@article{HofstetterOgunleyeKutschkeetal.2024, author = {Hofstetter, Julia and Ogunleye, Ayoola and Kutschke, Andr{\´e} and Buchholz, Lisa Marie and Wolf, Elmar and Raabe, Thomas and Gallant, Peter}, title = {Spt5 interacts genetically with Myc and is limiting for brain tumor growth in Drosophila}, series = {Life Science Alliance}, volume = {7}, journal = {Life Science Alliance}, number = {1}, issn = {2575-1077}, doi = {10.26508/lsa.202302130}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-350197}, year = {2024}, abstract = {The transcription factor SPT5 physically interacts with MYC oncoproteins and is essential for efficient transcriptional activation of MYC targets in cultured cells. Here, we use Drosophila to address the relevance of this interaction in a living organism. Spt5 displays moderate synergy with Myc in fast proliferating young imaginal disc cells. During later development, Spt5-knockdown has no detectable consequences on its own, but strongly enhances eye defects caused by Myc overexpression. Similarly, Spt5-knockdown in larval type 2 neuroblasts has only mild effects on brain development and survival of control flies, but dramatically shrinks the volumes of experimentally induced neuroblast tumors and significantly extends the lifespan of tumor-bearing animals. This beneficial effect is still observed when Spt5 is knocked down systemically and after tumor initiation, highlighting SPT5 as a potential drug target in human oncology.}, language = {en} } @article{HailerGrunewaldOrthetal.2014, author = {Hailer, Amelie and Grunewald, Thomas G. P. and Orth, Martin and Reiss, Cora and Kneitz, Burkhard and Spahn, Martin and Butt, Elke}, title = {Loss of tumor suppressor mir-203 mediates overexpression of LIM and SH3 Protein 1 (LASP1) in high-risk prostate cancer thereby increasing cell proliferation and migration}, series = {Oncotarget}, volume = {5}, journal = {Oncotarget}, number = {12}, issn = {1949-2553}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-120540}, pages = {4144-53}, year = {2014}, abstract = {Several studies have linked overexpression of the LIM and SH3 domain protein 1 (LASP1) to progression of breast, colon, liver, and bladder cancer. However, its expression pattern and role in human prostate cancer (PCa) remained largely undefined. Analysis of published microarray data revealed a significant overexpression of LASP1 in PCa metastases compared to parental primary tumors and normal prostate epithelial cells. Subsequent gene-set enrichment analysis comparing LASP1-high and -low PCa identified an association of LASP1 with genes involved in locomotory behavior and chemokine signaling. These bioinformatic predictions were confirmed in vitro as the inducible short hairpin RNA-mediated LASP1 knockdown impaired migration and proliferation in LNCaP prostate cancer cells. By immunohistochemical staining and semi-quantitative image analysis of whole tissue sections we found an enhanced expression of LASP1 in primary PCa and lymph node metastases over benign prostatic hyperplasia. Strong cytosolic and nuclear LASP1 immunoreactivity correlated with PSA progression. Conversely, qRT-PCR analyses for mir-203, which is a known translational suppressor of LASP1 in matched RNA samples revealed an inverse correlation of LASP1 protein and mir-203 expression. Collectively, our results suggest that loss of mir-203 expression and thus uncontrolled LASP1 overexpression might drive progression of PCa.}, language = {en} } @article{GarciaBetancurGoniMorenoHorgeretal.2017, author = {Garc{\´i}a-Betancur, Juan-Carlos and Go{\~n}i-Moreno, Angel and Horger, Thomas and Schott, Melanie and Sharan, Malvika and Eikmeier, Julian and Wohlmuth, Barbara and Zernecke, Alma and Ohlsen, Knut and Kuttler, Christina and Lopez, Daniel}, title = {Cell differentiation defines acute and chronic infection cell types in Staphylococcus aureus}, series = {eLife}, volume = {6}, journal = {eLife}, number = {e28023}, doi = {10.7554/eLife.28023}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-170346}, year = {2017}, abstract = {A central question to biology is how pathogenic bacteria initiate acute or chronic infections. Here we describe a genetic program for cell-fate decision in the opportunistic human pathogen Staphylococcus aureus, which generates the phenotypic bifurcation of the cells into two genetically identical but different cell types during the course of an infection. Whereas one cell type promotes the formation of biofilms that contribute to chronic infections, the second type is planktonic and produces the toxins that contribute to acute bacteremia. We identified a bimodal switch in the agr quorum sensing system that antagonistically regulates the differentiation of these two physiologically distinct cell types. We found that extracellular signals affect the behavior of the agr bimodal switch and modify the size of the specialized subpopulations in specific colonization niches. For instance, magnesium-enriched colonization niches causes magnesium binding to S. aureusteichoic acids and increases bacterial cell wall rigidity. This signal triggers a genetic program that ultimately downregulates the agr bimodal switch. Colonization niches with different magnesium concentrations influence the bimodal system activity, which defines a distinct ratio between these subpopulations; this in turn leads to distinct infection outcomes in vitro and in an in vivo murine infection model. Cell differentiation generates physiological heterogeneity in clonal bacterial infections and helps to determine the distinct infection types.}, language = {en} } @article{GambaryanSubramanianKehreretal.2016, author = {Gambaryan, Stepan and Subramanian, Hariharan and Kehrer, Linda and Mindukshev, Igor and Sudnitsyna, Julia and Reiss, Cora and Rukoyatkina, Natalia and Friebe, Andreas and Sharina, Iraida and Martin, Emil and Walter, Ulrich}, title = {Erythrocytes do not activate purified and platelet soluble guanylate cyclases even in conditions favourable for NO synthesis}, series = {Cell Communication and Signaling}, volume = {14}, journal = {Cell Communication and Signaling}, number = {16}, doi = {10.1186/s12964-016-0139-9}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-161223}, year = {2016}, abstract = {Background Direct interaction between Red blood cells (RBCs) and platelets is known for a long time. The bleeding time is prolonged in anemic patients independent of their platelet count and could be corrected by transfusion of RBCs, which indicates that RBCs play an important role in hemostasis and platelet activation. However, in the last few years, opposing mechanisms of platelet inhibition by RBCs derived nitric oxide (NO) were proposed. The aim of our study was to identify whether RBCs could produce NO and activate soluble guanylate cyclase (sGC) in platelets. Methods To test whether RBCs could activate sGC under different conditions (whole blood, under hypoxia, or even loaded with NO), we used our well-established and highly sensitive models of NO-dependent sGC activation in platelets and activation of purified sGC. The activation of sGC was monitored by detecting the phosphorylation of Vasodilator Stimulated Phosphoprotein (VASPS239) by flow cytometry and Western blot. ANOVA followed by Bonferroni's test and Student's t-test were used as appropriate. Results We show that in the whole blood, RBCs prevent NO-mediated inhibition of ADP and TRAP6-induced platelet activation. Likewise, coincubation of RBCs with platelets results in strong inhibition of NO-induced sGC activation. Under hypoxic conditions, incubation of RBCs with NO donor leads to Hb-NO formation which inhibits sGC activation in platelets. Similarly, RBCs inhibit activation of purified sGC, even under conditions optimal for RBC-mediated generation of NO from nitrite. Conclusions All our experiments demonstrate that RBCs act as strong NO scavengers and prevent NO-mediated inhibition of activated platelets. In all tested conditions, RBCs were not able to activate platelet or purified sGC.}, language = {en} } @article{FrietschKastnerGrunewaldetal.2014, author = {Frietsch, Jochen J. and Kastner, Carolin and Grunewald, Thomas G.P. and Schweigel, Hardy and Nollau, Peter and Ziermann, Janine and Clement, Joachim H. and La Res{\´e}e, Paul and Hochhaus, Andreas and Butt, Elke}, title = {LASP1 is a novel BCR-ABL substrate and a phosphorylation-dependent binding partner of CRKL in chronic myeloid leukemia}, series = {Oncotarget}, volume = {5}, journal = {Oncotarget}, number = {14}, issn = {1949-2553}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-120639}, pages = {5257-71}, year = {2014}, abstract = {Chronic myeloid leukemia (CML) is characterized by a genomic translocation generating a permanently active BCR-ABL oncogene with a complex pattern of atypically tyrosine-phosphorylated proteins that drive the malignant phenotype of CML. Recently, the LIM and SH3 domain protein 1 (LASP1) was identified as a component of a six gene signature that is strongly predictive for disease progression and relapse in CML patients. However, the underlying mechanisms why LASP1 expression correlates with dismal outcome remained unresolved. Here, we identified LASP1 as a novel and overexpressed direct substrate of BCR-ABL in CML. We demonstrate that LASP1 is specifically phosphorylated by BCR-ABL at tyrosine-171 in CML patients, which is abolished by tyrosine kinase inhibitor therapy. Further studies revealed that LASP1 phosphorylation results in an association with CRKL - another specific BCR-ABL substrate and bona fide biomarker for BCR-ABL activity. pLASP1-Y171 binds to non-phosphorylated CRKL at its SH2 domain. Accordingly, the BCR-ABL-mediated pathophysiological hyper-phosphorylation of LASP1 in CML disrupts normal regulation of CRKL and LASP1, which likely has implications on downstream BCR-ABL signaling. Collectively, our results suggest that LASP1 phosphorylation might serve as an additional candidate biomarker for assessment of BCR-ABL activity and provide a first step toward a molecular understanding of LASP1 function in CML.}, language = {en} } @article{FerberGerhardsSaueretal.2020, author = {Ferber, Elena and Gerhards, Julian and Sauer, Miriam and Krischke, Markus and Dittrich, Marcus T. and M{\"u}ller, Tobias and Berger, Susanne and Fekete, Agnes and Mueller, Martin J.}, title = {Chemical Priming by Isothiocyanates Protects Against Intoxication by Products of the Mustard Oil Bomb}, series = {Frontiers in Plant Science}, volume = {11}, journal = {Frontiers in Plant Science}, issn = {1664-462X}, doi = {10.3389/fpls.2020.00887}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-207104}, year = {2020}, abstract = {In Brassicaceae, tissue damage triggers the mustard oil bomb i.e., activates the degradation of glucosinolates by myrosinases leading to a rapid accumulation of isothiocyanates at the site of damage. Isothiocyanates are reactive electrophilic species (RES) known to covalently bind to thiols in proteins and glutathione, a process that is not only toxic to herbivores and microbes but can also cause cell death of healthy plant tissues. Previously, it has been shown that subtoxic isothiocyanate concentrations can induce transcriptional reprogramming in intact plant cells. Glutathione depletion by RES leading to breakdown of the redox potential has been proposed as a central and common RES signal transduction mechanism. Using transcriptome analyses, we show that after exposure of Arabidopsis seedlings (grown in liquid culture) to subtoxic concentrations of sulforaphane hundreds of genes were regulated without depletion of the cellular glutathione pool. Heat shock genes were among the most highly up-regulated genes and this response was found to be dependent on the canonical heat shock factors A1 (HSFA1). HSFA1-deficient plants were more sensitive to isothiocyanates than wild type plants. Moreover, pretreatment of Arabidopsis seedlings with subtoxic concentrations of isothiocyanates increased resistance against exposure to toxic levels of isothiocyanates and, hence, may reduce the autotoxicity of the mustard oil bomb by inducing cell protection mechanisms.}, language = {en} } @article{CochainChaudhariKochetal.2014, author = {Cochain, Clement and Chaudhari, Sweena M. and Koch, Miriam and Wiendl, Heinz and Eckstein, Hans-Henning and Zernecke, Alma}, title = {Programmed Cell Death-1 Deficiency Exacerbates T Cell Activation and Atherogenesis despite Expansion of Regulatory T Cells in Atherosclerosis-Prone Mice}, series = {PLoS ONE}, volume = {9}, journal = {PLoS ONE}, number = {4}, issn = {1932-6203}, doi = {10.1371/journal.pone.0093280}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119823}, pages = {e93280}, year = {2014}, abstract = {T cell activation represents a double-edged sword in atherogenesis, as it promotes both pro-inflammatory T cell activation and atheroprotective Foxp3(+) regulatory T cell (Treg) responses. Here, we investigated the role of the co-inhibitory receptor programmed cell death-1 (PD-1) in T cell activation and CD4(+) T cell polarization towards pro-atherogenic or atheroprotective responses in mice. Mice deficient for both low density lipoprotein receptor and PD-1 (Ldlr(-/-)Pd1(-/-)) displayed striking increases in systemic CD4(+) and CD8(+) T cell activation after 9 weeks of high fat diet feeding, associated with an expansion of both pro-atherogenic IFNγ-secreting T helper 1 cells and atheroprotective Foxp3+ Tregs. Importantly, PD-1 deficiency did not affect Treg suppressive function in vitro. Notably, PD-1 deficiency exacerbated atherosclerotic lesion growth and entailed a massive infiltration of T cells in atherosclerotic lesions. In addition, aggravated hypercholesterolemia was observed in Ldlr(-/-)Pd1(-/-) mice. In conclusion, we here demonstrate that although disruption of PD-1 signaling enhances both pro- and anti-atherogenic T cell responses in Ldlr(-/-) mice, pro-inflammatory T cell activation prevails and enhances dyslipidemia, vascular inflammation and atherosclerosis.}, language = {en} } @article{BuschWesthofenKochetal.2014, author = {Busch, Martin and Westhofen, Thilo C. and Koch, Miriam and Lutz, Manfred B. and Zernecke, Alma}, title = {Dendritic Cell Subset Distributions in the Aorta in Healthy and Atherosclerotic Mice}, series = {PLoS ONE}, volume = {9}, journal = {PLoS ONE}, number = {2}, issn = {1932-6203}, doi = {10.1371/journal.pone.0088452}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-119907}, pages = {e88452}, year = {2014}, abstract = {Dendritic cells (DCs) can be sub-divided into various subsets that play specialized roles in priming of adaptive immune responses. Atherosclerosis is regarded as a chronic inflammatory disease of the vessel wall and DCs can be found in non-inflamed and diseased arteries. We here performed a systematic analyses of DCs subsets during atherogenesis. Our data indicate that distinct DC subsets can be localized in the vessel wall. In C57BL/6 and low density lipoprotein receptor-deficient (Ldlr-/-) mice, CD11c+ MHCII+ DCs could be discriminated into CD103- CD11b+F4/80+, CD11b+F4/80- and CD11b-F4/80- DCs and CD103+ CD11b-F4/80- DCs. Except for CD103- CD11b- F4/80- DCs, these subsets expanded in high fat diet-fed Ldlr-/- mice. Signal-regulatory protein (Sirp)-α was detected on aortic macrophages, CD11b+ DCs, and partially on CD103- CD11b- F4/80- but not on CD103+ DCs. Notably, in FMS-like tyrosine kinase 3-ligand-deficient (Flt3l-/-) mice, a specific loss of CD103+ DCs but also CD103- CD11b+ F4/80- DCs was evidenced. Aortic CD103+ and CD11b+ F4/80- CD103- DCs may thus belong to conventional rather than monocyte-derived DCs, given their dependence on Flt3L-signalling. CD64, postulated to distinguish macrophages from DCs, could not be detected on DC subsets under physiological conditions, but appeared in a fraction of CD103- CD11b+ F4/80- and CD11b+ F4/80+ cells in atherosclerotic Ldlr-/- mice. The emergence of CD64 expression in atherosclerosis may indicate that CD11b+ F4/80- DCs similar to CD11b+ F4/80+ DCs are at least in part derived from immigrated monocytes during atherosclerotic lesion formation. Our data advance our knowledge about the presence of distinct DC subsets and their accumulation characteristics in atherosclerosis, and may help to assist in future studies aiming at specific DC-based therapeutic strategies for the treatment of chronic vascular inflammation.}, language = {en} } @article{BuschHoffjanBergmannetal.2016, author = {Busch, Albert and Hoffjan, Sabine and Bergmann, Frauke and Hartung, Birgit and Jung, Helena and Hanel, Daniela and Tzschach, Andeas and Kadar, Janos and von Kodolitsch, Yskert and Germer, Christoph-Thomas and Trobisch, Heiner and Strasser, Erwin and Wildenauer, Ren{\´e}}, title = {Vascular type Ehlers-Danlos syndrome is associated with platelet dysfunction and low vitamin D serum concentration}, series = {Orphanet Journal of Rare Diseases}, volume = {11}, journal = {Orphanet Journal of Rare Diseases}, number = {111}, doi = {10.1186/s13023-016-0491-2}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147757}, year = {2016}, abstract = {Background The vascular type represents a very rare, yet the clinically most fatal entity of Ehlers-Danlos syndrome (EDS). Patients are often admitted due to arterial bleedings and the friable tissue and the altered coagulation contribute to the challenge in treatment strategies. Until now there is little information about clotting characteristics that might influence hemostasis decisively and eventually worsen emergency situations. Results 22 vascular type EDS patients were studied for hemoglobin, platelet volume and count, Quick and activated partial thromboplastin time, fibrinogen, factor XIII, von Willebrand disease, vitamin D and platelet aggregation by modern standard laboratory methods. Results show a high prevalence of over 50 \% for platelet aggregation disorders in vascular type EDS patients, especially for collagen and epinephrine induced tests, whereas the plasmatic cascade did not show any alterations. Additionally, more than half of the tested subjects showed low vitamin D serum levels, which might additionally affect vascular wall integrity. Conclusion The presented data underline the importance of detailed laboratory screening methods in vascular type EDS patients in order to allow for targeted application of platelet-interacting substances that might be of decisive benefit in the emergency setting.}, language = {en} } @article{BeyrichLoefflerKobsaretal.2011, author = {Beyrich, Claudia and L{\"o}ffler, J{\"u}rgen and Kobsar, Anna and Speer, Christian P. and Kneitz, Susanne and Eigenthaler, Martin}, title = {Infection of Human Coronary Artery Endothelial Cells by Group B Streptococcus Contributes to Dysregulation of Apoptosis, Hemostasis, and Innate Immune Responses [Research Article]}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-68834}, year = {2011}, abstract = {Early onset sepsis due to group B streptococcus leads to neonatal morbidity, increased mortality, and long-term neurological deficencies. Interaction between septicemic GBS and confluent monolayers of human coronary artery endothelial cells (HCAECs) was analyzed by genome wide expression profiling. In total, 124 genes were differentially expressed (89 upregulated, 35 downregulated) based on a more than 3-fold difference to control HCAEC. Regulated genes are involved in apoptosis, hemostasis, oxidative stress response, infection, and inflammation. Regulation of selected genes and proteins identified in the gene array analysis was confirmed by Real-time RT-PCR assay (granulocy te chemotactic protein 2), ELISA (urokinase, cyclooxygenase 2, granulocyte chemotactic protein 1), and western blotting (Heme oxygenase1, BCL2 interacting protein) at various time points between 4 and 24 hours. These results indicate that GBS infection might influence signalling pathways leading to impaired function of the innate immune system and hemorrhagic and inflammatory complications during GBS sepsis.}, subject = {Medizin}, language = {en} }