5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Thymic Epithelial Cell Alterations and Defective Thymopoiesis Lead to Central and Peripheral Tolerance Perturbation in MHCII Deficiency

      research-article
      1 , 2 , 3 , 1 , 4 , 5 , 6 , 4 , 5 , 1 , 1 , 6 , 1 , 5 , 7 , 8 , 9 , 8 , 10 , 11 , 12 , 13 , 14 , 14 , 15 , 16 , 17 , 18 , 19 , 19 , 20 , 21 , 22 , 22 , 23 , 24 , 25 , 6 , 1 , 5 , 1 , 22
      Frontiers in Immunology
      Frontiers Media S.A.
      thymus, thymic epithelial cells, primary immunodeficiency, MHCII, central tolerance

      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

          Major Histocompatibility Complex (MHC) class II (MHCII) deficiency (MHCII-D), also known as Bare Lymphocyte Syndrome (BLS), is a rare combined immunodeficiency due to mutations in genes regulating expression of MHCII molecules. MHCII deficiency results in impaired cellular and humoral immune responses, leading to severe infections and autoimmunity. Abnormal cross-talk with developing T cells due to the absence of MHCII expression likely leads to defects in thymic epithelial cells (TEC). However, the contribution of TEC alterations to the pathogenesis of this primary immunodeficiency has not been well characterized to date, in particular in regard to immune dysregulation. To this aim, we have performed an in-depth cellular and molecular characterization of TEC in this disease. We observed an overall perturbation of thymic structure and function in both MHCII −/− mice and patients. Transcriptomic and proteomic profiling of murine TEC revealed several alterations. In particular, we demonstrated that impairment of lymphostromal cross-talk in the thymus of MHCII −/− mice affects mTEC maturation and promiscuous gene expression and causes defects of central tolerance. Furthermore, we observed peripheral tolerance impairment, likely due to defective Treg cell generation and/or function and B cell tolerance breakdown. Overall, our findings reveal disease-specific TEC defects resulting in perturbation of central tolerance and limiting the potential benefits of hematopoietic stem cell transplantation in MHCII deficiency.

          Related collections

          Most cited references59

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2

          In comparative high-throughput sequencing assays, a fundamental task is the analysis of count data, such as read counts per gene in RNA-seq, for evidence of systematic changes across experimental conditions. Small replicate numbers, discreteness, large dynamic range and the presence of outliers require a suitable statistical approach. We present DESeq2, a method for differential analysis of count data, using shrinkage estimation for dispersions and fold changes to improve stability and interpretability of estimates. This enables a more quantitative analysis focused on the strength rather than the mere presence of differential expression. The DESeq2 package is available at http://www.bioconductor.org/packages/release/bioc/html/DESeq2.html. Electronic supplementary material The online version of this article (doi:10.1186/s13059-014-0550-8) contains supplementary material, which is available to authorized users.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Near-optimal probabilistic RNA-seq quantification.

            We present kallisto, an RNA-seq quantification program that is two orders of magnitude faster than previous approaches and achieves similar accuracy. Kallisto pseudoaligns reads to a reference, producing a list of transcripts that are compatible with each read while avoiding alignment of individual bases. We use kallisto to analyze 30 million unaligned paired-end RNA-seq reads in <10 min on a standard laptop computer. This removes a major computational bottleneck in RNA-seq analysis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymic medullary microenvironment and self-tolerance.

              Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.
                Bookmark

                Author and article information

                Contributors
                URI : https://loop.frontiersin.org/people/89474
                URI : https://loop.frontiersin.org/people/1347927
                URI : https://loop.frontiersin.org/people/32464
                URI : https://loop.frontiersin.org/people/382659
                URI : https://loop.frontiersin.org/people/632748
                URI : https://loop.frontiersin.org/people/417734
                URI : https://loop.frontiersin.org/people/793289
                URI : https://loop.frontiersin.org/people/135156
                URI : https://loop.frontiersin.org/people/30766
                URI : https://loop.frontiersin.org/people/244027
                URI : https://loop.frontiersin.org/people/593219
                URI : https://loop.frontiersin.org/people/81470
                URI : https://loop.frontiersin.org/people/28385
                URI : https://loop.frontiersin.org/people/681474
                URI : https://loop.frontiersin.org/people/25314
                URI : https://loop.frontiersin.org/people/873547
                URI : https://loop.frontiersin.org/people/1348161
                URI : https://loop.frontiersin.org/people/669361
                URI : https://loop.frontiersin.org/people/23588
                URI : https://loop.frontiersin.org/people/595855
                URI : https://loop.frontiersin.org/people/28384
                URI : https://loop.frontiersin.org/people/474683
                URI : https://loop.frontiersin.org/people/1204638
                URI : https://loop.frontiersin.org/people/24896
                URI : https://loop.frontiersin.org/people/369904
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                15 June 2021
                2021
                : 12
                : 669943
                Affiliations
                [1] 1 San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute , Milan, Italy
                [2] 2 Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute , Milan, Italy
                [3] 3 Vita-Salute San Raffaele University , Milan, Italy
                [4] 4 Human Genome Department, Humanitas Clinical and Research Center , Rozzano, Milan, Italy
                [5] 5 Milan Unit, Institute of Genetic and Biomedical Research, National Research Council (CNR) , Milan, Italy
                [6] 6 Department of Biomedical Sciences, Institute for Biomedical Technologies-National Research Council (CNR) , Milan, Italy
                [7] 7 Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute , Milan, Italy
                [8] 8 Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children’s Hospital, AP-HP , Paris, France
                [9] 9 Laboratory “Genome Dynamics in the Immune System”, INSERM UMR1163, Université de Paris, Institut Imagine , Paris, France
                [10] 10 Centre d’Etude des Déficits Immunitaires, Necker-Enfants Malades Hospital, AP-HP , Paris, France
                [11] 11 Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, University Paris Descartes Sorbonne Paris Cité, Imagine Institute , Paris, France
                [12] 12 Department of Diagnostic Sciences, Ghent University Hospital, Ghent University , Ghent, Belgium
                [13] 13 Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Ghent University Hospital , Ghent, Belgium
                [14] 14 Department of Pediatrics, University Medical Center Ulm , Ulm, Germany
                [15] 15 Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden , Dresden, Germany
                [16] 16 Cytogenetics and Medical Genetics Unit and “A. Nocivelli” Institute for Molecular Medicine, Spedali Civili Hospital, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
                [17] 17 Unit of Pediatric Immunology, Pediatrics Clinic, University of Brescia, ASST-Spedali Civili Brescia , Brescia, Italy
                [18] 18 Translational and Clinical Research Institute, Newcastle University , Newcastle upon Tyne, United Kingdom
                [19] 19 Department of Pediatric Immunology and HSCT, Great North Children's Hospital , Newcastle upon Tyne, United Kingdom
                [20] 20 Autoinflammatory Diseases and Immunodeficiencies Center, IRCCS Istituto G. Gaslini, and Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, and Maternal and Children's Sciences, University of Genoa , Genoa, Italy
                [21] 21 Division of Blood and Marrow Transplantation, Children's National Hospital , Washington, DC, United States
                [22] 22 Laboratory of Clinical Immunology and Microbiology, NIAID, NIH , Bethesda, MD, United States
                [23] 23 Division of Immunology & Allergy, Department of Pediatrics, The Hospital for Sick Children, the Canadian Centre for Primary Immunodeficiency and the University of Toronto , Toronto, ON, Canada
                [24] 24 Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia , Brescia, Italy
                [25] 25 CRS4, Science and Technology Park Polaris , Pula, Cagliari, Italy
                Author notes

                Edited by: Michele Kay Anderson, University of Toronto, Canada

                Reviewed by: Magali Irla, INSERM U1104 Centre d'immunologie de Marseille-Luminy (CIML), France; Graham Anderson, University of Birmingham, United Kingdom

                *Correspondence: Marita Bosticardo, marita.bosticardo@ 123456nih.gov

                †These authors have contributed equally to this work

                This article was submitted to T Cell Biology, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2021.669943
                8239840
                34211466
                9277eaa3-844d-4c92-8a95-c6cfa25190f8
                Copyright © 2021 Ferrua, Bortolomai, Fontana, Di Silvestre, Rigoni, Marcovecchio, Draghici, Brambilla, Castiello, Delfanti, Moshous, Picard, Taghon, Bordon, Schulz, Schuetz, Giliani, Soresina, Gennery, Signa, Dávila Saldaña, Delmonte, Notarangelo, Roifman, Poliani, Uva, Mauri, Villa and Bosticardo

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 19 February 2021
                : 07 May 2021
                Page count
                Figures: 7, Tables: 4, Equations: 0, References: 59, Pages: 20, Words: 10901
                Categories
                Immunology
                Original Research

                Immunology
                thymus,thymic epithelial cells,primary immunodeficiency,mhcii,central tolerance
                Immunology
                thymus, thymic epithelial cells, primary immunodeficiency, mhcii, central tolerance

                Comments

                Comment on this article