@article{RiedelMofoloAvotaetal.2013, author = {Riedel, Alice and Mofolo, Boitumelo and Avota, Elita and Schneider-Schaulies, Sibylle and Meintjes, Ayton and Mulder, Nicola and Kneitz, Susanne}, title = {Accumulation of Splice Variants and Transcripts in Response to PI3K Inhibition in T Cells}, series = {PLoS ONE}, volume = {8}, journal = {PLoS ONE}, number = {2}, doi = {10.1371/journal.pone.0050695}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-130335}, pages = {e50695}, year = {2013}, abstract = {Background Measles virus (MV) causes T cell suppression by interference with phosphatidylinositol-3-kinase (PI3K) activation. We previously found that this interference affected the activity of splice regulatory proteins and a T cell inhibitory protein isoform was produced from an alternatively spliced pre-mRNA. Hypothesis Differentially regulated and alternatively splice variant transcripts accumulating in response to PI3K abrogation in T cells potentially encode proteins involved in T cell silencing. Methods To test this hypothesis at the cellular level, we performed a Human Exon 1.0 ST Array on RNAs isolated from T cells stimulated only or stimulated after PI3K inhibition. We developed a simple algorithm based on a splicing index to detect genes that undergo alternative splicing (AS) or are differentially regulated (RG) upon T cell suppression. Results Applying our algorithm to the data, 9\% of the genes were assigned as AS, while only 3\% were attributed to RG. Though there are overlaps, AS and RG genes differed with regard to functional regulation, and were found to be enriched in different functional groups. AS genes targeted extracellular matrix (ECM)-receptor interaction and focal adhesion pathways, while RG genes were mainly enriched in cytokine-receptor interaction and Jak-STAT. When combined, AS/RG dependent alterations targeted pathways essential for T cell receptor signaling, cytoskeletal dynamics and cell cycle entry. Conclusions PI3K abrogation interferes with key T cell activation processes through both differential expression and alternative splicing, which together actively contribute to T cell suppression.}, language = {en} } @article{KarulinKaracsonyZhangetal.2015, author = {Karulin, Alexey Y. and Karacsony, Kinga and Zhang, Wenji and Targoni, Oleg S. and Moldova, Ioana and Dittrich, Marcus and Sundararaman, Srividya and Lehmann, Paul V.}, title = {ELISPOTs produced by CD8 and CD4 cells follow Log Normal size distribution permitting objective counting}, series = {Cells}, volume = {4}, journal = {Cells}, number = {1}, doi = {10.3390/cells4010056}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-149648}, pages = {56-70}, year = {2015}, abstract = {Each positive well in ELISPOT assays contains spots of variable sizes that can range from tens of micrometers up to a millimeter in diameter. Therefore, when it comes to counting these spots the decision on setting the lower and the upper spot size thresholds to discriminate between non-specific background noise, spots produced by individual T cells, and spots formed by T cell clusters is critical. If the spot sizes follow a known statistical distribution, precise predictions on minimal and maximal spot sizes, belonging to a given T cell population, can be made. We studied the size distributional properties of IFN-γ, IL-2, IL-4, IL-5 and IL-17 spots elicited in ELISPOT assays with PBMC from 172 healthy donors, upon stimulation with 32 individual viral peptides representing defined HLA Class I-restricted epitopes for CD8 cells, and with protein antigens of CMV and EBV activating CD4 cells. A total of 334 CD8 and 80 CD4 positive T cell responses were analyzed. In 99.7\% of the test cases, spot size distributions followed Log Normal function. These data formally demonstrate that it is possible to establish objective, statistically validated parameters for counting T cell ELISPOTs.}, language = {en} } @phdthesis{Schuh2001, author = {Schuh, Kai}, title = {Erzeugung und Analyse NF-ATp-defizienter M{\"a}use}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-117}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2001}, abstract = {Ziel dieser Arbeit war es, NF-AT1-Gen-defiziente Mauslinien zu erzeugen und die Folgen dieser genetischen Manipulation in vivo zu untersuchen. Die Untersuchung sollte die durch die Gendefizienz erwarteten M{\"a}ngel w{\"a}hrend der Entwicklung (Embryogenese) und, im Besonderen, die Auswirkungen auf das Immunsytem und die Entwicklung und Differenzierung der T-Zellen aufzeigen. Zur Untersuchung der genomischen Organisation des Maus-NF-AT1-Gens wurde eine genomische l-Phagen-DNA-Bibliothek "gescreent" (durchgef{\"u}hrt von Dr. E. Jankevics, Universit{\"a}t von Riga, Lettland), die entsprechenden l-Phagen, die das NF-AT1-Gen enthielten, isoliert und die DNA pr{\"a}pariert. Nach Analyse der klonierten Phagen (Subklonierung und Sequenzierung) wurde eine Restriktionskarte der entsprechenden Bereiche erstellt und der "targeting-vector" erstellt. Der "targeting-vector" wurde durch Elektroporation in embryonale Stammzellen (ES-Zellen) eingebracht und die Integration in das Genom ("Homologe Rekombination") durch Southern Blotting- bzw. PCR-Analyse untersucht. Manipulierte ES-Zellklone wurden in C57Bl/6-Blastozysten injiziert, diese in scheinschwangere Ammentiere transferiert und die Nachkommen nach Geburt anhand der Fellfarben klassifiziert. Nachkommen mit einem hohen Anteil hellen Fells wurden mit C57 Bl/6-Tieren verpaart und die Integration des manipulierten Zellklons in die Keimbahn wurde anhand der wildtypischen Fellfarbe und Genotypisierung nachgewiesen. Bez{\"u}glich des manipulierten NF-ATp-Gens heterozygote F1-Tiere wurden miteinander verpaart, um eine homozygote NF-ATp-defiziente M{\"a}use zu erhalten. Die Deletion des NF-ATp-Proteins wurde durch in Western-Blotting-Experimenten und EMSAs ("electrophoretic mobility shift assays") nachgewiesen. Die NF-ATp-/--Tiere zeigten keine augenscheinlichen Ver{\"a}nderungen w{\"a}hrend der Entwicklung und, bei jungen Tieren, keine offensichtlichen Ver{\"a}nderungen bei der Entwicklung des Immunsystems. In {\"a}lteren Tieren (> 6 Wochen) war eine Hyperproliferation der Zellen des Immunsystems zu beobachten, was mit einer Splenomegalie, einer verst{\"a}rkten Bildung von Keimzentren in lymphatischen Organen, vergr{\"o}ßerten Lymphknoten und einer verlangsamten Involution des Thymus einherging. Weitergehende Untersuchungen der Ursache dieser hyperproliferativen Erkrankung offenbarten eine verminderte klonale Deletion nach Aktivierung. Die Ursachen dieses {\"u}berraschenden Effekts sind wahrscheinlich vielf{\"a}ltig, da NF-AT-Faktoren an der Regulation der Expression vieler Gene beteiligt sind, u.a. des Apoptose-assoziierten CD95-Liganden. Da sich bez{\"u}glich der IL-2-Expression keine Unterschiede zwischen NF-ATp-defizienten Tieren und Kontrollen zeigten, jedoch eine erh{\"o}hte IL-2-Konzentration im Medium kultivierter NF-ATp-defizienter T-Zellen beobachtet wurde, wurde die Bindung von NF-ATp an putative NF-AT-Bindungssequenzen des CD25-Promotors, die transkriptionelle Aktivierung des Promotors mittels Luciferase-Assays und die Expression der IL-2R-alpha-Kette (CD25) untersucht. Es konnte gezeigt werden, daß (1.) NF-ATp an zwei Regionen des CD25-Promotors bindet, (2.) der CD25-Promotor durch NF-ATp transkriptionell stark stimuliert wird und (3.) T-Zellen NF-ATp-defizienter Tiere nach Stimulation eine verminderte CD25-Expression zeigen. In NF-ATp-defizienten Tieren war die Expression von CD25 moderat reduziert, was eine Erkl{\"a}rung f{\"u}r den abgeschw{\"a}chten Ph{\"a}notyp dieser Tiere - im Vergleich zu IL-2- oder CD25-defizienten Tieren - sein kann. Die hyperproliferativen Erkrankungen dieser verschiedenen Mauslinien weisen auf eine Beteiligung der NF-AT-/IL-2-/IL-2R-Signalwege nicht nur w{\"a}hrend der T-Zell-Aktivierung hin, sondern auch auf eine Beteiligung an Signalwegen, die zur anschließenden Inaktivierung und Apoptose der T-Lymphozyten n{\"o}tig sind.}, subject = {Maus}, language = {de} } @article{SilvaVilchesPletinckxLohnertetal.2017, author = {Silva-Vilches, Cinthia and Pletinckx, Katrien and Lohnert, Miriam and Pavlovic, Vladimir and Ashour, Diyaaeldin and John, Vini and Vendelova, Emilia and Kneitz, Susanne and Zhou, Jie and Chen, Rena and Reinheckel, Thomas and Mueller, Thomas D. and Bodem, Jochen and Lutz, Manfred B.}, title = {Low doses of cholera toxin and its mediator cAMP induce CTLA-2 secretion by dendritic cells to enhance regulatory T cell conversion}, series = {PLoS ONE}, volume = {12}, journal = {PLoS ONE}, number = {7}, doi = {10.1371/journal.pone.0178114}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-158244}, pages = {e0178114}, year = {2017}, abstract = {Immature or semi-mature dendritic cells (DCs) represent tolerogenic maturation stages that can convert naive T cells into Foxp3\(^{+}\) induced regulatory T cells (iTreg). Here we found that murine bone marrow-derived DCs (BM-DCs) treated with cholera toxin (CT) matured by up-regulating MHC-II and costimulatory molecules using either high or low doses of CT (CT\(^{hi}\), CT\(^{lo}\)) or with cAMP, a known mediator CT signals. However, all three conditions also induced mRNA of both isoforms of the tolerogenic molecule cytotoxic T lymphocyte antigen 2 (CTLA-2α and CTLA-2β). Only DCs matured under CT\(^{hi}\) conditions secreted IL-1β, IL-6 and IL-23 leading to the instruction of Th17 cell polarization. In contrast, CT\(^{lo}\)- or cAMP-DCs resembled semi-mature DCs and enhanced TGF-β-dependent Foxp3\(^{+}\) iTreg conversion. iTreg conversion could be reduced using siRNA blocking of CTLA-2 and reversely, addition of recombinant CTLA-2α increased iTreg conversion in vitro. Injection of CT\(^{lo}\)- or cAMP-DCs exerted MOG peptide-specific protective effects in experimental autoimmune encephalomyelitis (EAE) by inducing Foxp3\(^{+}\) Tregs and reducing Th17 responses. Together, we identified CTLA-2 production by DCs as a novel tolerogenic mediator of TGF-β-mediated iTreg induction in vitro and in vivo. The CT-induced and cAMP-mediated up-regulation of CTLA-2 also may point to a novel immune evasion mechanism of Vibrio cholerae.}, language = {en} } @article{TuChenLimetal.2012, author = {Tu, Xiaolin and Chen, Jianquan and Lim, Joohyun and Karner, Courtney M. and Lee, Seung-Yon and Heisig, Julia and Wiese, Cornelia and Surendran, Kameswaran and Kopan, Raphael and Gessler, Manfred and Long, Fanxin}, title = {Physiological Notch Signaling Maintains Bone Homeostasis via RBPjk and Hey Upstream of NFATc1}, series = {PLoS Genetics}, volume = {8}, journal = {PLoS Genetics}, number = {3}, doi = {10.1371/journal.pgen.1002577}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-133490}, pages = {e1002577}, year = {2012}, abstract = {Notch signaling between neighboring cells controls many cell fate decisions in metazoans both during embryogenesis and in postnatal life. Previously, we uncovered a critical role for physiological Notch signaling in suppressing osteoblast differentiation in vivo. However, the contribution of individual Notch receptors and the downstream signaling mechanism have not been elucidated. Here we report that removal of Notch2, but not Notch1, from the embryonic limb mesenchyme markedly increased trabecular bone mass in adolescent mice. Deletion of the transcription factor RBPjk, a mediator of all canonical Notch signaling, in the mesenchymal progenitors but not the more mature osteoblast-lineage cells, caused a dramatic high-bone-mass phenotype characterized by increased osteoblast numbers, diminished bone marrow mesenchymal progenitor pool, and rapid age-dependent bone loss. Moreover, mice deficient in Hey1 and HeyL, two target genes of Notch-RBPjk signaling, exhibited high bone mass. Interestingly, Hey1 bound to and suppressed the NFATc1 promoter, and RBPjk deletion increased NFATc1 expression in bone. Finally, pharmacological inhibition of NFAT alleviated the high-bone-mass phenotype caused by RBPjk deletion. Thus, Notch-RBPjk signaling functions in part through Hey1-mediated inhibition of NFATc1 to suppress osteoblastogenesis, contributing to bone homeostasis in vivo.}, language = {en} } @article{BoertleinDraegerSchoenaueretal.2018, author = {B{\"o}rtlein, Charlene and Draeger, Annette and Schoenauer, Roman and Kuhlemann, Alexander and Sauer, Markus and Schneider-Schaulies, Sybille and Avota, Elita}, title = {The neutral sphingomyelinase 2 is required to polarize and sustain T Cell receptor signaling}, series = {Frontiers in Immunology}, volume = {9}, journal = {Frontiers in Immunology}, number = {815}, doi = {10.3389/fimmu.2018.00815}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-176572}, year = {2018}, abstract = {By promoting ceramide release at the cytosolic membrane leaflet, the neutral sphingomyelinase 2 (NSM) is capable of organizing receptor and signalosome segregation. Its role in T cell receptor (TCR) signaling remained so far unknown. We now show that TCR-driven NSM activation is dispensable for TCR clustering and initial phosphorylation, but of crucial importance for further signal amplification. In particular, at low doses of TCR stimulatory antibodies, NSM is required for Ca\(^{2+}\) mobilization and T cell proliferation. NSM-deficient T cells lack sustained CD3ζ and ZAP-70 phosphorylation and are unable to polarize and stabilize their microtubular system. We identified PKCζ as the key NSM downstream effector in this second wave of TCR signaling supporting dynamics of microtubule-organizing center (MTOC). Ceramide supplementation rescued PKCζ membrane recruitment and MTOC translocation in NSM-deficient cells. These findings identify the NSM as essential in TCR signaling when dynamic cytoskeletal reorganization promotes continued lateral and vertical supply of TCR signaling components: CD3ζ, Zap70, and PKCζ, and functional immune synapses are organized and stabilized via MTOC polarization.}, language = {en} } @phdthesis{Chan2002, author = {Chan, Gordon}, title = {The Role of Vav-1, Vav-2 and Lsc in NK T cell development and NK cell cytotoxicity}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-3645}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2002}, abstract = {The hematopoietic-specific Rho-family GTP exchange factor (GEF) Vav-1 is a regulator of lymphocyte antigen receptor signaling and mediates normal maturation and activation of B and T cells. Recent findings suggest that Vav-1 also forms part of signaling pathways required for natural and antibody dependent cellular cytotoxicity (ADCC) of human NK cells. In this study, I show that Vav-1 is also expressed in murine NK cells. Vav-1-/- mice had normal numbers of splenic NK cells, and these displayed a similar expression profile of NK cell receptors as cells from wild type mice. Unexpectedly, IL-2-activated Vav-1-/- NK cells retained normal ADCC. Fc-receptor mediated activation of ERK, JNK, and p38 was also normal. In contrast, Vav-1-/- NK cells exhibited reduced natural cytotoxicity against EL4, C4.4.25, RMA and RMA/S. Together, these results demonstrate that Vav-1 is dispensable for mainstream NK cell development, but is required for NK cell natural cytotoxicity. Vav-2, a protein homologous to Vav-1 has also been implicated in NK cell functions. However, NK cells from Vav-2-/- mice have normal cytotoxic activities and NK cells that lack both Vav-1 and Vav-2 exhibit similar defect as Vav-1-/- cells. Thus Vav-2 has no apparent function in the development and the activation of NK cells. Although NK cell development is normal in Vav-1-/- mice, their numbers of NKT cells were dramatically diminished. Furthermore, NKT cells from Vav-1 mutant mice failed to produce IL-4 and IFNg following in vivo CD3 stimulation. A similar loss of NKT cells was observed in Vav-1-/-Vav-2-/- mice, but not in Vav-2-/- mice, suggesting that only Vav-1, and not Vav-2, is an essential regulator of NKT cell development and NK cell cytotoxicity. Similar to Vav-1, Lsc is a Rho GEF that is expressed specifically in the hematopoietic system. It contains a regulator of G-protein signaling (RGS) domain which negatively regulates the Ga12 and Ga13 subunits of G-protein coupled receptors (GPCRs). This study shows that NK and NKT cell development are normal in Lsc-/- mice. However, NK cells from mutant mice display enhanced cytotoxic responses towards a panel of tumor cells. These data implicate for the first time a RGS-containing Rho GEF in cytotoxic responses and suggest that Lsc down-modulate NK cell activation.}, subject = {Maus}, language = {en} } @article{HaakeHaackSchaeferetal.2023, author = {Haake, Markus and Haack, Beatrice and Sch{\"a}fer, Tina and Harter, Patrick N. and Mattavelli, Greta and Eiring, Patrick and Vashist, Neha and Wedekink, Florian and Genssler, Sabrina and Fischer, Birgitt and Dahlhoff, Julia and Mokhtari, Fatemeh and Kuzkina, Anastasia and Welters, Marij J. P. and Benz, Tamara M. and Sorger, Lena and Thiemann, Vincent and Almanzar, Giovanni and Selle, Martina and Thein, Klara and Sp{\"a}th, Jacob and Gonzalez, Maria Cecilia and Reitinger, Carmen and Ipsen-Escobedo, Andrea and Wistuba-Hamprecht, Kilian and Eichler, Kristin and Filipski, Katharina and Zeiner, Pia S. and Beschorner, Rudi and Goedemans, Renske and Gogolla, Falk Hagen and Hackl, Hubert and Rooswinkel, Rogier W. and Thiem, Alexander and Romer Roche, Paula and Joshi, Hemant and P{\"u}hringer, Dirk and W{\"o}ckel, Achim and Diessner, Joachim E. and R{\"u}diger, Manfred and Leo, Eugen and Cheng, Phil F. and Levesque, Mitchell P. and Goebeler, Matthias and Sauer, Markus and Nimmerjahn, Falk and Schuberth-Wagner, Christine and Felten, Stefanie von and Mittelbronn, Michel and Mehling, Matthias and Beilhack, Andreas and van der Burg, Sjoerd H. and Riedel, Angela and Weide, Benjamin and Dummer, Reinhard and Wischhusen, J{\"o}rg}, title = {Tumor-derived GDF-15 blocks LFA-1 dependent T cell recruitment and suppresses responses to anti-PD-1 treatment}, series = {Nature Communications}, volume = {14}, journal = {Nature Communications}, doi = {10.1038/s41467-023-39817-3}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-357333}, year = {2023}, abstract = {Immune checkpoint blockade therapy is beneficial and even curative for some cancer patients. However, the majority don't respond to immune therapy. Across different tumor types, pre-existing T cell infiltrates predict response to checkpoint-based immunotherapy. Based on in vitro pharmacological studies, mouse models and analyses of human melanoma patients, we show that the cytokine GDF-15 impairs LFA-1/β2-integrin-mediated adhesion of T cells to activated endothelial cells, which is a pre-requisite of T cell extravasation. In melanoma patients, GDF-15 serum levels strongly correlate with failure of PD-1-based immune checkpoint blockade therapy. Neutralization of GDF-15 improves both T cell trafficking and therapy efficiency in murine tumor models. Thus GDF-15, beside its known role in cancer-related anorexia and cachexia, emerges as a regulator of T cell extravasation into the tumor microenvironment, which provides an even stronger rationale for therapeutic anti-GDF-15 antibody development.}, language = {en} }