12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Myocardial Infarction Primes Autoreactive T Cells through Activation of Dendritic Cells

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Peripheral tolerance is crucial for avoiding activation of self-reactive T cells to tissue-restricted antigens. Sterile tissue injury can break peripheral tolerance, but it is unclear how autoreactive T cells get activated in response to self. An example of a sterile injury is myocardial infarction (MI). We hypothesized that tissue necrosis is an activator of dendritic cells (DCs), which control tolerance to self-antigens. DC subsets of a murine healthy heart consisted of IRF8-dependent conventional (c)DC1, IRF4-dependent cDC2, and monocyte-derived DCs. In steady state, cardiac self-antigen α-myosin was presented in the heart-draining mediastinal lymph node (mLN) by cDC1s, driving the proliferation of antigen-specific CD4 + TCR-M T cells and their differentiation into regulatory cells (Tregs). Following MI, all DC subsets infiltrated the heart, whereas only cDCs migrated to the mLN. Here, cDC2s induced TCR-M proliferation and differentiation into interleukin-(IL)-17/interferon-(IFN)γ-producing effector cells. Thus, cardiac-specific autoreactive T cells get activated by mature DCs following myocardial infarction.

          Graphical Abstract

          Highlights

          • IRF8 + cDC1, IRF4 + cDC2, moDCs, and macrophages are the APCs of the murine heart

          • Self-antigen presentation in the steady state drives Treg development via cDC1s

          • Myocardial infarction promotes infiltration, activation, and maturation of all DCs

          • Myocardial infarction promotes priming of Th1/Th17 autoreactive T cells via cDC2s

          Abstract

          Van der Borght et al. demonstrate that myocardial infarction induces the priming of autoreactive CD4 + T cells specific for cardiac self-antigen α-myosin in the heart-draining lymph node through the maturation and migration of conventional dendritic cells. Using ex vivo co-culture systems, cDC2s are shown to be superior in presenting α-myosin.

          Related collections

          Most cited references34

          • Record: found
          • Abstract: found
          • Article: not found

          Notch–RBP-J signaling controls the homeostasis of CD8− dendritic cells in the spleen

          Signaling through Notch receptors and their transcriptional effector RBP-J is essential for lymphocyte development and function, whereas its role in other immune cell types is unclear. We tested the function of the canonical Notch–RBP-J pathway in dendritic cell (DC) development and maintenance in vivo. Genetic inactivation of RBP-J in the bone marrow did not preclude DC lineage commitment but caused the reduction of splenic DC fraction. The inactivation of RBP-J in DCs using a novel DC-specific deleter strain caused selective loss of the splenic CD8− DC subset and reduced the frequency of cytokine-secreting CD8− DCs after challenge with Toll-like receptor ligands. In contrast, other splenic DC subsets and DCs in the lymph nodes and tissues were unaffected. The RBP-J–deficient splenic CD8− DCs were depleted at the postprogenitor stage, exhibited increased apoptosis, and lost the expression of the Notch target gene Deltex1. In the spleen, CD8− DCs were found adjacent to cells expressing the Notch ligand Delta-like 1 in the marginal zone (MZ). Thus, canonical Notch–RBP-J signaling controls the maintenance of CD8− DCs in the splenic MZ, revealing an unexpected role of the Notch pathway in the innate immune system.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Deciphering the transcriptional network of the DC lineage

            Although, much progress has been made in our understanding of DC ontogeny and function, the transcriptional regulation of DC lineage commitment and functional specialization in vivo is poorly understood. We performed a comprehensive comparative analysis of CD8+, CD103+, CD11b+, and plasmacytoid DC subsets and the recently identified Macrophage DC precursors and Common DC precursors across the entire immune system. Here we characterize candidate transcriptional activators involved in myeloid progenitor commitment to the DC lineage and predicted regulators of DC functional diversity in tissues. We identify a molecular signature that distinguishes tissue DC from macrophages. We also identify a transcriptional program expressed specifically during steady-state tissue DC migration to the draining lymph nodes that may control tolerance to self-tissue antigens.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions.

              The CC chemokine receptor CCR7 has been identified as a key regulator of homeostatic B and T cell trafficking to secondary lymphoid organs. Data presented here demonstrate that CCR7 is also an essential mediator for entry of both dermal and epidermal dendritic cells (DC) into the lymphatic vessels within the dermis while this receptor is dispensable for the mobilization of Langerhans cells from the epidermis to the dermis. Moreover, a distinct population of CD11c(+)MHCII(high) DC showing low expression of the costimulatory molecules CD40, CD80, and CD86 in wild-type animals was virtually absent in skin-draining lymph nodes of CCR7-deficient mice under steady-state conditions. We provide evidence that these cells represent a semimature population of DC that is capable of initiating T cell proliferation under conditions known to induce tolerance. Thus, our data identify CCR7 as a key regulator that governs trafficking of skin DC under both inflammatory and steady-state conditions.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                21 March 2017
                21 March 2017
                21 March 2017
                : 18
                : 12
                : 3005-3017
                Affiliations
                [1 ]Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, 9052 Ghent, Belgium
                [2 ]Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
                [3 ]VIB Vesalius Research Center, 3000 Leuven, Belgium
                [4 ]Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
                [5 ]Institute of Immunobiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
                [6 ]Department of Pulmonary Medicine, ErasmusMC, 3015 Rotterdam, the Netherlands
                Author notes
                []Corresponding author peter.carmeliet@ 123456vib-kuleuven.be
                [∗∗ ]Corresponding author bart.lambrecht@ 123456ugent.be
                [7]

                Lead Contact

                Article
                S2211-1247(17)30299-1
                10.1016/j.celrep.2017.02.079
                5379012
                28329691
                19035503-a799-4178-9746-2965045095b7
                © 2017 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 3 October 2016
                : 18 January 2017
                : 27 February 2017
                Categories
                Article

                Cell biology
                dendritic cell,heart,tolerance,autoimmunity,myocardial infarction,tissue necrosis,irf4,irf8,cardiac myosin,gene expression

                Comments

                Comment on this article