TY - JOUR A1 - Siegmund, Daniela A1 - Kums, Juliane A1 - Ehrenschwender, Martin A1 - Wajant, Harald T1 - Activation of TNFR2 sensitizes macrophages for TNFR1-mediated necroptosis JF - Cell Death & Disease N2 - Macrophages express TNFR1 as well as TNFR2 and are also major producers of tumor necrosis factor (TNF), especially upon contact with pathogen-associated molecular patterns. Consequently, TNF not only acts as a macrophage-derived effector molecule but also regulates the activity and viability of macrophages. Here, we investigated the individual contribution of TNFR1 and TNFR2 to TNF-induced cell death in macrophages. Exclusive stimulation of TNFR1 showed no cytotoxic effect whereas selective stimulation of TNFR2 displayed mild cytotoxicity. Intriguingly, the latter was strongly enhanced by the caspase inhibitor zVAD-fmk. The strong cytotoxic activity of TNFR2 in the presence of zVAD-fmk was reversed by necrostatin-1, indicating necroptotic cell death. TNFR1- and TNF-deficient macrophages turned out to be resistant against TNFR2-induced cell death. In addition, the cIAP-depleting SMAC mimetic BV6 also enforced TNF/TNFR1-mediated necroptotic cell death in the presence of zVAD-fmk. In sum, our data suggest a model in which TNFR2 sensitizes macrophages for endogenous TNF-induced TNFR1-mediated necroptosis by the known ability of TNFR2 to interfere with the survival activity of TRAF2-cIAP1/2 complexes. KW - TNFR2 Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-162317 VL - 7 ER - TY - THES A1 - Shaikh, Muhammad Haroon T1 - Nicht-hämatopoetische lymphoide Stromazellen aktivieren alloreaktive CD4\(^+\) T-Zellen in der Initiierung der akuten Graft-versus-Host Disease T1 - Non-hematopoietic lymphoid stromal cells prime alloreactive CD4\(^+\) T cells during acute graft-versus-host disease N2 - In der Initiationsphase der akuten Graft-versus-Host Erkrankung (GvHD) werden CD4+ T-Zellen in den lymphatischen Organen durch hämatopoietische Antigen-präsentierende Zellen aktiviert. Im Gegensatz dazu, werden in der Effektorphase CD4+ T-Zellen von nicht-hämatopoetischen Zellen im Dünndarm aktiviert. Wir stellten die Hypothese auf, dass alloreaktive CD4+ T-Zellen nach allogener hämatopoetischer Zelltransplantation, welche in der Initiationsphase der aGvHD vorwiegend in die sekundären lymphatischen Organe migrieren, dort durch nicht-hämatopoetische Lymphknoten-Stromazellen über die Erkennung von MHC-Klasse II aktiviert werden. Um diese Hypothese zu testen, setzten wir ein von allogenen CD4+ T-Zellen-abhängiges MHC Major Mismatch aGvHD Mausmodell ein, um diese Zusammenhänge näher zu erforschen. Mittels Biolumineszenz-Bildgebung und dreidimensionale Lichtblattmikroskopie und Durchflusszytometrie-Analysen von früheren Zeitpunkten nach einer alloHCT bzw. im Anfangsstadium der aGvHD konnten wir zeigen, dass allogene T-Zellen exklusiv in die Milz, Lymphknoten und die Peyerschen Plaques migrieren und nicht in die intestinale Lamina propria. Indem wir transgene Mauslinien verwendeten, die keine oder eine nur partielle komplette hämatopoietische Antigenpräsentation aufwiesen, konnten wir eine sehr früh auf die alloHCT folgende allogene CD4+ T-Zellaktivierung in den lymphoiden Organen von MHCIIΔCD11c and MHCIIΔ Knochenmark-Chimären nachweisen. Aufgrund des, bei den MHCIIΔ Knochenmarks-Chimären auftretenden Versagens der negativen Thymusselektion und die daraus resultierende autoreaktive Immunreaktionen nach einer syngenen HCST stellte sich heraus, dass dies ein ungeeignetes Modell für die Untersuchung der Präsentation nicht-hämatopoetischer Antigene bei GvHD ist. Um diese Herausforderung zu bewältigen, generierten wir MHCIIΔVav1 Mäuse bei denen die MHC-Klasse-II-Expression auf allen hämatopoetischen Zellen fehlt. MHCIIΔVav1 Mäuse entwickelten eine aGvHD, wobei die Lymphknoten-Stromazellen dieser Tiere allogene CD4+ T-Zellen in gemischten Lymphozytenreaktionen aktivieren konnten. Ebenso konnten mesenteriale Lymphknoten von CD11c.DTR-Mäusen, die zuvor in eine MHCIIΔ Maus transplantiert wurden, CD4+ T-Zellen in vivo aktivieren, wodurch die Lymphknoten-Stromazellen eindeutig als nicht-hämatopoetische Antigen-präsentierende Zellen der lymphoiden Organe nachgewiesen werden konnten. Über das Cre/loxP-System konnten wir Knockout-Mäuse mit fehlender MHCII-Expression in Subpopulationen von Lymphknoten-Stromazellen generieren und verwendeten dann Einzelzell-RNA-Sequenzierung. Hier wählten wir Ccl19 und VE-Cadherin aus, um unsere Analyse spezifisch auf die fibroblastischen retikulären Zellen bzw. Endothelzellen der Lymphknoten zu konzentrieren. Bei MHCIIΔCcl19 Mäusen war die Aktivierung alloreaktiver CD4+ T-Zellen in der Initiationsphase der aGvHD mäßig reduziert, während das Fehlen von MHCII auf den fibroblastischen retikulären Zellen zu einer Hyperaktivierung allogener CD4+ T-Zellen führte, was wiederum eine schlechtere Überlebensrate der Mäuse zur Folge hatte. Dieser Phänotyp wurde durch regulatorische T-Zellen moduliert, die in der Lage waren, H2-Ab1fl Mäuse von den Folgen von GvHD zu retten, jedoch nicht die MHCIIΔCcl19. Ein Knock-out von MHCII auf Endothelzellen von MHCIIΔVE-Cadherin Mäusen, führte in der Initiationsphase der GvHD nur zu einer mäßig reduzierten Aktivierung von CD4+ T-Zellen. Umgekehrt zeigten MHCIIΔVE-Cadherin Mäuse im Langzeitüberleben jedoch einen protektiven Phänotyp verglichen mit wurfgeschwister H2-Ab1fl Mäusen. Um die Bedeutung der MHCII-Antigenpräsentation der Endothelzellen zu untersuchen, generierten wir außerdem MHCIIΔVE-CadherinΔVav1 Mäuse, bei welchen eine Antigenpräsentation, weder im endothelialen noch im hämatopoetischen Kompartiment möglich war. Lymphknoten-Stromazellen von MHCIIΔVE-CadherinΔVav1 Mäusen waren nicht in der Lage, alloreaktive CD4+ T-Zellen in einer gemischten Lymphozytenreaktion zu aktivieren. Insgesamt konnten wir zum ersten Mal beweisen, dass die MHC-Klassse II auf den Lymphknoten-Stromazellen eine entscheidende Rolle bei der Modulation allogener CD4+ T-Zellen in der Initiations- und schließlich in der Effektorphase der Graft-versus-Host-Disease spielt. N2 - In the initiation phase of acute graft-versus-host disease (aGvHD), CD4+ T cells are activated by hematopoietic antigen presenting cells in secondary lymphoid organs whereas in effector phase by non-hematopoietic cells in the small intestine. We hypothesized that alloreactive CD4+ T cells primarily home to the secondary lymphoid organs subsequent to allogeneic hematopoietic cell transplantation in the initiation phase of aGvHD and are activated by the non-hematopoietic lymph node stromal cells via MHC class II. To test this hypothesis, we employed CD4+ T cell-dependent major mismatch aGvHD mouse model to study this correlation. Upon analyzing the early events following allo-HCT with bioluminescence imaging, flow cytometry and whole-mount light sheet fluorescence microscopy, we found that allogeneic T cells exclusively home to the spleen, lymph nodes and the Peyer’s patches and not to the intestinal lamina propria in the initiation phase of aGvHD. Utilizing mice devoid of partial or complete hematopoietic antigen presentation we could show allogeneic CD4+ T cells activation in the lymphoid organs of MHCIIΔCD11c and MHCIIΔ BM chimeric mice early after allo-HCT. MHCIIΔ BM chimeras failure of thymic negative selection and developing tissue wasting disease upon syn-HCT deemed them unsuitable to study non-hematopoietic antigen presentation in aGvHD. To overcome this challenge, we generated MHCIIΔVav1 mice that lack MHC class II expression on all hematopoietic cells. MHCIIΔVav1 mice were susceptible to aGvHD and LNSCs from these animals activated allogeneic CD4+ T cells in mixed lymphocyte reaction. Likewise, mesenteric lymph nodes from CD11c.DTR mice surgically transplanted into a MHCIIΔ mouse could activate CD4+ T cells in vivo, clearly demonstrating LNSCs as non-hematopoietic APCs of the lymphoid organs. We specifically target lymph node stromal cell subsets via the Cre/loxP system, we employed single cell RNA sequencing and selected Ccl19 and VE-Cadherin to specifically target the fibroblastic reticular cells and endothelial cells of the lymph nodes respectively. In MHCIIΔCcl19 mice, alloreactive CD4+ T cells activation was discreetly reduced in the initiation phase of aGvHD whereas absence of MHCII on fibroblastic reticular cells resulted in hyper-activation of allogeneic CD4+ T cells leading to poor survival. This phenotype was modulated by the regulatory T cells that were able to rescue H2-Ab1fl mice but not the MHCIIΔCcl19 subsequent to GvHD. Knock-out of MHCII on endothelial cells MHCIIΔVE Cadherin, resulted only in modest reduction of CD4+ T cells activation in the initiation phase of GvHD, conversely MHCIIΔVE Cadherin mice showed a protective phenotype compared against littermates H2-Ab1fl mice in long-term survival. Furthermore, to pin-point endothelial cells MHCII antigen presentation we generated MHCIIΔVE Cadherin ΔVav1 animals devoid of antigen presentation in both endothelial and hematopoietic compartments. LNSCs from MHCIIΔVE Cadherin ΔVav1 were unable to activate alloreactive CD4+ T cells in mixed lymphocyte reaction. Altogether, we demonstrate for the first time that MHC class II on the lymph node stromal cells plays a crucial role in the modulation of allogeneic CD4+ T cells in the initiation and later in the effector phase of graft-versus-host-disease. KW - Transplantat-Wirt-Reaktion KW - Graft-versus-host disease KW - Hematopoietic cell transplantation KW - CD4+ T cell activation Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-252015 ER - TY - JOUR A1 - Seher, Axel A1 - Nickel, Joachim A1 - Mueller, Thomas D. A1 - Kneitz, Susanne A1 - Gebhardt, Susanne A1 - Meyer ter Vehn, Tobias A1 - Schlunck, Guenther A1 - Sebald, Walter T1 - Gene expression profiling of connective tissue growth factor (CTGF) stimulated primary human tenon fibroblasts reveals an inflammatory and wound healing response in vitro JF - Molecular Vision N2 - Purpose: The biologic relevance of human connective tissue growth factor (hCTGF) for primary human tenon fibroblasts (HTFs) was investigated by RNA expression profiling using affymetrix (TM) oligonucleotide array technology to identify genes that are regulated by hCTGF. Methods: Recombinant hCTGF was expressed in HEK293T cells and purified by affinity and gel chromatography. Specificity and biologic activity of hCTGF was confirmed by biosensor interaction analysis and proliferation assays. For RNA expression profiling HTFs were stimulated with hCTGF for 48h and analyzed using affymetrix (TM) oligonucleotide array technology. Results were validated by real time RT-PCR. Results: hCTGF induces various groups of genes responsible for a wound healing and inflammatory response in HTFs. A new subset of CTGF inducible inflammatory genes was discovered (e.g., chemokine [C-X-C motif] ligand 1 [CXCL1], chemokine [C-X-C motif] ligand 6 [CXCL6], interleukin 6 [IL6], and interleukin 8 [IL8]). We also identified genes that can transmit the known biologic functions initiated by CTGF such as proliferation and extracellular matrix remodelling. Of special interest is a group of genes, e.g., osteoglycin (OGN) and osteomodulin (OMD), which are known to play a key role in osteoblast biology. Conclusions: This study specifies the important role of hCTGF for primary tenon fibroblast function. The RNA expression profile yields new insights into the relevance of hCTGF in influencing biologic processes like wound healing, inflammation, proliferation, and extracellular matrix remodelling in vitro via transcriptional regulation of specific genes. The results suggest that CTGF potentially acts as a modulating factor in inflammatory and wound healing response in fibroblasts of the human eye. KW - Bone morphogenetic protein-2 KW - Smooth-muscle-cells KW - Myofibroblast differentiation KW - TGF-beta KW - CYR61 KW - Proliferation KW - Mechanisms KW - Apoptosis KW - Receptor KW - Cancer Y1 - 2011 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-140189 VL - 17 IS - 08. Okt ER - TY - JOUR A1 - Seher, Axel A1 - Lagler, Charlotte A1 - Stühmer, Thorsten A1 - Müller-Richter, Urs Dietmar Achim A1 - Kübler, Alexander Christian A1 - Sebald, Walter A1 - Müller, Thomas Dieter A1 - Nickel, Joachim T1 - Utilizing BMP-2 muteins for treatment of multiple myeloma JF - PLoS ONE N2 - Multiple myeloma (MM) represents a haematological cancer characterized by the pathological hyper proliferation of antibody-producing B-lymphocytes. Patients typically suffer from kidney malfunction and skeletal disorders. In the context of MM, the transforming growth factor β (TGFβ) member Activin A was recently identified as a promoter of both accompanying symptoms. Because studies have shown that bone morphogenetic protein (BMP)-2-mediated activities are counteracted by Activin A, we analysed whether BMP2, which also binds to the Activin A receptors ActRII and ActRIIB but activates the alternative SMAD-1/5/8 pathway, can be used to antagonize Activin A activities, such as in the context of MM. Therefore three BMP2 derivatives were generated with modified binding activities for the type II (ActRIIB) and/or type I receptor (BMPRIA) showing either increased or decreased BMP2 activity. In the context of MM these BMP2 muteins show two functionalities since they act as a) an anti-proliferative/apoptotic agent against neoplastic B-cells, b) as a bone-formation promoting growth factor. The molecular basis of both activities was shown in two different cellular models to clearly rely on the properties of the investigated BMP2 muteins to compete for the binding of Activin A to the Activin type II receptors. The experimental outcome suggests new therapeutic strategies using BMP2 variants in the treatment of MM-related pathologies. KW - multiple myeloma KW - signaling KW - cell proliferation KW - cell binding KW - membrane receptor signaling KW - BMP KW - gene expression KW - B cell receptors KW - B cells Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-158144 VL - 12 IS - 5 ER - TY - JOUR A1 - Schmid, Tobias A1 - Falter, Lena A1 - Weber, Sabine A1 - Müller, Nils A1 - Molitor, Konstantin A1 - Zeller, David A1 - Weber-Steffens, Dorothea A1 - Hehlgans, Thomas A1 - Wajant, Harald A1 - Mostböck, Sven A1 - Männel, Daniela N. T1 - Chronic inflammation increases the sensitivity of mouse Treg for TNFR2 costimulation JF - Frontiers in Immunology N2 - TNF receptor type 2 (TNFR2) has gained attention as a costimulatory receptor for T cells and as critical factor for the development of regulatory T cells (Treg) and myeloid suppressor cells. Using the TNFR2-specific agonist TNCscTNF80, direct effects of TNFR2 activation on myeloid cells and T cells were investigated in mice. \(In\) \(vitro\), TNCscTNF80 induced T cell proliferation in a costimulatory fashion, and also supported \(in\) \(vitro\) expansion of Treg cells. In addition, activation of TNFR2 retarded differentiation of bone marrow-derived immature myeloid cells in culture and reduced their suppressor function. \(In\) \(vivo\) application of TNCscTNF80-induced mild myelopoiesis in naïve mice without affecting the immune cell composition. Already a single application expanded Treg cells and improved suppression of CD4 T cells in mice with chronic inflammation. By contrast, multiple applications of the TNFR2 agonist were required to expand Treg cells in naïve mice. Improved suppression of T cell proliferation depended on expression of TNFR2 by T cells in mice repeatedly treated with TNCscTNF80, without a major contribution of TNFR2 on myeloid cells. Thus, TNFR2 activation on T cells in naïve mice can lead to immune suppression \(in\) \(vivo\). These findings support the important role of TNFR2 for Treg cells in immune regulation. KW - molecular medicine KW - inflammation KW - immune regulation KW - costimulation KW - MDSC KW - TNFR2 KW - regulatory T cell Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-173259 VL - 8 ER - TY - THES A1 - Schenkhoff, Felix Stephan T1 - Herstellung und Charakterisierung multifunktioneller Fusionsproteine des membranständigen Fas-Liganden und des membranständigen CD40-Liganden T1 - Development and Characterization of multifunctional fusionproteins of the membranous Fas-ligand and of the membranous CD40-ligand N2 - TNF-Liganden liegen primär in membranständiger Form mit trimerer Struktur vor und die meisten von ihnen können sekundär durch Metalloproteasen in lösliche trimere Liganden prozessiert werden. Während membranständige Formen der TNF-Liganden ihre korrespondierenden Rezeptoren aktivieren können, sind die löslichen Varianten einzelner TNF-Liganden unterschiedlich aktiv beziehungsweise inaktiv an den korrespondierenden Rezeptoren, obwohl sie ebenfalls zur Bindung in der Lage sind. Dies konnte bereits in Studien für TNF und TRAIL gezeigt werden. Die Unterschiede zwischen löslichen Varianten und der membranständigen Form des Liganden betreffen sowohl die Rezeptorselektivität als auch den Aktivierungsgrad am Rezeptor bis hin zu völliger Inaktivität der löslichen Form. Unterschiede finden sich jedoch nicht nur hinsichtlich der Aktivität, sondern auch in der Interaktion des Liganden mit dem Rezeptor. Für die membranständige und die lösliche Form von FasL konnten Unterschiede in der Notwendigkeit intrazellulärer Signalmoleküle bei der Ausbildung Ligand-induzierter Rezeptorsignalcluster gezeigt werden. Für die lösliche und membranständige Form des CD40L werden eine unterschiedliche Rezeptorinternalisierung und eine unterschiedliche Rekrutierung von TRAF-Molekülen angenommen. Bisherige Arbeiten zur Untersuchung der Rezeptor-Ligand-Interaktion stützen sich meist auf lösliche Varianten von TNF-Liganden, die durch artifizielle Multimerisierung oder Antikörper-induzierte Quervernetzung sekundär aktiviert werden müssen. Um für die Untersuchung der Rezeptor-Ligand-Interaktion in Zukunft realitätsnähere Bedingungen zu schaffen, sollten im Rahmen dieser Arbeit multifunktionelle Fusionsproteine des membranständigen FasL und CD40L hergestellt und charakterisiert werden. Die Fusionsproteine wurden im Rahmen der Klonierung so konstruiert, dass von der aminoterminalen Seite beginnend eine GST-Domäne, ein Flag-Tag, ein YFP-Tag und abschließend die vollständige membranständige Form des Liganden (FasL oder CD40L) aneinander gefügt wurden. In FACS-Analysen konnte sowohl die Funktion des YFP-Tag als auch die korrekte Expression des Liganden an der Zelloberfläche durch spezifische Antikörperfärbung nachgewiesen werden. Die funktionelle Aktivität der Liganden wurde durch IL-8-Induktion gezeigt, die eine Aktivierung des NFB-Signalweges durch die GST-Fusionsproteine des membranständigen FasL und des membranständigen CD40L beweist. Im Rahmen von Immunopräzipitationen wurde die Möglichkeit der Detektion der Fusionsproteine über ihr Flag-Tag getestet. Für das in GST-pull-down-Assays genauer untersuchte membranständige GST-Flag-YFP-CD40L-Fusionsprotein gelang eine Koimmunopräzipitation mit dem im Rezeptorkomplex gebundenen TRAF2. Für dieses in der Signaltransduktion des CD40 entscheidende Molekül konnte seine transiente Interaktion mit dem Rezeptorsignalkomplex sowie eine Veränderung in der Assoziation an den Rezeptor durch TNF-abhängige TRAF1-Induktion gezeigt werden. Im Zusammenhang der mit der Rezeptoraktivierung oftmals gleichgesetzten Bildung von Rezeptorsignalclustern durch den entsprechenden TNF-Liganden war die Beobachtung interessant, dass das Konstrukt GST-Flag-YFP-CD40L im Gegensatz zum analog konstruierten GST-Flag-YFP-FasL nicht in der Lage war, Signalcluster des korrespondierenden Rezeptors zu erzeugen, aber dennoch fähig war, eine Rezeptoraktivierung zu bewirken. Hinsichtlich der Untersuchung von Unterschieden in der Rezeptor-Ligand-Interaktion von löslichen und membranständigen Formen sind für FasL und CD40L noch viele Fragen offen, die beispielsweise auch die Stabilität von Rezeptorsignalclustern betreffen. Die in dieser Arbeit erzeugten und charakterisierten multifunktionellen Fusionsproteine sollten helfen, neue Erkenntnisse bezüglich der molekularen Grundlagen der Rezeptor-Ligand-Interaktion zu erzielen. N2 - TNF ligands primary are membranous ligands with a trimeric structure and the most of them can be cleaved to soluble trimeric forms by metalloproteases secondarily. While membranous forms of TNF ligands can activate their corresponding receptors, soluble variants ot some TNF ligands are insufficient for full activity or totally inactive. Nevertheless these variants are able to bind the corresponding receptor properly. This already could be shown in experiments with TNF and TRAIL. The differences between soluble variants and the membranous form of TNF ligands concern their receptor selectivity as well as their degree of activity at the receptor. There also are differences regarding the interaction itself between ligand and receptor. Comparing the membranous and the soluble form of Fas ligand there could be shown differences in terms of dependency for intracellular signaling moleculs regarding the formation of ligand induced clustering of the receptor. The soluble and the membranous form of CD40 ligand are supposed to have differences in receptor internalization and recruting of TRAF molecules. Previous research concentrating on receptor ligand interactions often is based on soluble variants of TNF ligands that have to be activated by artificial multimerization or antibody dependent aggregation. To provide more realistic conditions for the analysis of receptor ligand interactions, multifunctional fusionproteins of the membranous Fas ligand and the membranous CD40 ligand should be developed and characterized in this dissertation. Starting from the N-terminal end the fusionproteins contain a GST-domain, a Flag-tag, a YFP-tag and the complete membranous form of the ligand. In FACS analysis the activity of the YFP-tag as well as the correct expression of the ligand on the cell membrane could be demonstrated by specific antibody staining. The functional activity of the ligand itself has been shown by IL-8-induction, that proves the sucessful activation of the NFB signaling pathway by GST-fusionproteins of the membranous Fas ligand and the membranous CD40 ligand. In the context of immunoprecipitations the possibility of detection of the fusionproteins by Flag-tag has been tested. Concerning the GST-pull-down-assays of the fusionprotein GST-Flag-YFP-CD40L a coimmunoprecipitation with the intracellular adaptor protein TRAF2 could be shown. A transient interaction of TRAF2 with the receptor signaling complex as well as a change of its association with the receptor by TNF-induced TRAF1 upregulation also could be demonstrated. In context with the formation of high molecular receptor clusters that are often equated with receptor activation the observation was interesting that GST-Flag-YFP-CD40L in contrast to the analogous constructed GST-Flag-YFP-FasL could not induce signalling cluster of the corresponding receptor, but still was able to induce receptor activation. Regarding the analysis of differences in receptor ligand interaction between soluble and membranous forms of Fas ligand and CD40 ligand there are still many questions that have to be answered, e.g. in terms of stability of induced receptor clustering. The multifunctional fusionproteins provided by this dissertation shall contribute to gain new findings in respect of molecular fundamentals of the receptor ligand interaction. KW - Tumor-Necrose-Faktor KW - Entzündung KW - Apoptose KW - TNF-Superfamilie KW - CD40-Ligand KW - Fas-Ligand KW - Rezeptorsignalkomplexe KW - TNF superfamily KW - CD40L KW - FasL KW - receptor cluster Y1 - 2007 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-25978 SN - xxx ER - TY - JOUR A1 - Schanbacher, Constanze A1 - Hermanns, Heike M. A1 - Lorenz, Kristina A1 - Wajant, Harald A1 - Lang, Isabell T1 - Complement 1q/tumor necrosis factor-related proteins (CTRPs): structure, receptors and signaling JF - Biomedicines N2 - Adiponectin and the other 15 members of the complement 1q (C1q)/tumor necrosis factor (TNF)-related protein (CTRP) family are secreted proteins composed of an N-terminal variable domain followed by a stalk region and a characteristic C-terminal trimerizing globular C1q (gC1q) domain originally identified in the subunits of the complement protein C1q. We performed a basic PubMed literature search for articles mentioning the various CTRPs or their receptors in the abstract or title. In this narrative review, we briefly summarize the biology of CTRPs and focus then on the structure, receptors and major signaling pathways of CTRPs. Analyses of CTRP knockout mice and CTRP transgenic mice gave overwhelming evidence for the relevance of the anti-inflammatory and insulin-sensitizing effects of CTRPs in autoimmune diseases, obesity, atherosclerosis and cardiac dysfunction. CTRPs form homo- and heterotypic trimers and oligomers which can have different activities. The receptors of some CTRPs are unknown and some receptors are redundantly targeted by several CTRPs. The way in which CTRPs activate their receptors to trigger downstream signaling pathways is largely unknown. CTRPs and their receptors are considered as promising therapeutic targets but their translational usage is still hampered by the limited knowledge of CTRP redundancy and CTRP signal transduction. KW - adiponectin KW - AMPK KW - C1q/TNF related protein (CTRP) KW - inflammation KW - metabolism Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-304136 SN - 2227-9059 VL - 11 IS - 2 ER - TY - THES A1 - Schaffstein, Stella T1 - Molekulare Mechanismen der nicht-apoptotischen Signaltransduktion des Todesliganden TRAIL (Apo-2 Ligand) T1 - Molecular mechanisms of non-apoptotic signaling of the death-ligand TRAIL (Apo-2 ligand) N2 - TRAIL (tumor necrosis factor-related apoptosis-inducing ligand)/Apo-2 Ligand ist ein Mitglied der TNF (tumor necrosis factor)-Superfamilie, das in den vergangenen Jahren als potentielles Tumortherapeutikum breite Aufmerksamkeit auf sich gezogen hat. Denn über seine korrespondierenden Todesrezeptoren induziert TRAIL vornehmlich in Tumorzellen den apoptotischen Zelltod, während normale Zellen unbeschadet bleiben (Ashkenazi et al., 1999; Walczak et al., 1999; Griffith et al., 1998). Neuere Studien belegen allerdings, dass die TRAIL-Todesrezeptoren neben ihrer herausragenden Funktion als Auslöser der Apoptose zusätzlich die Fähigkeit zur Aktivierung nicht-apoptotischer Signalwege besitzen. In der vorliegenden Arbeit wurden vornehmlich die nicht-apoptotischen Signalwege wie die MAPK-Kaskaden sowie die NFkappaB-Signalwege in Verbindung mit der Aktivierung von Apoptose sowie der Induktion des Chemokins IL-8 analysiert. Hierfür wurde die humane Pankreasadenokarzinomzellinie Colo 357 verwendet. In den Experimenten konnte nachgewiesen werden, dass TRAIL die MAP Kinasen JNK, ERK und p38 in Colo 357 Zellen induziert. Die Induktion erfolgte hierbei unabhängig vom apoptotischen Zelltod aber abhängig von der Aktivierung der Caspasen. Desweiteren konnte eine TRAIL-vermittelte Aktivierung des Transkriptionsfaktors NFkappaB in Colo 357 Zellen demonstriert werden. Anhand von ELISA-Experimenten wurde gezeigt, dass sowohl die Aktivierung der MAP Kinasen als auch die Aktivierung von NFkappaB eine essentielle Rolle bei der TRAIL-vermittelten Induktion von IL-8 spielen. Durch die Induktion von IL-8 wiederum kann TRAIL inflammatorische Effekte induzieren. Im Hinblick auf eine potentielle Tumortherapie mit TRAIL legen die Daten dieser Studie die Notwendigkeit von Kombinationstherapien mit TRAIL nahe. So kann durch die Kombination von TRAIL mit anti-inflammatorisch wirkenden Medikamenten eine Reduktion entzündlicher Nebenwirkungen erzielt werden. Andererseits kann durch Verwendung von TRAIL zusammen mit Proteasom-Inhibitoren die Resistenz gegenüber TRAIL-vermittelter Apoptose vermindert werden und gleichzeitig eine anti-inflammatorische und NFkappaB-hemmende Wirkung erzielt werden. N2 - TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), also known as Apo-2 ligand, is one of several members of the TNF (tumor necrosis factor) superfamily that induce apoptosis through engagement of death receptors (Wiley et al., 1995). This protein has generated tremendous excitement as a potential tumor-specific cancer therapeutic because it selectively induces apoptosis in many transformed cells but not in normal cells (Ashkenazi et al., 1999; Walczak et al., 1999; Griffith et al., 1998). Since its discovery in 1995, TRAIL has been predominantly described as a potent inducer of apoptosis with functions in tumor surveillance and immune privilege through binding of its corresponding death receptors. More recent studies however point to additional, apoptosis-independent functions of the TRAIL-death-receptors. The experiments described in this study focused mainly on TRAIL-mediated activation of the non-apoptotic signaling pathways as the MAPK (mitogen-activated protein kinase) cascades and the transcriptional factor NFkappaB in connection with the induction of apoptotic cell death as well as the induction of the chemokine IL-8. For the described project the human pancreatic cancer cell line Colo 357 was predominantely used. The experiments demonstrated that TRAIL induces the activation of the MAPK cascades JNK, ERK and p38 in Colo 357 cells. This induction occured independently of the induction of apoptotic cell death but dependently of the activation of caspases. Furthermore a TRAIL-mediated activation of the transcriptional factor NFkappaB was demonstrated in Colo 357 cells. Both the activation of the MAP kinase JNK and the induction of NFkappaB were shown to play a decisive role in the TRAIL-induced expression of the chemokine IL-8. In correspondance to potential tumor-specific cancer therapy with TRAIL the data of this study suggest the use of combintion therapies of TRAIL, on the one hand combination with anti-inflammatory drugs to avoid side effects arising from the proinflammatory potential of TRAIL and on the other hand combintaion with drugs to overcome resistance against TRAIL-mediated apoptosis such as proteasomal inhibitors which were shown to not only reinforce TRAIL-mediated apoptosis but also have anti-inflammatory and NFkappaB inhibitory effects. KW - Apoptosis KW - TRAIL KW - Apoptose KW - c-Jun N-terminale Kinase KW - NFkappaB KW - Interleukin 8 KW - TRAIL KW - apoptosis KW - c-Jun N-terminal kinase KW - NFkappaB KW - interleukine 8 Y1 - 2010 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-55943 ER - TY - THES A1 - Salzmann, Steffen T1 - Regulation der TNF-Rezeptor Signaltransduktion durch das Zytokin TWEAK T1 - Regulation of the TNF-receptor signaltransduction through the cytokine TWEAK N2 - Das pleiotrope Zytokin TNF (tumor necrosis factor) kann an den TNF-Rezeptor 1 (TNFR1) und den TNF-Rezeptor 2 (TNFR2) binden und mit deren Hilfe seine biologischen Funktionen über verschiedene Signalwege, wie z.B. NFB- und MAPK-Aktivierung bzw. Apop¬toseinduktion, vermitteln. In früheren Arbeiten konnte gezeigt werden, dass die Aktivierung des TNFR2 zur proteasomalen Degradation des Adaterproteins TRAF2 führt und dadurch die TNFR1-induzierte Apoptose verstärkt wird. TWEAK (tumor necrosis like weak inducer of apoptosis), das ebenfalls der TNF-Ligandenfamilie angehört und die Interaktion mit dessen Rezeptor Fn14 (fibroblast growth factor-inducible 14), der wie der TNFR2 zur Untergruppe der TRAF-bindenden Rezeptoren der TNF-Rezeptorfamilie gehört, zeigten in verschiedenen Arbeiten auch eine TRAF2-degradierende Wirkung. In der vorliegenden Arbeit konnte nun gezeigt werden, dass dies auch im Falle des TWEAK/Fn14-Systems mit einem verstärkenden Effekt auf die TNFR1-vermittelte Apoptose einhergeht. Darüber hinaus konnte gezeigt werden, dass TWEAK zusätzlich auch die TNFR1-induzierte Nekrose verstärkt, die den Zelltod durch andere Mechanismen als bei der Apoptose induziert. Von anderen Arbeiten unserer Gruppe war bekannt, dass lösliches TWEAK (sTWEAK) und membranständiges TWEAK (mTWEAK) bezüglich der TRAF2-Depletion wirkungs¬gleich sind. Da der apoptotische Fn14-TNFR1-„crosstalk“ auf der Depletion von TRAF2-Komplexen beruht wurden auch keine signifikanten Unterschiede zwischen sTWEAK und mTWEAK in Bezug auf die Verstärkung der TNFR1-induzierten Apoptose beobachtet. Interessanter¬weise zeigte sich in der vorliegenden Arbeit jedoch, dass sTWEAK den klassischen NFB-Signalweg gar nicht bzw. nur schwach aktiviert, wohingegen mTWEAK diesen stark induziert. Bei der Aktivierung des alternativen NFB-Signalweges hingegen ließen sich keine Unterschiede zwischen sTWEAK und mTWEAK erkennen. Die Aktivierung eines Signalweges wird also durch die Oligomerisierung des Liganden nicht moduliert, demgegenüber aber erwies sich die Aktivierung eines anderen Signalweges als stark abhängig von der Liganden-Oligomerisierung. Vor dem Hintergrund, dass das Adapterprotein TRAF1 (TNF-receptor-associated factor 1) Heterotrimere mit TRAF2 bildet, wurde weiterhin untersucht, ob dieses Molekül einen Einfluss auf die Aktivität der TWEAK-induzierten Signalwege hat. Tatsächlich zeigte sich in TRAF1-exprimie¬renden Zellen eine Verstärkung der TWEAK-induzierten Aktivierung des klassischen NFB-Signalweges Zukünftige Studien müssen nun aufklären, inwieweit die hier gefundenen Mecha-nismen das Zusammenspiel von TNF und TWEAK in vivo bestimmen. N2 - The pleiotropic cytokine TNF (tumor necrosis factor) binds to two receptors, TNF-receptor 1 (TNFR1) and TNF-receptor 2 (TNFR2). TNF elicits its biological functions through different signaling pathways, e.g. NFB- and MAPK-activation and induction of apoptosis. Previous studies revealed that activation of TNFR2 leads to protea¬somal degradation of the adaptor protein TRAF2 and enhanced TNFR1-induced apoptosis. TWEAK (tumor necrosis like weak inducer of apoptosis), a member of the TNF-ligand family and Fn14 which belongs like TNFR2 to the TRAF-binding subgroup of the TNF-receptor family, showed also a TRAF2-degrading effect. In the present thesis it is shown that this is also associated with an enhancement of TNFR1-mediated apoptosis. Furthermore, it was shown that TWEAK also amplifies TNFR1-induced necrosis, a form of cell death that is induced independently and by different mechanisms than apoptosis. In earlier studies in our group it was found that soluble TWEAK (sTWEAK) and membrane bound TWEAK (mTWEAK) have the same TRAF2-depleting effect. Because the apoptotic crosstalk of Fn14 and TNFR1 bases on the depletion of TRAF2-containing complexes, there were no significant differences observed between sTWEAK and mTWEAK concerning the enhancement of TNFR1-induced apoptosis. Interestingly, it was shown in this thesis that sTWEAK can activate the classical NFB pathway not or just weak, whereas mTWEAK can do this very efficiently. However there were no differences between sTWEAK and mTWEAK in the activa¬tion of the alternative NFB pathway. Thus the activation of one type of signaling pathway is therefore not modulated through ligand oligomerisation, but in contrast the activation of another pathway turned out to be strongly dependent of ligand oligomerisation. It is known that the adaptor protein TRAF1 (TNF-receptor-associated factor 1) builds heterotrimers with TRAF2. It was furthermore investiga-ted, whether this molecule has an influence on the activity of TWEAK-induced pathways. Actually there was increased activity in TWEAK-mediated classical NFB signaling observed in TRAF1-expressing cells. Further studies have to clarify to which extend the here found mechanisms affect the TNF-TWEAK interplay in vivo. KW - Tumor-Nekrose-Faktor KW - Signaltransduktion KW - TNF KW - TWEAK Y1 - 2010 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-52525 ER - TY - JOUR A1 - Rödel, Mark-Oliver A1 - Brede, Christian A1 - Hirschfeld, Mareike A1 - Schmitt, Thomas A1 - Favreau, Philippe A1 - Stöcklin, Reto A1 - Wunder, Cora A1 - Mebs, Dietrich T1 - Chemical Camouflage - A Frog's Strategy to Co-Exist with Aggressive Ants JF - PLOS ONE N2 - Whereas interspecific associations receive considerable attention in evolutionary, behavioural and ecological literature, the proximate bases for these associations are usually unknown. This in particular applies to associations between vertebrates with invertebrates. The West-African savanna frog Phrynomantis microps lives in the underground nest of ponerine ants (Paltothyreus tarsatus). The ants usually react highly aggressively when disturbed by fiercely stinging, but the frog is not attacked and lives unharmed among the ants. Herein we examined the proximate mechanisms for this unusual association. Experiments with termites and mealworms covered with the skin secretion of the frog revealed that specific chemical compounds seem to prevent the ants from stinging. By HPLC-fractionation of an aqueous solution of the frogs' skin secretion, two peptides of 1,029 and 1,143 Da were isolated and found to inhibit the aggressive behaviour of the ants. By de novo sequencing using tandem mass spectrometry, the amino acid sequence of both peptides consisting of a chain of 9 and 11 residues, respectively, was elucidated. Both peptides were synthesized and tested, and exhibited the same inhibitory properties as the original frog secretions. These novel peptides most likely act as an appeasement allomone and may serve as models for taming insect aggression. KW - amphibian skin secretions KW - antimicrobial peptides KW - paltothyreus tarsatus KW - dendrobates pumilio KW - anurans KW - microhylidae KW - hymenoptera KW - formicidae KW - mutualisms KW - alkaloids Y1 - 2013 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-128181 SN - 1932-6203 VL - 8 IS - 12 ER - TY - THES A1 - Rumpf, Jost-Julian T1 - Mechanismen der TRAIL-induzierten Signaltransduktion T1 - Mechanisms of TRAIL-induced cell signaling N2 - TRAIL (TNF Related Apoptosis Inducing Ligand), ein Mitglied der TNF-Ligandenfamilie, wurde bislang hauptsächlich hinsichtlich seiner dominanten Funktion als Auslöser des apoptotischen Programms untersucht. In neueren Untersuchungen konnte allerdings gezeigt werden, dass TRAIL unter bestimmten Bedingungen auch eine starke Aktivierung nicht-apoptotischer Signalwege induzieren kann. Um die Mechanismen der TRAIL-induzierten nicht-apoptotischen Signaltransduktion genauer zu untersuchen, wurde in der hier vorliegenden Arbeit besonderes Augenmerk auf die TRAIL-vermittelte Aktivierung des Transkriptionsfaktors NFkB und deren Modulation durch Interferon gamma und FLIP gelegt. Hierbei konnte gezeigt werden, dass Interferon gamma, neben einer synergistischen Wirkung hinsichtlich der TRAIL-induzierten Apoptose, unter nicht-apoptotischen Bedingungen, die durch Caspase-Inhibition oder Bcl2-Überexpression geschaffen wurden, auch eine verstärkende Wirkung auf die TRAIL-induzierten NFkB-Aktivierung in KB-Zellen entfaltet. Weiterhin konnte gezeigt werden, dass FLIP, ein Inhibitor der Caspase-8-Aktivierung, dessen Expression unter anderem durch Interferon gamma reguliert wird, neben einer Apoptose-inhibierenden Wirkung auch die TRAIL-induzierte NFkB-Aktivierung in KB-Zellen inhibiert, was auf eine gemeinsame Regulation beider Mechanismen auf der Ebene des DISC (Death Inducin Signaling Complex) hindeutet. N2 - TRAIL (TNF Related Apoptosis Inducing Ligand) is a member of the TNF superfamily and is primarily known for its strong apoptosis inducing capability. However, it could be shown that besides its strong apoptotic potential, TRAIL can also induce non-apoptotic signaling pathways under certain conditions. In this dissertation it is shown that interferon gamma synergistically enhances the TRAIL induced activation of NFkB under non-apoptotic conditions, which were achieved by inhibition of caspases or stable expression of Bcl2 in KB-cells. Furthermore, FLIP, an inhibitor of caspase-8-activation, which is amongst others regulated by interferon gamma, is shown not only to block TRAIL-induced apoptosis but also to inhibit TRAIL-induced NFkB-activation in KB-cells, indicating that both mechanisms are coregulated at the level of the DISC (Death Inducing Signaling Complex). KW - Interferon KW - Apoptosis KW - Tumor-Nekrose-Faktor KW - Entzündung KW - TRAIL KW - NFkB KW - cFLIP KW - Todesrezeptor KW - TRAIL KW - NFkB KW - cFLIP KW - Death Receptor Y1 - 2007 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-27112 ER - TY - THES A1 - Roos, Claudia T1 - Characterization of tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-induced signaling pathways T1 - Charakterisierung TWEAK induzierter Signalwege N2 - TWEAK ist ein typischer Vertreter der TNF Ligandenfamilie. TWEAK wird als Typ II Transmembranprotein exprimiert, kann jedoch durch proteolytische Prozessierung auch als lösliches Protein freigesetzt werden. In dieser Arbeit wird gezeigt, dass oligomerisiertes TWEAK in Hinblick auf die Aktivierung des klassischen NFκB Signalweges deutlich aktiver ist als lösliches, trimeres TWEAK. Jedoch sind beide TWEAK-Varianten in der Lage, die Depletion von TRAF2 und die Prozessierung von p100, beides Kennzeichen für die Aktivierung des alternativen NFκB Signalweges, zu induzieren. Ebenso wie andere lösliche TNF-Liganden, die ihren entsprechenden Rezeptor nur schwach aktivieren, erlangt lösliches TWEAK durch Oligomerisierung vergleichbare Aktivität zum membrangebundenen Liganden. TRAF2 spielt eine Schlüsselrolle in der TWEAK-vermittelten NFκB Aktivierung. Durch Depletion oder Degradation von TRAF2 fällt die Entscheidung, ob lediglich der alternative oder beide, der klassische und der alternative NFκB Signalweg aktiviert werden. Die Blockade des TWEAK-Rezeptors Fn14 inhibiert die Aktivierung der NFκB Signalwege, ungeachtet welche Form von TWEAK zur Stimulation genutzt wird. Das weist darauf hin, dass die unterschiedlichen Aktivitäten der beiden TWEAK-Varianten in der Induktion des klassischen und alternativen NFκB Signalweges nicht durch die Nutzung verschiedener Rezeptoren verursacht sind. Damit wird in dieser Arbeit anhand von TWEAK zum ersten mal gezeigt, dass ein TNF Ligand in unterschiedlichen Varianten qualitativ unterschiedliche Aktivitäten des entsprechenden TNF Rezeptors auslöst. N2 - TWEAK is a typical member oft he TNF ligand family. Therefore it is initially expressed as a type II transmembrane protein, but a soluble variant can be released by proteolytic processing. In this work it is shown that oligomerized TWEAK is more competent than soluble, trimeric TWEAK regarding the activation of classical NFκB signaling pathway. However, both TWEAK variants are able to induce depletion of TRAF2 and processing of p100, which are hallmarks for the activation of the noncanonical NFκB pathway. Like other solube TNF ligands with no or poor activity on their corresponding receptor, TWEAK gains high activity upon oligomerization resembling the activity of the transmembrane ligand. TRAF2 has a key role in TWEAK-induced NFκB signaling. Depletion or degradation of TRAF2 is crucial for activation of the noncanonial or both, the classical and the noncanonical NFκB pathway. Blocking the TWEAK receptor Fn14 inhibits the activation of NFκB signaling, irrespective of the TWEAK form used for stimulation. This indicates that the different activities of the two TWEAK variants in activation of classical and noncanonical NFκB signaling are not caused by the use of different receptors. Therefore this study on TWEAK is the first reported case where one TNF ligand in different variants induces qualitatively different activities of the corresponding TNF receptor. KW - Tumor-Nekrose-Faktor KW - Signaltransduktion KW - Fn14 KW - NFkappaB KW - p100 KW - TRAF2 KW - TWEAK KW - Fn14 KW - NFkappaB KW - p100 KW - TRAF2 KW - TWEAK Y1 - 2009 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-45295 ER - TY - JOUR A1 - Rauert-Wunderlich, Hilka A1 - Siegmund, Daniela A1 - Maier, Eduard A1 - Giner, Tina A1 - Bargou, Ralf C. A1 - Wajant, Harald A1 - Stühmer, Thorsten T1 - The IKK Inhibitor Bay 11-7082 Induces Cell Death Independent from Inhibition of Activation of NF kappa B Transcription Factors JF - PLoS ONE N2 - Multiple myeloma (MM) displays an NFκB activity-related gene expression signature and about 20% of primary MM samples harbor genetic alterations conducive to intrinsic NFκB signaling activation. The relevance of blocking the classical versus the alternative NFκB signaling pathway and the molecular execution mechanisms involved, however, are still poorly understood. Here, we comparatively tested NFκB activity abrogation through TPCA-1 (an IKK2 inhibitor), BAY 11-7082 (an IKK inhibitor poorly selective for IKK1 and IKK2), and MLN4924 (an NEDD8 activating enzyme (NAE)-inhibitor), and analyzed their anti-MM activity. Whereas TPCA-1 interfered selectively with activation of the classical NFκB pathway, the other two compounds inhibited classical and alternative NFκB signaling without significant discrimination. Noteworthy, whereas TPCA-1 and MLN4924 elicited rather mild anti-MM effects with slight to moderate cell death induction after 1 day BAY 11-7082 was uniformly highly toxic to MM cell lines and primary MM cells. Treatment with BAY 11-7082 induced rapid cell swelling and its initial effects were blocked by necrostatin-1 or the ROS scavenger BHA, but a lasting protective effect was not achieved even with additional blockade of caspases. Because MLN4924 inhibits the alternative NFκB pathway downstream of IKK1 at the level of p100 processing, the quite discordant effects between MLN4924 and BAY 11-7082 must thus be due to blockade of IKK1-mediated NFκB-independent necrosis-inhibitory functions or represent an off-target effect of BAY 11-7082. In accordance with the latter, we further observed that concomitant knockdown of IKK1 and IKK2 did not have any major short-term adverse effect on the viability of MM cells. KW - signal inhibition KW - necrotic cell death KW - cell viability testing KW - cell death KW - small interfering RNAs KW - HT29 cells KW - phosphorylation KW - multiple myeloma Y1 - 2013 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-130140 VL - 8 IS - 3 ER - TY - THES A1 - Rauert-Wunderlich, Hilka T1 - Apoptoseregulation durch TNF im Multiplen Myelom T1 - Regulation of apoptosis via TNF in multiple myeloma N2 - Der Tumornekrosefaktor (TNF) entfaltet seine vielfältigen biologischen Aktivitäten durch die Stimulation der beiden TNF-Rezeptoren TNFR1 und TNFR2. Die TNFR1-vermittelte Signaltransduktion ist in vielen Details gut verstanden, wohingegen die TNFR2-vermittelte Signaltransduktion bis heute kaum untersucht ist. Mit Hilfe einer in unserer Gruppe entwickelten hochaktiven TNFR2-spezifischen TNF-Variante sowie einer bereits länger bekannten TNFR1-spezifischen TNF-Variante wurde in dieser Arbeit die TNF-Signaltransduktion insbesondere im Mutiplen Myelom untersucht. Mit Hilfe der beiden TNF-Varianten konnte gezeigt werden, dass die alleinige Stimulation des TNFR2 die Aktivierung des alternativen NFkappaB-Signalweges vermittelt, wohingegen TNFR1 nicht dazu in der Lage ist. So zeigte sich im Einklang mit der inhibitorischen Funktion des Adapterproteins TRAF2 in der Signaltransduktion des alternativen NFkappaB-Signalweges, dass die TNFR2-Stimulation in einer TRAF2-Depletion resultiert. Dies führt weiterhin zur Akkumulation von NIK und der Prozessierung von p100 zu seiner aktiven Form p52, den klassischen biochemisch nachweisbaren Ereignissen der Aktivierung des alternativen NFkappaB-Signalweges. Aufgrund der Rolle des NFkappaB-Systems im Multiplen Myelom (MM) und der stimulierenden Wirkung des TNFR1 und TNFR2 auf das NFkappaB-System wurde die Expression und Funktion dieser beiden Rezeptoren auf Myelomzelllinien untersucht. Insbesondere wurde analysiert, welchen Effekt eine spezifische Stimulation der beiden TNF-Rezeptoren auf die apoptotische Sensitivität von Myelomzellen hat. Mit einer Ausnahme wiesen alle untersuchten Myelomzelllinien eine eindeutige TNFR2-Oberflächenexpression auf, die TNFR1-Expression hingegen war heterogen. Die TNFR1-Stimulation in den TNFR1-positiven Zelllinien zeigte keinen wesentlichen Einfluss auf die Zellviabilität. Allerdings resultierte eine Vorstimulation mit TNF in einer gesteigerten Sensitivität für den CD95L-induzierten Zelltod, schützte aber gleichzeitig vor der TRAIL-vermittelten Induktion der Apoptose. Der gegenläufige Effekt der TNF-Vorstimulation auf den CD95L- und TRAIL-induzierten Zelltod konnte auf die Hochregulation der CD95-Oberflächenexpression und der gesteigerten Expression des antiapoptotischen cFLIPLong-Proteins zurückgeführt werden. Beide Effekte basieren auf der TNF-induzierten Aktivierung des klassischen NFkappaB-Signalweges. Im CD95L-induzierten Zelltod überkompensierte die Induktion der CD95-Expression offensichtlich die Hochregulation von cFLIPLong und resultierte in gesteigertem Zelltod. Der TRAIL-induzierte Zelltod hingegen wurde durch die TNF-Vorstimulation abgeschwächt, da hier lediglich die durch den klassischen NFkappaB-Signalweg vermittelte gesteigerte Expression des antiapoptotischen cFLIPLong eine Rolle spielte. Desweiteren zeigten die Analysen in dieser Arbeit, dass die TNFR2-Stimulation zu einer Depletion von TRAF2 und z. B. in JJN3-Zellen zu einer Sensitivierung für den TNFR1-induzierten Zelltod führte. Die Ergebnisse dieser Arbeit zeigten in der Summe somit, dass das TNF-TNFR-Signaling durch verschiedene Mechanismen Einfluss auf den Ausgang der extrinsischen Apoptoseinduktion hat, und dass der Effekt von TNF auf das Überleben von MM-Zellen kontextabhängig ist. N2 - TNF mediates its biological functions by stimulation of the two TNF receptors TNFR1 and TNFR2. TNFR1-mediated signaling has already been studied in detail, whereas TNFR2-mediated signal transduction is poorly understood. In this work a newly developed TNFR2-specific variant and an established TNFR1-specific variant was used to study TNF signaling especially in myeloma cells. With the help of these TNF-variants it is shown here that TNFR2, but not TNFR1, induces activation of the alternative NFkappaB-pathway. Thus in consent with the inhibitory function of TRAF2 in alternative NFkappaB signal transduction, stimulation of TNFR2 resulted in depletion of TRAF2, accumulation of NIK and p100 processing to p52, the biochemical hallmarks of this pathway. Due to the relevance of the NFkappaB-system for multiple myeloma (MM) and the NFkappaB stimulatory activities of TNFR1 and TNFR2, the expression of these two receptors and their effect on apoptotic sensitivity was analyzed in myeloma cell lines. A huge majority of myeloma cell lines express TNFR2 whereas TNFR1 expression is rather restricted. Stimulation of TNFR1 in the TNFR1-positive subset of MM cell lines showed nearly no impact on cellular viability. However, TNF stimulation enhanced CD95L-induced cell death and in parallel reduced the TRAIL-mediated induction of apoptosis. This opposed regulation of TRAIL- and CD95L-induced cell death by TNF based on upregulation of the death receptor CD95 via the classical NFkappaB-pathway and by upregulation of the antiapoptotic protein cFLIPLong via the same pathway. The induction of CD95 expression appeared to overcompensate the upregulation of cFLIPLong and consequently TNF-induced NFkappaB activation resulted, in context of CD95 signaling, in apoptosis enhancement. TRAIL-mediated cell death induction, however, was reduced after TNF prestimulation, due to the fact that here only upregulation of cFLIPLong was relevant. Furthermore the experiments in this study showed that TNFR2-mediated depletion of TRAF2 resulted in a sensitization for TNFR1-induced cell death, for example in JJN3-cells. Taken together, this study revealed that the TNF-TNFR system influenced the outcome of activation of the extrinsic apoptotic pathway in myeloma cells by various mechanisms and the effect of TNF on MM cell survival is thus context dependent. KW - Apoptosis KW - Tumor-Nekrose-Faktor KW - Plasmozytom KW - apoptosis KW - TNF KW - multiple myeloma Y1 - 2012 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-73998 ER - TY - JOUR A1 - Rauert, H. A1 - Stühmer, T. A1 - Bargou, R. A1 - Wajant, H. A1 - Siegmund, D. T1 - TNFR1 and TNFR2 regulate the extrinsic apoptotic pathway in myeloma cells by multiple mechanisms JF - Cell Death and Disease N2 - The huge majority of myeloma cell lines express TNFR2 while a substantial subset of them failed to show TNFR1 expression. Stimulation of TNFR1 in the TNFR1-expressing subset of MM cell lines had no or only a very mild effect on cellular viability. Surprisingly, however, TNF stimulation enhanced cell death induction by CD95L and attenuated the apoptotic effect of TRAIL. The contrasting regulation of TRAIL- and CD95L-induced cell death by TNF could be traced back to the concomitant NFjBmediated upregulation of CD95 and the antiapoptotic FLIP protein. It appeared that CD95 induction, due to its strength, overcompensated a rather moderate upregulation of FLIP so that the net effect of TNF-induced NFjB activation in the context of CD95 signaling is pro-apoptotic. TRAIL-induced cell death, however, was antagonized in response to TNF because in this context only the induction of FLIP is relevant. Stimulation of TNFR2 in myeloma cells leads to TRAF2 depletion. In line with this, we observed cell death induction in TNFR1-TNFR2-costimulated JJN3 cells. Our studies revealed that the TNF-TNF receptor system adjusts the responsiveness of the extrinsic apoptotic pathway in myeloma cells by multiple mechanisms that generate a highly context-dependent net effect on myeloma cell survival KW - apoptosis KW - CD95 KW - multiple myeloma KW - NFkB KW - TNF KW - TRAIL KW - NF-Kappa-B KW - Tumor-necrosis-factor KW - Factor receptor KW - Factor-alpha KW - Activation KW - Polymorphisms KW - Inhibitor KW - Promoter KW - Transcription KW - Expression Y1 - 2011 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-133486 VL - 2 ER - TY - JOUR A1 - Philipp-Abbrederis, Kathrin A1 - Herrmann, Ken A1 - Knop, Stefan A1 - Schottelius, Margret A1 - Eiber, Matthias A1 - Lückerath, Katharina A1 - Pietschmann, Elke A1 - Habringer, Stefan A1 - Gerngroß, Carlos A1 - Franke, Katharina A1 - Rudelius, Martina A1 - Schirbel, Andreas A1 - Lapa, Constantin A1 - Schwamborn, Kristina A1 - Steidle, Sabine A1 - Hartmann, Elena A1 - Rosenwald, Andreas A1 - Kropf, Saskia A1 - Beer, Ambros J A1 - Peschel, Christian A1 - Einsele, Hermann A1 - Buck, Andreas K A1 - Schwaiger, Markus A1 - Götze, Katharina A1 - Wester, Hans-Jürgen A1 - Keller, Ulrich T1 - In vivo molecular imaging of chemokine receptor CXCR4 expression in patients with advanced multiple myeloma JF - EMBO Molecular Medicine N2 - CXCR4 is a G-protein-coupled receptor that mediates recruitment of blood cells toward its ligand SDF-1. In cancer, high CXCR4 expression is frequently associated with tumor dissemination andpoor prognosis. We evaluated the novel CXCR4 probe [\(^{68}\)Ga]Pentixafor for invivo mapping of CXCR4 expression density in mice xenografted with human CXCR4-positive MM cell lines and patients with advanced MM by means of positron emission tomography (PET). [\(^{68}\)Ga]Pentixafor PET provided images with excellent specificity and contrast. In 10 of 14 patients with advanced MM [\(^{68}\)Ga]Pentixafor PET/CT scans revealed MM manifestations, whereas only nine of 14 standard [\(^{18}\)F]fluorodeoxyglucose PET/CT scans were rated visually positive. Assessment of blood counts and standard CD34\(^{+}\) flow cytometry did not reveal significant blood count changes associated with tracer application. Based on these highly encouraging data on clinical PET imaging of CXCR4 expression in a cohort of MM patients, we conclude that [\(^{68}\)Ga]Pentixafor PET opens a broad field for clinical investigations on CXCR4 expression and for CXCR4-directed therapeutic approaches in MM and other diseases. KW - FDG PET/CT KW - cells KW - CXCR4/SDF-1 KW - CXCR4 KW - multiple myeloma KW - positron emission tomography KW - chemokine receptor KW - in vivo imaging KW - malignancies KW - involvement KW - microenvironment KW - survival KW - cancer KW - autologous transplantation KW - bone disease Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-148738 VL - 7 IS - 4 ER - TY - JOUR A1 - Othman, Eman M. A1 - Bekhit, Amany A. A1 - Anany, Mohamed A. A1 - Dandekar, Thomas A1 - Ragab, Hanan M. A1 - Wahid, Ahmed T1 - Design, Synthesis, and Anticancer Screening for Repurposed Pyrazolo[3,4-d]pyrimidine Derivatives on Four Mammalian Cancer Cell Lines JF - Molecules N2 - The present study reports the synthesis of new purine bioisosteres comprising a pyrazolo[3,4-d]pyrimidine scaffold linked to mono-, di-, and trimethoxy benzylidene moieties through hydrazine linkages. First, in silico docking experiments of the synthesized compounds against Bax, Bcl-2, Caspase-3, Ki67, p21, and p53 were performed in a trial to rationalize the observed cytotoxic activity for the tested compounds. The anticancer activity of these compounds was evaluated in vitro against Caco-2, A549, HT1080, and Hela cell lines. Results revealed that two (5 and 7) of the three synthesized compounds (5, 6, and 7) showed high cytotoxic activity against all tested cell lines with IC50 values in the micro molar concentration. Our in vitro results show that there is no significant apoptotic effect for the treatment with the experimental compounds on the viability of cells against A549 cells. Ki67 expression was found to decrease significantly following the treatment of cells with the most promising candidate: drug 7. The overall results indicate that these pyrazolopyrimidine derivatives possess anticancer activity at varying doses. The suggested mechanism of action involves the inhibition of the proliferation of cancer cells. KW - pyrazolo[3,4-d]pyrimidine KW - anticancer activity KW - apoptosis KW - Ki67 Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-239734 SN - 1420-3049 VL - 26 IS - 10 ER - TY - JOUR A1 - Medler, Juliane A1 - Kucka, Kirstin A1 - Wajant, Harald T1 - Tumor necrosis factor receptor 2 (TNFR2): an emerging target in cancer therapy JF - Cancers N2 - Despite the great success of TNF blockers in the treatment of autoimmune diseases and the identification of TNF as a factor that influences the development of tumors in many ways, the role of TNFR2 in tumor biology and its potential suitability as a therapeutic target in cancer therapy have long been underestimated. This has been fundamentally changed with the identification of TNFR2 as a regulatory T-cell (Treg)-stimulating factor and the general clinical breakthrough of immunotherapeutic approaches. However, considering TNFR2 as a sole immunosuppressive factor in the tumor microenvironment does not go far enough. TNFR2 can also co-stimulate CD8\(^+\) T-cells, sensitize some immune and tumor cells to the cytotoxic effects of TNFR1 and/or acts as an oncogene. In view of the wide range of cancer-associated TNFR2 activities, it is not surprising that both antagonists and agonists of TNFR2 are considered for tumor therapy and have indeed shown overwhelming anti-tumor activity in preclinical studies. Based on a brief summary of TNFR2 signaling and the immunoregulatory functions of TNFR2, we discuss here the main preclinical findings and insights gained with TNFR2 agonists and antagonists. In particular, we address the question of which TNFR2-associated molecular and cellular mechanisms underlie the observed anti-tumoral activities of TNFR2 agonists and antagonists. KW - NFkappaB KW - regulatory T-cell (Treg) KW - tumor necrosis factor (TNF) KW - TNF receptor 2 (TNFR2) KW - TNF receptor associated factor 1 and 2 (TRAF1, TRAF2) Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-275143 SN - 2072-6694 VL - 14 IS - 11 ER - TY - JOUR A1 - Lückerath, Katharina A1 - Lapa, Constantin A1 - Albert, Christa A1 - Herrmann, Ken A1 - Jörg, Gerhard A1 - Samnick, Samuel A1 - Einsele, Herrmann A1 - Knop, Stefan A1 - Buck, Andreas K. T1 - \(^{11}\)C-Methionine-PET: a novel and sensitive tool for monitoring of early response to treatment in multiple myeloma JF - Oncotarget N2 - Multiple myeloma (MM) remains an essentially incurable hematologic malignancy. However, new treatment modalities and novel drugs have been introduced and thus additional tools for therapy monitoring are increasingly needed. Therefore, we evaluated the radiotracers \(^{11}\)C-Methionine (paraprotein-biosynthesis) and \(^{18}\)F-FDG (glucose-utilization) for monitoring response to anti-myeloma-therapy and outcome prediction. Influence of proteasome-inhibition on radiotracer-uptake of different MM cell-lines and patient-derived CD138\(^{+}\) plasma cells was analyzed and related to tumor-biology. Mice xenotransplanted with MM. 1S tumors underwent MET- and FDG-\(\mu\)PET. Tumor-to-background ratios before and after 24 h, 8 and 15 days treatment with bortezomib were correlated to survival. Treatment reduced both MET and FDG uptake; changes in tracer-retention correlated with a switch from high to low CD138-expression. In xenotransplanted mice, MET-uptake significantly decreased by 30-79% as early as 24 h after bortezomib injection. No significant differences were detected thus early with FDG. This finding was confirmed in patient-derived MM cells. Importantly, early reduction of MET-but not FDG-uptake correlated with improved survival and reduced tumor burden in mice. Our results suggest that MET is superior to FDG in very early assessment of response to anti-myeloma-therapy. Early changes in MET-uptake have predictive potential regarding response and survival. MET-PET holds promise to individualize therapies in MM in future. KW - positron emission tomography KW - imaging techniques KW - experience KW - \(^{11}\)C-Methionine-PET KW - treatment response KW - molecular imaging KW - multiple myeloma KW - management KW - \(^{18}\)F-FDG PET/CT KW - bone disease KW - stem-cell transplantation KW - esophagogastric junction Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-148688 VL - 6 IS - 10 ER - TY - JOUR A1 - Lapa, Constantin A1 - Kircher, Malte A1 - Hänscheid, Heribert A1 - Schirbel, Andreas A1 - Grigoleit, Götz Ulrich A1 - Klinker, Erdwine A1 - Böck, Markus A1 - Samnick, Samuel A1 - Pelzer, Theo A1 - Buck, Andreas K T1 - Peptide receptor radionuclide therapy as a new tool in treatment-refractory sarcoidosis - initial experience in two patients JF - Theranostics N2 - Sarcoidosis is a multisystem granulomatous disorder of unknown etiology that can involve virtually all organ systems. Whereas most patients present without symptoms, progressive and disabling organ failure can occur in up to 10% of subjects. Somatostatin receptor (SSTR)-directed peptide receptor radionuclide therapy (PRRT) has recently received market authorization for treatment of SSTR-positive neuroendocrine tumors. Methods: We describe the first case series comprising two patients with refractory multi-organ involvement of sarcoidosis who received 4 cycles of PRRT. Results: PRRT was well-tolerated without any acute adverse effects. No relevant toxicities could be recorded during follow-up. Therapy resulted in partial response accompanied by a pronounced reduction in pain (patient #1) and stable disease regarding morphology as well as disease activity (patient #2), respectively. Conclusion: Peptide receptor radionuclide therapy in sarcoidosis is feasible and might be a new valuable tool in patients with otherwise treatment-refractory disease. Given the long experience with and good tolerability of PRRT, further evaluation of this new treatment option for otherwise treatment-refractory sarcoidosis in larger patient cohorts is warranted. KW - peptide receptor KW - PRRT KW - sarcoidosis KW - somatostatin receptors KW - radionuclide therapy Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-158983 VL - 8 IS - 3 ER -