@article{JodeleitPalamidesBeigeletal.2017, author = {Jodeleit, Henrika and Palamides, Pia and Beigel, Florian and Mueller, Thomas and Wolf, Eckhard and Siebeck, Matthias and Gropp, Roswitha}, title = {Design and validation of a disease network of inflammatory processes in the NSG-UC mouse model}, series = {Journal of Translational Medicine}, volume = {15}, journal = {Journal of Translational Medicine}, doi = {10.1186/s12967-017-1368-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-225516}, year = {2017}, abstract = {Background: Ulcerative colitis (UC) is a highly progressive inflammatory disease that requires the interaction of epithelial, immune, endothelial and muscle cells and fibroblasts. Previous studies suggested two inflammatory conditions in UC-patients: 'acute' and 'remodeling' and that the design of a disease network might improve the understanding of the inflammatory processes. The objective of the study was to design and validate a disease network in the NOD-SCID IL2rγ\(^{null}\) (NSG)-UC mouse model to get a better understanding of the inflammatory processes. Methods: Leukocytes were isolated from the spleen of NSG-UC mice and subjected to flow cytometric analysis. RT-PCR and RNAseq analysis were performed from distal parts of the colon. Based on these analyses and the effects of interleukins, chemokines and growth factors described in the literature, a disease network was designed. To validate the disease network the effect of infliximab and pitrakinra was tested in the NSG-UC model. A clinical- and histological score, frequencies of human leukocytes isolated from spleen and mRNA expression levels from distal parts of the colon were determined. Results: Analysis of leukocytes isolated from the spleen of challenged NSG-UC mice corroborated CD64, CD163 and CD1a expressing CD14+ monocytes, CD1a expressing CD11b+ macrophages and HGF, TARC, IFNγ and TGFß1 mRNA as inflammatory markers. The disease network suggested that a proinflammatory condition elicited by IL-17c and lipids and relayed by cytotoxic T-cells, Th17 cells and CD1a expressing macrophages and monocytes. Conversely, the remodeling condition was evoked by IL-34 and TARC and promoted by Th2 cells and M2 monocytes. Mice benefitted from treatment with infliximab as indicated by the histological- and clinical score. As predicted by the disease network infliximab reduced the proinflammatory response by suppressing M1 monocytes and CD1a expressing monocytes and macrophages and decreased levels of IFNγ, TARC and HGF mRNA. As predicted by the disease network inflammation aggravated in the presence of pitrakinra as indicated by the clinical and histological score, elevated frequencies of CD1a expressing macrophages and TNFα and IFNγ mRNA levels. Conclusions: The combination of the disease network and the NSG-UC animal model might be developed into a powerful tool to predict efficacy or in-efficacy and potential mechanistic side effects.}, language = {en} } @article{KleistMohrGaikwadetal.2016, author = {Kleist, Christian and Mohr, Elisabeth and Gaikwad, Sadanand and Dittmar, Laura and Kuerten, Stefanie and Platten, Michael and Mier, Walter and Schmitt, Michael and Opelz, Gerhard and Terness, Peter}, title = {Autoantigen-specific immunosuppression with tolerogenic peripheral blood cells prevents relapses in a mouse model of relapsing-remitting multiple sclerosis}, series = {Journal of Translational Medicine}, volume = {14}, journal = {Journal of Translational Medicine}, number = {99}, doi = {10.1186/s12967-016-0860-6}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-165787}, pages = {1-14}, year = {2016}, abstract = {Background: Dendritic cells (DCs) rendered suppressive by treatment with mitomycin C and loaded with the autoantigen myelin basic protein demonstrated earlier their ability to prevent experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis (MS). This provides an approach for prophylactic vaccination against autoimmune diseases. For clinical application such DCs are difficult to generate and autoantigens hold the risk of exacerbating the disease. Methods: We replaced DCs by peripheral mononuclear cells and myelin autoantigens by glatiramer acetate (Copaxone ®), a drug approved for the treatment of MS. Spleen cells were loaded with Copaxone®, incubated with mitomycin C (MICCop) and injected into mice after the first bout of relapsing-remitting EAE. Immunosuppression mediated by MICCop was investigated in vivo by daily assessment of clinical signs of paralysis and in in vitro restimulation assays of peripheral immune cells. Cytokine profiling was performed by enzyme-linked immunosorbent assay (ELISA). Migration of MICCop cells after injection was examined by biodistribution analysis of 111Indium-labelled MICCop. The number and inhibitory activity of CD4+CD25+FoxP3+ regulatory T cells were analysed by histology, flow cytometry and in vitro mixed lymphocyte cultures. In order to assess the specificity of MICCop-induced suppression, treated EAE mice were challenged with the control protein ovalbumin. Humoral and cellular immune responses were then determined by ELISA and in vitro antigen restimulation assay. Results: MICCop cells were able to inhibit the harmful autoreactive T-cell response and prevented mice from further relapses without affecting general immune responses. Administered MICCop migrated to various organs leading to an increased infiltration of the spleen and the central nervous system with CD4+CD25+FoxP3+ cells displaying a suppressive cytokine profile and inhibiting T-cell responses. Conclusion: We describe a clinically applicable cell therapeutic approach for controlling relapses in autoimmune encephalomyelitis by specifically silencing the deleterious autoimmune response.}, language = {en} } @phdthesis{Boerner2020, author = {B{\"o}rner, Kevin}, title = {How CLEC16A modifies the function of thymic epithelial cells}, doi = {10.25972/OPUS-20023}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-200230}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Genomweite Assoziationsstudien haben CLEC16A als ein Suszeptibilit{\"a}tsgen f{\"u}r Typ 1 Diabetes und weitere Autoimmunerkrankungen identifiziert. Die genaue Funktion von CLEC16A bleibt jedoch ungekl{\"a}rt. Studien zeigten, dass sowohl das Drosophila Ortholog ema als auch das murine Clec16a eine Rolle in Autophagie spielen. Autophagie tr{\"a}gt zur Beladung der MHC-Klasse-II Molek{\"u}le und somit der Antigenpr{\"a}sentation bei. Dar{\"u}ber hinaus konnten Studien belegen, dass Autophagie zur Antigenpr{\"a}sentation w{\"a}hrend der T-Zell Selektion in Thymus-Epithelzellen ben{\"o}tigt wird. Dies schl{\"a}gt eine m{\"o}gliche Funktion von CLEC16A in Thymus-Epithelzellen w{\"a}hrend der T-Zell Selektion vor. Außerdem berichteten Arbeiten, dass CLEC16A als quantitativer Trait Locus f{\"u}r seine Nachbargene fungiert und dass Clec16a KD in Langerhans Inseln im Pankreas die Insulinsekretion und den Glukosestoffwechsel beeintr{\"a}chtigt. Dieser Arbeit vorausgehend hatten Schuster et al. eine Clec16a KD NOD Maus generiert, welche vor spontanem autoimmunem Diabetes gesch{\"u}tzt war. F{\"u}r diese Arbeit wurde vermutet, dass CLEC16A als Suszeptibilit{\"a}tsgen f{\"u}r Typ 1 Diabetes den Prozess der Autophagie in Thymus-Epithelzellen beeintr{\"a}chtigt und somit Antigenpr{\"a}sentation und das T-Zell Repertoire beeinflusst. Um auf der Vorarbeit von Schuster et al. aufzubauen und diese zu erg{\"a}nzen, zielte diese Arbeit darauf ab, den Einfluss von CLEC16A auf Thymus-Epithelzellen zu untersuchen. Hierf{\"u}r wurde ein CLEC16A KD in menschlichen Zellen mittels RNA Interferenz erzeugt und Autophagie durch Immunoblotting untersucht. Zus{\"a}tzlich wurde die Entz{\"u}ndung im Pankreasgewebe von Clec16a KD NOD M{\"a}usen mittels H.E. F{\"a}rbung beurteilt und bewertet. Thymus-Transplanationen wurden durchgef{\"u}hrt, um zu sehen, ob der Einfluss von Clec16a KD T-Zell intrinsisch ist. Außerdem wurden intraperitoneale Glukosetoleranztests durchgef{\"u}hrt, um den Blutzuckerstoffwechsel in Clec16a KD M{\"a}usen zu beurteilen. Schließlich wurden mittels qPCR Expressionslevel der benachbarten Gene, wie zum Beispiel Dexi und Socs1, erhoben, um die Eigenschaften von CLEC16A als quantitativer Trait Locus einzuordnen. Gemeinsam mit den Ergebnissen von Schuster et al. kann diese Arbeit aufzeigen, dass Clec16a KD die Auspr{\"a}gung von Insulitis im Pankreas reduziert und Clec16a KD NOD M{\"a}use vor spontanem Autoimmundiabetes sch{\"u}tzt. Dieser Schutz vor Erkrankung wird durch beeintr{\"a}chtigte Autophagie in Thymus-Epithelzellen hervorgerufen, welche die T-Zell Selektion beeinflusst und die Reaktivit{\"a}t von T-Zellen reduziert. Der Einfluss des Clec16a KD ist innerhalb des Thymus wirksam. Der Blutzuckerstoffwechsel in Clec16a KD NOD M{\"a}usen bleibt unver{\"a}ndert und kann deshalb als Ursache f{\"u}r den Schutz vor Type 1 Diabetes ausgeschlossen werden. Clec16a und Dexi zeigen {\"a}hnliche Expressionslevel auf, dennoch ben{\"o}tigt es weitere detaillierte Studien, um eine Beziehung zwischen den beiden Genen etablieren zu k{\"o}nnen. Letztlich konnte die Beeintr{\"a}chtigung von Autophagie in menschlichen CLEC16A KD Zellen nachgewiesen werden, was bedeutet, dass die Funktion von CLEC16A evolution{\"a}r konserviert ist und ein m{\"o}glicher Zusammenhang zwischen CLEC16A Polymorphismen und einem erh{\"o}hten Risiko f{\"u}r Typ 1 Diabetes im Menschen besteht.}, subject = {Thymus}, language = {en} } @phdthesis{HerreroGonzalez2007, author = {Herrero-Gonz{\´a}lez, Josep E.}, title = {Pathogenic relevance of autoantibodies to type XVII collagen from pemphigoid gestationis patients}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-22329}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2007}, abstract = {Pemphigoid gestationis (PG) and bullous pemphigoid (BP) are subepidermal autoimmune blistering diseases characterized by self-reactive T and B cells specific for the transmembrane hemidesmosomal protein type XVII collagen/BP180. Major T and B cell epitopes are located within the immunodominant 16th non-collagenous domain A (NC16A) of type XVII collagen. It has been suggested that pathogenically relevant autoantibodies also bind to this immunodominant region. The aim of this study was to map the epitopes targeted by blister-inducing human autoantibodies. For this purpose, we used an in vitro model of autoantibody-induced leucocyte-dependent dermal-epidermal separation. In contrast to the majority of patients with BP (7 of 10), preadsorption against a recombinant form of the NC16A region abolished the blister-inducing potential of autoantibodies from all PG patients tested (n=5). Using overlapping synthetic peptides, we demonstrate that PG autoantibodies bind to 2 defined epitopes within the NC16A region (aa 500-514 and aa 511-523). Preadsorption using an affinity matrix containing these two epitopes completely abolished dermal-epidermal separation induced by PG autoantibodies (in 8 of 9 patients). These findings provide new insights into the pathogenesis of pemphigoid diseases and should prove helpful for the development of an antigen-specific immunoadsorption therapy in PG.}, language = {en} }