101
|
Tizian C, Lahmann A, Hölsken O, Cosovanu C, Kofoed-Branzk M, Heinrich F, Mashreghi MF, Kruglov A, Diefenbach A, Neumann C. c-Maf restrains T-bet-driven programming of CCR6-negative group 3 innate lymphoid cells. eLife 2020; 9:52549. [PMID: 32039762 PMCID: PMC7025824 DOI: 10.7554/elife.52549] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
RORγt+ group 3 innate lymphoid cells (ILC3s) maintain intestinal homeostasis through secretion of type 3 cytokines such as interleukin (IL)−17 and IL-22. However, CCR6- ILC3s additionally co-express T-bet allowing for the acquisition of type 1 effector functions. While T-bet controls the type 1 programming of ILC3s, the molecular mechanisms governing T-bet are undefined. Here, we identify c-Maf as a crucial negative regulator of murine T-bet+ CCR6- ILC3s. Phenotypic and transcriptomic profiling of c-Maf-deficient CCR6- ILC3s revealed a hyper type 1 differentiation status, characterized by overexpression of ILC1/NK cell-related genes and downregulation of type 3 signature genes. On the molecular level, c-Maf directly restrained T-bet expression. Conversely, c-Maf expression was dependent on T-bet and regulated by IL-1β, IL-18 and Notch signals. Thus, we define c-Maf as a crucial cell-intrinsic brake in the type 1 effector acquisition which forms a negative feedback loop with T-bet to preserve the identity of CCR6- ILC3s.
Collapse
Affiliation(s)
- Caroline Tizian
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Annette Lahmann
- Chronic Immune Reactions, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Oliver Hölsken
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Catalina Cosovanu
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Michael Kofoed-Branzk
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Frederik Heinrich
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Andrey Kruglov
- Chronic Inflammation, Deutsches Rheuma-Forschungszentrum, Berlin, Germany.,Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russian Federation
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Christian Neumann
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
102
|
Muscari I, Adorisio S, Liberati AM, Thuy TT, Van Sung T, Cannarile L, Ayroldi E, Riccardi C, Delfino DV. Bcl-xL overexpression decreases GILZ levels and inhibits glucocorticoid-induced activation of caspase-8 and caspase-3 in mouse thymocytes. J Transl Autoimmun 2020; 3:100035. [PMID: 32803151 PMCID: PMC7423577 DOI: 10.1016/j.jtauto.2020.100035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/20/2019] [Accepted: 01/05/2020] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids promote thymocyte apoptosis and modulate transcription of numerous regulators of thymic apoptosis. Among these, glucocorticoid-induced leucine zipper (GILZ) is strongly upregulated in the thymus. We have previously demonstrated that GILZ decreases Bcl-xL expression, activates caspase-8 and caspase-3, and augments apoptosis in mice thymocytes. To better understand the causal links between glucocorticoids, GILZ, Bcl-xL, caspase-8, and caspase-3, we analyzed the thymocytes of Bcl-xL-overexpressing transgenic mice with or without glucocorticoid stimulation in vitro. Overexpression of Bcl-xL inhibited the glucocorticoid-induced up-regulation of GILZ in murine thymocytes as well as the glucocorticoid-dependent activation of caspase-8 and caspase-3. By contrast, no appreciable change in caspase-9 activation was observed upon Bcl-xL overexpression. Thus, these experiments highlighted a novel thymocyte apoptotic pathway in which Bcl-xL overexpression inhibited the glucocorticoid-induced activation of caspase-8 and caspase-3, but not caspase-9, as well as the accumulation of GILZ protein. These findings, together with our previous results showing that caspase-8 protects GILZ from proteasomal degradation, suggest the presence of a glucocorticoid-induced apoptosis self-amplification loop in which GILZ decreases Bcl-xL expression with a subsequent activation of caspase-8 and caspase-3; caspase-8 activation then enhances the stability and accumulation of GILZ and ensures the unimpeded and irreversible progression of apoptosis. By contrast, inappropriate increases in Bcl-xL levels could have catastrophic effects on thymic apoptosis as it would shut down caspase-8/3 activation, diminish the expression of GILZ, and impair the fine control necessary for thymic generation of a healthy immune repertoire. Bcl-xL is the main anti-apoptotic molecule of the BCl-2 family expressed in the thymus. Bcl-xL and GILZ are linked in a loop to influence each other’s expression. Overexpression of Bcl-xL inhibits GC-induced expression of Gilz, activation of caspase-8 and blocks thymocyte apoptosis.
Collapse
Affiliation(s)
- Isabella Muscari
- Department of Medicine, University of Perugia, Santa Maria Hospital, Terni, Italy
| | - Sabrina Adorisio
- Foligno Nursing School, Department of Medicine, University of Perugia, Foligno (PG), Italy
| | - Anna Marina Liberati
- Department of Medicine, University of Perugia, Santa Maria Hospital, Terni, Italy
| | - Trinh Thi Thuy
- Institute of Chemistry, Vietnam Academy of Science and Technology, Ha Noi, Viet Nam
| | - Tran Van Sung
- Institute of Chemistry, Vietnam Academy of Science and Technology, Ha Noi, Viet Nam
| | - Lorenza Cannarile
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Emira Ayroldi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Domenico V Delfino
- Foligno Nursing School, Department of Medicine, University of Perugia, Foligno (PG), Italy.,Section of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
103
|
Amir M, Chaudhari S, Wang R, Campbell S, Mosure SA, Chopp LB, Lu Q, Shang J, Pelletier OB, He Y, Doebelin C, Cameron MD, Kojetin DJ, Kamenecka TM, Solt LA. REV-ERBα Regulates T H17 Cell Development and Autoimmunity. Cell Rep 2019; 25:3733-3749.e8. [PMID: 30590045 PMCID: PMC6400287 DOI: 10.1016/j.celrep.2018.11.101] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 09/19/2018] [Accepted: 11/29/2018] [Indexed: 11/19/2022] Open
Abstract
RORγt is well recognized as the lineage-defining transcription factor for T helper 17 (TH17) cell development. However, the cell-intrinsic mechanisms that negatively regulate TH17 cell development and autoimmunity remain poorly understood. Here, we demonstrate that the transcriptional repressor REV-ERBα is exclusively expressed in TH17 cells, competes with RORγt for their shared DNA consensus sequence, and negatively regulates TH17 cell development via repression of genes traditionally characterized as RORγt dependent, including Il17a. Deletion of REV-ERBα enhanced TH17-mediated pro-inflammatory cytokine expression, exacerbating experimental autoimmune encephalomyelitis (EAE) and colitis. Treatment with REV-ERB-specific synthetic ligands, which have similar phenotypic properties as RORγ modulators, suppressed TH17 cell development, was effective in colitis intervention studies, and significantly decreased the onset, severity, and relapse rate in several models of EAE without affecting thymic cellularity. Our results establish that REV-ERBα negatively regulates pro-inflammatory TH17 responses in vivo and identifies the REV-ERBs as potential targets for the treatment of TH17-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Mohammed Amir
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Sweena Chaudhari
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Ran Wang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Sean Campbell
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Sarah A Mosure
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA; Scripps Research, Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, California 92037, USA; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Laura B Chopp
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Qun Lu
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA; Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Jinsai Shang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Oliver B Pelletier
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Yuanjun He
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Christelle Doebelin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Michael D Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Douglas J Kojetin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida 33458, USA; Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, USA.
| |
Collapse
|
104
|
Rafei-Shamsabadi DA, Klose CSN, Halim TYF, Tanriver Y, Jakob T. Context Dependent Role of Type 2 Innate Lymphoid Cells in Allergic Skin Inflammation. Front Immunol 2019; 10:2591. [PMID: 31781103 PMCID: PMC6851052 DOI: 10.3389/fimmu.2019.02591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022] Open
Abstract
The discovery of innate lymphoid cells (ILC) has profoundly influenced the understanding of innate and adaptive immune crosstalk in health and disease. ILC and T cells share developmental and functional characteristics such as the lineage-specifying transcription factors and effector cytokines, but importantly ILC do not display rearranged antigen-specific receptors. Similar to T cells ILC are subdivided into 3 different helper-like subtypes, namely ILC1-3, and a killer-like subtype comprising natural killer (NK) cells. Increasing evidence supports the physiological relevance of ILC, e.g., in wound healing and defense against parasites, as well as their pathogenic role in allergy, inflammatory bowel diseases or psoriasis. Group 2 ILC have been attributed to the pathogenesis of allergic diseases like asthma and atopic dermatitis. Other inflammatory skin diseases such as allergic contact dermatitis are profoundly shaped by inflammatory NK cells. This article reviews the role of ILC in allergic skin diseases with a major focus on ILC2. While group 2 ILC are suggested to contribute to the pathogenesis of type 2 dominated inflammation as seen in atopic dermatitis, we have shown that lack of ILC2 in type 1 dominated contact hypersensitivity results in enhanced inflammation, suggesting a regulatory role of ILC2 in this context. We provide a concept of how ILC2 may influence context dependent the mutual counterbalance between type I and type II immune responses in allergic skin diseases.
Collapse
Affiliation(s)
- David A. Rafei-Shamsabadi
- Allergy Research Group, Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph S. N. Klose
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Yakup Tanriver
- Institute of Medical Microbiology and Hygiene, University Medical Center Freiburg, Freiburg, Germany
- Department of Internal Medicine IV, University Medical Center Freiburg, Freiburg, Germany
| | - Thilo Jakob
- Experimental Dermatology and Allergy Research Group, Department of Dermatology and Allergology, University Medical Center Giessen, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
105
|
Jiang Y, Wang X, Dong C. Molecular mechanisms of T helper 17 cell differentiation: Emerging roles for transcription cofactors. Adv Immunol 2019; 144:121-153. [PMID: 31699215 DOI: 10.1016/bs.ai.2019.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T helper 17 (Th17) cells, characterized by secretion of IL-17 and IL-17F, are a specialized CD4+ effector T cell lineage that not only facilitates host defense against pathogen infection and maintenance of mucosal barrier, but also potently induces tissue inflammation and autoimmune diseases. Since its discovery in 2005, the developmental program of Th17 cells has been characterized, which involves a number of key cytokines, transcription factors and multiple layers of epigenetic modifications. However, how these mechanisms integrate into the complex regulatory network in Th17 cells has not been well defined. Emerging evidences have revealed essential roles of cofactors in controlling chromosome accessibilities and activities of Th17-specific transcription factors. Moreover, cofactors also act as critical signaling integrators to coordinate multiple signaling pathways and transcriptional programs. Deficiency or dysregulation of these cofactors results in defects in Th17 responses and induction of associated autoimmune diseases. Our lab has recently reported several important cofactors in Th17 cells. Here we summarize our findings regarding this new scenario of developmental regulation of Th17 cells. These findings may benefit the development of innovative strategies to treat autoimmune diseases.
Collapse
Affiliation(s)
- Yu Jiang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing, China.
| |
Collapse
|
106
|
Yahia-Cherbal H, Rybczynska M, Lovecchio D, Stephen T, Lescale C, Placek K, Larghero J, Rogge L, Bianchi E. NFAT primes the human RORC locus for RORγt expression in CD4 + T cells. Nat Commun 2019; 10:4698. [PMID: 31619674 PMCID: PMC6795897 DOI: 10.1038/s41467-019-12680-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
T helper 17 (Th17) cells have crucial functions in mucosal immunity and the pathogenesis of several chronic inflammatory diseases. The lineage-specific transcription factor, RORγt, encoded by the RORC gene modulates Th17 polarization and function, as well as thymocyte development. Here we define several regulatory elements at the human RORC locus in thymocytes and peripheral CD4+ T lymphocytes, with CRISPR/Cas9-guided deletion of these genomic segments supporting their role in RORγt expression. Mechanistically, T cell receptor stimulation induces cyclosporine A-sensitive histone modifications and P300/CBP acetylase recruitment at these elements in activated CD4+ T cells. Meanwhile, NFAT proteins bind to these regulatory elements and activate RORγt transcription in cooperation with NF-kB. Our data thus demonstrate that NFAT specifically regulate RORγt expression by binding to the RORC locus and promoting its permissive conformation. The master transcription factor RORγt, encoded by the RORC gene, controls the polarization of CD4+ T cells expressing interleukin-17 (Th17). Here the authors describe several regulatory elements at the RORC locus that are recognized by NFAT and NFkB to induce a permissive epigenetic configuration of the RORC gene for RORγt expression and Th17 differentiation.
Collapse
Affiliation(s)
- Hanane Yahia-Cherbal
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Magda Rybczynska
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France.,Laboratoire Colloides et Matériaux Divisés, École supérieure de Physique et de Chimie industrielles, Paris, France
| | - Domenica Lovecchio
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France
| | - Tharshana Stephen
- Institut Pasteur, Unité de Technologie et Service Cytométrie et Biomarqueurs (UTechS CB), Centre de recherche translationnelle (CRT), Paris, France
| | - Chloé Lescale
- Institut Pasteur, Genome Integrity, Immunity and Cancer Unit, Equipe Labellisée Ligue Contre le Cancer, Department of Immunology, Department of Genomes and Genetics, Paris, France
| | - Katarzyna Placek
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France.,Immunology and Metabolism, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jérome Larghero
- Assistance Publique-Hopitaux de Paris, Hôpital Saint-Louis, Cell Therapy Unit and Cord Blood Bank; CIC de Biothérapies, CBT501, Paris, France
| | - Lars Rogge
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France
| | - Elisabetta Bianchi
- Institut Pasteur, Immunoregulation Unit, Department of Immunology, Paris, France.
| |
Collapse
|
107
|
Gauld SB, Jacquet S, Gauvin D, Wallace C, Wang Y, McCarthy R, Goess C, Leys L, Huang S, Su Z, Edelmayer R, Wetter J, Salte K, McGaraughty SP, Argiriadi MA, Honore P, Luccarini JM, Bressac D, Desino K, Breinlinger E, Cusack K, Potin D, Kort ME, Masson PJ. Inhibition of Interleukin-23-Mediated Inflammation with a Novel Small Molecule Inverse Agonist of ROR γt. J Pharmacol Exp Ther 2019; 371:208-218. [PMID: 31375639 DOI: 10.1124/jpet.119.258046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/18/2019] [Indexed: 03/08/2025] Open
Abstract
Blockade of interleukin (IL)-23 or IL-17 with biologics is clinically validated as a treatment of psoriasis. However, the clinical impact of targeting other nodes within the IL-23/IL-17 pathway, especially with small molecules, is less defined. We report on a novel small molecule inverse agonist of retinoid acid-related orphan receptor (ROR) γt and its efficacy in preclinical models of psoriasis and arthritis. 1-(2,4-Dichloro-3-((1,4-dimethyl-6-(trifluoromethyl)-1H-indol-2-yl)methyl)benzoyl)piperidine-4-carboxylic acid (A-9758) was optimized from material identified from a high-throughput screening campaign. A-9758 is selective for RORγt and exhibits robust potency against IL-17A release both in vitro and in vivo. In vivo, we also show that IL-23 is sufficient to drive the accumulation of RORγt+ cells, and inhibition of RORγt significantly attenuates IL-23-driven psoriasiform dermatitis. Therapeutic treatment with A-9758 (i.e., delivered during active disease) was also effective in blocking skin and joint inflammation. Finally, A-9758 exhibited efficacy in an ex vivo human whole blood assay, suggesting small molecule inverse agonists of RORγt could be efficacious in human IL-17-related diseases. SIGNIFICANCE STATEMENT: Using a novel small molecule inverse agonist, and preclinical assays, we show that RORγt is a viable target for the inhibition of RORγt/Th17-driven diseases such as psoriasis. Preclinical models of psoriasis show that inhibition of RORγt blocks both the accumulation and effector function of IL-17-producing T cells.
Collapse
Affiliation(s)
- Stephen B Gauld
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Sebastien Jacquet
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Donna Gauvin
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Craig Wallace
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Yibing Wang
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Richard McCarthy
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Christian Goess
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Laura Leys
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Susan Huang
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Zhi Su
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Rebecca Edelmayer
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Joseph Wetter
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Katherine Salte
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Steven P McGaraughty
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Maria A Argiriadi
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Prisca Honore
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Jean-Michel Luccarini
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Didier Bressac
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Kelly Desino
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Eric Breinlinger
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Kevin Cusack
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Dominique Potin
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Michael E Kort
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| | - Philippe J Masson
- AbbVie Inc., North Chicago, Illinois (S.B.G., Y.W., L.L., S.H., Z.S., J.W., K.S., S.P.M., P.H., K.D., M.E.K., D.G., R.E.); Inventiva, Daix, France (J.-M.L., D.B., D.P., P.J.M., S.J.); and AbbVie Bioresearch Center, Worcester, Massachusetts (C.W., R.M., C.G., M.A.A., E.B., K.C.)
| |
Collapse
|
108
|
Lao C, Zhou X, Chen H, Wei F, Huang Z, Bai C. 5,6,7,8-Tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidine derivatives as inhibitors of full-length RORγt. Bioorg Chem 2019; 90:103077. [DOI: 10.1016/j.bioorg.2019.103077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
|
109
|
Notch and the pre-TCR coordinate thymocyte proliferation by induction of the SCF subunits Fbxl1 and Fbxl12. Nat Immunol 2019; 20:1381-1392. [PMID: 31451788 PMCID: PMC6754294 DOI: 10.1038/s41590-019-0469-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 07/16/2019] [Indexed: 01/05/2023]
Abstract
Proliferation is tightly regulated during T cell development and is limited to immature CD4−CD8− thymocytes. The major proliferative event is initiated at the ‘β-selection’ stage following successful rearrangement of Tcrβ and is triggered by and dependent on concurrent signaling by Notch and the pre-TCR; however, it is unclear how these signals cooperate to promote cell proliferation. Here we found that β-selection-associated proliferation required the combined activity of two SCF ubiquitin ligase complexes that included as substrate recognition subunits the F-box proteins Fbxl1 or Fbxl12. Both SCF complexes targeted the cyclin-dependent kinase inhibitor Cdkn1b for ubiquitinylaton and degradation. We found that Notch signals induced the transcription of Fbxl1 whereas pre-TCR signals induced the transcription of Fbxl12. Thus, concurrent Notch and pre-TCR signaling induced the expression of two genes, Fbxl1 and Fbxl12, whose products functioned identically but additively to promote degradation of Cdkn1b, cell cycle progression, and proliferation of β-selected thymocytes.
Collapse
|
110
|
Sun N, Guo H, Wang Y. Retinoic acid receptor-related orphan receptor gamma-t (RORγt) inhibitors in clinical development for the treatment of autoimmune diseases: a patent review (2016-present). Expert Opin Ther Pat 2019; 29:663-674. [DOI: 10.1080/13543776.2019.1655541] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nannan Sun
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Huimin Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
111
|
Nagatake T, Suzuki H, Hirata SI, Matsumoto N, Wada Y, Morimoto S, Nasu A, Shimojou M, Kawano M, Ogami K, Tsujimura Y, Kuroda E, Iijima N, Hosomi K, Ishii KJ, Nosaka T, Yasutomi Y, Kunisawa J. Immunological association of inducible bronchus-associated lymphoid tissue organogenesis in Ag85B-rHPIV2 vaccine-induced anti-tuberculosis mucosal immune responses in mice. Int Immunol 2019; 30:471-481. [PMID: 30011025 PMCID: PMC6153728 DOI: 10.1093/intimm/dxy046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/12/2018] [Indexed: 12/30/2022] Open
Abstract
We previously reported that Ag85B-expressing human parainfluenza type 2 virus (Ag85B-rHPIV2) was effective as a nasal vaccine against tuberculosis in mice; however, the mechanism by which it induces an immune response remains to be investigated. In the present study, we found that organogenesis of inducible bronchus-associated lymphoid tissue (iBALT) played a role in the induction of antigen-specific T cells and IgA antibody responses in the lung of mice intra-nasally administered Ag85B-rHPIV2. We found that expression of Ag85B was dispensable for the development of iBALT, suggesting that HPIV2 acted as an iBALT-inducing vector. When iBALT organogenesis was disrupted in Ag85B-rHPIV2-immunized mice, either by neutralization of the lymphotoxin pathway or depletion of CD11b+ cells, Ag85B-specific immune responses (i.e. IFN γ-producing T cells and IgA antibody) were diminished in the lung. Furthermore, we found that immunization with Ag85B-rHPIV2 induced neutrophil and eosinophil infiltration temporally after the immunization in the lung. Thus, our results show that iBALT organogenesis contributes to the induction of antigen-specific immune responses by Ag85B-rHPIV2 and that Ag85B-rHPIV2 provokes its immune responses without inducing long-lasting inflammation.
Collapse
Affiliation(s)
- Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Hidehiko Suzuki
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - So-Ichiro Hirata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan.,Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, Japan
| | - Naomi Matsumoto
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Yasuko Wada
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Sakiko Morimoto
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Ayaka Nasu
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Michiko Shimojou
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Mitsuo Kawano
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Kentaro Ogami
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, NIBIOHN, Hachimandai, Tsukuba, Ibaraki, Japan
| | - Yusuke Tsujimura
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, NIBIOHN, Hachimandai, Tsukuba, Ibaraki, Japan
| | - Etsushi Kuroda
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, NIBIOHN, Ibaraki, Osaka, Japan
| | - Norifumi Iijima
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, NIBIOHN, Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, NIBIOHN, Ibaraki, Osaka, Japan
| | - Tetsuya Nosaka
- Department of Microbiology and Molecular Genetics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Yasuhiro Yasutomi
- Laboratory of Immunoregulation and Vaccine Research, Tsukuba Primate Research Center, NIBIOHN, Hachimandai, Tsukuba, Ibaraki, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan.,Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan.,Graduate School of Medicine, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
112
|
Jetten AM, Cook DN. (Inverse) Agonists of Retinoic Acid-Related Orphan Receptor γ: Regulation of Immune Responses, Inflammation, and Autoimmune Disease. Annu Rev Pharmacol Toxicol 2019; 60:371-390. [PMID: 31386594 DOI: 10.1146/annurev-pharmtox-010919-023711] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Retinoic acid-related orphan receptor γt (RORγt) functions as a ligand-dependent transcription factor that regulates multiple proinflammatory genes and plays a critical role in several inflammatory and autoimmune diseases. Various endogenous and synthetic RORγ (inverse) agonists have been identified that regulate RORγ transcriptional activity, including many cholesterol intermediates and oxysterols. Changes in cholesterol biosynthesis and metabolism can therefore have a significant impact on the generation of oxysterol RORγ ligands and, consequently, can control RORγt activity and inflammation. These observations contribute to a growing literature that connects cholesterol metabolism to the regulation of immune responses and autoimmune disease. Loss of RORγ function in knockout mice and in mice treated with RORγ inverse agonists results in reduced production of proinflammatory cytokines, such as IL-17A/F, and increased resistance to autoimmune disease in several experimental rodent models. Thus, RORγt inverse agonists might provide an attractive therapeutic approach to treat a variety of autoimmune diseases.
Collapse
Affiliation(s)
- Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA;
| | - Donald N Cook
- Immunogenetics Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
113
|
Chuang HC, Chen YM, Chen MH, Hung WT, Yang HY, Tseng YH, Tan TH. AhR-ROR-γt complex is a therapeutic target for MAP4K3/GLK highIL-17A high subpopulation of systemic lupus erythematosus. FASEB J 2019; 33:11469-11480. [PMID: 31318609 PMCID: PMC6766655 DOI: 10.1096/fj.201900105rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The cytokine IL-17A plays critical roles in the pathogenesis of autoimmune diseases. The frequencies of MAP kinase kinase kinase kinase 3 [also named germinal center kinase-like kinase (GLK)]-overexpressing T cells are correlated with disease severity of systemic lupus erythematosus (SLE). T-cell-specific GLK-transgenic mice develop spontaneous autoimmune responses through IL-17A. GLK signaling selectively stimulates IL-17A production in murine T cells through inducing aryl hydrocarbon receptor (AhR)-retinoic acid receptor-related orphan nuclear receptor-γt (ROR-γt) complex formation. Here, we investigated whether GLK-induced AhR-ROR-γt complex in T cells is a therapeutic target for human SLE. The population of GLK+IL-17A+ T cells was enhanced in the peripheral blood from patients with SLE compared with that of healthy controls using flow cytometry. The receiver operating characteristic curve analysis showed that increased GLK+IL-17A+ T-cell population in peripheral blood reflected an active stage of SLE. In addition, peripheral blood T cells from patients with SLE displayed induction of ROR-γt phosphorylation and the AhR-ROR-γt (and AhR-phosphorylated ROR-γt) complex. Moreover, we identified a small-molecule inhibitor, verteporfin, that inhibited GLK kinase activity and AhR-ROR-γt interaction. The small-molecule inhibitor verteporfin suppressed the disease severity in autoimmune mouse models and IL-17A production in T cells from patients with SLE. Collectively, the GLK-induced AhR-ROR-γt (and AhR-phosphorylated ROR-γt) complex is a therapeutic target for the GLKhighIL-17Ahigh subpopulation of human patients with SLE.-Chuang, H.-C., Chen, Y.-M., Chen, M.-H., Hung, W.-T., Yang, H.-Y., Tseng, Y.-H., Tan, T.-H. AhR-ROR-γt complex is a therapeutic target for MAP4K3/GLKhighIL-17Ahigh subpopulation of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Ming Chen
- Division of Allergy, Immunology, and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Han Chen
- Division of Allergy, Immunology, and Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wei-Ting Hung
- Division of Allergy, Immunology, and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Huang-Yu Yang
- Department of Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yang-Hao Tseng
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
114
|
Hirose S, Wang S, Tormanen K, Wang Y, Tang J, Akbari O, Ghiasi H. Roles of Type 1, 2, and 3 Innate Lymphoid Cells in Herpes Simplex Virus 1 Infection In Vitro and In Vivo. J Virol 2019; 93:e00523-19. [PMID: 31019056 PMCID: PMC6580970 DOI: 10.1128/jvi.00523-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/16/2019] [Indexed: 01/14/2023] Open
Abstract
Innate lymphoid cells (ILCs) play important roles in host defense and inflammation. They are classified into three distinct groups based on their cytokine and chemokine secretion patterns and transcriptome profiles. Here, we show that ILCs isolated from mice can be infected with herpes simplex virus 1 (HSV-1) but that subsequent replication of the virus is compromised. After infection, type 2 ILCs expressed significantly higher levels of granulocyte colony-stimulating factor (G-CSF), interleukin 1α (IL-1α), IL-6, IL-9, RANTES, tumor necrosis factor alpha (TNF-α), CXCL1, CXCL2, CXCL10, CCL3, and CCL4 than infected type 1 or type 3 ILCs. Transcriptome-sequencing (RNA-seq) analysis of the ILCs 24 h after HSV-1 infection revealed that 77 herpesvirus genes were detected in the infected type 3 ILCs, whereas only 11 herpesvirus genes were detected in infected type 1 ILCs and 27 in infected type 2 ILCs. Compared with uninfected cells, significant upregulation of over 4,000 genes was seen in the HSV-1-infected type 3 ILCs, whereas 414 were upregulated in the infected type 1 ILCs and 128 in the infected type 2 ILCs. In contrast, in all three cell types, only a limited number of genes were significantly downregulated. Type 1, type 2, and type 3 ILC-deficient mice were used to gain insights into the effects of the ILCs on the outcome of ocular HSV-1 infection. No significant differences were found on comparison with similarly infected wild-type mice or on comparison of the three strains of deficient mice in terms of virus replication in the eyes, levels of corneal scarring, latency-reactivation in the trigeminal ganglia, or T-cell exhaustion. Although there were no significant differences in the survival rates of infected ILC-deficient mice and wild-type mice, there was significantly reduced survival of the infected type 1 or type 3 ILC-deficient mice compared with type 2 ILC-deficient mice. Adoptive transfer of wild-type T cells did not alter survival or any other parameters tested in the infected mice. Our results indicate that type 1, 2, and 3 ILCs respond differently to HSV-1 infection in vitro and that the absence of type 1 or type 3, but not type 2, ILCs affects the survival of ocularly infected mice.IMPORTANCE In this study, we investigated for the first time what roles, if any, innate lymphoid cells (ILCs) play in HSV-1 infection. Analysis of isolated ILCs in vitro revealed that all three subtypes could be infected with HSV-1 but that they were resistant to replication. The expression profiles of HSV-1-induced cytokines/chemokines and cellular and viral genes differed among the infected type 1, 2, and 3 ILCs in vitro While ILCs play no role or a redundant role in the outcomes of latency-reactivation in infected mice, absence of type 1 and type 3, but not type 2, ILCs affects the survival of infected mice.
Collapse
Affiliation(s)
- Satoshi Hirose
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shaohui Wang
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kati Tormanen
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yizhou Wang
- Genomics Core, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jie Tang
- Genomics Core, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Homayon Ghiasi
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
115
|
Sasaki T, Moro K, Kubota T, Kubota N, Kato T, Ohno H, Nakae S, Saito H, Koyasu S. Innate Lymphoid Cells in the Induction of Obesity. Cell Rep 2019; 28:202-217.e7. [DOI: 10.1016/j.celrep.2019.06.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 04/16/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022] Open
|
116
|
Shahoei SH, Nelson ER. Nuclear receptors, cholesterol homeostasis and the immune system. J Steroid Biochem Mol Biol 2019; 191:105364. [PMID: 31002862 PMCID: PMC6589364 DOI: 10.1016/j.jsbmb.2019.04.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Cholesterol is essential for maintaining membrane fluidity in eukaryotes. Additionally, the synthetic cascade of cholesterol results in precursor molecules important for cellular function such as lipid raft formation and protein prenylation. As such, cholesterol homeostasis is tightly regulated. Interestingly, it is now known that some cholesterol precursors and many metabolites serve as active signaling molecules, binding to different classes of receptors including the nuclear receptors. Furthermore, many cholesterol metabolites or their nuclear receptors have been implicated in the regulation of the immune system in normal physiology and disease. Therefore, in this focused review, cholesterol homeostasis and nuclear receptors involved in this regulation will be discussed, with particular emphasis on how these cascades influence the immune system.
Collapse
Affiliation(s)
- Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States.
| |
Collapse
|
117
|
Branzk N, Gronke K, Diefenbach A. Innate lymphoid cells, mediators of tissue homeostasis, adaptation and disease tolerance. Immunol Rev 2019; 286:86-101. [PMID: 30294961 DOI: 10.1111/imr.12718] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/05/2018] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILC) are a recently identified group of tissue-resident innate lymphocytes. Available data support the view that ILC or their progenitors are deposited and retained in tissues early during ontogeny. Thereby, ILC become an integral cellular component of tissues and organs. Here, we will review the intriguing relationships between ILC and basic developmental and homeostatic processes within tissues. Studying ILC has already led to the appreciation of the integral roles of immune cells in tissue homeostasis, morphogenesis, metabolism, regeneration, and growth. This area of immunology has not yet been studied in-depth but is likely to reveal important networks contributing to disease tolerance and may be harnessed for future therapeutic approaches.
Collapse
Affiliation(s)
- Nora Branzk
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Konrad Gronke
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| |
Collapse
|
118
|
Markiewicz A, Brożyna AA, Podgórska E, Elas M, Urbańska K, Jetten AM, Slominski AT, Jóźwicki W, Orłowska-Heitzman J, Dyduch G, Romanowska-Dixon B. Vitamin D receptors (VDR), hydroxylases CYP27B1 and CYP24A1 and retinoid-related orphan receptors (ROR) level in human uveal tract and ocular melanoma with different melanization levels. Sci Rep 2019; 9:9142. [PMID: 31235702 PMCID: PMC6591242 DOI: 10.1038/s41598-019-45161-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/23/2019] [Indexed: 12/27/2022] Open
Abstract
In recent years, a significant number of studies have investigated the preventive role of vitamin D in a number of different neoplasms. In this study, we analyze various components of the vitamin D signaling pathways in the human uveal tract and uveal melanoma, including analysis of the expression of vitamin D receptors (VDR), the activating and inactivating hydroxylases, respectively, CYP27B1 and CYP24A1, and the retinoic acid-related orphan receptors (ROR) α (RORα) and γ (RORγ) in these tissues. We further analyzed the expression of VDR, CYP27B1, CYP24A1, and ROR in relation to melanin levels, clinical stage and prognosis. Our study indicated that the uveal melanoma melanin level inversely correlated with VDR expression. We further showed that vitamin D is metabolized in uveal melanoma. This is significant because until now there has been no paper published, that would describe presence of VDR, hydroxylases CYP27B1 and CYP24A1, and RORα and RORγ in the human uveal tract and uveal melanomas. The outcomes of our research can contribute to the development of new diagnostic and therapeutic methods in uveal tract disorders, especially in uveal melanoma. The presented associations between vitamin D signaling elements and uveal melanoma in comparison to uveal tract encourage future clinical research with larger patients' population.
Collapse
Affiliation(s)
- Anna Markiewicz
- Department of Ophthalmology and Ocular Oncology, Medical College, Jagiellonian University in Kraków, 31-501, Kraków, Poland.
| | - Anna A Brożyna
- Department of Human Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University, 87-100, Toruń, Poland
| | - Ewa Podgórska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007, Kraków, Poland
| | - Martyna Elas
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007, Kraków, Poland
| | - Krystyna Urbańska
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007, Kraków, Poland
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Andrzej T Slominski
- Department of Dermatology, Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- VA Medical Center, Birmingham, AL, 35294, USA
| | - Wojciech Jóźwicki
- Department of Tumor Pathology and Pathomorphology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
- Department of Tumor Pathology and Pathomorphology, Faculty of Health Sciences, Nicolaus Copernicus University Collegium Medicum in Bydgoszcz, 85-796, Bydgoszcz, Poland
| | - Jolanta Orłowska-Heitzman
- Clinical and Experimental Pathomorphology, Jagiellonian University, Medical College, 31-531, Kraków, Poland
| | - Grzegorz Dyduch
- Clinical and Experimental Pathomorphology, Jagiellonian University, Medical College, 31-531, Kraków, Poland
| | - Bożena Romanowska-Dixon
- Department of Ophthalmology and Ocular Oncology, Medical College, Jagiellonian University in Kraków, 31-501, Kraków, Poland
| |
Collapse
|
119
|
Mahalingam D, Wang JS, Hamilton EP, Sarantopoulos J, Nemunaitis J, Weems G, Carter L, Hu X, Schreeder M, Wilkins HJ. Phase 1 Open-Label, Multicenter Study of First-in-Class RORγ Agonist LYC-55716 (Cintirorgon): Safety, Tolerability, and Preliminary Evidence of Antitumor Activity. Clin Cancer Res 2019; 25:3508-3516. [PMID: 30819679 DOI: 10.1158/1078-0432.ccr-18-3185] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/23/2019] [Accepted: 02/25/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Transcription factor retinoic acid receptor-related orphan receptor γ (RORγ) regulates type 17 effector T-cell differentiation and function and is key to immune cell regulation. Synthetic RORγ agonists modulate immune cell gene expression to increase effector T-cell activity and decrease immune suppression. A phase 1 study evaluated the safety and tolerability of LYC-55716 (cintirorgon), a first-in-class, oral, small-molecule RORγ agonist in adults with relapsed/refractory metastatic cancer. PATIENTS AND METHODS Patients received 28-day treatment cycles of oral LYC-55716; dose and dosing regimen were determined according to pharmacokinetic profile and safety. Primary endpoints were safety and tolerability. Secondary endpoints included pharmacokinetics and objective tumor response rate. RESULTS No dose-limiting toxicities occurred among the 32 enrolled patients who received LYC-55716 150 mg BID to 450 mg BID. Treatment-related adverse events (AE) were primarily grade 1-2 and included diarrhea (n = 11), fatigue (n = 7), anemia (n = 4), decreased appetite (n = 4), and nausea (n = 4). Grade 3 AEs were anemia (n = 2), elevated gamma-glutamyl transferase (n = 1), and hypophosphatemia (n = 1). Pharmacokinetic concentrations achieved levels expected for target gene regulation. Pharmacodynamic results indicated RORγ pathway engagement. Two patients (NSCLC and sarcomatoid breast cancer) had confirmed partial responses; 11 had disease stabilization for 2 to 12 months (6 received >4 months of treatment). CONCLUSIONS These data support the safety and tolerability of LYC-55716 and selection of 450 mg BID dose for a phase 2a study assessing LYC-55716 clinical activity, safety, and biomarkers in patients with NSCLC, head and neck, gastroesophageal, renal cell, urothelial, and ovarian cancers.
Collapse
Affiliation(s)
- Devalingam Mahalingam
- Department of Medicine, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Judy S Wang
- Drug Development Unit, Sarah Cannon Research Institute at Florida Cancer Specialists, Sarasota, Florida
| | - Erika P Hamilton
- Breast Cancer and Gynecologic Cancer Research Program, Sarah Cannon Research Institute, Nashville, Tennessee
| | - John Sarantopoulos
- Division of Hematology-Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - John Nemunaitis
- Executive Medical Director, Mary Crowley Cancer Research, Dallas, Texas
| | - Garry Weems
- Clinical Development, Lycera Corp., Plymouth Meeting, Pennsylvania.
| | | | - Xiao Hu
- Biology, Lycera Corp., Ann Arbor, Michigan
| | | | | |
Collapse
|
120
|
Wenink MH, Leijten EFA, Cupedo T, Radstake TRDJ. Review: Innate Lymphoid Cells: Sparking Inflammatory Rheumatic Disease? Arthritis Rheumatol 2019; 69:885-897. [PMID: 28217945 DOI: 10.1002/art.40068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Tom Cupedo
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
121
|
Klepsch V, Moschen AR, Tilg H, Baier G, Hermann-Kleiter N. Nuclear Receptors Regulate Intestinal Inflammation in the Context of IBD. Front Immunol 2019; 10:1070. [PMID: 31139192 PMCID: PMC6527601 DOI: 10.3389/fimmu.2019.01070] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/26/2019] [Indexed: 12/26/2022] Open
Abstract
Gastrointestinal (GI) homeostasis is strongly dependent on nuclear receptor (NR) functions. They play a variety of roles ranging from nutrient uptake, sensing of microbial metabolites, regulation of epithelial intestinal cell integrity to shaping of the intestinal immune cell repertoire. Several NRs are associated with GI pathologies; therefore, systematic analysis of NR biology, the underlying molecular mechanisms, and regulation of target genes can be expected to help greatly in uncovering the course of GI diseases. Recently, an increasing number of NRs has been validated as potential drug targets for therapeutic intervention in patients with inflammatory bowel disease (IBD). Besides the classical glucocorticoids, especially PPARγ, VDR, or PXR-selective ligands are currently being tested with promising results in clinical IBD trials. Also, several pre-clinical animal studies are being performed with NRs. This review focuses on the complex biology of NRs and their context-dependent anti- or pro-inflammatory activities in the regulation of gastrointestinal barrier with special attention to NRs already pharmacologically targeted in clinic and pre-clinical IBD treatment regimens.
Collapse
Affiliation(s)
- Victoria Klepsch
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander R. Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Gottfried Baier
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
122
|
|
123
|
Pantazi E, Powell N. Group 3 ILCs: Peacekeepers or Troublemakers? What's Your Gut Telling You?! Front Immunol 2019; 10:676. [PMID: 31024537 PMCID: PMC6460375 DOI: 10.3389/fimmu.2019.00676] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
A complex network of interactions exists between the microbiome, the epithelium, and immune cells that reside along the walls of the gastrointestinal tract. The intestinal immune system has been assigned with the difficult task of discriminating between commensal, harmless bacteria, and invading pathogens that translocate across the epithelial monolayer. Importantly, it is trained to maintain tolerance against commensals, and initiate protective immune responses against pathogens to secure intestinal homeostasis. Breakdown of this fine balance between the host and its intestinal microbiota can lead to intestinal inflammation and subsequently to development of inflammatory bowel disease (IBD). A decade since their discovery, innate lymphoid cells (ILCs) are now recognized as important regulators of intestinal homeostasis. ILC3s have emerged as a critical subset in the gut. They are the most phenotypically diverse ILC population and interact directly with numerous different cell types (haematopoietic and non-haematopoeitic), as well as interface with the bacterial flora. In addition to their contribution to intestinal pathogen immunity, they also mitigate against tissue damage occurring following acute injury, by facilitating tissue repair and regeneration, a key function in the maintenance of intestinal homeostasis. However, in chronic inflammation the tables are turned and ILC3s may acquire a pro-inflammatory phenotype in the gut. Chronic ILC activation can lead to persistent inflammation contributing to IBD and/or colorectal cancer. In this review, we discuss current knowledge of group 3 ILCs and their contributions to intestinal homeostasis and disease leading to novel therapeutic targets and clinical approaches that may inform novel treatment strategies for immune-mediated disorders, including IBD.
Collapse
Affiliation(s)
- Eirini Pantazi
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nick Powell
- Department of Inflammation Biology, Centre for Inflammation and Cancer Immunology, King's College London, London, United Kingdom
| |
Collapse
|
124
|
Carvalho AMS, Heimfarth L, Santos KA, Guimarães AG, Picot L, Almeida JRGS, Quintans JSS, Quintans-Júnior LJ. Terpenes as possible drugs for the mitigation of arthritic symptoms - A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:137-147. [PMID: 30668316 DOI: 10.1016/j.phymed.2018.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 05/11/2023]
Abstract
BACKGROUND Arthritis is a syndrome associated with exacerbated inflammation, joint destruction and chronic pain and disability. Chronic treatment of arthritis is associated with several side effects and high abandonment. Therefore, there has been an ongoing search for alternative treatments to overcome these problems. PURPOSE Natural products, which are already widely used for their biological, cosmetic and pharmacotechnic properties, are a possible source for new drugs. Terpenes, a large class of organic compounds produced mainly by plants and trees, are a promising natural product and have already been shown to be effective in treating chronic pain, particularly of an inflammatory origin. STUDY DESIGN AND METHODS This review identifies the main terpenes with anti-arthritic activity reported in the last 10 years. A survey was conducted between December 2017 and June 2018 in the PUBMED, SCOPUS and Science Direct databases using combinations of the descriptors terpenes, arthritis and inflammation. RESULTS The results showed that terpenes have promising biological effects in relation to the treatment of arthritis, with the 24 terpenes identified in our survey being effective in the modulation of inflammatory mediators important to the physiopathology of arthritis, such as IL-6, IL-17, TNF-α, NFκB, and COX-2, among others. It is important to note that most of the studies used animal models, which limits, at least in part, the direct translation to humans of the experimental evidence produced by the studies. CONCLUSION Together, our finds suggest that terpenes can modulate the immuno-regulatory and destructive tissue events that underlie the clinical presentation and the progression of arthritis and are worthy of further clinical investigation.
Collapse
Affiliation(s)
- Alexandra M S Carvalho
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil
| | - Luana Heimfarth
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil
| | - Klécia A Santos
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil
| | - Adriana G Guimarães
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil
| | - Laurent Picot
- UMRi CNRS 7266 LIENSs, University of La Rochelle, 17042 La Rochelle, France.
| | | | - Jullyana S S Quintans
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil.
| | - Lucindo J Quintans-Júnior
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Av. Marechal Rondon, SN, Rosa Elze, São Cristóvão, SE, Brazil.
| |
Collapse
|
125
|
Hirahara K, Shinoda K, Morimoto Y, Kiuchi M, Aoki A, Kumagai J, Kokubo K, Nakayama T. Immune Cell-Epithelial/Mesenchymal Interaction Contributing to Allergic Airway Inflammation Associated Pathology. Front Immunol 2019; 10:570. [PMID: 30972065 PMCID: PMC6443630 DOI: 10.3389/fimmu.2019.00570] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/04/2019] [Indexed: 11/13/2022] Open
Abstract
The primary function of the lung is efficient gas exchange between alveolar air and alveolar capillary blood. At the same time, the lung protects the host from continuous invasion of harmful viruses and bacteria by developing unique epithelial barrier systems. Thus, the lung has a complex architecture comprising a mixture of various types of cells including epithelial cells, mesenchymal cells, and immune cells. Recent studies have revealed that Interleukin (IL-)33, a member of the IL-1 family of cytokines, is a key environmental cytokine that is derived from epithelial cells and induces type 2 inflammation in the barrier organs, including the lung. IL-33 induces allergic diseases, such as asthma, through the activation of various immune cells that express an IL-33 receptor, ST2, including ST2+ memory (CD62LlowCD44hi) CD4+ T cells. ST2+ memory CD4+ T cells have the capacity to produce high levels of IL-5 and Amphiregulin and are involved in the pathology of asthma. ST2+ memory CD4+ T cells are maintained by IL-7- and IL-33-produced lymphatic endothelial cells within inducible bronchus-associated lymphoid tissue (iBALT) around the bronchioles during chronic lung inflammation. In this review, we will discuss the impact of these immune cells-epithelial/mesenchymal interaction on shaping the pathology of chronic allergic inflammation. A better understanding of pathogenic roles of the cellular and molecular interaction between immune cells and non-immune cells is crucial for the development of new therapeutic strategies for intractable allergic diseases.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,AMED-PRIME, AMED, Chiba, Japan
| | - Kenta Shinoda
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Laboratory of Genome Integrity, National Institutes of Health, Bethesda, MD, United States
| | - Yuki Morimoto
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ami Aoki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jin Kumagai
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,AMED-CREST, AMED, Chiba, Japan
| |
Collapse
|
126
|
Roch B, Abramowski V, Chaumeil J, de Villartay JP. Cernunnos/Xlf Deficiency Results in Suboptimal V(D)J Recombination and Impaired Lymphoid Development in Mice. Front Immunol 2019; 10:443. [PMID: 30923523 PMCID: PMC6426757 DOI: 10.3389/fimmu.2019.00443] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/19/2019] [Indexed: 12/23/2022] Open
Abstract
Xlf/Cernunnos is unique among the core factors of the non-homologous end joining (NHEJ) DNA double strand breaks (DSBs) repair pathway, in the sense that it is not essential for V(D)J recombination in vivo and in vitro. Unlike other NHEJ deficient mice showing a SCID phenotype, Xlf−/− mice present a unique immune phenotype with a moderate B- and T-cell lymphopenia, a decreased cellularity in the thymus, and a characteristic TCRα repertoire bias associated with the P53-dependent apoptosis of CD4+CD8+ DP thymocytes. Here, we thoroughly analyzed Xlf−/− mice immune phenotype and showed that it is specifically related to the DP stage but independent of the MHC-driven antigen presentation and T-cell activation during positive selection. Instead, we show that V(D)J recombination is subefficient in Xlf−/− mice in vivo, exemplified by the presence of unrepaired DSBs in the thymus. This results in a moderate developmental delay of both B- and T-lymphocytes at key V(D)J recombination dependent stages. Furthermore, subefficient V(D)J recombination waves are accumulating during TCRα rearrangement, causing the typical TCRα repertoire bias with loss of distal Vα and Jα rearrangements.
Collapse
Affiliation(s)
- Benoit Roch
- Laboratory "Genome Dynamics in the Immune System", INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Vincent Abramowski
- Laboratory "Genome Dynamics in the Immune System", INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Julie Chaumeil
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris-Descartes, Paris, France
| | - Jean-Pierre de Villartay
- Laboratory "Genome Dynamics in the Immune System", INSERM UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| |
Collapse
|
127
|
Tugues S, Ducimetiere L, Friebel E, Becher B. Innate lymphoid cells as regulators of the tumor microenvironment. Semin Immunol 2019; 41:101270. [PMID: 30871769 DOI: 10.1016/j.smim.2019.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/19/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
As crucial players in innate immunity, Innate Lymphoid Cells (ILCs) have been distinctly associated with either tumor-promoting or tumor-inhibiting activities. This dichotomy arises from the high degree of heterogeneity and plasticity between the ILC family subsets. Also, the tissue microenvironment is crucial for the function of ILCs. Especially within the tumor niche, each of the ILC subsets participates in a complex network of interactions with other cells and molecules. Although extensive research has unraveled several aspects of the crosstalk ILCs establish with the tumor microenvironment (TME), numerous questions remain to be answered. Here, we will discuss a role for the different ILC subsets that goes beyond their direct effects on the tumor cells. Instead, we will highlight the ability of ILCs to communicate with the surrounding milieu and the impact this has on tumor progression.
Collapse
Affiliation(s)
- Sonia Tugues
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | - Laura Ducimetiere
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ekaterina Friebel
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
128
|
Imura C, Ueyama A, Sasaki Y, Shimizu M, Furue Y, Tai N, Tsujii K, Katayama K, Okuno T, Shichijo M, Yasui K, Yamamoto M. A novel RORγt inhibitor is a potential therapeutic agent for the topical treatment of psoriasis with low risk of thymic aberrations. J Dermatol Sci 2019; 93:176-185. [PMID: 30905492 DOI: 10.1016/j.jdermsci.2019.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Retinoic acid receptor-related orphan receptor gamma t (RORγt) has critical roles in the development, maintenance and function of interleukin (IL)-17-producing cells and is a highly attractive target for the treatment of IL-17-mediated autoimmune disease, particularly psoriasis. On the other hand, RORγt is also critical for controlling apoptosis during thymopoiesis, and genetic RORγt ablation or systematic RORγt inhibition cause progressive thymic aberrations leading to T cell lymphomas. OBJECTIVE We investigated whether topical administration of our novel RORγt inhibitor, S18-000003 has therapeutic potential for psoriasis with low risk of thymic aberrations. METHODS We evaluated the effect of topical S18-000003 on psoriasis-like skin inflammation and influence on the thymus in a 12-O-tetradecanoylphorbol-13-acetate-induced K14.Stat3C mouse psoriasis model. RESULTS S18-000003 markedly inhibited the development of psoriatic skin inflammation via suppression of the IL-17 pathway. In the skin, S18-000003 suppressed all subsets of IL-17-producing cells that we previously identified in this psoriasis model: Th17 cells, Tc17 cells, dermal γδ T cells, TCR- cells that probably included innate lymphoid cells, and CD4-CD8- double-negative αβ T cells. Notably, neither reduction of CD4+CD8+ double-positive thymocytes nor dysregulation of cell cycling was observed in S18-000003-treated mice, even at a high dose. CONCLUSION Our topically administered RORγt inhibitor is a potential therapeutic agent for psoriasis with low risk of thymic lymphoma.
Collapse
Affiliation(s)
- Chihiro Imura
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Azumi Ueyama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan.
| | - Yoshikazu Sasaki
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Masaya Shimizu
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Yoko Furue
- Shionogi Techno Advance Research Co., Ltd., Toyonaka, Japan
| | - Nobuyuki Tai
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Kenichiro Tsujii
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Kazufumi Katayama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Takayuki Okuno
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | | | - Kiyoshi Yasui
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Mina Yamamoto
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Toyonaka, Japan
| |
Collapse
|
129
|
Cao D, Qi Z, Pang Y, Li H, Xie H, Wu J, Huang Y, Zhu Y, Shen Y, Zhu Y, Dai B, Hu X, Ye D, Wang Z. Retinoic Acid-Related Orphan Receptor C Regulates Proliferation, Glycolysis, and Chemoresistance via the PD-L1/ITGB6/STAT3 Signaling Axis in Bladder Cancer. Cancer Res 2019; 79:2604-2618. [PMID: 30808674 DOI: 10.1158/0008-5472.can-18-3842] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/01/2019] [Accepted: 02/21/2019] [Indexed: 12/29/2022]
Abstract
Retinoic acid-related orphan receptor C (RORC) is a member of the nuclear orphan receptor family and performs critical regulatory functions in cell proliferation, metastasis, and chemoresistance in various types of malignant tumors. Here we showed that expression of RORC is lost in tumor tissues of bladder cancer patients. Enhanced expression of RORC suppressed cell proliferation and glucose metabolism and increased cisplatin-induced apoptosis in vitro and in vivo. RORC bound the promoter region of programmed death ligand-1 (PD-L1) and negatively regulated PD-L1 expression. PD-L1 directly interacted with integrin β6 (ITGB6) and activated the ITGB6/FAK signaling pathway. RORC prevented the nuclear translocation of STAT3 via suppression of the PD-L1/ITGB6 signaling pathway, which further inhibited bladder cell proliferation and glucose metabolism and increased cisplatin-induced apoptosis. These findings reveal that RORC regulates bladder cancer cell proliferation, glucose metabolism, and chemoresistance by participating in the PD-L1/ITGB6/STAT3 signaling axis. Moreover, this new understanding of PD-L1 signaling may guide the selection of therapeutic targets to prevent tumor recurrence. SIGNIFICANCE: These findings suggest that RORC-mediated regulation of a PD-L1/ITGB6/FAK/STAT3 signaling axis in bladder cancer provides several potential therapeutic targets to prevent tumor progression.
Collapse
Affiliation(s)
- Dalong Cao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zihao Qi
- Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yangyang Pang
- Department of Urology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Haoran Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Huyang Xie
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongqiang Huang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yijun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Hu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ziliang Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
130
|
Martínez-López M, Iborra S, Conde-Garrosa R, Mastrangelo A, Danne C, Mann ER, Reid DM, Gaboriau-Routhiau V, Chaparro M, Lorenzo MP, Minnerup L, Saz-Leal P, Slack E, Kemp B, Gisbert JP, Dzionek A, Robinson MJ, Rupérez FJ, Cerf-Bensussan N, Brown GD, Bernardo D, LeibundGut-Landmann S, Sancho D. Microbiota Sensing by Mincle-Syk Axis in Dendritic Cells Regulates Interleukin-17 and -22 Production and Promotes Intestinal Barrier Integrity. Immunity 2019; 50:446-461.e9. [PMID: 30709742 PMCID: PMC6382412 DOI: 10.1016/j.immuni.2018.12.020] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 07/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Production of interleukin-17 (IL-17) and IL-22 by T helper 17 (Th17) cells and group 3 innate lymphoid cells (ILC3s) in response to the gut microbiota ensures maintenance of intestinal barrier function. Here, we examined the mechanisms whereby the immune system detects microbiota in the steady state. A Syk-kinase-coupled signaling pathway in dendritic cells (DCs) was critical for commensal-dependent production of IL-17 and IL-22 by CD4+ T cells. The Syk-coupled C-type lectin receptor Mincle detected mucosal-resident commensals in the Peyer's patches (PPs), triggered IL-6 and IL-23p19 expression, and thereby regulated function of intestinal Th17- and IL-17-secreting ILCs. Mice deficient in Mincle or with selective depletion of Syk in CD11c+ cells had impaired production of intestinal RegIIIγ and IgA and increased systemic translocation of gut microbiota. Consequently, Mincle deficiency led to liver inflammation and deregulated lipid metabolism. Thus, sensing of commensals by Mincle and Syk signaling in CD11c+ cells reinforces intestinal immune barrier and promotes host-microbiota mutualism, preventing systemic inflammation.
Collapse
Affiliation(s)
- María Martínez-López
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain; Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| | - Ruth Conde-Garrosa
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Annalaura Mastrangelo
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Camille Danne
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Delyth M Reid
- Medical Research Council Centre for Medical Mycology at the University of Aberdeen, Aberdeen Fungal Group, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Valérie Gaboriau-Routhiau
- INRA Micalis Institut, UMR1319, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France; INSERM UMR1163, Institut Imagine, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75006 Paris, France
| | - Maria Chaparro
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | - María P Lorenzo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, Urbanización Montepríncipe, km 0, M501, Alcorcón 28925, Spain
| | | | - Paula Saz-Leal
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Emma Slack
- Institute of Food, Nutrition, and Health, ETH Zurich, Vladimir-Prelog-Weg 4, Zürich 8093, Switzerland
| | | | - Javier P Gisbert
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | | | | | - Francisco J Rupérez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, Urbanización Montepríncipe, km 0, M501, Alcorcón 28925, Spain
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Institut Imagine, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75006 Paris, France
| | - Gordon D Brown
- Medical Research Council Centre for Medical Mycology at the University of Aberdeen, Aberdeen Fungal Group, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - David Bernardo
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a Zurich 8057, Switzerland
| | - David Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain.
| |
Collapse
|
131
|
Philips RL, McCue SA, Rajcula MJ, Shapiro VS. Cutting Edge: HDAC3 Protects Double-Positive Thymocytes from P2X7 Receptor-Induced Cell Death. THE JOURNAL OF IMMUNOLOGY 2019; 202:1033-1038. [PMID: 30626694 DOI: 10.4049/jimmunol.1801438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/10/2018] [Indexed: 11/19/2022]
Abstract
Intricate life-versus-death decisions are programmed during T cell development, and the regulatory mechanisms that coordinate their activation and repression are still under investigation. In this study, HDAC3-deficient double-positive (DP) thymocytes exhibit a severe decrease in numbers. The thymic cortex is rich in ATP, which is released by macrophages that clear apoptotic DP thymocytes that fail to undergo positive selection. We demonstrate that HDAC3 is required to repress expression of the purinergic receptor P2X7 to prevent DP cell death. HDAC3-deficient DP thymocytes upregulate the P2X7 receptor, increasing sensitivity to ATP-induced cell death. P2rx7/HDAC3-double knockout mice show a partial rescue in DP cell number. HDAC3 directly binds to the P2rx7 enhancer, which is hyperacetylated in the absence of HDAC3. In addition, RORγt binds to the P2rx7 enhancer and promotes P2X7 receptor expression in the absence of HDAC3. Therefore, HDAC3 is a critical regulator of DP thymocyte survival and is required to suppress P2X7 receptor expression.
Collapse
|
132
|
Philips RL, Lee JH, Gaonkar K, Chanana P, Chung JY, Romero Arocha SR, Schwab A, Ordog T, Shapiro VS. HDAC3 restrains CD8-lineage genes to maintain a bi-potential state in CD4 +CD8 + thymocytes for CD4-lineage commitment. eLife 2019; 8:43821. [PMID: 30657451 PMCID: PMC6338460 DOI: 10.7554/elife.43821] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/30/2018] [Indexed: 12/20/2022] Open
Abstract
CD4 and CD8 T cells are vital components of the immune system. We found that histone deacetylase 3 (HDAC3) is critical for the development of CD4 T cells, as HDAC3-deficient DP thymocytes generate only CD8SP thymocytes in mice. In the absence of HDAC3, MHC Class II-restricted OT-II thymocytes are redirected to the CD8 cytotoxic lineage, which occurs with accelerated kinetics. Analysis of histone acetylation and RNA-seq reveals that HDAC3-deficient DP thymocytes are biased towards the CD8 lineage prior to positive selection. Commitment to the CD4 or CD8 lineage is determined by whether persistent TCR signaling or cytokine signaling predominates, respectively. Despite elevated IL-21R/γc/STAT5 signaling in HDAC3-deficient DP thymocytes, blocking IL-21R does not restore CD4 lineage commitment. Instead, HDAC3 binds directly to CD8-lineage promoting genes. Thus, HDAC3 is required to restrain CD8-lineage genes in DP thymocytes for the generation of CD4 T cells.
Collapse
Affiliation(s)
| | - Jeong-Heon Lee
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, United States
| | - Krutika Gaonkar
- Department of Health Science Research, Division of Biostatistics and Informatics, Mayo Clinic, Rochester, United States
| | - Pritha Chanana
- Department of Health Science Research, Division of Biostatistics and Informatics, Mayo Clinic, Rochester, United States
| | - Ji Young Chung
- Department of Immunology, Mayo Clinic, Rochester, United States
| | | | - Aaron Schwab
- Department of Immunology, Mayo Clinic, Rochester, United States
| | - Tamas Ordog
- Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, United States
| | | |
Collapse
|
133
|
He Z, Zhang J, Du Q, Xu J, Gwack Y, Sun Z. SRC3 Is a Cofactor for RORγt in Th17 Differentiation but Not Thymocyte Development. THE JOURNAL OF IMMUNOLOGY 2018; 202:760-769. [PMID: 30567733 DOI: 10.4049/jimmunol.1801187] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/19/2018] [Indexed: 01/13/2023]
Abstract
SRC3, a highly conserved member of the steroid receptor coactivator (SRC) family, is recruited by transcription factors to regulate cellular function. Previously, we demonstrated that SRC1, another highly conserved member of the SRC family, interacts with RORγt to regulate Th17 differentiation. However, the relationship between SRC1 and SRC3 in the regulation of Th17 cell function remains unknown. In this study, we demonstrate that mouse SRC3 interacts with RORγt in Th17 cells but not in thymocytes. In addition, Src3-/- mice exhibited defective Th17 differentiation and induction of experimental autoimmune encephalomyelitis but normal thymocyte development. Furthermore, a K313 to arginine mutation of RORγt (RORγt-K313R), which disrupts the interaction of RORγt with SRC3 but not with SRC1, impairs Th17 differentiation but not thymocyte development. These data suggest that SRC3 works with SRC1 to regulate RORγt-dependent Th17 differentiation but is not essential for RORγt-dependent thymocyte development.
Collapse
Affiliation(s)
- Zhiheng He
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Jing Zhang
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, CA 91010.,Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010
| | - Qian Du
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, CA 91010
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030; and
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095
| | - Zuoming Sun
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, CA 91010;
| |
Collapse
|
134
|
Li S, Bostick JW, Ye J, Qiu J, Zhang B, Urban JF, Avram D, Zhou L. Aryl Hydrocarbon Receptor Signaling Cell Intrinsically Inhibits Intestinal Group 2 Innate Lymphoid Cell Function. Immunity 2018; 49:915-928.e5. [PMID: 30446384 PMCID: PMC6249058 DOI: 10.1016/j.immuni.2018.09.015] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/03/2018] [Accepted: 09/20/2018] [Indexed: 12/17/2022]
Abstract
Innate lymphoid cells (ILCs) are important for mucosal immunity. The intestine harbors all ILC subsets, but how these cells are balanced to achieve immune homeostasis and mount appropriate responses during infection remains elusive. Here, we show that aryl hydrocarbon receptor (Ahr) expression in the gut regulates ILC balance. Among ILCs, Ahr is most highly expressed by gut ILC2s and controls chromatin accessibility at the Ahr locus via positive feedback. Ahr signaling suppresses Gfi1 transcription-factor-mediated expression of the interleukin-33 (IL-33) receptor ST2 in ILC2s and expression of ILC2 effector molecules IL-5, IL-13, and amphiregulin in a cell-intrinsic manner. Ablation of Ahr enhances anti-helminth immunity in the gut, whereas genetic or pharmacological activation of Ahr suppresses ILC2 function but enhances ILC3 maintenance to protect the host from Citrobacter rodentium infection. Thus, the host regulates the gut ILC2-ILC3 balance by engaging the Ahr pathway to mount appropriate immunity against various pathogens.
Collapse
Affiliation(s)
- Shiyang Li
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - John W Bostick
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Jian Ye
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhang
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph F Urban
- USDA/ARS, Beltsville Human Nutrition Research Center, Diet, Genomics, & Immunology Lab, Beltsville, MD 20705, USA
| | - Dorina Avram
- Department of Medicine, Division of Pulmonary Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA.
| |
Collapse
|
135
|
He Z, Zhang J, Huang Z, Du Q, Li N, Zhang Q, Chen Y, Sun Z. Sumoylation of RORγt regulates T H17 differentiation and thymocyte development. Nat Commun 2018; 9:4870. [PMID: 30451821 PMCID: PMC6242824 DOI: 10.1038/s41467-018-07203-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer’s patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity. The transcription factor RORγt is essential for the differentiation of TH17 cells, thymocyte development and lymphoid organogenesis. Here the authors show that the function of RORγt is regulated by PIAS4-mediated sumoylation that stabilize the interaction with SRC1 and KAT2A, to enhance the transcriptional activity of RORγt.
Collapse
Affiliation(s)
- Zhiheng He
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Jing Zhang
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA.,Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, 91010, CA, USA
| | - Zhaofeng Huang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Qian Du
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan Univerity, Shanghai, 200040, China
| | - Qiang Zhang
- Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| | - Yuan Chen
- Division of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Zuoming Sun
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA.
| |
Collapse
|
136
|
Billon C, Murray MH, Avdagic A, Burris TP. RORγ regulates the NLRP3 inflammasome. J Biol Chem 2018; 294:10-19. [PMID: 30455347 DOI: 10.1074/jbc.ac118.002127] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 11/05/2018] [Indexed: 12/21/2022] Open
Abstract
RAR-related orphan receptor γ (RORγ) is a nuclear receptor that plays an essential role in the development of T helper 17 (Th17) cells of the adaptive immune system. The NLRP3 inflammasome is a component of the innate immune system that processes interleukin (IL)-1β into a mature cytokine. Elevated activity of the NLRP3 inflammasome contributes to the progression of an array of inflammatory diseases. Bone marrow-derived macrophages (BMDMs) isolated from RORγ-null mice displayed reduced capacity to secrete IL-1β, and they also displayed a reduction in Nlrp3 and Il1b gene expression. Examination of the promoters of the Il1b and Nlrp3 genes revealed multiple putative ROR response elements (ROREs) that were occupied by RORγ. RORγ inverse agonists were effective inhibitors of the inflammasome. RORγ inverse agonists suppressed lipopolysaccharide (LPS)/ATP-stimulated IL-1β secretion and expression of Il1b and Nlrp3 in BMDMs. Additionally, the ability of the RORγ inverse agonists to suppress IL-1β secretion was lost in Nlrp3-null macrophages. The potential for targeting the NLRP3 inflammasome in vivo using RORγ inverse agonists was examined in two models: LPS-induced sepsis and fulminant hepatitis. Pharmacological inhibition of RORγ activity reduced plasma IL-1β as well as IL-1β production by peritoneal macrophages in a model of LPS-induced sepsis. Additionally, RORγ inverse agonists reduced mortality in an LPS/d-galactosamine-induced fulminant hepatitis mouse model. These results illustrate a major role for RORγ in regulation of innate immunity via modulation of NLRP3 inflammasome activity. Furthermore, these data suggest that inhibiting the NLRP3 inflammasome with RORγ inverse agonists may be an effective method to treat NLRP3-associated diseases.
Collapse
Affiliation(s)
- Cyrielle Billon
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110
| | - Meghan H Murray
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104
| | - Amer Avdagic
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110
| | - Thomas P Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110.
| |
Collapse
|
137
|
Suo S, Zhu Q, Saadatpour A, Fei L, Guo G, Yuan GC. Revealing the Critical Regulators of Cell Identity in the Mouse Cell Atlas. Cell Rep 2018; 25:1436-1445.e3. [PMID: 30404000 PMCID: PMC6281296 DOI: 10.1016/j.celrep.2018.10.045] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/06/2018] [Accepted: 10/11/2018] [Indexed: 12/19/2022] Open
Abstract
Recent progress in single-cell technologies has enabled the identification of all major cell types in mouse. However, for most cell types, the regulatory mechanism underlying their identity remains poorly understood. By computational analysis of the recently published mouse cell atlas data, we have identified 202 regulons whose activities are highly variable across different cell types, and more importantly, predicted a small set of essential regulators for each major cell type in mouse. Systematic validation by automated literature and data mining provides strong additional support for our predictions. Thus, these predictions serve as a valuable resource that would be useful for the broad biological community. Finally, we have built a user-friendly, interactive web portal to enable users to navigate this mouse cell network atlas.
Collapse
Affiliation(s)
- Shengbao Suo
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Qian Zhu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Assieh Saadatpour
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA.
| |
Collapse
|
138
|
Onder L, Ludewig B. A Fresh View on Lymph Node Organogenesis. Trends Immunol 2018; 39:775-787. [DOI: 10.1016/j.it.2018.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 01/18/2023]
|
139
|
Ligons DL, Hwang S, Waickman AT, Park JY, Luckey MA, Park JH. RORγt limits the amount of the cytokine receptor γc through the prosurvival factor Bcl-x L in developing thymocytes. Sci Signal 2018; 11:11/545/eaam8939. [PMID: 30154103 DOI: 10.1126/scisignal.aam8939] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The cytokine receptor subunit γc provides critical signals for T cell survival and differentiation. We investigated the molecular mechanism that controls the cell surface abundance of γc during T cell development in the thymus. We found that the amount of γc was low on CD4+CD8+ double-positive (DP) thymocytes before their positive selection to become mature T cells. The transcription factor RORγt was abundant in immature DP thymocytes, and its loss resulted in an increase in the abundance of surface γc, specifically on preselection DP cells. Rather than directly repressing expression of the gene encoding γc, RORγt acted through the antiapoptotic protein Bcl-xL to reduce the abundance of surface γc, which resulted in decreased cytokine signaling and was associated with inhibition of cell metabolism and mitochondrial biogenesis. Accordingly, overexpression of Bcl-xL in RORγt-deficient thymocytes restored the amount of surface γc to that present on normal preselection DP cells. Together, these data highlight a previously unappreciated role for RORγt and Bcl-xL in limiting γc abundance at the cell surface and reveal a signaling circuit in which survival factors control cytokine signaling by limiting the abundance and surface distribution of a receptor subunit shared by several cytokines.
Collapse
Affiliation(s)
- Davinna L Ligons
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - SuJin Hwang
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam T Waickman
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joo-Young Park
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Megan A Luckey
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
140
|
Link VM, Romanoski CE, Metzler D, Glass CK. MMARGE: Motif Mutation Analysis for Regulatory Genomic Elements. Nucleic Acids Res 2018; 46:7006-7021. [PMID: 29893919 PMCID: PMC6101580 DOI: 10.1093/nar/gky491] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022] Open
Abstract
Cell-specific patterns of gene expression are determined by combinatorial actions of sequence-specific transcription factors at cis-regulatory elements. Studies indicate that relatively simple combinations of lineage-determining transcription factors (LDTFs) play dominant roles in the selection of enhancers that establish cell identities and functions. LDTFs require collaborative interactions with additional transcription factors to mediate enhancer function, but the identities of these factors are often unknown. We have shown that natural genetic variation between individuals has great utility for discovering collaborative transcription factors. Here, we introduce MMARGE (Motif Mutation Analysis of Regulatory Genomic Elements), the first publicly available suite of software tools that integrates genome-wide genetic variation with epigenetic data to identify collaborative transcription factor pairs. MMARGE is optimized to work with chromatin accessibility assays (such as ATAC-seq or DNase I hypersensitivity), as well as transcription factor binding data collected by ChIP-seq. Herein, we provide investigators with rationale for each step in the MMARGE pipeline and key differences for analysis of datasets with different experimental designs. We demonstrate the utility of MMARGE using mouse peritoneal macrophages, liver cells, and human lymphoblastoid cells. MMARGE provides a powerful tool to identify combinations of cell type-specific transcription factors while simultaneously interpreting functional effects of non-coding genetic variation.
Collapse
Affiliation(s)
- Verena M Link
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, USA
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilian Universität München, Planegg-Martinsried, Germany
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | - Dirk Metzler
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilian Universität München, Planegg-Martinsried, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, USA
- Department of Medicine, University of California, San Diego, San Diego, USA
| |
Collapse
|
141
|
Chalmin F, Humblin E, Ghiringhelli F, Végran F. Transcriptional Programs Underlying Cd4 T Cell Differentiation and Functions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:1-61. [PMID: 30262030 DOI: 10.1016/bs.ircmb.2018.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the basis of cellular differentiation is a fundamental issue in developmental biology but also for the comprehension of pathological processes. In fact, the palette of developmental decisions for naive CD4 T cells is a critical aspect of the development of appropriate immune responses which could control infectious processes or cancer growth. However, the current accumulation of data on CD4 T cells biology reveals a complex world with different helper populations. Naive CD4 T cells can differentiate into different subtypes in response to cytokine stimulation. This stimulation involves a complex transcriptional network implicating the activation of Signal Transducer and Activator of Transcription but also master regulator transcription factors allowing the functions of each helper T lymphocyte subtype. In this review, we will present an overview of the transcriptional regulation which controls process of helper T cells differentiation. We will focus on the role of initiator transcriptional factors and on master regulators but also on other nonspecific transcriptional factors which refine the T helper polarization to stabilize or modulate the differentiation program.
Collapse
Affiliation(s)
- Fanny Chalmin
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France; Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Univ. Bourgogne Franche-Comté, Dijon, France
| | - Etienne Humblin
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France; Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Univ. Bourgogne Franche-Comté, Dijon, France
| | - François Ghiringhelli
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France; Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Univ. Bourgogne Franche-Comté, Dijon, France; Platform of Transfer in Cancer Biology, Centre Georges-François Leclerc, Dijon, France
| | - Frédérique Végran
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Univ. Bourgogne Franche-Comté, Dijon, France; Platform of Transfer in Cancer Biology, Centre Georges-François Leclerc, Dijon, France
| |
Collapse
|
142
|
Cording S, Medvedovic J, Lécuyer E, Aychek T, Déjardin F, Eberl G. Mouse models for the study of fate and function of innate lymphoid cells. Eur J Immunol 2018; 48:1271-1280. [PMID: 29974461 DOI: 10.1002/eji.201747388] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/05/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022]
Abstract
Natural killer (NK) cells and lymphoid tissue inducer (LTi) cells were discovered more than 40 and 20 years ago, respectively. These two cell types were initially studied for their unique functions in the elimination of infected or transformed cells, and in the development of lymphoid tissues. It took an additional 10 years to realize that NK cells and LTi cells were members of a larger family of innate lymphoid cells (ILCs), whose phenotypes and functions mirror those of T cells. Many mouse models have since been developed to identify and isolate ILCs, map their developmental pathways and characterize their functions. Because of the similarity between ILCs and T cells, this exploration remains a challenge. In spite of this, a broad range of mouse models available to researchers has lead to significant progress in untangling the unique roles of ILCs early in defense, regulation of adaptive immunity and homeostasis. Here, we review these mouse models, and discuss their strengths and limitations.
Collapse
Affiliation(s)
- Sascha Cording
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Laboratory of Intestinal Immunity, Institute IMAGINE, INSERM 1163, Paris, France
| | - Jasna Medvedovic
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France
| | - Emelyne Lécuyer
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France
| | - Tegest Aychek
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France
| | - François Déjardin
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France
| | - Gérard Eberl
- Institut Pasteur, Microenvironment & Immunity Unit, Paris, France.,INSERM U1224, Paris, France
| |
Collapse
|
143
|
Pandya VB, Kumar S, Sachchidanand, Sharma R, Desai RC. Combating Autoimmune Diseases With Retinoic Acid Receptor-Related Orphan Receptor-γ (RORγ or RORc) Inhibitors: Hits and Misses. J Med Chem 2018; 61:10976-10995. [DOI: 10.1021/acs.jmedchem.8b00588] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Vrajesh B. Pandya
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH8A, Moraiya, Ahmedabad 382210, India
| | - Sanjay Kumar
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH8A, Moraiya, Ahmedabad 382210, India
| | - Sachchidanand
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH8A, Moraiya, Ahmedabad 382210, India
| | - Rajiv Sharma
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH8A, Moraiya, Ahmedabad 382210, India
| | - Ranjit C. Desai
- Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH8A, Moraiya, Ahmedabad 382210, India
| |
Collapse
|
144
|
Schaeuble K, Britschgi MR, Scarpellino L, Favre S, Xu Y, Koroleva E, Lissandrin TKA, Link A, Matloubian M, Ware CF, Nedospasov SA, Tumanov AV, Cyster JG, Luther SA. Perivascular Fibroblasts of the Developing Spleen Act as LTα1β2-Dependent Precursors of Both T and B Zone Organizer Cells. Cell Rep 2018; 21:2500-2514. [PMID: 29186687 DOI: 10.1016/j.celrep.2017.10.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/08/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022] Open
Abstract
T and B cell compartmentalization is a hallmark of secondary lymphoid organs and is maintained by chemokine-expressing stromal cells. How this stromal cell network initially develops and differentiates into two distinct subsets is poorly known, especially for the splenic white pulp (WP). Here, we show that perivascular fibroblast precursors are triggered by LTα1β2 signals to expand, express CCL19/21, and then differentiate into two functionally distinct fibroblast subsets responsible for B and T cell clustering and WP compartmentalization. Failure to express or sense CCL19 leads to impaired T zone development, while lack of B cells or LTα1β2 leads to an earlier and stronger impairment in WP development. We therefore propose that WP development proceeds in multiple steps, with LTα1β2+ B cells acting as major inducer cells driving the expansion and gradual differentiation of perivascular fibroblasts into T and B zone organizer cells.
Collapse
Affiliation(s)
- Karin Schaeuble
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mirjam R Britschgi
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Leo Scarpellino
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stéphanie Favre
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ekaterina Koroleva
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | - Alexander Link
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mehrdad Matloubian
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Carl F Ware
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, Moscow 119991, Russia
| | - Alexei V Tumanov
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
145
|
Jones R, Cosway EJ, Willis C, White AJ, Jenkinson WE, Fehling HJ, Anderson G, Withers DR. Dynamic changes in intrathymic ILC populations during murine neonatal development. Eur J Immunol 2018; 48:1481-1491. [PMID: 29851080 PMCID: PMC6174991 DOI: 10.1002/eji.201847511] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/19/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
Members of the innate lymphoid cell (ILC) family have been implicated in the development of thymic microenvironments and the recovery of this architecture after damage. However, a detailed characterization of this family in the thymus is lacking. To better understand the thymic ILC compartment, we have utilized multiple in vivo models including the fate mapping of inhibitor of DNA binding‐2 (Id2) expression and the use of Id2 reporter mice. Our data demonstrate that ILCs are more prominent immediately after birth, but were rapidly diluted as the T‐cell development program increased. As observed in the embryonic thymus, CCR6+NKp46− lymphoid tissue inducer (LTi) cells were the main ILC3 population present, but numbers of these cells swiftly declined in the neonate and ILC3 were barely detectable in adult thymus. This loss of ILC3 means ILC2 are the dominant ILC population in the thymus. Thymic ILC2 were able to produce IL‐5 and IL‐13, were located within the medulla, and did not result from ILC3 plasticity. Furthermore, in WT mice, thymic ILC2 express little RANKL (receptor activator of nuclear factor kappa‐B ligand) arguing that functionally, these cells provide different signals to LTi cells in the thymus. Collectively, these data reveal a dynamic switch in the ILC populations of the thymus during neonatal development.
Collapse
Affiliation(s)
- Rhys Jones
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emilie J Cosway
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Claire Willis
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrea J White
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - William E Jenkinson
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Hans J Fehling
- Institute of Immunology, University of Ulm, Ulm, Germany
| | - Graham Anderson
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David R Withers
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
146
|
Xie Z, Li Y, Long L, Liang H, Cai W, Shen T. Polymorphisms in the Th17 cell-related RORC gene are associated with spontaneous clearance of HCV in Chinese women. BMC Infect Dis 2018; 18:254. [PMID: 29866105 PMCID: PMC5987631 DOI: 10.1186/s12879-018-3153-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 05/18/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Female gender and favorable IFNL3 genotypes are the primary independent predictors of spontaneous clearance of HCV infection. However, chronic hepatitis C infection occurs in numerous women carrying favorable IFNL3 genotypes, indicating that other host and/or virological factors contribute to the prognosis of infection. METHODS A cohort of 137 anti-HCV-positive female Han Chinese cases, including 64 chronic HCV carriers and 73 HCV spontaneous resolvers, was recruited in the study. 111 SNPs in 23 genes encoding HCV co-receptors, transcription factors, Toll-like receptors, co-stimulating molecules, and cytokines were selected for SNP analysis. RESULTS After comparison of genotypes and allelotype frequencies of 111 SNPs in 23 genes in the primary cohort, the SNPs rs9826 (P = 0.024 for CC/TT/CT; P = 0.015 for C allele/T allele) and rs1521177 (P = 0.017 for GG/TT/GT; P = 0.006 for G allele/T allele) in the RORC gene were significantly associated with spontaneous HCV clearance. In the sub-cohort carrying favorable IFNL3 genotypes (rs12979860CC, rs8099917 TT, rs12980275 AA), rs1521177 (genotype: P = 0.040; allelotype: P = 0.021) remained significantly associated with spontaneous HCV clearance. Importantly, the most common RORC haplotype rs9826-T/rs1521177-T was presented at significantly different frequencies in resolvers and carriers in both the primary cohort (P = 0.0027) and the IFNL3 favorable sub-cohort (P = 0.0117). CONCLUSIONS This study indicates that genetic polymorphisms in human Th17-related RORC gene are associated with different natural prognosis of HCV infection. The RORC haplotype, rs9826-T/rs1521177-T, was favorable for spontaneous clearance of HCV infection.
Collapse
Affiliation(s)
- Zhe Xie
- Department of Microbiology and Center of Infectious Diseases, School of Basic Medical Sciences, Peking University, Beijing, 100191 China
| | - Yuantao Li
- Department of Microbiology and Center of Infectious Diseases, School of Basic Medical Sciences, Peking University, Beijing, 100191 China
| | - Lu Long
- Department of Microbiology and Center of Infectious Diseases, School of Basic Medical Sciences, Peking University, Beijing, 100191 China
| | - Hua Liang
- State Key laboratory of Infectious Disease Prevention and Control, National Center for AIDS/STD Control and Prevention, China CDC, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing, China
| | - Weiping Cai
- Department of Infectious Diseases, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, 510060 Guangdong China
| | - Tao Shen
- Department of Microbiology and Center of Infectious Diseases, School of Basic Medical Sciences, Peking University, Beijing, 100191 China
| |
Collapse
|
147
|
Paul R, Jaiswal S, Mahalakshmi N, Kaliraj P. Elucidation of immunological response and its regulatory network by P-TUFT-ALT-2: a promising fusion protein vaccine for human lymphatic filariasis. ROYAL SOCIETY OPEN SCIENCE 2018; 5:172039. [PMID: 29892388 PMCID: PMC5990782 DOI: 10.1098/rsos.172039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Human lymphatic filariasis, a mosquito-borne neglected tropical parasitic disease, needs an early development of prophylactic agents such as a vaccine for its successful elimination. Our earlier study suggested the enhanced immunological response by fusion protein (P-TUFT-ALT-2) of Tuftsin and ALT-2 in a mice model. We cultured human peripheral blood mononuclear cells (PBMCs) and treated cells with Escherichia coli-expressed ALT-2 (E-ALT-2) and P-TUFT-ALT-2. Real-time polymerase chain reaction was performed to identify the mRNA copy number of various cytokine and transcription factor genes. The recombinant vaccine candidate was also validated for humans by immunoreactivity with human sera samples of natural infection. In this study, P-TUFT-ALT-2 stimulated 12% higher PBMC proliferation in endemic normal (EN) individuals than E-ALT-2 alone. There was enhanced production of IFN γ, IL-2, IL-5 and IL-12, indicating a balanced Th1/Th2 response. However, higher expression of IL-5 and lower IL-4 validate the humoral response through an IL-5-dependent manner. Also, high level of IL-17 indicates a strong Th/Treg regulation over T-cell activation. The upregulated T-bet might have enhanced IFN-γ production, whereas GATA-3 was supposed to enhance IL-5 expression. The fusion protein also exhibited 15-16% higher reactivity with EN clinical sera, exposing the upregulation of IgG1 and IgM in natural infection. The higher reactivity of P-TUFT-ALT-2 with sera of natural infection (EN) was validated indirectly by B-cell activation through various cytokines and regulatory genes produced from different T cells. Thus, these findings endorse P-TUFT-ALT-2 as a potential vaccine candidate for human lymphatic filariasis.
Collapse
|
148
|
Hosokawa H, Rothenberg EV. Cytokines, Transcription Factors, and the Initiation of T-Cell Development. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028621. [PMID: 28716889 DOI: 10.1101/cshperspect.a028621] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multipotent blood progenitor cells migrate into the thymus and initiate the T-cell differentiation program. T-cell progenitor cells gradually acquire T-cell characteristics while shedding their multipotentiality for alternative fates. This process is supported by extracellular signaling molecules, including Notch ligands and cytokines, provided by the thymic microenvironment. T-cell development is associated with dynamic change of gene regulatory networks of transcription factors, which interact with these environmental signals. Together with Notch or pre-T-cell-receptor (TCR) signaling, cytokines always control proliferation, survival, and differentiation of early T cells, but little is known regarding their cross talk with transcription factors. However, recent results suggest ways that cytokines expressed in distinct intrathymic niches can specifically modulate key transcription factors. This review discusses how stage-specific roles of cytokines and transcription factors can jointly guide development of early T cells.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
149
|
Zhang J, He Z, Sen S, Wang F, Zhang Q, Sun Z. TCF-1 Inhibits IL-17 Gene Expression To Restrain Th17 Immunity in a Stage-Specific Manner. THE JOURNAL OF IMMUNOLOGY 2018; 200:3397-3406. [PMID: 29632143 DOI: 10.4049/jimmunol.1800193] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/14/2018] [Indexed: 12/22/2022]
Abstract
T cell factor 1 (TCF-1) is expressed in both developing and mature T cells and has been shown to restrain mature T cell-mediated Th17 responses by inhibiting IL-17 expression. However, it is not clear when TCF-1 is required in vivo to restrain the magnitude of peripheral Th17 responses and what the molecular mechanisms responsible for TCF-1-regulated IL-17 gene expression are. In this study, we showed that conditional deletion of TCF-1 at the early but not later CD4+CD8+ double-positive stage in mice enhanced Th17 differentiation and aggravated experimental autoimmune encephalomyelitis, which correlates with abnormally high IL-17 expression. Expression of TCF-1 in TCF-1-deficient thymocytes but not TCF-1-deficient Th17 cells inhibited IL-17 expression. TCF-1 binds to IL-17 promoter regions, and deletion of two TCF-1 binding sites relieves TCF-1-mediated inhibition of IL-17 promoter activity. Lastly, wild-type TCF-1, but not a TCF-1 mutant that has no intrinsic histone deacetylase activity, was able to inhibit IL-17 expression in TCF-1 deficient mouse thymocytes. Thus, our study demonstrates the requirement of TCF-1 in vivo at stages earlier than double-positive cells to restrain peripheral Th17 immunity by directly binding and inhibiting IL-17 promoter in its intrinsic histone deacetylase-dependent manner.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010.,Irell and Manella Graduate School of Biological Sciences at City of Hope, Duarte, CA 91010; and
| | - Zhiheng He
- Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Subha Sen
- Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Fei Wang
- Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin 300052, China
| | - Zuoming Sun
- Department of Molecular Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010;
| |
Collapse
|
150
|
Identification of novel lncRNAs regulated by the TAL1 complex in T-cell acute lymphoblastic leukemia. Leukemia 2018; 32:2138-2151. [PMID: 29654272 DOI: 10.1038/s41375-018-0110-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 02/05/2018] [Accepted: 03/09/2018] [Indexed: 12/13/2022]
Abstract
TAL1/SCL is one of the most prevalent oncogenes in T-cell acute lymphoblastic leukemia (T-ALL). TAL1 and its regulatory partners (GATA3, RUNX1, and MYB) positively regulate each other and coordinately regulate the expression of their downstream target genes in T-ALL cells. However, long non-coding RNAs (lncRNAs) regulated by these factors are largely unknown. Here we established a bioinformatics pipeline and analyzed RNA-seq datasets with deep coverage to identify lncRNAs regulated by TAL1 in T-ALL cells. Our analysis predicted 57 putative lncRNAs that are activated by TAL1. Many of these transcripts were regulated by GATA3, RUNX1, and MYB in a coordinated manner. We identified two novel transcripts that were activated in multiple T-ALL cell samples but were downregulated in normal thymocytes. One transcript near the ARID5B gene locus was specifically expressed in TAL1-positive T-ALL cases. The other transcript located between the FAM49A and MYCN gene locus was also expressed in normal hematopoietic stem cells and T-cell progenitor cells. In addition, we identified a subset of lncRNAs that were negatively regulated by TAL1 and positively regulated by E-proteins in T-ALL cells. This included a known lncRNA (lnc-OAZ3-2:7) located near the RORC gene, which was expressed in normal thymocytes but repressed in TAL1-positive T-ALL cells.
Collapse
|