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 -