TY - JOUR A1 - Jarick, Katja J. A1 - Mokhtari, Zeinab A1 - Scheller, Lukas A1 - Hartweg, Julia A1 - Thusek, Sina A1 - Le, Duc-Dung A1 - Ranecky, Maria A1 - Shaikh, Haroon A1 - Qureischi, Musga A1 - Heinze, Katrin G. A1 - Beilhack, Andreas T1 - Photoconversion of Alloreactive T Cells in Murine Peyer’s Patches During Acute Graft-Versus-Host Disease: Tracking the Homing Route of Highly Proliferative Cells In Vivo JF - Frontiers in Immunology N2 - The regulation of immune cell migration throughout the body is essential to warrant immunosurveillance and to maintain immune homeostasis. Marking and tracking of these cells has proven important to study mechanisms of immune cell trafficking and cell interaction in vivo. Photoconversion is a well-suited technique for intravital application because it enables contactless time- and location-specific marking of cells in the tissue without surgically manipulating the microenvironment of the cells in question. However, in dividing cells the converted fluorescent protein may decline quickly. Here, we provide a detailed description of the photoconversion technique and its applicability to tracking highly proliferating T cells from the priming site of T cell activation to peripheral target organs of effector function in a preclinical model. Dendra2+ T cells were photoconverted in the Peyer’s patches during the initiation phase of acute graft-versus-host disease (GvHD) and tracked through the mesenteric lymph nodes and the peripheral blood to the small intestine with flow cytometry and intravital two-photon microscopy. Photoconverted alloreactive T cells preserved the full proliferative capacity, homing, and migration of alloreactive T cells in the intestinal lamina propria. We conclusively proved that photoconversion of highly proliferative alloreactive T cells in the Peyer’s patches is an effective tool to study trafficking of alloreactive T cells under physiologic conditions and to GvHD target tissues. This technique can also be applied to the study of immune cell tracking under inflammatory and non-inflammatory conditions. KW - T cell migration KW - acute graft-versus-host disease KW - mouse models KW - photoconversion KW - Dendra2 KW - Peyer's patch KW - in vivo cell tracking KW - lymphocyte homing Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-323309 VL - 9 ER - TY - JOUR A1 - Herster, Franziska A1 - Bittner, Zsofia A1 - Codrea, Marius Cosmin A1 - Archer, Nathan K. A1 - Heister, Martin A1 - Löffler, Markus W. A1 - Heumos, Simon A1 - Wegner, Joanna A1 - Businger, Ramona A1 - Schindler, Michael A1 - Stegner, David A1 - Schäkel, Knut A1 - Grabbe, Stephan A1 - Ghoreschi, Kamran A1 - Miller, Lloyd S. A1 - Weber, Alexander N. R. T1 - Platelets Aggregate With Neutrophils and Promote Skin Pathology in Psoriasis JF - Frontiers in Immunology N2 - Psoriasis is a frequent systemic inflammatory autoimmune disease characterized primarily by skin lesions with massive infiltration of leukocytes, but frequently also presents with cardiovascular comorbidities. Especially polymorphonuclear neutrophils (PMNs) abundantly infiltrate psoriatic skin but the cues that prompt PMNs to home to the skin are not well-defined. To identify PMN surface receptors that may explain PMN skin homing in psoriasis patients, we screened 332 surface antigens on primary human blood PMNs from healthy donors and psoriasis patients. We identified platelet surface antigens as a defining feature of psoriasis PMNs, due to a significantly increased aggregation of neutrophils and platelets in the blood of psoriasis patients. Similarly, in the imiquimod-induced experimental in vivo mouse model of psoriasis, disease induction promoted PMN-platelet aggregate formation. In psoriasis patients, disease incidence directly correlated with blood platelet counts and platelets were detected in direct contact with PMNs in psoriatic but not healthy skin. Importantly, depletion of circulating platelets in mice in vivo ameliorated disease severity significantly, indicating that both PMNs and platelets may be relevant for psoriasis pathology and disease severity. KW - psoriasis KW - neutrophil KW - platelet KW - platelet-neutrophil complexes (PNCs) KW - imiquimod Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-320175 VL - 10 ER - TY - JOUR A1 - Schurr, Yvonne A1 - Spindler, Markus A1 - Kurz, Hendrikje A1 - Bender, Markus T1 - The cytoskeletal crosslinking protein MACF1 is dispensable for thrombus formation and hemostasis JF - Scientific Reports N2 - Coordinated reorganization of cytoskeletal structures is critical for key aspects of platelet physiology. While several studies have addressed the role of microtubules and filamentous actin in platelet production and function, the significance of their crosstalk in these processes has been poorly investigated. The microtubule-actin cross-linking factor 1 (MACF1; synonym: Actin cross-linking factor 7, ACF7) is a member of the spectraplakin family, and one of the few proteins expressed in platelets, which possess actin and microtubule binding domains thereby facilitating actin-microtubule interaction and regulation. We used megakaryocyte- and platelet-specific Macf1 knockout (Macf1fl/fl, Pf4-Cre) mice to study the role of MACF1 in platelet production and function. MACF1 deficient mice displayed comparable platelet counts to control mice. Analysis of the platelet cytoskeletal ultrastructure revealed a normal marginal band and actin network. Platelet spreading on fibrinogen was slightly delayed but platelet activation and clot traction was unaffected. Ex vivo thrombus formation and mouse tail bleeding responses were similar between control and mutant mice. These results suggest that MACF1 is dispensable for thrombopoiesis, platelet activation, thrombus formation and the hemostatic function in mice. KW - actin KW - microtubules Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-234966 VL - 9 ER - TY - JOUR A1 - Lee, Hong-Jen A1 - Li, Chien-Feng A1 - Ruan, Diane A1 - He, Jiabei A1 - Montal, Emily D. A1 - Lorenz, Sonja A1 - Girnun, Geoffrey D. A1 - Chan, Chia-Hsin T1 - Non-proteolytic ubiquitination of Hexokinase 2 by HectH9 controls tumor metabolism and cancer stem cell expansion JF - Nature Communications N2 - Enormous efforts have been made to target metabolic dependencies of cancer cells for developing new therapies. However, the therapeutic efficacy of glycolysis inhibitors is limited due to their inability to elicit cell death. Hexokinase 2 (HK2), via its mitochondrial localization, functions as a central nexus integrating glycolysis activation and apoptosis resilience. Here we identify that K63-linked ubiquitination by HectH9 regulates the mitochondrial localization and function of HK2. Through stable isotope tracer approach and functional metabolic analyses, we show that HectH9 deficiency impedes tumor glucose metabolism and growth by HK2 inhibition. The HectH9/HK2 pathway regulates cancer stem cell (CSC) expansion and CSC-associated chemoresistance. Histological analyses show that HectH9 expression is upregulated and correlated with disease progression in prostate cancer. This work uncovers that HectH9 is a novel regulator of HK2 and cancer metabolism. Targeting HectH9 represents an effective strategy to achieve long-term tumor remission by concomitantly disrupting glycolysis and inducing apoptosis. KW - cancer KW - cancer metabolism KW - molecular biology Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-236445 VL - 10 ER - TY - JOUR A1 - Hines, Rochelle M. A1 - Maric, Hans Michael A1 - Hines, Dustin J. A1 - Modgil, Amit A1 - Panzanelli, Patrizia A1 - Nakamura, Yasuko A1 - Nathanson, Anna J. A1 - Cross, Alan A1 - Deeb, Tarek A1 - Brandon, Nicholas J. A1 - Davies, Paul A1 - Fritschy, Jean-Marc A1 - Schindelin, Hermann A1 - Moss, Stephen J. T1 - Developmental seizures and mortality result from reducing GABAA receptor α2-subunit interaction with collybistin JF - Nature Communications N2 - Fast inhibitory synaptic transmission is mediated by γ-aminobutyric acid type A receptors (GABAARs) that are enriched at functionally diverse synapses via mechanisms that remain unclear. Using isothermal titration calorimetry and complementary methods we demonstrate an exclusive low micromolar binding of collybistin to the α2-subunit of GABAARs. To explore the biological relevance of collybistin-α2-subunit selectivity, we generate mice with a mutation in the α2-subunit-collybistin binding region (Gabra2-1). The mutation results in loss of a distinct subset of inhibitory synapses and decreased amplitude of inhibitory synaptic currents. Gabra2–1 mice have a striking phenotype characterized by increased susceptibility to seizures and early mortality. Surviving Gabra2-1 mice show anxiety and elevations in electroencephalogram δ power, which are ameliorated by treatment with the α2/α3-selective positive modulator, AZD7325. Taken together, our results demonstrate an α2-subunit selective binding of collybistin, which plays a key role in patterned brain activity, particularly during development. KW - cellular neuroscience KW - ion channels in the nervous system KW - neurotransmitters KW - synaptic development Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-320719 VL - 9 ER - TY - JOUR A1 - Heil, Hannah S. A1 - Schreiber, Benjamin A1 - Götz, Ralph A1 - Emmerling, Monika A1 - Dabauvalle, Marie-Christine A1 - Krohne, Georg A1 - Höfling, Sven A1 - Kamp, Martin A1 - Sauer, Markus A1 - Heinze, Katrin G. T1 - Sharpening emitter localization in front of a tuned mirror JF - Light: Science & Applications N2 - Single-molecule localization microscopy (SMLM) aims for maximized precision and a high signal-to-noise ratio1. Both features can be provided by placing the emitter in front of a metal-dielectric nanocoating that acts as a tuned mirror2,3,4. Here, we demonstrate that a higher photon yield at a lower background on biocompatible metal-dielectric nanocoatings substantially improves SMLM performance and increases the localization precision by up to a factor of two. The resolution improvement relies solely on easy-to-fabricate nanocoatings on standard glass coverslips and is spectrally and spatially tunable by the layer design and wavelength, as experimentally demonstrated for dual-color SMLM in cells. KW - imaging and sensing KW - super-resolution microscopy Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-228080 VL - 7 ER - TY - THES A1 - Nair, Radhika Karal T1 - Structural and biochemical characterization of USP28 inhibition by small molecule inhibitors T1 - Strukturelle und biochemische Charakterisierung der Hemmung von USP28 durch niedermolekulare Inhibitoren N2 - Ubiquitination is an important post-translational modification that maintains cellular homeostasis by regulating various biological processes. Deubiquitinases (DUBs) are enzymes that reverse the ubiquitination process by catalyzing the removal of ubiquitin from a substrate. Abnormal expression or function of DUBs is often associated with the onset and progression of various diseases, including cancer. Ubiquitin specific proteases (USPs), which constitute the largest family of DUBs in humans, have become the center of interest as potential targets in cancer therapy as many of them display increased activity or are overexpressed in a range of malignant tumors or the tumor microenvironment. Two related members of the USP family, USP28 and USP25, share high sequence identities but play diverse biological roles. USP28 regulates cell proliferation, oncogenesis, DNA damage repair and apoptosis, whereas USP25 is involved in the anti-viral response, innate immunity and ER-associated degradation in addition to carcinogenesis. USP28 and USP25 also exhibit different oligomeric states – while USP28 is a constitutively active dimer, USP25 assumes an auto-inhibited tetrameric structure. The catalytic domains of both USP28 and USP25 comprise the canonical, globular USP-domain but contain an additional, extended insertion site called USP25/28 catalytic domain inserted domain (UCID) that mediates oligomerization of the proteins. Disruption of the USP25 tetramer leads to the formation of an activated dimeric protein. However, it is still not clear what triggers its activation. Due to their role in maintaining and stabilizing numerous oncoproteins, USP28 and USP25 have emerged as interesting candidates for anti-cancer therapy. Recent advances in small-molecular inhibitor development have led to the discovery of relatively potent inhibitors of USP28 and USP25. This thesis focuses on the structural elucidation of USP28 and the biochemical characterization of USP28/USP25, both in complex with representatives of three out of the eight compound classes reported as USP28/USP25-specific inhibitors. The crystal structures of USP28 in complex with the AZ compounds, Vismodegib and FT206 reveal that all three inhibitor classes bind into the same allosteric pocket distant from the catalytic center, located between the palm and the thumb subdomains (the S1-site). Intriguingly, this binding pocket is identical to the UCID-tip binding interface in the USP25 tetramer, rendering the protein in a locked, inactive conformation. Formation of the binding pocket in USP28 requires a shift in the helix α5, which induces conformational changes and local distortion of the binding channel that typically accommodates the C-terminal tail of Ubiquitin, thus preventing catalysis and abrogating USP28 activity. The key residues of the USP28-inhibitor binding pocket are highly conserved in USP25. Mutagenesis studies of these residues accompanied by biochemical and biophysical assays confirm the proposed mechanism of inhibition and similar binding to USP25. This work provides valuable insights into the inhibition mechanism of the small molecule compounds specifically for the DUBs USP28 and USP25. The USP28-inhibitor complex structures offer a framework to develop more specific and potent inhibitors. N2 - Ubiquitinierung ist eine wichtige posttranslationale Modifikation, die die zelluläre Homöostase aufrechterhält, indem sie verschiedene biologische Prozesse reguliert. Deubiquitinasen (DUBs) sind Enzyme, die den Ubiquitinierungsprozess umkehren, indem sie die Entfernung von Ubiquitin von einem Substrat katalysieren. Eine abnorme Expression oder Funktion von DUBs wird häufig mit dem Auftreten und Fortschreiten verschiedener Krankheiten, einschließlich Krebs, in Verbindung gebracht. Ubiquitin-spezifische Proteasen (USPs), die im Menschen die größte Familie der DUBs bilden, sind als potenzielle Ziele in der Krebstherapie von besonderem Interesse, da viele von ihnen in bösartigen Tumoren oder deren Mikroumgebung abnormal aktiv oder überexprimiert sind. Die zwei eng verwandten Mitglieder der USP-Familie, USP28 und USP25, weisen eine hohe Sequenzidentität auf, sind aber an unterschiedlichen biologischen Prozessen beteiligt. USP28 reguliert die Zellproliferation, die Onkogenese, die Reparatur von DNA-Schäden und die Apoptose, während USP25 eine Rolle bei der antiviralen Reaktion, der angeborenen Immunität, dem ER-assoziierten Abbau und der Carcinogenese spielt. USP28 und USP25 weisen auch unterschiedliche oligomere Zustände auf. Während USP28 ein konstitutiv aktives Dimer bildet, tritt USP25 als auto-inhibiertes Tetramer auf. Strukturell bestehen die katalytischen Domänen sowohl von USP28 als auch von USP25 aus der kanonischen globulären USP-Domäne enthalten jedoch eine zusätzliche Insertion, die als „USP25/28 catalytic domain inserted domain (UCID)“ bezeichnet wird und die Oligomerisierung der Proteine vermittelt. Die Dissoziation des USP25 Tetramers in Dimere führt zu einem aktivierten USP25-Protein. Es ist jedoch immer noch nicht klar, was seine Aktivierung auslöst. Aufgrund ihrer Rolle bei der Aufrechterhaltung und Stabilisierung zahlreicher Onkoproteine haben sich USP28 und USP25 als interessante Kandidaten für die Entwicklung von Medikamenten in der Krebstherapie erwiesen. Jüngste Fortschritte in der Entwicklung von niedermolekularen Inhibitoren haben zur Entdeckung von relativ potenten Inhibitoren von USP28 und USP25 geführt. Diese Arbeit konzentriert sich auf die Strukturaufklärung von USP28 und die biochemische Charakterisierung von USP28/USP25, beide im Komplex mit Vertretern von drei der acht Verbindungsklassen, die als USP28/USP25-spezifische Inhibitoren bekannt sind. Die Kristallstrukturen von USP28 im Komplex mit den AZ-Verbindungen, Vismodegib und FT206 zeigen, dass alle Inhibitoren in einer ähnlichen Region an USP28 binden - einer allosterischen Tasche, die in der Nähe des katalytischen Zentrums liegt und sich zwischen der Handflächen- und der Daumen-Subdomäne befindet. Diese Bindungstasche ist identisch mit der Position, an der der „UCID-tip“ im USP25-Tetramer bindet und das Protein in eine verschränkte, inaktive Konformation versetzt. Die Bildung der Bindungstasche in USP28 erfordert eine Verschiebung der α5-Helix, die zu Konformationsänderungen und einer lokalen Verzerrung des Bindungskanalsführt, der normalerweise den C-terminus des Ubiquitin-Moleküls bindet und so die Katalyse verhindert und die Aktivität von USP28 hemmt. Die Schlüsselreste der USP28-Inhibitor-Bindungstasche sind in USP25 hoch konserviert. Mutagenese-Studien dieser Aminosäuren, begleitet von biochemischen und biophysikalischen Analysen, bestätigen den vorgeschlagenen Mechanismus der Hemmung und eine ähnliche Bindung der Inhibitoren an USP25. Diese Arbeit liefert wertvolle Einblicke in den Hemmungsmechanismus der Kleinmolekülverbindungen, die spezifisch für die DUBs USP28 und USP25 entwickelt worden sind. Die Strukturen der USP28-Inhibitor-Komplexe bieten eine Grundlage für die zukünftige Entwicklung spezifischerer und wirksamerer Inhibitoren. KW - USP KW - Inhibition KW - enzyme KW - crystallography KW - Unique Selling Proposition KW - Inhibition KW - Enzym KW - Kristallographie Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-281742 ER - TY - JOUR A1 - Harnoš, Jakub A1 - Cañizal, Maria Consuelo Alonso A1 - Jurásek, Miroslav A1 - Kumar, Jitender A1 - Holler, Cornelia A1 - Schambony, Alexandra A1 - Hanáková, Kateřina A1 - Bernatík, Ondřej A1 - Zdráhal, Zbynêk A1 - Gömöryová, Kristína A1 - Gybeľ, Tomáš A1 - Radaszkiewicz, Tomasz Witold A1 - Kravec, Marek A1 - Trantírek, Lukáš A1 - Ryneš, Jan A1 - Dave, Zankruti A1 - Fernández-Llamazares, Ana Iris A1 - Vácha, Robert A1 - Tripsianes, Konstantinos A1 - Hoffmann, Carsten A1 - Bryja, Vítězslav T1 - Dishevelled-3 conformation dynamics analyzed by FRET-based biosensors reveals a key role of casein kinase 1 JF - Nature Communications N2 - Dishevelled (DVL) is the key component of the Wnt signaling pathway. Currently, DVL conformational dynamics under native conditions is unknown. To overcome this limitation, we develop the Fluorescein Arsenical Hairpin Binder- (FlAsH-) based FRET in vivo approach to study DVL conformation in living cells. Using this single-cell FRET approach, we demonstrate that (i) Wnt ligands induce open DVL conformation, (ii) DVL variants that are predominantly open, show more even subcellular localization and more efficient membrane recruitment by Frizzled (FZD) and (iii) Casein kinase 1 ɛ (CK1ɛ) has a key regulatory function in DVL conformational dynamics. In silico modeling and in vitro biophysical methods explain how CK1ɛ-specific phosphorylation events control DVL conformations via modulation of the PDZ domain and its interaction with DVL C-terminus. In summary, our study describes an experimental tool for DVL conformational sampling in living cells and elucidates the essential regulatory role of CK1ɛ in DVL conformational dynamics. KW - biological techniques KW - cell signalling KW - phosphorylation Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-227837 VL - 10 ER - TY - JOUR A1 - Gotru, Sanjeev Kiran A1 - van Geffen, Johanna P. A1 - Nagy, Magdolna A1 - Mammadova-Bach, Elmina A1 - Eilenberger, Julia A1 - Volz, Julia A1 - Manukjan, Georgi A1 - Schulze, Harald A1 - Wagner, Leonard A1 - Eber, Stefan A1 - Schambeck, Christian A1 - Deppermann, Carsten A1 - Brouns, Sanne A1 - Nurden, Paquita A1 - Greinacher, Andreas A1 - Sachs, Ulrich A1 - Nieswandt, Bernhard A1 - Hermanns, Heike M. A1 - Heemskerk, Johan W. M. A1 - Braun, Attila T1 - Defective Zn2+ homeostasis in mouse and human platelets with α- and δ-storage pool diseases JF - Scientific Reports N2 - Zinc (Zn2+) can modulate platelet and coagulation activation pathways, including fibrin formation. Here, we studied the (patho)physiological consequences of abnormal platelet Zn2+ storage and release. To visualize Zn2+ storage in human and mouse platelets, the Zn2+ specific fluorescent dye FluoZin3 was used. In resting platelets, the dye transiently accumulated into distinct cytosolic puncta, which were lost upon platelet activation. Platelets isolated from Unc13d−/− mice, characterized by combined defects of α/δ granular release, showed a markedly impaired Zn2+ release upon activation. Platelets from Nbeal2−/− mice mimicking Gray platelet syndrome (GPS), characterized by primarily loss of the α-granule content, had strongly reduced Zn2+ levels, which was also confirmed in primary megakaryocytes. In human platelets isolated from patients with GPS, Hermansky-Pudlak Syndrome (HPS) and Storage Pool Disease (SPD) altered Zn2+ homeostasis was detected. In turbidity and flow based assays, platelet-dependent fibrin formation was impaired in both Nbeal2−/− and Unc13d−/− mice, and the impairment could be partially restored by extracellular Zn2+. Altogether, we conclude that the release of ionic Zn2+ store from secretory granules upon platelet activation contributes to the procoagulant role of Zn2+ in platelet-dependent fibrin formation. KW - coagulation system KW - metals Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-227455 VL - 9 ER - TY - JOUR A1 - El-Mesery, Mohamed A1 - Rosenthal, Tina A1 - Rauert-Wunderlich, Hilka A1 - Schreder, Martin A1 - Stühmer, Thorsten A1 - Leich, Ellen A1 - Schlosser, Andreas A1 - Ehrenschwender, Martin A1 - Wajant, Harald A1 - Siegmund, Daniela T1 - The NEDD8-activating enzyme inhibitor MLN4924 sensitizes a TNFR1+ subgroup of multiple myeloma cells for TNF-induced cell death JF - Cell Death & Disease N2 - The NEDD8-activating enzyme (NAE) inhibitor MLN4924 inhibits cullin-RING ubiquitin ligase complexes including the SKP1-cullin-F-box E3 ligase βTrCP. MLN4924 therefore inhibits also the βTrCP-dependent activation of the classical and the alternative NFĸB pathway. In this work, we found that a subgroup of multiple myeloma cell lines (e.g., RPMI-8226, MM.1S, KMS-12BM) and about half of the primary myeloma samples tested are sensitized to TNF-induced cell death by MLN4924. This correlated with MLN4924-mediated inhibition of TNF-induced activation of the classical NFκB pathway and reduced the efficacy of TNF-induced TNFR1 signaling complex formation. Interestingly, binding studies revealed a straightforward correlation between cell surface TNFR1 expression in multiple myeloma cell lines and their sensitivity for MLN4924/TNF-induced cell death. The cell surface expression levels of TNFR1 in the investigated MM cell lines largely correlated with TNFR1 mRNA expression. This suggests that the variable levels of cell surface expression of TNFR1 in myeloma cell lines are decisive for TNF/MLN4924 sensitivity. Indeed, introduction of TNFR1 into TNFR1-negative TNF/MLN4924-resistant KMS-11BM cells, was sufficient to sensitize this cell line for TNF/MLN4924-induced cell death. Thus, MLN4924 might be especially effective in myeloma patients with TNFR1+ myeloma cells and a TNFhigh tumor microenvironment. KW - cancer therapy KW - tumour-necrosis factors Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-226666 VL - 10 ER - TY - INPR A1 - Hennig, Thomas A1 - Prusty, Archana B. A1 - Kaufer, Benedikt A1 - Whisnant, Adam W. A1 - Lodha, Manivel A1 - Enders, Antje A1 - Thomas, Julius A1 - Kasimir, Francesca A1 - Grothey, Arnhild A1 - Herb, Stefanie A1 - Jürges, Christopher A1 - Meister, Gunter A1 - Erhard, Florian A1 - Dölken, Lars A1 - Prusty, Bhupesh K. T1 - Selective inhibition of miRNA 1 processing by a herpesvirus encoded miRNA N2 - Herpesviruses have mastered host cell modulation and immune evasion to augment productive infection, life-long latency and reactivation thereof 1,2. A long appreciated, yet elusively defined relationship exists between the lytic-latent switch and viral non-coding RNAs 3,4. Here, we identify miRNA-mediated inhibition of miRNA processing as a thus far unknown cellular mechanism that human herpesvirus 6A (HHV-6A) exploits to disrupt mitochondrial architecture, evade intrinsic host defense and drive the lytic-latent switch. We demonstrate that virus-encoded miR-aU14 selectively inhibits the processing of multiple miR-30 family members by direct interaction with the respective pri-miRNA hairpin loops. Subsequent loss of miR-30 and activation of the miR-30/p53/Drp1 axis triggers a profound disruption of mitochondrial architecture. This impairs induction of type I interferons and is necessary for both productive infection and virus reactivation. Ectopic expression of miR-aU14 triggered virus reactivation from latency, identifying viral miR-aU14 as a readily drugable master regulator of the herpesvirus lytic-latent switch. Our results show that miRNA-mediated inhibition of miRNA processing represents a generalized cellular mechanism that can be exploited to selectively target individual members of miRNA families. We anticipate that targeting miR-aU14 provides exciting therapeutic options for preventing herpesvirus reactivations in HHV-6-associated disorders. KW - Herpesvirus KW - HHV-6A KW - miRNA processing KW - miR-30 KW - mitochondria KW - fusion and fission KW - type I interferon KW - latency KW - virus reactivation Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-267862 ET - accepted version ER - TY - JOUR A1 - Brünnert, Daniela A1 - Seupel, Raina A1 - Goyal, Pankaj A1 - Bach, Matthias A1 - Schraud, Heike A1 - Kirner, Stefanie A1 - Köster, Eva A1 - Feineis, Doris A1 - Bargou, Ralf C. A1 - Schlosser, Andreas A1 - Bringmann, Gerhard A1 - Chatterjee, Manik T1 - Ancistrocladinium A induces apoptosis in proteasome inhibitor-resistant multiple myeloma cells: a promising therapeutic agent candidate JF - Pharmaceuticals N2 - The N,C-coupled naphthylisoquinoline alkaloid ancistrocladinium A belongs to a novel class of natural products with potent antiprotozoal activity. Its effects on tumor cells, however, have not yet been explored. We demonstrate the antitumor activity of ancistrocladinium A in multiple myeloma (MM), a yet incurable blood cancer that represents a model disease for adaptation to proteotoxic stress. Viability assays showed a potent apoptosis-inducing effect of ancistrocladinium A in MM cell lines, including those with proteasome inhibitor (PI) resistance, and in primary MM cells, but not in non-malignant blood cells. Concomitant treatment with the PI carfilzomib or the histone deacetylase inhibitor panobinostat strongly enhanced the ancistrocladinium A-induced apoptosis. Mass spectrometry with biotinylated ancistrocladinium A revealed significant enrichment of RNA-splicing-associated proteins. Affected RNA-splicing-associated pathways included genes involved in proteotoxic stress response, such as PSMB5-associated genes and the heat shock proteins HSP90 and HSP70. Furthermore, we found strong induction of ATF4 and the ATM/H2AX pathway, both of which are critically involved in the integrated cellular response following proteotoxic and oxidative stress. Taken together, our data indicate that ancistrocladinium A targets cellular stress regulation in MM and improves the therapeutic response to PIs or overcomes PI resistance, and thus may represent a promising potential therapeutic agent. KW - multiple myeloma KW - ancistrocladinium A KW - naphthylisoquinoline alkaloids KW - proteasome inhibitor resistance KW - RNA splicing KW - cellular stress response KW - proteasome subunit beta type-5 (PSMB5) KW - activating transcription factor 4 (ATF4) KW - ataxia teleagiectasia mutated (ATM) KW - H2A histone family member X (H2AX) Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-362887 SN - 1424-8247 VL - 16 IS - 8 ER - TY - JOUR A1 - Rauschenberger, Vera A1 - Piro, Inken A1 - Kasaragod, Vikram Babu A1 - Hörlin, Verena A1 - Eckes, Anna-Lena A1 - Kluck, Christoph J. A1 - Schindelin, Hermann A1 - Meinck, Hans-Michael A1 - Wickel, Jonathan A1 - Geis, Christian A1 - Tüzün, Erdem A1 - Doppler, Kathrin A1 - Sommer, Claudia A1 - Villmann, Carmen T1 - Glycine receptor autoantibody binding to the extracellular domain is independent from receptor glycosylation JF - Frontiers in Molecular Neuroscience N2 - Glycine receptor (GlyR) autoantibodies are associated with stiff-person syndrome and the life-threatening progressive encephalomyelitis with rigidity and myoclonus in children and adults. Patient histories show variability in symptoms and responses to therapeutic treatments. A better understanding of the autoantibody pathology is required to develop improved therapeutic strategies. So far, the underlying molecular pathomechanisms include enhanced receptor internalization and direct receptor blocking altering GlyR function. A common epitope of autoantibodies against the GlyRα1 has been previously defined to residues 1A-33G at the N-terminus of the mature GlyR extracellular domain. However, if other autoantibody binding sites exist or additional GlyR residues are involved in autoantibody binding is yet unknown. The present study investigates the importance of receptor glycosylation for binding of anti-GlyR autoantibodies. The glycine receptor α1 harbors only one glycosylation site at the amino acid residue asparagine 38 localized in close vicinity to the identified common autoantibody epitope. First, non-glycosylated GlyRs were characterized using protein biochemical approaches as well as electrophysiological recordings and molecular modeling. Molecular modeling of non-glycosylated GlyRα1 did not show major structural alterations. Moreover, non-glycosylation of the GlyRα1N38Q did not prevent the receptor from surface expression. At the functional level, the non-glycosylated GlyR demonstrated reduced glycine potency, but patient GlyR autoantibodies still bound to the surface-expressed non-glycosylated receptor protein in living cells. Efficient adsorption of GlyR autoantibodies from patient samples was possible by binding to native glycosylated and non-glycosylated GlyRα1 expressed in living not fixed transfected HEK293 cells. Binding of patient-derived GlyR autoantibodies to the non-glycosylated GlyRα1 offered the possibility to use purified non-glycosylated GlyR extracellular domain constructs coated on ELISA plates and use them as a fast screening readout for the presence of GlyR autoantibodies in patient serum samples. Following successful adsorption of patient autoantibodies by GlyR ECDs, binding to primary motoneurons and transfected cells was absent. Our results indicate that the glycine receptor autoantibody binding is independent of the receptor’s glycosylation state. Purified non-glycosylated receptor domains harbouring the autoantibody epitope thus provide, an additional reliable experimental tool besides binding to native receptors in cell-based assays for detection of autoantibody presence in patient sera. KW - glycine receptor KW - autoantibodies KW - glycosylation KW - extracellular domain KW - adsorption Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-304206 VL - 16 ER - TY - JOUR A1 - Osmanoglu, Özge A1 - Gupta, Shishir K. A1 - Almasi, Anna A1 - Yagci, Seray A1 - Srivastava, Mugdha A1 - Araujo, Gabriel H. M. A1 - Nagy, Zoltan A1 - Balkenhol, Johannes A1 - Dandekar, Thomas T1 - Signaling network analysis reveals fostamatinib as a potential drug to control platelet hyperactivation during SARS-CoV-2 infection JF - Frontiers in Immunology N2 - Introduction Pro-thrombotic events are one of the prevalent causes of intensive care unit (ICU) admissions among COVID-19 patients, although the signaling events in the stimulated platelets are still unclear. Methods We conducted a comparative analysis of platelet transcriptome data from healthy donors, ICU, and non-ICU COVID-19 patients to elucidate these mechanisms. To surpass previous analyses, we constructed models of involved networks and control cascades by integrating a global human signaling network with transcriptome data. We investigated the control of platelet hyperactivation and the specific proteins involved. Results Our study revealed that control of the platelet network in ICU patients is significantly higher than in non-ICU patients. Non-ICU patients require control over fewer proteins for managing platelet hyperactivity compared to ICU patients. Identification of indispensable proteins highlighted key subnetworks, that are targetable for system control in COVID-19-related platelet hyperactivity. We scrutinized FDA-approved drugs targeting indispensable proteins and identified fostamatinib as a potent candidate for preventing thrombosis in COVID-19 patients. Discussion Our findings shed light on how SARS-CoV-2 efficiently affects host platelets by targeting indispensable and critical proteins involved in the control of platelet activity. We evaluated several drugs for specific control of platelet hyperactivity in ICU patients suffering from platelet hyperactivation. The focus of our approach is repurposing existing drugs for optimal control over the signaling network responsible for platelet hyperactivity in COVID-19 patients. Our study offers specific pharmacological recommendations, with drug prioritization tailored to the distinct network states observed in each patient condition. Interactive networks and detailed results can be accessed at https://fostamatinib.bioinfo-wuerz.eu/. KW - signaling network KW - controllability KW - platelet KW - SARS-CoV-2 KW - fostamatinib KW - drug repurposing KW - COVID-19 Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-354158 VL - 14 ER - TY - INPR A1 - Brenner, Marian A1 - Zink, Christoph A1 - Witzinger, Linda A1 - Keller, Angelika A1 - Hadamek, Kerstin A1 - Bothe, Sebastian A1 - Neuenschwander, Martin A1 - Villmann, Carmen A1 - von Kries, Jens Peter A1 - Schindelin, Hermann A1 - Jeanclos, Elisabeth A1 - Gohla, Antje T1 - 7,8-Dihydroxyflavone is a direct inhibitor of pyridoxal phosphatase T2 - eLife N2 - Vitamin B6 deficiency has been linked to cognitive impairment in human brain disorders for decades. Still, the molecular mechanisms linking vitamin B6 to these pathologies remain poorly understood, and whether vitamin B6 supplementation improves cognition is unclear as well. Pyridoxal phosphatase (PDXP), an enzyme that controls levels of pyridoxal 5’-phosphate (PLP), the co-enzymatically active form of vitamin B6, may represent an alternative therapeutic entry point into vitamin B6-associated pathologies. However, pharmacological PDXP inhibitors to test this concept are lacking. We now identify a PDXP and age-dependent decline of PLP levels in the murine hippocampus that provides a rationale for the development of PDXP inhibitors. Using a combination of small molecule screening, protein crystallography and biolayer interferometry, we discover and analyze 7,8-dihydroxyflavone (7,8-DHF) as a direct and potent PDXP inhibitor. 7,8-DHF binds and reversibly inhibits PDXP with low micromolar affinity and sub-micromolar potency. In mouse hippocampal neurons, 7,8-DHF increases PLP in a PDXP-dependent manner. These findings validate PDXP as a druggable target. Of note, 7,8-DHF is a well-studied molecule in brain disorder models, although its mechanism of action is actively debated. Our discovery of 7,8-DHF as a PDXP inhibitor offers novel mechanistic insights into the controversy surrounding 7,8-DHF-mediated effects in the brain. KW - 7,8-dihydroxyflavone (7,8-DHF) KW - pyridoxal phosphatase (PDXP) KW - vitamin B6 KW - PDXP inhibitors Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-350446 ER - TY - JOUR A1 - Tessmer, Ingrid A1 - Margison, Geoffrey P. T1 - The DNA alkyltransferase family of DNA repair proteins: common mechanisms, diverse functions JF - International Journal of Molecular Sciences N2 - DNA alkyltransferase and alkyltransferase-like family proteins are responsible for the repair of highly mutagenic and cytotoxic O\(^6\)-alkylguanine and O\(^4\)-alkylthymine bases in DNA. Their mechanism involves binding to the damaged DNA and flipping the base out of the DNA helix into the active site pocket in the protein. Alkyltransferases then directly and irreversibly transfer the alkyl group from the base to the active site cysteine residue. In contrast, alkyltransferase-like proteins recruit nucleotide excision repair components for O\(^6\)-alkylguanine elimination. One or more of these proteins are found in all kingdoms of life, and where this has been determined, their overall DNA repair mechanism is strictly conserved between organisms. Nevertheless, between species, subtle as well as more extensive differences that affect target lesion preferences and/or introduce additional protein functions have evolved. Examining these differences and their functional consequences is intricately entwined with understanding the details of their DNA repair mechanism(s) and their biological roles. In this review, we will present and discuss various aspects of the current status of knowledge on this intriguing protein family. KW - DNA repair KW - O6-alkylguanine-DNA alkyltransferase KW - alkylation damage Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-355790 SN - 1422-0067 VL - 25 IS - 1 ER - TY - JOUR A1 - Meinert, Madlen A1 - Jessen, Christina A1 - Hufnagel, Anita A1 - Kreß, Julia Katharina Charlotte A1 - Burnworth, Mychal A1 - Däubler, Theo A1 - Gallasch, Till A1 - Da Xavier Silva, Thamara Nishida A1 - Dos Santos, Ancély Ferreira A1 - Ade, Carsten Patrick A1 - Schmitz, Werner A1 - Kneitz, Susanne A1 - Friedmann Angeli, José Pedro A1 - Meierjohann, Svenja T1 - Thiol starvation triggers melanoma state switching in an ATF4 and NRF2-dependent manner JF - Redox Biology N2 - The cystine/glutamate antiporter xCT is an important source of cysteine for cancer cells. Once taken up, cystine is reduced to cysteine and serves as a building block for the synthesis of glutathione, which efficiently protects cells from oxidative damage and prevents ferroptosis. As melanomas are particularly exposed to several sources of oxidative stress, we investigated the biological role of cysteine and glutathione supply by xCT in melanoma. xCT activity was abolished by genetic depletion in the Tyr::CreER; Braf\(^{CA}\); Pten\(^{lox/+}\) melanoma model and by acute cystine withdrawal in melanoma cell lines. Both interventions profoundly impacted melanoma glutathione levels, but they were surprisingly well tolerated by murine melanomas in vivo and by most human melanoma cell lines in vitro. RNA sequencing of human melanoma cells revealed a strong adaptive upregulation of NRF2 and ATF4 pathways, which orchestrated the compensatory upregulation of genes involved in antioxidant defence and de novo cysteine biosynthesis. In addition, the joint activation of ATF4 and NRF2 triggered a phenotypic switch characterized by a reduction of differentiation genes and induction of pro-invasive features, which was also observed after erastin treatment or the inhibition of glutathione synthesis. NRF2 alone was capable of inducing the phenotypic switch in a transient manner. Together, our data show that cystine or glutathione levels regulate the phenotypic plasticity of melanoma cells by elevating ATF4 and NRF2. KW - thiol starvation KW - ATF4 KW - NRF2 KW - melanoma Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-350328 VL - 70 ER - TY - JOUR A1 - Kreß, Julia Katharina Charlotte A1 - Jessen, Christina A1 - Hufnagel, Anita A1 - Schmitz, Werner A1 - Da Xavier Silva, Thamara Nishida A1 - Ferreira Dos Santos, Ancély A1 - Mosteo, Laura A1 - Goding, Colin R. A1 - Friedmann Angeli, José Pedro A1 - Meierjohann, Svenja T1 - The integrated stress response effector ATF4 is an obligatory metabolic activator of NRF2 JF - Cell Reports N2 - Highlights • The integrated stress response leads to a general ATF4-dependent activation of NRF2 • ATF4 causes a CHAC1-dependent GSH depletion, resulting in NRF2 stabilization • An elevation of NRF2 transcript levels fosters this effect • NRF2 supports the ISR/ATF4 pathway by improving cystine and antioxidant supply Summary The redox regulator NRF2 becomes activated upon oxidative and electrophilic stress and orchestrates a response program associated with redox regulation, metabolism, tumor therapy resistance, and immune suppression. Here, we describe an unrecognized link between the integrated stress response (ISR) and NRF2 mediated by the ISR effector ATF4. The ISR is commonly activated after starvation or ER stress and plays a central role in tissue homeostasis and cancer plasticity. ATF4 increases NRF2 transcription and induces the glutathione-degrading enzyme CHAC1, which we now show to be critically important for maintaining NRF2 activation. In-depth analyses reveal that NRF2 supports ATF4-induced cells by increasing cystine uptake via the glutamate-cystine antiporter xCT. In addition, NRF2 upregulates genes mediating thioredoxin usage and regeneration, thus balancing the glutathione decrease. In conclusion, we demonstrate that the NRF2 response serves as second layer of the ISR, an observation highly relevant for the understanding of cellular resilience in health and disease. KW - NRF2 KW - ATF4 KW - integrated stress response KW - CHAC1 KW - melanoma KW - SLC7A11 KW - GSH Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-350312 VL - 42 IS - 7 ER - TY - RPRT ED - Nieswandt, Bernhard T1 - Platelets – Molecular, cellular and systemic functions in health and disease T1 - Thrombozyten – molekulare, zelluläre und systemische Funktionen unter physiologischen und pathologischen Bedingungen (SFB/TR240 - Abschlussbericht BT - Final Report (2018/2 - 2023/1) N2 - Besides their central role in haemostasis and thrombosis, platelets are increasingly recognised as versatile effector cells in inflammation, the innate and adaptive immune response, extracellular matrix reorganisation and fibrosis, maintenance of barrier and organ integrity, and host response to pathogens. These platelet functions, referred to as thrombo-inflammation and immunothrombosis, have gained major attention in the COVID-19 pandemic, where patients develop an inflammatory disease state with severe and life-threatening thromboembolic complications. In the CRC/TR 240, a highly interdisciplinary team of basic, translational and clinical scientists explored these emerging roles of platelets with the aim to develop novel treatment concepts for cardiovascular disorders and beyond. We have i) unravelled mechanisms leading to life-threatening thromboembolic complica-tions following vaccination against SARS-CoV-2 with adenoviral vector-based vaccines, ii) identified unrecognised functions of platelet receptors and their regulation, offering new potential targets for pharmacological intervention and iii) developed new methodology to study the biology of megakar-yocytes (MKs), the precursor cells of platelets in the bone marrow, which lay the foundation for the modulation of platelet biogenesis and function. The projects of the CRC/TR 240 built on the unique expertise of our research network and focussed on the following complementary fields: (A) Cell bi-ology of megakaryocytes and platelets and (B) Platelets as regulators and effectors in disease. To achieve this aim, we followed a comprehensive approach starting out from in vitro systems and animal models to clinical research with large prospective patient cohorts and data-/biobanking. Despite the comparably short funding period the CRC/TR 240 discovered basic new mechanisms of platelet biogenesis, signal transduction and effector function and identified potential MK/platelet-specific molecular targets for diagnosis and therapy of thrombotic, haemorrhagic and thrombo-inflammatory disease states. N2 - Thrombozyten sind von zentraler Bedeutung für die Hämostase, aber auch bei der Entstehung akuter thrombotischer Erkrankungen wie Herzinfarkt oder Schlaganfall. Darüber hinaus sind Thrombozyten aber auch vielseitige Effektorzellen von Entzündungsprozessen, der angeborenen Immunität, bei zellulären Abwehrmechanismen sowie bei der Aufrechterhaltung der Gefäß- und Organintegrität. Diese neuen, als Thrombo-Inflammation und Immunothrombose bezeichneten Funktionen haben im Rahmen der COVID-19 Pandemie große Aufmerksamkeit erlangt, da betroffene Patienten systemische Entzündungszustände in Verbindung mit thromboembolischen Komplikationen aufweisen, die oft auch tödlich verlaufen. Im SFB/TR 240 arbeitete ein interdisziplinäres Team von grundlagenorientierten, translationalen und klinischen Wissenschaftlern zusammen an der Erforschung dieser neuartigen Thrombozytenfunktionen mit dem Ziel, neue verbesserte Therapiemöglichkeiten für kardiovaskuläre, aber auch andere Erkrankungen zu entwickeln. Während der Förderphase haben wir i) die Mechanismen aufgeklärt, die in seltenen Fällen nach Impfung mit Adenovirus-basierten Vakzinen gegen Sars-CoV-2 zu lebensbedrohlichen thromboembolischen Komplikationen führten, ii) neue Funktionen und Regulationsmechanismen thrombozytärer Rezeptoren identifiziert, die Grundlage zur therapeutischen Intervention sein könnten und iii) neue Technologien entwickelt, die vertiefte Studien zur Biologie der Megakaryozyten, den Vorläuferzellen der Thrombozyten im Knochenmark, ermöglichen und den Weg zu einer gezielten Beeinflussung der Thrombozytenbiogenese und –funktion ebnen könnten. Die Projekte des TR 240 konzentrierten sich auf die folgenden komplementären Forschungsgebiete: (A) Zellbiologie der Megakaryozyten und Thrombozyten mit dem Ziel eines verbesserten Verständnisses der grundlegenden Funktionen beider Zelltypen und (B) Thrombozyten als Modulatoren und Effektoren bei Erkrankungen. Um dieses Ziel zu erreichen, wurde ein sehr umfassender Ansatz verfolgt, der sich von in vitro Systemen über Tiermodelle bis hin zur klinischen Forschung mit Biobanken und großen, prospektiven Patientenkohorten erstreckte. Der SFB/TR 240 konnte in der vergleichsweisen kurzen Zeit seiner Förderung grundlegend neue Erkenntnisse zu den Mechanismen der Thrombozytenbiogenese, Thrombozyten-Signaltransduktion und -Effektorfunktionen erarbeiten und neue MK/Thrombozyten-spezifische Angriffspunkte für Diag-nose und Therapie thrombotischer, hämorrhagischer und thrombo-inflammatorischer Erkrankungen identifizieren. KW - Thrombozyt KW - platelets KW - thrombo-inflammation KW - haemostasis KW - stroke KW - megakaryocytes KW - Sonderforschungsbereich Transregio 240 KW - Bericht KW - Collaborative Research Center KW - Experimental Biomedicine Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-359636 ER - TY - JOUR A1 - Liu, Ruiqi A1 - Friedrich, Mike A1 - Hemmen, Katherina A1 - Jansen, Kerstin A1 - Adolfi, Mateus C. A1 - Schartl, Manfred A1 - Heinze, Katrin G. T1 - Dimerization of melanocortin 4 receptor controls puberty onset and body size polymorphism JF - Frontiers in Endocrinology N2 - Xiphophorus fish exhibit a clear phenotypic polymorphism in puberty onset and reproductive strategies of males. In X. nigrensis and X. multilineatus, puberty onset is genetically determined and linked to a melanocortin 4 receptor (Mc4r) polymorphism of wild-type and mutant alleles on the sex chromosomes. We hypothesized that Mc4r mutant alleles act on wild-type alleles by a dominant negative effect through receptor dimerization, leading to differential intracellular signaling and effector gene activation. Depending on signaling strength, the onset of puberty either occurs early or is delayed. Here, we show by Förster Resonance Energy Transfer (FRET) that wild-type Xiphophorus Mc4r monomers can form homodimers, but also heterodimers with mutant receptors resulting in compromised signaling which explains the reduced Mc4r signaling in large males. Thus, hetero- vs. homo- dimerization seems to be the key molecular mechanism for the polymorphism in puberty onset and body size in male fish. KW - fluorescence lifetime imaging microscopy KW - Förster Resonance Energy Transfer KW - Mc4r KW - puberty KW - Xiphophorus Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-354261 SN - 1664-2392 VL - 14 ER - TY - THES A1 - Karwen, Till T1 - Platelets promote insulin secretion of pancreatic β-cells T1 - Thrombozyten fördern die Insulinsekretion von pankreatischen β-Zellen N2 - The pancreas is the key organ for the maintenance of euglycemia. This is regulated in particular by α-cell-derived glucagon and β-cell-derived insulin, which are released in response to nutrient deficiency and elevated glucose levels, respectively. Although glucose is the main regulator of insulin secretion, it is significantly enhanced by various potentiators. Platelets are anucleate cell fragments in the bloodstream that are essential for hemostasis to prevent and stop bleeding events. Besides their classical role, platelets were implemented to be crucial for other physiological and pathophysiological processes, such as cancer progression, immune defense, and angiogenesis. Platelets from diabetic patients often present increased reactivity and basal activation. Interestingly, platelets store and release several substances that have been reported to potentiate insulin secretion by β-cells. For these reasons, the impact of platelets on β-cell functioning was investigated in this thesis. Here it was shown that both glucose and a β-cell-derived substance/s promote platelet activation and binding to collagen. Additionally, platelet adhesion specifically to the microvasculature of pancreatic islets was revealed, supporting the hypothesis of their influence on glucose homeostasis. Genetic or pharmacological ablation of platelet functioning and platelet depletion consistently resulted in reduced insulin secretion and associated glucose intolerance. Further, the platelet-derived lipid fraction was found to enhance glucose-stimulated insulin secretion, with 20-hydroxyeicosatetraenoic acid (20-HETE) and possibly also lyso-precursor of platelet-activating factor (lysoPAF) being identified as crucial factors. However, the acute platelet-stimulated insulin secretion was found to decline with age, as did the levels of platelet-derived 20-HETE. In addition to their direct stimulatory effect on insulin secretion, specific defects in platelet activation have also been shown to affect glucose homeostasis by potentially influencing islet vascular development. Taking together, the results of this thesis suggest a direct and indirect mechanism of platelets in the regulation of insulin secretion that ensures glucose homeostasis, especially in young individuals. N2 - Der Pankreas ist das Schlüsselorgan für die Aufrechterhaltung der Glukosehomöostase. Diese wird insbesondere durch das von α-Zellen stammende Glukagon und von β-Zellen stammende Insulin reguliert, die als Reaktion auf Nährstoffmangel beziehungsweise erhöhte Glukosespiegel freigesetzt werden. Obwohl Glukose der Hauptregulator der Insulinsekretion ist, wird sie durch verschiedene Potentiatoren erheblich gesteigert. Thrombozyten sind kernlose Zellfragmente im Blutkreislauf, die für die Hämostase unerlässlich sind. Neben ihrer klassischen Funktion sind sie auch an anderen physiologischen und pathophysiologischen Prozessen beteiligt, etwa an der Tumorentwicklung, der Immunabwehr und der Angiogenese. Thrombozyten von Diabetikern weisen häufig eine erhöhte Reaktivität und basale Aktivierung auf. Außerdem speichern und sekretieren sie Substanzen, von denen bekannt ist, dass sie die Insulinsekretion durch β-Zellen verstärken. Aus diesen Gründen wurde in dieser Arbeit der Einfluss von Thrombozyten auf die Funktion von β-Zellen untersucht. Es konnte gezeigt werden, dass sowohl Glukose als auch eine aus β-Zellen stammende Substanz/en die Thrombozytenaktivierung und die Bindung an Kollagen fördern. Darüber hinaus wurde eine spezifische Thrombozytenadhäsion an der Mikrovaskulatur der pankreatischen Inseln festgestellt, was die Hypothese ihres Einflusses auf die Glukosehomöostase unterstützt. Eine genetische oder pharmakologische Ablation der Thrombozytenfunktion sowie eine Depletion von Thrombozyten führten zu einer verminderten Insulinsekretion und einer damit verbundenen Glukoseintoleranz. Hierbei erwies sich die Lipidfraktion von Thrombozyten als essentieller Potentiator für die glukosestimulierte Insulinsekretion, wobei 20-Hydroxyeicosatetraensäure (20-HETE) und die Lyso-Vorstufe des Plättchen-Aktivierenden Faktors (LysoPAF) als entscheidende Faktoren identifiziert werden konnten. Weiterhin wurde festgestellt, dass sowohl der direkte stimulierende Effekt von Thrombozyten auf die Insulinsekretion, als auch deren 20-HETE Sekretion mit zunehmendem Alter abnimmt. Thrombozyten beeinflussten außerdem die Inselvaskularisierung, welche mutmaßlich zusätzlich zu Glukoseintoleranz führt. Insgesamt deuten die Ergebnisse dieser Arbeit auf einen direkten und indirekten Mechanismus der Thrombozyten bei der Regulierung der Insulinsekretion hin, der die Glukosehomöostase insbesondere bei jungen Menschen gewährleistet. KW - platelet KW - β cell KW - insulin KW - pancreas KW - diabetes KW - Thrombozyt KW - Insulinsekretion Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-313933 ER - TY - JOUR A1 - Nishida Xavier da Silva, Thamara A1 - Schulte, Clemens A1 - Nunes Alves, Ariane A1 - Maric, Hans Michael A1 - Friedmann Angeli, José Pedro T1 - Molecular characterization of AIFM2/FSP1 inhibition by iFSP1-like molecules JF - Cell Death & Disease N2 - Ferroptosis is a form of cell death characterized by phospholipid peroxidation, where numerous studies have suggested that the induction of ferroptosis is a therapeutic strategy to target therapy refractory cancer entities. Ferroptosis suppressor protein 1 (FSP1), an NAD(P)H-ubiquinone reductase, is a key determinant of ferroptosis vulnerability, and its pharmacological inhibition was shown to strongly sensitize cancer cells to ferroptosis. A first generation of FSP1 inhibitors, exemplified by the small molecule iFSP1, has been reported; however, the molecular mechanisms underlying inhibition have not been characterized in detail. In this study, we explore the species-specific inhibition of iFSP1 on the human isoform to gain insights into its mechanism of action. Using a combination of cellular, biochemical, and computational methods, we establish a critical contribution of a species-specific aromatic architecture that is essential for target engagement. The results described here provide valuable insights for the rational development of second-generation FSP1 inhibitors combined with a tracer for screening the druggable pocket. In addition, we pose a cautionary notice for using iFSP1 in animal models, specifically murine models. KW - cell biology KW - chemical libraries Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-357943 VL - 14 ER - TY - JOUR A1 - Wiessler, Anna-Lena A1 - Talucci, Ivan A1 - Piro, Inken A1 - Seefried, Sabine A1 - Hörlin, Verena A1 - Baykan, Betül B. A1 - Tüzün, Erdem A1 - Schaefer, Natascha A1 - Maric, Hans M. A1 - Sommer, Claudia A1 - Villmann, Carmen T1 - Glycine receptor β–targeting autoantibodies contribute to the pathology of autoimmune diseases JF - Neurology: Neuroimmunology & Neuroinflammation N2 - Background and Objectives Stiff-person syndrome (SPS) and progressive encephalomyelitis with rigidity and myoclonus (PERM) are rare neurologic disorders of the CNS. Until now, exclusive GlyRα subunit–binding autoantibodies with subsequent changes in function and surface numbers were reported. GlyR autoantibodies have also been described in patients with focal epilepsy. Autoimmune reactivity against the GlyRβ subunits has not yet been shown. Autoantibodies against GlyRα1 target the large extracellular N-terminal domain. This domain shares a high degree of sequence homology with GlyRβ making it not unlikely that GlyRβ-specific autoantibody (aAb) exist and contribute to the disease pathology. Methods In this study, we investigated serum samples from 58 patients for aAb specifically detecting GlyRβ. Studies in microarray format, cell-based assays, and primary spinal cord neurons and spinal cord tissue immunohistochemistry were performed to determine specific GlyRβ binding and define aAb binding to distinct protein regions. Preadsorption approaches of aAbs using living cells and the purified extracellular receptor domain were further used. Finally, functional consequences for inhibitory neurotransmission upon GlyRβ aAb binding were resolved by whole-cell patch-clamp recordings. Results Among 58 samples investigated, cell-based assays, tissue analysis, and preadsorption approaches revealed 2 patients with high specificity for GlyRβ aAb. Quantitative protein cluster analysis demonstrated aAb binding to synaptic GlyRβ colocalized with the scaffold protein gephyrin independent of the presence of GlyRα1. At the functional level, binding of GlyRβ aAb from both patients to its target impair glycine efficacy. Discussion Our study establishes GlyRβ as novel target of aAb in patients with SPS/PERM. In contrast to exclusively GlyRα1-positive sera, which alter glycine potency, aAbs against GlyRβ impair receptor efficacy for the neurotransmitter glycine. Imaging and functional analyses showed that GlyRβ aAbs antagonize inhibitory neurotransmission by affecting receptor function rather than localization. KW - autoantibody (aAb) KW - glycine receptor (GlyR) KW - stiff-person syndrome (SPS) KW - clinical neurology KW - movement disorders KW - progressive encephalitis with rigidity and myoclonus (PERM) Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-349958 VL - 11 IS - 2 ER - TY - THES A1 - Weigel [verh. Hoffmann], Mathis Leonard T1 - Thrombozytenfunktionsanalyse als potenzielles Instrument zur Früherkennung von Sepsis T1 - Platelet function analysis as a potential tool for early sepsis diagnosis N2 - Sepsis ist ein häufiges und akut lebensbedrohliches Syndrom, das eine Organfunktionsstörung in Folge einer dysregulierten Immunantwort auf eine Infektion beschreibt. Eine frühzeitige Diagnosestellung und Therapieeinleitung sind von zentraler Bedeutung für das Überleben der Patient:innen. In einer Pilotstudie konnte unsere Forschungsgruppe mittels Durchflusszytometrie eine ausgeprägte Hyporeaktivität der Thrombozyten bei Sepsis nachweisen, die einen potenziell neuen Biomarker zur Sepsis-Früherkennung darstellt. Zur Evaluation des Ausmaßes und Entstehungszeitpunktes der detektierten Thrombozytenfunktionsstörung wurden im Rahmen der vorliegenden Arbeit zusätzlich zu Patient:innen mit Sepsis (SOFA-Score ≥ 2; n=13) auch hospitalisierte Patient:innen mit einer Infektion ohne Sepsis (SOFA-Score < 2; n=12) rekrutiert. Beide Kohorten wurden zu zwei Zeitpunkten (t1: <24h; t2: Tag 5-7) im Krankheitsverlauf mittels Durchflusszytometrie und PFA-200 untersucht und mit einer gesunden Kontrollgruppe (n=28) verglichen. Phänotypische Auffälligkeiten der Thrombozyten bei Sepsis umfassten: (i) eine veränderte Expression verschiedener Untereinheiten des GPIb-IX-V-Rezeptorkomplexes, die auf ein verstärktes Rezeptor-Shedding hindeutet; (ii) ein ausgeprägtes Mepacrin-Beladungsdefizit, das auf eine zunehmend reduzierte Anzahl von δ-Granula entlang des Infektion-Sepsis Kontinuums hinweist; (iii) eine Reduktion endständig gebundener Sialinsäure im Sinne einer verstärkten Desialylierung. Die funktionelle Analyse der Thrombozyten bei Sepsis ergab bei durchflusszytometrischer Messung der Integrin αIIbβ3-Aktivierung (PAC-1-Bindung) eine ausgeprägte generalisierte Hyporeaktivität gegenüber multiplen Agonisten, die abgeschwächt bereits bei Infektion nachweisbar war und gemäß ROC-Analysen gut zwischen Infektion und Sepsis diskriminierte (AUC >0.80 für alle Agonisten). Im Gegensatz dazu zeigten Thrombozyten bei Sepsis und Analyse mittels PFA-200 unter Einfluss physiologischer Scherkräfte eine normale bis gar beschleunigte Aggregation. Die Reaktivitätsmessung von Thrombozyten mittels Durchflusszytometrie stellt weiterhin einen vielversprechenden Biomarker für die Sepsis-Früherkennung dar. Für weitere Schlussfolgerungen ist jedoch eine größere Kohorte erforderlich. In nachfolgenden Untersuchungen sollten zudem mechanistische Ursachen der beschriebenen phänotypischen und funktionellen Auffälligkeiten von Thrombozyten bei Infektion und Sepsis z.B. mittels Koinkubationsexperimenten untersucht werden. N2 - Sepsis is a frequent and life-threatening condition that describes organ dysfunction resulting from a dysregulated host immune response to infection. Early diagnosis and treatment are essential to improve patient survival. In a previous pilot study with sepsis patients, our research identified a severe platelet hyporeactivity using flow cytometry which could become a potential new biomarker for early sepsis diagnosis. To evaluate onset and extend of the detected platelet dysfunction in this study, we extended our patient cohort in addition to sepsis (SOFA-score ≥2; n=13) also to hospitalized patients with infection without sepsis (SOFA-score <2; n=12). Both cohorts were assessed at two time points during the disease (t1: <24h; t2: day 5-7) by flow cytometry and PFA-200 and compared with a healthy control group (n=28). Platelet phenotypic abnormalities during sepsis included: (i) altered expression of subunits of the GPIb-IX-V receptor complex, pointing to increased receptor shedding; (ii) a severe mepacrine loading deficit, indicating an increasingly reduced number of δ-granules along the infection-sepsis continuum; (iii) a reduction of terminally bound sialic acid, suggesting increased desialylation. Functional analysis of platelets in sepsis revealed a marked and generalized hyporeactivity toward multiple agonists when integrin αIIbβ3 activation (PAC-1 binding) was measured by flow cytometry, which was already to a lesser extend present in patients with infection and discriminated well between infection and sepsis according to ROC analysis (AUC >0.80 for all agonists). In contrast, platelets from septic patients showed normal to even accelerated aggregation when measured under flow condition and physiological shear forces by PFA-200. Analysis of platelet reactivity by flow cytometry remains a promising biomarker for early sepsis detection, but a larger cohort is needed for further conclusions. In subsequent studies, mechanistic causes of the described alterations in platelet phenotype and function during infection and sepsis should be investigated, e.g. by means of co-incubation experiments. KW - Sepsis KW - Thrombozyt KW - Biomarker KW - Frühdiagnostik KW - Durchflusscytometrie KW - Thrombozytenfunktionsanalyse Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-358193 ER - TY - THES A1 - Neagoe, Raluca Alexandra Iulia T1 - Development of techniques for studying the platelet glycoprotein receptors GPVI and GPIb localisation and signalling T1 - Entwicklung von Methoden zur Untersuchung zur der Lokalisation und Signaltransduktion der Thrombozytenrezeptoren GPVI und GPIb N2 - Platelets play an important role in haemostasis by mediating blood clotting at sites of blood vessel damage. Platelets, also participate in pathological conditions including thrombosis and inflammation. Upon vessel damage, two glycoprotein receptors, the GPIb-IX-V complex and GPVI, play important roles in platelet capture and activation. GPIb-IX-V binds to von Willebrand factor and GPVI to collagen. This initiates a signalling cascade resulting in platelet shape change and spreading, which is dependent on the actin cytoskeleton. This thesis aimed to develop and implement different super-resolution microscopy techniques to gain a deeper understanding of the conformation and location of these receptors in the platelet plasma membrane, and to provide insights into their signalling pathways. We suggest direct stochastic optical reconstruction microscopy (dSTORM) and structured illumination microscopy (SIM) as the best candidates for imaging single platelets, whereas expansion microscopy (ExM) is ideal for imaging platelets aggregates. Furthermore, we highlighted the role of the actin cytoskeleton, through Rac in GPVI signalling pathway. Inhibition of Rac, with EHT1864 in human platelets induced GPVI and GPV, but not GPIbα shedding. Furthermore, EHT1864 treatment did not change GPVI dimerisation or clustering, however, it decreased phospholipase Cγ2 phosphorylation levels, in human, but not murine platelets, highlighting interspecies differences. In summary, this PhD thesis demonstrates that; 1) Rac alters GPVI signalling pathway in human but not mouse platelets; 2) our newly developed ExM protocol can be used to image platelet aggregates labelled with F(ab’) fragments N2 - Thrombozyten, spielen in der Hämostase eine entscheidende Rolle, indem sie die Blutstillung bei Gefäßverletzung vermitteln. Sie sind jedoch auch an pathologischen Prozessen wie zum Beispiel der Thrombose und Entzündungen beteiligt. Bei einer Gefäßverletzung spielen zwei Glykoproteinrezeptoren eine wichtige Rolle bei der Adhäsion und Aktivierung von Thrombozyten: der GPIb-IX-V-Komplex und GPVI. GPIb-IX-V bindet an den von-Willebrand-Faktor und GPVI an Kollagen. Dies initiiert eine Signalkaskade, die zu einer Änderung der Morphologie der Thrombozyten führt, welche vom Aktin-Zytoskelett abhängig ist. Ziel dieser Doktorarbeit war die Entwicklung und Anwendung verschiedener hochauflösender Mikroskopietechniken, um ein tieferes Verständnis der Konformation und Lokalisation dieser Rezeptoren in der Plasmamembran der Thrombozyten zu erlangen und Einblicke in ihre Signalwege zu gewinnen. Hierbei etablierten wir dSTORM und die structured illumination microscopy (SIM) als die geeignetsten Methoden für die mikroskopische Untersuchung einzelner Thrombozyten, während die Expansionsmikroskopie (ExM) ideal für die Darstellung von Thrombozytenaggregaten ist. Darüber heben unsere Ergebnisse zur Funktion von Rac im GPVI Signalweg die wichtige Rolle des Aktin-Zytoskeletts hervor. Die Hemmung von Rac mit EHT1864 in menschlichen Thrombozyten induzierte das Abscheiden (shedding) von GPVI und GPV, nicht jedoch von GPIbα. Darüber hinaus blieb die GPVI Dimerisierung und GPVI-Clusterbildung durch EHT1864-Behandlung unverändert, jedoch verringerte sich die Phosphorylierung der Phospholipase Cγ2 in humanen, aber nicht in murinen Thrombozyten, was Unterschiede zwischen den Spezies aufzeigt. Zusammenfassend zeigen die Ergebnisse dieser Doktorarbeit, dass; 1) Rac den GPVI Signalweg in humanen aber nicht in murinen Thrombozyten beeinflusst; 2) unser neu entwickeltes ExM-Protokoll zur Darstellung von F(ab’)-Fragment markierten Thrombozytenaggregaten verwendet werden kann. KW - Platelet-Membranglykoprotein p62 KW - Platelets KW - Microscopy KW - GPVI KW - Rac1 Y1 - 2024 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-313064 ER - TY - JOUR A1 - Rasmussen, Tim T1 - The potassium efflux system Kef: bacterial protection against toxic electrophilic compounds JF - Membranes N2 - Kef couples the potassium efflux with proton influx in gram-negative bacteria. The resulting acidification of the cytosol efficiently prevents the killing of the bacteria by reactive electrophilic compounds. While other degradation pathways for electrophiles exist, Kef is a short-term response that is crucial for survival. It requires tight regulation since its activation comes with the burden of disturbed homeostasis. Electrophiles, entering the cell, react spontaneously or catalytically with glutathione, which is present at high concentrations in the cytosol. The resulting glutathione conjugates bind to the cytosolic regulatory domain of Kef and trigger activation while the binding of glutathione keeps the system closed. Furthermore, nucleotides can bind to this domain for stabilization or inhibition. The binding of an additional ancillary subunit, called KefF or KefG, to the cytosolic domain is required for full activation. The regulatory domain is termed K+ transport–nucleotide binding (KTN) or regulator of potassium conductance (RCK) domain, and it is also found in potassium uptake systems or channels in other oligomeric arrangements. Bacterial RosB-like transporters and K+ efflux antiporters (KEA) of plants are homologs of Kef but fulfill different functions. In summary, Kef provides an interesting and well-studied example of a highly regulated bacterial transport system. KW - potassium homeostasis KW - monovalent cation:proton antiporter-2 (CPA2) family KW - gram-negative bacteria KW - stress response KW - RCK domain KW - KEA Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-313686 SN - 2077-0375 VL - 13 IS - 5 ER - TY - JOUR A1 - Goeritzer, Madeleine A1 - Kuentzel, Katharina B. A1 - Beck, Sarah A1 - Korbelius, Melanie A1 - Rainer, Silvia A1 - Bradić, Ivan A1 - Kolb, Dagmar A1 - Mussbacher, Marion A1 - Schrottmaier, Waltraud C. A1 - Assinger, Alice A1 - Schlagenhauf, Axel A1 - Rost, René A1 - Gottschalk, Benjamin A1 - Eichmann, Thomas O. A1 - Züllig, Thomas A1 - Graier, Wolfgang F. A1 - Vujić, Nemanja A1 - Kratky, Dagmar T1 - Monoglyceride lipase deficiency is associated with altered thrombogenesis in mice JF - International Journal of Molecular Sciences N2 - Monoglyceride lipase (MGL) hydrolyzes monoacylglycerols (MG) to glycerol and one fatty acid. Among the various MG species, MGL also degrades 2-arachidonoylglycerol, the most abundant endocannabinoid and potent activator of the cannabinoid receptors 1 and 2. We investigated the consequences of MGL deficiency on platelet function using systemic (Mgl\(^{−/−}\)) and platelet-specific Mgl-deficient (platMgl\(^{−/−}\)) mice. Despite comparable platelet morphology, loss of MGL was associated with decreased platelet aggregation and reduced response to collagen activation. This was reflected by reduced thrombus formation in vitro, accompanied by a longer bleeding time and a higher blood volume loss. Occlusion time after FeCl\(_3\)-induced injury was markedly reduced in Mgl\(^{−/−}\) mice, which is consistent with contraction of large aggregates and fewer small aggregates in vitro. The absence of any functional changes in platelets from platMgl\(^{−/−}\) mice is in accordance with lipid degradation products or other molecules in the circulation, rather than platelet-specific effects, being responsible for the observed alterations in Mgl\(^{−/−}\) mice. We conclude that genetic deletion of MGL is associated with altered thrombogenesis. KW - platelets KW - MGL KW - in vitro and in vivo thrombus formation KW - platelet activation KW - platelet aggregation Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-304052 SN - 1422-0067 VL - 24 IS - 4 ER - TY - THES A1 - Brown, Helena Charlotte T1 - Investigating the role of the platelet receptor C-type lectin-like receptor 2 in models of thrombosis T1 - Untersuchungen zur Rolle des Thrombozytenrezeptors CLEC-2 (C- type lectin-like receptor 2) in Thrombosemodellen N2 - Platelets have a key physiological role in haemostasis however, inappropriate thrombus formation can lead to cardiovascular diseases such as myocardial infarction or stroke. Although, such diseases are common worldwide there are comparatively few anti-platelet drugs, and these are associated with an increased risk of bleeding. Platelets also have roles in thrombo-inflammation, immuno-thrombosis and cancer, in part via C-type lectin-like receptor 2 (CLEC-2) and its ligand podoplanin. Although CLEC-2 contributes to these diseases in mice, as well as to thrombus stability, it is unclear whether CLEC-2 has similar roles in humans, particularly as human CLEC-2 (hCLEC-2) cannot be investigated experimentally in vivo. To investigate hCLEC-2 in vivo, we generated a humanised CLEC-2 mouse (hCLEC-2KI) model, as well as a novel monoclonal antibody, HEL1, that binds to a different site than an existing antibody, AYP1. Using these antibodies, we have provided proof of principle for the use of hCLEC-2KI mice to test potential therapeutics targeting hCLEC-2, and shown for the first time that hCLEC-2 can be immunodepleted, with little effect on haemostasis. However, our results have also suggested that there are species differences in the role of CLEC-2 in arterial thrombosis. We further confirmed this using human blood where blocking CLEC-2 ligand binding had no effect on thrombosis, whereas we confirmed a minor role for mouse CLEC-2 in thrombus stability. We also investigated the effect of blocking CLEC-2 signalling using the Bruton’s tyrosine kinase inhibitor PRN473 on CLEC-2 mediated immuno-thrombosis in a Salmonella typhimurium infection model. However, no effect on thrombosis was observed suggesting that CLEC-2 signalling is not involved. Overall, our results suggest that there may be differences in the role of human and mouse CLEC-2, at least in arterial thrombosis, which could limit the potential of CLEC-2 as an anti-thrombotic target. However, it appears that the interaction between CLEC-2 and podoplanin is conserved and therefore CLEC-2 could still be a therapeutic target in immuno-thrombosis, thrombo-inflammation and cancer. Furthermore, any potential human specific therapeutics could be investigated in vivo using hCLEC-2KI mice. N2 - Thrombozyten sind ein wichtiger Bestandteil der Hämostase, können allerdings durch die Bildung eines Blutgerinnsels auch kardiovaskuläre Krankheitsbilder wie Myokardinfarkte oder Schlaganfälle hervorrufen. Obwohl diese Erkrankungen weltweit zu den führenden Todesursachen zählen, gibt es vergleichsweise wenig Thrombozyteninhibitoren und die bislang verfügbaren Wirkstoffe gehen mit einem erhöhten Blutungsrisiko einher. Darüber hinaus spielen Thrombozyten auch bei thrombo-inflammatorischen oder malignen Erkrankungen eine Rolle und sind maßgeblich an Entzündungs-vermittelten Thrombosen (Immunothrombosen) beteiligt. Daten aus Mausmodellen legen nahe, dass die Interaktion zwischen dem Thrombozytenrezeptor CLEC-2 (C-type lectin-like receptor 2) und seinem Liganden Podoplanin von Bedeutung für diese Krankheitsbilder, und die Thrombusstabilität ist. Allerdings ist bislang unklar, ob CLEC-2 im Menschen eine ähnliche Rolle spielt, da die Rolle des menschlichen CLEC-2 (hCLEC-2) in diesen Prozessen bislang nicht experimentell in vivo erforscht werden kann. Um hCLEC-2 in vivo zu erforschen, haben wir Mäuse generiert, die humanes CLEC-2 exprimieren (hCLEC-2KI), sowie einen neuen, monoklonalen Antikörper (HEL1) entwickelt, der an eine andere Bindungsstelle als der zuvor generierter Antikörper (AYP1) bindet. Mit Hilfe dieser Antikörper haben wir erstmalig gezeigt, dass hCLEC-2KI Mäuse geeignet sind, um potenzielle Therapeutika zu testen, die auf hCLEC-2 abzielen. Des Weiteren konnten wir erstmalig zeigen, dass auch hCLEC-2 immunodepletiert werden kann und dass der Verlust des Rezeptors in zirkulierenden Thrombozyten die Hämostase nur minimal beeinträchtigt. Allerdings deuten unsere Ergebnisse auch darauf hin, dass es hinsichtlich der Bedeutung CLEC-2 für die arterielle Thrombose artspezifische Unterschiede gibt: Während Maus CLEC-2 zur Stabilität der Thromben beiträgt, hatte die Blockade der Ligandenbindungsstelle von hCLEC-2 keinen Einfluss auf Thrombose. Des Weiteren wurde mit Hilfe des Bruton’s tyrosine kinase Inhibitors PRN473 der Effekt einer Blockierung des CLEC-2 Signalwegs auf die durch CLEC-2 hervorgerufene Immuno-Thrombose in einem Salmonella typhimurium Infektionsmodel erforscht. Da jedoch keine Effekte nachgewiesen werde konnten, schlussfolgern wir, dass der CLEC-2 Signalweg nicht in diesen Prozess involviert ist. Insgesamt deuten unsere Ergebnisse darauf hin, dass es Unterschiede in der Rolle von CLEC-2 zwischen Mensch und Maus gibt, zumindest im Kontext der arteriellen Thrombose, was das Potenzial von CLEC-2 als antithrombotisches Ziel einschränken könnte. Da allem Anschein nach die Interaktion zwischen CLEC-2 und Podoplanin konserviert ist, könnte CLEC-2 dennoch als Therapeutikum für Thrombo-Inflammation, Immunothrombose und Krebsbildungen genutzt werden. Des Weiteren könnten für den Menschen entwickelte Therapieansätze mit Hilfe von hCLEC-2KI Mäusen in vivo untersucht werden. KW - Thrombozyt KW - Thrombose KW - Platelet KW - Thrombosis KW - Rezeptor Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-293108 ER - TY - JOUR A1 - Schwan, Carsten A1 - Lang, Alexander E. A1 - Schlosser, Andreas A1 - Fujita-Becker, Setsuko A1 - AlHaj, Abdulatif A1 - Schröder, Rasmus R. A1 - Faix, Jan A1 - Aktories, Klaus A1 - Mannherz, Hans Georg T1 - Inhibition of Arp2/3 complex after ADP-ribosylation of Arp2 by binary Clostridioides toxins JF - Cells N2 - Clostridioides bacteria are responsible for life threatening infections. Here, we show that in addition to actin, the binary toxins CDT, C2I, and Iota from Clostridioides difficile, botulinum, and perfrigens, respectively, ADP-ribosylate the actin-related protein Arp2 of Arp2/3 complex and its additional components ArpC1, ArpC2, and ArpC4/5. The Arp2/3 complex is composed of seven subunits and stimulates the formation of branched actin filament networks. This activity is inhibited after ADP-ribosylation of Arp2. Translocation of the ADP-ribosyltransferase component of CDT toxin into human colon carcinoma Caco2 cells led to ADP-ribosylation of cellular Arp2 and actin followed by a collapse of the lamellipodial extensions and F-actin network. Exposure of isolated mouse colon pieces to CDT toxin induced the dissolution of the enterocytes leading to luminal aggregation of cellular debris and the collapse of the mucosal organization. Thus, we identify the Arp2/3 complex as hitherto unknown target of clostridial ADP-ribosyltransferases. KW - actin KW - ADP-ribosyltransferases KW - Arp2/3 complex KW - Clostridioides binary toxins Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-297454 SN - 2073-4409 VL - 11 IS - 22 ER - TY - THES A1 - Pacios Michelena, Anabel T1 - Molecular insights into the complex formed by the actin cytoskeleton related protein VASP and the inhibitory postsynaptic scaffolding protein gephyrin T1 - Molekulare Einblicke in den Komplex, der durch das mit dem Aktin-Zytoskelett verwandte Protein VASP und Gephyrin, einem Gerüstprotein inhibitorischer postsynaptischer Strukturen, gebildet wird N2 - Gephyrin is a 93 kDa moonlighting protein, which is involved in the last two steps of the molybdenum cofactor (Moco) biosynthesis pathway while at the same time playing a central role in the anchoring, clustering and stabilization of glycine receptors (GlyRs) ... N2 - Gephyrin ist ein multifunktionales 93 kDa-Protein. Dieses Protein katalysiert die letzten beiden Schritte des Biosynthesewegs des Molybdän-Cofaktors (Moco). Gleichzeitig spielt es eine zentrale Rolle bei der Verankerung, Clusterbildung und Stabilisierung sowohl von Glycinrezeptoren (GlyRs) als auch von ... KW - gephyrin KW - vasp KW - Inhibitory-postsynapse KW - Actin cytoskeleton-related protein Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-213373 ER - TY - JOUR A1 - Bieber, Michael A1 - Schuhmann, Michael K. A1 - Bellut, Maximilian A1 - Stegner, David A1 - Heinze, Katrin G. A1 - Pham, Mirko A1 - Nieswandt, Bernhard A1 - Stoll, Guido T1 - Blockade of platelet glycoprotein Ibα augments neuroprotection in Orai2-deficient mice during middle cerebral artery occlusion JF - International Journal of Molecular Sciences N2 - During ischemic stroke, infarct growth before recanalization diminishes functional outcome. Hence, adjunct treatment options to protect the ischemic penumbra before recanalization are eagerly awaited. In experimental stroke targeting two different pathways conferred protection from penumbral tissue loss: (1) enhancement of hypoxic tolerance of neurons by deletion of the calcium channel subunit Orai2 and (2) blocking of detrimental lymphocyte–platelet responses. However, until now, no preclinical stroke study has assessed the potential of combining neuroprotective with anti-thrombo-inflammatory interventions to augment therapeutic effects. We induced focal cerebral ischemia in Orai2-deficient (Orai2\(^{-/-}\)) mice by middle cerebral artery occlusion (MCAO). Animals were treated with anti-glycoprotein Ib alpha (GPIbα) Fab fragments (p0p/B Fab) blocking GPIbα–von Willebrand factor (vWF) interactions. Rat immunoglobulin G (IgG) Fab was used as the control treatment. The extent of infarct growth before recanalization was assessed at 4 h after MCAO. Moreover, infarct volumes were determined 6 h after recanalization (occlusion time: 4 h). Orai2 deficiency significantly halted cerebral infarct progression under occlusion. Inhibition of platelet GPIbα further reduced primary infarct growth in Orai2\(^{-/-}\) mice. During ischemia–reperfusion, upon recanalization, mice were likewise protected. All in all, we show that neuroprotection in Orai2\(^{-/-}\) mice can be augmented by targeting thrombo-inflammation. This supports the clinical development of combined neuroprotective/anti-platelet strategies in hyper-acute stroke. KW - ischemic penumbra KW - Orai2 KW - glycoprotein receptor Ibα KW - ischemic stroke KW - thrombo-inflammation KW - middle cerebral artery occlusion Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-286038 SN - 1422-0067 VL - 23 IS - 16 ER - TY - JOUR A1 - Diebold, Mathias A1 - Schönemann, Lars A1 - Eilers, Martin A1 - Sotriffer, Christoph A1 - Schindelin, Hermann T1 - Crystal structure of a covalently linked Aurora-A-MYCN complex JF - Acta Crystallographica N2 - Formation of the Aurora-A–MYCN complex increases levels of the oncogenic transcription factor MYCN in neuroblastoma cells by abrogating its degradation through the ubiquitin proteasome system. While some small-molecule inhibitors of Aurora-A were shown to destabilize MYCN, clinical trials have not been satisfactory to date. MYCN itself is considered to be `undruggable' due to its large intrinsically disordered regions. Targeting the Aurora-A–MYCN complex rather than Aurora-A or MYCN alone will open new possibilities for drug development and screening campaigns. To overcome the challenges that a ternary system composed of Aurora-A, MYCN and a small molecule entails, a covalently cross-linked construct of the Aurora-A–MYCN complex was designed, expressed and characterized, thus enabling screening and design campaigns to identify selective binders. KW - MYCNv KW - neuroblastoma cell KW - proteasome system Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-318855 VL - D79 SP - 1 EP - 9 ER - TY - THES A1 - Göb [née Klaus], Vanessa Aline Domenica T1 - Pathomechanisms underlying ischemic stroke T1 - Pathomechanismen des ischämischen Schlaganfalles N2 - Every year, stroke affects over 100 million people worldwide and the number of cases continues to grow. Ischemic stroke is the most prevalent form of stroke and rapid restoration of blood flow is the primary therapeutic aim. However, recanalization might fail or reperfusion itself induces detrimental processes leading to infarct progression. Previous studies identified platelets and immune cells as drivers of this so-called ischemia/reperfusion (I/R) injury, establishing the concept of ischemic stroke as thrombo-inflammatory disease. Reduced cerebral blood flow despite recanalization promoted the hypothesis that thrombus formation within the cerebral microcirculation induces further tissue damage. The results presented in this thesis refute this: using complementary methodologies, it was shown that infarct growth precedes the occurrence of thrombi excluding them as I/R injury-underlying cause. Blood brain barrier disruption is one of the hallmarks of ischemic stroke pathology and was confirmed as early event during reperfusion injury in the second part of this study. Abolished platelet α-granule release protects mice from vascular leakage in the early reperfusion phase resulting in smaller infarcts. Using in vitro assays, platelet α-granule-derived PDGF-AB was identified as one factor contributing to blood-brain barrier disruption. In vivo visualization of platelet activation would provide important insights in the spatio-temporal context of platelet activation in stroke pathology. As platelet signaling results in elevated intracellular Ca2+ levels, this is an ideal readout. To overcome the limitations of chemical calcium indicators, a mouse line expressing an endogenous calcium reporter specifically in platelets and megakaryocytes was generated. Presence of the reporter did not interfere with platelet function, consequently these mice were characterized in in vivo and ex vivo models. Upon ischemic stroke, neutrophils are among the first cells that are recruited to the brain. Since for neutrophils both, beneficial and detrimental effects are described, their role was investigated within this thesis. Neither neutrophil depletion nor absence of NADPH-dependent ROS production (Ncf-/- mice) affected stroke outcome. In contrast, abolished NET-formation in Pad4-/- mice resulted in reduced infarct sizes, revealing detrimental effects of NETosis in the context of ischemic stroke, which might become a potential therapeutic target. Cerebral venous (sinus) thrombosis, CV(S)T is a rare type of stroke with mainly idiopathic onset. Whereas for arterial thrombosis a critical contribution of platelets is known and widely accepted, for venous thrombosis this is less clear but considered more and more. In the last part of this thesis, it was shown that fab-fragments of the anti-CLEC-2 antibody INU1 trigger pathological platelet activation in vivo, resulting in foudroyant CVT accompanied by heavy neurological symptoms. Using this novel animal model for CVT, cooperative signaling of the two platelet receptors CLEC-2 and GPIIb/IIIa was revealed as major trigger of CVT and potential target for treatment. N2 - Jährlich sind weltweit über 100 Millionen Menschen von einem Schlaganfall betroffen, wobei die Zahl der Fälle weiter zunimmt. Der ischämische Schlaganfall ist die häufigste Form des Schlaganfalls, und die sofortige Wiederherstellung des Blutflusses ist das oberste Therapie¬ziel. Allerdings kommt es vor, dass die Rekanalisierung des betroffenen Gefäßes fehlschlägt oder die Reperfusion selbst zu schädlichen Prozessen führt, die das Fortschreiten des Infarkts begünstigen. In vorangegangen Studien wurden Thrombozyten und Immunzellen als treibende Kräfte dieser so genannten Ischämie/Reperfusion (I/R)-Schädigung identifiziert und der ischämische Schlaganfall als thrombo-inflammatorische Erkrankung definiert. Eine verminderte zerebrale Durchblutung trotz Rekanalisation führte zu der Hypothese, dass die Bildung von Thromben in der zerebralen Mikrozirkulation zu weiteren Gewebeschäden führt. Die hier vorgestellten Ergebnisse widerlegen dies: Mit Hilfe komplementärer Methoden konnte gezeigt werden, dass das Infarktwachstum dem Auftreten von Thromben vorausgeht, was diese als Ursache für die I/R-Verletzung ausschließt. Die Störung der Blut-Hirn-Schranke ist eines der charakteristischen Kennzeichen der Pathologie des ischämischen Schlaganfalls und wurde im zweiten Teil dieser Studie als frühes Ereignis während des Reperfusionsschadens bestätigt. Mit Hilfe transgener Mäuse konnte gezeigt werden, dass die Ausschüttung von α-Granula aus Thrombozyten in der frühen Reperfusionsphase an Störungen der Blut-Hirn-Schranke beteiligt ist und somit zum Infarktwachstum beiträgt. In in-vitro-Versuchen konnte gezeigt werden, dass PDGF-AB, ein Bestandteil der α-Granula, an Prozessen, die für die Beeinträchtigung der Blut-Hirn-Schranke verantwortlich sind, beteiligt ist. Die Sichtbarmachung von aktivierten Thrombozyten in vivo, würde wichtige Erkenntnisse über den räumlichen und zeitlichen Kontext der Aktivierung von Thrombozyten im Verlauf des Schlaganfalls liefern. Da alle aktivierenden Signalwege zum Anstieg des intrazellulären Kalziumspiegels führen, ist Kalzium ein idealer Indikator der Thrombozytenaktivierung. Um die Grenzen chemischer Kalziumindikatoren zu überwinden, wurde eine transgene Mauslinie erzeugt, welche einen endogenen Kalziumreporter speziell in Thrombozyten und Megakaryozyten exprimiert. Die Anwesenheit des Reporters hatte keine Auswirkung auf die Funktionalität der Thrombozyten und die Mäuse wurden in vivo sowie ex vivo in verschiedenen Experimenten charakterisiert. In der Folge eines ischämischen Schlaganfalles gehören Neutrophile zu den am frühesten ins Gehirn einwandernden Zellen. Dabei werden Neutrophilen sowohl günstige als auch schädliche Wirkungen auf den Verlauf des ischämischen Schlaganfalls zugeschrieben. Aus diesem Grund wurde ihre Rolle in dieser Arbeit näher untersucht. Weder die Abwesenheit von Neutrophilen noch das Fehlen der NADPH-abhängigen Produktion von reaktiven Sauerstoffspezies (Ncf1-/- Mäuse) beeinflussen den Ausgang eines Schlaganfalls. Im Gegensatz dazu, führte die Verhinderung der NET-Bildung (NET = neutrophil extracellular traps) in Pad4-/- Mäusen zu verringerten Infarktgrößen, was auf eine schädliche Wirkung der NETose im Zusammenhang des Schlaganfalls hinweist und somit ein therapeutisches Angriffsziel darstellen könnte. Sinusvenenthrombosen sind eine seltene Form des Schlaganfalls, die meist ohne bekannte Ursache auftreten. Während für die arterielle Thrombose ein kritischer Beitrag der Thrombozyten bekannt und weithin akzeptiert ist, ist dies für venöse Thrombosen weniger klar, wird aber immer mehr in Betracht gezogen. Im letzten Teil dieser Arbeit wurde gezeigt, dass Fab-Fragmente des anti-CLEC-2-Antikörpers INU1 in vivo eine pathologische Aktivierung von Thrombozyten auslösen, die zu einer fulminanten Sinusvenenthrombose mit schweren neurologischen Symptomen führt. Mit Hilfe dieses neuartigen Tiermodells wurde die zusammenwirkende Signalübertragung der beiden Thrombozytenrezeptoren CLEC-2 und GPIIb/IIIa als Hauptauslöser der Sinusvenenthrombose und damit potenzielles Ziel für eine Behandlung identifiziert. KW - Schlaganfall KW - Thrombozyt KW - Maus KW - Blut-Hirn-Schranke KW - Sinusthrombose KW - thrombo-inflammation KW - ischemic stroke KW - blood brain barrier KW - CVT KW - platelets Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-286727 ER - TY - JOUR A1 - Neagoe, Raluca A. I. A1 - Gardiner, Elizabeth E. A1 - Stegner, David A1 - Nieswandt, Bernhard A1 - Watson, Steve P. A1 - Poulter, Natalie S. T1 - Rac inhibition causes impaired GPVI signalling in human platelets through GPVI shedding and reduction in PLCγ2 phosphorylation JF - International Journal of Molecular Sciences N2 - Rac1 is a small Rho GTPase that is activated in platelets upon stimulation with various ligands, including collagen and thrombin, which are ligands for the glycoprotein VI (GPVI) receptor and the protease-activated receptors, respectively. Rac1-deficient murine platelets have impaired lamellipodia formation, aggregation, and reduced PLCγ2 activation, but not phosphorylation. The objective of our study is to investigate the role of Rac1 in GPVI-dependent human platelet activation and downstream signalling. Therefore, we used human platelets stimulated using GPVI agonists (collagen and collagen-related peptide) in the presence of the Rac1-specific inhibitor EHT1864 and analysed platelet activation, aggregation, spreading, protein phosphorylation, and GPVI clustering and shedding. We observed that in human platelets, the inhibition of Rac1 by EHT1864 had no significant effect on GPVI clustering on collagen fibres but decreased the ability of platelets to spread or aggregate in response to GPVI agonists. Additionally, in contrast to what was observed in murine Rac1-deficient platelets, EHT1864 enhanced GPVI shedding in platelets and reduced the phosphorylation levels of PLCγ2 following GPVI activation. In conclusion, Rac1 activity is required for both human and murine platelet activation in response to GPVI-ligands, but Rac1’s mode of action differs between the two species. KW - platelets KW - Rac1 KW - glycoprotein VI KW - EHT1864 KW - GPVI shedding KW - phospholipase C gamma 2 Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-284350 SN - 1422-0067 VL - 23 IS - 7 ER - TY - JOUR A1 - Koo, Chek Ziu A1 - Matthews, Alexandra L. A1 - Harrison, Neale A1 - Szyroka, Justyna A1 - Nieswandt, Bernhard A1 - Gardiner, Elizabeth E. A1 - Poulter, Natalie S. A1 - Tomlinson, Michael G. T1 - The platelet collagen receptor GPVI is cleaved by Tspan15/ADAM10 and Tspan33/ADAM10 molecular scissors JF - International Journal of Molecular Sciences N2 - The platelet-activating collagen receptor GPVI represents the focus of clinical trials as an antiplatelet target for arterial thrombosis, and soluble GPVI is a plasma biomarker for several human diseases. A disintegrin and metalloproteinase 10 (ADAM10) acts as a ‘molecular scissor’ that cleaves the extracellular region from GPVI and many other substrates. ADAM10 interacts with six regulatory tetraspanin membrane proteins, Tspan5, Tspan10, Tspan14, Tspan15, Tspan17 and Tspan33, which are collectively termed the TspanC8s. These are emerging as regulators of ADAM10 substrate specificity. Human platelets express Tspan14, Tspan15 and Tspan33, but which of these regulates GPVI cleavage remains unknown. To address this, CRISPR/Cas9 knockout human cell lines were generated to show that Tspan15 and Tspan33 enact compensatory roles in GPVI cleavage, with Tspan15 bearing the more important role. To investigate this mechanism, a series of Tspan15 and GPVI mutant expression constructs were designed. The Tspan15 extracellular region was found to be critical in promoting GPVI cleavage, and appeared to achieve this by enabling ADAM10 to access the cleavage site at a particular distance above the membrane. These findings bear implications for the regulation of cleavage of other ADAM10 substrates, and provide new insights into post-translational regulation of the clinically relevant GPVI protein. KW - ADAM10 KW - GPVI KW - tetraspanin KW - platelet KW - shedding KW - TspanC8 KW - metalloproteinase Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-284468 SN - 1422-0067 VL - 23 IS - 5 ER - TY - JOUR A1 - Schanbacher, Constanze A1 - Bieber, Michael A1 - Reinders, Yvonne A1 - Cherpokova, Deya A1 - Teichert, Christina A1 - Nieswandt, Bernhard A1 - Sickmann, Albert A1 - Kleinschnitz, Christoph A1 - Langhauser, Friederike A1 - Lorenz, Kristina T1 - ERK1/2 activity is critical for the outcome of ischemic stroke JF - International Journal of Molecular Sciences N2 - Ischemic disorders are the leading cause of death worldwide. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) are thought to affect the outcome of ischemic stroke. However, it is under debate whether activation or inhibition of ERK1/2 is beneficial. In this study, we report that the ubiquitous overexpression of wild-type ERK2 in mice (ERK2\(^{wt}\)) is detrimental after transient occlusion of the middle cerebral artery (tMCAO), as it led to a massive increase in infarct volume and neurological deficits by increasing blood–brain barrier (BBB) leakiness, inflammation, and the number of apoptotic neurons. To compare ERK1/2 activation and inhibition side-by-side, we also used mice with ubiquitous overexpression of the Raf-kinase inhibitor protein (RKIP\(^{wt}\)) and its phosphorylation-deficient mutant RKIP\(^{S153A}\), known inhibitors of the ERK1/2 signaling cascade. RKIP\(^{wt}\) and RKIP\(^{S153A}\) attenuated ischemia-induced damages, in particular via anti-inflammatory signaling. Taken together, our data suggest that stimulation of the Raf/MEK/ERK1/2-cascade is severely detrimental and its inhibition is rather protective. Thus, a tight control of the ERK1/2 signaling is essential for the outcome in response to ischemic stroke. KW - ERK1/2 KW - tMCAO KW - ischemic stroke KW - RKIP Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-283991 SN - 1422-0067 VL - 23 IS - 2 ER - TY - JOUR A1 - Navarro, Stefano A1 - Stegner, David A1 - Nieswandt, Bernhard A1 - Heemskerk, Johan W. M. A1 - Kuijpers, Marijke J. E. T1 - Temporal roles of platelet and coagulation pathways in collagen- and tissue factor-induced thrombus formation JF - International Journal of Molecular Sciences N2 - In hemostasis and thrombosis, the complex process of thrombus formation involves different molecular pathways of platelet and coagulation activation. These pathways are considered as operating together at the same time, but this has not been investigated. The objective of our study was to elucidate the time-dependency of key pathways of thrombus and clot formation, initiated by collagen and tissue factor surfaces, where coagulation is triggered via the extrinsic route. Therefore, we adapted a microfluidics whole-blood assay with the Maastricht flow chamber to acutely block molecular pathways by pharmacological intervention at desired time points. Application of the technique revealed crucial roles of glycoprotein VI (GPVI)-induced platelet signaling via Syk kinase as well as factor VIIa-induced thrombin generation, which were confined to the first minutes of thrombus buildup. A novel anti-GPVI Fab EMF-1 was used for this purpose. In addition, platelet activation with the protease-activating receptors 1/4 (PAR1/4) and integrin αIIbβ3 appeared to be prolongedly active and extended to later stages of thrombus and clot formation. This work thereby revealed a more persistent contribution of thrombin receptor-induced platelet activation than of collagen receptor-induced platelet activation to the thrombotic process. KW - coagulation KW - fibrin KW - glycoprotein VI KW - platelet receptors KW - spatiotemporal thrombus KW - thrombin Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-284219 SN - 1422-0067 VL - 23 IS - 1 ER - TY - THES A1 - Khayenko, Vladimir T1 - Functional peptide-based probes for the visualization of inhibitory synapses T1 - Funktionelle peptidbasierte Sonden zur Visualisierung von hemmenden Synapsen N2 - Short functional peptidic probes can maximize the potential of high-end microscopy techniques and multiplex imaging assays and provide new insights into normal and aberrant molecular, cellular and tissue function. Particularly, the visualization of inhibitory synapses requires protocol tailoring for different sample types and imaging techniques and relies either on genetic manipulation or on antibodies that underperform in tissue immunofluorescence. Starting from an endogenous activity-related ligand of gephyrin, a universal marker of the inhibitory post-synapse, I developed a short peptidic multivalent binder with exceptional affinity and selectivity to gephyrin. By tailoring fluorophores to the binder, I have obtained Sylite, a probe for the visualization of inhibitory synapses, with an outstanding signal-to-background ratio, that bests the “gold standard” gephyrin antibodies both in selectivity and in tissue immunofluorescence. In tissue Sylite benefits from simplified handling, provides robust synaptic labeling in record-short time and, unlike antibodies, is not affected by staining artefacts. In super-resolution microscopy Sylite precisely localizes the post-synapse and enables accurate pre- to post-synapse measurements. Combined with complimentary tracing techniques Sylite reveals inhibitory connectivity and profiles inhibitory inputs and synapse sizes of excitatory and inhibitory neurons in the periaqueductal gray brain region. Lastly, upon probe optimization for live cell application and with the help of novel thiol-reactive cell penetrating peptide I have visualized inhibitory synapses in living neurons. Taken together, my work provided a versatile probe for conventional and super-resolution microscopy and a workflow for the development and application of similar compact functional synthetic probes. N2 - Kurze funktionelle peptidische Sonden können das Potenzial von High-End-Mikroskopietechniken und Multiplex-Imaging-Assays maximieren und neue Erkenntnisse über normale und abweichende Molekulare-, Zelluläre- und Gewebefunktionen liefern. Insbesondere die Visualisierung inhibitorischer Synapsen erfordert eine Anpassung des Protokolls an verschiedene Probentypen und Bildgebungsverfahren und ist entweder auf genetische Manipulationen oder auf Antikörper angewiesen, die in der Gewebeimmunfluoreszenz unterdurchschnittlich abschneiden. Ausgehend von einem endogenen aktivitätsbezogenen Liganden von Gephyrin, einem universellen Marker der hemmenden Postsynapse, habe ich einen kurzen peptidischen multivalenten Binder mit außergewöhnlicher Affinität und Selektivität zu Gephyrin entwickelt. Durch die Anpassung von Fluorophoren an das Bindemittel habe ich Sylite erhalten, eine Sonde für die Visualisierung inhibitorischer Synapsen mit einem hervorragenden Signal-Hintergrund-Verhältnis, das die "Goldstandard"-Gephyrin-Antikörper sowohl in der Selektivität als auch in der Gewebe-Immunfluoreszenz übertrifft. Im Gewebe profitiert Sylite von einer vereinfachten Handhabung, bietet eine robuste synaptische Markierung in rekordverdächtig kurzer Zeit und wird im Gegensatz zu Antikörpern nicht durch Färbungsartefakte beeinträchtigt. In der Super-Resolution-Mikroskopie lokalisiert Sylite präzise die Post-Synapse und ermöglicht genaue Messungen von Prä- zu Postsynapse. In Kombination mit ergänzenden Tracing-Techniken deckt Sylite die hemmende Konnektivität auf und erstellt Profile der hemmenden Eingänge und Synapsengrößen von erregenden und hemmenden Neuronen in der periaquäduktalen Grau Hirnregion. Schließlich habe ich nach Optimierung der Sonde für die Anwendung in lebenden Zellen und mit Hilfe eines neuartigen thiolreaktiven zelldurchdringenden Peptids hemmende Synapsen in lebenden Neuronen visualisiert. Insgesamt lieferte meine Arbeit eine vielseitige Sonde für konventionelle und superauflösende Mikroskopie und einen Arbeitsablauf für die Entwicklung und Anwendung ähnlicher kompakter funktioneller synthetischer Sonden. KW - Fluoreszenzsonde KW - Peptidsynthese KW - Neurowissenschaften KW - Inhibitorische Synapse KW - Gephyrin KW - Peptide KW - Fluorescent probes KW - Neuroscience KW - Inhibitory synapse KW - Super-Resolution Microscopy KW - Tissue staining Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-320438 ER - TY - JOUR A1 - Vogelsang, Anna A1 - Eichler, Susann A1 - Huntemann, Niklas A1 - Masanneck, Lars A1 - Böhnlein, Hannes A1 - Schüngel, Lisa A1 - Willison, Alice A1 - Loser, Karin A1 - Nieswandt, Bernhard A1 - Kehrel, Beate E. A1 - Zarbock, Alexander A1 - Göbel, Kerstin A1 - Meuth, Sven G. T1 - Platelet inhibition by low-dose acetylsalicylic acid reduces neuroinflammation in an animal model of multiple sclerosis JF - International Journal of Molecular Sciences N2 - Aside from the established immune-mediated etiology of multiple sclerosis (MS), compelling evidence implicates platelets as important players in disease pathogenesis. Specifically, numerous studies have highlighted that activated platelets promote the central nervous system (CNS)-directed adaptive immune response early in the disease course. Platelets, therefore, present a novel opportunity for modulating the neuroinflammatory process that characterizes MS. We hypothesized that the well-known antiplatelet agent acetylsalicylic acid (ASA) could inhibit neuroinflammation by affecting platelets if applied at low-dose and investigated its effect during experimental autoimmune encephalomyelitis (EAE) as a model to study MS. We found that oral administration of low-dose ASA alleviates symptoms of EAE accompanied by reduced inflammatory infiltrates and less extensive demyelination. Remarkably, the percentage of CNS-infiltrated CD4\(^+\) T cells, the major drivers of neuroinflammation, was decreased to 40.98 ± 3.28% in ASA-treated mice compared to 56.11 ± 1.46% in control animals at the disease maximum as revealed by flow cytometry. More interestingly, plasma levels of thromboxane A\(_2\) were decreased, while concentrations of platelet factor 4 and glycoprotein VI were not affected by low-dose ASA treatment. Overall, we demonstrate that low-dose ASA could ameliorate the platelet-dependent neuroinflammatory response in vivo, thus indicating a potential treatment approach for MS. KW - acetylsalicylic acid KW - experimental autoimmune encephalomyelitis KW - platelets KW - multiple sclerosis KW - thromboxane KW - glycoprotein VI KW - platelet factor 4 Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-284535 SN - 1422-0067 VL - 22 IS - 18 ER - TY - THES A1 - Truongvan, Ngoc T1 - Understanding the dual specificity of UBA6 T1 - Einblick in die duale Spezifität von UBA6 N2 - Ubiquitylation is a protein post translational modification, in which ubiquitin is covalently attached to target protein substrates resulting in diverse cellular outcomes. Besides ubiquitin, various ubiquitin-like proteins including FAT10 exist, which are also conjugated to target proteins. The underlying modification mechanisms are conserved. In the initial step, ubiquitin or a ubiquitin-like protein is thioester-linked to a catalytic cysteine in the E1activating enzyme in an ATP-dependent manner. The respective protein modifier is then transferred to an E2 conjugating enzyme in a transthioesterification reaction. Finally, an E3 ubiquitin ligase E3 catalyzes the covalent attachment of the protein modifier to a substrate. In the case of ubiquitin, multiple ubiquitin molecules can be attached to a substrate in the form of either linear or branched polyubiquitin chains but also as single ubiquitin modifications. Depending on the nature of the ubiquitin chain, the substrates are destined to various cellular processes such as their targeted destruction by the proteasome but also non-degradative outcomes may occur. As stated above FAT10 is a ubiquitin-like protein modifier which typically targets proteins for proteasomal degradation. It consists of two ubiquitin-like domains and is mainly expressed in cells of the human immune system. The reported involvement of FAT10 modifications in cancers and other diseases has caught the attention of the scientific community as an inhibition of the FAT10ylation process may provide avenues for novel therapeutic approaches. UBA6 is the E1 activating enzyme that resides at the apex of the FAT10 proteasomal degradation pathway. UBA6 not only recognizes FAT10 but can also activate ubiquitin as efficiently as the ubiquitin specific E1 UBA1. The dual specificity of UBA6 may complicate the inhibition FAT10ylation since targeting the active site of UBA6 will also inhibit the UBA6-catalyzed ubiquitin activation. Therefore, it is important to understand the underlying principles for the dual specificity of UBA6 prior to the development of compounds interfering with FAT10ylation. In this thesis important novel insights into the structure and function of UBA6 were derived by X-ray crystallography and biochemical methods. The first crystal structure of UBA6 reveals the multidomain architecture of this enzyme in atomic detail. The enzyme is composed of a rigid core including its active and inactive adenylation domains as well as a 4 helix bundle. Overall, the molecule adopts a “Y” shape architecture with the core at the base and the first and second catalytic half domains forming one arm of the “Y” and the ubiquitin fold domain constituting the other arm. While UBA6 shares the same domain architecture as UBA1, substantial differences were revealed by the crystal structure. In particular, the first catalytic half domain undergoes a significant shift to a position more distal from the core. This rigid body movement is assumed to generate room to accommodate the second ubiquitin-like domain of FAT10. Differences are also observed in a hydrophobic platform between the core and the first catalytic half domain and the adenylation active site in the core, which together from the binding sites for ubiquitin and FAT10. Site directed mutagenesis of key residues in these areas altered the UBA6-catalyzed activation of ubiquitin and FAT10. UBA6 variants were generated with the goal of trying to block the activation of FAT10 while still maintaining that of ubiquitin activation, in order to fully explain the dual specificity of UBA6. However, none of these mutations could block the activation of FAT10, while some of these UBA6 variants blocked ubiquitin activation. Preliminary inhibition assays with a group of E1 inhibitors belonging to the adenosyl sulfamate family demonstrated potent inhibition of FAT10ylation for two compounds. The dual specificity of UBA6 hence needs to be further examined by biochemical and structural methods. In particular, the structure of a complex between UBA6 and ubiquitin or FAT10 would provide key insights for further biochemical studies, ultimately allowing the targeted inhibition of the FAT10ylation machinery. N2 - Der Prozess der Ubiquitinierung stellt eine posttranslationale Modifikation dar, bei der das kleine Protein Ubiquitin kovalent an ein Zielprotein angehängt wird, was zu verschiedenen zellulären Effekten führt. Neben Ubiquitin existieren verschiedene ubiquitinähnliche Proteine, wie z.B. FAT10, die an Zielproteine angehängt werden können. Die der Modifikation zugrunde liegenenden Mechanismen der Proteinmodifikation sind konserviert. Im ersten Schritt wird Ubiquitin oder das ubiquitinähnliche Protein in einer ATP-abhängigen Reaktion kovalent an das katalytische Cystein des aktivierenden Enzyms (E1) gebunden. Danach wird es durch Transthioestherifizierung an ein konjugierendes Enzym (E2) übertagen und schließlich durch eine Ligase (E3) kovalent an das Substrat gehängt. Ubiquitin kann entweder einzeln oder in Form linearer oder verzweigter Ketten an ein Substrat angehängt werden, was wiederum zu verschiedenen funktionalen Konsequenzen wie dem Abbau das Proteins durch das Proteasom führen kann. Wie schon erwähnt ist FAT10 ein ubiquitinähliches Protein, das üblicherweise Zielproteine für den Abbau durch das Proteasom markiert. Es besteht aus zwei ubiquitinähnlichen Domänen und wird im Menschen hauptsächlich in Zellen des Immunsystems exprimiert. Die Beteiligung von FAT10 an der Entstehung von Krebs and anderen Krankheiten hat die Aufmerksamkeit der wissenschaftlichen Gemeinschaft erregt, da Inhibition des ‚FAT10ylation‘ Prozesses einen neuen therapeutischen Ansatz zur Behandlung dieser Krankheiten darstellen könnte. UBA6 fungiert hierbei als E1 s Enzym, das am Anfang des FAT10-abhängigen proteasomalen Abbaus steht. UBA6 aktiviert neben FAT10 auch Ubiquitin mit ähnlicher Effizienz wie das ubiquitinspezifische E1 UBA1. Diese Bispezifität von UBA6 könnte die Inhibition der FAT10ylierung erschweren, da die Inhibition der katalytischen UBA6 Aktivität gleichzeitig UBA6-abhängige Ubiquitinaktivierung behindern würde. Daher ist für die zukünfitge Entwicklung FAT10-spezifischer UBA6 Inhibitoren ein grundlegendes Verständnis der UBA6 Bispezifität unerlässlich. In dieser Dissertation wurden wichtige, neue Einsichten in die Struktur und Funktion von UBA6 durch Röntgenkristallographie und biochemische Methoden gewonnen. Die erste Kristallstruktur von UBA6 zeigt die Multidomänenarchitektur des Enzyms bei atomarer Auflösung. Das Protein besteht aus einem starren Kern, der sowohl seine aktive als auch inaktive Adenylierungsdomäne sowie ein 4-Helix Bündel enthält. Das Molekül nimmt eine an ein Y erinnnernde Form ein, in der der Kern die Basis, die erste und zweite katalytischen Halbdomänen einen Arm und die ubiquitinähnliche gefaltete Domäne den zweiten Arm darstellen. Zwar ähneln sich der Domänenaufbau von UBA6 und UBA1, jedoch zeigte die Kristallstruktur bedeutende Unterschiede zwischen den beiden auf. Speziell die erste katalytische Halbdomäne ist in UBA6 im Vergleich zu UBA1weiter vom Enzymkern entfernt. Diese ‚Bewegung‘ erlaubt wahrscheinlich die Platzierung der zweiten UBL-Domäne von FAT10. Weitere Unterschiede konnten auch in der hydrophoben Oberfläche zwischen Kern, erster katalytischer Halbdomäne und dem aktiven Zentrum für die Adenylierung im Kern beobachtet werden, die zusammen die Bindestelle für Ubiquitin und FAT10 bilden. Durch ortsgerichtete Mutagenese von Schlüsselpositionen in dieser Region kontte die UBA6-katalysierte Aktivierung von entweder Ubiquitin oder FAT10 unterbunden werden. Um die Bispezifität von UBA6 zu entschlüsseln wurden UBA6 Varianten mit dem Ziel erzeugt, die Aktivierung von FAT10 unter Aufrechterhaltung der von Ubiquitin zu blockieren. Obwohl keine dieser Mutationen die FAT10-Aktivierung unterband, verhinderten einige jedoch die Aktivierung von Ubiquitin. Vorläufige Inhibitionsexperimente mit E1-Inhibitoren aus der Adenosylsulfamat Klasse zeigten starke Inhibition der FAT10ylierung durch zwei Verbindungen. Die Bispezifität von UBA6 bedarf weiterer strukturbiologischer und biochemischer Untersuchungen. Vor allem Kristallstrukturen von UBA6 in FAT10 und Ubiquitin-gebundener Form würden wichtige Erkenntnisse für weiteregehende biochemische Untersuchungen und schließlich die gezielte Unterdrückung der FAT10ylierungsmaschinerie liefern. KW - Ubiquitylation KW - FAT10ylation KW - UBA6 KW - UBE2Z Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-244579 ER - TY - JOUR A1 - Mammadova-Bach, Elmina A1 - Braun, Attila T1 - Zinc homeostasis in platelet-related diseases JF - International Journal of Molecular Sciences N2 - Zn\(^{2+}\) deficiency in the human population is frequent in underdeveloped countries. Worldwide, approximatively 2 billion people consume Zn\(^{2+}\)-deficient diets, accounting for 1–4% of deaths each year, mainly in infants with a compromised immune system. Depending on the severity of Zn\(^{2+}\) deficiency, clinical symptoms are associated with impaired wound healing, alopecia, diarrhea, poor growth, dysfunction of the immune and nervous system with congenital abnormalities and bleeding disorders. Poor nutritional Zn\(^{2+}\) status in patients with metastatic squamous cell carcinoma or with advanced non-Hodgkin lymphoma, was accompanied by cutaneous bleeding and platelet dysfunction. Forcing Zn\(^{2+}\) uptake in the gut using different nutritional supplementation of Zn\(^{2+}\) could ameliorate many of these pathological symptoms in humans. Feeding adult rodents with a low Zn\(^{2+}\) diet caused poor platelet aggregation and increased bleeding tendency, thereby attracting great scientific interest in investigating the role of Zn\(^{2+}\) in hemostasis. Storage protein metallothionein maintains or releases Zn\(^{2+}\) in the cytoplasm, and the dynamic change of this cytoplasmic Zn\(^{2+}\) pool is regulated by the redox status of the cell. An increase of labile Zn\(^{2+}\) pool can be toxic for the cells, and therefore cytoplasmic Zn\(^{2+}\) levels are tightly regulated by several Zn\(^{2+}\) transporters located on the cell surface and also on the intracellular membrane of Zn\(^{2+}\) storage organelles, such as secretory vesicles, endoplasmic reticulum or Golgi apparatus. Although Zn\(^{2+}\) is a critical cofactor for more than 2000 transcription factors and 300 enzymes, regulating cell differentiation, proliferation, and basic metabolic functions of the cells, the molecular mechanisms of Zn\(^{2+}\) transport and the physiological role of Zn\(^{2+}\) store in megakaryocyte and platelet function remain elusive. In this review, we summarize the contribution of extracellular or intracellular Zn\(^{2+}\) to megakaryocyte and platelet function and discuss the consequences of dysregulated Zn\(^{2+}\) homeostasis in platelet-related diseases by focusing on thrombosis, ischemic stroke and storage pool diseases. KW - Zinc KW - platelets KW - hemostasis KW - thrombosis KW - ischemic stroke KW - storage-pool diseases Y1 - 2019 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-285554 SN - 1422-0067 VL - 20 IS - 21 ER - TY - THES A1 - Imam, Nasir T1 - Molecular basis of collybistin conformational activation T1 - Molekulare Prinzipien der konformellen Aktivierung von Collybistin N2 - The nervous system relies on an orchestrated assembly of complex cellular entities called neurons, which are specifically committed to information management and transmission. Inter-neuronal communication takes place via synapses, membrane-membrane junctions which ensure efficient signal transfer. Synaptic neurotransmission involves release of presynaptic neurotransmitters and their reception by cognate receptors at postsynaptic terminals. Inhibitory neurotransmission is primarily mediated by the release of neurotransmitters GABA (γ-Aminobutyric acid) and glycine, which are precisely sensed by GABA type-A receptors (GABAARs) and glycine receptors (GlyRs), respectively. GABAAR assembly and maintenance is coordinated by various postsynaptic neuronal factors including the scaffolding protein gephyrin, the neuronal adaptor collybistin (CB) and cell adhesion proteins of the neuroligin (NL) family, specifically NL2 and NL4. At inhibitory postsynaptic specializations, gephyrin has been hypothesized to form extended structures underneath the plasma membrane, where its interaction with the receptors leads to their stabilization and impedes their lateral movement. Gephyrin mutations have been associated with various brain disorders, including autism, schizophrenia, Alzheimer’s disease, and epilepsy. Furthermore, gephyrin loss is lethal and causes mice to die within the first post-natal day. Gephyrin recruitment from intracellular deposits to postsynaptic membranes primarily relies on the adaptor protein CB. As a moonlighting protein, CB, a guanine nucleotide exchange factor (GEF), also catalyzes a nucleotide exchange reaction, thereby regenerating the GTP-bound state of the small GTPase Cdc42 from its GDP-bound form. The CB gene undergoes alternative splicing with the majority of CB splice variants featuring an N-terminal SH3 domain followed by tandem Dbl-homology (DH) and pleckstrin-homology (PH) domains. Previous studies demonstrated that the most widely expressed, SH3-domain containing splice variant (CB2SH3+) preferentially adopts a closed conformation, in which the N-terminally located SH3 domain forms intra-molecular interaction with the DH-PH domain tandem. Previous cell-based studies indicated that SH3 domain-encoding CB variants remain untargeted and colocalize with intracellular gephyrin deposits and hence require additional factors which interact with the SH3 domain, thus inducing an open or active conformation. The SH3 domain-deficient CB isoform (CB2SH3-), on the contrary, adopts an open conformation, which possess enhanced postsynaptic gephyrin-clustering and also effectively replenishes the GTP-bound small GTPase-Cdc42 from its GDP-bound state. Despite the fundamental role of CB as a neuronal adaptor protein maintaining the proper function of inhibitory GABAergic synapses, its interactions with the neuronal scaffolding protein gephyrin and other post synaptic neuronal factors remain poorly understood. Moreover, CB interaction studies with the small GTPase Cdc42 and TC10, a closely related member of Cdc42 subfamily, remains poorly characterized. Most importantly, the roles of the neuronal factors and small GTPases in CB conformational activation have not been elucidated. This PhD dissertation primarily focuses on delineating the molecular basis of the interactions between CB and postsynaptic neuronal factors. During the course of my PhD dissertation, I engineered a series of CB FRET (Förster Resonance Energy Transfer) sensors to characterize the CB interaction with its binding partners along with outlining their role in CB conformational activation. Through the aid of these CB FRET sensors, I analyzed the gephyrin-CB interaction, which, due to technical limitations remained unaddressed for more than two decades (refer Chapter 2 for more details). Subsequently, I also unraveled the molecular basis of the interactions between CB and the neuronal cell adhesion factor neuroligin 2 (refer chapter 2) and the small GTPases Cdc42 and TC10 (refer chapter 3) and describe how these binding partners induce a conformational activation of CB. In summary, this PhD dissertation provides strong evidence of a closely knit CB communication network with gephyrin, neuroligin and the small GTPase TC10, wherein CB activation from closed/inactive to open/active states is effectively triggered by these ligands. N2 - Das Nervensystem ist eine komplexe Ansammlung zellulärer Einheiten, darunter sind die Neuronen, die speziell für die Verarbeitung und Übertragung von Informationen zuständig sind. Die Kommunikation zwischen Neuronen erfolgt über Synapsen, spezialisierte Membran-Membran-Kontakte, die eine effiziente Signalübertragung gewährleisten. Die synaptische Neurotransmission umfasst die präsynaptische Freisetzung von Neurotransmitters und deren Empfang durch entsprechende Rezeptoren in den Postsynapsen. Die inhibitorische Neurotransmission wird in erster Linie durch die Freisetzung der Neurotransmitter GABA (γ-Aminobuttersäure-Typ) und Glycin vermittelt, die von GABA-Typ-A-Rezeptor (GABAAR) bzw. Glycinrezeptoren (GlyR) präzise wahrgenommen werden. Der Aufbau und die Aufrechterhaltung von GABAAR Clustern wird durch verschiedene postsynaptische neuronale Faktoren koordiniert, darunter das Gerüstprotein Gephyrin, das neuronale Adaptorprotein Collybistin (CB) und Zelladhäsionsproteine der Neuroligin (NL)-Familie, insbesondere NL2 und NL4. Es wird angenommen, dass Gephyrin an hemmenden postsynaptischen Spezialisierungen ausgedehnte Strukturen unterhalb der Plasmamembran bildet, und durch Interaktion mit den Rezeptoren deren laterale Diffusion verhindert. Gephyrin-Mutationen wurden mit verschiedenen Hirnkrankheiten in Verbindung gebracht, darunter Autismus, Schizophrenie, Alzheimer und Epilepsie. Der Verlust von Gephyrin ist tödlich und führt dazu, dass Mäuse innerhalb des ersten postnatalen Tages sterben. Die Rekrutierung von Gephyrin aus intrazellulären Ablagerungen zu postsynaptischen Membranen hängt in erster Linie von CB ab. Als Moonlighting-Protein katalysiert CB, ein Guanin-Nukleotid-Austauschfaktor (GEF), auch den Nukleotidaustausch und somit die Reaktivierung der kleinen GTPase Cdc42 . Das CB-Gen wird durch alternatives Spleißen modifiziert; die meisten CB-Spleißvarianten weisen eine N-terminale SH3-Domäne auf, gefolgt von Tandem aus einer Dbl-Homologie (DH)- und einer Pleckstrin-Homologie (PH)-Domäne. Frühere Studien zeigten, dass die am häufigsten exprimierte Spleißvariante, die eine SH3-Domäne enthält (CB2SH3+), vorzugsweise eine geschlossene Konformation annimmt, bei der die N-terminal gelegene SH3-Domäne eine intra-molekulare Interaktion mit dem DH-PH- Tandem eingeht. Zellbasierte Studien zeigten, dass CB-Varianten, die für die SH3-Domäne kodieren, sich innerhalb der Zelle nicht an spezifischen Orten anreichern und stattdessen mit intrazellulären Gephyrin-Ablagerungen kolokalisieren. Zusätzliche Faktoren werden benötigt, die mit der SH3-Domäne interagieren und so eine offene oder aktive Konformation hervorrufen. Die SH3-Domänen-defiziente CB-Isoform (CB2SH3-) hingegen nimmt eine offene Konformation an, die eine verstärkte postsynaptische Gephyrin-Anhäufung aufweist und die GTP-gebundene kleine GTPase Cdc42 aus ihrem GDP-gebundenen Zustand effektiv wieder regeneriert. Trotz der grundlegenden Rolle von CB als neuronales Adaptorprotein, das die ordnungsgemäße Funktion hemmender GABAerger Synapsen aufrechterhält, ist seine Interaktion mit dem neuronalen Gerüstprotein Gephyrin und anderen post-synaptischen neuronalen Faktoren nach wie vor unzureichend verstanden. Darüber hinaus sind die Interaktionsstudien von CB mit der kleinen GTPase Cdc42 und TC10, einem eng verwandten Mitglied der Cdc42-Unterfamilie, noch immer unzureichend charakterisiert. Somit war die Frage, ob diese neuronalen Faktoren sowie die kleinen GTPasen an der CB-Konformationsaktivierung beteiligt sind. Diese Dissertation konzentriert sich in erster Linie auf die Beschreibung der molekularen Grundlagen der Interaktion von CB mit postsynaptischen neuronalen Faktoren. Im Rahmen meiner Dissertation habe ich eine Reihe von CB-FRET-Sensoren (Förster-Resonanz-Energie-Transfer) entwickelt, um die CB-Interaktion mit seinen Bindungspartnern zu charakterisieren und ihre Rolle bei der CB-Konformationsaktivierung zu beschreiben. Mit Hilfe der CB-FRET-Sensoren entschlüsselte ich das langjährige Rätsel der Gephyrin-CB-Interaktion, das aufgrund technischer Beschränkungen mehr als zwei Jahrzehnte lang ungelöst blieb (siehe Kapitel 2 für weitere Einzelheiten). In der Folge habe ich auch die molekularen Grundlagen der CB-Wechselwirkung und damit ihre konformelle Aktivierung durch den neuronalen Zelladhäsionsfaktor Neuroligin 2 (siehe Kapitel 2) und die kleinen GTPasen Cdc42 und TC10 (siehe Kapitel 3) analysiertt. Zusammengefasst liefert diese Dissertation starke Beweise für ein engmaschiges CB-Kommunikationsnetzwerk mit Gephyrin, Neuroligin und der kleinen GTPase TC10, in dem der CB-Konformationswechsel vom geschlossenen/inaktiven zum offenen/aktiven Zustand effektiv durch die Liganden ausgelöst wird. KW - Guanine nucleotide exchange factor (GEF) KW - Rho GTPasw KW - inhibitory postsynapse KW - Autoinhibition KW - conformational activation KW - collybistin KW - fluorescence resonance energy transfer (FRET) KW - gephyrin KW - neurologin-2 KW - time-correlated single photon counting (TCSPC) Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-311458 ER - TY - JOUR A1 - Truongvan, Ngoc A1 - Li, Shurong A1 - Misra, Mohit A1 - Kuhn, Monika A1 - Schindelin, Hermann T1 - Structures of UBA6 explain its dual specificity for ubiquitin and FAT10 JF - Nature Communications N2 - The covalent modification of target proteins with ubiquitin or ubiquitin-like modifiers is initiated by E1 activating enzymes, which typically transfer a single modifier onto cognate conjugating enzymes. UBA6 is an unusual E1 since it activates two highly distinct modifiers, ubiquitin and FAT10. Here, we report crystal structures of UBA6 in complex with either ATP or FAT10. In the UBA6-FAT10 complex, the C-terminal domain of FAT10 binds to where ubiquitin resides in the UBA1-ubiquitin complex, however, a switch element ensures the alternate recruitment of either modifier. Simultaneously, the N-terminal domain of FAT10 interacts with the 3-helix bundle of UBA6. Site-directed mutagenesis identifies residues permitting the selective activation of either ubiquitin or FAT10. These results pave the way for studies investigating the activation of either modifier by UBA6 in physiological and pathophysiological settings. KW - enzyme mechanisms KW - post-translational modifications KW - X-ray crystallography Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-301161 VL - 13 ER - TY - JOUR A1 - Schulte, Clemens A1 - Soldà, Alice A1 - Spänig, Sebastian A1 - Adams, Nathan A1 - Bekić, Ivana A1 - Streicher, Werner A1 - Heider, Dominik A1 - Strasser, Ralf A1 - Maric, Hans Michael T1 - Multivalent binding kinetics resolved by fluorescence proximity sensing JF - Communications Biology N2 - Multivalent protein interactors are an attractive modality for probing protein function and exploring novel pharmaceutical strategies. The throughput and precision of state-of-the-art methodologies and workflows for the effective development of multivalent binders is currently limited by surface immobilization, fluorescent labelling and sample consumption. Using the gephyrin protein, the master regulator of the inhibitory synapse, as benchmark, we exemplify the application of Fluorescence proximity sensing (FPS) for the systematic kinetic and thermodynamic optimization of multivalent peptide architectures. High throughput synthesis of +100 peptides with varying combinatorial dimeric, tetrameric, and octameric architectures combined with direct FPS measurements resolved on-rates, off-rates, and dissociation constants with high accuracy and low sample consumption compared to three complementary technologies. The dataset and its machine learning-based analysis deciphered the relationship of specific architectural features and binding kinetics and thereby identified binders with unprecedented protein inhibition capacity; thus, highlighting the value of FPS for the rational engineering of multivalent inhibitors. KW - combinatorial libraries KW - kinetics KW - peptides KW - screening KW - thermodynamics Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-301157 VL - 5 ER - TY - INPR A1 - Neitz, Hermann A1 - Bessi, Irene A1 - Kuper, Jochen A1 - Kisker, Caroline A1 - Höbartner, Claudia T1 - Programmable DNA interstrand crosslinking by alkene-alkyne [2+2] photocycloaddition T2 - Journal of the American Chemical Society N2 - Covalent crosslinking of DNA strands provides a useful tool for medical, biochemical and DNA nanotechnology applications. Here we present a light-induced interstrand DNA crosslinking reaction using the modified nucleoside 5-phenylethynyl-2’-deoxyuridine (\(^{Phe}\)dU). The crosslinking ability of \(^{Phe}\)dU was programmed by base pairing and by metal ion interaction at the Watson-Crick base pairing site. Rotation to intrahelical positions was favored by hydrophobic stacking and enabled an unexpected photochemical alkene-alkyne [2+2] cycloaddition within the DNA duplex, resulting in efficient formation of a \(^{Phe}\)dU-dimer after short irradiation times of a few seconds. A \(^{Phe}\)dU dimer-containing DNA was shown to efficiently bind a helicase complex, but the covalent crosslink completely prevented DNA unwinding, suggesting possible applications in biochemistry or structural biology. KW - light-induced interstrand DNA crosslinking KW - alkene-alkyne [2+2] photocycloaddition KW - DNA-based nanostructures KW - DNA-processing enzymes Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-311822 N1 - This document is the unedited Author's version of a Submitted Work that was subsequently accepted for publication in Journal of the American Chemical Society, copyright © 2023 The Authors. Published by American Chemical Society. after peer review. To access the final edited and published work see https://doi.org/10.1021/jacs.3c01611. ET - submitted version ER - TY - JOUR A1 - Navarro, Stefano A1 - Starke, Andreas A1 - Heemskerk, Johan W. M. A1 - Kuijpers, Marijke J. E. A1 - Stegner, David A1 - Nieswandt, Bernhard T1 - Targeting of a conserved epitope in mouse and human GPVI differently affects receptor function JF - International Journal of Molecular Sciences N2 - Glycoprotein (GP) VI is the major platelet collagen receptor and a promising anti-thrombotic target. This was first demonstrated in mice using the rat monoclonal antibody JAQ1, which completely blocks the Collagen-Related Peptide (CRP)-binding site on mouse GPVI and efficiently inhibits mouse platelet adhesion, activation and aggregation on collagen. Here, we show for the first time that JAQ1 cross-reacts with human GPVI (huGPVI), but not with GPVI in other tested species, including rat, rabbit, guinea pig, swine, and dog. We further demonstrate that JAQ1 differently modulates mouse and human GPVI function. Similar to its effects on mouse GPVI (mGPVI), JAQ1 inhibits CRP-induced activation in human platelets, whereas, in stark contrast to mouse GPVI, it does not inhibit the adhesion, activation or aggregate formation of human platelets on collagen, but causes instead an increased response. This effect was also seen with platelets from newly generated human GPVI knockin mice (hGP6\(^{tg/tg\)). These results indicate that the binding of JAQ1 to a structurally conserved epitope in GPVI differently affects its function in human and mouse platelets. KW - glycoprotein VI KW - JAQ1 KW - platelet receptors KW - platelet activation KW - platelet inhibition Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-286227 SN - 1422-0067 VL - 23 IS - 15 ER - TY - JOUR A1 - Imam, Nasir A1 - Choudhury, Susobhan A1 - Heinze, Katrin G. A1 - Schindelin, Hermann T1 - Differential modulation of collybistin conformational dynamics by the closely related GTPases Cdc42 and TC10 JF - Frontiers in Synaptic Neuroscience N2 - Interneuronal synaptic transmission relies on the proper spatial organization of presynaptic neurotransmitter release and its reception on the postsynaptic side by cognate neurotransmitter receptors. Neurotransmitter receptors are incorporated into and arranged within the plasma membrane with the assistance of scaffolding and adaptor proteins. At inhibitory GABAergic postsynapses, collybistin, a neuronal adaptor protein, recruits the scaffolding protein gephyrin and interacts with various neuronal factors including cell adhesion proteins of the neuroligin family, the GABAA receptor α2-subunit and the closely related small GTPases Cdc42 and TC10 (RhoQ). Most collybistin splice variants harbor an N-terminal SH3 domain and exist in an autoinhibited/closed state. Cdc42 and TC10, despite sharing 67.4% amino acid sequence identity, interact differently with collybistin. Here, we delineate the molecular basis of the collybistin conformational activation induced by TC10 with the aid of recently developed collybistin FRET sensors. Time-resolved fluorescence-based FRET measurements reveal that TC10 binds to closed/inactive collybistin leading to relief of its autoinhibition, contrary to Cdc42, which only interacts with collybistin when forced into an open state by the introduction of mutations destabilizing the closed state of collybistin. Taken together, our data describe a TC10-driven signaling mechanism in which collybistin switches from its autoinhibited closed state to an open/active state. KW - autoinhibition KW - fluorescence resonance energy transfer (FRET) KW - gephyrin KW - guanine nucleotide exchange factor (GEF) KW - inhibitory postsynapse KW - Rho GTPase Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-282816 SN - 1663-3563 VL - 14 ER - TY - THES A1 - Aigner, Max T1 - Establishing successful protocols and imaging pipelines for Expansion Microscopy in murine blood platelets T1 - Etablierung erfolgreicher Protokolle zur Probenpräparation und Bildgebung für die ‚Expansion Microscopy‘ in murinen Thrombozyten N2 - Platelets play an important role in the body, since they are part of the hemostasis system, preventing and stopping blood loss. Nevertheless, when platelet or coagulation system function are impaired, uncontrolled bleedings but also irreversible vessel occlusion followed by ischemic tissue damage can occur. Therefore, understanding platelet function and activation, mechanisms which are controlled by a variety of platelet membrane receptors and other factors is important to advance out knowledge of hemostasis and platelet malfunction. For a complete picture of platelet function and their modulating behavior it is desired to be able to quantify receptor distributions and interactions of these densely packed molecular ensembles in the membrane. This challenges scientists for several reasons. Most importantly, platelets are microscopically small objects, challenging the spatial resolution of conventional light microscopy. Moreover, platelet receptors are highly abundant on the membrane so even super-resolution microscopy struggles with quantitative receptor imaging on platelets. With Expansion microscopy (ExM), a new super-resolution technique was introduced, allowing resolutions to achieve super-resolution without using a super-resolution microscope, but by combining a conventional confocal microscopy with a highly processed sample that has been expanded physically. In this doctoral thesis, I evaluated the potential of this technique for super-resolution platelet imaging by optimizing the sample preparation process and establishing an imaging and image processing pipeline for dual-color 3D images of different membrane receptors. The analysis of receptor colocalization using ExM demonstrated a clear superiority compared to conventional microscopy. Furthermore, I identified a library of fluorescently labeled antibodies against different platelet receptors compatible with ExM and showed the possibility of staining membrane receptors and parts of the cytoskeleton at the same time. N2 - Thrombozyten spielen eine wichtige Rolle im Körper, denn als Teil des Gerinnungssystems, sind sie daran beteiligt Blutverlust vorzubeugen und zu stoppen. Gleichwohl können sie bei Störungen des Gerinnungssystems zu unkontrollierbaren Blutungen und auch durch Aggregation zu kardiovaskulären Ereignissen, wie Herzinfarkt und Schlaganfällen führen. Für ein besseres Verständnis von Hämostase und Gerinnungsstörungen ist es deshalb nötig die Funktion und Aktivierung von Thrombozyten zu verstehen, welche durch eine Vielzahl von Membranrezeptoren und anderen Faktoren gesteuert wird. Eine Methode, um weitere Einblicke in diese Prozesse zu bekommen ist die mikroskopische Darstellung von Rezeptorverteilungen auf der Zellmembran und deren Interaktionen. Dies zu realisieren ist aus verschiedenen Gründen anspruchsvoll. Der mikroskopisch kleine Durchmesser der Thrombozyten macht es konventioneller Lichtmikroskopie schwer, einzelne Rezeptoren auf der Membran darzustellen. Außerdem befinden sich sehr viele Rezeptoren dicht gepackt auf der Membran, sodass sogar superhochauflösende Mikroskope Schwierigkeiten haben, die Rezeptoren quantitativ zu beurteilen. Mit ‚Expansion microscopy‘ (ExM) wurde eine relativ junge superhochaufösende Technik auf Thrombozyten angewendet. Diese Technik erreicht Auflösungen vergleichbar mit sogenannten ‚super-resolution‘ Mikroskopen, ohne die Benutzung selbiger, sondern durch die Kombination von konfokaler Mikroskopie mit einer physikalisch expandierten Probe. In dieser Arbeit evaluierte ich das Potential dieser Technik für superhochauflösende Bilder von Thrombozytenrezeptoren und optimierte die Probenvorbereitung, sodass zweifarbige 3D Bilder von verschiedenen Membranrezeptoren möglich waren. Die Ergebnisse der Kolokationsanalyse zeigten einen deutlich vergrößerten Dynamikumfang durch ExM. Außerdem katalogisierte ich fluoreszenzmarkierte Antikörper gegen verschiedene Thrombozyten Rezeptoren bezüglich ihrer Tauglichkeit mit ExM und zeigte, dass es möglich ist Membranrezeptoren und Bestandteile des Zytoskeletts gleichzeitig zu färben. KW - Expansion Microscopy KW - platelets KW - Mikroskopie KW - Microscopy Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-309003 ER - TY - JOUR A1 - Jeanclos, Elisabeth A1 - Schlötzer, Jan A1 - Hadamek, Kerstin A1 - Yuan-Chen, Natalia A1 - Alwahsh, Mohammad A1 - Hollmann, Robert A1 - Fratz, Stefanie A1 - Yesilyurt-Gerhards, Dilan A1 - Frankenbach, Tina A1 - Engelmann, Daria A1 - Keller, Angelika A1 - Kaestner, Alexandra A1 - Schmitz, Werner A1 - Neuenschwander, Martin A1 - Hergenröder, Roland A1 - Sotriffer, Christoph A1 - von Kries, Jens Peter A1 - Schindelin, Hermann A1 - Gohla, Antje T1 - Glycolytic flux control by drugging phosphoglycolate phosphatase JF - Nature Communications N2 - Targeting the intrinsic metabolism of immune or tumor cells is a therapeutic strategy in autoimmunity, chronic inflammation or cancer. Metabolite repair enzymes may represent an alternative target class for selective metabolic inhibition, but pharmacological tools to test this concept are needed. Here, we demonstrate that phosphoglycolate phosphatase (PGP), a prototypical metabolite repair enzyme in glycolysis, is a pharmacologically actionable target. Using a combination of small molecule screening, protein crystallography, molecular dynamics simulations and NMR metabolomics, we discover and analyze a compound (CP1) that inhibits PGP with high selectivity and submicromolar potency. CP1 locks the phosphatase in a catalytically inactive conformation, dampens glycolytic flux, and phenocopies effects of cellular PGP-deficiency. This study provides key insights into effective and precise PGP targeting, at the same time validating an allosteric approach to control glycolysis that could advance discoveries of innovative therapeutic candidates. KW - phosphoglycolate phosphatase KW - glycolytic flux control KW - intrinsic metabolism Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-300928 VL - 13 IS - 1 ER - TY - JOUR A1 - Fusi, Lorenza A1 - Paudel, Rupesh A1 - Meder, Katharina A1 - Schlosser, Andreas A1 - Schrama, David A1 - Goebeler, Matthias A1 - Schmidt, Marc T1 - Interaction of transcription factor FoxO3 with histone acetyltransferase complex subunit TRRAP modulates gene expression and apoptosis JF - Journal of Biological Chemistry N2 - Forkhead box O (FoxO) transcription factors are conserved proteins involved in the regulation of life span and age-related diseases, such as diabetes and cancer. Stress stimuli or growth factor deprivation promotes nuclear localization and activation of FoxO proteins, which—depending on the cellular context—can lead to cell cycle arrest or apoptosis. In endothelial cells (ECs), they further regulate angiogenesis and may promote inflammation and vessel destabilization implicating a role of FoxOs in vascular diseases. In several cancers, FoxOs exert a tumor-suppressive function by regulating proliferation and survival. We and others have previously shown that FoxOs can regulate these processes via two different mechanisms: by direct binding to forkhead-responsive elements at the promoter of target genes or by a poorly understood alternative process that does not require direct DNA binding and regulates key targets in primary human ECs. Here, we performed an interaction study in ECs to identify new nuclear FoxO3 interaction partners that might contribute to FoxO-dependent gene regulation. Mass spectrometry analysis of FoxO3-interacting proteins revealed transformation/transcription domain–associated protein (TRRAP), a member of multiple histone acetyltransferase complexes, as a novel binding partner of FoxO family proteins. We demonstrate that TRRAP is required to support FoxO3 transactivation and FoxO3-dependent G1 arrest and apoptosis in ECs via transcriptional activation of the cyclin-dependent kinase inhibitor p27\(^{kip1}\) and the proapoptotic B-cell lymphoma 2 family member, BIM. Moreover, FoxO–TRRAP interaction could explain FoxO-induced alternative gene regulation via TRRAP-dependent recruitment to target promoters lacking forkhead-responsive element sequences. KW - FoxO3 KW - TRRAP KW - transcription factors Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-299820 VL - 298 IS - 3 ER -