45
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access
      research-article
      1 , 2 , 1 , 1 , 1 , 1 , 3 , 4 , 5 , 6 , 8 , 8 , 7 , 9 , 10 , 11 , 10 , 12 , 13 , 13 , 13 , 15 , 16 , 15 , 16 , 4 , 17 , 4 , 17 , 18 , 19 , 20 , 10 , 21 , 15 , 16 , 1 , 1 , 1 , 1 , 22 , 1 , 15 , 16 , 15 , 16 , 23 , 24 , 25 , 26 , 26 , 27 , 27 , 28 , 29 , 1 , 15 , 16 , 30 , 18 , 15 , 16 , 21 , 1 , 15 , 16 , 31 , 13 , 14 , 32 , 10 , 11 , 15 , 16 , 30 , 1 , 15 , 16 , 33 , 34 , 1 , 15 , 16 ,
      The Journal of Experimental Medicine
      The Rockefeller University Press

      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

          Kreins et al. report the identification and immunological characterization of a group of TYK2-deficient patients.

          Abstract

          Autosomal recessive, complete TYK2 deficiency was previously described in a patient (P1) with intracellular bacterial and viral infections and features of hyper-IgE syndrome (HIES), including atopic dermatitis, high serum IgE levels, and staphylococcal abscesses. We identified seven other TYK2-deficient patients from five families and four different ethnic groups. These patients were homozygous for one of five null mutations, different from that seen in P1. They displayed mycobacterial and/or viral infections, but no HIES. All eight TYK2-deficient patients displayed impaired but not abolished cellular responses to (a) IL-12 and IFN-α/β, accounting for mycobacterial and viral infections, respectively; (b) IL-23, with normal proportions of circulating IL-17 + T cells, accounting for their apparent lack of mucocutaneous candidiasis; and (c) IL-10, with no overt clinical consequences, including a lack of inflammatory bowel disease. Cellular responses to IL-21, IL-27, IFN-γ, IL-28/29 (IFN-λ), and leukemia inhibitory factor (LIF) were normal. The leukocytes and fibroblasts of all seven newly identified TYK2-deficient patients, unlike those of P1, responded normally to IL-6, possibly accounting for the lack of HIES in these patients. The expression of exogenous wild-type TYK2 or the silencing of endogenous TYK2 did not rescue IL-6 hyporesponsiveness, suggesting that this phenotype was not a consequence of the TYK2 genotype. The core clinical phenotype of TYK2 deficiency is mycobacterial and/or viral infections, caused by impaired responses to IL-12 and IFN-α/β. Moreover, impaired IL-6 responses and HIES do not appear to be intrinsic features of TYK2 deficiency in humans.

          Related collections

          Most cited references74

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

          IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex.

          We report here the identification of a ligand-receptor system that, upon engagement, leads to the establishment of an antiviral state. Three closely positioned genes on human chromosome 19 encode distinct but paralogous proteins, which we designate interferon-lambda1 (IFN-lambda1), IFN-lambda2 and IFN-lambda3 (tentatively designated as IL-29, IL-28A and IL-28B, respectively, by HUGO). The expression of IFN-lambda mRNAs was inducible by viral infection in several cell lines. We identified a distinct receptor complex that is utilized by all three IFN-lambda proteins for signaling and is composed of two subunits, a receptor designated CRF2-12 (also designated as IFN-lambdaR1) and a second subunit, CRF2-4 (also known as IL-10R2). Both receptor chains are constitutively expressed on a wide variety of human cell lines and tissues and signal through the Jak-STAT (Janus kinases-signal transducers and activators of transcription) pathway. This receptor-ligand system may contribute to antiviral or other defenses by a mechanism similar to, but independent of, type I IFNs.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity.

            Chronic mucocutaneous candidiasis disease (CMCD) is characterized by recurrent or persistent infections of the skin, nails, and oral and genital mucosae caused by Candida albicans and, to a lesser extent, Staphylococcus aureus, in patients with no other infectious or autoimmune manifestations. We report two genetic etiologies of CMCD: autosomal recessive deficiency in the cytokine receptor, interleukin-17 receptor A (IL-17RA), and autosomal dominant deficiency of the cytokine interleukin-17F (IL-17F). IL-17RA deficiency is complete, abolishing cellular responses to IL-17A and IL-17F homo- and heterodimers. By contrast, IL-17F deficiency is partial, with mutant IL-17F-containing homo- and heterodimers displaying impaired, but not abolished, activity. These experiments of nature indicate that human IL-17A and IL-17F are essential for mucocutaneous immunity against C. albicans, but otherwise largely redundant.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genetic dissection of immunity to mycobacteria: the human model.

              Humans are exposed to a variety of environmental mycobacteria (EM), and most children are inoculated with live Bacille Calmette-Guérin (BCG) vaccine. In addition, most of the world's population is occasionally exposed to human-borne mycobacterial species, which are less abundant but more virulent. Although rarely pathogenic, mildly virulent mycobacteria, including BCG and most EM, may cause a variety of clinical diseases. Mycobacterium tuberculosis, M. leprae, and EM M. ulcerans are more virulent, causing tuberculosis, leprosy, and Buruli ulcer, respectively. Remarkably, only a minority of individuals develop clinical disease, even if infected with virulent mycobacteria. The interindividual variability of clinical outcome is thought to result in part from variability in the human genes that control host defense. In this well-defined microbiological and clinical context, the principles of mouse immunology and the methods of human genetics can be combined to facilitate the genetic dissection of immunity to mycobacteria. The natural infections are unique to the human model, not being found in any of the animal models of experimental infection. We review current genetic knowledge concerning the simple and complex inheritance of predisposition to mycobacterial diseases in humans. Rare patients with Mendelian disorders have been found to be vulnerable to BCG, a few EM, and M. tuberculosis. Most cases of presumed Mendelian susceptibility to these and other mycobacterial species remain unexplained. In the general population leprosy and tuberculosis have been shown to be associated with certain human genetic polymorphisms and linked to certain chromosomal regions. The causal vulnerability genes themselves have yet to be identified and their pathogenic alleles immunologically validated. The studies carried out to date have been fruitful, initiating the genetic dissection of protective immunity against a variety of mycobacterial species in natural conditions of infection. The human model has potential uses beyond the study of mycobacterial infections and may well become a model of choice for the investigation of immunity to infectious agents.
                Bookmark

                Author and article information

                Journal
                J Exp Med
                J. Exp. Med
                jem
                jem
                The Journal of Experimental Medicine
                The Rockefeller University Press
                0022-1007
                1540-9538
                21 September 2015
                : 212
                : 10
                : 1641-1662
                Affiliations
                [1 ]St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065
                [2 ]Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
                [3 ]Department of Pediatric Immunology, Uludağ University Faculty of Medicine, 16059 Görükle, Bursa, Turkey
                [4 ]Genetics Unit, Military Hospital Mohamed V, Hay Riad, 10100 Rabat, Morocco
                [5 ]Department of Pediatrics, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
                [6 ]Pediatric Respiratory Diseases Research Center; and [7 ]Department of Clinical Immunology and Infectious Diseases, Masih Daneshvari Hospital; National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, 141556153 Tehran, Iran
                [8 ]Clinical Immunology Unit, Department of Pediatrics, King Hassan II University, CHU Ibn Rochd, 20000 Casablanca, Morocco
                [9 ]Immunology Unit, Pediatric Hospital A. Fleming-OSEP, Mendoza 5500, Argentina
                [10 ]Immunology Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
                [11 ]St. Vincent’s Clinical School, University of New South Wales, Darlinghurst, New South Wales 2010, Australia
                [12 ]Immunology and Rheumatology Service, Garrahan Hospital, Buenos Aires 1408, Argentina
                [13 ]Department of Laboratory Medicine, Clinical Center; and [14 ]Primary Immunodeficiency Clinic, National Institute of Allergy and Infectious Diseases; National Institutes of Health, Bethesda, MD 20892
                [15 ]Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Enfants Malades Hospital, 75015 Paris, France
                [16 ]University Paris Descartes, Imagine Institute, 75006 Paris, France
                [17 ]Faculty of Science-Kenitra, Ibn Tofaïl University, 14000 Kenitra, Morocco
                [18 ]Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, 141 52 Stockholm, Sweden
                [19 ]Department of Clinical Research, Singapore General Hospital, Singapore 169856
                [20 ]Pediatric Hematology/Immunology, Astrid Lindgrens Children’s Hospital and Karolinska Institutet, 141 86 Stockholm, Sweden
                [21 ]Asthma Research Chair and Prince Naif Center for Immunology Research, Department of Pediatrics, College of Medicine, King Saud University, Riyadh 12372, Saudi Arabia
                [22 ]Group of Primary Immunodeficiencies, Institute of Biology, University of Antioquia UdeA, 1226 Medellín, Colombia
                [23 ]Baylor Institute for Immunology Research and [24 ]Baylor Research Institute, Dallas, TX 75204
                [25 ]Allergy and Clinical Immunology Department, Hospital Sant Joan de Deu, Barcelona University, 08950 Barcelona, Spain
                [26 ]Department of Pediatrics, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, D-80337 Munich, Germany
                [27 ]Institute of Clinical and Molecular Virology, University of Erlangen-Nuremberg, D-91054 Erlangen, Germany
                [28 ]Department of Genetics, INSERM U1163, University Paris Descartes–Sorbonne Paris Cite, Imagine Institute, Necker Enfants Malades Hospital, 75015 Paris, France
                [29 ]Institute of Biochemistry, University of Kiel, D-24098 Kiel, Germany
                [30 ]Center for the Study of Primary Immunodeficiencies, Assistance Publique–Hôpitaux de Paris, Necker Enfants Malades Hospital, 75015 Paris, France
                [31 ]Systems Biology Department, Sidra Medical and Research Center, Doha, Qatar
                [32 ]Department of Immune Regulation, Graduate School, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
                [33 ]Pediatric Immunology and Hematology Unit, Necker Enfants Malades Hospital, 75015 Paris, France
                [34 ]Howard Hughes Medical Institute, New York, NY 10065
                Author notes
                CORRESPONDENCE Stéphanie Boisson-Dupuis: stbo603@ 123456rockefeller.edu
                [*]

                M.J. Ciancanelli, S. Okada, X.-F. Kong, and N. Ramírez-Alejo contributed equally to this paper.

                [**]

                S.S. Kilic, J. El Baghdadi, S. Nonoyama, S.A. Mahdaviani, F. Ailal, A. Bousfiha, D. Mansouri, and E. Nievas contributed equally to this paper.

                [***]

                L. Hammarstrom, A. Puel, S. Al-Muhsen, L. Abel, D. Chaussabel, S.D. Rosenzweig, Y. Minegishi, S.G. Tangye, and J. Bustamante contributed equally to this paper.

                [****]

                J.-L. Casanova and S. Boisson-Dupuis contributed equally to this paper.

                Article
                20140280
                10.1084/jem.20140280
                4577846
                26304966
                3178da19-3529-4d5f-bd72-d9370dd9a9a5
                © 2015 Kreins 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.rupress.org/terms). 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
                : 11 February 2014
                : 4 August 2015
                Categories
                Article

                Medicine
                Medicine

                Comments

                Comment on this article