26
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome

      research-article

      Read this article at

          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

          Meuwissen and collaborators define a novel genetic cause of pseudo-TORCH syndrome, which resembles the sequelae of congenital infection and represents a novel type I interferonopathy.

          Abstract

          Pseudo-TORCH syndrome (PTS) is characterized by microcephaly, enlarged ventricles, cerebral calcification, and, occasionally, by systemic features at birth resembling the sequelae of congenital infection but in the absence of an infectious agent. Genetic defects resulting in activation of type 1 interferon (IFN) responses have been documented to cause Aicardi-Goutières syndrome, which is a cause of PTS. Ubiquitin-specific peptidase 18 (USP18) is a key negative regulator of type I IFN signaling. In this study, we identified loss-of-function recessive mutations of USP18 in five PTS patients from two unrelated families. Ex vivo brain autopsy material demonstrated innate immune inflammation with calcification and polymicrogyria. In vitro, patient fibroblasts displayed severely enhanced IFN-induced inflammation, which was completely rescued by lentiviral transduction of USP18. These findings add USP18 deficiency to the list of genetic disorders collectively termed type I interferonopathies. Moreover, USP18 deficiency represents the first genetic disorder of PTS caused by dysregulation of the response to type I IFNs. Therapeutically, this places USP18 as a promising target not only for genetic but also acquired IFN-mediated CNS disorders.

          Related collections

          Most cited references12

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

          Aicardi-Goutieres syndrome and related phenotypes: linking nucleic acid metabolism with autoimmunity.

          Aicardi-Goutières syndrome (AGS) is a genetically determined encephalopathy demonstrating phenotypic overlap both with the sequelae of congenital infection and with systemic lupus erythematosus (SLE). Recent molecular advances have revealed that AGS can be caused by mutations in any one of five genes, most commonly on a recessive basis but occasionally as a dominant trait. Like AGS, SLE is associated with a perturbation of type I interferon metabolism. Interestingly then, heterozygous mutations in the AGS1 gene TREX1 underlie a cutaneous subtype of SLE-called familial chilblain lupus, and mutations in TREX1 represent the single most common cause of monogenic SLE identified to date. Evidence is emerging to show that the nucleases defective in AGS are involved in removing endogenously produced nucleic acid (NA) species, and that a failure of this removal results in activation of the immune system. This hypothesis explains the phenotypic overlap of AGS with congenital infection and some aspects of SLE, where an equivalent type I interferon-mediated innate immune response is triggered by viral and self NAs, respectively. The combined efforts of clinicians, geneticists, immunologists and cell biologists are producing rapid progress in the understanding of AGS and overlapping autoimmune disorders. These studies provide important insights into the pathogenesis of SLE and beg urgent questions about the development and use of immunosuppressive therapies in AGS and related phenotypes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Type I interferonopathies: mendelian type I interferon up-regulation.

            The concept of grouping Mendelian disorders associated with an up-regulation of type I interferon has only recently been suggested. Here we discuss the progress being made in the delineation and understanding of this novel set of inborn errors of immunity, the human type I interferonopathies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Microcephaly with simplified gyration, epilepsy, and infantile diabetes linked to inappropriate apoptosis of neural progenitors.

              We describe a syndrome of primary microcephaly with simplified gyral pattern in combination with severe infantile epileptic encephalopathy and early-onset permanent diabetes in two unrelated consanguineous families with at least three affected children. Linkage analysis revealed a region on chromosome 18 with a significant LOD score of 4.3. In this area, two homozygous nonconserved missense mutations in immediate early response 3 interacting protein 1 (IER3IP1) were found in patients from both families. IER3IP1 is highly expressed in the fetal brain cortex and fetal pancreas and is thought to be involved in endoplasmic reticulum stress response. We reported one of these families previously in a paper on Wolcott-Rallison syndrome (WRS). WRS is characterized by increased apoptotic cell death as part of an uncontrolled unfolded protein response. Increased apoptosis has been shown to be a cause of microcephaly in animal models. An autopsy specimen from one patient showed increased apoptosis in the cerebral cortex and pancreas beta cells, implicating premature cell death as the pathogenetic mechanism. Both patient fibroblasts and control fibroblasts treated with siRNA specific for IER3IP1 showed an increased susceptibility to apoptotic cell death under stress conditions in comparison to controls. This directly implicates IER3IP1 in the regulation of cell survival. Identification of IER3IP1 mutations sheds light on the mechanisms of brain development and on the pathogenesis of infantile epilepsy and early-onset permanent diabetes. Copyright © 2011 The American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.
                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
                27 June 2016
                : 213
                : 7
                : 1163-1174
                Affiliations
                [1 ]Department of Clinical Genetics, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [2 ]Department of Bioinformatics, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [3 ]Department of Radiology, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [4 ]Department of Pathology, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [5 ]Erasmus Center for Biomics, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [6 ]Department of Child Neurology, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [7 ]Department of Neonatology, Erasmus University Medical Center, 3015 CE Rotterdam, the Netherlands
                [8 ]Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
                [9 ]Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
                [10 ]The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
                [11 ]Medical Faculty Carl Gustav Carus, Technical University of Dresden, 01069 Dresden, Germany
                [12 ]Division of Human Genetics, Medical University Innsbruck, 6020 Innsbruck, Austria
                [13 ]Institut Pasteur, Cytokine Signaling Unit, Centre National de la Recherche Scientifique URA 1961, INSERM U 1221, 75724, Paris, France
                [14 ]Institute of Neuropathology and BIOSS Center for Biological Signaling Studies, University of Freiburg, 79085 Freiburg, Germany
                [15 ]INSERM UMR 1163, Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Necker Hospital, Paris Descartes University, 75015 Paris, France
                [16 ]Manchester Centre for Genomic Medicine and Academic Health Science Centre, University of Manchester, Manchester M13 9PL, England, UK
                Author notes
                Correspondence to Grazia M.S. Mancini: g.mancini@ 123456erasmusmc.nl ; or Dusan Bogunovic: Dusan.Bogunovic@ 123456mssm.edu
                [*]

                M.E.C. Meuwissen, R. Schot, F.W. Verheijen, and S. Pellegrini contributed equally to this paper.

                [**]

                D. Bogunovic and G.M.S. Mancini contributed equally to this paper.

                M.E.C. Meuwissen’s present address is Dept. of Medical Genetics, Antwerp University Hospital, 2650 Edegen, Belgium.

                A. Bertoli Avella’s present address is Centogene, 18057 Rostock, Germany

                Author information
                http://orcid.org/0000-0001-9226-8159
                http://orcid.org/0000-0001-9578-4095
                http://orcid.org/0000-0001-7662-3851
                http://orcid.org/0000-0001-7256-2481
                http://orcid.org/0000-0002-7584-5265
                http://orcid.org/0000-0003-1174-9140
                http://orcid.org/0000-0001-9754-9144
                http://orcid.org/0000-0001-7488-1734
                http://orcid.org/0000-0002-4727-0724
                http://orcid.org/0000-0002-7470-847X
                http://orcid.org/0000-0002-0421-8301
                http://orcid.org/0000-0003-4671-6349
                http://orcid.org/0000-0003-0533-7422
                http://orcid.org/0000-0003-0446-3646
                http://orcid.org/0000-0002-3991-1800
                http://orcid.org/0000-0001-5837-7589
                Article
                20151529
                10.1084/jem.20151529
                4925017
                27325888
                f6956b9d-b3ba-40b8-95e2-6f0cd95f5a7b
                © 2016 Meuwissen 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
                : 22 September 2015
                : 17 May 2016
                Funding
                Funded by: NutsOhra Funds http://dx.doi.org/10.13039/501100003142
                Award ID: 1203-030
                Funded by: National Institute of Allergy and Infectious Diseases http://dx.doi.org/10.13039/100000060
                Award ID: R00AI106942-02
                Funded by: Institut Pasteur http://dx.doi.org/10.13039/501100003762
                Funded by: Centre National de la Recherche Scientifique http://dx.doi.org/10.13039/501100004794
                Funded by: Institut National de la Santè et de la Recherche http://dx.doi.org/10.13039/501100001677
                Funded by: European Research Council http://dx.doi.org/10.13039/501100000781
                Award ID: GA 309449
                Categories
                Research Articles
                Brief Definitive Report

                Medicine
                Medicine

                Comments

                Comment on this article