas all cells of the body express gp130, only a few

cell types (that is, hepatocytes, some epithelial cells and some leukocytes) express IL-6R and

can therefore respond to IL-6 directly. All other cells are dependent on IL-6 trans-signaling for

their response to IL-6.

Blockade of IL-6 family cytokines has been shown to be beneficial in autoimmune diseases,

but bacterial infections and metabolic side effects have been observed. Using global blockade

of IL-6 and specific blockade of IL-6 trans-signaling in various mouse models of human

diseases led to the view that IL-6 responses mediated by the membrane-bound IL-6R are

protective and regenerative whereas IL-6 responses mediated by the sIL-6R are rather pro-

inflammatory. This might explain how a single cytokine can have pro- and anti-inflammatory

characteristics.

C.2 Development of an IL-6 based novel drug for the treatment of type II diabetes

Based on earlier structure-function work on the cytokine IL-6, we have generated a chimeric

protein of the gp130 cytokines IL-6 and Ciliary Neurotrophic Factor (CNTF). In the chimeric

protein, one gp130 binding site was removed from the IL-6 protein and replaced with the

leukemia inhibitory factor receptor (LIFR) binding site from CNTF. To increase in vivo half-

life, this chimeric protein was fused with the Fc-portion of human IgG1. This procedure created

a new cytokine with CNTF-like, but IL-6R-dependent receptor binding and signaling.

Consequently, this chimeric cytokine assembles a receptor combination (IL-6R, gp130, LIF-R)

which is not used by any natural cytokine. Since it had been shown that CNTF exhibits

beneficial metabolic functions, we analyzed the metabolic properties of the new protein. It

turned out that the novel designer protein significantly improved glucose tolerance and

hyperglycemia and prevented weight gain and liver steatosis in mice and in non-human

primates. Moreover, in multiple relevant mouse models of insulin resistance and type II

diabetes, treatment with the designer protein either increased skeletal muscle mass, or prevented

the loss of skeletal muscle mass via activation of the YAP-Hippo signaling pathway.

Additionally, in comprehensive human cell based assays, we could show that treatment with

the designer protein resulted in no signs of inflammation or immunogenicity. Thus, the new

designer protein is a realistic next generation biological for the treatment of obesity, type II

diabetes, and muscle atrophy. These are all disorders that are currently pandemic world-wide

(Findeisen et al, 2019).

C.3 The soluble interleukin-6 receptor: generation and physiologic function

The soluble interleukin-6 receptor (IL-6R) in complex with interleukin-6 (IL-6) stimulates cells

which express the signaling receptor subunit gp130 but no ligand binding IL-6R. Such cells in

the absence of the soluble IL-6R are unable to respond to IL-6. This process has been named

'trans-signaling'. Trans-signaling has been shown to be important for inflammation reactions,

neuronal survival hematopoiesis and tumor rejection.

We have characterized the metalloproteinase, which is responsible for the release of the soluble

IL-6R by biochemical and genetic means. This protease belongs to the Metalloproteinase with

Page 11: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

7

Disintegrin Domain (ADAM) family of metalloproteinases. We perform experiments to better

understand the biochemical and structural prerequisites of limited proteolysis of the IL-6R by

members of the ADAM family. We also try to understand the regulation of the cleavage

reaction. In this respect it is interesting that we could show that the induction of apoptosis leads

to activation of the shedding protease ADAM17. This seems to be a general phenomenon,

which might play an important role in the regulation of the inflammatory process. We could

show that neutrophils which are the first line of defense of the body during infection and

inflammation. Neutrophils are a major source of the soluble IL-6R in vivo. Interestingly,

induction of apoptosis leads to a selective activation of ADAM17, which in turn is responsible

for shedding of the IL-6R.

As mentioned above we could show that the protease ADAM17 (also called TACE), which is

responsible for cleavage of TNFa is also responsible for shedding of the IL-6R. Using a novel

homologous recombination strategy we have generated ADAM17 hypomorphic mice (called

ADAM17ex/ex mice) to analyze the involvement of this protease in various shedding processes.

Using these ADAM17ex/ex mice we have studied the physiologic role of cleavage of the ligands

of the EGF-Receptor (EGF-R) and other substrates.

The phenotype of the ADAM17 hypomorphic mice clearly shows an involvement of ADAM17

in inflammatory and regenerative processes. Therefore, these mice are an excellent

experimental tool to study the overall physiologic role of the ADAM17 metalloprotease and its

involvement in inflammation and cancer.

Furthermore, we could show that ADAM17 is necessary for tumor formation in the intestine.

Interestingly, ADAM17 is needed for the cleavage of ligands of the EGF-R on macrophages.

The subsequent stimulation of the EGF-R leads to the secretion of IL-6. Moreover, ADAM17

on macrophages cleaves the IL-6R. Consequently, IL-6 trans-signaling via IL-6 and sIL-6R

leads to tumor formation.

This scenario seems to be a general phenomenon since also tumor formation in the lung depends

on IL-6 trans-signaling and on the activity of ADAM17. The fact that IL-6 trans-signaling acts

downstream of the EGF-R is intriguing. The activity of the EGF-R is therapeutically inhibited

in many cancers but patients develop resistance to the treatment within few months. Inhibition

of IL-6 trans-signaling might open a second therapeutic window after failure of anti-EGF-R

drugs.

C.4 Viral Interleukin-6: Structure, Pathophysiology and Strategies of Neutralisation

On target cells, Interleukin-6 (IL-6) binds to a receptor complex consisting of the ligand binding

subunit Interleukin-6-receptor (IL-6R) and the signal transducing subunit gp130. The complex

of soluble IL-6R and IL-6 acts on cells, which do not express IL-6R. Such cells would not be

able to respond to IL-6 alone. Such cells comprise hematopoietic progenitor cells, endothelial

cells, smooth muscle cells, T-cells and neural cells. Interestingly, the recently identified viral

IL-6 (vIL-6) encoded by Human Herpes Virus 8 (HHV8) binds to and activates gp130 directly.

Therefore, vIL-6 activates a significantly larger spectrum of target cells than human IL-6. We

characterized the biochemical and physiological properties of vIL-6. Furthermore, we have

generated transgenic mice, which overexpress vIL-6. Using these mice we evaluate the

involvement of vIL-6 in human diseases like Castleman disease primary effusion lymphoma

and multiple myeloma.

Using a novel strategy we used our recently generated recombinant antibodies against vIL-6 to

target the vIL-6 within cells expressing the protein. The underlying principle of this strategy is

to anchor the recombinant vIL-6 antibodies within the endoplasmic reticulum (ER) with the

help of the canonical ER retention sequence KDEL. Indeed we could demonstrate that vIL-6

can induce signaling from within the cell and that such signaling can be completely blocked

from within the cell.

Page 12: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

8

We have performed structure-function analysis to clarify how vIL-6 can bind directly to gp130

whereas human IL-6 needs the IL-6R in order to bind to and activate gp130. These data clearly

show that the so-called site III of vIL-6 is responsible for this property and that the ability to

directly bind to gp130 can be transferred to human IL-6 by transferring site III.

Transgenic mice, which over-express the vIL-6 protein show a phenotype resembling human

Castleman disease. These data indicate that vIL-6 is strongly involved in the pathophysiology

of the HHV8 virus. Interestingly, the Castleman disease phenotype of the vIL-6 transgenic mice

depends on endogenous IL-6 since vIL-6 transgenic mice on an IL-6-/- background do not

develop such a Castleman disease phenotype. These results explain why in HHV8 associated

human Castleman disease therapy with anti-human-IL-6R neutralizing antibody (Tocilizumab)

was successful.

C.5 Generation of constitutively active gp130

We have generated a constitutively active gp130 protein by exchanging at the cDNA level the

entire extracellular domain of gp130 with a leucin zipper. This leads to dimerization of gp130

and subsequently to permanent activation of gp130 mediated signaling. We could show that the

constitutively active gp130 cDNA, when transfected into IL-6 dependent cells, rendered these

cells cytokine independent. The constitutively active gp130 protein was named L-gp130. These

experiments showed that positive signaling of gp130 was dominant over the negative feed-back

loop mediated by SOCS3.

Interestingly, when introduced into mouse bone marrow cells led to the development of multiple

myeloma with high penetrance. L-gp130–expressing mice recapitulated all of the

characteristics of human disease, including monoclonal gammopathy, bone marrow infiltration

with lytic bone lesions, and protein deposition in the kidney. The disease was transplantable

and allowed evaluation of therapeutic strategies in vivo.

We recently generated mice in which the L-gp130 cDNA was introduced into the ROSA26

locus of mice. This allows the cell autonomous activation of gp130signaling in vivo using

appropriate cre-transgenic animals. This novel mouse model will help to analyze the precise

role of cell-specific gp130 signaling in many IL-6 target cells without flooding an animal with

the cytokine IL-6, which leads to the activation of many target cells.

In a first example we studied the cell-autonomous activation of gp130 in B-cells. Regardless of

the timing of gp130 activation during B-cell development, constitutively active gp130 signaling

resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders

with full penetrance. constitutively active gp130 signaling in all adult hematopoietic cells also

led to the development specifically of largely mature, aggressive B cell cancers. We concluded

from these studies that gp130 signaling selectively provides a strong growth and differentiation

advantage for mature B cells, and directs lymphomagenesis specifically towards terminally

differentiated B cell cancers (for details, see Scherger et al, 2019).

C.6 Development of the IL-6 trans-signaling antagonist sgp130Fc

We could show in the past years that IL-6 trans-signaling can specifically be inhibited by the

sgp130Fc protein without affecting IL-6 signaling via the membrane bound IL-6R. These data

established the sgp130Fc protein not only as a molecular tool to experimentally distinguish

between classic- and trans-signaling. The sgp130Fc protein can also be used to block the course

of inflammatory diseases in animal models of rheumatoid arthritis, peritonitis, inflammatory

bowel disease and many animal models of inflammation induced cancer.

We have improved the properties of the sgp130Fc protein in terms of protein stability, affinity

towards the IL-6/sIL-6R complex and feasibility of production.

Since the sgp130Fc protein has a considerable therapeutic potential, Stefan Rose-John together

with Prof. Stefan Schreiber (Director of the Clinic of Internal Medicine at the University

Hospital in Kiel) founded a biotechnology company (Conaris Research Institute), which

Page 13: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

9

developed the sgp130Fc protein into a drug. In December 2008 Conaris Research Institute and

the company Ferring have signed an exclusive worldwide license agreement for the

development of sgp130Fc for inflammatory conditions such as inflammatory bowel disease and

rheumatoid arthritis. This led to the GMP-production of the sgp130Fc protein. The sgp130Fc

protein has already been successfully tested in the clinic in phase I clinical trials in 2013 and

2014. Currently, a phase II clinical trial with the sgp130Fc protein, which has been renamed by

the WHO to 'Olamkicept', is running with inflammatory bowel disease patients at the Clinic of

Internal Medicine at the University Hospital in Kiel. Moreover, in collaboration with the

Chinese Company iMAB, a phase II clinical trial with inflammatory bowel disease patients is

currently running in China, Taiwan and South Korea.

D Publications 2018/2019

Publications 2018 Impact Factor

Armacki M, Trugenberger AK, Ellwanger AK, Eiseler T, Schwerdt C, Bettac L,

Langgartner D, Azoitei N, Halbgebauer R, Groß R, Barth T, Lechel A, Walter

BM, Kraus JM, Wiegreffe C, Grimm J, Scheffold A, Schneider MR, Peuker K,

Zeißig S, Britsch S, Rose-John S, Vettorazzi S, Wolf E, Tannapfel A, Steinestel

K, Reber SO, Walther P, Kestler HA, Radermacher P, Barth TF, Huber-Lang M,

Kleger A, Seufferlein T (2018) Thirty-eight-negative kinase 1 mediates trauma-

induced intestinal injury and multi-organ failure. J Clin Invest 128: 5056-5072

12.282

Cabron AS, El Azzouzi K, Boss M, Arnold P, Schwarz J, Rosas M, Dobert JP,

Pavlenko E, Schumacher N, Renné T, Taylor PR, Linder S, Rose-John S, Zunke

F (2018) Structural and Functional Analyses of the Shedding Protease ADAM17

in HoxB8-Immortalized Macrophages and Dendritic-like Cells. J Immunol 201:

3106-3118

4.718

Stahl FR, Jung R, Jazbutyte V, Ostermann E, Tödter S, Brixel R, Kemmer A,

Halle S, Rose-John S, Messerle M, Arck PC, Brune W, Renné T (2018)

Laboratory diagnostics of murine blood for detection of mouse cytomegalovirus

(MCMV)-induced hepatitis. Sci Rep 8: 14823

4.011

Lücke K, Yan I, Krohn S, Volmari A, Klinge S, Schmid J, Schumacher V,

Steinmetz OM, Rose-John S, Mittrücker H-W (2018) Control of Listeria

monocytogenes infection requires classical IL-6 signaling in myeloid cells.

PlosOne 13: e0203395

2.776

Agthe M, Brugge J, Garbers Y, Wandel M, Kespohl B, Arnold P, Flynn CM,

Lokau J, Aparicio-Siegmund S, Bretscher C, Rose-John S, Waetzig GH, Putoczki

T, Grötzinger J, Garbers C (2018) Mutations in Craniosynostosis Patients Cause

Defective Interleukin-11 Receptor Maturation and Drive Craniosynostosis-like

Disease in Mice. Cell Rep 25: 10-18.e5

7.815

Holz K, Prinz M, Bredecke SM, Mittrücker H-W, Rose-John S, Hölscher C

(2018) Differing outcome of experimental autoimmune encephalitis in

macrophage/neutrophil- and T cell-specific gp130-deficient mice. Front

Immunol 9: 836

4.716

Kaiser K, Prystaz K, Vikman A, Haffner-Luntzer M, Bergdolt S, Strauss G,

Waetzig GH, Rose-John S, Ignatius A (2018) Pharmacological inhibition of IL-

6 trans-signaling improves compromised fracture healing after severe trauma.

Naunyn Schmiedebergs Arch Pharmacol 391: 523-536

2.058

Fuchslocher Chico J, Falk-Paulsen M, Luzius A, Saggau C, Ruder B, Bolik J,

Schmidt-Arras D, Linkermann A, Becker C, Rosenstiel P, Rose-John S, Adam D

(2018) The enhanced susceptibility of ADAM-17 hypomorphic mice to DSS-

induced colitis is not ameliorated by loss of RIPK3, revealing an unexpected

function of ADAM-17 in necroptosis. Oncotarget 9: 12941-12958

Rose-John S (2018) IL-6 family cytokines. Cold Spring Harb Perspect Biol 10:

pii: a028415. doi: 10.1101/cshperspect.a028415 9.110

Prystaz K, Kaiser K, Kovtun A, Haffner-Luntzer M, Fischer V, Strauss G,

Waetzig GH, Rose-John S, Ignatius A (2018) Distinct effects of interleukin-6

classic and trans-signaling in bone fracture healing. Am J Pathol 188: 474-490

3.762

Page 14: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

10

Garbers C, Rose-John S (2018) Dissecting lnterleukin-6 Classic- and Trans-

Signaling in Inflammation and Cancer. Meth Mol Biol 1725: 127-140 1.290

Garbers C, Heink S, Korn T, and Rose-John S (2018) Interleukin-6: Designing

specific therapeutics for a complex cytokine. Nat Rev Drug Discov 17: 395-412 57.618

Schmidt S, Schumacher N, Schwarz J, Roos S, Kenner L, Schlederer M, Sibilia

M, Linder M, Altendorf-Hofmann A, Knoesel T, Gruber E, Oberhuber G,

Rehman A, Sinha A, Arnold P, Zunke F, Becker-Pauly C, Preaudet A, Nguyen

P, Huynh J, Chand A, Westermann J, Dempsey PJ, Garbers C, Rosenstiel

P, Putoczki T, Ernst M, Rose-John S (2018) ADAM17 is required for EGF-R

induced intestinal tumors via IL-6 trans-signaling. J Exp Med 215: 1205-1225

10.892

Ullrich E, Abendroth B, Rothamer J, Huber C, Büttner-Herold M, Kitowski V,

Vogler T, Longerich T, Zundler S, Völkl S, Beilhack A, Rose-John S, Wirtz

S,Weber GF, Ghimire S, Kreutz M, Holler E, Mackensen A, Neurath MF,

Hildner K (2018) BATF-dependent IL-7RhiGM-CSF+ T cells control intestinal

graft-versus-host disease. J Clin Invest 128: 916-930

12.282

Dixit A, Bottek J, Beerlage AL, Schuettpelz J, Thiebes S, Brenzel A, Squire A,

Garbers G, Rose-John S, Mittruecker HW, Engel DR (2018) Proliferation

of Ly6C+ monocytes during urinary tract infection is regulated by IL-6 trans-

signaling. J Leukoc Biol 103: 13-22

4.012

Nicolaou A, Northoff BH, Zhao Z, Kohlmaier A, Sass K, Rose-John S, Steffens

S, Weber C, Teupser D, Holdt LM (2018) The ADAM17 metalloproteinase

maintains arterial elasticity. Thromb Haemost. 118: 210-213

4.662

Publications 2019 Impact Factor

dos Santos Guilherme M, Stoye NM, Rose-John S, Garbers C, Fellgiebel A,

Endres K (2019) The synthetic retinoid acitretin increases IL-6 in the central

nervous system of Alzheimer disease model mice and human patients. Front

Aging Neurosci 11: 182

4.504

Scharfenberg F, Helbig A, Sammel M, Benzel J, Schlomann U, Peters F, Wichert

R, Bettendorff M, Schmidt-Arras D, Rose-John S, Moali C, Lichtenthaler SF,

Pietrzik CU, Bartsch JW, Tholey A, Becker-Pauly C (2019) Degradome of

soluble ADAM10 and ADAM17 metalloproteases. Cellular and Molecular Life

Sciences, doi: 10.1007/s00018-019-03184-4

7.014

Scherger AK, Al-Maarri M, Maurer HC, Schick M, Maurer S, Öllinger R,

Gonzalez-Menendez I, Martella M, Thaler M, Pechloff K, Steiger K, Sander S,

Ruland J, Rad R, Quintanilla-Martinez L, Wunderlich FT, Rose-John S, Keller U

(2019) Activated gp130 signaling selectively targets B cell differentiation to

promote transformation of mature lymphoma and plasmacytoma. JCI Insight 4:

pii: 128435

6.014

Saad MI, McLeod L, Yu L, Ebi H, Ruwanpura S, Sagi I, Rose-John S, Jenkins BJ

(2019) The ADAM17 protease promotes tobacco smoke carcinogen-induced lung

tumorigenesis. Carcinogenesis doi: 10.1093/carcin/bgz123

4.004

Aparicio-Siegmund S, Garbers Y, Flynn CM, Waetzig GH, Gouni-Berthold I,

Krone W, Berthold HK, Laudes M, Rose-John S, Garbers C (2019) The IL-6-

neutralizing sIL-6R/sgp130 buffer system is disturbed in patients with type 2

diabetes mellitus. Am J Physiol 317(2):E411-E420

4.125

Sommer D, Corstjens I, Sanchez S, Dooley D, Lemmens S, Van Broeckhoven J,

Bogie J, Vanmierlo T, Vidal PM, Rose-John S, Gou-Fabregas M, Hendrix S

(2019) ADAM17-deficiency on microglia but not on macrophages promotes

phagocytosis and functional recovery after spinal cord injury. Brain Behav

Immun 80:129-145

6.170

Pavlenko E, Cabron A-S, Arnold P, Dobert JP, Rose-John S Zunke F (2019)

Functional characterization of colon cancer-associated mutations in ADAM17:

modifications in the pro-domain interfere with trafficking and maturation. Int J

Mol Sci 20: pii: E2198

4.183

Escrig A, Molinero A, Méndez B, Sanchis P, Fernández-Gayol O, Montilla A,

Comes G, Giralt M, LaFerla FM, Giménez-Llort L, Becker-Pauly C, Rose-John

S, Hidalgo J (2019) IL-6 trans-signaling in the brain influences the behavioral

6.170

Page 15: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

11

and physiological phenotype of the 3xTgAD mouse model of Alzheimer’s

disease. Brain Behav Immun 82: 145-159

Servais FA, Kirchmeyer M, Hamdorf M, Minoungou N, Rose-John S, Kreis S,

Haan C, Behrmann I. Modulation of the IL-6 signaling pathway in liver cells by

miRNAs targeting gp130, JAK1 and/or STAT3 (2019) Molecular Therapy -

Nucleic Acids 16: 419-433

5.919

Saad MI, Alhayyani S, McLeod L, Yu L, Alanazi M, Deswaerte V, Tang K,

Jarde T, Smith JA, Prodanovic Z, Watkins N, Cain JE, Bozinovski S, Algar E,

Ferlin W, Garbers C, Ruwanpura S, Sagi I, Rose-John S, Jenkins BJ (2019)

ADAM17 selectively activates the IL-6 trans-signaling/ERK MAPK axis in

KRAS-addicted lung cancer. EMBO Mol Med pii: e9976

10.624

Ziegler L, Gajulapuri A, Frumento P, Bonomi A, Wallén H, de Faire U, Rose-

John S, Gigante B (2019) Interleukin 6 Trans-Signalling and Risk of Future

Cardiovascular Events. Cardiovasc Res 115: 213-221

7.014

Aden K, Bartsch K, Dahl J, Reijns MAM, Esser D, Sheibani-Tezerji R, Sinha A,

Wottawa F, Ito G, Mishra N, Knittler K, Burkholder A, Welz L, van Es J, Tran F,

Lipinski S, Kakavand N, Boeger C, Lucius R, von Schoenfels W, Schafmayer C,

Lenk L, Chalaris A, Clevers H, Röcken C, Kaleta C, Rose-John S, Schreiber S,

Kunkel T, Rabe B, Rosenstiel P (2019) Epithelial RNase H2 Maintains Genome

Integrity and Prevents Intestinal Tumorigenesis in Mice. Gastroenterology 156:

145-159.e19

19.233

Kleinegger F, Hofer E, Wodlej C, Golob-Schwarzl N, Birkl-Toeglhofer AM,

Stallinger A, Petzold J, Orlova A, Krassnig S, Reihs R, Niedrist T, Mangge H,

Park YN, Thalhammer M, Aigelsreiter A, Lax S, Garbers C, Fickert P, Rose-

John S, Moriggl R, Rinner B, Haybaeck J (2019) Pharmacologic IL-6Rα

inhibition in cholangiocarcinoma promotes cancer cell growth and survival.

Biochim Biophys Acta Mol Basis Dis 1865: 308-321

4.328

Saad MI, Rose-John S, Jenkins BJ (2019) ADAM17: An emerging therapeutic

target for lung adenocarcinoma. Cancers 11: E1218 6.162

Quarta S, Mitrić M, Kalpachidou T, Mair N, Schiefermeier-Mach N, Andratsch

M, Qi Y, Langeslag M, Malsch P, Rose-John S, Kress M (2019) Impaired

mechanical, heat and cold nociception in a murine model of genetic

TACE/ADAM17 knockdown. FASEB J 33: 4418-4431

5.391

Schmidt-Arras D, Rose-John S (2019) Regulation of fibrotic processes in the

liver by ADAM proteases. Cells 8: 1226 5.656

Prenissl N, Lokau J, Rose-John S, Haybaeck J, Garbers C (2019) Therapeutic

blockade of the interleukin-6 receptor (IL-6R) allows sIL-6R generation by

proteolytic cleavage. Cytokine 114: 1-5

3.078

Paige E, Clément M, Lareyre F, Sweeting M, Raffort J, Grenier C, Finigan A,

Harrison J, Peters JE, Sun BB, Butterworth AS, Harrison SC, Bown MJ, Lindholt

JS, Badger SA, Kullo IJ, Powell J, Norman PE, Scott JA, Bailey MA, Rose-John

S, Danesh J, Freitag DF, Paul DS, Mallat Z (2019) Interleukin-6 Receptor

Signalling and Abdominal Aortic Aneurysm Growth Rates. Circ Genom Precis

Med 12: e002413

4.743

Schumacher N, Rose-John S (2019) ADAM17 activity and IL-6 Trans-Signaling

are required for EGF-R driven Colon Cancer. Cancers 11: E1736 6.162

Findeisen M, Allen TL, Henstridge DC, Kammoun HL, Brandon AE, Baggio LL,

Watt KI, Pal M, Cron L, Estevez E, Yang C, Kowalski GM, O’Reilly L, Egan C,

Sun E, Thai LM, Krippner G, Adams TE, Lee RS, Grötzinger J, Garbers C, Risis

S, Kraakman MJ, Mellet N, Sligar J, Kimber ET, Young RL, Cowley MA, Bruce

CR, Meikle PJ, Baldock PA, Gregorevic P, Biden TJ, Cooney GJ, Keating DJ,

Drucker DJ, Rose-John S & Febbraio MA (2019) Treatment of type 2 diabetes

and muscle atrophy with the designer cytokine IC7Fc. Nature 574: 63-68

43.070

Wilkinson A, Gartlan K, Kuns R, Chang K, Minnie S, Ensbey K, Clouston A,

Zhang P, Koyama M, Hidalgo J, Rose-John S, Varelias A, Vuckovic S, Hill G

(2019) IL-6 dysregulation originates in dendritic cells and initiates graft-versus-

host disease via classical signaling. Blood 134: 2092-2106

16.562

Page 16: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

12

Heichler C, Scheibe K, Schmied A, Geppert C, Schmid B, Wirtz S, Thoma A-M,

Kramer V, Waldner M, Büttner , Henner F, Farin H, Pesic M, Knieling F, Merkel

S, Grüneboom A, Gunzer M, Grützmann R, Rose-John S, Koralov SB, Kollias G,

Vieth M, Hartmann A, Greten FR, Neurath MF, Neufert C (2019) STAT3

activation through IL-6/IL-11 in cancer-associated fibroblasts promotes

colorectal tumor development and correlates with poor prognosis. Gut, pii:

gutjnl-2019-319200

17.943

Liao C-W; Chou C-H; Wu X-M; Chen Z-W; Chen Y-H; Chang Y-Y; Wu V-C;

Rose-John S; Hung C-S; Lin Y-H (2019) Interleukin-6 plays a critical role in

aldosterone-induced macrophage recruitment and infiltration in the myocardium.

Biochim Biophys Acta – Molecular Basis of Disease165627

4.328

Sammel M, Peters F, Lokau J, Scharfenberg F, Werny L, Linder S, Garbers C,

Rose-John S, Becker-Pauly C (20190) Differences in shedding of the interleukin-

11 receptor by the proteases ADAM9, ADAM10, ADAM17, meprin α, meprin β

and MT1-MMP. Int J Mol Sci 20: E3677

4.183

Wichert R, Scharfenberg F, Koudelka T, Colmorgen C, Schwarz J, Wetzel S,

Potempa B, Potempa J, Bartsch JW, Sagi I, Tholey A, Saftig P, Rose-John S,

Becker-Pauly C (2019) Meprin β induces activities of A Disintegrin and

Metalloproteinases 9, 10 and 17 by specific prodomain cleavage. FASEB J 33:

11925-11940

5.391

Impact factors 2018: 142.004

Impact factors 2019: 211.761

Total impact factors 2018/2019: 353.965

E Grants

E.1 The role of gp130-Trans-Signaling in liver-regeneration and -cancer: therapeutic perspectives

(Together with Dirk Schmidt-Arras), Collaborative Research Centre 841. Project C1 (DFG. Germany)

Total granted sum: (2018 – 2021) 488,800 €

E.2 Analysis of the role of the shedding protease ADAM17 in vivo

Collaborative Research Centre 877. Project A1 (DFG, Germany)

Total granted sum: (2018 – 2022) 380,300 €

E.3 Collaborative Research Centre 877. Central Tasks (DFG, Germany)

Total granted sum: (2018 – 2022) 1,230,300 €

E.4 Genetic Analysis of the Role of IL-6 in Prostate Cancer

German Cancer Aid (Grant 70112589)

Granted Amount (2017 – 2020): 265,780 €

E.5 Inhibiting malignant EGF-R activity by targeting ADAM17 (TACE) as a novel alternative therapeutic

strategy (Together with Prof. Irit Sagi, Weizmann Institute, Israel)

German-Israeli Foundation for Scientific Research and Development

Granted Amount (2017 – 2020): 200,000 €

Page 17: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

13

2. Research Group Prof. Dr. Joachim Grötzinger

A Group Leader: Prof. Dr. rer. nat. Joachim Grötzinger

B Lab Members:

Doctoral students

Theis Hülsebus

Miriam Schäfer

Anne-Kathrin Bartels

Markus Schrader

Diploma/Bachelor/Master students:

Fabian Uhrlaub

Technicians

Alyna Lycke (Trainee Lab. Technician)

Lea Egli (Trainee Lab. Technician)

C Research Report

C.1 A disintegrin and metalloprotease 17 (ADAM17)

In contrast to many other signalling mechanisms shedding of membrane-anchored proteins is

an irreversible process. A Disintegrin And Metalloproteinase (ADAM) 17 is one of the major

sheddases involved in a variety of physiological and pathophysiological processes including

regeneration, differentiation, and cancer progression. Due to its central role in signalling the

shedding activity of ADAM17 is tightly regulated, especially on the cell surface, where

shedding events take place. The activity of ADAM17 can be subdivided into a catalytic activity

and the actual shedding activity. Whereas the catalytic activity is constitutively present, the

shedding activity has to be induced and is tightly controlled to prevent pathological situations

induced by the release of ist substrates. The regulation of the shedding activity of ADAM17 is

multilayered and different regions of the protease are involved. Intriguingly, its extracellular

domains play crucial roles in different regulatory mechanisms.

Page 18: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

14

Figure 1: ADAM17 activity has to be tightly controlled to allow physiological processes and to prevent

pathological situations. ADAM17 sheds more than 70 different substrates; therefore, the enzyme acts as a major

sheddase in various physiological and protective processes, such as immune defence, coagulation, and

regeneration. By contrast, the uncontrolled release of such potent substrates is associated with numerous

pathological conditions, such as uncontrolled inflammation (autoimmune diseases and chronic inflammation) and

cancer progression. Hence, ADAM17 is initially kept in an inactive state, e.g, by interaction with inhibitory

proteins such as β1-integrin. Only upon activation by various stimuli, such as LPS, ATP, and thrombin, does

ADAM17 shed its substrates from the cell surface, thereby changing their activities. Later, the enzyme is switched

off. This switching off is catalysed by protein disulfide isomerases (PDIs) and is accompanied by a structural

change within the extracellular part of ADAM17, which prevents substrate recognition and shedding.

A prominent class of ADAM17 substrates are growth factors of the EGFR ligand family. These

substrates are expressed as membrane-bound preforms; therefore, they are not active prior to

shedding. The significance of this process in development is displayed by the perinatality of

ADAM17-deficient mice, which show comparable phenotypes to EGFR ligand-deficient mice,

including malformations of the skin, hair, eye, heart, and lung. In adult organisms, generation

of soluble EGFR ligands is essential for proper regeneration processes.

These few examples (and ADAM17 processes more than 70 substrates) illustrate the necessity

of this enzyme for appropriate physiological processes such as immune defence and

regeneration. However, they also indicate that uncontrolled ADAM17 activity supports

misguided inflammation, such as occurs in autoimmune diseases (e.g, rheumatoid arthritis),

chronic inflammation (e.g, Crohn's disease), and cancer progression, due to mistimed and

enhanced release of pro-inflammatory mediators and growth factors. Hence, ADAM17 has to

be tightly controlled to avoid pathophysiological situations and, accordingly, it is of

extraordinarily high therapeutic interest. A prerequisite to modulate ADAM17 activity is an in-

depth knowledge of the molecular basis of its functioning, including its on and off switches,

because ADAM17 has to be activated to become proteolytically active and inactivated to

prevent damage. The cytoplasmic tail of ADAM17 is dispensable for its activation by numerous

stimuli; therefore, we focus on its extracellular part.

In its mature form, ADAM17 lacks its pro-domain, which retains the zymogen in an inactive

state and is removed during maturation in the Golgi apparatus. As the names suggest, the

catalytic domain cleaves the substrates and the disintegrin domain interacts with β1-integrin.

This interaction supports cell-cell contacts between fibroblasts and tumour cells and keeps the

enzyme inactive, most likely due to steric hindrance of substrate recognition. Activation of β1-

integrin leads to the release of ADAM17 accompanied by the catalytically active enzyme.

Page 19: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

15

Figure 2: (A) Schematic drawing of the domain structure of ADAM17. (B) A structural model of the

extracellular region of ADAM17.

Due to the impact of the MPD in ADAM17 regulation and activity, we were interested in what

it actually looks like. Hence, we produced recombinant MPD using bacterial expression

systems for structural determination by multidimensional heteronuclear NMR spectroscopy.

Soluble expressed MPD is a disulfide isomer that is only partially structured and is a flexible

elongated domain (Figure 3). The MPD comprises a classical thioredoxin motif (C600KVC),

which is essential for ADAM17 shedding activity, and exogenous PDI supresses ADAM17

activity. Therefore, we hypothesise that PDI targets this specific motif. Indeed, PDI catalyses

disulfide isomerisation within the flexible open MPD. Two disulfide bonds thereby undergo

specific isomerisation. This process changes the affected disulfide pattern from a sequential

arrangement (C600-C630 and C635-C641), which is present in the elongated MPD structure,

to an overlapping pattern (C600-635 and C630-641), which exists in a compact, rigid-structured

MPD. Whereas the latter isoform is associated with the inactive enzyme, the flexible form is

associated with active ADAM17. Hence, the MPD acts as the off-switch of ADAM17, which

is operated by exogenous PDI.

Fig. 3. Ribbon representation of the MPD structure. The

upper, green depicted part of the MPD is not altered by the

PDI-mediated isomerisation, while the lower, red depicted

part, is flexible in the open isoform but becomes rigid in

the closed one. The disulphide bonds, which undergo

isomerisation, as well as the positive charged residues,

which form the phosphatidylserine interaction site, are

shown in the open isoform.

Recently, the crucial role of the MPD in the shedding mechanism beyond substrate recognition

was revealed. Since the absence of the cytoplasmic region does not impair ADAM17 activity

but all stimulators of ADAM17-mediated shedding activate intracellular signalling pathways,

other ways must exist to transmit the activation signal through the membrane to the cell surface.

We revealed that ADAM17 stimuli such as PMA or Melittin also lead to the activation of

Page 20: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

16

scramblases, which transfer the negatively charged phospholipid phosphatidylserine to the cell

surface. Under normal conditions, phosphatidylserines are located in the inner leaflet of the cell

membrane. Upon translocation to the outer leaflet, they act as negatively charged interaction

hubs for proteins, which either possess clusters of positively charged residues or bind cations.

The positively charged sequence R625-K626-G627-K628 within the MPD binds to these

negative hubs on the membrane which leads to the shedding process (Fig. 4). It is assumed that

the shedding process is initialised because the binding of the MPD to the membrane is

accompanied by a conformational change within the whole ectodomain and thereby brings the

catalytic site in close proximity to the membrane-proximal cleavage sites of ADAM17

substrates (Fig. 4). Notably, the phosphatidylserine binding sequence in the MPD is in the

flexible part of the domain, which becomes rigid after PDI-mediated isomerisation. Only the

open MPD conformation binds to phosphatidylserine, while the closed form does not. This

explains why the closed conformation corresponds to the shedding inactive state of the protease

(Fig. 4).

Fig. 4. Schematic depiction of the shedding process. Under normal conditions ADAM17 rests in cholesterol-rich

membrane microdomains. Upon scramblase activation a lipid redistribution between the outer and inner leaflet of

the membrane takes place which leads to an exposure of phosphatidylserine at the cell surface. As a result,

CANDIS as well as the open MPD, through its RKGK-motif, bind to the membrane. This interaction results in a

different positioning of the cleavage site of the substrate within the active site of ADAM17. In case of type-1

transmembrane substrates another crucial step of the shedding process is the binding of parts of their stalk region

to CANDIS.

D Publications 2018/2019

Publications 2018 Impact Factor

Machado-Pineda, Y., Reyes, R., Cardenes, B., Loppez-Martin, S., Toriba, V. Sanchez-

Organero, P., Grötzinger, J., Lorenzen, I., Yanez-Mo, M., Cabanas, C. (2018) CD9

controls integrin a5b1-mediated cell adhesion by modulating its assciciation with the

metalloproteinase ADAM17. Front. Immunol. 9, 2474.

5.511

Page 21: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

17

Agthe, M., Brügge, J., Garbers, Y., Wandel, M., Kespohl, B., Arnold, P., Flynn, C.M.,

Lokau, J., Bretscher, C., Waetzig, G.H., Putoczki, C.G., Grötzinger, J., Garbers, C.

(2018) Mutations in Craniosynostosis Patients Cause Defective Interleukin-11 Receptor

Maturation and Drive Craniosynostosis-like Disease in Mice. Cell Rep 25, 10-18.

8.282

Agthe, M., Garbers, Y., Grötzinger, J., Garbers, C. (2018) Two N-linked glycans

differentially control maturation and trafficking, but not activity of the interleukin-11

receptor. Cell Physiol Biochem 45, 2071-2085.

5.104

Krossa, S., Scheidig, A.J., Grötzinger, J., Lorenzen, I. (2018) Redundancy of protein

disulfide isomerases in the catalysis of the inactivating disulfide switch in A Disintegrin

and Metalloprotease 17. Sci Rep 8, 1103.

4.259

Lokau, J. Göttert, S., Arnold, P., Düsterhöft, S., Massa López, D., Grötzinger, J.,

Garbers, C. (2018) The SNP rs4252548 (R112H) which is associated with reduced

human height compromises the stability of IL-11. BBA-Mol Cell Res 1865, 496-506.

4.521

Publications 2019 Impact Factor

Marques, A.R.A., Di Spiezio, A., Thießen, N., Schmidt, L., Grötzinger, J., Lüllmann-

Rauch, R., Storck, S.E., Pietrzik, C.U., Fogh, J., Bär, J., Mikhaylova, M., Glatzel, M.,

Bassal, M., Bartsch, U., Paul Saftig, P. (2019) Enzyme replacement therapy with

recombinant pro-cathepsin-D corrects defective proteolysis and autophagy in Neuronal

Ceroid Lipofuscinosis. Autophagy 16, 1-15.

11.100

Bartels, A-K., Göttert, S., Desel, C., Schäfer, M., Krossa, S., Scheidig, A.J., Grötzinger,

J., Lorenzen, I. (2019) KDEL receptor 1 contributes to cell surface association of protein

disulfide isomerases. Cell Physiol Biochem 52,850-868

5.104

Findeisen, M., Allen, T.L., Henstridge, D.C., Kammoun, H., Brandon, A.E., Baggio,

L.LWatt, K.I., Pal, M., Cron, L., Estevez, E., Yang, C., Kowalski, G.M., O’Reilly, L.,

Egan, Sun, C., Thai, L.M., Krippner, G., Adams, T.E., Lee, R.S., Grötzinger, J., Garbers,

C., Risis, S., Kraakman, M.J., Mellet, N.A., Sligar, J., Kimber, E.T.,. Young, R.L.,

Cowley, M.A., Bruce, C.R., Meikle, P.J., Baldock, P.A., Gregorevic, P., Biden, T.J.,

Cooney, G.J., Keating, D.J., Drucker, D.J., Rose-John., S., Febbraio, M.A. (2019)

Treatment of type 2 diabetes with the designer cytokine IC7Fc. Nature 574, 63-68

43.070

Bleibaum, F., Sommer, A., Veit, M., Rabe, B., Andrä, J., Kunzelmann, K., Nehls, C.,

Correa, W., Gutsmann, T., Grötzinger, J., Bhakdi, S., Reiss, K. (2019) ADAM10

sheddase activation is controlled by cell membrane symmetry. J Mol Cell Biol. 11:979-

993.

3.813

Impact factors 2018: 27.677

Impact factors 2019: 63.087

Total impact factors 2016/2017: 90.764

E Grants E.1 Structure-function analysis of the extracellular part of ADAM17. DFG SFB 877-A6, Total granted sum:

(2014-2018): 301.200 €.

Page 22: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

18

Page 23: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

19

3. Research Group Dr. Friederike Zunke

A Group Leader: Dr. rer. nat. Friederike Zunke

B Lab Members: Doctoral students

Alice Drobny (M.Sc.)

Susy Prieto Huarcaya (M.Sc.)

Theresia Schlothauer (medical student)

Josina Bunk (medical student;

10/18-10/19)

Anne-Sophie Cabron (M.Sc.)

(10/16- 08/19)

Scientists

Jan Dobert (B.Sc.)

Annika Kluge Dr. med.

Master students

Egor Pavlenko (B.Sc.)

Bisher Eymsh (B.Sc.; joint project with

Philipp Arnold, Anatomical Institute)

Bachelor students

Lina Walther

Deniz Caylioglu (finished 07/18)

Niklas Meyer (finished 10/19)

Technicians

Melanie Boss

Dwayne Götze (trainee)

C Research Report

C.1 The role of lysosomal cathepsins on -synuclein homeostasis

Within the cell, lysosomes are the central compartments of degradation and recycling. A

decrease in its proteolytic capacity has been implicated in a number of neurodegenerative

disorders. In Parkinson disease (PD) -the second most common neurodegenerative disorder-

degeneration of dopaminergic neurons accounts for the disease-specific motor symptoms. PD

pathology is characterized by the accumulation of -synuclein, which converts from a soluble

Page 24: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

20

synaptic protein into insoluble amyloid fibrils eventually leading to pathogenic Lewy body

formation.

Although the lysosomal cathepsins D, B and L (CTSD, CTSB, CTSL) have been recently

suspected to be involved in lysosomal -synuclein degradation (Figure 1), their precise

molecular role in regulating a-synuclein homeostasis is still elusive. Moreover, recent genome

screenings in PD patients uncovered a genetic correlation between CTSD as well as CTSB with

PD, emphasizing a role of both proteases in PD development. To date it is unknown to what

extent CTSD, CTSB or CTSL are involved in -synuclein turnover in human neurons and thus

might contribute to PD development.

Hence, we are studying the effects of deficiencies as well as upregulation of lysosomal

cathepsins on -synuclein homeostasis.

Our tools mainly comprise established human neuronal cell lines as well as dopaminergic

neurons derived from induced pluripotent stem cells (iPSC) from Parkinson’s disease patients.

Taken together, we want to understand the role of CTSD, CTSB and CTSL activity on -

synuclein degradation and evaluate if they could be exploited as therapeutic target in PD.

Figure 1: Lysosomal degradation and aggregation of a-

synuclein.

This overview shows the lysosomal degradation of a-

synuclein by CSTD, CTSB and CTSL. The acidic

lysosomal pH accelerates -synuclein to aggregate,

leading to oligomeric species as well as amyloidogenic a-

synuclein fibrils. Hereby, also glycolipids can interfere

with a-synuclein leading to soluble oligomeric structures.

These oligomeric -synuclein species have been shown to

exert negative effects on cell homeostasis, like for example

the ER-Golgi transport pathway.

C.2 The role of -synuclein on cell homeostsis

The intracellular aggregation of -synuclein has been shown to exert negative effects on cell

homeostasis. For instance, -synuclein oligomers have been shown to block intracellular

protein trafficking. However, the underlying molecular mechanisms are still elusive. Hence, we

are studying intracellular protein trafficking in human -synuclein overexpressing neuronal cell

lines. Moreover, we are also interested in other cellular pathways that seem to be affected by

pathological -synuclein conformers.

C.3 -Synuclein structure and aggregation pathways

Surprisingly, a central event in Parkinson’s disease pathology -the aggregation of -synuclein-

is not well understood. Hence, we are studying the aggregation pathways of the protein in cells

as well as with in vitro methodology, including CD-spectroscopy, electron microscopy and

biochemical analyses like cell toxicity, dot blots and seeding assays.

Moreover, in a project together with the Neurology Department (Prof. Daniela Berg) we are

investigating and biochemically characterizing -synuclein conformers found in colon biopsies

of Parkinson disease patients as well as control individuals. We hope that this will help to

understand the role of the intestine in Parkinson disease progression.

C.4 Cryo-EM analysis of the GCase-LIMP-2 protein complex

Page 25: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

21

In a project funded by the Michael J. Fox Foundation we are analyzing the complex of the

lysosomal hydrolase -Glucocerebrosidase (GCase) and the lysosomal membrane protein

LIMP-2. In this project we aim to decipher the structure of the complex of both proteins by

cryo-electron microscopy (in collaboration with Dr. Philipp Arnold (Anatomy Kiel) and Dr.

Arne Möller (MPI Frankfurt)). Since GCase has been shown to be a therapeutic target for

Parkinson’s disease, we hope to better understand the interaction with LIMP-2, which has been

shown to stabilize/chaperone the GCase enzyme.

C.5 Understanding regulation of ADAM17 and its role in neurogenesis

Members of the ADAM family have emerged as major ectodomain shedding proteases. Within

the ADAM family, ADAM17 and its closest relative ADAM10 mediate most cleavage events

on the cell surface. Thus, to date more than 80 substrates have been identified for ADAM17.

Among these substrates are growth factors and cytokines such as TNF, ErbB-ligands and their

receptors, as well as the Interleukin-6 Receptor (IL-6R) and cell adhesion molecules. We use

the ADAM17ex/ex mice, which exhibit only minimal ADAM17 expression to study the role of

ADAM17 in inflammatory disorders, cancer development as well as neuronal differentiation

and homeostasis. Recent studies indicated regulation of proteolytic ADAM17 activity by

cellular processes such as cytoplasmic phosphorylation and removal of the pro-domain by furin

cleavage. The goal of our group is to shed light on biological mechanisms by which ADAM17

recognizes stimuli and becomes catalytically active. Therefore, we are analyzing maturation

and activity of various ADAM17 mutants. So far, studies of ADAM17 maturation have been

mainly limited to mouse embryonic fibroblasts (mEF) or transfected cell lines relying on non-

physiologic stimuli such as the phorbolester (PMA), making interpretation of the results

difficult in a physiologic context. Hence, we introduced a robust cell system to study ADAM17

maturation and function in primary cells of the immune system. This hoxB8-conditionally-

immortalized macrophage precursor (MØP) cell lines derived from bone marrow of wildtype

and hypomorphic ADAM17ex/ex mice, lacking measurable ADAM17 activity. ADAM17

mutants were stably expressed in these MØP cells, which can be further differentiated to

macrophage- and dendritic-like cells by applying different growth factor conditions (M-

CSF/GM-CSF). Our aim is to study maturation, activation and function of the respective

ADAM17 mutants in this physiological cell system to gain more information about ADAM17

regulation and function in immune cells.

Moreover, we utilize various human cell models to study the role of ADAM17 in neuronal

development. By inhibiting and knocking down the protease in cell models of SH-SY5Y, iPS

cells and gingival mesenchymal stem cells (GMSC; taken from gum of patients) we study the

effect of ADAM17 deficiency in neuronal differentiation and homeostasis. Moreover, we

monitor the neuronal development in mice with ADAM17ex/ex background.

C.6 Functional analysis of human colon cancer-associated ADAM17 mutants

As stated above, ADAM17 was shown to be involved in the development of EGF-R-induced

intestinal cancer. Thereby the precise mechanistic role of ADAM17 in colon cancer

development still remains elusive. In this part of the project we are focusing on identified

ADAM17 point mutations found in genomic screening of human patients diagnosed with colon

cancer. In total 12 exonic mutations have been identified in colon cancer patients and found

localized spread all over the protein. To better understand the role of ADAM17 in colon cancer

development, we are interested in the biology of each ADAM17 mutant. Therefore, we are

analyzing cellular localization, maturation and activity of each mutant. Initial experiments

revealed a strong interference with ADAM17 activity when mutations were localized in the

catalytic domain, but also in other parts of the protein.

D Publications 2018/2019

Page 26: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

22

Publications 2018

Cabron AS, El Azzouzi K, Boss M, Arnold P, Schwarz J, Rosas M, Dobert JP, Pavlenko E,

Schumacher N, Renne T, Taylor PR, Linder S, Rose-John S, Zunke F (2018) Structural and

Functional Analyses of the Shedding Protease ADAM17 in HoxB8-Immortalized

Macrophages and Dendritic-like Cells. J Immunol 201: 3106-3118

4.718

Schmidt S, Schumacher N, Schwarz J, Roos S, Kenner L, Schlederer M, Sibilia M, Linder M,

Altendorf-Hofmann A, Knösel T, Gruber ES, Oberhuber G, Rehman A, Sinha A, Arnold P,

Zunke F, Becker-Pauly C, Preaudet A, Nguyen P, Huynh J, Chand AL, Westermann J,

Dempsey PJ, Garbers C, Rosenstiel P, Putoczki T, Ernst M, Rose-John S (2018) ADAM17 is

required for EGF-R induced intestinal tumors via IL-6 trans-signaling. J Exp Med,

215(4):1205-1225

10.892

Zunke F, Moise AC, Belur NR, Gelyana E, Stojkovska I, Dzaferbegovic H, Toker NJ, Jeon S,

Fredriksen K, Mazzulli JR (2018) Reversible Conformational Conversion of alpha-Synuclein

into Toxic Assemblies by Glucosylceramide. Neuron 97: 92-107. 14.403

Publications 2019 Impact Factor

Cuddy LK, Wani WY, Morella ML, Pitcairn C, Tsutsumi K, Fredriksen K, Justman CJ,

Grammatopoulos TN, Belur NR, Zunke F, Subramanian A, Affaneh A, Lansbury PT Jr,

Mazzulli JR. (2019) Stress-Induced Cellular Clearance Is Mediated by the SNARE Protein

ykt6 and Disrupted by α-Synuclein. Neuron. 104: 869-884.

14.403

Zunke F, Mazzulli JR (2019) Modeling neuronopathic storage diseases with patient-derived

culture systems. Neurobiol Dis 127: 147-162 5.16

Riederer P, Berg D, Casadei N, Cheng F, Classen J, Dresel C, Jost W, Kruger R, Muller T,

Reichmann H, Riess O, Storch A, Strobel S, van Eimeren T, Volker HU, Winkler J,

Winklhofer KF, Wullner U, Zunke F, Monoranu CM (2019) alpha-Synuclein in Parkinson's

disease: causal or bystander? J Neural Transm (Vienna) 126: 815-840

2.903

Pavlenko E, Cabron AS, Arnold P, Dobert JP, Rose-John S, Zunke F (2019) Functional

Characterization of Colon Cancer-Associated Mutations in ADAM17: Modifications in the

Pro-Domain Interfere with Trafficking and Maturation. Int J Mol Sci 20

4.183

Impact factors 2018: 30.013

Impact factors 2019: 26.649

Total impact factors 2018/2019: 56.662

E Grants E.1 ParkinsonFonds Germany (in collaboration with Prof. D. Berg, Neurology Department, UKSH Kiel);

01.11.2019 – 30.10.2021;

188.600 €

E.2 Michael J Fox Foundation / Silverstein Foundation; 01.01.2019 – 31.03.2020;

128.322 €

E.3 The role of lysosomal Cathepsins in -Synuclein Metabolism and Parkinson’s Disease, Collaborative

Research Center 877, Project B11; 01.07.2018 – 31.06.2022;

236.000 €

E.4 Junior support (Collaborative Research Center 877); 01.07.2018 - 31.06.2021;

176.775 €

E.5 Kiel Life Science Research Grant (in collaboration with Dr. Philipp Arnold (Anatomy) and Dr. Julia

Keppler (Food Technology); 01.01.2019 – 31.05.2020;

18.000 €

E.6 Kiel Life Science Early Career Postdoc Award; 6.000 €

E.7 Intramurale Forschungsförderung der Medizinischen Fakultät (CAU Kiel); 01.01.2018 – 28.02.2019;

30.000 €

Schilling Research Award 2019 of the German Neuroscience Society

Page 27: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

23

4. Research Group PD Dr. Dirk Schmidt-Arras

A Group Leader: PD Dr. rer. nat. Dirk Schmidt-Arras

B Lab Members: PostDocs:

Dr. rer. nat. Neele Schumacher

Doctoral students:

Julia Bolik

Monja Gandraß

Master students:

Sagar Ajmera

Freia Krause

Bachelor students:

Mouhamad Khouja

Ilka Thomsen

Sarah Schacht

Luise Gorki

Niklas Weißer

Technicians:

Fabian Neumann

Pit Christoffersen

Elias Diallo (tech. trainee)

C Research Report:

Our group is interested in the molecular cues underlying cancer formation and metastasis with

the general aim of finding novel concepts for therapeutic intervention. Using state-of-the-art

biochemical and cell biological methods as well as genetic mouse models we are trying to

understand the complex nature of tumor-stroma and tumor-metastatic niche interactions as well

as the role of cytokine signaling in gastroenterological pathologies predisposing to tumour

formation. Especially the formation of malignant hepatic lesions like HCC and liver metastasis

of solid tumors are in the focus of our interest.

Page 28: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

24

C.1 Inflammatory signals driving gastroenterological pathologies

Inflammatory cytokines are involved in many gastrointestinal pathologies including

inflammatory bowel disease, chronic liver disease and intestinal and hepatic tumorigenesis. Our

group is interested in deciphering molecular cues underlying these processes.

Aging is characterised by a progressive of loss of many functions including a decline in DNA-

damage repair. In cooperation with the laboratory of Prof. Jacob Rachmilewitz we

demonstrated that age- and diet-induced changes in intestinal microbiota are responsible for a

chronic low tone inflammatory response in the liver, characterised by the secretion of IL-1β

and TNFα by liver-resident Kupffer cells (Fig. 1 A+B). This phenomenon, also called

“inflamm-aging” is reverted in germ-free mice (Fig. 1 B) and mice treated with antibiotics.

TNFα, as well as epidermal growth factor (EGF) receptor ligands are released from the surface

of macrophages through limited proteolysis mediated by the metalloprotease ADAM17.

ADAM17-mediated release of EGFR ligands is necessary to induce IL-6 expression and

secretion in tumour-associated macrophages (Schmidt et al. 2018). ADAM17 is furthermore

able to proteolytically release the soluble form of the IL-6 receptor α (sIL-6RA). In complex

with IL-6 the sIL-6RA is able to induce signalling in cells that express the IL-6 receptor β-

subunit gp130 but not the IL-6RA, a process termed IL-6 trans-signalling. We previously

demonstrated that IL-6 trans-signalling promotes hepatocarcinogenesis (Bergmann et al. 2017).

Similarly, ADAM17 and IL-6 trans-signalling also promotes intestinal tumorigenesis (Schmidt

et al. 2018).

Using both, ADAM17ex/ex mice with ubiquitous ADAM17-deficiency or LysM-

Cre::ADAM17fl/fl mice with myeloid-specific ADAM17-deficiency, we furthermore detected

that myeloid ADAM17 is essential for HB-EGF mediated DNA-damage response (data not

shown) and regulates the susceptibility to develop hepatocellular cancer (data not shown).

Figure 1: Inflamm-aging

impairs DNA-damage repair.

A. During aging and Western-

type diet, alterations in intestinal

microbiota induces activation of

liver-resident Kupffer cells and

subsequent TNFα secretion

which impairs HB-EGF

mediated increase in DNA-

damage repair. B. Inflammatory

cytokines TNFα and IL-1β are

reduced in germ-free mice

compared to mice kept under

specified pathogen free (SPF)

conditions. C. DNA damage

repair, as assessed by γH2Ax

staining, is enhanced in aged

germ-free mice as compared to

aged mice kept under specified

pathogen free conditions.

Page 29: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

25

However, gp130 is ubiquitously expressed in the body. It is therefore difficult to delineate

effects of IL-6 signaling in a cell type-specific manner. In order to circumvent this problem, we

employ a synthetic biology approach. Lgp130 is an artificial gp130 variant, in which the

extracellular domain of gp130 is replaced by the c-Jun leucin zipper. We inserted a Cre-

inducible expression cassette coding for Lgp130 and the ZsGreen fluorescent reporter protein

into the ROSA26 locus. We bred these mice to Alb-CreERT2 and Vil-CreERT2 mice to induce

ligand-independent cell-autonomous gp130 activation in hepatocytes or intestinal epithelial

cells, respectively (Fig. 2 C+D). We used a multi-omics approach to analyze mice with

hepatocytic gp130 activation. These animals display significant alterations in gene expression

with an enrichment of genes associated with the innate immune system (Fig. 2E). Accordingly,

we observed alterations in acute-phase protein translation in the liver and secretion to the

plasma, as well as alterations in the cellular composition of the innate immune system (data not

shown). Consequently, these mice display an increased ability to defend bacterial pathogens

(Fig. 2 E). A manuscript summarizing these data is currently in preparation. Mice with gp130

activation in intestinal epithelial cells show alterations in local production of acute-phase

proteins and anti-microbial peptides in both, small intestine and colon (data not shown). We are

currently using both mouse models to analyze the effects of systemic and local elevation in

acute-phase proteins and anti-microbial peptides on intestinal microbiota composition and the

consequence on intestinal and hepatic inflammation, DNA-damage repair and tumorigenesis.

Figure 2: A novel synthetic biology-based mouse model to study cell-autonomous gp130 activation. A.

Schematic representation of Lgp130. B. LSL-Lgp130 targeting construct. C. Lgp130 expression exclusively

localises to hepatocytes upon tamoxifen (TAM) expression in Rosa26lgp130/lgp130::Alb-CreERT2 mice. D. Genes

involved in the innate immune system are significantly enriched in the liver of mice with hepatocytic Lgp130-

expression. E. Analysis of CFSE-labeled listeria clearance via liver-resident Kupffer cells. F. Lgp130 expression

localises to intestinal epithelial cells upon tamoxifen (TAM) expression in Rosa26lgp130/lgp130::Vil-CreERT2 mice.

G. Both mouse models with either systemic elevation in acute-phase proteins (APP, left panel), as well as local

elevation in acute-phase proteins (APP) or anti-microbial peptides (AMPs) are currently used to analyse their

effects on intestinal microbiota composition, gastrointestinal inflammation, DNA-damage repair and tumour

formation.

We furthermore identified and characterized a novel cytokine loss-of-function mutation in

gp130 which was associated with craniosynostosis. We demonstrate an exclusive defect in IL-

11 signaling of this mutant. Using a CRISPR/Cas approach we generated mice carrying a gp130

point mutation equivalent to the human mutation. These mice also display impaired IL-11

signaling and in consequence develop facial synostosis (data not shown). A manuscript

summarizing these data has been recently submitted.

Page 30: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

26

Cooperations:

Jacob Rachmilewitz, Eithan Galun, Hadassah Medical Center, Hebrew University Jerusalem,

Israel

Hans-Willi Mittrücker, Institute of Immunology, University Medical Center Hamburg-

Eppendorf, Germany

Robert Häsler, Philipp Rosenstiel, Institute of Clinical Molecular Biology, Christian-

Albrechts-University Kiel, Germany

Hartmut Schlüter, Thomas Renné, Department of Clinical Chemistry, University Medical

Center Hamburg-Eppendorf, Germany

Holm Uhlig, Translational Gastroenterology Unit, John Radcliffe Hospital, University of

Oxford, Oxford, UK

Irm Hermans-Borgmeyer, Center for Molecular Neurobiology Hamburg (ZMNH), Hamburg,

Germany

Holger Glüers, Section Biomedical Imaging, Department of Radiology and Neuroradiology,

University Medical Center Schleswig-Holstein, Kiel, Germany

Jürgen Scheller, Institute of Biochemistry and Molecular Biology II, Medical Faculty,

Heinrich-Heine-University, Düsseldorf, Germany

C.2 The impact of ADAM proteases on tumour growth and metastasis

Members of the A Disintegrin and Metalloprotease (ADAM) family are major mediators of

receptor shedding. The family member ADAM10 is well known as major sheddase for the

Notch receptor. We previously identified ADAM10 as a major regulator of liver tissue

homeostasis and potential regulator of liver progenitor cells (Müller et al. 2016). We now

identified a non-catalytic function of ADAM10 in the regulation of liver progenitor cell biology.

We detected a C-terminal proteolytic fragment of ADAM10 that translocated to the nucleus to

regulate gene expression in liver progenitor cells (Fig. 3 A). Also in liver tissue sections from

Figure 3: Tumorigenesis and

metastasis is driven by

ADAM proteases. A. A C-

terminal fragment of ADAM10

localises to the nucleus of

BMOL liver progenitor cells

upon cleavage by ADAM9 or

15. B. A6+ liver progenitor

cells accumulate in livers of

ADAM10-deficient mice in the

thioacetamide (TAA) chronic

liver damage/tumorigenesis

model. C. Expression of stem

cell-associated Cd133 is

increased in mice with

ADAM10-deficient liver

progenitor cells. D. Ubiquitous

ADAM17-deficiency leads to

reduced lung metastasis. E.

ADAM17 on endothelial cells

promotes metastasis to the

lung. LLC: Lewis lung

carcinoma cells, B16F1:

melanoma cells.

Page 31: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

27

patients with chronic liver disease, nuclear ADAM10 can be detected (data not shown). Genetic

loss of ADAM10 in biliary epithelial cells and liver progenitor cells led to an elevation of A6+

liver progenitor cells in these mice and increased the susceptibility to develop hepatic fibrosis

(Fig. 3 B). Furthermore, we confirmed our in vitro findings in these mice that a non-catalytic

function of ADAM10 suppresses expression of stem cell-associated genes such as Cd133 (Fig.

3 C). A manuscript containing these data has been recently submitted.

Metastasis is the leading cause of death in cancer patients. ADAM17 has been shown to be

upregulated on a number of tumor cells. However, little is known about the role of ADAM17

in the tumor stroma and cells of the metastatic niche. Using a murine lung metastasis model we

observed that mice developed significantly less metastasis in the absence of ADAM17 in the

metastatic niche. This effect was irrespective of the metastasizing tumour entity and is therefore

a general mechanism (Fig. 3 D). We furthermore demonstrate that ADAM17 on endothelial

cells controls tumour cell extravasation and metastatic outgrowth in the lung (Fig. 3 E). We link

ADAM17 activity to cell death receptor signalling in endothelial cells (data not shown). A

manuscript summarizing the data of this project has been recently submitted.

Collaborations:

Henning Walczak, University College of London

Ingo Ringshausen, Department of Hematology, School of Clinical Medicine, University of

Cambridge, UK

Achim Krüger, Institute of Molecular Immunology and Experimental Oncology, TU München

Irit Sagi, Department of Biological Regulation, Weizmann Institute Rehovot, Israel

Tom Lüdde, University Medical Center, RWTH Aachen

Dieter Adam, Stefan Schütze, Institute of Immunology, UKSH Kiel

Paul Saftig, Institute of Biochemistry, CAU Kiel

Karel Chalupsky, Radislav Sedlacek, Institute of Molecular Genetics of the ASCR, Prague,

Czech Republic

Nina Tirnitz-Parker, School of Biomedicine, Curtin University, Perth, Australia

Grant Ramm, QIMR Berghofer Medical Research Institute, Brisbane, Australia

Jörg Heeren, Christoph Schramm, University Medical Center Hamburg-Eppendorf

D Publications 2018/2019:

Publications 2018 Impact Factor

Hedemann N, Rogmans C, Sebens S, Wesch D, Schmidt-Arras D, Pecks U, van

Mackelenbergh M, Weimer J, Arnold N, Maas N, Bauerschlag DO. ADAM17 inhibition

enhances platinum efficience in ovarian cancer. (2018) Oncotarget 9(22):16043-16058

Fuchslocher-Chico J, Falk-Paulsen M, Luzius A, Saggau A, Ruder B, Bolik J, Schmidt-

Arras D, Linkermann A, Becker C, Rosenstiel P, Rose-John S, Adam D, The enhanced

susceptibility of ADAM-17 hypomorphic mice to DSS-induced colitis is not ameliorated

by loss of RIPK3, revealing an unexpected function of ADAM-17 in necroptosis. (2018)

Oncotarget 9(16):12941-12958

Barikbin R, Berkhout L, Bolik J, Schmidt-Arras D, Ernst T, Ittrich H, Adam G, Parplys

A, Casar C, Krech T, Karimi K, Sass G, Tiegs G. Early HO-1 induction has beneficial

effects on inflammation and malignant transformation in mdr2-/- mice (2018) Sci Rep

Nov 2;8(1):16238

4.1

Schmidt S, Schumacher N, Schwarz J, Tangermann S, Kenner L, Schlederer M, Sibilia

M, Linder M, Altendorf-Hofmann A, Knösel T, Gruber ES, Oberhuber G, Bolik J,

Rehman A, Sinha A, Lokau J, Arnold P, Cabron AS, Zunke F, Becker-Pauly C, Preaudet

10.892

Page 32: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

28

A, Nguyen P, Huynh J, Afshar-Sterle A, Chand AL, Westermann J, Dempsey P, Garbers

C, Schmidt-Arras D, Rosenstiel P, Putoczki T, Ernst M, Rose-John S. ADAM17 is

required for EGF-R-induced intestinal tumors via IL-6 trans-signaling, (2018) J Exp Med

215(4):1205-1225.

Publications 2019 Impact Factor

Schmidt-Arras, D and Rose-John, S. Regulation of Fibrotic Processes in the Liver by

ADAM Proteases. (2019) Cells 8(10) doi: 10.3390/cells8101226. epub ahead of print 5.6

Bolik J, Tirnitz-Parker JEE and Schmidt-Arras D, ADAM and ADAMTS proteases in

hepatic disorders, J Ren Hepat Disord 2019;3(1):23–32 not yet assigned

Scharfenberg F, Helbig A, Sammel M, Benzel J, Schlomann U, Peters F, Wiechert R,

Bettendorff M, Schmidt-Arras D, Rose-John S, Moali C, Lichtenthaler SF, Pietrzik CU,

Bartsch JW, Tholey A and Becher-Pauly D. Degradome of soluble ADAM10 and

ADAM17 metalloproteases (2019) Cell Mol Lif Sci doi: 10.1007/s00018-019-03184-4.

epub ahead of print

5.9

Guedj A, Volman Y, Geiger-Maor A, Bolik J, Kuenzel S, Nevo Y, Baines J, Elgavish S,

Galun E, Amsalem H, Schmidt-Arras D, Rachmilewitz J. Gut microbiota shape 'inflamm-

aging' cytokines and account for age-dependent decline in DNA damage repair (2019)

Gut doi: 10.1136/gutjnl-2019

17.9

Impact factors 2018: 16.0 Impact factors 2019: 28.5

Total impact factors 2018/2019: 44.5

E. Grants E.1 The role of gp130-Trans-Signaling in liver-regeneration and -cancer: therapeutic perspectives together with Stefan Rose-John

Collaborative Research Centre 841. Project C1. (DFG. Germany). Total granted sum: (2018-2021) €

479,200 E.2 Metabolic functions of the metalloprotease ADAM10 in the liver together with Karel Chalupsky (Institute of Molecular Genetics of the ASCR, Prague, Czech Republic) CAS-DAAD 57315783 exchange program. Total granted sum: (2017-2018) €8,648 E.3 gp130 as essential regulator of the mamma epithelium together with Dirk Bauerschlag (Clinic of Gynecology, University hospital Kiel), Medical Faculty, CAU Kiel F358901. Total granted sum: €60,800

Page 33: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

29

5. Research Group Dr. Matthias Voss

A Group Leader: Dr. rer. nat. Matthias Voss

B Lab Members:

Doctoral students

Laura Hobohm

Anna Hofmann

Diploma/Bachelor/Master students:

Lukas Heintz

Technicians

Stefanie Schnell

Amelie Mies (trainee)

C Research Report

The research group was established late in 2018 when Dr. Matthias Voss was recruited as a

junior group leader from Karolinska Institutet, Stockholm, Sweden. The group’s research

interests focus on the proteolytic cleavage and secretion of Golgi-resident glycan-modifying

enzymes and its physiological implications (C.1) as well as the biological function of SAMD9

and SAMD9L, two tumor suppressors that have recently come into spotlight due to their

association with severe human disease (C.2).

C.1 Proteolytic processing of Golgi-resident glycan-modifying enzymes

Glycosylation is a posttranslational modification commonly found on secreted and membrane

proteins in eukaryotic cells. In particular, in multicellular organisms it contributes to various

processes ranging from protein quality control in the secretory pathway to intercellular

communication and alterations in glycosylation have been observed in numerous pathological

conditions, e.g. in cancer. While protein N-glycosylation is initiated in the ER, the vast diversity

of glycan structures observed in eukaryotes is generated in the Golgi network through the

activity of > 300 glycosyltransferases and other glycan-modifying enzymes (Fig. 1A). With

only few exceptions, these enzymes are type II membrane proteins which are distributed in the

Golgi stacks in an asymmetric, yet orderly fashion to ensure correct glycosylation. Soluble

forms of many – if not all – of these enzymes can, however, be detected in bodily fluids and

cell culture supernatants demonstrating that they are secreted in vitro and in vivo. For this this

to occur, the Golgi-resident proteins need to be proteolytically cleaved off their membrane

anchors (Fig. 1B). The protease(s) catalyzing Golgi glycan-modifying enzymes cleavage, thus

Page 34: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

30

Fig. 1: Golgi glycan-modifying enzymes and their proteolysis. (A) Glycosylation in the Golgi network. Glycans found on

nascent glycoproteins passing through the Golgi network are subject to additional modifications by numerous Golgi-resident

glycosylation enzymes which are distributed asymmetrically (orange, pink, green, blue). In addition tot he organelle-resident

enzymes, numerous Golgi glycosylation enzymes have been observed in the extracellular space. (B) Proteolytic processing of

e.g. Golgi glycosyltransferases (GT) and their subsequent secretion affects cellular Golgi glycosylation.

enabling their secretion, have been elusive for a long time until BACE1 emerged as a first

candidate protease implicated in proteolysis of sialyltransferases. More recently, however, we

showed that SPPL3, an evolutionarily conserved, Golgi-resident intramembrane protease,

cleaves numerous Golgi glycan-modifying enzymes.

In spite of these findings, proteolysis-dependent secretion of glycan-modifying enzymes

remains a unique and poorly understood process. On the one hand, activity of

glycosyltransferase and other glycan-modifying enzymes is required within the Golgi stack to

ensure a sufficient degree of glycosylation of nascent proteins and lipids. In line with this,

alterations in SPPL3 expression, i.e. following overexpression or knockdown/knockout led to

global changes in the cellular glycome. On the other hand, among the > 300 membrane-

anchored Golgi glycosylation enzymes, proteolytic processing and secretion is highly

prevalent, suggesting a fundamental physiological importance of this process warranting further

investigation.

Our newly established group is establishing molecular tools to further dissect Golgi enzyme

proteolysis and secretion and aims to understand the physiological implications of this

phenomenon. We assume that latter relates to the regulation of Golgi network homeostasis.

Therefore, we want to dissect further to which extent BACE1 and SPPL3 contribute to this

process and whether other – and, if so, which – proteases similarly cleave Golgi enzyme

substrates. Moreover, we want to dissect in finer detail on a spatiotemporal scale where and

when cleavage of certain glycosyltransferase substrates by BACE1 and SPPL3 occurs and how

this can be aligned with our current understanding of Golgi organization and function. For these

purposes we are currently developing suitable overexpression-based cell culture models but at

the same time are also exploring endogenous tagging using genome editing techniques.

C.2 The cellular function of the twin tumor suppressors SAMD9 and SAMD9L

SAMD9 and SAMD9L, located in direct adjacency on chromosome 7q in humans, encode two

unique, yet evolutionarily conserved tumor suppressors that have recently moved into the

spotlight as mutations in both genes have been linked to severe human diseases. Of note, both

gain-of-function and loss-of-function variants have been described in distinct disease contexts,

Page 35: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

31

suggesting that tightly controlled

activity of both tumor suppressors is important. In spite of the disease relevance, the precise

cellular function of SAMD9 and SAMD9L has until now not been determined. Both genes are,

however, interferon-regulated and appear to be active against a wide range of DNA and RNA

viruses suggesting that SAMD9 and SAMD9L are part of the cellular innate antiviral defense.

The mechanisms underlying this antiviral activity have yet also remained elusive.

Homology-based domain predictions suggest a multi-domain structure shared by SAMD9 and

SAMD9L which includes domains which could engage in interactions with protein or nucleic

acid interaction partners. This suggests that SAMD9 and SAMD9L may form a multimeric

intracellular complex to exert their anti-proliferative and potentially antiviral functions.

Identification of cellular interaction partners thus will be instrumental to specifically pinpoint

cellular pathways activated by SAMD9 and SAMD9L. To capture protein-protein interactions

that dynamically and selectively change upon virus infection, we are performing proximity

biotinylation experiments. These datasets will enable us to more precisely dissect SAMD9 and

SAMD9L biology in the context of virus infection, which will ultimately help to pinpoint the

mechanisms underlying the diseases associated with SAMD9 and SAMD9L variants.

D Publications 2018/2019

Publications 2018 Impact

Factor

Tesi B, Rascon J, Chiang SCC, Burnyte B, Löfstedt A, Fasth A, Heizmann M, Juozapaite S,

Kiudeliene R, Kvedaraite E, Miseviciene V, Muleviciene A, Müller ML, Nordenskjöld M,

Matuzeviciene R, Samaitiene R, Speckmann C, Stakeviciene S, Zekas V, Voss M, Ehl S, Vaiciene-

Magistris N, Henter JI, Meeths M, Bryceson YT (2018). A RAB27A 5’UTR structural variant

associated with late-onset hemophagocytic lymphohistiocytosis and normal pigmentation. J Allergy

Clin Immunol 142, 317-321.

14.110

Pastor VB, Sahoo SS, Boklan J, Schabe GC, Saribeyoglu E, Strahm B,Lebrecht D, Voss M,

Bryceson YT, Erlacher M, Ehninger C, Niewisch M, Schlegelberger B, Baumann I, Achermann

JC, Shimamura A, Hochrein J, Tedgård U, Nilsson L, Hasle H, Boerries M, Busch H, Niemeyer

CM, Wlodarski MW (2018). Constitutional SAMD9L mutations cause familial myelodysplastic

syndrome and transient monosomy 7. Haematologica 103, 427-437.

7.570

Publications 2019 Impact

Factor - -

Impact factors 2018: 21.680

Impact factors 2019: -

Total impact factors 2018/2019: 21.680

E Grants E.1 Der Zusammenhang der onkogenen und der antiviralen Eigenschaften von SAMD9 und SAMD9L.

Medical Faculty, Kiel University, Total granted sum (2019-2020): 58,000 €.

E.2 Systematic analysis of proteolysis-dependent secretion of Golgi-resident enzymes. Kiel Life Science

Young Scientist Programme, Total granted sum: (2019): 5,000 €.

E.3 Dissecting the role of SAMD9 and SAMD9L in cell intrinsic defence against viruses and myeloid

malignancy. Svenska Sällskapet för Medicinsk Forskning, Total granted sum (2018-2019): 80,000 €. E.4 Junior support (Collaborative Research Center 877); 01.07.2018 - 31.06.2021; 176.775 €

Fig. 2: Schematic overview of SAMD9

and SAMD9L domain structure as

predicted by Mekhedov et al (2017).

Hotspots for patient gain-of-function

(GoF) and loss-of-function (LoF)

variants are depicted.

Page 36: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

32

Page 37: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

33

6. Research Group Prof. Dr. Paul Saftig

A Group Leader: Prof. Dr. rer. nat. Paul Saftig

B Lab Members: Group leaders

PD Dr. rer. nat. Markus Damme

PostDoc

Dr. rer. nat. Florian Bleibaum

Dr. rer. nat. Alessandro Di Spiezio

Dr. rer. nat. David Massa Lopez until

Dr. rer. nat. Andre Marques until

Dr. rer. nat. Sandra Kissing until

Doctoral students

Maria Diez Tellez

Lisa Gallwitz

Saskia Heybrock

Mara Riechmann

Sönke Rudnik

Cedric Cappel

Florenica Cabrera

Adriana Gonzalez

Lisa Seipold

Ann-Christin Gradtke

Torben Mentrup

Wolfram Grieb

Niklas Thiessen

Technician

Marlies Rusch

Sebastian Held

Nadja Peter

Maike Langer until 3/19

Meryem Senkara until 6/18

Technician trainee

Sophie Reiher

Annika Detje until 8/19

Katarina Wieben until 6/18

Page 38: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

34

Secretary

Rita Latosch

C Research Report

C.1 Lysosome biology: Insight into how lysosomes mediate macromolecule degradation,

transport of metabolites and communication with the cytosol

Lysosomes are surrounded by a single membrane with a unique composition of different and

usually highly glycosylated membrane proteins. They also display various functions including

the regulation of acidification, protection against potent lysosomal hydrolases, transport of

specific hydrolases, transport of metabolites and ions as well as controlling membrane

fusion processes. Based on sub-proteomic isolation procedures the number of lysosomal

membrane proteins is estimated to reach up to 250 different proteins. Some of these proteins

are very stable in the lysosomal compartment whereas others tend to be degraded rather quickly.

The identification of new lysosomal membrane proteins by subproteomic approaches, in which

new members of the lysosomal membrane are being investigated is another focus in the lab.

The functional characterization of these new members of the lysosomal membrane is performed

using biochemical and mouse genetic approaches.

Figure 1: The lysosomal membrane protein LIMP-2 involved in lysosomal cholesterol efflux (Heybrock et al.

(2019) Nat. Comm.). Depicted is the intraluminal structure of LIMP-2 with the intramolecular hydrophobic tunnel

able to transport lipids.

C.2 Lysosome storage disorders: Towards new therapies

The pivotal role of lysosomes in cellular processes is increasingly appreciated. An

understanding of the balanced interplay between the activity of acidic hydrolases, lysosomal

membrane proteins and cytosolic proteins is required. Lysosomal storage diseases (LSDs) are

characterized by disturbances in this network and by intralysosomal accumulation of substrates,

often only in certain cell types. Even though our knowledge of these diseases has increased and

therapies have been established, many aspects of the molecular pathology of LSDs remain

obscure. This review aims to discuss how lysosomal storage affects functions linked to

lysosomes, such as membrane repair, autophagy, exocytosis, lipid homeostasis, signalling

cascades and cell viability. The possible roles of lysosomal positioning, lysosome contact sites

and lysophagy in theprogression of LSDs remain to be explored. Therapies must aim to correct

lysosomal storage not only morphologically but revert ist (patho)biochemical consequences.

As different LSDs have different molecular causes, this requires custom-tailoring of therapies.

Page 39: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

35

We will discuss the major advantages and drawbacks of current and possible future therapies

for LSDs. Study of the pathological molecular mechanisms underlying these “experiments of

nature" often yields information that is relevant for other conditions found in the general

population. Therefore, more common diseases may profit from a correction of impaired

lysosomal functions.

Figure 2: Lysosomes in the center of health and disease. The lysosomal compartment provides the tools for the

enzymatic degradation of extracellular molecules. Many phagocytosed pathogens or intracellular molecules during

autophagy end up in lysosomal degradation. (Marques and Saftig, J. Cell Biol. 2019)

C.3 Horizon Impact Award by the European Union

The European Commission has announced the winners of the first Horizon Impact Award, a

prize dedicated to EU-funded projects that have created societal impact across Europe and

beyond. The winning project have come up with a new drug for a rare disease. Jean-Eric Paquet,

Director-General for Research and Innovation of the Commission, announced the winners at

the European Research and Innovation Days in Brussels. An independent jury selected the

winning projects. MANNO-CURE (Kiel Germany) has produced the first long-term drug

therapy to treat a rare disease called Alpha-Mannosidosis.

Alpha-Mannosidosis is a rare inborn disorder caused by the lack of the lysosomal enzyme

alpha-Mannosidase, resulting in mental retardation, skeletal changes, hearing loss and recurrent

infections. Patients are often born apparently normal, and their conditions worsen

progressively, without any possibility to prevent this evolution. Already within the 5th EU

framework the collaboration EURAMAN (2001-2005) described the pathophysiology of the

disease and succesfully established an enzyme replacement therapy (ERT) for a mouse model

of Alpha-Mannosidosis. A correction of storage in many tissues including brain was found after

administration of lysosomal acid -Mannosidase (LAMAN). Within the subsequent 6th EU

framework the collaboration HUE-MAN (2006-2009) successfully established the large scale

Page 40: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

36

production of recombinant human LAMAN as a therapeutic agent for ERT in alpha-

Mannosidosis. The most effective dose was determined in preclinical trials. An immune-

tolerant mouse model that allowed chronic ERT treatment was developed and the natural

history study of alpha-Mannosidosis patients was completed. The promising results of these

two previous networks were the basis for the ALPHA-MAN project (2010-2015) within the 7th

framework program. The ALPHA-MAN network was successful to transfer and expand the

information and knowledge gained from the previous projects, to enable to perform “First in

Man” clinical trials (phase 1-3) in alpha-Mannosidosis patients, using the medicinal enzyme

product rhLAMAN as a therapeutic agent. The final goal of ALPHA-MAN to make a future

treatment for ALL alpha-Mannosidosis patients available and thereby improving their life

expectancy and quality of life became succesful. Due to the positive results of the clinical trials

within ALPHA-MAN Chiesi-Pharmaceutical took over and since April 2018 Velmanase alpha

(LAMZEDE) has been approved by the EC as the first long-term treatment therapy drug of

patients suffering from alpha-mannosidosis.

C.4 Proteolysis in or at membranes

Proteolytical processing of membrane-bound molecules is emerging as a fundamental

mechanism for controlling the strength and timing of cell-to-cell communication. Proteins

belonging to the ‘A Disintegrin And Metalloproteinase’ (ADAM) family are membrane-

anchored proteases that are able to cleave the extracellular domains of several membrane-bound

proteins in a process known as ‘ectodomain shedding’. Substrates for ADAMs include growth

factors, cytokines, chemokines and adhesion molecules. Therfore many ADAM proteins play

important roles in cell-cell adhesion, extracellular and intracellular signaling, cell

differentiation and cell proliferation. It has been shown that ADAMs are widely expressed and

are of required during developmental processes, by regulating cell-cell and cell-matrix

interactions and by modulating differentiation, migration or receptor-ligand-activated

signaling. In most cases, ectodomain shedding leads to the modulation of signaling activity on

host and neighbouring cells either through downregulation of cell surface receptors or increased

liberation of soluble ligands such as tumour necrosis factor-alpha (TNF-alpha) or epidermal

growth factor receptor (EGFR)-ligands. Dysregulation of a properly regulated shedding activity

is a critical factor in the development of complex pathologies such as cancer, cardiovascular

disease, inflammation and neurodegeneration.

Figure 2: ADAM proteases and their substrates are tightly embedded in large multi-protein and tetraspanin-

containing complexes (Seipold and Saftig, Front. Neurocsi. 2016)

Page 41: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

37

D Publications in 2018/2019

Publications 2018 Impact Factor

Gonzalez, AC, Jagdmann, S., Schweizer, M., Bernreuther, C.,Reinheckel, T.,Saftig,

P, Damme, M. (2018) Unconventional trafficking of mammalian phospholipase D3 to

lysosomes. Cell Rep., 22(4):1040-1053.

7.8

Seipold, L., Altmeppen,H., Koudelka, T., Tholey,A.,Kasparek,P., Sedlacek,R.,

Schweizer, M., Bär,J., Mikhaylova,M., Glatzel, M.,Saftig, P. (2018) In Vivo

Regulation of the A Disintegrin And Metalloproteinase 10 (ADAM10) by the

Tetraspanin 15, Cell Mol. Life Sci., 75(17):3251-3267.

5.7

Linsenmeier,L., Mohammadi, B., Wetzel,S., Puig,B.,Jackson,W.S., Hartmann,A.,

Uchiyama,K., Sakaguchi,S., Endres, K., Tatzelt, J.,Saftig,P., Glatzel, M.,Altmeppen,

H.C. (2018) Structural and mechanistic aspects influencing the ADAM10-mediated

shedding of the prion protein. Mol. Neurodegen., 13(1):18

6.7

Pohl, S., Angermann, A., Jeschke, A., Hendickx, Yorgan, T.A., Makrypidi-Fraune,

G., Steigert, A. Kuehn, S.C., Rolvien, T., Schweizer, M., Koehne, T., Neven, M.,

Winter, O., Voltolini Velho, R., Albers, J., Streichert, T., Pestka, J.M., Baldauf, C.,

Breyer, S., Stuecker, R., Muschol, N., Cox, T.M.,Saftig, P., Paganini, C., Rossi, A.,

Amling, M., Braulke, T., Schinke, T. (2018) The lysosomal protein arylsulfatase B is

a key enzyme involved in skeletal turnover. J. Bone Miner. Res., 33(12):2186-2201

6.2

Gonzalez AC, Stroobants S, Reisdorf P, Gavin AL, Nemazee D, Schwudke D,

D'Hooge R, Saftig P, Damme M. (2018) PLD3 and spinocerebellar ataxia. Brain.

141:e78

11.8

Haas A, Hensel M, Lührmann A, Rudel T, Saftig P, Schaible UE Intracellular

compartments of pathogens: Highways to hell or stairways to heaven? (2018) Int J

Med Microbiol, 308(1):1-2

3.4

Kissing S, Saftig P, Haas A. Vacuolar ATPase in phago(lyso)some biology. (2018)

Int J Med Microbiol, 308(1):58-67 3.4

Publications 2019 Impact Factor

Marques, A.R.A. & Saftig, P. (2019) Lysosomal storage disorders: challenges,

concepts and avenues for therapy - beyond rare diseases. J. Cell Sci., 132(2) pii:

jcs221739

4.4

Niemeyer, J., Mentrup, T., Heidasch, R., Mueller, S., Biswas, U., Meyer,

R., Papadopoulou, A., Dederer, V., Haug-Kroeper, M., Adamski,V., Lüllmann-

Rauch, R., Bergmann, M., Mayerhofer, A., Saftig, P., Wennemuth, G., Jessberger,

R., Fluhrer, R., Lichtenthaler,S., Lemberg, M., Schröder, B. (2019) The

intramembrane protease SPPL2c promotes male germ cell development by cleaving

phospholamban. EMBO REP., 20(3) pii: e46449

9.7

Reinicke, A.T., Laban, K., Sachs, M., Kraus, V., Walden, M., Damme, M., Sachs,

W., Reichelt, J., Schweizer, M., Janiesch, P.C., Duncan, K.E., Saftig, P., Rinschen,

M.M., Morellini, F., Meyer-Schwesinger, C. (2019) Ubiquitin C-Terminal Hydrolase

L1 (UCH-L1) loss causes neurodegeneration by altering protein turnover in the first

postnatal weeks. Proc. Natl. Acad. Sci, USA, 116(16):7963-7972

9.58

Vezzoli, E., Caron, I., Talpo, F., Besusso, D., Conforti, P., Battaglia, E., Sogne, E.,

Falqui, A.,Petricca, L., Verani, M., Martufi, P., Caricasole, A., Bresciani,

A.,Cecchetti, O., Rivetti di Val Cervo, P., Sancini, G., Riess, O., Nguyen, H.,

Seipold, L., Saftig, P., Biella, G., Cattaneo, E., Zuccato, C. (2019) Inhibiting

pathologically active ADAM10 rescues synaptic and cognitive decline

in Huntington’s disease. J. Clin. Invest., 129(6):2390-2403

12.25

Liebsch, F., Kulic, L., Teunissen, C., Shobo, A., Ulku, I., Engelschalt, V., Hancock,

M.A., van der Flier, W.M., Kunach, P., Rosa-Neto, P.,Scheltens, P., Poirier, J., Saftig,

P., Bateman, R.J., Breitner, J., Hock, C., Multhaup, G., PREVENT-AD Research

Group (2019) Aβ34 is a BACE1-derived degradation intermediate associated with

amyloid clearance and Alzheimer’s disease progression Nat. Comm., 10(1):2240

12.35

Page 42: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

38

Pasquire, A., , Vivot, K., Erbs, E., Spiegelhalter, C., Zhanh, Z., Aubert, V., Senkara,

M., Maillard, E., Pinget, M., Kerr-Conte, J., Pattou, F., Marciniak, G., Ganzhorn, A.,

Muzet, N., Rooney, E., Ronchi, P., Schieber, N.L., Schwab, Y., Saftig, P.,

Goginashvili, A., Ricci, R. (2019) Lysosomal degradation of newly formed insulin

granules contributes to cell failure in type 2 diabetes Nat. Comm., 10(1):3312

12.35

Marques, A.R.A., Di Spiezio, A., Thießen,T., Schmidt, L., Grötzinger, J., Lüllmann-

Rauch, R., Damme, M., Storck, S.E., Pietrzik, C.U., Fogh, J., Bär,J., Mikhaylova, M.,

Glatzel, M., Bassal, M., Bartsch, U., Saftig, P. (2019) Enzyme replacement therapy

with recombinant pro-CTSD (cathepsin D) corrects defective proteolysis and

autophagy in neuronal ceroid lipofuscinosis. Autophagy, 16:1-15

11.10

Wichert, R., Scharfenberg, F., Colmorgen, C., Koudelka, T., Schwarz, J., Wetzel,

S., Potempa, B., Potempa, J., Bartsch, J.W., Sagi, I., Tholey, A., Saftig,P., Rose-John,

S., Becker-Pauly, C. (2019) Meprin β induces activities of A Disintegrin and

Metalloproteinases 9, 10 and 17 by specific prodomain cleavage. FASEB J.,

33(11):11925-11940

5.94

Li, X., Qin, L., Li, Y., Yu, H., Zhang, Z., Tao, C., Liu, Y., Xue, Y., Zhang, X., Xu, Z.,

Wang, Y., Lou, H., Tan, Z., Saftig, P., Chen, Z., Xu, T., Bi, G., Duan, S., Gao, Z.

(2019) Presynaptic endosomal cathepsin D regulates the biogenesis of GABAeric

synaptic vesicles. Cell Rep. 28(4):1015-1028.

7.81

Heybrock, S., Kanerva, K., Meng, Y., Ing, C., Liang, A., Xiong, Z.J., Wenig, X., Kim,

Y.A., Collins, R.A., Trimble, W., Pomès, R., Privé, G.G., Annaert, W., Schwake, M.,

Heeren, J., Lüllmann-Rauch, R., Grinstein, S.*, Ikonen, E.*, Saftig, P.*, Neculai, D.4*

(2019) Lysosomal Integral Membrane Protein-2 (LIMP-2/SCARB2) is involved in

lysosomal cholesterol export. Nat. Comm., 10(1):3521

12.35

Massa Lopez, D., Thelen, M., Stahl, F., Thiel, C., Linhorst, A., Sylvester, M.,

Hermanns-Borgmeyer, I., ´Lüllmann-Rauch, R., Eskild, W., Saftig, P., Damme, M.

(2019) The lysosomal transporter MFSD1 is essential for liver homeostasis and

critically depends on its accessory subunit GLMP. ELife, 8. pii: e50025

7.55

Notomi, S., Ishihara, K., Efstathiou,N.E., Jong-Jer, L., Hisatomi, T., Tachibana, T.,

Konstantinou, E., Ueta, T., Murakami, Y., Maidana, D.E., Ikeda,Y., Kume, S.,

Terasaki, H., Sonoda, S., Blanz, J., Young, L., Sakamoto, T., Sonoda,K.H., Saftig, P.,

Ishibashi, T., Miller, J.W., Kroemer, G., Vavvas, D.G. (2019) Genetic LAMP2

deficiency accelerates the age-associated formation of basal laminar deposits in the

retina. Proc. Natl. Acad. Sci., 116:23724-23734

9.58

Impact factors 2018: 45.5 Impact factors 2019: 116.9

Total impact factors 2018/2019: 162.38

E Grants

E.1 Analysis of the postnatal and tissue specific role of the protease ADAM10

Sonderforschungsbereich 877, Teilprojekt A3

E.2 Microscopy core facility: Analysis of protease structure, cell biology and function

Sonderforschungsbereich 877, Teilprojekt Z3

E.3 Role of ADAM10-mediated shedding of the prion protein in prion disease in vivo

Sonderforschungsbereich 877, Teilprojekt A12

E.4 Lysosomal Membrane Proteins and their Roles in Phagosome Maturation. SPP1580 (DFG)

E.5 EUROCLAST— Exploring Functional and Developmental Osteoclast Heterogeneity in Health and

Disease; Marie Curie Actions— Initial Training Networks (ITN)

E.6 NCL2TREAT "Entwicklung einer therapeutischen Korrektur des autophagozytotischen Flux in einem

NCL Model durch preklinische Enzymersatztherapie mittels rekombinanten cathepsin-D"

E.7 ACE BIOSCIENCES "Method development to demonstrate CNS effect, Characterization of animal

models, Demonstration of Proof of Principle"

E.8 Forschergruppe "Mechanismen der Lysosomalen Homöostase, TP2: Untersuchung der Funktion der

lysosomalen Phospatidylinositol 4,5-bisphosphat 4 Phosphatasen TMEM55A und TMEM55B"

E.9 ACE BIOSCIENCES "Method development to demonstrate CNS effect, Characterization of animal

models, Demonstration of Proof of Principle" Part II

E.10 Analyse von Proteasen und Proteolyse durch Mikroskopie

Sonderforschungsbereich 877, Teilprojekt Z3

Page 43: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

39

E.11 Development of a therapy for human Alpha-Mannosidosis - Horizon Impact Award 2019 - H2020

Page 44: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

40

Page 45: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

41

7. Research Group PD Dr. Markus Damme

A Group Leader: PD Dr. rer. nat. Markus Damme

B Lab Members: PostDocs

Dr. rer. nat. David Massa Lopez

Doctoral Students

Adriana Gonzalez

Sönke Rudnik

Mara Riechmann

Medical/Master Students

Cedric Cappel

Technicians

Sebastian Held

C Research Report

C.1 Dysfunction of lysosomal proteins in neurodegenerative diseases

Advanced genomic techniques like genome wide association (GWAS) analyses and whole

exome sequencing have revealed a number of causative genes and risk factors for common

neurodegenerative diseases like Alzheimer`s disease, frontotemporal lobar dementia (FTLD)

and hereditary neuropathies. Interestingly, a number of these genes are coding for lysosomal

proteins. We are investigating the contribution of several genes coding for lysosomal proteins

in the context of such diseases in knockout and transgenic mouse models. In the focus of our

studies is the type II transmembrane proteins TMEM106B.

C.2 Characterization of new lysosomal membrane proteins of unknown function

Lysosomes are membrane-bound organelles in eukaryotic cells in which (mostly soluble) acidic

hydrolases mediate the catabolic degradation of macromolecules like proteins, lipids or

oligosaccharides to lower molecular weight metabolites like amino acids, monosaccharides or

fatty acids. These metabolites become eventually exported from the lysosomal lumen to the

cytosol by specific membranous exporter proteins, where they can be reused for a new round

of synthesis. The lysosomal membrane is furthermore protected by highly glycosylated integral

membrane proteins from self-digestion by the formation of a glycocalyx.

Page 46: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

42

Although a couple of lysosomal exporters like Sialin, Cystinosin or the Cobalamin exporter

where identified and thoroughly characterized in the last years (mostly due to diseases

characterized by a deficiency of such exporter proteins and resulting lysosomal storage of the

cargo in the lysosomal lumen), a great majority of such proteins was described so far only by

means of biochemistry, without identifying the corresponding genes.

Several recent proteomics studies have addressed the identification of new lysosomal

membrane proteins including such putative polytopic exporter proteins and integral membrane

proteins with unlikely transporter function with only one or two transmembrane domains. Goal

of our research is to elucidate the function of such lysosomal membrane proteins of so far

unknown function, particularly those whose dysfunction leads to devastating diseases like

lysosomal storage diseases or neurodegeneration. We are currently working on several new

lysosomal membrane proteins including the putative transporters Mfsd1, the single

transmembrane domain containing proteins Ncu-g1/Glmp, Lamp3 and Tmem106B and the

membrane-bound putative Phosphatidyl-Inositol-Phosphatases TMEM55A and TMEM55B.

Mouse models with loss-of-function are analyzed in order to identify putative substrates or to

reveal impaired pathological pathways. We are seeking for interaction partners to elucidate the

function of the non-transporter proteins.

Figure 1. The lysosomal membrane contains of different classes of integral membrane proteins including the

structural proteins Lamp1, Lamp2, enzymes (Sppl2a, Hgsnat), the V-ATPase composed of several subunits,

proteins mediating vesicular fusion, proteins of unknown function or function of none of the above mentioned

categories and finally a large group of lysosomal transporters and ion channels, including Sialin, Cystinosin,

Lmbrd1, Npc1, Clc7 and Dirc2. Prototypical members of each group are depicted. The function of a number of

lysosomal membrane proteins is unknown, including Tmem106b, Mfsd1 and NCU-G1/ Glmp.

C.3 (Patho-)physiologic function of the novel lysosomal 5’ exonuclease Phospholipase

D3 (PLD3)

PLD3 is a recently described lysosomal nuclease that specifically cleaves single stranded short

nucleic acids in lysosomes. We have shown that PLD3 is synthesized as a type II

Structure

- Lamp1

- Lamp2

Transporter/

Channels

- Sialin

- Cystinosin

- Lmbrd1

- Npc1

- Dirc2

- CLCN-7

- TAPL

- SLC17A9

V-ATPase

Enzymes

- Hgsnat

- Sppl2a

Other/ Unknown

- CD63

- Ncu-G1

- Limp2

- Tmem106b

Y

LumenH+

H+

H+

H+

Glycocalyx

Substrate

turnover

Vesicular trafficking/

fusion

- Vti1b

- Vamp8

Page 47: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

43

transmembrane protein that is proteolytically cleaved yielding a stable soluble luminal domain.

This luminal domain is active as a nuclease. We are investigating the physiological function of

PLD3 in a knockout mouse model.

D Publications 2018/2019

Publications 2018 Impact Factor

Gonzalez AC, Stroobants S, Reisdorf P, Gavin AL, Nemazee D, Schwudke D, D'Hooge

R, Saftig P, Damme M. (2018) PLD3 and spinocerebellar ataxia.. Brain. 141(11):e78. 11.814

Khateb S, Kowalewski B, Bedoni N, Damme M, Pollack N, Saada A, Obolensky A, Ben-

Yosef T, Gross M, Dierks T, Banin E, Rivolta C, Sharon D. (2018). A homozygous

founder missense variant in arylsulfatase G abolishes its enzymatic activity causing

atypical Usher syndrome in humans. Genet Med. (9):1004-1012.

8.683

Gavin AL, Huang D, Huber C, Mårtensson A, Tardif V, Skog PD, Blane TR, Thinnes

TC, Osborn K, Chong HS, Kargaran F, Kimm P, Zeitjian A, Sielski RL, Briggs M, Schulz

SR, Zarpellon A, Cravatt B, Pang ES, Teijaro J, de la Torre JC, O'Keeffe M, Hochrein H,

Damme M, Teyton L, Lawson BR, Nemazee D. (2018). PLD3 and PLD4 are single-

stranded acid exonucleases that regulate endosomal nucleic-acid sensing. Nat Immunol.

(9):942-953.

23.530

De Pace R, Skirzewski M, Damme M, Mattera R, Mercurio J, Foster AM, Cuitino L,

Jarnik M, Hoffmann V, Morris HD, Han TU, Mancini GMS, Buonanno A, Bonifacino

JS. (2018). Altered distribution of ATG9A and accumulation of axonal aggregates in

neurons from a mouse model of AP-4 deficiency syndrome. PLoS Genet. 14(4):e1007363

5.224

Bartsch K, Damme M, Regen T, Becker L, Garrett L, Hölter SM, Knittler K, Borowski

C, Waisman A, Glatzel M, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Rabe B.

(2018). RNase H2 Loss in Murine Astrocytes Results in Cellular Defects Reminiscent of

Nucleic Acid-Mediated Autoinflammation. Front Immunol. 9:587.

4.716

Gonzalez AC, Schweizer M, Jagdmann S, Bernreuther C, Reinheckel T, Saftig P, Damme

M. (2018). Unconventional Trafficking of Mammalian Phospholipase D3 to Lysosomes. Cell Rep. 22(4):1040-1053.

7.815

Publications 2019 Impact Factor

Massa Lopez, D., Thelen, M., Stahl, F., Thiel, C., Linhorst, A., Sylvester, M., Hermanns-

Borgmeyer, I., Lüllmann-Rauch, R., Eskild, W., Saftig, P., Damme, M. (2019). The

lysosomal transporter MFSD1 is essential for liver homeostasis and critically depends on

its accessory subunit GLMP. Elife. 8. pii: e50025.

7.551

Marques ARA, Di Spiezio A, Thießen N, Schmidt L, Grötzinger J, Lüllmann-Rauch R,

Damme M, Storck SE, Pietrzik CU, Fogh J, Bär J, Mikhaylova M, Glatzel M, Bassal M,

Bartsch U, Saftig P. (2019). Enzyme replacement therapy with recombinant pro-CTSD

(cathepsin D) corrects defective proteolysis and autophagy in neuronal ceroid

lipofuscinosis. Autophagy. 16:1-15

11.059

Moreau D, Vacca F, Vossio S, Scott C, Colaco A, Paz Montoya J, Ferguson C, Damme

M, Moniatte M, Parton RG, Platt FM, Gruenberg J. (2019). Drug-induced increase in

lysobisphosphatidic acid reduces the cholesterol overload in Niemann-Pick type C cells

and mice. EMBO Rep. 20(7):e47055.

8.383

Khundadze M, Ribaudo F, Hussain A, Rosentreter J, Nietzsche S, Thelen M, Winter D,

Hoffmann B, Afzal MA, Hermann T, de Heus C, Piskor EM, Kosan C, Franzka P, von

Kleist L, Stauber T, Klumperman J, Damme M, Proikas-Cezanne T, Hübner CA. (2019).

A mouse model for SPG48 reveals a block of autophagic flux upon disruption of adaptor

protein complex five. Neurobiol Dis.127:419-431.

5.160

Reinicke AT, Laban K, Sachs M, Kraus V, Walden M, Damme M, Sachs W, Reichelt J,

Schweizer M, Janiesch PC, Duncan KE, Saftig P, Rinschen MM, Morellini F, Meyer-

Schwesinger C. (2019). Ubiquitin C-terminal hydrolase L1 (UCH-L1) loss causes

neurodegeneration by altering protein turnover in the first postnatal weeks. Proc Natl

Acad Sci U S A. 116(16):7963-7972.

9.504

Page 48: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

44

Impact factors 2018: 61.782

Impact factors 2019: 41.657 Total impact factors 2018/2019: 103.439

E Grants

E.1 Industry collaboration project: 510.749 €

E.2 Transcriptome Analysis of TMEM106B knockout mice (Intramural Funding of the Medical

Faculty of the CAU (with PD Dr. Robert Häsler): 70.000 €

E.3 Functional characterization of the putative lysosomal transporter protein “Major facilitator

superfamily domain containing 1” (Mfsd1) and its role in sinusoidal obstruction syndrome

(DFG-Sachbeihilfe Fördersumme (2017 -2020): 244.450

E.4 Deciphering the physiological roles of the lysosomal phosphatidylinositol-4,5-bisphosphate 4-

phosphatases TMEM55A and TMEM55B; DFG-Forschergruppe “Mechanismen der

Lysosomalen Homöostase” – Teilprojektleiter (with Prof. Paul Saftig): 459.950 €

Page 49: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

45

8. Research Group Prof. Dr. Christoph Becker-Pauly

A Group Leader: Prof. Dr. rer nat. Christoph Becker-Pauly

B Lab Members: Post-Doc:

Dr. rer. nat. Franka Scharfenberg

Dr. rer. nat. Florian Peters

Dr. rer. nat. Sascha Rahn

Doctoral Students:

Fred Armbrust

Cynthia Colmorgen

Kriti Pathak

Ludwig Werny

Martin Sammel (med)

Friederike Schrell (med)

Anne Winkelmann (med)

Luisa Wessolowski (med)

Technicians:

Ingrid Berg

Inez Götting

Britta Hansen

Lennard Arp (Trainee)

Annika Malien (Trainee)

Bachelor/Master students:

Kira Bickenbach

Marion Mengel

Nicolas Steen

Max Bettendorff

Page 50: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

46

C Research Report

C1. Regulation of the alternative β-secretase meprin β by ADAM-mediated shedding

Neurotoxic amyloid-β (Aβ) plaques are one hallmark of Alzheimer disease (AD). Aβ deposits

in the brain are composed of peptides derived from APP and consist of up to 42 amino acids.

Several publications support different molecular mechanisms for Aβ mediated synaptic

dysfunction and neuronal cell death, such as membrane disruption, ion dysregulation or

oxidative stress induction. However, the Aβ biology is rather complex, mainly due to its great

hydrophobic interacting potential. Thus, the entire and exact role of Aβ remains elusive. Aβ

peptides derive from APP by sequential cleavage at the β- and γ-secretase cleavage sites.

Intramembranous γ-cleavage is accomplished by the aspartic peptidases presenilin 1 and 2

(PS1/2) within the γ-secretase complex at position 40 or 42 (numbering according to Aβ

sequence). Aβ1-40 is the major species, whereas Aβ1-42 levels are low in healthy brains,

however, strongly increase during progression of AD. Of note, conditional PS1/2 double knock-

out mice exhibit significantly reduced Aβ levels. Further, more than one hundred PS1 related

mutations were identified that lead to increased Aβ levels suggesting PS1 as susceptibility gene

for AD. However, β-secretase cleavage is rate limiting for the Aβ formation. The first identified

β-secretase is the aspartic protease β-site cleaving enzyme 1 (BACE-1). It is predominantly

expressed in acidic compartments and exhibits a low pH optimum. Thus, BACE-1 dependent

APP cleavage occurs in endosomal/lysosomal compartments. The major cleavage event by

BACE-1 at the β-site of APP is at position 1 resulting in the dominant Aβ species Aβ1-40/42.

Since the Aβ formation in BACE-1 knock-out mice is strongly reduced, BACE-1 is thought to

be the major β-secretase. Thus, BACE-1 is one of the most promising therapeutic targets for

AD treatment. However, all clinical trials using specific BACE-1 inhibitors have failed so far

and have not shown any cognitive benefits for the patients

(https://www.alzforum.org/news/conference-coverage/bump-road-or-disaster-bace-inhibitors-

worsen-cognition; 02.11.2018). Therefore, the investigation of alternative β-secretases as

potential drug targets is of great interest.

Besides the BACE-1-generated Aβ1-x species N-terminal truncated Aβ forms came into focus

of research (Figure 1). Already many years ago, Konrad Beyreuther and Colin Masters

described N-terminal truncated Aβ peptides in the core of amyloid plaques of AD patients.

Other groups have shown an increase of Aβ peptides starting at position 2 (Aβ2-x) in the brains

of AD patients compared to other dementias or non-demented subjects. A number of N-

terminally truncated Aβ variants starting at different other positions have been reported in the

cerebrospinal fluid (CSF), blood and brain tissue of AD patients. Since BACE-1 is incapable

to generate such peptides the hunt for these enzymes was evident. For instance, cathepsin B, S

and L as lysosomal proteases are discussed as alternative β-secretases generating various Aβ

species. Inhibitor studies in cells and mice indicate a direct involvement of these proteinases in

Aβ generation. Of note, cathepsin B is thought to be involved in Aβ3-x formation, which is the

progenitor of highly neurotoxic pyroglutamate-modified Aβ3-x. However, cathepsin D is

involved in the clearance of Aβ. The detailed role of cathepsins in this context is not revealed

so far, however, their low cleavage specificity on APP suggest an Aβ degrading role. Other

candidates generating N-terminally truncated Aβ peptides are the Aminopeptidases A and N

(APA/APN). APA generates Aβ2-x from Aβ1-x, whereas APN is thought to convert Aβ2-x to

Aβ3-x. A promising alternative β-secretase that directly cleaves at p2 within full length APP is

the metalloprotease meprin β. We and others could show that mRNA and protein levels of

meprin β are significantly increased in AD brain, which is in line with increased Aβ2-42. Of

note, Aβ2-40 peptides not only exhibit a greater aggregation potential than Aβ1-40 but

additionally induce Aβ1-40 aggregation. Mass spectrometry-based degradomics identified APP

as a substrate for meprin β at three different sites in the N-terminal region between

Page 51: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

47

Ser124/Asp125, Glu380/Thr381, and Gly383/Asp384 (Figure 1). After incubation with all APP

isoforms and meprin β, two fragments of 20 and 11 kDa were identified either in vitro or in cell

culture-based assays. Interestingly, these fragments derived from APP processing were found

in human and wild-type mice brain lysates, but not in the brain of Mep1b-/- mice, proving APP

as a physiological target of meprin β. The major cleavage site of meprin β in APP695 is between

Asp597 and Ala598 resulting in the formation of Aβ2-x, and to a minor extend between Met596

and Asp597 at the BACE-1 site. Meprin β knock-out mice brains show increased sAPPα levels

which could indicate that the absence of meprin β leads to more available substrate to α-

secretase. It has been shown that meprin β and APP co-localize at the cell surface and in the

secretory pathway leading to APP processing by meprin β in these cellular compartments.

Therefore, meprin β may compete at the cell surface with ADAM10, the main α-secretase in

the brain. A recent publication indicates that meprin β may also act as dipeptidyl-peptidase

being able to convert Aβ1-x to Aβ3-x, which is the progenitor of highly neurotoxic

pyroglutamate-modified Aβ3-x. However, this observation is based on in vitro cleavage using

truncated Aβ-peptides. Hence, further cell-based assays are necessary to validate these findings.

Figure 1: APP processing by BACE-1 and meprin β in the canonical and non-canonical pathway.

BACE-1 and meprin β are shown to be involved in the generation of Aβ peptides in the canonical and non-

canonical pathway. The so far well described APP processing pathways by α-, β- and δ-secretases are described

in green, orange and blue. Meprin β is involved in the generation of two APP N-terminal fragments of 20 and 11

kDa as well as the cleavage of APP at the β-secretase cleavage site, providing a substrate for γ-secretase releasing

Aβ peptides (red).

We identified meprin β as an alternative β-secretase predominantly generating Aβ2-x peptides.

This cleavage event requires membrane-bound meprin β and is prevented for the soluble shed

protease. Thus, the meprin β sheddases ADAM10 and 17 may exhibit a dual function in

Page 52: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

48

preventing from amyloidation in AD. On the one hand, ADAM10 acts as α-secretase cleaving

APP within the Aβ sequence and, thus, counteracts against Aβ formation. On the other hand,

ADAM10/17 prevent from amyloidation by shedding the β-secretase meprin β from the cell

surface (Figure 2). However, whether ADAM proteases prefer APP over meprin β as shedding

substrate or vice versa is completely unknown. Of note, meprin β itself was identified as an

inducer of ADAM10 activity. This finding complicates the protease network around

ADAM10/17-mediated prevention of Aβ generation. Thus, further research on the exact

mechanism of the dual protective role of ADAM 10 and 17 is required.

Figure 2: Extracellular regulation of meprin β activity with respect to β-site cleavage of APP.

(A) Cartoon representation of a membrane-bound meprin β model (blue) based on the crystal structure of the

ectodomain (PDB: 4GWN) in complex with part of APP (grey, Aβ peptide in red). Structures of additional

N-terminal domains of APP695 are also shown as cartoons: E1 (PDB: 3KTM), E2 (PDB: 3NYL). Sequence

stretches of unknown structure and the AICD domain are illustrated as dashed lines. The close up views in the

right panel highlight determined meprin β cleavage sites, while P1 and P1´ residues are depicted as sticks. (B)

Model of a membrane-bound ADAM10 based on the ectodomain (yellow, PDB: 6BE6) and pro-meprin β (blue).

The pro-peptide of meprin β is shown in orange. The red arrow indicates the shedding site within pro-meprin β.

E1/2: extracellular domains 1/2; Acd: acidic domain; JMR: juxtamembrane region ; AICD: APP intracellular

domain

C2. Tethering soluble meprin α in an enzyme complex to the cell surface affects IBD

associated genes

Biological activity of proteases is mainly characterized by their substrate specificity, tissue

distribution and cellular localization. The human metalloproteases meprin α and meprin β share

41% sequence identity and exhibit a similar cleavage specificity with a preference for

Page 53: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

49

negatively charged amino acids. However, shedding of meprin α by furin on the secretory

pathway makes it a secreted enzyme in comparison to the membrane bound meprin β. In this

study, we identified the human meprin α and meprin β to form covalently linked membrane

tethered heterodimers in the early ER thereby preventing furin-mediated secretion of meprin α

(Figure 3).

Figure 3: Assembly of meprin oligomers

(A) Cartoon representation of meprin β shedding by ADAM10 and ADAM17 as homo- or heterodimer. Meprin α

is shed by furin in the Golgi apparatus and secreted into the extracellular space when expressed alone. (B)

Immunofluorescence microscopy of small intestine from wt, Mep1b-/- and Mep1a-/- mice using specific meprin

antibodies. Staining revealed absence of meprin α at the cell surface in Mep1b-/- mice. Scale bar = 40 µm.

Within this newly formed enzyme complex meprin α was able to be activated on the cell surface

detected by cleavage of a novel specific fluorogenic peptide substrate. However, the known

meprin β substrates amyloid precursor protein (APP) and CD99 were not shed by membrane

tethered meprin α. On the other hand, being linked to meprin α, activation of or substrate

cleavage by meprin β on the cell surface was not altered. Interestingly, proteolytic activity of

both proteases was increased in the heteromeric complex, indicating an increased proteolytic

potential at the plasma membrane. Since meprins are susceptibility genes for inflammatory

bowel disease (IBD), and to investigate the physiological impact of the enzyme complex, we

performed transcriptome analyses of intestinal mucosa from meprin knock-out mice.

Comparison of the RNAseq data with gene analyses of IBD patients revealed that different gene

subsets were dysregulated if meprin α was expressed alone or in the enzyme complex,

demonstrating the physiological and pathophysiological relevance of the meprin heterodimer

formation.

Page 54: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

50

C3. Role of Meprin Metalloproteases in Metastasis and Tumor Microenvironment

A crucial step for tumor cell extravasation and metastasis is the migration through the

extracellular matrix, which requires proteolytic activity. Hence, proteases, particularly matrix

metalloproteases (MMPs), have been discussed as therapeutic targets and their inhibition

should diminish tumor growth and metastasis.

The metalloproteases meprin α and meprin β are highly abundant on intestinal enterocytes and

their expression was associated with different stages of colorectal cancer. Due to their ability

to cleave extracellular matrix (ECM) components, they were suggested as protumorigenic

enzymes (Figure 4). Additionally, both meprins were shown to have proinflammatory activity

by cleaving cytokines and their receptors, which correlates with chronic intestinal inflammation

and associated conditions. On the other hand, meprin β was identified as an essential enzyme

for the detachment and renewal of the intestinal mucus, important to prevent bacterial

overgrowth and infection. Considering this, it is hard to estimate whether high activity of

meprins is generally detrimental or if these enzymes have also protective functions in certain

cancer types. For instance, in colorectal cancer, patients with high meprin β expression in tumor

tissue exhibit a better survival prognosis, which is completely different to prostate cancer. This

demonstrates that the very same enzyme may have contrary effects on tumor initiation and

growth, depending on its tissue and subcellular localization. Hence, precise knowledge about

proteolytic enzymes is required to design the most efficient therapeutic options for cancer

treatment.

Figure 4: Meprin Metalloproteases in Metastasis and Tumor Microenvironment

Under physiological conditions, meprins are mainly located at apical sites of epithelial cells and expressed as

homo- or hetero-dimers. Upon mislocalization due to the loss of cell polarity and their activation by tryptic

proteases, meprins can degrade and remodel ECM components on the basolateral site and promote tumor growth

via induction of angiogenesis and inflammation. Furthermore, cleavage of adhesion molecules can induce loss of

intercellular adhesion and promote invasion and metastasis of cancer cells.

Page 55: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

51

D Publications 2018/2019

Biasin V, Wygrecka M, Bärnthaler T, Jandl K, Bálint Z, Kovacs G, Leitinger G,

Kolb-Lenz D, Kornmueller K, Peters F, Sinn K, Klepetko W, Heinemann A,

Olschewski A, Becker-Pauly C, Kwapiszewska G. (2018) Docking of meprin α to

heparan sulphate protects the endothelium from inflammatory cell extravasation.

Thrombosis and Haemostasis, 118(10):1790-1802.

4.899

Talantikite M, Lécorché P, Beau F, Damour O, Becker-Pauly C, Ho WB, Dive V,

Vadon-Le Goff S, Moali C. (2018) Inhibitors of BMP-1/tolloid-like proteinases:

efficacy, selectivity and cellular toxicity. FEBS OpenBio, 8(12):2011-2021.

1.782

Schmidt S, Schumacher N, Schwarz J, Roos S, Kenner L, Schlederer M, Sibilia M,

Linder M, Altendorf-Hofmann A, Knösel T, Gruber ES, Oberhuber G, Rehman A,

Sinha A, Arnold P, Zunke F, Becker-Pauly C, Preaudet A, Nguyen P, Huynh J,

Chand AL, Westermann J, Dempsey PJ, Garbers C, Rosenstiel P, Putoczki T, Ernst

M, Rose-John S (2018) ADAM17 is required for EGF-R induced intestinal tumors

via IL-6 trans-signaling. J Exp Med, 215(4):1205-1225

10.892

Publications 2019 Impact Factor

Karmilin K, Schmitz C, Kuske M, Körschgen H, Olf M, Meyer K, Hildebrand A,

Felten M, Fridrich S, Yiallouros I, Becker-Pauly C, Weiskirchen R, Jahnen-Dechent

W, Floehr J, Stöcker W. (2019) Mammalian plasma fetuin-B is a selective inhibitor

of ovastacin and meprin metalloproteinases. Sci Rep. 9(1):546.

4.011

Boon L, Ugarte-Berzal E, Martens E, Vandooren J, Rybakin V, Colau D, Gordon-

Alonso M, van der Bruggen P, Stöcker W, Becker-Pauly C, Witters P, Morava E,

Jaeken J, Proost P, Opdenakker, G. (2019) Propeptide glycosylation and galectin-3

binding decrease proteolytic activation of human progelatinase B/proMMP-9. FEBS

J, 286(5):930-945.

4.739

Walter S, Jumpertz T, Hüttenrauch M, Gerber H, Dimitrov M, Ogorek I, Lehmann

S, Lepka K, Berndt C, Wiltfang J, Becker-Pauly C, Beher D, Pietrzik CU, Fraering

PC, Wirths O, Weggen S. (2019) The metalloprotease ADAMTS4 generates N-

truncated Aβ4-x peptides and marks oligodendrocytes as pro-amyloidogenic in

Alzheimer’s disease. Acta Neuropathologica, 137:239–257.

18.174

Peters F, Scharfenberg F, Colmorgen C, Armbrust F, Wichert R, Arnold P, Potempa

B, Potempa J, Pietrzik CU, Häsler R, Rosenstiel P, Becker-Pauly C*. (2019)

Tethering soluble meprin α in an enzyme complex to the cell surface affects IBD

associated genes. FASEB J, 9(1):546..

5.391

Scharfenberg F, Helbig A, Sammel M, Benzel J, Schlomann U, Peters F, Wichert R,

Bettendorff M, Schmidt-Arras D, Rose-John S, Moali C, Lichtenthaler SF, Pietrzik

CU, Bartsch JW, Tholey A, and Becker-Pauly C*. (2019) Degradome of soluble

ADAM10 and ADAM17 metalloproteases. Cell Mol Life Sci. 2019 Jun 17. doi:

10.1007/s00018-019-03184-4.

7.014

Scharfenberg F, Armbrust F, Marengo L, Pietrzik CU, Becker-Pauly C*. (2019)

Regulation of the alternative β-secretase meprin β by ADAM-mediated shedding.

Cell Mol Life Sci, 76(16):3193-3206.

7.014

Wünnemann F, Ta-Shma A, Preuss C, Leclerc S, van Vliet PP, Oneglia A, Thibeault

M, Nordquist E, Lincoln J, Scharfenberg F, Becker-Pauly C, Hofmann P, Hoff K,

Audain E, Kramer HH, Makalowski W, Nir A, Gerety S, Hurles M, Comes J,

Fournier A, Osinska H, Robins J, Puceat M, MIBAVA Leducq Consortium, Elpeleg

O, Hitz MP*, Andelfinger G*. (2019) Loss of ADAMTS19 causes progressive non-

syndromic heart valve disease. Nat Genet. doi: 10.1038/s41588-019-0536-2

25.455

Wichert R, Scharfenberg F, Koudelka T, Colmorgen C, Schwarz J, Wetzel S,

Potempa B, Potempa J, Bartsch JW, Sagi I, Tholey A, Saftig P, Rose-John S, Becker-

Pauly C*. (2019) Meprin β induces activities of A Disintegrin and

Metalloproteinases 9, 10 and 17 by specific prodomain cleavage. FASEB J,

33(11):11925-11940.

5.391

Schäffler H, Li W, Helm O, Krüger S, Böger C, Peters F, Röcken C, Sebens S, Lucius

R, Becker-Pauly C, Arnold P. (2019) The cancer-associated meprin β variant G32R 4.517

Publications 2018 Impact Factor

Page 56: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

52

provides an additional activation site and promotes cancer cell invasion. J Cell Sci,

132(11).

Sammel M, Peters F, Lokau J, Scharfenberg F, Werny L, Linder S, Garbers C, Rose-

John S, Becker-Pauly C*. (2019) Differences in Shedding of the Interleukin-11

Receptor by the Proteases ADAM9, ADAM10, ADAM17, Meprin α, Meprin β and

MT1-MMP. Int J Mol Sci. 20: 15

4.183

Peters F & Becker-Pauly C*. Role of meprin metalloproteases in metastasis and

tumor microenvironment. Cancer Metastasis Rev. 2019 Sep;38(3):347-356 6.667

Escrig A, Canal C, Sanchis P, Fernández-Gayol O, Montilla A, Comes G, Molinero

A, Giralt M, Giménez-Llort L, Becker-Pauly C, Rose-John S, Hidalgo J. (2019) IL-

6 trans-signaling in the brain influences the behavioral and physio-pathological

phenotype of the Tg2576 and 3xTgAD mouse models of Alzheimer's disease. Brain

Behav Immun. S0889-1591(19)30150-3

6.306

Impact factors 2019: 98.89 Impact factors 2018: 18.67 Total impact factors 2018/2019: 117.5

E Grants E.1 2014-2018 DFG (SFB877, A9) 661.000 €

“Proteolytic network of meprin metalloproteases and ADAMs with regard to

fibrosis, neurodegeneration and inflammation”

E.2 2016-2019 DFG (BE 4086/5-1) 241.000 €

“Role of astacin-like proteinases in physiological wound healing and scarring”

E.3 2017-2020 DFG (BE 4086/2-2) 190.000 €

“Functional role of meprin beta in Alzheimer s disease”

E.4• 2018-20 Alzheimer Forschung Initiative e.V (#18007) 120.000 €

“Inhibition of meprin β to prevent generation of N-terminal truncated Aβ-peptides”

E.5 2018-2022 DFG (SFB877, A9) 490.000 €

“Regulation of meprin metalloproteases in inflammation and fibrosis”

E.6 2018-2022 DFG (SFB877, A15) 496.000 €

“Functional role of meprin beta in Alzheimer’s disease”

Page 57: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

53

9. Research Group Prof. Dr. Hilmar Lemke

A Group Leader: Prof. Dr. Hilmar Lemke

B Lab Members: ---

C Research Report

Antigen receptors – self-made but of somatically created Non-Self nature

Antigen determinants (epitopes) are recognized by the combining sites (paratopes) of B

and T cell antigen receptors, which exist in three different forms BCR, a:ß-TCR and g:d-TCR.

Their diversity is generated through four different processes:

1. combination of variable (V), diversity (D) and joining (J) gene segments gives the combinatorial diversity,

2. inaccuracies at the recombination sites result in junctional diversity,

3. association of BCR heavy and light chains as well as association of a:ß-chains for creation of a:ß-TCR and

g:d-chain for creation of g:d-TCR creates the full receptors and

4. somatic hypermutations that are introduced during antigen-induced immune responses into BCR, but not

both TCR, add additional variability.

Remarkably, the somatically generated junctional diversity is many orders of magnitude

higher than the combinatorial diversity. Sites of junctional diversity are created at the third

complementarity-determining regions of BCR (CDRH3), but not the light chain, and at both

CDR3 of a:ß- and g:d-TCR. As these somatically created CDR3s are decisive for antigen-

specificity, it follows that the entire repertoire of antigen receptors belong to the Non-Self. In

addition, with all likelihood a combinatorial repertoire that is assembled from purely

genomically encoded antigen-receptors does not exist.

Moreover, sites of junctional diversity and somatically created hypermutations in BCR

harbor clone-specific epitopes (idiotopes) of any antigen receptor. Additional idiotypic sites are

created through the conformational impact of both. Idiotopes can be recognized by BCR/TCR

not only of genetically different donors but also within the autologous immune system. While

xenogeneic and allogeneic anti-idiotypic BCR/TCR are broadly cross-reactive, only autologous

anti-idiotypes are truly specific and of functional regulatory relevance within a particular

immune system. However, although idiotypic characters are per se non-immunogenic, in

conjunction with immunogenicity- and adjuvanticity-providing antigen-induced immune

responses, they induce abating regulatory idiotypic chain reactions. The dualistic nature of

antigen receptors of seeing antigens (self and nonself alike) and being nonself at the same time

has far reaching consequences for an understanding of the regulation of adaptive immune

responses.

D Publications 2018/2019

Page 58: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

54

Publications 2018 Impact Factor

Lemke H. (2018) Immune Response Regulation by Antigen Receptors’ Clone-Specific

Non-Self Parts. Front. Immunol. 9:1471 6.429

Publications 2019 Impact Factor

---

Impact factors 2018: 6.429

Impact factors 2019: -----

Total impact factors 2018/2019: 6.429

Page 59: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

55

Appendix

Biochemisches Kolloquium 2018/2019 16. 01. 2018 Dr. Dante Neculai, College of Medicine, Zhejiang University, China

"NODs palmitoylation is required for bacterial sensing"

24.04.2018 Prof. Dr. Peter Schu, Biochemie 2, Universität Göttingen

"Synaptic Signaling and Plasticity regulated by AP-1& AP-2 dependent Protein

Sorting"

19.06.2018 Prof. Dr. Thorsten Maretzky, Carver College of Medicine,Department of Internal

Medicine, University of Iowa, USA

"iRhom2 deficiency results in an increased CD4+Th1/IFNgg mediated immune

response in early onset spontaneous murine colitis"

10.07.2018 Dr. Fedor Berditchevski, Institute of Cancer and Genomic Sciences, The University of

Birmingham

" Different faces of tetraspanin proteins in cancer"

12. 07. 2018 Prof. Dr. Gisou van der Goot, Ecole Polytechnique Federale de Lausanne

" Functions and dynamics of protein palmitoylation"

24.09.2018 Sergei Grivennikov, Fox Chase Cancer Center, Philadelphia, USA

"Microbes and cytokines in regulation of tumor elicited inflammation"

22.01.2019 Dr. Björn Schröder, The Wallenberg Laboratory for cardipvasculkar and Metabolic

Research, University of Göteborg

"How dietary modulation of the gut bacteria affects the intestinal mucus layer"

12.03.2019 Prof. Dr. Henning Walczak, University College London, UK

"Cell death and inflammation by TNFa and related factors in cancer and

autoimmunity“

18.03.2019 Prof. Dr. Jacob Rachmilewitz, Hadassah Medical Centre, Hebrew University

Jerusalem

"Macrophage-Assisted DNA Damage Response: A novel systemic mechanism that

maintains genome integrity“

09.04.2019 Prof. Dr. Aymelt Itzen,UKE Hamburg, Biochemie

"Manipulation of human proteins by bacterial pathogens"

16.07.2019 Prof. Dr. Jürgen Scheller, Institut für Biochemie und Molekularbiologie II

Heinrich-Heine Universität Düsseldorf

"Synthetic Cytokine Signaling“

Page 60: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

56

Publications 2018/2019

Original Papers and Reviews

2018 Agthe, M., Brügge, J., Garbers, Y., Wandel, M., Kespohl, B., Arnold, P., Flynn, C.M., Lokau, J., Bretscher, C.,

Waetzig, G.H., Putoczki, C.G., Grötzinger, J., Garbers, C. (2018) Mutations in Craniosynostosis Patients

Cause Defective Interleukin-11 Receptor Maturation and Drive Craniosynostosis-like Disease in Mice.

Cell Rep 25, 10-18. 8.282

Agthe, M., Garbers, Y., Grötzinger, J., Garbers, C. (2018) Two N-linked glycans differentially control

maturation and trafficking, but not activity of the interleukin-11 receptor. Cell Physiol Biochem 45, 2071-

2085. 5.104

Armacki M, Trugenberger AK, Ellwanger AK, Eiseler T, Schwerdt C, Bettac L, Langgartner D, Azoitei N,

Halbgebauer R, Groß R, Barth T, Lechel A, Walter BM, Kraus JM, Wiegreffe C, Grimm J, Scheffold A,

Schneider MR, Peuker K, Zeißig S, Britsch S, Rose-John S, Vettorazzi S, Wolf E, Tannapfel A, Steinestel

K, Reber SO, Walther P, Kestler HA, Radermacher P, Barth TF, Huber-Lang M, Kleger A, Seufferlein T

(2018) Thirty-eight-negative kinase 1 mediates trauma-induced intestinal injury and multi-organ failure. J

Clin Invest 128: 5056-5072 12.282

Barikbin R, Berkhout L, Bolik J, Schmidt-Arras D, Ernst T, Ittrich H, Adam G, Parplys A, Casar C, Krech T,

Karimi K, Sass G, Tiegs G. Early HO-1 induction has beneficial effects on inflammation and malignant

transformation in mdr2-/- mice (2018) Sci Rep Nov 2;8(1):16238 4.1

Bartsch K, Damme M, Regen T, Becker L, Garrett L, Hölter SM, Knittler K, Borowski C, Waisman A, Glatzel

M, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Rabe B. (2018). RNase H2 Loss in Murine

Astrocytes Results in Cellular Defects Reminiscent of Nucleic Acid-Mediated Autoinflammation. Front

Immunol. 9:587. 4.716

Biasin V, Wygrecka M, Bärnthaler T, Jandl K, Bálint Z, Kovacs G, Leitinger G, Kolb-Lenz D, Kornmueller K,

Peters F, Sinn K, Klepetko W, Heinemann A, Olschewski A, Becker-Pauly C, Kwapiszewska G. (2018)

Docking of meprin α to heparan sulphate protects the endothelium from inflammatory cell extravasation.

Thrombosis and Haemostasis, 118(10):1790-1802. 4.899

Cabron AS, El Azzouzi K, Boss M, Arnold P, Schwarz J, Rosas M, Dobert JP, Pavlenko E, Schumacher N,

Renne T, Taylor PR, Linder S, Rose-John S, Zunke F (2018) Structural and Functional Analyses of the

Shedding Protease ADAM17 in HoxB8-Immortalized Macrophages and Dendritic-like Cells. J Immunol

201: 3106-3118 4.718

De Pace R, Skirzewski M, Damme M, Mattera R, Mercurio J, Foster AM, Cuitino L, Jarnik M, Hoffmann V,

Morris HD, Han TU, Mancini GMS, Buonanno A, Bonifacino JS. (2018). Altered distribution of ATG9A

and accumulation of axonal aggregates in neurons from a mouse model of AP-4 deficiency syndrome.

PLoS Genet. 14(4):e1007363 5.224

Dixit A, Bottek J, Beerlage AL, Schuettpelz J, Thiebes S, Brenzel A, Squire A, Garbers G, Rose-John S,

Mittruecker HW, Engel DR (2018) Proliferation of Ly6C+ monocytes during urinary tract infection is

regulated by IL-6 trans-signaling. J Leukoc Biol 103: 13-22 4.012

Fuchslocher-Chico J, Falk-Paulsen M, Luzius A, Saggau A, Ruder B, Bolik J, Schmidt-Arras D, Linkermann A,

Becker C, Rosenstiel P, Rose-John S, Adam D, The enhanced susceptibility of ADAM-17 hypomorphic

mice to DSS-induced colitis is not ameliorated by loss of RIPK3, revealing an unexpected function of

ADAM-17 in necroptosis. (2018) Oncotarget 9(16):12941-12958

Garbers C, Heink S, Korn T, and Rose-John S (2018) Interleukin-6: Designing specific therapeutics for a

complex cytokine. Nat Rev Drug Discov 17: 395-412 57.618

Garbers C, Rose-John S (2018) Dissecting lnterleukin-6 Classic- and Trans-Signaling in Inflammation and

Cancer. Meth Mol Biol 1725: 127-140 1.290

Gavin AL, Huang D, Huber C, Mårtensson A, Tardif V, Skog PD, Blane TR, Thinnes TC, Osborn K, Chong HS,

Kargaran F, Kimm P, Zeitjian A, Sielski RL, Briggs M, Schulz SR, Zarpellon A, Cravatt B, Pang ES,

Teijaro J, de la Torre JC, O'Keeffe M, Hochrein H, Damme M, Teyton L, Lawson BR, Nemazee D.

(2018). PLD3 and PLD4 are single-stranded acid exonucleases that regulate endosomal nucleic-acid

sensing. Nat Immunol. (9):942-953. 23.530

Gonzalez AC, Schweizer M, Jagdmann S, Bernreuther C, Reinheckel T, Saftig P, Damme M. (2018).

Unconventional Trafficking of Mammalian Phospholipase D3 to Lysosomes. Cell Rep. 22(4):1040-1053.

7.815

Gonzalez AC, Stroobants S, Reisdorf P, Gavin AL, Nemazee D, Schwudke D, D'Hooge R, Saftig P, Damme M.

(2018) PLD3 and spinocerebellar ataxia.. Brain. 141(11):e78. 11.814

Page 61: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

57

Gonzalez, AC, Jagdmann, S., Schweizer, M., Bernreuther,C., Reinheckel, T., Saftig, P, Damme, M. (2018)

Unconventional trafficking of mammalian phospholipase D3 to lysosomes. Cell Rep., 22(4):1040-1053.

8.3

Haas A, Hensel M, Lührmann A, Rudel T, Saftig P, Schaible UE Intracellular compartments of pathogens:

Highways to hell or stairways to heaven? (2018) Int J Med Microbiol, 308(1):1-2 3.4

Hedemann N, Rogmans C, Sebens S, Wesch D, Schmidt-Arras D, Pecks U, van Mackelenbergh M, Weimer J,

Arnold N, Maas N, Bauerschlag DO. ADAM17 inhibition enhances platinum efficience in ovarian

cancer. (2018) Oncotarget 9(22):16043-16058

Holz K, Prinz M, Bredecke SM, Mittrücker H-W, Rose-John S, Hölscher C (2018) Differing outcome of

experimental autoimmune encephalitis in macrophage/neutrophil- and T cell-specific gp130-deficient

mice. Front Immunol 9: 836 4.716

Kaiser K, Prystaz K, Vikman A, Haffner-Luntzer M, Bergdolt S, Strauss G, Waetzig GH, Rose-John S, Ignatius

A (2018) Pharmacological inhibition of IL-6 trans-signaling improves compromised fracture healing after

severe trauma. Naunyn Schmiedebergs Arch Pharmacol 391: 523-536 2.058

Khateb S, Kowalewski B, Bedoni N, Damme M, Pollack N, Saada A, Obolensky A, Ben-Yosef T, Gross M,

Dierks T, Banin E, Rivolta C, Sharon D. (2018). A homozygous founder missense variant in arylsulfatase

G abolishes its enzymatic activity causing atypical Usher syndrome in humans. Genet Med. (9):1004-

1012. 8.683

Kissing S, Saftig P, Haas A. Vacuolar ATPase in phago(lyso)some biology. (2018) Int J Med Microbiol,

308(1):58-67 3.4

Krossa, S., Scheidig, A.J., Grötzinger, J., Lorenzen, I. (2018) Redundancy of protein disulfide isomerases in the

catalysis of the inactivating disulfide switch in A Disintegrin and Metalloprotease 17. Sci Rep 8, 1103.

4.259

Lemke H. (2018) Immune Response Regulation by Antigen Receptors’ Clone-Specific Non-Self Parts. Front.

Immunol. 9:1471 6.429

Linsenmeier,L., Mohammadi, B., Wetzel,S., Puig,B., Jackson,W.S., Hartmann,A., Uchiyama,K., Sakaguchi,S.,

Endres, K., Tatzelt, J., Saftig,P., Glatzel, M., Altmeppen, H.C. (2018) Structural and mechanistic

aspects influencing the ADAM10-mediated shedding of the prion protein. Mol. Neurodegen., 13(1):18

6.7

Lokau, J. Göttert, S., Arnold, P., Düsterhöft, S., Massa López, D., Grötzinger, J., Garbers, C. (2018) The SNP

rs4252548 (R112H) which is associated with reduced human height compromises the stability of IL-11.

BBA-Mol Cell Res 1865, 496-506. 4.521

Lücke K, Yan I, Krohn S, Volmari A, Klinge S, Schmid J, Schumacher V, Steinmetz OM, Rose-John S,

Mittrücker H-W (2018) Control of Listeria monocytogenes infection requires classical IL-6 signaling in

myeloid cells. PlosOne 13: e0203395 2.776

Machado-Pineda, Y., Reyes, R., Cardenes, B., Loppez-Martin, S., Toriba, V. Sanchez-Organero, P., Grötzinger,

J., Lorenzen, I., Yanez-Mo, M., Cabanas, C. (2018) CD9 controls integrin a5b1-mediated cell adhesion

by modulating its assciciation with the metalloproteinase ADAM17. Front. Immunol. 9, 2474. 5.511

Nicolaou A, Northoff BH, Zhao Z, Kohlmaier A, Sass K, Rose-John S, Steffens S, Weber C, Teupser D, Holdt

LM (2018) The ADAM17 metalloproteinase maintains arterial elasticity. Thromb Haemost. 118: 210-213

4.662

Pastor VB, Sahoo SS, Boklan J, Schabe GC, Saribeyoglu E, Strahm B,Lebrecht D, Voss M, Bryceson YT,

Erlacher M, Ehninger C, Niewisch M, Schlegelberger B, Baumann I, Achermann JC, Shimamura A,

Hochrein J, Tedgård U, Nilsson L, Hasle H, Boerries M, Busch H, Niemeyer CM, Wlodarski MW (2018).

Constitutional SAMD9L mutations cause familial myelodysplastic syndrome and transient monosomy 7.

Haematologica 103, 427-437. 7.570

Pohl, S., Angermann, A., Jeschke, A., Hendickx, Yorgan, T.A., Makrypidi-Fraune, G., Steigert, A. Kuehn, S.C.,

Rolvien, T., Schweizer, M., Koehne, T., Neven, M., Winter, O., Voltolini Velho, R., Albers, J., Streichert,

T., Pestka, J.M., Baldauf, C., Breyer, S., Stuecker, R., Muschol, N., Cox, T.M., Saftig, P., Paganini, C.,

Rossi, A., Amling, M., Braulke, T., Schinke, T. (2018) The lysosomal protein arylsulfatase B is a key

enzyme involved in skeletal turnover. J. Bone Miner. Res., 33(12):2186-2201 6.2

Prystaz K, Kaiser K, Kovtun A, Haffner-Luntzer M, Fischer V, Strauss G, Waetzig GH, Rose-John S, Ignatius A

(2018) Distinct effects of interleukin-6 classic and trans-signaling in bone fracture healing. Am J Pathol

188: 474-490 3.762

Rose-John S (2018) IL-6 family cytokines. Cold Spring Harb Perspect Biol 10: pii: a028415. doi:

10.1101/cshperspect.a028415 9.110

Schmidt S, Schumacher N, Schwarz J, Roos S, Kenner L, Schlederer M, Sibilia M, Linder M, Altendorf-

Hofmann A, Knoesel T, Gruber E, Oberhuber G, Rehman A, Sinha A, Arnold P, Zunke F, Becker-Pauly

C, Preaudet A, Nguyen P, Huynh J, Chand A, Westermann J, Dempsey PJ, Garbers C, Rosenstiel

P, Putoczki T, Ernst M, Rose-John S (2018) ADAM17 is required for EGF-R induced intestinal tumors

via IL-6 trans-signaling. J Exp Med 215: 1205-1225 10.892

Page 62: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

58

Seipold, L., Altmeppen,H., Koudelka, T., Tholey,A., Kasparek,P., Sedlacek,R., Schweizer, M., Bär,J.,

Mikhaylova,M., Glatzel, M., Saftig, P. (2018) In Vivo Regulation of the A Disintegrin And

Metalloproteinase 10 (ADAM10) by the Tetraspanin 15, Cell Mol. Life Sci., 75(17):3251-3267. 5.7

Stahl FR, Jung R, Jazbutyte V, Ostermann E, Tödter S, Brixel R, Kemmer A, Halle S, Rose-John S, Messerle M,

Arck PC, Brune W, Renné T (2018) Laboratory diagnostics of murine blood for detection of mouse

cytomegalovirus (MCMV)-induced hepatitis. Sci Rep 8: 14823 4.011

Talantikite M, Lécorché P, Beau F, Damour O, Becker-Pauly C, Ho WB, Dive V, Vadon-Le Goff S, Moali C.

(2018) Inhibitors of BMP-1/tolloid-like proteinases: efficacy, selectivity and cellular toxicity. FEBS

OpenBio, 8(12):2011-2021. 1.782

Tesi B, Rascon J, Chiang SCC, Burnyte B, Löfstedt A, Fasth A, Heizmann M, Juozapaite S, Kiudeliene R,

Kvedaraite E, Miseviciene V, Muleviciene A, Müller ML, Nordenskjöld M, Matuzeviciene R, Samaitiene

R, Speckmann C, Stakeviciene S, Zekas V, Voss M, Ehl S, Vaiciene-Magistris N, Henter JI, Meeths M,

Bryceson YT (2018). A RAB27A 5’UTR structural variant associated with late-onset hemophagocytic

lymphohistiocytosis and normal pigmentation. J Allergy Clin Immunol 142, 317-321. 14.110

Ullrich E, Abendroth B, Rothamer J, Huber C, Büttner-Herold M, Kitowski V, Vogler T, Longerich T, Zundler

S, Völkl S, Beilhack A, Rose-John S, Wirtz S, Weber GF, Ghimire S, Kreutz M, Holler E, Mackensen A,

Neurath MF, Hildner K (2018) BATF-dependent IL-7RhiGM-CSF+ T cells control intestinal graft-

versus-host disease. J Clin Invest 128: 916-930 12.282

Zunke F, Moise AC, Belur NR, Gelyana E, Stojkovska I, Dzaferbegovic H, Toker NJ, Jeon S, Fredriksen K,

Mazzulli JR (2018) Reversible Conformational Conversion of alpha-Synuclein into Toxic Assemblies by

Glucosylceramide. Neuron 97: 92-107. 14.403

2019 Aden K, Bartsch K, Dahl J, Reijns MAM, Esser D, Sheibani-Tezerji R, Sinha A, Wottawa F, Ito G, Mishra N,

Knittler K, Burkholder A, Welz L, van Es J, Tran F, Lipinski S, Kakavand N, Boeger C, Lucius R, von

Schoenfels W, Schafmayer C, Lenk L, Chalaris A, Clevers H, Röcken C, Kaleta C, Rose-John S,

Schreiber S, Kunkel T, Rabe B, Rosenstiel P (2019) Epithelial RNase H2 Maintains Genome Integrity

and Prevents Intestinal Tumorigenesis in Mice. Gastroenterology 156: 145-159.e19 19.233

Aparicio-Siegmund S, Garbers Y, Flynn CM, Waetzig GH, Gouni-Berthold I, Krone W, Berthold HK, Laudes

M, Rose-John S, Garbers C (2019) The IL-6-neutralizing sIL-6R/sgp130 buffer system is disturbed in

patients with type 2 diabetes mellitus. Am J Physiol 317(2):E411-E420 4.125

Bartels, A-K., Göttert, S., Desel, C., Schäfer, M., Krossa, S., Scheidig, A.J., Grötzinger, J., Lorenzen, I. (2019)

KDEL receptor 1 contributes to cell surface association of protein disulfide isomerases. Cell Physiol

Biochem 52,850-868 5.104

Bleibaum, F., Sommer, A., Veit, M., Rabe, B., Andrä, J., Kunzelmann, K., Nehls, C., Correa, W., Gutsmann, T.,

Grötzinger, J., Bhakdi, S., Reiss, K. (2019) ADAM10 sheddase activation is controlled by cell membrane

symmetry. J Mol Cell Biol. 11:979-993. 3.813

Bolik J, Tirnitz-Parker JEE and Schmidt-Arras D, ADAM and ADAMTS proteases in hepatic disorders, J Ren

Hepat Disord 2019;3(1):23–32

Boon L, Ugarte-Berzal E, Martens E, Vandooren J, Rybakin V, Colau D, Gordon-Alonso M, van der Bruggen P,

Stöcker W, Becker-Pauly C, Witters P, Morava E, Jaeken J, Proost P, Opdenakker, G. (2019) Propeptide

glycosylation and galectin-3 binding decrease proteolytic activation of human progelatinase B/proMMP-

9. FEBS J, 286(5):930-945. 4.739

Cuddy LK, Wani WY, Morella ML, Pitcairn C, Tsutsumi K, Fredriksen K, Justman CJ, Grammatopoulos TN,

Belur NR, Zunke F, Subramanian A, Affaneh A, Lansbury PT Jr, Mazzulli JR. (2019) Stress-Induced

Cellular Clearance Is Mediated by the SNARE Protein ykt6 and Disrupted by α-Synuclein. Neuron. 104:

869-884. 14.403 dos Santos Guilherme M, Stoye NM, Rose-John S, Garbers C, Fellgiebel A, Endres K (2019) The synthetic

retinoid acitretin increases IL-6 in the central nervous system of Alzheimer disease model mice and

human patients. Front Aging Neurosci 11: 182 4.504

Escrig A, Canal C, Sanchis P, Fernández-Gayol O, Montilla A, Comes G, Molinero A, Giralt M, Giménez-Llort

L, Becker-Pauly C, Rose-John S, Hidalgo J. (2019) IL-6 trans-signaling in the brain influences the

behavioral and physio-pathological phenotype of the Tg2576 and 3xTgAD mouse models of Alzheimer's

disease. Brain Behav Immun. S0889-1591(19)30150-3 6.306

Escrig A, Molinero A, Méndez B, Sanchis P, Fernández-Gayol O, Montilla A, Comes G, Giralt M, LaFerla FM,

Giménez-Llort L, Becker-Pauly C, Rose-John S, Hidalgo J (2019) IL-6 trans-signaling in the brain

influences the behavioral and physiological phenotype of the 3xTgAD mouse model of Alzheimer’s

disease. Brain Behav Immun 82: 145-159 6.170

Findeisen M, Allen TL, Henstridge DC, Kammoun HL, Brandon AE, Baggio LL, Watt KI, Pal M, Cron L,

Estevez E, Yang C, Kowalski GM, O’Reilly L, Egan C, Sun E, Thai LM, Krippner G, Adams TE, Lee

Page 63: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

59

RS, Grötzinger J, Garbers C, Risis S, Kraakman MJ, Mellet N, Sligar J, Kimber ET, Young RL, Cowley

MA, Bruce CR, Meikle PJ, Baldock PA, Gregorevic P, Biden TJ, Cooney GJ, Keating DJ, Drucker DJ,

Rose-John S & Febbraio MA (2019) Treatment of type 2 diabetes and muscle atrophy with the designer

cytokine IC7Fc. Nature 574: 63-68 43.070

Guedj A, Volman Y, Geiger-Maor A, Bolik J, Kuenzel S, Nevo Y, Baines J, Elgavish S, Galun E, Amsalem H,

Schmidt-Arras D, Rachmilewitz J. Gut microbiota shape 'inflamm-aging' cytokines and account for age-

dependent decline in DNA damage repair (2019) Gut doi: 10.1136/gutjnl-2019-318491 epub ahead of

print 17.943

Heichler C, Scheibe K, Schmied A, Geppert C, Schmid B, Wirtz S, Thoma A-M, Kramer V, Waldner M, Büttner

, Henner F, Farin H, Pesic M, Knieling F, Merkel S, Grüneboom A, Gunzer M, Grützmann R, Rose-John

S, Koralov SB, Kollias G, Vieth M, Hartmann A, Greten FR, Neurath MF, Neufert C (2019) STAT3

activation through IL-6/IL-11 in cancer-associated fibroblasts promotes colorectal tumor development

and correlates with poor prognosis. Gut, pii: gutjnl-2019-319200 17.943

Heybrock, S., Kanerva, K., Meng, Y., Ing, C., Liang, A., Xiong, Z.J., Wenig, X., Kim, Y.A., Collins, R.A.,

Trimble, W., Pomès, R., Privé, G.G., Annaert, W., Schwake, M., Heeren, J., Lüllmann-Rauch, R.,

Grinstein, S.*, Ikonen, E.*, Saftig, P.*, Neculai, D.4* (2019) Lysosomal Integral Membrane Protein-2

(LIMP-2/SCARB2) is involved in lysosomal cholesterol export. Nat. Comm., 10(1):3521 12.35

Karmilin K, Schmitz C, Kuske M, Körschgen H, Olf M, Meyer K, Hildebrand A, Felten M, Fridrich S,

Yiallouros I, Becker-Pauly C, Weiskirchen R, Jahnen-Dechent W, Floehr J, Stöcker W. (2019)

Mammalian plasma fetuin-B is a selective inhibitor of ovastacin and meprin metalloproteinases. Sci Rep.

9(1):546. 4.011

Khundadze M, Ribaudo F, Hussain A, Rosentreter J, Nietzsche S, Thelen M, Winter D, Hoffmann B, Afzal MA,

Hermann T, de Heus C, Piskor EM, Kosan C, Franzka P, von Kleist L, Stauber T, Klumperman J,

Damme M, Proikas-Cezanne T, Hübner CA. (2019). A mouse model for SPG48 reveals a block of

autophagic flux upon disruption of adaptor protein complex five. Neurobiol Dis.127:419-431. 5.160

Kleinegger F, Hofer E, Wodlej C, Golob-Schwarzl N, Birkl-Toeglhofer AM, Stallinger A, Petzold J, Orlova A,

Krassnig S, Reihs R, Niedrist T, Mangge H, Park YN, Thalhammer M, Aigelsreiter A, Lax S, Garbers C,

Fickert P, Rose-John S, Moriggl R, Rinner B, Haybaeck J (2019) Pharmacologic IL-6Rα inhibition in

cholangiocarcinoma promotes cancer cell growth and survival. Biochim Biophys Acta Mol Basis Dis

1865: 308-321 4.328

Li, X., Qin, L., Li, Y., Yu, H., Zhang, Z., Tao, C., Liu, Y., Xue, Y., Zhang, X., Xu, Z., Wang, Y., Lou, H., Tan,

Z., Saftig, P., Chen, Z., Xu, T., Bi, G., Duan, S., Gao, Z. (2019) Presynaptic endosomal cathepsin D

regulates the biogenesis of GABAeric synaptic vesicles. Cell Rep. 28(4):1015-1028. 7.81

Liao C-W; Chou C-H; Wu X-M; Chen Z-W; Chen Y-H; Chang Y-Y; Wu V-C; Rose-John S; Hung C-S; Lin Y-

H (2019) Interleukin-6 plays a critical role in aldosterone-induced macrophage recruitment and

infiltration in the myocardium. Biochim Biophys Acta – Molecular Basis of Disease165627 4.328

Liebsch, F., Kulic, L., Teunissen, C., Shobo, A., Ulku, I., Engelschalt, V., Hancock, M.A., van der Flier,

W.M., Kunach, P., Rosa-Neto, P.,Scheltens, P., Poirier, J., Saftig, P., Bateman, R.J., Breitner, J., Hock,

C., Multhaup, G., PREVENT-AD Research Group (2019) Aβ34 is a BACE1-derived degradation

intermediate associated with amyloid clearance and Alzheimer’s disease progression Nat. Comm.,

10(1):2240 12.35

Marques ARA, Di Spiezio A, Thießen N, Schmidt L, Grötzinger J, Lüllmann-Rauch R, Damme M, Storck SE,

Pietrzik CU, Fogh J, Bär J, Mikhaylova M, Glatzel M, Bassal M, Bartsch U, Saftig P. (2019). Enzyme

replacement therapy with recombinant pro-CTSD (cathepsin D) corrects defective proteolysis and

autophagy in neuronal ceroid lipofuscinosis. Autophagy. 16:1-15 11.059

Marques, A.R.A. & Saftig, P. (2019) Lysosomal storage disorders: challenges, concepts and avenues for therapy

- beyond rare diseases. J. Cell Sci., 132(2) pii: jcs221739 4.4

Marques, A.R.A., Di Spiezio, A., Thießen,T., Schmidt, L., Grötzinger, J., Lüllmann-Rauch, R., Damme, M.,

Storck, S.E., Pietrzik, C.U., Fogh, J., Bär,J., Mikhaylova, M., Glatzel, M., Bassal, M., Bartsch, U., Saftig,

P. (2019) Enzyme replacement therapy with recombinant pro-CTSD (cathepsin D) corrects defective

proteolysis and autophagy in neuronal ceroid lipofuscinosis. Autophagy, 16:1-15 11.059

Massa Lopez, D., Thelen, M., Stahl, F., Thiel, C., Linhorst, A., Sylvester, M., Hermanns-Borgmeyer, I.,

Lüllmann-Rauch, R., Eskild, W., Saftig, P., Damme, M. (2019). The lysosomal transporter MFSD1 is

essential for liver homeostasis and critically depends on its accessory subunit GLMP. Elife. 8. pii:

e50025. 7.551

Moreau D, Vacca F, Vossio S, Scott C, Colaco A, Paz Montoya J, Ferguson C, Damme M, Moniatte M, Parton

RG, Platt FM, Gruenberg J. (2019). Drug-induced increase in lysobisphosphatidic acid reduces the

cholesterol overload in Niemann-Pick type C cells and mice. EMBO Rep. 20(7):e47055. 8.383

Niemeyer, J., Mentrup, T., Heidasch, R., Mueller, S., Biswas, U., Meyer, R., Papadopoulou, A., Dederer, V.,

Haug-Kroeper, M., Adamski,V., Lüllmann-Rauch, R., Bergmann, M., Mayerhofer, A., Saftig,

P., Wennemuth, G., Jessberger, R., Fluhrer, R., Lichtenthaler,S., Lemberg, M., Schröder, B. (2019) The

Page 64: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

60

intramembrane protease SPPL2c promotes male germ cell development by cleaving phospholamban.

EMBO REP., 20(3) pii: e46449 8.383

Notomi, S., Ishihara, K., Efstathiou,N.E., Jong-Jer, L., Hisatomi, T., Tachibana, T., Konstantinou, E., Ueta, T.,

Murakami, Y., Maidana, D.E., Ikeda,Y., Kume, S., Terasaki, H., Sonoda, S., Blanz, J., Young, L.,

Sakamoto, T., Sonoda,K.H., Saftig, P., Ishibashi, T., Miller, J.W., Kroemer, G., Vavvas, D.G. (2019)

Genetic LAMP2 deficiency accelerates the age-associated formation of basal laminar deposits in the

retina. Proc. Natl. Acad. Sci., 116:23724-23734 9.58

Paige E, Clément M, Lareyre F, Sweeting M, Raffort J, Grenier C, Finigan A, Harrison J, Peters JE, Sun BB,

Butterworth AS, Harrison SC, Bown MJ, Lindholt JS, Badger SA, Kullo IJ, Powell J, Norman PE, Scott

JA, Bailey MA, Rose-John S, Danesh J, Freitag DF, Paul DS, Mallat Z (2019) Interleukin-6 Receptor

Signalling and Abdominal Aortic Aneurysm Growth Rates. Circ Genom Precis Med 12: e002413 4.743

Pasquire, A., , Vivot, K., Erbs, E., Spiegelhalter, C., Zhanh, Z., Aubert, V., Senkara, M., Maillard, E., Pinget, M.,

Kerr-Conte, J., Pattou, F., Marciniak, G., Ganzhorn, A., Muzet, N., Rooney, E., Ronchi, P., Schieber,

N.L., Schwab, Y., Saftig, P., Goginashvili, A., Ricci, R. (2019) Lysosomal degradation of newly formed

insulin granules contributes to cell failure in type 2 diabetes Nat. Comm., 10(1):3312 12.35

Pavlenko E, Cabron A-S, Arnold P, Dobert JP, Rose-John S Zunke F (2019) Functional characterization of colon

cancer-associated mutations in ADAM17: modifications in the pro-domain interfere with trafficking and

maturation. Int J Mol Sci 20: pii: E2198 4.183

Pavlenko E, Cabron AS, Arnold P, Dobert JP, Rose-John S, Zunke F (2019) Functional Characterization of

Colon Cancer-Associated Mutations in ADAM17: Modifications in the Pro-Domain Interfere with

Trafficking and Maturation. Int J Mol Sci 20 4.183 Peters F & Becker-Pauly C*. Role of meprin metalloproteases in metastasis and tumor microenvironment.

Cancer Metastasis Rev. 2019 Sep;38(3):347-356 6.667

Peters F, Scharfenberg F, Colmorgen C, Armbrust F, Wichert R, Arnold P, Potempa B, Potempa J, Pietrzik CU,

Häsler R, Rosenstiel P, Becker-Pauly C*. (2019) Tethering soluble meprin α in an enzyme complex to the

cell surface affects IBD associated genes. FASEB J, 9(1):546.. 5.391

Prenissl N, Lokau J, Rose-John S, Haybaeck J, Garbers C (2019) Therapeutic blockade of the interleukin-6

receptor (IL-6R) allows sIL-6R generation by proteolytic cleavage. Cytokine 114: 1-5 3.078

Quarta S, Mitrić M, Kalpachidou T, Mair N, Schiefermeier-Mach N, Andratsch M, Qi Y, Langeslag M, Malsch

P, Rose-John S, Kress M (2019) Impaired mechanical, heat and cold nociception in a murine model of

genetic TACE/ADAM17 knockdown. FASEB J 33: 4418-4431 5.391

Reinicke, A.T., Laban, K., Sachs, M., Kraus, V., Walden, M., Damme, M., Sachs, W., Reichelt, J., Schweizer,

M., Janiesch, P.C., Duncan, K.E., Saftig, P., Rinschen, M.M., Morellini, F., Meyer-Schwesinger, C.

(2019) Ubiquitin C-Terminal Hydrolase L1 (UCH-L1) loss causes neurodegeneration by altering protein

turnover in the first postnatal weeks. Proc. Natl. Acad. Sci, USA, 116(16):7963-7972 9.58

Riederer P, Berg D, Casadei N, Cheng F, Classen J, Dresel C, Jost W, Kruger R, Muller T, Reichmann H, Riess

O, Storch A, Strobel S, van Eimeren T, Volker HU, Winkler J, Winklhofer KF, Wullner U, Zunke F,

Monoranu CM (2019) alpha-Synuclein in Parkinson's disease: causal or bystander? J Neural Transm

(Vienna) 126: 815-840 2.903 Saad MI, Alhayyani S, McLeod L, Yu L, Alanazi M, Deswaerte V, Tang K, Jarde T, Smith JA, Prodanovic Z,

Watkins N, Cain JE, Bozinovski S, Algar E, Ferlin W, Garbers C, Ruwanpura S, Sagi I, Rose-John S,

Jenkins BJ (2019) ADAM17 selectively activates the IL-6 trans-signaling/ERK MAPK axis in KRAS-

addicted lung cancer. EMBO Mol Med pii: e9976 10.624

Saad MI, McLeod L, Yu L, Ebi H, Ruwanpura S, Sagi I, Rose-John S, Jenkins BJ (2019) The ADAM17 protease

promotes tobacco smoke carcinogen-induced lung tumorigenesis. Carcinogenesis doi:

10.1093/carcin/bgz123 4.004

Saad MI, Rose-John S, Jenkins BJ (2019) ADAM17: An emerging therapeutic target for lung adenocarcinoma.

Cancers 11: E1218 6.162

Sammel M, Peters F, Lokau J, Scharfenberg F, Werny L, Linder S, Garbers C, Rose-John S, Becker-Pauly C*.

(2019) Differences in Shedding of the Interleukin-11 Receptor by the Proteases ADAM9, ADAM10,

ADAM17, Meprin α, Meprin β and MT1-MMP. Int J Mol Sci. 20: 15 4.183

Schäffler H, Li W, Helm O, Krüger S, Böger C, Peters F, Röcken C, Sebens S, Lucius R, Becker-Pauly C,

Arnold P. (2019) The cancer-associated meprin β variant G32R provides an additional activation site and

promotes cancer cell invasion. J Cell Sci, 132(11). 4.517

Scharfenberg F, Armbrust F, Marengo L, Pietrzik CU, Becker-Pauly C*. (2019) Regulation of the alternative β-

secretase meprin β by ADAM-mediated shedding. Cell Mol Life Sci, 76(16):3193-3206. 7.014

Scharfenberg F, Helbig A, Sammel M, Benzel J, Schlomann U, Peters F, Wichert R, Bettendorff M, Schmidt-

Arras D, Rose-John S, Moali C, Lichtenthaler SF, Pietrzik CU, Bartsch JW, Tholey A, Becker-Pauly C

(2019) Degradome of soluble ADAM10 and ADAM17 metalloproteases. Cellular and Molecular Life

Sciences, doi: 10.1007/s00018-019-03184-4 7.014

Page 65: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

61

Scherger AK, Al-Maarri M, Maurer HC, Schick M, Maurer S, Öllinger R, Gonzalez-Menendez I, Martella M,

Thaler M, Pechloff K, Steiger K, Sander S, Ruland J, Rad R, Quintanilla-Martinez L, Wunderlich FT,

Rose-John S, Keller U (2019) Activated gp130 signaling selectively targets B cell differentiation to

promote transformation of mature lymphoma and plasmacytoma. JCI Insight 4: pii: 128435 6.014

Schmidt-Arras D, Rose-John S (2019) Regulation of fibrotic processes in the liver by ADAM proteases. Cells 8:

1226 5.656

Schumacher N, Rose-John S (2019) ADAM17 activity and IL-6 Trans-Signaling are required for EGF-R driven

Colon Cancer. Cancers 11: E1736 6.162

Servais FA, Kirchmeyer M, Hamdorf M, Minoungou N, Rose-John S, Kreis S, Haan C, Behrmann I. Modulation

of the IL-6 signaling pathway in liver cells by miRNAs targeting gp130, JAK1 and/or STAT3 (2019)

Molecular Therapy - Nucleic Acids 16: 419-433 5.919

Sommer D, Corstjens I, Sanchez S, Dooley D, Lemmens S, Van Broeckhoven J, Bogie J, Vanmierlo T, Vidal

PM, Rose-John S, Gou-Fabregas M, Hendrix S (2019) ADAM17-deficiency on microglia but not on

macrophages promotes phagocytosis and functional recovery after spinal cord injury. Brain Behav

Immun 80:129-145 6.170

Vezzoli, E., Caron, I., Talpo, F., Besusso, D., Conforti, P., Battaglia, E., Sogne, E., Falqui, A.,Petricca, L.,

Verani, M., Martufi, P., Caricasole, A., Bresciani, A.,Cecchetti, O., Rivetti di Val Cervo, P., Sancini,

G., Riess, O., Nguyen, H., Seipold, L., Saftig, P., Biella, G., Cattaneo, E., Zuccato, C. (2019) Inhibiting

pathologically active ADAM10 rescues synaptic and cognitive decline in Huntington’s disease. J. Clin.

Invest., 129(6):2390-2403 12.282

Walter S, Jumpertz T, Hüttenrauch M, Gerber H, Dimitrov M, Ogorek I, Lehmann S, Lepka K, Berndt C,

Wiltfang J, Becker-Pauly C, Beher D, Pietrzik CU, Fraering PC, Wirths O, Weggen S. (2019) The

metalloprotease ADAMTS4 generates N-truncated Aβ4-x peptides and marks oligodendrocytes as pro-

amyloidogenic in Alzheimer’s disease. Acta Neuropathologica, 137:239–257. 18.174

Wichert R, Scharfenberg F, Koudelka T, Colmorgen C, Schwarz J, Wetzel S, Potempa B, Potempa J, Bartsch

JW, Sagi I, Tholey A, Saftig P, Rose-John S, Becker-Pauly C*. (2019) Meprin β induces activities of A

Disintegrin and Metalloproteinases 9, 10 and 17 by specific prodomain cleavage. FASEB J,

33(11):11925-11940. 5.391

Wilkinson A, Gartlan K, Kuns R, Chang K, Minnie S, Ensbey K, Clouston A, Zhang P, Koyama M, Hidalgo J,

Rose-John S, Varelias A, Vuckovic S, Hill G (2019) IL-6 dysregulation originates in dendritic cells and

initiates graft-versus-host disease via classical signaling. Blood 134: 2092-210616.562

Wünnemann F, Ta-Shma A, Preuss C, Leclerc S, van Vliet PP, Oneglia A, Thibeault M, Nordquist E, Lincoln J,

Scharfenberg F, Becker-Pauly C, Hofmann P, Hoff K, Audain E, Kramer HH, Makalowski W, Nir A,

Gerety S, Hurles M, Comes J, Fournier A, Osinska H, Robins J, Puceat M, MIBAVA Leducq

Consortium, Elpeleg O, Hitz MP*, Andelfinger G*. (2019) Loss of ADAMTS19 causes progressive non-

syndromic heart valve disease. Nat Genet. doi: 10.1038/s41588-019-0536-2 25.455

Ziegler L, Gajulapuri A, Frumento P, Bonomi A, Wallén H, de Faire U, Rose-John S, Gigante B (2019)

Interleukin 6 Trans-Signalling and Risk of Future Cardiovascular Events. Cardiovasc Res 115: 213-221

7.014

Zunke F, Mazzulli JR (2019) Modeling neuronopathic storage diseases with patient-derived culture systems.

Neurobiol Dis 127: 147-162 5.16

Page 66: The Biochemical Institute Christian-Albrechts-University

Research Report Biochemical Institute, Christian-Albrechts-University Kiel

62

Accumulated Impact Factors of publications from the Biochemical Institute

2018: 368.949

2019: 492.377

0

100

200

300

400

500

600

2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 2017 2018 2019