@phdthesis{Weirather2014, author = {Weirather, Johannes}, title = {Role of CD4+ T lymphocytes in cardiac wound healing and remodeling after experimental myocardial infarction in mice}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-107225}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {Cardiac healing after myocardial infarction (MI) represents the cardinal prerequisite for proper replacement of the irreversibly injured myocardium. In contrast to innate immunity, the functional role of adaptive immunity in postinfarction healing has not been systematically addressed. The present study focused on the influence of CD4+ T lymphocytes on wound healing and cardiac remodeling after experimental myocardial infarction in mice. Both conventional and Foxp3+ regulatory CD4+ T cells (Treg cells) became activated in heart draining lymph nodes after MI and accumulated in the infarcted myocardium. T cell activation was strictly antigen-dependant as T cell receptor-transgenic OT-II mice in which CD4+ T cells exhibit a highly limited T cell receptor repertoire did not expand in heart-draining lymph nodes post-MI. Both OT-II and major histocompatibility complex class II-deficient mice lacking a CD4+ T cell compartment showed a fatal clinical postinfarction outcome characterized by disturbed scar tissue construction that resulted in impaired survival due to a prevalence of left-ventricular ruptures. To assess the contribution of anti-inflammatory Treg cells on wound healing after MI, the Treg cell compartment was depleted using DEREG mice that specifically express the human diphtheria toxin receptor in Foxp3-positive cells, resulting in Treg cell ablation after diphtheria toxin administration. In a parallel line of experiments, a second model of anti-CD25 antibody-mediated Treg cell immuno-depletion was used. Treg cell ablation prior to MI resulted in adverse postinfarction left-ventricular dilatation associated with cardiac deterioration. Mechanistically, Treg cell depletion resulted in an increased recruitment of pro-inflammatory neutrophils and Ly-6Chigh monocytes into the healing myocardium. Furthermore, Treg cell-ablated mice exhibited an adverse activation of conventional non-regulatory CD4+ and CD8+ T cells that showed a reinforced infiltration into the infarct zone. Increased synthesis of TNFα and IFNγ by conventional CD4+ and CD8+ T cells in hearts of Treg cell-depleted mice provoked an M1-like macrophage polarization characterized by heightened expression of healing-compromising induced NO synthase, in line with a reduced synthesis of healing-promoting transglutaminase factor XIII (FXIII), osteopontin (OPN) and transforming growth factor beta 1 (TGFβ1). Therapeutic Treg cell activation by a superagonistic anti-CD28 monoclonal antibody stimulated Treg cell accumulation in the infarct zone and led to an increased expression of mediators inducing an M2-like macrophage polarization state, i.e. interleukin-10, interleukin-13 and TGFβ1. M2-like macrophage differentiation in the healing infarct was associated with heightened expression of scar-forming procollagens as well as scar-stabilizing FXIII and OPN, resulting in improved survival due to a reduced incidence of left-ventricular ruptures. Therapeutic Treg cell activation and the induction of a beneficial M2-like macrophage polarization was further achieved by employing a treatment modality of high clinical potential, i.e. by therapeutic administration of IL-2/ anti-IL-2 monoclonal antibody complexes. The findings of the present study suggest that therapeutic Treg cell activation and the resulting improvement of healing may represent a suitable strategy to attenuate adverse infarct expansion, left-ventricular remodeling, or infarct ruptures in patients with MI.}, subject = {Antigen CD4}, language = {en} } @phdthesis{Heinrichs2018, author = {Heinrichs, Susanne Margarete}, title = {Myocardial B-cell infiltration following occlusion of the left anterior descending artery in mice is driven by CXCL13}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-168554}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2018}, abstract = {Myocardial B-cell infiltration after LAD occlusion in mice is driven by CXCL13 After myocardial infarction, the immune system is activated and regulates wound healing and remodeling processes in the heart. While the role of T cells has been elucidated already, the function of B cells in myocardial infarction remained relatively unclear until now. It is, however, already known that B cells are of importance in healing processes in other tissues, for example in the skin. Our studies therefore addressed the role and function of B cells in healing and early remodeling processes in the myocardium after infarction. Under physiological conditions, only few B cells can be found in the heart. After myocardial infarction, however, which we modelled with a permanent ligation of the left anterior descending artery (LAD) in C57BL/6J mice, we could demonstrate that B lymphocytes accumulate in the early phase after tissue injury (days one to seven) in the myocardium. To detect B cells, we performed immunofluorescence stainings on cryosections of infarcted hearts using an anti-B220 antibody. Quantitative analysis of tissue infiltration revealed that B cells peaked at day seven. In flow cytometry, we further characterized the B cells infiltrating infarcted tissue. We found that most of them were mature B cells (IgM+, IgD+). Next, we wanted to outline a potential mechanism responsible for B-cell infiltration to the site of tissue injury. We therefore performed ELISA experiments revealing that CXCL13 was upregulated in scar tissue. Antibody-mediated neutralization of CXCL13 verifiably attenuated B-cell infiltration. Treated mice also showed - in the tendency - smaller infarct sizes and an improved survival. In conclusion, we could show that B lymphocytes infiltrate the myocardium after MI in mice following a local CXCL13 gradient and that it is, most likely, beneficial to inhibit this process.}, subject = {Maus}, language = {en} } @phdthesis{vandenBerg2020, author = {van den Berg, Anne Maria}, title = {Age-related alterations of the immune system aggravate the myocardial aging process}, doi = {10.25972/OPUS-19362}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-193622}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {The prevalence of cardiovascular diseases (CVD) increases dramatically with age. Nevertheless, most of the basic research in cardiology has been conducted on young healthy animals which may not necessarily reflect the situation observed in the clinic. The heart undergoes profound changes in elderly, including molecular alterations, myocardial hypertrophy, interstitial fibrosis and functional decline. To date, numerous approaches exist to explain mechanisms of the cardiac aging process whereupon inflammation and immune activity are of increasing interest. Myocardial aging is temporally associated with chronic low-grade systemic inflammation and accumulation of memory T-cells. However, a possible causal relationship between these two phenomena has not yet been investigated. Thus, aim of the present study was to assess how immunological mechanisms contribute to the myocardial aging process. Herein, the healthy murine heart was found to harbor all major resident leukocyte populations, including macrophages (CD45+CD11b+Ly6G-), granulocytes (CD45+ CD11b+Ly6G+), T-cells (CD45+CD11b-CD3e+), B-cells (CD45+CD11b-B220+) at frequencies that largely surpass those found in skeletal muscles. Age-related structural alterations and functional impairment occur simultaneously with significant shifts of the tissue resident leukocyte composition. Gene expression analyses performed on bulk myocardial samples revealed higher expression levels of TNF and INF- suggesting that in situ inflammation plays a role in the myocardial aging process. Aging was furthermore accompanied by a significant increase in size and cellularity of mediastinal, heart draining lymph nodes (med LN). Moreover, the med LNs harvested from aged mice showed a strong accumulation of effector-memory T-cells (CD44+CD62L-), mainly exhibiting a pro-inflammatory phenotype (Foxp3-, TNF+, IFN- γ+). None of these alterations were observed in popliteal lymph nodes of aged mice, indicating that they might be site-specific. Next, to go beyond mere associative evidence and examine underlying mechanisms, the myocardial aging process was comprehensively characterized in mice lacking B- (µMT) or CD4+ T-cells (CD4ko). Our analyses revealed that aged CD4+ T-cell-deficient, but not B-cell-deficient mice, exhibit a lower in situ inflammatory tone and preserved ventricular function, as compared to age-matched wild type controls. No differences in the expression levels of genes related to fibrosis were observed in the groups. Taken together, the results of this study indicate that heart-directed immune responses may spontaneously arise in the elderly, even in the absence of a clear tissue damage or concomitant infection. The T-cell-mediated immunosenescence profile might be particularly associated with age-related myocardial inflammation and functional decline, but not with tissue remodeling. These observations might shed new light on the emerging role of T cells in myocardial diseases, which primarily affect the elderly population.}, language = {en} } @phdthesis{Gaal2022, author = {Gaal, Chiara Claudia}, title = {Cardiac Antigens and T cell Specificity after Experimental Myocardial Infarction in Mice}, doi = {10.25972/OPUS-26004}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-260047}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2022}, abstract = {Cardiovascular diseases (CVD), subsuming atherosclerosis of the coronary arteries and subsequent myocardial infarction, are the leading cause of death in the European Union (over 4 million deaths annually), with devastating individual and economic consequences. Recent studies revealed that T cells play a crucial role in post-MI inflammation, healing and remodelling processes. Nevertheless, the specificity profile of adaptive immune responses in the infarcted myocardium has not yet been differentiated. The experiments portrayed in this thesis sought to assess whether post-MI CD4+ T cell responses in mice are triggered by heart specific antigens, and eventually identify relevant epitopes. We were able to create a murine antigen atlas including a list of 206 epitopes for I-Ab and 193 epitopes for I-Ad presented on MHC-II in the context of MI. We sought to consecutively test this panel by in vitro T cell proliferation and antigen recall assays ex vivo. The elispot assay was used as a readout for antigen-specific stimulation by measurement of IL-2 and IFN-γ production, currently the most sensitive approach available to detect even small counts of antigen producing cells. Splenocytes as well as lymphocytes from mediastinal lymph nodes were purified from animals 7 days or 56 days after EMI conducted by ligation of the left anterior descending artery. We were able to provide evidence that post-MI T cell responses in Balb/c mice are triggered by heart-specific antigens and that MYHCA, especially MYHCA614-628, is relevant for that response. Moreover, a significant specific T cell response after MI in C57BL/6J mice was observed for α actin, cardiac muscle 1 [ACTC1], myosin-binding protein C3 [MYBPC3] and myosin heavy chain α [MYHCA] derived heart specific antigens. Generally, the epitopes of interest for Balb/c as well as C57BL/6J could be further investigated and may eventually be modulated in the future.}, subject = {Regulatorische T-Lymphozyt}, language = {en} } @phdthesis{Hapke2023, author = {Hapke, Nils}, title = {Cardiac antigen derived T cell epitopes in the frame of myocardial infarction}, doi = {10.25972/OPUS-30196}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-301963}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Cardiovascular disease and the acute consequence of myocardial infarc- tion remain one of the most important causes of morbidity and mortality in all western societies. While much progress has been made in mitigating the acute, life-threatening ischemia caused by infarction, heart failure of the damaged my- ocardium remains prevalent. There is mounting evidence for the role of T cells in the healing process after myocardial infarction, but relevant autoantigens, which might trigger and regulate adaptive immune involvement have not been discov- ered in patients. In this work, we discovered an autoantigenic epitope in the adrenergic receptor beta 1, which is highly expressed in the heart. This autoantigenic epitope causes a pro-inflammatory immune reaction in T cells isolated from pa- tients after myocardial infarction (MI) but not in control patients. This immune reaction was only observed in a subset of MI patients, which carry at least one allele of the HLA-DRB1*13 family. Interestingly, HLA-DRB1*13 was more com- monly expressed in patients in the MI group than in the control group. Taken together, our data suggests antigen-specific priming of T cells in MI patients, which leads to a pro-inflammatory phenotype. The primed T cells react to a cardiac derived autoantigen ex vivo and are likely to exhibit a similar phenotype in vivo. This immune phenotype was only observed in a certain sub- set of patients sharing a common HLA-allele, which was more commonly ex- pressed in MI patients, suggesting a possible role as a risk factor for cardiovas- cular disease. While our results are observational and do not have enough power to show strong clinical associations, our discoveries provide an essential tool to further our understanding of involvement of the immune system in cardiovascu- lar disease. We describe the first cardiac autoantigen in the clinical context of MI and provide an important basis for further translational and clinical research in cardiac autoimmunity.}, subject = {Immunologie}, language = {en} }