19
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Human Inborn Errors of Immunity: an Expanding Universe

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          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

          Molecular, cellular, and clinical studies of human inborn errors of immunity have revolutionized our understanding of their pathogenesis, considerably broadened their spectrum of immunological and clinical phenotypes, and enabled successful targeted therapeutic interventions. These studies have also been of great scientific merit, challenging a number of immunological notions initially established in inbred mice, while revealing previously unrecognized mechanisms of host defense by leukocytes and other cells, and of both innate and adaptive tolerance to self.

          Related collections

          Most cited references108

          • 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: not found

            A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function.

            Antigen stimulation of immune cells triggers Ca2+ entry through Ca2+ release-activated Ca2+ (CRAC) channels, promoting the immune response to pathogens by activating the transcription factor NFAT. We have previously shown that cells from patients with one form of hereditary severe combined immune deficiency (SCID) syndrome are defective in store-operated Ca2+ entry and CRAC channel function. Here we identify the genetic defect in these patients, using a combination of two unbiased genome-wide approaches: a modified linkage analysis with single-nucleotide polymorphism arrays, and a Drosophila RNA interference screen designed to identify regulators of store-operated Ca2+ entry and NFAT nuclear import. Both approaches converged on a novel protein that we call Orai1, which contains four putative transmembrane segments. The SCID patients are homozygous for a single missense mutation in ORAI1, and expression of wild-type Orai1 in SCID T cells restores store-operated Ca2+ influx and the CRAC current (I(CRAC)). We propose that Orai1 is an essential component or regulator of the CRAC channel complex.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Human Inborn Errors of Immunity: 2019 Update on the Classification from the International Union of Immunological Societies Expert Committee

              We report the updated classification of Inborn Errors of Immunity/Primary Immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee. This report documents the key clinical and laboratory features of 430 inborn errors of immunity, including 64 gene defects that have either been discovered in the past 2 years since the previous update (published January 2018) or were characterized earlier but have since been confirmed or expanded upon in subsequent studies. The application of next-generation sequencing continues to expedite the rapid identification of novel gene defects, rare or common; broaden the immunological and clinical phenotypes of conditions arising from known gene defects and even known variants; and implement gene-specific therapies. These advances are contributing to greater understanding of the molecular, cellular, and immunological mechanisms of disease, thereby enhancing immunological knowledge while improving the management of patients and their families. This report serves as a valuable resource for the molecular diagnosis of individuals with heritable immunological disorders and also for the scientific dissection of cellular and molecular mechanisms underlying inborn errors of immunity and related human diseases.
                Bookmark

                Author and article information

                Journal
                101688624
                45516
                Sci Immunol
                Sci Immunol
                Science immunology
                2470-9468
                31 October 2020
                10 July 2020
                10 January 2021
                : 5
                : 49
                : eabb1662
                Affiliations
                [1 ]Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
                [2 ]Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
                [3 ]St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY 10065, USA.
                [4 ]Laboratory of Human Genetics of Infectious Diseases, Necker Branch INSERM, Necker Hospital for Sick Children, Paris, France, EU.
                [5 ]Paris University, Imagine Institute, Paris, France, EU.
                [6 ]Pediatrics Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France, EU.
                [7 ]Howard Hughes Medical Institute, New York, NY 10065, USA.
                Author notes

                Author contributions: All authors (LDN, RB, JLC, HS) equally contributed to writing of the manuscript.

                [^]

                Equally contributed

                [* ]Corresponding author: Luigi D. Notarangelo, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 5-3950, 10 Center Drive, Bethesda, MD 20892, USA, Ph: (301)-761-7550, luigi.notarangelo2@ 123456nih.gov
                Article
                PMC7647049 PMC7647049 7647049 nihpa1641282
                10.1126/sciimmunol.abb1662
                7647049
                32651211
                c7030682-09b2-4175-9535-b6dc66ae451e
                History
                Categories
                Article

                Comments

                Comment on this article