148
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines

      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.

          Abstract

          Chronic mucocutaneous candidiasis (CMC) is frequently associated with T cell immunodeficiencies. Specifically, the proinflammatory IL-17A–producing Th17 subset is implicated in protection against fungi at epithelial surfaces. In autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED, or autoimmune polyendocrine syndrome 1), CMC is often the first sign, but the underlying immunodeficiency is a long-standing puzzle. In contrast, the subsequent endocrine features are clearly autoimmune, resulting from defects in thymic self-tolerance induction caused by mutations in the autoimmune regulator ( AIRE). We report severely reduced IL-17F and IL-22 responses to both Candida albicans antigens and polyclonal stimulation in APECED patients with CMC. Surprisingly, these reductions are strongly associated with neutralizing autoantibodies to IL-17F and IL-22, whereas responses were normal and autoantibodies infrequent in APECED patients without CMC. Our multicenter survey revealed neutralizing autoantibodies against IL-17A (41%), IL-17F (75%), and/ or IL-22 (91%) in >150 APECED patients, especially those with CMC. We independently found autoantibodies against these Th17-produced cytokines in rare thymoma patients with CMC. The autoantibodies preceded the CMC in all informative cases. We conclude that IL-22 and IL-17F are key natural defenders against CMC and that the immunodeficiency underlying CMC in both patient groups has an autoimmune basis.

          Related collections

          Most cited references32

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

          Projection of an immunological self shadow within the thymus by the aire protein.

          Humans expressing a defective form of the transcription factor AIRE (autoimmune regulator) develop multiorgan autoimmune disease. We used aire- deficient mice to test the hypothesis that this transcription factor regulates autoimmunity by promoting the ectopic expression of peripheral tissue- restricted antigens in medullary epithelial cells of the thymus. This hypothesis proved correct. The mutant animals exhibited a defined profile of autoimmune diseases that depended on the absence of aire in stromal cells of the thymus. Aire-deficient thymic medullary epithelial cells showed a specific reduction in ectopic transcription of genes encoding peripheral antigens. These findings highlight the importance of thymically imposed "central" tolerance in controlling autoimmunity.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis

            The commensal fungus Candida albicans causes oropharyngeal candidiasis (OPC; thrush) in settings of immunodeficiency. Although disseminated, vaginal, and oral candidiasis are all caused by C. albicans species, host defense against C. albicans varies by anatomical location. T helper 1 (Th1) cells have long been implicated in defense against candidiasis, whereas the role of Th17 cells remains controversial. IL-17 mediates inflammatory pathology in a gastric model of mucosal candidiasis, but is host protective in disseminated disease. Here, we directly compared Th1 and Th17 function in a model of OPC. Th17-deficient (IL-23p19−/−) and IL-17R–deficient (IL-17RA−/−) mice experienced severe OPC, whereas Th1-deficient (IL-12p35−/−) mice showed low fungal burdens and no overt disease. Neutrophil recruitment was impaired in IL-23p19−/− and IL-17RA−/−, but not IL-12−/−, mice, and TCR-αβ cells were more important than TCR-γδ cells. Surprisingly, mice deficient in the Th17 cytokine IL-22 were only mildly susceptible to OPC, indicating that IL-17 rather than IL-22 is vital in defense against oral candidiasis. Gene profiling of oral mucosal tissue showed strong induction of Th17 signature genes, including CXC chemokines and β defensin-3. Saliva from Th17-deficient, but not Th1-deficient, mice exhibited reduced candidacidal activity. Thus, the Th17 lineage, acting largely through IL-17, confers the dominant response to oral candidiasis through neutrophils and antimicrobial factors.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Production of interleukin 22 but not interleukin 17 by a subset of human skin-homing memory T cells.

              Interleukin 22 (IL-22) is a cytokine produced by the T(H)-17 lineage of helper T cells and NK-22 subset of natural killer cells that acts on epithelial cells and keratinocytes and has been linked to skin homeostasis and inflammation. Here we characterize a population of human skin-homing memory CD4(+) T cells that expressed the chemokine receptors CCR10, CCR6 and CCR4 and produced IL-22 but neither IL-17 nor interferon-gamma (IFN-gamma). Clones isolated from this population produced IL-22 only and had low or undetectable expression of the T(H)-17 and T helper type 1 (T(H)1) transcription factors RORgammat and T-bet. The differentiation of T cells producing only IL-22 was efficiently induced in naive T cells by plasmacytoid dendritic cells in an IL-6- and tumor necrosis factor-dependent way. Our findings delineate a previously unknown subset of human CD4(+) effector T cells dedicated to skin pathophysiology.
                Bookmark

                Author and article information

                Journal
                J Exp Med
                J. Exp. Med
                jem
                The Journal of Experimental Medicine
                The Rockefeller University Press
                0022-1007
                1540-9538
                15 February 2010
                : 207
                : 2
                : 299-308
                Affiliations
                [1 ]Molecular Pathology Group and [2 ]Immunology Group, Institute of General and Molecular Pathology, University of Tartu, 50411 Tartu, Estonia
                [3 ]Institute of Medicine, University of Bergen and [4 ]Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
                [5 ]Centre for Medical Genetics and [6 ]Department of Pediatric Endocrinology, Diabetes and Metabolism, University Children's Hospital, 1000 Ljubljana, Slovenia
                [7 ]The Hospital for Children and Adolescents, University of Helsinki, 00290 Helsinki, Finland
                [8 ]Pediatric Clinic II, Ospedale Microcitemico and Department of Biomedical and Biotechnological Science, University of Cagliari, 09121 Cagliari, Italy
                [9 ]Department of Endocrinology and Metabolism, University of Pisa, 56124 Pisa, Italy
                [10 ]Department of Paediatrics-Neonatal Intensive Care, V. Fazzi Regional Hospital, 73100 Lecce, Italy
                [11 ]Division of Pediatric Endocrinology, University of Medicine and Dentistry of New Jersey/Robert Wood Johnson Medical School, New Brunswick, NJ 08901
                [12 ]Bioseek Clinics, New York, NY 10021
                [13 ]Department of Pathology, Tampere University Hospital, 33521 Tampere, Finland
                [14 ]Department of Endocrinology, National Institute of Endocrinology and Diabetology, 03491 Lubochna, Slovakia
                [15 ]Department of Neurology, University of Regensburg, 93053 Regensburg, Germany
                [16 ]Mannheim Medical Center, University of Heidelberg, 68135 Mannheim, Germany
                [17 ]Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, England, UK
                [18 ]Biotherapeutics Group, National Institute for Biological Standards and Control, South Mimms, EN6 3QG Hertfordshire, England, UK
                Author notes
                CORRESPONDENCE Kai Kisand: kai.kisand@ 123456ut.ee OR Anthony Meager: Anthony.Meager@ 123456nibsc.hpa.org.uk

                K. Kisand and A.S. Bøe Wolff contributed equally to this paper.

                Article
                20091669
                10.1084/jem.20091669
                2822605
                20123959
                2bb82c39-c5c9-4341-abba-3fb3b3b4069f
                © 2010 Kisand et al.

                This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.jem.org/misc/terms.shtml). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).

                History
                : 30 July 2009
                : 4 January 2010
                Categories
                Brief Definitive Report

                Medicine
                Medicine

                Comments

                Comment on this article