@article{HanitschBaumannBoztugetal.2020, author = {Hanitsch, Leif and Baumann, Ulrich and Boztug, Kaan and Burkhard-Meier, Ulrike and Fasshauer, Maria and Habermehl, Pirmin and Hauck, Fabian and Klock, Gerd and Liese, Johannes and Meyer, Oliver and M{\"u}ller, Rainer and Pachlopnik-Schmid, Jana and Pfeiffer-Kascha, Dorothea and Warnatz, Klaus and Wehr, Claudia and Wittke, Kirsten and Niehues, Tim and von Bernuth, Horst}, title = {Treatment and management of primary antibody deficiency: German interdisciplinary evidence-based consensus guideline}, series = {European Journal of Immunology}, volume = {50}, journal = {European Journal of Immunology}, number = {10}, doi = {10.1002/eji.202048713}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-225731}, pages = {1432 -- 1446}, year = {2020}, abstract = {This evidence-based clinical guideline provides consensus-recommendations for the treatment and care of patients with primary antibody deficiencies (PADs). The guideline group comprised 20 clinical and scientific expert associations of the German, Swiss, and Austrian healthcare system and representatives of patients. Recommendations were based on results of a systematic literature search, data extraction, and evaluation of methodology and study quality in combination with the clinical expertise of the respective representatives. Consensus-based recommendations were determined via nominal group technique. PADs are the largest clinically relevant group of primary immunodeficiencies. Most patients with PADs present with increased susceptibility to infections, however immune dysregulation, autoimmunity, and cancer affect a significant number of patients and may precede infections. This guideline therefore covers interdisciplinary clinical and therapeutic aspects of infectious (e.g., antibiotic prophylaxis, management of bronchiectasis) and non-infectious manifestations (e.g., management of granulomatous disease, immune cytopenia). PADs are grouped into disease entities with definitive, probable, possible, or unlikely benefit of IgG-replacement therapy. Summary and consensus-recommendations are provided for treatment indication, dosing, routes of administration, and adverse events of IgG-replacement therapy. Special aspects of concomitant impaired T-cell function are highlighted as well as clinical data on selected monogenetic inborn errors of immunity formerly classified into PADs (APDS, CTLA-4-, and LRBA-deficiency).}, language = {en} } @phdthesis{Dreykluft2013, author = {Dreykluft, Angela}, title = {The PD-1/B7-H1 Pathway in a Transgenic Mouse Model for Spontaneous Autoimmune Neuroinflammation: Immunological Studies on Devic B7-H1-/- Mice}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-83288}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {Multiple sclerosis is an autoimmune disease of the central nervous system characterized by inflammatory, demyelinating lesions and neuronal death. Formerly regarded as a variant of MS, neuromyelitis optica (NMO)/Devic's disease is now recognized as a distinct neurological disorder exhibiting characteristic inflammatory and demyelinated foci in the optic nerves and the spinal cord sparing the brain. With the introduction of the double-transgenic "Devic mouse" model featuring spontaneous, adjuvant-free incidence of autoimmune neuroinflammation due to the interaction of transgenic MOG-specific T and B cells, a promising tool was found for the analysis of factors triggering or preventing autoimmunity. The co-inhibitory molecule B7-H1 has been proposed to contribute to the maintenance of peripheral tolerance and to confine autoimmune inflammatory damage via the PD-1/B7-H1 pathway. Compared to Devic B7-H1+/+ mice, Devic B7-H1-/- mice developed clinical symptoms with a remarkably higher incidence rate and faster kinetics emphasized by deteriorated disease courses and a nearly quadrupled mortality rate. Remarkably enlarged immune-cell accumulation in the CNS of Devic B7-H1-/- mice, in particular of activated MOG-specific CD4+ T cells, correlated with the more severe clinical features. Our studies showed that the CNS not only was the major site of myelin-specific CD4+ T-cell activation but also that B7-H1 expression within the target organ significantly influenced T-cell activation and differentiation levels. Analysis at disease maximum revealed augmented accumulation of MOG-specific CD4+ T cells in the peripheral lymphoid organs of Devic B7-H1-/- mice partly due to increased T-cell proliferation rates. Transgenic MOG-specific B cells of Devic B7-H1-/- mice activated MOG-specific CD4+ T cells more efficiently than B cells of Devic B7-H1+/+ mice. This observation indicated a relevant immune-modulating role of B7-H1 on APCs (antigen-presenting cells) in this mouse model. We also assumed altered thymic selection processes to be involved in increased peripheral CD4+ T-cell numbers of Devic B7-H1-/- mice as we found more thymocytes expressing the transgenic MOG-specific T-cell receptor (TCR). Moreover, preliminary in vitro experiments hinted on an enhanced survival of TCRMOG-transgenic CD4+ T cells of Devic B7-H1-/- mice; a mechanism that might as well have led to higher peripheral T-cell accumulation. Elevated levels of MOG-specific CD4+ T cells in the periphery of Devic B7-H1-/- mice could have entailed the higher quantities in the CNS. However, mechanisms such as CNS-specific proliferation and/or apoptosis/survival could also have contributed. This should be addressed in future investigations. Judging from in vitro migration assays and adoptive transfer experiments on RAG-1-/- recipient mice, migratory behavior of MOG-specific CD4+ T cells of Devic B7-H1+/+ and Devic B7-H1-/- mice seemed not to differ. However, enhanced expression of the transmigration-relevant integrin LFA-1 on CD4+ T cells in young symptom-free Devic B7-H1-/- mice might hint on temporally differently pronounced transmigration capacities during the disease course. Moreover, we attributed the earlier conversion of CD4+ T cells into Th1 effector cells in Devic B7-H1-/- mice during the initiation phase to the lack of co-inhibitory signaling via PD-1/B7-H1 possibly leading to an accelerated disease onset. Full blown autoimmune inflammatory processes could have masked these slight effects of B7-H1 in the clinical phase. Accordingly, at peak of the disease, Th1 and Th17 effector functions of peripheral CD4+ T cells were comparable in both mouse groups. Moreover, judging from titers of MOG-specific IgG1 and IgM antibodies, alterations in humoral immunity were not detected. Therefore, clinical differences could not be explained by altered T-cell or B-cell effector functions at disease maximum. B7-H1 rather seemed to take inhibitory effect in the periphery during the initiation phase only and consistently within the target organ by parenchymal expression. Our observations indicate that B7-H1 plays a relevant role in the regulation of T-cell responses in this mouse model for spontaneous CNS autoimmunity. By exerting immune-modulating effects in the preclinical as well as the clinical phase of the disease, B7-H1 contributed to the confinement of the immunopathological tissue damage in Devic B7-H1+/+ mice mirrored by later disease onsets and lower disease scores. As a model for spontaneous autoimmunity featuring a close to 100 \% incidence rate, the Devic B7-H1-/- mouse may prove instrumental in clarifying disease-triggering and -limiting factors and in validating novel therapeutic approaches in the field of autoimmune neuroinflammation, in particular the human Devic's disease.}, subject = {Autoimmunit{\"a}t}, language = {en} } @article{LodhaErhardDoelkenetal.2022, author = {Lodha, Manivel and Erhard, Florian and D{\"o}lken, Lars and Prusty, Bhupesh K.}, title = {The hidden enemy within: non-canonical peptides in virus-induced autoimmunity}, series = {Frontiers in Microbiology}, volume = {13}, journal = {Frontiers in Microbiology}, issn = {1664-302X}, doi = {10.3389/fmicb.2022.840911}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-263053}, year = {2022}, abstract = {Viruses play a key role in explaining the pathogenesis of various autoimmune disorders, whose underlying principle is defined by the activation of autoreactive T-cells. In many cases, T-cells escape self-tolerance due to the failure in encountering certain MHC-I self-peptide complexes at substantial levels, whose peptides remain invisible from the immune system. Over the years, contribution of unstable defective ribosomal products (DRiPs) in immunosurveillance has gained prominence. A class of unstable products emerge from non-canonical translation and processing of unannotated mammalian and viral ORFs and their peptides are cryptic in nature. Indeed, high throughput sequencing and proteomics have revealed that a substantial portion of our genomes comprise of non-canonical ORFs, whose generation is significantly modulated during disease. Many of these ORFs comprise short ORFs (sORFs) and upstream ORFs (uORFs) that resemble DRiPs and may hence be preferentially presented. Here, we discuss how such products, normally "hidden" from the immune system, become abundant in viral infections activating autoimmune T-cells, by discussing their emerging role in infection and disease. Finally, we provide a perspective on how these mechanisms can explain several autoimmune disorders in the wake of the COVID-19 pandemic.}, language = {en} } @article{GirschickWolfMorbachetal.2015, author = {Girschick, Hermann and Wolf, Christine and Morbach, Henner and Hertzberg, Christoph and Lee-Kirsch, Min Ae}, title = {Severe immune dysregulation with neurological impairment and minor bone changes in a child with spondyloenchondrodysplasia due to two novel mutations in the ACP5 gene}, series = {Pediatric Rheumatology}, volume = {13}, journal = {Pediatric Rheumatology}, number = {37}, doi = {10.1186/s12969-015-0035-7}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-149990}, year = {2015}, abstract = {Spondyloenchondrodysplasia (SPENCD) is a rare skeletal dysplasia, characterized by metaphyseal lesions, neurological impairment and immune dysregulation associated with lupus-like features. SPENCD is caused by biallelic mutations in the ACP5 gene encoding tartrate-resistant phosphatase. We report on a child, who presented with spasticity, multisystem inflammation, autoimmunity and immunodeficiency with minimal metaphyseal changes due to compound heterozygosity for two novel ACP5 mutations. These findings extend the phenotypic spectrum of SPENCD and indicate that ACP5 mutations can cause severe immune dysregulation and neurological impairment even in the absence of metaphyseal dysplasia.}, language = {en} } @article{LangenhorstTabaresGuldeetal.2018, author = {Langenhorst, Daniela and Tabares, Paula and Gulde, Tobias and Becklund, Bryan R. and Berr, Susanne and Surh, Charles D. and Beyersdorf, Niklas and H{\"u}nig, Thomas}, title = {Self-recognition sensitizes mouse and human regulatory T cells to low-dose CD28 superagonist stimulation}, series = {Frontiers in Immunology}, volume = {8}, journal = {Frontiers in Immunology}, number = {1985}, doi = {10.3389/fimmu.2017.01985}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-159387}, year = {2018}, abstract = {In rodents, low doses of CD28-specific superagonistic monoclonal antibodies (CD28 superagonists, CD28SA) selectively activate regulatory T cells (Treg). This observation has recently been extended to humans, suggesting an option for the treatment of autoimmune and inflammatory diseases. However, a mechanistic explanation for this phenomenon is still lacking. Given that CD28SA amplify T cell receptor (TCR) signals, we tested the hypothesis that the weak tonic TCR signals received by conventional CD4\(^{+}\) T cells (Tconv) in the absence of cognate antigen require more CD28 signaling input for full activation than the stronger TCR signals received by self-reactive Treg. We report that in vitro, the response of mouse Treg and Tconv to CD28SA strongly depends on MHC class II expression by antigen-presenting cells. To separate the effect of tonic TCR signals from self-peptide recognition, we compared the response of wild-type Treg and Tconv to low and high CD28SA doses upon transfer into wild-type or H-2M knockout mice, which lack a self-peptide repertoire. We found that the superior response of Treg to low CD28SA doses was lost in the absence of self-peptide presentation. We also tested if potentially pathogenic autoreactive Tconv would benefit from self-recognition-induced sensitivity to CD28SA stimulation by transferring TCR transgenic OVA-specific Tconv into OVA-expressing mice and found that low-dose CD28SA application inhibited, rather than supported, their expansion, presumably due to the massive concomitant activation of Treg. Finally, we report that also in the in vitro response of human peripheral blood mononuclear cells to CD28SA, HLA II blockade interferes with the expansion of Treg by low-dose CD28SA stimulation. These results provide a rational basis for the further development of low-dose CD28SA therapy for the improvement of Treg activity.}, language = {en} } @article{MurakawaHinzMothesetal.2015, author = {Murakawa, Yasuhiro and Hinz, Michael and Mothes, Janina and Schuetz, Anja and Uhl, Michael and Wyler, Emanuel and Yasuda, Tomoharu and Mastrobuoni, Guido and Friedel, Caroline C. and D{\"o}lken, Lars and Kempa, Stefan and Schmidt-Supprian, Marc and Bl{\"u}thgen, Nils and Backofen, Rolf and Heinemann, Udo and Wolf, Jana and Scheidereit, Claus and Landthaler, Markus}, title = {RC3H1 post-transcriptionally regulates A20 mRNA and modulates the activity of the IKK/NF-\(\kappa\)B pathway}, series = {Nature Communications}, volume = {6}, journal = {Nature Communications}, number = {7367}, doi = {10.1038/ncomms8367}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-151596}, year = {2015}, abstract = {The RNA-binding protein RC3H1 (also known as ROQUIN) promotes TNF\(\alpha\) mRNA decay via a 3'UTR constitutive decay element (CDE). Here we applied PAR-CLIP to human RC3H1 to identify ~3,800 mRNA targets with >16,000 binding sites. A large number of sites are distinct from the consensus CDE and revealed a structure-sequence motif with U-rich sequences embedded in hairpins. RC3H1 binds preferentially short-lived and DNA damage-induced mRNAs, indicating a role of this RNA-binding protein in the post-transcriptional regulation of the DNA damage response. Intriguingly, RC3H1 affects expression of the NF-\(\kappa\)B pathway regulators such as I\(\kappa\)B\(\alpha\) and A20. RC3H1 uses ROQ and Zn-finger domains to contact a binding site in the A20 3'UTR, demonstrating a not yet recognized mode of RC3H1 binding. Knockdown of RC3H1 resulted in increased A20 protein expression, thereby interfering with I\(\kappa\)B kinase and NF-\(\kappa\)B activities, demonstrating that RC3H1 can modulate the activity of the IKK/NF-\(\kappa\)B pathway.}, language = {en} } @phdthesis{Visan2003, author = {Visan, Ion Lucian}, title = {P0 specific T-cell repertoire in wild-type and P0 deficient mice}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-5734}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2003}, abstract = {Zusammenfassung Das Myelinprotein P0 stellt eine zentrale Komponente f{\"u}r die Stabilit{\"a}t und Funktionalit{\"a}t der Myelinscheiden des peripheren Nervensystems dar. Mutationen des P0-Proteins f{\"u}hren zu verschiedenen, schwer behindernden peripheren Neuropathien wie der Charcot-Marie-Tooth- oder der Dejerine-Sotas-Erkrankung. Wir haben das Tiermodell der P0-Knock-Out-M{\"a}use verwendet, um im Vergleich zu den C57BL/6-Wildtyp-Tieren Selektionsmechanismen des P0-spezifischen T-Zell-Repertoires zu untersuchen. Dazu wurde eine Reihe von {\"u}berlappenden 20-mer-Peptiden benutzt, die die gesamte Aminos{\"a}uresequenz von P0 abdeckten. Mit Hilfe dieser Peptide wurde ein sog. „Epitop-Mapping" der H2-Ab-restringierten T-Zell-Antwort durchgef{\"u}hrt. Auf diese Weise konnte das P0-Peptid 5 (Aminos{\"a}ure 41-60) in der extrazellul{\"a}ren P0-Dom{\"a}ne als immunogene Determinante identifiziert werden. Dieses immunogene Peptid wurde dann f{\"u}r Untersuchungen der Toleranzmechanismen verwendet und zeigte, dass in P0-Knock-Out-M{\"a}usen ein hochreaktives P0-spezifisches T-Zell-Repertoire vorliegt, w{\"a}hrend es in Wildtyp-Tieren inaktiviert ist und so Selbsttoleranz erzeugt wird. Die Toleranzerzeugung in Wildtyp- und heterozygoten P0 +/- M{\"a}usen h{\"a}ngt nicht von der Gen-Dosis ab. P0 ist ein gewebespezifisches Antigen, dessen Expression normalerweise auf myelinisierende Schwann-Zellen beschr{\"a}nkt ist. Die klassischen Vorstellungen zu Toleranzmechanismen gegen{\"u}ber gewebsspezifischen Antigenen schrieben diese vor allem peripheren Immunmechanismen zu. Durch den erstmaligen Nachweis von intrathymischer Expression gewebsspezifischer Antigene wie P0 konnten wir best{\"a}tigen, dass f{\"u}r P0 offensichtlich die Expression deutlich weiter verbreitet ist, insbesondere auch auf Thymus-Stroma-Zellen. Unter Verwendung von Knochenmarkschim{\"a}ren haben wir weitere Untersuchungen durchgef{\"u}hrt, wie Knochenmarks-abstammende Zellen im Vergleich zu nicht-h{\"a}matopoetischen Zellen Toleranz gegen{\"u}ber P0 erzeugen k{\"o}nnen. Unsere Befunde zeigen, dass Knochenmarks-abh{\"a}ngige Zellen nicht ausreichen, um v{\"o}llige Toleranz zu erzeugen. Zus{\"a}tzlich wurde eine P0-Expression auf anderen Geweben wie dem Thymus ben{\"o}tigt, um komplette Toleranz zu erhalten. Wir identifizierten ein kryptisches P0-Peptid 8 und zwei subdominante P0-Peptide 1 und 3. W{\"a}hrend das Peptid 8 sowohl in Wildtyp- als auch Knock-Out-M{\"a}usen erkannt wurde, wurden die Peptide 1 und 3 in Wildtyp-M{\"a}usen nicht als Immunogen erkannt. Die genannten Peptide wurden verwendet, um eine experimentelle autoimmune Neuritis (EAN) zu erzeugen. Mit keinem der experimentellen Ans{\"a}tze konnten wir klinische Zeichen einer EAN generieren, allerdings mit dem Peptid 3 doch Entz{\"u}ndung im peripheren Nerven beobachten. Es werden zuk{\"u}nftig weitere Untersuchungen ben{\"o}tigt, um P0-spezifische T-Zell-Linien zu etablieren und so mit h{\"o}herer Effizienz eine EAN zu erzeugen. Unsere Untersuchungen sprechen daf{\"u}r, dass bei gentherapeutischen Ans{\"a}tzen bei erblichen Neuropathien vorsichtig und schrittweise vorgegangen werden muss, da mit sekund{\"a}rer Autoimmunit{\"a}t und damit Inflammation im peripheren Nerven zu rechnen ist.}, subject = {Myelin}, language = {en} } @phdthesis{Probst2015, author = {Probst, Lilli Teresa}, title = {Immune cell function in the Clec16a Knock-down Mouse}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-122513}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Genome wide association studies (GWAS) have identified Clec16a as disease suscepti-bility gene for numerous auto-immune disorders in particular type 1 diabetes. In spite of this strong genetic link, the role of Clec16a for immune regulation continues to be largely unknown. To study the function of Clec16a in an environment susceptible to autoimmune diseases a Clec16a deficient non obese diabetic (NOD) mouse strain was generated by means of lentiviral RNA interference. Clec16a knock down (KD) mice prove to be strongly protected against developing type 1 diabetes, an effect that is mediated by hyporeactive T effector cells. T cell hyporeactivity seems to result from an impairment of proximal TCR signalling and its cause is likely to be external to T cells. Given evidence on the involvement of the Clec16a Drosophila ortholog ema in endo- and autophagosomal processes, alterations in peripheral and/or central antigen presenting cells appeared to be potential reasons for the observed T cell hyporeactivity. While we are not able to identify any changes in quantity and quality of peripheral antigen presenting cells due to Clec16a silencing activation status of thymic epithelial cells in Clec16a KD mice deviates from NOD WT. The findings presented here suggest that thymic T cell development is affected by Clec16a variation. Such a relationship could explain the genetic association between Clec16a variations in humans and susceptibility to immune-mediated diseases, yet further investigations are needed to confirm this notion.}, language = {en} } @article{vonLukowiczSchlegelHaerteletal.2021, author = {von Lukowicz, Hannah and Schlegel, Paul-Gerhardt and H{\"a}rtel, Christoph and Morbach, Henner and Haubitz, Imme and Wiegering, Verena}, title = {ESPED survey on newly diagnosed immune thrombocytopenia in childhood: how much treatment do we give?}, series = {Molecular and Cellular Pediatrics}, volume = {8}, journal = {Molecular and Cellular Pediatrics}, doi = {10.1186/s40348-021-00121-z}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-261832}, year = {2021}, abstract = {Background Immune thrombocytopenia (ITP) is an autoimmune disease associated with isolated thrombocytopenia, which is caused by an imbalance between platelet production and platelet destruction. Petechial and mucous membrane hemorrhages are characteristic of ITP, but life-threatening bleeding rarely occurs. Depending on the bleeding symptoms, ITP can be treated with glucocorticoids (GC), intravenous immunoglobulins (IVIG), or in severe cases, platelet transfusions. Mild bleeding does not necessarily require therapy. Using the German Surveillance Unit for rare Pediatric Diseases (ESPED) we conducted a prospective survey on ITP patients in all German Children's Hospitals between September 2018 and August 2019. We collected data on ITP, including the clinical course, therapy implementation recommendations (according to the Association of German Scientific Medical Societies guidelines), outcome, and influence of treatment regimens depending on the treating physician´s experience with ITP patients. Results Of the 287 recorded cases of children with ITP, 268 questionnaires were sent to the authors. Two hundred seventeen of the questionnaires fulfilled the inclusion criteria. ITP affected boys and girls similarly, and the median age of manifestation was 3.5 years. The main reasons for hospitalization were thrombocytopenia, bleeding signs, hematomas, and/or petechiae. Bleeding scores were ≤ 3 in 96\% of children, which corresponded to a low-to-moderately low risk of bleeding. No life-threatening bleeding was documented. The most common therapies were IVIG (n = 59), GC (n = 33), or a combination of these (n = 17). Blood products (i.e., red blood cells, platelet concentrate, and fresh frozen plasma) were given to 13 patients. Compared to the established guidelines, 67 patients were over-treated, and 2 patients were under-treated. Conclusions Adherence to German ITP treatment guidelines is currently limited. To improve patient safety and medical care, better medical training and dissemination of the guidelines are required in line with targeted analyses of patients with serious bleeding events to identify potential risk constellations.}, language = {en} } @phdthesis{Werner2014, author = {Werner, Christian}, title = {Effect of autoantibodies targeting amphiphysin or glutamate decarboxylase 65 on synaptic transmission of GABAergic neurons}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-105648}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {The number of newly detected autoantibodies (AB) targeting synaptic proteins in neurological disorders of the central nervous system (CNS) is steadily increasing. Direct interactions of AB with their target antigens have been shown in first studies but the exact pathomecha-nisms for most of the already discovered AB are still unclear. The present study investigates pathophysiological mechanisms of AB-fractions that are associated with the enigmatic CNS disease Stiff person syndrome (SPS) and target the synaptically located proteins amphiphysin or glutamate decarboxylase 65 (GAD65). In the first part of the project, effects of AB to the presynaptic endocytic protein amphiphysin were investigated. Ultrastructural investigations of spinal cord presynaptic boutons in an es-tablished in-vivo passive-transfer model after intrathecal application of human anti-amphiphysin AB showed a defect of endocytosis. This defect was apparent at high synaptic activity and was characterized by reduction of the synaptic vesicle pool, clathrin coated vesi-cles (CCVs), and endosome like structures (ELS) in comparison to controls. Molecular inves-tigation of presynaptic boutons in cultured murine hippocampal neurons with dSTORM microscopy after pretreatment with AB to amphiphysin revealed that marker proteins involved in vesicle exocytosis (synaptobrevin 2 and synaptobrevin 7) had an altered expression in GA-BAergic presynapses. Endophilin, a direct binding partner of amphiphysin also displayed a disturbed expression pattern. Together, these results point towards an anti-amphiphysin AB-induced defective organization in GABAergic synapses and a presumably compensatory rearrangement of proteins responsible for CME. In the second part, functional consequences of SPS patient derived IgG fractions containing AB to GAD65, the rate limiting enzyme for GABA synthesis, were investigated by patch clamp electrophysiology and immunohistology. GABAergic neurotransmission at low and high activity as well as short term plasticity appeared normal but miniature synaptic potentials showed an enhanced frequency with constant amplitudes. SPS patient IgG after preabsorption of GAD65-AB using recombinant GAD65 still showed specific synaptic binding to neu-rons and brain slices supporting the hypothesis that additional, not yet characterized AB are present in patient IgG responsible for the exclusive effect on frequency of miniature potentials. In conclusion, the present thesis uncovered basal pathophysiological mechanisms underlying paraneoplastic SPS induced by AB to amphiphysin leading to disturbed presynaptic architec-ture. In idiopathic SPS, the hypothesis of a direct pathophysiological role of AB to GAD65 was not supported and additional IgG AB are suspected to induce distinct synaptic malfunction.}, subject = {Autoaggressionskrankheit}, language = {en} } @article{RovitusoSchefflerWunschetal.2016, author = {Rovituso, Damiano M. and Scheffler, Laura and Wunsch, Marie and Kleinschnitz, Christoph and D{\"o}rck, Sebastian and Ulzheimer, Jochen and Bayas, Antonios and Steinman, Lawrence and Erg{\"u}n, S{\"u}leyman and Kuerten, Stefanie}, title = {CEACAM1 mediates B cell aggregation in central nervous system autoimmunity}, series = {Scientific Reports}, volume = {6}, journal = {Scientific Reports}, doi = {10.1038/srep29847}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-147690}, pages = {29847}, year = {2016}, abstract = {B cell aggregates in the central nervous system (CNS) have been associated with rapid disease progression in patients with multiple sclerosis (MS). Here we demonstrate a key role of carcinoembryogenic antigen-related cell adhesion molecule1 (CEACAM1) in B cell aggregate formation in MS patients and a B cell-dependent mouse model of MS. CEACAM1 expression was increased on peripheral blood B cells and CEACAM1\(^+\) B cells were present in brain infiltrates of MS patients. Administration of the anti-CEACAM1 antibody T84.1 was efficient in blocking aggregation of B cells derived from MS patients. Along these lines, application of the monoclonal anti-CEACAM1 antibody mCC1 was able to inhibit CNS B cell aggregate formation and significantly attenuated established MS-like disease in mice in the absence of any adverse effects. CEACAM1 was co-expressed with the regulator molecule T cell immunoglobulin and mucin domain -3 (TIM-3) on B cells, a novel molecule that has recently been described to induce anergy in T cells. Interestingly, elevated coexpression on B cells coincided with an autoreactive T helper cell phenotype in MS patients. Overall, these data identify CEACAM1 as a clinically highly interesting target in MS pathogenesis and open new therapeutic avenues for the treatment of the disease.}, language = {en} } @article{FroehlichSchwaneckGernertetal.2020, author = {Froehlich, Matthias and Schwaneck, Eva C. and Gernert, Michael and Gadeholt, Ottar and Strunz, Patrick-Pascal and Morbach, Henner and Tony, Hans-Peter and Schmalzing, Marc}, title = {Autologous Stem Cell Transplantation in Common Variable Immunodeficiency: A Case of Successful Treatment of Severe Refractory Autoimmune Encephalitis}, series = {Frontiers in Immunology}, volume = {11}, journal = {Frontiers in Immunology}, number = {1317}, issn = {1664-3224}, doi = {10.3389/fimmu.2020.01317}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-206972}, year = {2020}, abstract = {Common variable immunodeficiency (CVID) is the most common primary immunodeficiency in adults. It is associated with hypogammaglobulinemia, recurring infections and autoimmune phenomena. Treatment includes immunoglobulin substitution and immunosuppressants. Autoimmune neurological manifestations of CVID are rare and occur predominantly as granulomatous disease. We report the case of a 35-year-old woman with CVID who developed autoimmune encephalitis as demonstrated by double cerebral biopsy. Infectious or malignant causes could be excluded. Despite intensive immunosuppressive therapy with common regimens no significant improvement could be achieved. Ultimately, an autologous hematopoietic stem cell transplantation (HSCT) was performed, resulting in lasting complete remission of the encephalitis. To our knowledge, this is the first report of refractory autoimmune phenomena in CVID treated by autologous HSCT.}, language = {en} } @article{KoutsilieriLutzScheller2013, author = {Koutsilieri, E. and Lutz, M. B. and Scheller, C.}, title = {Autoimmunity, dendritic cells and relevance for Parkinson's disease}, series = {Journal of Neural Transmission}, volume = {120}, journal = {Journal of Neural Transmission}, doi = {10.1007/s00702-012-0842-7}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-132308}, pages = {75-81}, year = {2013}, abstract = {Innate and adaptive immune responses in neurodegenerative diseases have become recently a focus of research and discussions. Parkinson's disease (PD) is a neurodegenerative disorder without known etiopathogenesis. The past decade has generated evidence for an involvement of the immune system in PD pathogenesis. Both inflammatory and autoimmune mechanisms have been recognized and studies have emphasized the role of activated microglia and T-cell infiltration. In this short review, we focus on dendritic cells, on their role in initiation of autoimmune responses, we discuss aspects of neuroinflammation and autoimmunity in PD, and we report new evidence for the involvement of neuromelanin in these processes.}, language = {en} }