1
|
Rao G, Mack CD, Nguyen T, Wong N, Payne K, Worley L, Gray PE, Wong M, Hsu P, Stormon MO, Preece K, Suan D, O'Sullivan M, Blincoe AK, Sinclair J, Okada S, Hambleton S, Arkwright PD, Boztug K, Stepensky P, Cooper MA, Bezrodnik L, Nadeau KC, Abolhassani H, Abraham RS, Seppänen MRJ, Béziat V, Bustamante J, Forbes Satter LR, Leiding JW, Meyts I, Jouanguy E, Boisson-Dupuis S, Uzel G, Puel A, Casanova JL, Tangye SG, Ma CS. Inborn errors of immunity reveal molecular requirements for generation and maintenance of human CD4 + IL-9-expressing cells. J Allergy Clin Immunol 2024:S0091-6749(24)01283-1. [PMID: 39622295 DOI: 10.1016/j.jaci.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND CD4+ T cells play essential roles in adaptive immunity. Distinct CD4+ T-cell subsets-TH1, TH2, TH17, TH22, T follicular helper, and regulatory T cells-have been identified, and their contributions to host defense and immune regulation are increasingly well defined. IL-9-producing TH9 cells were first described in 2008 and appear to play both protective and pathogenic roles in human immunity. However, key requirements for generating human TH9 cells remain incompletely defined. OBJECTIVE We sought to define signaling pathways that regulate IL-9 production by human CD4+ T cells. METHODS Human naive and memory CD4+ T cells were cultured under different conditions, and the molecular mechanisms regulating IL-9 induction were determined by assessing the ability of CD4+ T cells from a broad range of patients (n = 92) with pathogenic variants in key immune genes (n = 21) to differentiate into IL-9+ cells. RESULTS We identified 2 culture conditions that yielded IL-9-expressing cells from naive CD4+ T cells and amplified IL-9 production by in vivo-generated memory CD4+ T cells: TGF-β plus IL-4 (ie, TH9 polarizing condition), and the combination of IL-21, IL-23, IL-6, IL-1β, and TGF-β (ie, TH17 polarizing condition). Combining these conditions had a synergistic effect in generating IL-9+CD4+ T cells. IL-9 induction required STAT3-activating cytokines as well as intact signaling via the T-cell receptor and STAT5. Importantly, IL-9 induction was restrained by IFN-γ/STAT1 and IL-10. CONCLUSIONS Our findings revealed critical molecules involved in inducing/restraining IL-9 production by human CD4+ T cells, thereby identifying pathways that could be targeted to modulate IL-9 in health and disease.
Collapse
Affiliation(s)
- Geetha Rao
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Corinne D Mack
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Tina Nguyen
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia
| | - Natalie Wong
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Kathryn Payne
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Lisa Worley
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia
| | - Paul E Gray
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, Australia
| | - Melanie Wong
- Children's Hospital at Westmead, Westmead, Australia; Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Peter Hsu
- Children's Hospital at Westmead, Westmead, Australia; Faculty of Medicine, University of Sydney, Sydney, Australia
| | | | - Kahn Preece
- John Hunter Children's Hospital, Newcastle, Australia
| | - Daniel Suan
- Garvan Institute of Medical Research, Darlinghurst, Australia
| | | | | | - Jan Sinclair
- Starship Children's Hospital, Auckland, New Zealand
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sophie Hambleton
- Primary Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom; Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Peter D Arkwright
- Lydia Becker Institute for Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Kaan Boztug
- St Anna Children's Cancer Research Institute (CCRI), Vienna, Austria; Medical University of Vienna, Department of Paediatrics and Adolescent Medicine, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Polina Stepensky
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University School of Medicine, St Louis, Mo
| | - Liliana Bezrodnik
- Grupo de Inmunología-Instituto Multidisciplinario de Investigaciones en Patologias Pediatricas (IMIPP-CONICET), Hospital de Niños "Dr. Ricardo Gutierrez," Buenos Aires, Argentina; Center for Clinical Immunology, Buenos Aires, Argentina
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, Calif; Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, Calif
| | - Hassan Abolhassani
- Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Rare Diseases Center and Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; ERN-RITA Core Center, RITAFIN, Helsinki, Finland
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Paris, France
| | - Lisa R Forbes Satter
- Department of Pediatrics, Baylor College of Medicine, and Texas Children's Hospital, William T. Shearer Center for Human Immunobiology, Department of Allergy, Immunology, and Retrovirology, Houston, Tex
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, Md; Institute for Clinical and Translational Research and the Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | - Isabelle Meyts
- Department of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium; Department of Pediatrics, Division of Inborn Errors of Immunity, University Hospitals Leuven, Leuven, Belgium; FWO Vlaanderen, Brussels, Belgium
| | - Emmanuelle Jouanguy
- Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Paris, France
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France; Imagine Institute, Université Paris Cité, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Howard Hughes Medical Institute, New York, NY; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales (UNSW), Sydney, Australia.
| |
Collapse
|
2
|
Niese ML, Pajulas AL, Rostron CR, Cheung CCL, Krishnan MS, Zhang J, Cannon AM, Kaplan MH. TL1A priming induces a multi-cytokine Th9 cell phenotype that promotes robust allergic inflammation in murine models of asthma. Mucosal Immunol 2024; 17:537-553. [PMID: 38493956 PMCID: PMC11354665 DOI: 10.1016/j.mucimm.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Multi-cytokine-producing Th9 cells secrete IL-9 and type 2 cytokines and mediate mouse and human allergic inflammation. However, the cytokines that promote a multi-cytokine secreting phenotype have not been defined. Tumor necrosis factor superfamily member TL1A signals through its receptor DR3 to increase IL-9. Here we demonstrate that TL1A increases expression of IL-9 and IL-13 co-expressing cells in murine Th9 cell cultures, inducing a multi-cytokine phenotype. Mechanistically, this is linked to histone modifications allowing for increased accessibility at the Il9 and Il13 loci. We further show that TL1A alters the transcription factor network underlying expression of IL-9 and IL-13 in Th9 cells and increases binding of transcription factors to Il9 and Il13 loci. TL1A-priming enhances the pathogenicity of Th9 cells in murine models of allergic airway disease through the increased expression of IL-9 and IL-13. Lastly, in both chronic and memory-recall models of allergic airway disease, blockade of TL1A signaling decreases the multi-cytokine Th9 cell population and attenuates the allergic phenotype. Taken together, these data demonstrate that TL1A promotes the development of multi-cytokine Th9 cells that drive allergic airway diseases and that targeting pathogenic T helper cell-promoting cytokines could be an effective approach for modifying disease.
Collapse
Affiliation(s)
- Michelle L Niese
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abigail L Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cameron R Rostron
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cherry C L Cheung
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maya S Krishnan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony M Cannon
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Li H, Bradbury JA, Edin ML, Gruzdev A, Li H, Graves JP, DeGraff LM, Lih FB, Feng C, Wolf ER, Bortner CD, London SJ, Sparks MA, Coffman TM, Zeldin DC. TXA2 attenuates allergic lung inflammation through regulation of Th2, Th9, and Treg differentiation. J Clin Invest 2024; 134:e165689. [PMID: 38483511 PMCID: PMC11060738 DOI: 10.1172/jci165689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024] Open
Abstract
In lung, thromboxane A2 (TXA2) activates the TP receptor to induce proinflammatory and bronchoconstrictor effects. Thus, TP receptor antagonists and TXA2 synthase inhibitors have been tested as potential asthma therapeutics in humans. Th9 cells play key roles in asthma and regulate the lung immune response to allergens. Herein, we found that TXA2 reduces Th9 cell differentiation during allergic lung inflammation. Th9 cells were decreased approximately 2-fold and airway hyperresponsiveness was attenuated in lungs of allergic mice treated with TXA2. Naive CD4+ T cell differentiation to Th9 cells and IL-9 production were inhibited dose-dependently by TXA2 in vitro. TP receptor-deficient mice had an approximately 2-fold increase in numbers of Th9 cells in lungs in vivo after OVA exposure compared with wild-type mice. Naive CD4+ T cells from TP-deficient mice exhibited increased Th9 cell differentiation and IL-9 production in vitro compared with CD4+ T cells from wild-type mice. TXA2 also suppressed Th2 and enhanced Treg differentiation both in vitro and in vivo. Thus, in contrast to its acute, proinflammatory effects, TXA2 also has longer-lasting immunosuppressive effects that attenuate the Th9 differentiation that drives asthma progression. These findings may explain the paradoxical failure of anti-thromboxane therapies in the treatment of asthma.
Collapse
Affiliation(s)
- Hong Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - J. Alyce Bradbury
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Artiom Gruzdev
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Huiling Li
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Joan P. Graves
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Laura M. DeGraff
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Fred B. Lih
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Chiguang Feng
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Erin R. Wolf
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Carl D. Bortner
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Stephanie J. London
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| | - Matthew A. Sparks
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
| | - Thomas M. Coffman
- Department of Nephrology, Duke University Medical Center, Durham, North Carolina, USA
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/NIH, Research Triangle Park, North Carolina, USA
| |
Collapse
|
4
|
Roostaee A, Yaghobi R, Afshari A, Jafarinia M. Regulatory role of T helper 9/interleukin-9: Transplantation view. Heliyon 2024; 10:e26359. [PMID: 38420400 PMCID: PMC10900956 DOI: 10.1016/j.heliyon.2024.e26359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
T helper 9 (Th9) cells, a subset of CD4+ T helper cells, have emerged as a valuable target for immune cell therapy due to their potential to induce immunomodulation and tolerance. The Th9 cells mainly produce interleukin (IL)-9 and are known for their defensive effects against helminth infections, allergic and autoimmune responses, and tumor suppression. This paper explores the mechanisms involved in the generation and differentiation of Th9 cells, including the cytokines responsible for their polarization and stabilization, the transcription factors necessary for their differentiation, as well as the role of Th9 cells in inflammatory and autoimmune diseases, allergic reactions, and cancer immunotherapies. Recent research has shown that the differentiation of Th9 cells is coregulated by the transcription factors transforming growth factor β (TGF-β), IL-4, and PU.1, which are also known to secrete IL-10 and IL-21. Multiple cell types, such as T and B cells, mast cells, and airway epithelial cells, are influenced by IL-9 due to its pleiotropic effects.
Collapse
Affiliation(s)
- Azadeh Roostaee
- Department of Genetics, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| | - Ramin Yaghobi
- Shiraz Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Jafarinia
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran
| |
Collapse
|
5
|
Frueh JT, Campe J, Sunaga-Franze DY, Verheyden NA, Ghimire S, Meedt E, Haslinger D, Harenkamp S, Staudenraus D, Sauer S, Kreft A, Schubert R, Lohoff M, Krueger A, Bonig H, Chiocchetti AG, Zeiser R, Holler E, Ullrich E. Interferon regulatory factor 4 plays a pivotal role in the development of aGVHD-associated colitis. Oncoimmunology 2023; 13:2296712. [PMID: 38170159 PMCID: PMC10761041 DOI: 10.1080/2162402x.2023.2296712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Interferon regulatory factor 4 (IRF4) is a master transcription factor that regulates T helper cell (Th) differentiation. It interacts with the Basic leucine zipper transcription factor, ATF-like (BATF), depletion of which in CD4+ T cells abrogates acute graft-versus-host disease (aGVHD)-induced colitis. Here, we investigated the immune-regulatory role of Irf4 in a mouse model of MHC-mismatched bone marrow transplantation. We found that recipients of allogenic Irf4-/- CD4+ T cells developed less GVHD-related symptoms. Transcriptome analysis of re-isolated donor Irf4-/- CD4+ T helper (Th) cells, revealed gene expression profiles consistent with loss of effector T helper cell signatures and enrichment of a regulatory T cell (Treg) gene expression signature. In line with these findings, we observed a high expression of the transcription factor BTB and CNC homolog 2; (BACH2) in Irf4-/- T cells, which is associated with the formation of Treg cells and suppression of Th subset differentiation. We also found an association between BACH2 expression and Treg differentiation in patients with intestinal GVHD. Finally, our results indicate that IRF4 and BACH2 act as counterparts in Th cell polarization and immune homeostasis during GVHD. In conclusion, targeting the BACH2/IRF4-axis could help to develop novel therapeutic approaches against GVHD.
Collapse
Affiliation(s)
- Jochen T. Frueh
- Department of Pediatrics, Experimental Immunology and Cell Therapy, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Julia Campe
- Department of Pediatrics, Experimental Immunology and Cell Therapy, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Daniele Yumi Sunaga-Franze
- Genomics Platform, Max Delbrueck Center for Molecular Medicine, Berlin Institute of Health, Berlin, Germany
| | - Nikita A. Verheyden
- Institute for Molecular Medicine, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Molecular Immunology, Justus Liebig University Giessen, Giessen, Germany
| | - Sakhila Ghimire
- Hematology and Oncology Department, Medical Clinic 3, University Hospital Regensburg, Regensburg, Germany
| | - Elisabeth Meedt
- Hematology and Oncology Department, Medical Clinic 3, University Hospital Regensburg, Regensburg, Germany
| | - Denise Haslinger
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Sabine Harenkamp
- German Red Cross Blood Service BaWüHe, Frankfurt am Main, Germany
| | | | - Sascha Sauer
- Genomics Platform, Max Delbrueck Center for Molecular Medicine, Berlin Institute of Health, Berlin, Germany
| | - Andreas Kreft
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Ralf Schubert
- Department of Pediatric Medicine, Division of Pneumology, Allergology, Infectious diseaes und Gastroenterology. Frankfurt am Main, Goethe University Frankfurt, Frankfurt, Germany
| | - Michael Lohoff
- Institute for Microbiology, Philipps University, Marburg, Germany
| | - Andreas Krueger
- Institute for Molecular Medicine, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Molecular Immunology, Justus Liebig University Giessen, Giessen, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| | - Halvard Bonig
- German Red Cross Blood Service BaWüHe, Frankfurt am Main, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt am Main, Germany
| | - Andreas G. Chiocchetti
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ernst Holler
- Hematology and Oncology Department, Medical Clinic 3, University Hospital Regensburg, Regensburg, Germany
| | - Evelyn Ullrich
- Department of Pediatrics, Experimental Immunology and Cell Therapy, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, a partnership between DKFZ, University Hospital Frankfurt, Frankfurt, Germany
- University Cancer Center (UCT), Frankfurt am Main, Germany
| |
Collapse
|
6
|
Taber A, Konecny A, Oda SK, Scott-Browne J, Prlic M. TGF-β broadly modifies rather than specifically suppresses reactivated memory CD8 T cells in a dose-dependent manner. Proc Natl Acad Sci U S A 2023; 120:e2313228120. [PMID: 37988468 PMCID: PMC10691214 DOI: 10.1073/pnas.2313228120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/16/2023] [Indexed: 11/23/2023] Open
Abstract
Transforming growth factor β (TGF-β) directly acts on naive, effector, and memory T cells to control cell fate decisions, which was shown using genetic abrogation of TGF-β signaling. TGF-β availability is altered by infections and cancer; however, the dose-dependent effects of TGF-β on memory CD8 T cell (Tmem) reactivation are still poorly defined. We examined how activation and TGF-β signals interact to shape the functional outcome of Tmem reactivation. We found that TGF-β could suppress cytotoxicity in a manner that was inversely proportional to the strength of the activating TCR or proinflammatory signals. In contrast, even high doses of TGF-β had a comparatively modest effect on IFN-γ expression in the context of weak and strong reactivation signals. Since CD8 Tmem may not always receive TGF-β signals concurrently with reactivation, we also explored whether the temporal order of reactivation versus TGF-β signals is of importance. We found that exposure to TGF-β before or after an activation event were both sufficient to reduce cytotoxic effector function. Concurrent ATAC-seq and RNA-seq analysis revealed that TGF-β altered ~10% of the regulatory elements induced by reactivation and also elicited transcriptional changes indicative of broadly modulated functional properties. We confirmed some changes on the protein level and found that TGF-β-induced expression of CCR8 was inversely proportional to the strength of the reactivating TCR signal. Together, our data suggest that TGF-β is not simply suppressing CD8 Tmem but modifies functional and chemotactic properties in context of their reactivation signals and in a dose-dependent manner.
Collapse
Affiliation(s)
- Alexis Taber
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
| | - Andrew Konecny
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
- Department of Immunology, University of Washington, Seattle, WA98195
| | - Shannon K. Oda
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA98101
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA98105
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO80206
- Department of Immunology and Microbiology, University of Colorado, Anschutz Medical Campus, Aurora, CO80045
| | - Martin Prlic
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA98109
- Department of Immunology, University of Washington, Seattle, WA98195
| |
Collapse
|
7
|
Li Y, Liu H, He C, Lin Y, Ma L, Xue H. IL-9-Producing Th9 Cells Participate in the Occurrence and Development of Iodine-Induced Autoimmune Thyroiditis. Biol Trace Elem Res 2023; 201:5298-5308. [PMID: 36773201 DOI: 10.1007/s12011-023-03598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023]
Abstract
Iodine excess may cause and aggravate autoimmune thyroiditis (AIT), which is regarded as a typical kind of autoimmune disease mainly mediated by CD4+ T cells. Thus far, it is unclear whether T helper (Th) 9 cells, a novel subpopulation of CD4+ T cells, play a potential role in AIT. Therefore, in the present study, changes in Th9 cells were detected in murine models of AIT induced by excess iodine intake to explore the possible immune mechanism. Female C57BL/6 mice were divided into 7 groups (n = 8) and were supplied with water containing 0.005% sodium iodide for 0, 2, 4, 6, 8, 10, and 12 weeks. With the extension of the high-iodine intake duration, the incidence of thyroiditis and the spleen index were significantly increased, and serum thyroglobulin antibody (TgAb) titers and interleukin 9 (IL-9, major cytokine from Th9 cells) concentrations were also increased. Additionally, it was revealed that the percentages of Th9 cells in spleen mononuclear cells (SMCs) and thyroid tissues were both markedly elevated and accompanied by increased mRNA and protein expression of IL-9 and key transcription factors of Th9 cells (PU.1 and IRF-4). Significantly, dynamic changes in Th9 cells were found, with a peak at 8 weeks after high iodine intake, the time point when thyroiditis was the most serious. Importantly, Th9 cells were detected in the areas of infiltrating lymphocytes in thyroid sections. In conclusion, the continuously increasing proportions of Th9 cells may play an important role in the occurrence and development of AIT induced by high iodine intake.
Collapse
Affiliation(s)
- Yiwen Li
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China
| | - Hao Liu
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China
| | - Chengyan He
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China
| | - Yawen Lin
- Department of Dermatology, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China
| | - Lei Ma
- Department of Dermatology, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China
| | - Haibo Xue
- Department of Endocrinology and Metabolism, Binzhou Medical University Hospital, No. 661 Second Huanghe Road, Binzhou, 256603, China.
| |
Collapse
|
8
|
Chen H, Han Z, Fan Y, Chen L, Peng F, Cheng X, Wang Y, Su J, Li D. CD4+ T-cell subsets in autoimmune hepatitis: A review. Hepatol Commun 2023; 7:e0269. [PMID: 37695088 PMCID: PMC10497257 DOI: 10.1097/hc9.0000000000000269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 09/12/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic autoimmune liver disease that can lead to hepatocyte destruction, inflammation, liver fibrosis, cirrhosis, and liver failure. The diagnosis of AIH requires the identification of lymphoblast cell interface hepatitis and serum biochemical abnormalities, as well as the exclusion of related diseases. According to different specific autoantibodies, AIH can be divided into AIH-1 and AIH-2. The first-line treatment for AIH is a corticosteroid and azathioprine regimen, and patients with liver failure require liver transplantation. However, the long-term use of corticosteroids has obvious side effects, and patients are prone to relapse after drug withdrawal. Autoimmune diseases are characterized by an imbalance in immune tolerance of self-antigens, activation of autoreactive T cells, overactivity of B cells, and increased production of autoantibodies. CD4+ T cells are key players in adaptive immunity and can secrete cytokines, activate B cells to produce antibodies, and influence the cytotoxicity of CD8+ T cells. According to their characteristics, CD4+ T cells can be divided into different subsets. In this review, we discuss the changes in T helper (Th)1, Th2, Th17, Th9, Th22, regulatory T cell, T follicular helper, and T peripheral helper cells and their related factors in AIH and discuss the therapeutic potential of targeting CD4+ T-cell subsets in AIH.
Collapse
Affiliation(s)
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiyue Fan
- Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Liuyan Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Peng
- Chengdu Xinhua Hospital, Chengdu, China
| | | | - Yi Wang
- Chengdu Xinhua Hospital, Chengdu, China
| | - Junyan Su
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | | |
Collapse
|
9
|
Pajulas A, Zhang J, Kaplan MH. The World according to IL-9. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:7-14. [PMID: 37339404 PMCID: PMC10287031 DOI: 10.4049/jimmunol.2300094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/24/2023] [Indexed: 06/22/2023]
Abstract
Among the cytokines regulating immune cells, IL-9 has gained considerable attention for its ability to act on multiple cell types as a regulator of beneficial and pathologic immune responses. Yet, it is still not clearly defined how IL-9 impacts immune responses. IL-9 demonstrates a remarkable degree of tissue-specific functionality and has cellular sources that vary by tissue site and the context of the inflammatory milieu. Here, we provide perspective to summarize the biological activities of IL-9 and highlight cell type-specific roles in the immune pathogenesis of diseases. This perspective will be important in defining the diseases where targeting IL-9 as a therapeutic strategy would be beneficial and where it has the potential to complicate clinical outcomes.
Collapse
Affiliation(s)
- Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jilu Zhang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Shafi T, Rasool R, Ayub S, Bhat IA, Gull A, Hussain S, Hassan Shah I, Shah ZA. Analysis of intronic SNP (rs4147358) and expression of SMAD3 gene in Atopic Dermatitis: A case-control study. Immunobiology 2023; 228:152390. [PMID: 37100019 DOI: 10.1016/j.imbio.2023.152390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND Atopic Dermatitis (AD) is a multifactorial cutaneous disorder associated with chronic inflammation of the skin. Growing evidence points to TGF-β/SMAD signaling as a key player in mediating inflammation and the subsequent tissue remodeling, often resulting in fibrosis. This study investigates the role of a core transcription factor involved in TGF-β signaling i.e., SMAD3 genetic variants (rs4147358) in AD predisposition and its association with SMAD3 mRNA expression, serum IgE levels, and sensitization to various allergens in AD patients. METHODS A total of 246 subjects including 134 AD cases and 112 matched healthy controls were genotyped for SMAD3 intronic SNP by PCR-RFLP. mRNA expression of SMAD3 was determined by quantitative Real-Time PCR (qRT-PCR), Vitamin-D levels by chemiluminescence, and total serum IgE levels by ELISA. In-vivo allergy testing was performed for the evaluation of allergic reactions to house dust mites (HDM) and food allergens. RESULTS A significantly higher frequency of mutant genotype AA (cases: 19.4% vs controls: 8.9%) (OR = 2.8, CI = 1.2 - 6.7, p = 0.01) was observed in AD cases. The mutant allele 'A' also showed a 1.9-fold higher risk for AD compared to the wild allele 'C' indicating that the carriers of the A allele have a higher risk for AD predisposition (OR-1.9, CI = 1.3-2.8, p < 0.001). In addition, quantitative analysis of SMAD3 mRNA in peripheral blood showed 2.8-fold increased expression in AD cases as compared to healthy controls. Stratification analysis revealed the association of the mutant AA genotype with deficient serum Vitamin D levels (p = 0.02) and SMAD3 mRNA overexpression with HDM sensitization (p = 0.03). Furthermore, no significant association of genotypes with SMAD3 mRNA expression was observed. CONCLUSION Our study indicates that SMAD3 intronic SNP bears a significant risk of AD development. Moreover, overexpression of SMAD3 mRNA and its association with HDM sensitization highlights the possible role of this gene in AD pathogenesis.
Collapse
Affiliation(s)
- Tabasum Shafi
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Roohi Rasool
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India.
| | - Sakeena Ayub
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Imtiyaz A Bhat
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Ayaz Gull
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Showkat Hussain
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| | - Iffat Hassan Shah
- Department of Dermatology, Venereology, and Leprosy, GMC- Srinagar 190010, India
| | - Zafar A Shah
- Department of Immunology & Molecular Medicine, SKIMS, Srinagar 190011, India
| |
Collapse
|
11
|
Qiu H, Wang R, Xing J, Li L, Gao Z, Li J, Fang C, Shi F, Mo F, Liu L, Zhao Y, Xie H, Zhao S, Huang J. Characteristics of Th9 cells in Schistosoma japonicum-infected C57BL/6 mouse mesenteric lymph node. Mol Biochem Parasitol 2023; 254:111561. [PMID: 37086898 DOI: 10.1016/j.molbiopara.2023.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
Interleukin 9 (IL-9) is an effective cytokine secreted by newly defined Th9 cells, which is involved in allergic and infectious diseases. In this study, lymphocytes were isolated from mesenteric lymph node (MLN), spleen, liver, lung, and Peyer's patches (PP) of C57BL/6 mice 5-6 weeks after S. japonicum infection, intracellular cytokine staining was done to detect the percentage of IL-9-producing CD4+ T cells. The qPCR and ELISA were used to verify the content of IL-9 in MLN. The population of IL-9-producing lymphocyte subset was identified by FACS. In addition, the dynamic changes and cytokine profiles of Th9 cells in the MLN of infected mice were detected by FACS. ELISA was used to detect IL-9 induced by soluble egg antigen (SEA) from isolated lymphocytes in mouse MLN. The results showed that the percentage of IL-9-secreting Th9 cells in the MLN of the infected mouse was higher than that in the spleen, liver, lung, or PP. Though CD8+ Tc cells, NKT cells, and γδT cells could secrete IL-9, CD4+ Th cells were the main source of IL-9 in S. japonicum-infected C57BL/6 mice (P < 0.05). The percentage of Th9 cells in MLN of infected mouse increased from week 3-4, and reached a peak at week 5-6, then began to decrease from week 7-8 (P < 0.05). Moreover, Th9 cells could also secrete a small amount of IL-4, IFN-γ, IL-5, and IL-10. Our results suggested a higher percentage of Th9 cells was induced in the MLN of S. japonicum-infected mice, which might play an important role in the early stage of S. japonicum-induced disease.
Collapse
Affiliation(s)
- Huaina Qiu
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Ruohan Wang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Junmin Xing
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Lu Li
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhiyan Gao
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiajie Li
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Chao Fang
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Feihu Shi
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Feng Mo
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Lin Liu
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Zhao
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Hongyan Xie
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| | - Shan Zhao
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| | - Jun Huang
- China Sino-French Hoffmann Institute, Department of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
12
|
Wang YH, Peng YJ, Liu FC, Lin GJ, Huang SH, Sytwu HK, Cheng CP. Interleukin 26 Induces Macrophage IL-9 Expression in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24087526. [PMID: 37108686 PMCID: PMC10139149 DOI: 10.3390/ijms24087526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/15/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with chronic inflammation, bone erosion, and joint deformation. Synovial tissue in RA patients is full of proinflammatory cytokines and infiltrated immune cells, such as T help (Th) 9, Th17, macrophages, and osteoclasts. Recent reports emphasized a new member of the interleukin (IL)-10 family, IL-26, an inducer of IL-17A that is overexpressed in RA patients. Our previous works found that IL-26 inhibits osteoclastogenesis and conducts monocyte differentiation toward M1 macrophages. In this study, we aimed to clarify the effect of IL-26 on macrophages linking to Th9 and Th17 in IL-9 and IL-17 expression and downstream signal transduction. Murine and human macrophage cell lines and primary culture cells were used and stimulated by IL26. Cytokines expressions were evaluated by flow cytometry. Signal transduction and transcription factors expression were detected by Western blot and real time-PCR. Our results show that IL-26 and IL-9 colocalized in macrophage in RA synovium. IL-26 directly induces macrophage inflammatory cytokines IL-9 and IL-17A expression. IL-26 increases the IL-9 and IL-17A upstream mechanisms IRF4 and RelB expression. Moreover, the AKT-FoxO1 pathway is also activated by IL-26 in IL-9 and IL-17A expressing macrophage. Blockage of AKT phosphorylation enhances IL-26 stimulating IL-9-producing macrophage cells. In conclusion, our results support that IL-26 promotes IL-9- and IL-17-expressing macrophage and might initiate IL-9- and IL-17-related adaptive immunity in rheumatoid arthritis. Targeting IL-26 may a potential therapeutic strategy for rheumatoid arthritis or other IL-9 plus IL-17 dominant diseases.
Collapse
Affiliation(s)
- Yi-Hsun Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Gu-Jiun Lin
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan
| | - Shing-Hwa Huang
- Division of Breast Surgery, Department of Surgery, New Taipei City Hospital, New Taipei City 241204, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan 35053, Taiwan
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chia-Pi Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
13
|
Khokhar M, Purohit P, Gadwal A, Tomo S, Bajpai NK, Shukla R. The Differentially Expressed Genes Responsible for the Development of T Helper 9 Cells From T Helper 2 Cells in Various Disease States: Immuno-Interactomics Study. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2023; 4:e42421. [PMID: 38935935 PMCID: PMC11135241 DOI: 10.2196/42421] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 06/29/2024]
Abstract
BACKGROUND T helper (Th) 9 cells are a novel subset of Th cells that develop independently from Th2 cells and are characterized by the secretion of interleukin (IL)-9. Studies have suggested the involvement of Th9 cells in variable diseases such as allergic and pulmonary diseases (eg, asthma, chronic obstructive airway disease, chronic rhinosinusitis, nasal polyps, and pulmonary hypoplasia), metabolic diseases (eg, acute leukemia, myelocytic leukemia, breast cancer, lung cancer, melanoma, pancreatic cancer), neuropsychiatric disorders (eg, Alzheimer disease), autoimmune diseases (eg, Graves disease, Crohn disease, colitis, psoriasis, systemic lupus erythematosus, systemic scleroderma, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, atopic dermatitis, eczema), and infectious diseases (eg, tuberculosis, hepatitis). However, there is a dearth of information on its involvement in other metabolic, neuropsychiatric, and infectious diseases. OBJECTIVE This study aims to identify significant differentially altered genes in the conversion of Th2 to Th9 cells, and their regulating microRNAs (miRs) from publicly available Gene Expression Omnibus data sets of the mouse model using in silico analysis to unravel various pathogenic pathways involved in disease processes. METHODS Using differentially expressed genes (DEGs) identified from 2 publicly available data sets (GSE99166 and GSE123501) we performed functional enrichment and network analyses to identify pathways, protein-protein interactions, miR-messenger RNA associations, and disease-gene associations related to significant differentially altered genes implicated in the conversion of Th2 to Th9 cells. RESULTS We extracted 260 common downregulated, 236 common upregulated, and 634 common DEGs from the expression profiles of data sets GSE99166 and GSE123501. Codifferentially expressed ILs, cytokines, receptors, and transcription factors (TFs) were enriched in 7 crucial Kyoto Encyclopedia of Genes and Genomes pathways and Gene Ontology. We constructed the protein-protein interaction network and predicted the top regulatory miRs involved in the Th2 to Th9 differentiation pathways. We also identified various metabolic, allergic and pulmonary, neuropsychiatric, autoimmune, and infectious diseases as well as carcinomas where the differentiation of Th2 to Th9 may play a crucial role. CONCLUSIONS This study identified hitherto unexplored possible associations between Th9 and disease states. Some important ILs, including CCL1 (chemokine [C-C motif] ligand 1), CCL20 (chemokine [C-C motif] ligand 20), IL-13, IL-4, IL-12A, and IL-9; receptors, including IL-12RB1, IL-4RA (interleukin 9 receptor alpha), CD53 (cluster of differentiation 53), CD6 (cluster of differentiation 6), CD5 (cluster of differentiation 5), CD83 (cluster of differentiation 83), CD197 (cluster of differentiation 197), IL-1RL1 (interleukin 1 receptor-like 1), CD101 (cluster of differentiation 101), CD96 (cluster of differentiation 96), CD72 (cluster of differentiation 72), CD7 (cluster of differentiation 7), CD152 (cytotoxic T lymphocyte-associated protein 4), CD38 (cluster of differentiation 38), CX3CR1 (chemokine [C-X3-C motif] receptor 1), CTLA2A (cytotoxic T lymphocyte-associated protein 2 alpha), CTLA28, and CD196 (cluster of differentiation 196); and TFs, including FOXP3 (forkhead box P3), IRF8 (interferon regulatory factor 8), FOXP2 (forkhead box P2), RORA (RAR-related orphan receptor alpha), AHR (aryl-hydrocarbon receptor), MAF (avian musculoaponeurotic fibrosarcoma oncogene homolog), SMAD6 (SMAD family member 6), JUN (Jun proto-oncogene), JAK2 (Janus kinase 2), EP300 (E1A binding protein p300), ATF6 (activating transcription factor 6), BTAF1 (B-TFIID TATA-box binding protein associated factor 1), BAFT (basic leucine zipper transcription factor), NOTCH1 (neurogenic locus notch homolog protein 1), GATA3 (GATA binding protein 3), SATB1 (special AT-rich sequence binding protein 1), BMP7 (bone morphogenetic protein 7), and PPARG (peroxisome proliferator-activated receptor gamma, were able to identify significant differentially altered genes in the conversion of Th2 to Th9 cells. We identified some common miRs that could target the DEGs. The scarcity of studies on the role of Th9 in metabolic diseases highlights the lacunae in this field. Our study provides the rationale for exploring the role of Th9 in various metabolic disorders such as diabetes mellitus, diabetic nephropathy, hypertensive disease, ischemic stroke, steatohepatitis, liver fibrosis, obesity, adenocarcinoma, glioblastoma and glioma, malignant neoplasm of stomach, melanoma, neuroblastoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, and stomach carcinoma.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Nitin Kumar Bajpai
- Department of Nephrology, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Ravindra Shukla
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| |
Collapse
|
14
|
Salmond RJ. Regulation of T Cell Activation and Metabolism by Transforming Growth Factor-Beta. BIOLOGY 2023; 12:297. [PMID: 36829573 PMCID: PMC9953227 DOI: 10.3390/biology12020297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023]
Abstract
Transforming growth factor beta (TGFβ) receptor signalling regulates T cell development, differentiation and effector function. Expression of the immune-associated isoform of this cytokine, TGFβ1, is absolutely required for the maintenance of immunological tolerance in both mice and humans, whilst context-dependent TGFβ1 signalling regulates the differentiation of both anti- and pro-inflammatory T cell effector populations. Thus, distinct TGFβ-dependent T cell responses are implicated in the suppression or initiation of inflammatory and autoimmune diseases. In cancer settings, TGFβ signals contribute to the blockade of anti-tumour immune responses and disease progression. Given the key functions of TGFβ in the regulation of immune responses and the potential for therapeutic targeting of TGFβ-dependent pathways, the mechanisms underpinning these pleiotropic effects have been the subject of much investigation. This review focuses on accumulating evidence suggesting that modulation of T cell metabolism represents a major mechanism by which TGFβ influences T cell immunity.
Collapse
Affiliation(s)
- Robert J Salmond
- Leeds Institute of Medical Research at St. James's, School of Medicine, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
15
|
Huang F, Tong X, Hu C, Zhang Q, Wei Y, Hu M, Kong L, Fu R, Li X, Xie Y, Ming X, Chen B, Lin Y, Xiong L. CAVO Inhibits Airway Inflammation and ILC2s in OVA-Induced Murine Asthma Mice. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8783078. [PMID: 39282108 PMCID: PMC11401656 DOI: 10.1155/2023/8783078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/25/2022] [Accepted: 12/05/2022] [Indexed: 09/18/2024]
Abstract
Cang-ai volatile oil (CAVO) is an aromatic Chinese medicine and is widely used to treat upper respiratory tract infections in children. However, the mechanism of CAVO in asthma treatment is unclear. In this study, we investigated the effects of CAVO on airway inflammation and the mechanism of inhibiting Group-2 innate lymphoid cells (ILC2s) in asthmatic mice, which was induced with Ovalbumin (OVA). CAVO improved AHR and airway inflammation in asthmatic mice. CAVO reduced the production of interleukin (IL)-2, IL-4, IL-5, IL-6, IL-7, IL-9, IL-13, IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) in the bronchoalveolar lavage fluid (BALF), while increased the production of IL-10, significantly. CAVO also inhibited the suppressor of tumorigenicity 2 (ST2) and IL-33 expressions in the lung tissue. Moreover, flow analyses demonstrated that CAVO inhibited ILC2s activation by reducing the sedimentation of its upstream cytokines, thus alleviating downstream cytokines. This could be because of the downregulated microRNA-155 and upregulated microRNA-146a. CAVO inhibits ILC2s activation, thus further attenuating airway inflammation and AHR in asthmatic mice. These effects may be related to the downregulation of microRNA-155 and upregulation of microRNA-146a.
Collapse
Affiliation(s)
- Feng Huang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaoyun Tong
- The First Affiliated Hospital of Yunnan University of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Chunyan Hu
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Qiushi Zhang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Yijie Wei
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
- Department of Pharmacy, Tengchong Hospital of Chinese Medicine, Baoshan, China
| | - Min Hu
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Lingqi Kong
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Rongbing Fu
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
- Department of Ethnic Medicine, Youjiang Medical University for Nationalities, Baise, China
| | - Xiaohong Li
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuhuan Xie
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, China
| | - Xi Ming
- The First Affiliated Hospital of Yunnan University of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Bojun Chen
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuping Lin
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Lei Xiong
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
16
|
The oncogenic JAG1 intracellular domain is a transcriptional cofactor that acts in concert with DDX17/SMAD3/TGIF2. Cell Rep 2022; 41:111626. [DOI: 10.1016/j.celrep.2022.111626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
|
17
|
Park SA, Lim YJ, Ku WL, Zhang D, Cui K, Tang LY, Chia C, Zanvit P, Chen Z, Jin W, Wang D, Xu J, Liu O, Wang F, Cain A, Guo N, Nakatsukasa H, Wu C, Zhang YE, Zhao K, Chen W. Opposing functions of circadian protein DBP and atypical E2F family E2F8 in anti-tumor Th9 cell differentiation. Nat Commun 2022; 13:6069. [PMID: 36241625 PMCID: PMC9568563 DOI: 10.1038/s41467-022-33733-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Interleukin-9 (IL-9)-producing CD4+ T helper cells (Th9) have been implicated in allergy/asthma and anti-tumor immunity, yet molecular insights on their differentiation from activated T cells, driven by IL-4 and transforming growth factor-beta (TGF-β), is still lacking. Here we show opposing functions of two transcription factors, D-binding protein (DBP) and E2F8, in controlling Th9 differentiation. Specifically, TGF-β and IL-4 signaling induces phosphorylation of the serine 213 site in the linker region of the Smad3 (pSmad3L-Ser213) via phosphorylated p38, which is necessary and sufficient for Il9 gene transcription. We identify DBP and E2F8 as an activator and repressor, respectively, for Il9 transcription by pSmad3L-Ser213. Notably, Th9 cells with siRNA-mediated knockdown for Dbp or E2f8 promote and suppress tumor growth, respectively, in mouse tumor models. Importantly, DBP and E2F8 also exhibit opposing functions in regulating human TH9 differentiation in vitro. Thus, our data uncover a molecular mechanism of Smad3 linker region-mediated, opposing functions of DBP and E2F8 in Th9 differentiation.
Collapse
Affiliation(s)
- Sang-A Park
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Yun-Ji Lim
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Wai Lim Ku
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - Dunfang Zhang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Kairong Cui
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - Liu-Ya Tang
- grid.94365.3d0000 0001 2297 5165Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Cheryl Chia
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Peter Zanvit
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Zuojia Chen
- grid.94365.3d0000 0001 2297 5165Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Wenwen Jin
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Dandan Wang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Junji Xu
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Ousheng Liu
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Fu Wang
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Alexander Cain
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Nancy Guo
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Hiroko Nakatsukasa
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| | - Chuan Wu
- grid.94365.3d0000 0001 2297 5165Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Ying E. Zhang
- grid.94365.3d0000 0001 2297 5165Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, 20892 MD USA
| | - Keji Zhao
- grid.94365.3d0000 0001 2297 5165Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, 20892 MD USA
| | - WanJun Chen
- grid.94365.3d0000 0001 2297 5165Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, 20892 MD USA
| |
Collapse
|
18
|
Abstract
Inflammation is a biological process that dynamically alters the surrounding microenvironment, including participating immune cells. As a well-protected organ surrounded by specialized barriers and with immune privilege properties, the central nervous system (CNS) tightly regulates immune responses. Yet in neuroinflammatory conditions, pathogenic immunity can disrupt CNS structure and function. T cells in particular play a key role in promoting and restricting neuroinflammatory responses, while the inflamed CNS microenvironment can influence and reshape T cell function and identity. Still, the contraction of aberrant T cell responses within the CNS is not well understood. Using autoimmunity as a model, here we address the contribution of CD4 T helper (Th) cell subsets in promoting neuropathology and disease. To address the mechanisms antagonizing neuroinflammation, we focus on the control of the immune response by regulatory T cells (Tregs) and describe the counteracting processes that preserve their identity under inflammatory challenges. Finally, given the influence of the local microenvironment on immune regulation, we address how CNS-intrinsic signals reshape T cell function to mitigate abnormal immune T cell responses.
Collapse
Affiliation(s)
- Nail Benallegue
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Hania Kebir
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jorge I. Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
19
|
Musiol S, Alessandrini F, Jakwerth CA, Chaker AM, Schneider E, Guerth F, Schnautz B, Grosch J, Ghiordanescu I, Ullmann JT, Kau J, Plaschke M, Haak S, Buch T, Schmidt-Weber CB, Zissler UM. TGF-β1 Drives Inflammatory Th Cell But Not Treg Cell Compartment Upon Allergen Exposure. Front Immunol 2022; 12:763243. [PMID: 35069535 PMCID: PMC8777012 DOI: 10.3389/fimmu.2021.763243] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
TGF-β1 is known to have a pro-inflammatory impact by inducing Th9 and Th17 cells, while it also induces anti-inflammatory Treg cells (Tregs). In the context of allergic airway inflammation (AAI) its dual role can be of critical importance in influencing the outcome of the disease. Here we demonstrate that TGF-β is a major player in AAI by driving effector T cells, while Tregs differentiate independently. Induction of experimental AAI and airway hyperreactivity in a mouse model with inducible genetic ablation of the gene encoding for TGFβ-receptor 2 (Tgfbr2) on CD4+T cells significantly reduced the disease phenotype. Further, it blocked the induction of pro-inflammatory T cell frequencies (Th2, Th9, Th17), but increased Treg cells. To translate these findings into a human clinically relevant context, Th2, Th9 and Treg cells were quantified both locally in induced sputum and systemically in blood of allergic rhinitis and asthma patients with or without allergen-specific immunotherapy (AIT). Natural allergen exposure induced local and systemic Th2, Th9, and reduced Tregs cells, while therapeutic allergen exposure by AIT suppressed Th2 and Th9 cell frequencies along with TGF-β and IL-9 secretion. Altogether, these findings support that neutralization of TGF-β represents a viable therapeutic option in allergy and asthma, not posing the risk of immune dysregulation by impacting Tregs cells.
Collapse
Affiliation(s)
- Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany.,Department of Otorhinolaryngology, Klinikum rechts der Isar, TUM School of Medicine, Technical University Munich, Munich, Germany
| | - Evelyn Schneider
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Johanna Grosch
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ileana Ghiordanescu
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Julia T Ullmann
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Josephine Kau
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Mirjam Plaschke
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Stefan Haak
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Members of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
20
|
SANTOS SOBRINHO ELIANEM, SANTOS HÉRCULESO, MARTINS ERNANER, FONSECA FRANCINESOUZAALVESDA, FARIAS LUCYANAC, AGUILAR CHARLESM, PEREIRA ULISSESA, NICOLAU JUNIOR NILSON, GOMES MATHEUSS, SOUZA CINTYANDE, RAVNJAK JOÃOMATHEUSA, PORTO RAPHAELR, ALMEIDA ANNACHRISTINADE. Protein-coding gene interaction network prediction of bioactive plant compound action against SARS-CoV-2: a novel hypothesis using bioinformatics analysis. AN ACAD BRAS CIENC 2022; 94:e20201380. [DOI: 10.1590/0001-3765202220201380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/23/2021] [Indexed: 11/23/2022] Open
|
21
|
Askari AA, Feizollahi P, Rezaiemanesh A, Salari F, Gorgin Karaji A. The effect of treatment with fexofenadine and fluticasone propionate on the gene expression levels of Th9 transcription factors in patients with allergic rhinitis. Heliyon 2021; 7:e08556. [PMID: 34917820 PMCID: PMC8665345 DOI: 10.1016/j.heliyon.2021.e08556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/20/2021] [Accepted: 12/02/2021] [Indexed: 12/02/2022] Open
Abstract
T helper-9 (Th9) is a new T cell subset involved in allergic rhinitis (AR) pathogenesis. Fexofenadine and fluticasone propionate are the first effective line of AR treatment. This study aimed to evaluate the effect of fexofenadine and fluticasone propionate on the gene expression levels of Interferon regulatory factor 4 (IRF4), B cell-activating transcription factor-like (BATF), and SPI1 gene-encoded protein (PU.1), essential transcription factors for Th9 cell differentiation, in AR patients. Twenty-six AR patients (aged 32.8 ± 9.1 years, 13 men and 13 women) were treated with fexofenadine and fluticasone propionate for one month. Expression levels of PU.1, IRF4, and BATF genes were measured using Real-Time PCR. Our results showed that after one month of treatment, the expression level of IRF4 and BATF genes decreased significantly (P < 0.001, P < 0.01, respectively), while PU.1 gene expression was not remarkably different. Overall, our results showed that after one month of treatment with fexofenadine and fluticasone propionate, the expression levels of IRF4 and BATF genes in AR patients decreased, which may be due to this treatment regimen. However, the exact mechanism of action of fexofenadine and fluticasone propionate needs further study.
Collapse
Affiliation(s)
- Ali Asghar Askari
- Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Feizollahi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Rezaiemanesh
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farhad Salari
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Gorgin Karaji
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
22
|
Naik A, Dalpatraj N, Thakur N. Global Histone H3 Lysine 4 Trimethylation (H3K4me3) Landscape Changes in Response to TGFβ. Epigenet Insights 2021; 14:25168657211051755. [PMID: 34671716 PMCID: PMC8521735 DOI: 10.1177/25168657211051755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/21/2021] [Indexed: 01/12/2023] Open
Abstract
TGFβ expression acts as a biomarker of poor prognosis in prostate cancer. It plays a dual functional role in prostate cancer. In the early stages of the tumor, it acts as a tumor suppressor while at the later stages of tumor development, it promotes metastasis. The molecular mechanisms of action of TGFβ are largely understood through the canonical and non-canonical signal transduction pathways. Our understanding of the mechanisms that establish transient TGFβ stimulation into stable gene expression patterns remains incomplete. Epigenetic marks like histone H3 modifications are directly linked with gene expression and they play an important role in tumorigenesis. In this report, we performed chromatin immunoprecipitation-sequencing (ChIP-Seq) to identify the genome-wide regions that undergo changes in histone H3 Lysine 4 trimethylation (H3K4me3) occupancy in response to TGFβ stimulation. We also show that TGFβ stimulation can induce acute epigenetic changes through the modulation of H3K4me3 signals at genes belonging to special functional categories in prostate cancer. TGFβ induces the H3K4me3 on its own ligands like TGFβ, GDF1, INHBB, GDF3, GDF6, BMP5 suggesting a positive feedback loop. The majority of genes were found to be involved in the positive regulation of transcription from the RNA polymerase II promoter in response to TGFβ. Other functional categories were intracellular protein transport, brain development, EMT, angiogenesis, antigen processing, antigen presentation via MHC class II, lipid transport, embryo development, histone H4 acetylation, positive regulation of cell cycle arrest, and genes involved in mitotic G2 DNA damage checkpoints. Our results link TGFβ stimulation to acute changes in gene expression through an epigenetic mechanism. These findings have broader implications on epigenetic bases of acute gene expression changes caused by growth factor stimulation.
Collapse
Affiliation(s)
- Ankit Naik
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad, Gujarat, India
| | - Nidhi Dalpatraj
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad, Gujarat, India
| | - Noopur Thakur
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Navrangpura, Ahmedabad, Gujarat, India
| |
Collapse
|
23
|
Lourenço LO, Ribeiro AM, Lopes FDTQDS, Tibério IDFLC, Tavares-de-Lima W, Prado CM. Different Phenotypes in Asthma: Clinical Findings and Experimental Animal Models. Clin Rev Allergy Immunol 2021; 62:240-263. [PMID: 34542807 DOI: 10.1007/s12016-021-08894-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
Asthma is a respiratory allergic disease presenting a high prevalence worldwide, and it is responsible for several complications throughout life, including death. Fortunately, asthma is no longer recognized as a unique manifestation but as a very heterogenic manifestation. Its phenotypes and endotypes are known, respectively, as pathologic and molecular features that might not be directly associated with each other. The increasing number of studies covering this issue has brought significant insights and knowledge that are constantly expanding. In this review, we intended to summarize this new information obtained from clinical studies, which not only allowed for the creation of patient clusters by means of personalized medicine and a deeper molecular evaluation, but also created a connection with data obtained from experimental models, especially murine models. We gathered information regarding sensitization and trigger and emphasizing the most relevant phenotypes and endotypes, such as Th2-high asthma and Th2-low asthma, which included smoking and obesity-related asthma and mixed and paucigranulocytic asthma, not only in physiopathology and the clinic but also in how these phenotypes can be determined with relative similarity using murine models. We also further investigated how clinical studies have been treating patients using newly developed drugs focusing on specific biomarkers that are more relevant according to the patient's clinical manifestation of the disease.
Collapse
Affiliation(s)
- Luiz Otávio Lourenço
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil
| | - Alessandra Mussi Ribeiro
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil
| | | | | | - Wothan Tavares-de-Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Carla Máximo Prado
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil. .,Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
24
|
Pei S, Huang M, Huang J, Zhu X, Wang H, Romano S, Deng X, Wang Y, Luo Y, Hao S, Xu J, Yu T, Zhu Q, Yuan J, Shen K, Liu Z, Hu G, Peng C, Luo Q, Wen Z, Dai D, Xiao Y. BFAR coordinates TGFβ signaling to modulate Th9-mediated cancer immunotherapy. J Exp Med 2021; 218:212036. [PMID: 33914044 PMCID: PMC8091105 DOI: 10.1084/jem.20202144] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/25/2021] [Accepted: 03/04/2021] [Indexed: 01/05/2023] Open
Abstract
TGFβ is essential for the generation of anti-tumor Th9 cells; on the other hand, it causes resistance against anti-tumor immunity. Despite recent progress, the underlying mechanism reconciling the double-edged effect of TGFβ signaling in Th9-mediated cancer immunotherapy remains elusive. Here, we find that TGFβ-induced down-regulation of bifunctional apoptosis regulator (BFAR) represents the key mechanism preventing the sustained activation of TGFβ signaling and thus impairing Th9 inducibility. Mechanistically, BFAR mediates K63-linked ubiquitination of TGFβR1 at K268, which is critical to activate TGFβ signaling. Thus, BFAR deficiency or K268R knock-in mutation suppresses TGFβR1 ubiquitination and Th9 differentiation, thereby inhibiting Th9-mediated cancer immunotherapy. More interestingly, BFAR-overexpressed Th9 cells exhibit promising therapeutic efficacy to curtail tumor growth and metastasis and promote the sensitivity of anti–PD-1–mediated checkpoint immunotherapy. Thus, our findings establish BFAR as a key TGFβ-regulated gene to fine-tune TGFβ signaling that causes Th9 induction insensitivity, and they highlight the translational potential of BFAR in promoting Th9-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Siyu Pei
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingzhu Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jia Huang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hui Wang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, Federico II, Naples, Italy
| | - Xiuyu Deng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yixiao Luo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shumeng Hao
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing Xu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tao Yu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingchen Zhu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Yuan
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiqiang Liu
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Guohong Hu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai, China
| | - Qingquan Luo
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenzhen Wen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongfang Dai
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yichuan Xiao
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Ma C, Qi Y, Liu H, Wu C, Cui X, Liu Z. Inhibitory effect of activin A on IL-9 production by mouse NK cells through Smad3 signaling. Biol Chem 2021; 401:297-308. [PMID: 31400749 DOI: 10.1515/hsz-2019-0245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022]
Abstract
Interleukin-9 (IL-9) is a cytokine secreted by T-helper (Th)9 cells, and activin A can enhance Th9 cell differentiation. However, whether activin A affects IL-9 production by natural killer (NK) cells remains unclear. Herein, we found that not only Th cells, but also CD3-CD49b+NKp46+ NK cells of Balb/c mice produced IL-9. Although activin A promoted IL-9 expression in CD4+ Th cells, it inhibited IL-9 production by CD49b+NKp46+ NK cells in mice. Furthermore, the enzyme-linked immunosorbent assay (ELISA) results showed that mouse NK cells could secrete mature IL-9 protein, and activin A inhibited IL-9 release by NK cells. Additionally, activin A inhibited interferon (IFN)-γ production in splenic NK cells in mice, but promoted IL-2 production, and did not alter the production of IL-10. Western blotting results showed that levels of activin type IIA receptor (ActRIIA), Smad3 and phosphorylated-Smad3 (p-SMAD3) protein increased in activin A-treated splenic NK cells, compared with that in control NK cells. The inhibitory effects of activin A on IL-9 production by NK cells were attenuated in the presence of activin antagonist follistatin (FST) or Smad3 knockdown to NK cells. These data suggest that although activin A up-regulates IL-9 expression in Th cells, it inhibits IL-9 production in NK cells through Smad3 signaling.
Collapse
Affiliation(s)
- Chunhui Ma
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Yan Qi
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Haiyan Liu
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Chengdong Wu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Xueling Cui
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhonghui Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| |
Collapse
|
26
|
Chen J, Ding Y, Huang F, Lan R, Wang Z, Huang W, Chen R, Wu B, Fu L, Yang Y, Liu J, Hong J, Zhang W, Zhang L. Irradiated whole-cell vaccine suppresses hepatocellular carcinoma growth in mice via Th9 cells. Oncol Lett 2021; 21:409. [PMID: 33841570 PMCID: PMC8020379 DOI: 10.3892/ol.2021.12670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Liver cancer is one of the most common malignant tumors with no available satisfactory treatment. The aim of the present study was to investigate the anti-tumor effect of an irradiated hepatocellular carcinoma (HCC) whole-cell vaccine and its underlying mechanisms. Hepa1-6 and H22 HCC cell lines were irradiated in preparation for whole-cell vaccine production. Subsequently, two HCC tumor-bearing mouse models were created by injecting these Hepa1-6 and H22 cells into the abdominal skin of C57BL/6 and ICR mice, respectively. The mice were immunized with the corresponding whole-cell vaccine the next day, and then once a week until the end of the experimental period. Tumor growth, blood T helper (Th)9 cells and plasma interleukin (IL)-9 levels were monitored during the immunization period. Th9 cells were also induced by in vitro co-culture of the whole-cell vaccine with lymphocytes from the spleen and lymph nodes of the corresponding mice. Alterations of gene expression in transcription factor (TF) were determined by reverse transcription-quantitative PCR, and Th9 cells were detected using flow cytometry. The whole-cell vaccine effectively suppressed HCC tumor growth, as indicated by slower tumor growth and a smaller tumor size in the immunized group compared with the control. The percentage of blood Th9 cells and the concentration of plasma IL-9 were significantly increased in the immunized group. The whole-cell vaccine also induced Th9 cell differentiation and upregulated the expression of TFs PU.1, interferon regulatory factor 4 and basic leucine zipper transcriptional factor ATF-like. These results suggest that the irradiated HCC whole-cell vaccine inhibited tumor growth by increasing Th9 cell numbers in HCC mice
Collapse
Affiliation(s)
- Junying Chen
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yuxiong Ding
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Fei Huang
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ruilong Lan
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zeng Wang
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Weikang Huang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Ruiqing Chen
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Bing Wu
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Lengxi Fu
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Yunhua Yang
- Department of Otolaryngology, Fujian Provincial Geriatric Hospital, Fuzhou, Fujian 350009, P.R. China
| | - Jun Liu
- Laboratory of Radiobiology, Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| | - Jinsheng Hong
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Weijian Zhang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Lurong Zhang
- Central Laboratory, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Key Laboratory of Radiation Biology of Fujian Province Universities, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China.,Laboratory of Radiobiology, Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, P.R. China
| |
Collapse
|
27
|
Critical Roles of Balanced T Helper 9 Cells and Regulatory T Cells in Allergic Airway Inflammation and Tumor Immunity. J Immunol Res 2021; 2021:8816055. [PMID: 33748292 PMCID: PMC7943311 DOI: 10.1155/2021/8816055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 01/02/2023] Open
Abstract
CD4+T helper (Th) cells are important mediators of immune responses in asthma and cancer. When counteracted by different classes of pathogens, naïve CD4+T cells undergo programmed differentiation into distinct types of Th cells. Th cells orchestrate antigen-specific immune responses upon their clonal T-cell receptor (TCR) interaction with the appropriate peptide antigen presented on MHC class II molecules expressed by antigen-presenting cells (APCs). T helper 9 (Th9) cells and regulatory T (Treg) cells and their corresponding cytokines have critical roles in tumor and allergic immunity. In the context of asthma and cancer, the dynamic internal microenvironment, along with chronic inflammatory stimuli, influences development, differentiation, and function of Th9 cells and Treg cells. Furthermore, the dysregulation of the balance between Th9 cells and Treg cells might trigger aberrant immune responses, resulting in development and exacerbation of asthma and cancer. In this review, the development, differentiation, and function of Th9 cells and Treg cells, which are synergistically regulated by various factors including cytokine signals, transcriptional factors (TFs), costimulatory signals, microenvironment cues, metabolic pathways, and different signal pathways, will be discussed. In addition, we focus on the recent progress that has helped to achieve a better understanding of the roles of Th9 cells and Treg cells in allergic airway inflammation and tumor immunity. We also discuss how various factors moderate their responses in asthma and cancer. Finally, we summarize the recent findings regarding potential mechanisms for regulating the balance between Th9 and Treg cells in asthma and cancer. These advances provide opportunities for novel therapeutic strategies that are aimed at reestablishing the balance of these cells in the diseases.
Collapse
|
28
|
Chen T, Guo J, Cai Z, Li B, Sun L, Shen Y, Wang S, Wang Z, Wang Z, Wang Y, Zhou H, Cai Z, Ye Z. Th9 Cell Differentiation and Its Dual Effects in Tumor Development. Front Immunol 2020; 11:1026. [PMID: 32508847 PMCID: PMC7251969 DOI: 10.3389/fimmu.2020.01026] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
With the improved understanding of the molecular pathogenesis and characteristics of cancers, the critical role of the immune system in preventing tumor development has been widely accepted. The understanding of the relationship between the immune system and cancer progression is constantly evolving, from the cancer immunosurveillance hypothesis to immunoediting theory and the delicate balance in the tumor microenvironment. Currently, immunotherapy is regarded as a promising strategy against cancers. Although adoptive cell therapy (ACT) has shown some exciting results regarding the rejection of tumors, the effect is not always satisfactory. Cellular therapy with CD4+ T cells remains to be further explored since the current ACT is mainly focused on CD8+ cytotoxic T lymphocytes (CTLs). Recently, Th9 cells, a subgroup of CD4+ T helper cells characterized by the secretion of IL-9 and IL-10, have been reported to be effective in the elimination of solid tumors and to exhibit superior antitumor properties to Th1 and Th17 cells. In this review, we summarize the most recent advances in the understanding of Th9 cell differentiation and the dual role, both anti-tumor and pro-tumor effects, of Th9 cells in tumor progression.
Collapse
Affiliation(s)
- Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenhai Cai
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Binghao Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Lingling Sun
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Yingying Shen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zenan Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Yucheng Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| | - Zhijian Cai
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China.,Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institute of Orthopedic Research, Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Crepeau RL, Ford ML. Programmed T cell differentiation: Implications for transplantation. Cell Immunol 2020; 351:104099. [PMID: 32247511 DOI: 10.1016/j.cellimm.2020.104099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/27/2022]
Abstract
While T cells play a critical role in protective immunity against infection, they are also responsible for graft rejection in the setting of transplantation. T cell differentiation is regulated by both intrinsic transcriptional pathways as well as extrinsic factors such as antigen encounter and the cytokine milieu. Herein, we review recent discoveries in the transcriptional regulation of T cell differentiation and their impact on the field of transplantation. Recent studies uncovering context-dependent differentiation programs that differ in the setting of infection or transplantation will also be discussed. Understanding the key transcriptional pathways that underlie T cell responses in transplantation has important clinical implications, including development of novel therapeutic agents to mitigate graft rejection.
Collapse
Affiliation(s)
- Rebecca L Crepeau
- Emory Transplant Center, Department of Surgery, Emory University, 101 Woodruff Circle, Suite 5208, Atlanta, GA 30322, United States
| | - Mandy L Ford
- Emory Transplant Center, Department of Surgery, Emory University, 101 Woodruff Circle, Suite 5208, Atlanta, GA 30322, United States.
| |
Collapse
|
30
|
Abdelaziz MH, Wang H, Cheng J, Xu H. Th2 cells as an intermediate for the differentiation of naïve T cells into Th9 cells, associated with the Smad3/Smad4 and IRF4 pathway. Exp Ther Med 2020; 19:1947-1954. [PMID: 32104253 DOI: 10.3892/etm.2020.8420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 06/26/2019] [Indexed: 01/12/2023] Open
Abstract
Type 9 T-helper (Th9) cells are associated with atopic and inflammatory diseases. Their increased levels and functions contribute to a number of inflammatory disorders, where they are accompanied by enhanced Th2-cell activity. However, there is currently no consensus regarding the association between Th9 and Th2 cells. Th9 cells may be induced from naïve T (Th0) cells under specific polarization conditions in vitro, a process driven by the addition of specific cytokines. In the present study, Th0 cells were cultured under Th9-polarizing conditions to promote differentiation into interleukin (IL)-4+ IL-9- or IL-4- IL-9+ T cells after 3 or 5 days in culture, respectively; the mRNA expression levels of IL-9 and IL-4 were consistent with the induced cell types. Simultaneously, the levels of interferon-regulatory factor 4 (IRF-4) and Smad3/Smad4 were significantly increased following Th9-cell polarization. It was therefore proposed that Th2 cells may be generated in the early stages of Th9-cell differentiation, and then ultimately differentiate into Th9 cells via the Smad3/Smad4 and IRF-4 activation pathway.
Collapse
Affiliation(s)
- Mohamed Hamed Abdelaziz
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Huixuan Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jianjun Cheng
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Laboratory Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huaxi Xu
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
31
|
Jang YW, Gil KC, Lee JS, Kang W, Park SY, Hwang KW. T-Cell Differentiation to T Helper 9 Phenotype is Elevated by Extremely Low-Frequency Electromagnetic Fields Via Induction of IL-2 Signaling. Bioelectromagnetics 2019; 40:588-601. [PMID: 31663626 DOI: 10.1002/bem.22219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 08/27/2019] [Indexed: 01/16/2023]
Abstract
Owing to the development of information technology and the electronics industry, and the increase in the use of electronic products, an increasing number of people are exposed to electromagnetic fields (EMFs) in daily life. There has been concern about the effects of EMFs on the human body. Th9 cells, which are characterized by the generation of interleukin-(IL-9), are a recently defined subset of T helper (Th) cells. In this study, we investigated the effect of extremely low-frequency (60 Hz) EMFs, such as those generated by household power sources, at 0.8 mT intensity on CD4+ T cells. The exposure of CD4+ T cells to such EMFs under Th9-polarizing conditions increased IL-9 secretion and gene expression of transcription factors that are important for Th9 development. The expression of GATA3 increased in the early stage, and the phosphorylation of STAT5 and STAT6, which regulate the expression of GATA3, increased. In addition, EMFs increased the expression of IL-2 by the T cells. In conclusion, the differentiation of CD4+ T cells to the Th9 phenotype was increased by exposure to extremely low-frequency EMFs, and this appeared to be dependent on the IL-2 signaling pathway. Furthermore, co-cultures of EMF-exposed Th9 cells and mast cells showed an increased expression of mast cell proteases, FcεR1α, and mast cell-derived inflammatory cytokines compared with co-cultures of non-EMF-exposed Th9 cells and mast cells. Our results suggest that EMFs enhance the differentiation of CD4+ T cells to the Th9 phenotype, resulting in mast cell activation and inflammation. Bioelectromagnetics. 2019;40:588-601. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Ye Won Jang
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ki Cheol Gil
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Soo Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - WonKu Kang
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - So-Young Park
- Laboratory of Pharmacognosy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Kwang Woo Hwang
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Fu Y, Koh B, Kuwahara M, Ulrich BJ, Kharwadkar R, Yamashita M, Kaplan MH. BATF-Interacting Proteins Dictate Specificity in Th Subset Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:1989-1998. [PMID: 31451674 PMCID: PMC6761015 DOI: 10.4049/jimmunol.1900128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 08/02/2019] [Indexed: 12/24/2022]
Abstract
The basic leucine zipper (bZIP) transcription factor BATF is expressed in multiple Th subsets and cooperates with other factors to regulate gene transcription. BATF activates lineage-specific cytokines in Th subsets, activating IL-9 in Th9 cells and IL-17 in Th17 cells, but not IL-9 or IL-17 in the reciprocal subset. The mechanism for this restricted activity is unclear. In this report, we define BATF binding partners that contribute to Th subset-specific functions. Although BATF and IRF4 are expressed in greater amounts in Th9 than Th17, increased expression of both factors is not sufficient to induce IL-9 in Th17 cells. BATF also requires heterodimer formation with Jun family members to bind DNA and induce gene expression. Using primary mouse T cell culture, we observed that JunB and c-Jun, but not JunD, promote IL-9 production in Th9 cells. Ectopic expression of BATF with either JunB or c-Jun generates modest, but significant, increases in IL-9 production in Th17 cells, suggesting that the low expression of Jun family members is one factor limiting the ability of BATF to induce IL-9 in Th17 cells. We further identified that Bach2 positively regulates IL-9 production by directly binding to the Il9 gene and by increasing transcription factor expression in Th9 cells. Strikingly, cotransduction of Bach2 and BATF significantly induces IL-9 production in both Th9 and Th17 cells. Taken together, our results reveal that JunB, c-Jun, and Bach2 cooperate with BATF to contribute to the specificity of BATF-dependent cytokine induction in Th subsets.
Collapse
Affiliation(s)
- Yongyao Fu
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN 46202
- Department of Microbiology and Immunology, School of Medicine, Indiana University, Indianapolis, IN 46202
| | - Byunghee Koh
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN 46202
| | - Makoto Kuwahara
- Department of Immunology, Ehime University, Shitsukawa, Toon-Shi, Ehime 791-0295, Japan; and
| | - Benjamin J Ulrich
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN 46202
- Department of Microbiology and Immunology, School of Medicine, Indiana University, Indianapolis, IN 46202
| | - Rakshin Kharwadkar
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN 46202
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, IN 46202
| | - Masakatsu Yamashita
- Department of Immunology, Ehime University, Shitsukawa, Toon-Shi, Ehime 791-0295, Japan; and
| | - Mark H Kaplan
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN 46202;
- Department of Microbiology and Immunology, School of Medicine, Indiana University, Indianapolis, IN 46202
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, IN 46202
| |
Collapse
|
33
|
Hu B, Li G, Ye Z, Gustafson CE, Tian L, Weyand CM, Goronzy JJ. Transcription factor networks in aged naïve CD4 T cells bias lineage differentiation. Aging Cell 2019; 18:e12957. [PMID: 31264370 PMCID: PMC6612640 DOI: 10.1111/acel.12957] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/17/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
With reduced thymic activity, the population of naïve T cells in humans is maintained by homeostatic proliferation throughout adult life. In young adults, naïve CD4 T cells have enormous proliferative potential and plasticity to differentiate into different lineages. Here, we explored whether naïve CD4 T-cell aging is associated with a partial loss of this unbiased multipotency. We find that naïve CD4 T cells from older individuals have developed a propensity to develop into TH9 cells. Two major mechanisms contribute to this predisposition. First, responsiveness to transforming growth factor β (TGFβ) stimulation is enhanced with age due to an upregulation of the TGFβR3 receptor that results in increased expression of the transcription factor PU.1. Secondly, aged naïve CD4 T cells display altered transcription factor profiles in response to T-cell receptor stimulation, including enhanced expression of BATF and IRF4 and reduced expression of ID3 and BCL6. These transcription factors are involved in TH9 differentiation as well as IL9 transcription suggesting that the aging-associated changes in the transcription factor profile favor TH9 commitment.
Collapse
Affiliation(s)
- Bin Hu
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Guangjin Li
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Zhongde Ye
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Claire E. Gustafson
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Lu Tian
- Department of Biomedical Data ScienceStanford University School of MedicineStanfordCaliforniaUSA
| | - Cornelia M. Weyand
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| | - Jörg J. Goronzy
- Department of Medicine, Division of Immunology and RheumatologyStanford UniversityStanfordCaliforniaUSA
- Department of MedicinePalo Alto Veterans Administration Healthcare SystemPalo AltoCaliforniaUSA
| |
Collapse
|
34
|
Abstract
PURPOSES OF REVIEW Th9 cells are recognized as a novel subset of effector T helper cells that preferentially produce IL-9. Here, we provide a current update on the reports related to the function of Th9 cells in allergic inflammatory diseases. RECENT FINDINGS The effector Th9 cells differentiating from naïve T helper cells have recently been identified. Because of accumulating findings of Th9 cells in many inflammatory diseases, including allergic diseases, diverse functions of Th9 cells in regulating immune responses have been suggested. Related reports indicate multiple sources of IL-9 besides Th9 cells and their association with the pathogenesis of allergic rhinitis, asthma, atopic dermatitis, contact dermatitis, and food allergy. More recently, elements of the epigenetic landscape involving in the regulation of IL-9 by Th9 cells have been identified to be the potential target for allergic inflammation. This review provides the most recent information about Th9 cells and their contribution in airway allergic disease, skin, and food allergy.
Collapse
Affiliation(s)
- Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
35
|
Yazdani R, Shapoori S, Rezaeepoor M, Sanaei R, Ganjalikhani-Hakemi M, Azizi G, Rae W, Aghamohammadi A, Rezaei N. Features and roles of T helper 9 cells and interleukin 9 in immunological diseases. Allergol Immunopathol (Madr) 2019; 47:90-104. [PMID: 29703631 DOI: 10.1016/j.aller.2018.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/28/2018] [Accepted: 02/09/2018] [Indexed: 02/08/2023]
Abstract
T helper 9 (TH9) cells are considered as newly classified helper T cells that have an important role in the regulation of immune responses. Since these cells preferentially produce IL-9, these cells are termed TH9 cells. Recently, the role of TH9 and its signature cytokine (IL-9) has been investigated in a wide range of diseases, including autoimmunity, allergy, infections, cancer and immunodeficiency. Herein, we review the most recent data concerning TH9 cells and IL-9 as well as their roles in disease. These insights suggest that TH9 cells are a future target for therapeutic intervention.
Collapse
|
36
|
Worley L, Tangye SG, Ma CS. What can primary immunodeficiencies teach us about Th9 cell differentiation and function? Immunol Cell Biol 2018; 97:380-388. [PMID: 30357921 DOI: 10.1111/imcb.12215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022]
Abstract
Interleukin-9 (IL-9) producing CD4+ Th9 cells are a unique subset of effector cells involved in both health and disease. Th9 cells have been associated with protective immunity during parasitic infections with helminths, protozoans and extracellular pathogens, but implicated in disease states such as allergic asthma, atopic dermatitis, food allergy and autoimmune conditions including multiple sclerosis and ulcerative colitis. Here, we review the cytokine signaling pathways and downstream transcription factors required for IL-9 expression and how human primary immunodeficiencies caused by monogenic mutations can help elucidate the complex requirements for human Th9 cell differentiation. Primary immunodeficiencies are a platform for investigating IL-9 expression in primary human lymphocytes and by inference whether Th9 cells are implicated in the clinical phenotype characteristic of these patients.
Collapse
Affiliation(s)
- Lisa Worley
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Clinical Immunogenomics Research Consortia of Australia, Sydney, NSW, Australia
| | - Cindy S Ma
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia.,Clinical Immunogenomics Research Consortia of Australia, Sydney, NSW, Australia
| |
Collapse
|
37
|
A conserved enhancer regulates Il9 expression in multiple lineages. Nat Commun 2018; 9:4803. [PMID: 30442929 PMCID: PMC6237898 DOI: 10.1038/s41467-018-07202-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022] Open
Abstract
Cytokine genes are regulated by multiple regulatory elements that confer tissue-specific and activation-dependent expression. The cis-regulatory elements of the gene encoding IL-9, a cytokine that promotes allergy, autoimmune inflammation and tumor immunity, have not been defined. Here we identify an enhancer (CNS-25) upstream of the Il9 gene that binds most transcription factors (TFs) that promote Il9 gene expression. Deletion of the enhancer in the mouse germline alters transcription factor binding to the remaining Il9 regulatory elements, and results in diminished IL-9 production in multiple cell types including Th9 cells, and attenuates IL-9-dependent immune responses. Moreover, deletion of the homologous enhancer (CNS-18) in primary human Th9 cultures results in significant decrease of IL-9 production. Thus, Il9 CNS-25/IL9 CNS-18 is a critical and conserved regulatory element for IL-9 production. Interleukin-9 (IL-9) is important for allergy, autoimmunity and tumor immunity, but how its expression is regulated is unclear. Here the authors show the essential function of an enhancer, CNS-25 in mouse and CNS-18 in human, for IL-9 expression, with the deletion of this enhancer severely hampering IL-9 production in mice or human cells.
Collapse
|
38
|
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: 14] [Impact Index Per Article: 2.3] [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
|
39
|
Yoshimura A, Ito M, Chikuma S, Akanuma T, Nakatsukasa H. Negative Regulation of Cytokine Signaling in Immunity. Cold Spring Harb Perspect Biol 2018; 10:a028571. [PMID: 28716890 PMCID: PMC6028070 DOI: 10.1101/cshperspect.a028571] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are key modulators of immunity. Most cytokines use the Janus kinase and signal transducers and activators of transcription (JAK-STAT) pathway to promote gene transcriptional regulation, but their signals must be attenuated by multiple mechanisms. These include the suppressors of cytokine signaling (SOCS) family of proteins, which represent a main negative regulation mechanism for the JAK-STAT pathway. Cytokine-inducible Src homology 2 (SH2)-containing protein (CIS), SOCS1, and SOCS3 proteins regulate cytokine signals that control the polarization of CD4+ T cells and the maturation of CD8+ T cells. SOCS proteins also regulate innate immune cells and are involved in tumorigenesis. This review summarizes recent progress on CIS, SOCS1, and SOCS3 in T cells and tumor immunity.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takashi Akanuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
40
|
Shohan M, Sabzevary-Ghahfarokhi M, Bagheri N, Shirzad H, Rahimian G, Soltani A, Ghatreh-Samani M, Deris F, Tahmasbi K, Shahverdi E, Fathollahi F. Intensified Th9 Response is Associated with the Immunopathogenesis of Active Ulcerative Colitis. Immunol Invest 2018; 47:700-711. [DOI: 10.1080/08820139.2018.1486411] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Mojtaba Shohan
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Milad Sabzevary-Ghahfarokhi
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ghorbanali Rahimian
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amin Soltani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatreh-Samani
- Department of Microbiology and Immunology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Deris
- Department of Epidemiology and Biostatistics, School of Health, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Kamran Tahmasbi
- Department of Pathology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elahe Shahverdi
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fereshteh Fathollahi
- Department of Internal Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
41
|
Someya K, Nakatsukasa H, Ito M, Kondo T, Tateda KI, Akanuma T, Koya I, Sanosaka T, Kohyama J, Tsukada YI, Takamura-Enya T, Yoshimura A. Improvement of Foxp3 stability through CNS2 demethylation by TET enzyme induction and activation. Int Immunol 2018; 29:365-375. [PMID: 29048538 PMCID: PMC5890887 DOI: 10.1093/intimm/dxx049] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/05/2017] [Indexed: 12/12/2022] Open
Abstract
Since induced regulatory T cells (iTregs) can be produced in a large quantity in vitro, these cells are expected to be clinically useful to induce immunological tolerance in various immunological diseases. Foxp3 (Forkhead box P3) expression in iTregs is, however, unstable due to the lack of demethylation of the CpG island in the conserved non-coding sequence 2 (CNS2) of the Foxp3 locus. To facilitate the demethylation of CNS2, we over-expressed the catalytic domain (CD) of the ten-eleven translocation (TET) protein, which catalyzes the steps of the iterative demethylation of 5-methylcytosine. TET-CD over-expression in iTregs resulted in partial demethylation of CNS2 and stable Foxp3 expression. We also discovered that TET expression was enhanced under low oxygen (5%) culture conditions, which facilitated CNS2 DNA demethylation and stabilization of Foxp3 expression in a TET2- and TET3-dependent manner. In combination with vitamin C treatment, which has been reported to enhance TET catalytic activity, iTregs generated under low oxygen conditions retained more stable Foxp3 expression in vitro and in vivo and exhibited stronger suppression activity in a colitis model compared with untreated iTregs. Our data indicate that the induction and activation of TET enzymes in iTregs would be an effective method for Treg-mediated adoptive immunotherapy.
Collapse
Affiliation(s)
| | | | - Minako Ito
- Department of Microbiology and Immunology
| | | | | | | | - Ikuko Koya
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tsukasa Sanosaka
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Jun Kohyama
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yu-Ichi Tsukada
- Advanced Biological Information Research Division, Inamori Frontier Research Center, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Takeji Takamura-Enya
- Department of Applied Chemistry, Kanagawa Institute of Technology, Shimo-Ogino 1030, Atsugi-shi 243-0292, Japan
| | | |
Collapse
|
42
|
Malik S, Awasthi A. Transcriptional Control of Th9 Cells: Role of Foxo1 in Interleukin-9 Induction. Front Immunol 2018; 9:995. [PMID: 29867972 PMCID: PMC5954031 DOI: 10.3389/fimmu.2018.00995] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL) 9-producing helper T (Th) 9 cells play a major role in contributing immunity against extracellular pathogens. In addition, the role of Th9 cells was demonstrated in the pathogenesis of allergic, skin, and intestinal inflammation. The functions of Th9 cells were further extended in antitumor immune response, as Th9 cells were suggested to be potent antitumor Th cells. Given the pleotropic functions of IL-9 in various pathophysiological conditions, it is essential to understand the differentiation and stability of Th9 cells and other IL-9-producing T cells. In addition to Th9 cells, Th2 and Th17 cells as well as induced Foxp3+ regulatory T cells (iTregs) cells also produce IL-9, but how IL-9 production is regulated in these cell types is not yet clearly defined. Although Th2, Th9 and Th17 cells as well as iTregs develop in the presence of distinct differentiating factors, yet they all express IL-9 together with their own lineage specific cytokines. Here, in this review, we summarize the current understanding of signaling pathways that lead to the promotion of differentiation of Th9 cells and IL-9 induction in Th2 and Th17 cells, as well as in iTregs. We further discuss the transcriptional regulation of Th9 cells in context of Foxo1, as an essential transcription factor required for the development and functions of Th9 and other IL-9-producing T cells.
Collapse
Affiliation(s)
| | - Amit Awasthi
- Immuno-Biology Laboratory, Center for Human Microbial Ecology, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
43
|
Shohan M, Elahi S, Shirzad H, Rafieian-Kopaei M, Bagheri N, Soltani E. Th9 Cells: Probable players in ulcerative colitis pathogenesis. Int Rev Immunol 2018; 37:192-205. [PMID: 29672174 DOI: 10.1080/08830185.2018.1457659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T lymphocytes represent an important part of adaptive immune system undertaking different functions to regulate immune responses. CD4+ T cells are the most important activator cells in inflammatory conditions. Depending on the type of induced cells and inflamed sites, expression and activity of different subtypes of helper T cells are changed. Recent studies have confirmed the existence of a new subset of helper T lymphocytes called Th9. Naive T cells can differentiate into Th9 subtypes if they are exposed simultaneously by interleukin (IL) 4 and transforming growth factor β and also secondary activation of a complicated network of transcription factors such as interferon regulatory factor 4 (IRF4) and Smads which are essential for adequate induction of this phenotype. Th9 cells specifically produce interleukin 9 and their probable roles in promoting intestinal inflammation are being investigated in human subjects and experimental models of ulcerative colitis (UC). Recently, infiltration of Th9 cells, overexpression of IL-9, and certain genes associated with Th9 differentiation have been demonstrated in inflammatory microenvironment of UC. Intestinal oversecretion of IL-9 protein is likely to break down epithelial barriers and compromise tolerance to certain commensal microorganisms which leads to inflammation. Th9 pathogenicity has not yet been adequately explored in UC and they are far from being considered as inflammatory cells in this milieu, therefore precise understanding the role of these newly identified cells in particular their potential role in gut pathogenesis may enable us to develop novel therapeutic approaches for inflammatory bowel disease. So, this article tries to discuss the latest knowledge on the above-mentioned field.
Collapse
Affiliation(s)
- Mojtaba Shohan
- a Department of Microbiology and Immunology , Faculty of Medicine, Shahrekord University of Medical Sciences , Shahrekord , Iran
| | - Shokrollah Elahi
- b Department of Dentistry , Department of Medical Microbiology and Immunology , Faculty of Medicine and Dentistry, University of Alberta , Edmonton , Alberta , Canada
| | - Hedayatollah Shirzad
- c Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences , Shahrekord , Iran
| | - Mahmoud Rafieian-Kopaei
- d Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences , Shahrekord , Iran
| | - Nader Bagheri
- a Department of Microbiology and Immunology , Faculty of Medicine, Shahrekord University of Medical Sciences , Shahrekord , Iran
| | - Emad Soltani
- a Department of Microbiology and Immunology , Faculty of Medicine, Shahrekord University of Medical Sciences , Shahrekord , Iran
| |
Collapse
|
44
|
Pang N, Zhang F, Li S, Zhu Y, Zhang C, An M, Wang H, Mamuti W, Ding J, Fan H. TGF-β/Smad signaling pathway positively up-regulates the differentiation of Interleukin-9-producing CD4 + T cells in human Echinococcus granulosus infection. J Infect 2018; 76:406-416. [DOI: 10.1016/j.jinf.2018.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/15/2017] [Accepted: 01/07/2018] [Indexed: 12/17/2022]
|
45
|
Cherian MA, Olson S, Sundaramoorthi H, Cates K, Cheng X, Harding J, Martens A, Challen GA, Tyagi M, Ratner L, Rauch D. An activating mutation of interferon regulatory factor 4 (IRF4) in adult T-cell leukemia. J Biol Chem 2018. [PMID: 29540473 DOI: 10.1074/jbc.ra117.000164] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The human T-cell leukemia virus-1 (HTLV-1) oncoprotein Tax drives cell proliferation and resistance to apoptosis early in the pathogenesis of adult T-cell leukemia (ATL). Subsequently, probably as a result of specific immunoediting, Tax expression is down-regulated and functionally replaced by somatic driver mutations of the host genome. Both amplification and point mutations of interferon regulatory factor 4 (IRF4) have been previously detected in ATL., K59R is the most common single-nucleotide variation of IRF4 and is found exclusively in ATL. High-throughput whole-exome sequencing revealed recurrent activating genetic alterations in the T-cell receptor, CD28, and NF-κB pathways. We found that IRF4, which is transcriptionally activated downstream of these pathways, is frequently mutated in ATL. IRF4 RNA, protein, and IRF4 transcriptional targets are uniformly elevated in HTLV-1-transformed cells and ATL cell lines, and IRF4 was bound to genomic regulatory DNA of many of these transcriptional targets in HTLV-1-transformed cell lines. We further noted that the K59R IRF4 mutant is expressed at higher levels in the nucleus than WT IRF4 and is transcriptionally more active. Expression of both WT and the K59R mutant of IRF4 from a constitutive promoter in retrovirally transduced murine bone marrow cells increased the abundance of T lymphocytes but not myeloid cells or B lymphocytes in mice. IRF4 may represent a therapeutic target in ATL because ATL cells select for a mutant of IRF4 with higher nuclear expression and transcriptional activity, and overexpression of IRF4 induces the expansion of T lymphocytes in vivo.
Collapse
Affiliation(s)
- Mathew A Cherian
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Sydney Olson
- the Department of Biology, University of Wisconsin, Madison, Wisconsin 53706, and
| | - Hemalatha Sundaramoorthi
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kitra Cates
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xiaogang Cheng
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Harding
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Andrew Martens
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Grant A Challen
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Manoj Tyagi
- the Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland 20892
| | - Lee Ratner
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110,
| | - Daniel Rauch
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
46
|
Li Y, Yu Q, Zhang Z, Wang J, Li S, Zhang J, Liu G. TH9 cell differentiation, transcriptional control and function in inflammation, autoimmune diseases and cancer. Oncotarget 2018; 7:71001-71012. [PMID: 27589682 PMCID: PMC5342605 DOI: 10.18632/oncotarget.11681] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/26/2016] [Indexed: 12/31/2022] Open
Abstract
Naïve CD4+T cells differentiate into various T cell subsets depending on the specific cytokine environment. TH9 cells are less well-characterized than other T cell subsets, and factors that control their development and function have only recently been identified. It is now clear that TH9 cells play critical roles in immune-mediated diseases, including allergic airway, autoimmune and inflammatory bowel diseases, and cancer. Thus, the promotion or suppression of TH9 cell differentiation, transcriptional control and function may provide novel treatments for clinical inflammation, autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Yan Li
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Yu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Zhengguo Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jian Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Simin Li
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jiangyuan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China.,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Buttrick TS, Wang W, Yung C, Trieu KG, Patel K, Khoury SJ, Ai X, Elyaman W. Foxo1 Promotes Th9 Cell Differentiation and Airway Allergy. Sci Rep 2018; 8:818. [PMID: 29339772 PMCID: PMC5770389 DOI: 10.1038/s41598-018-19315-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/21/2017] [Indexed: 12/31/2022] Open
Abstract
T helper 9 (Th9) cells are effector CD4+ T cells that are characterized by the production of interleukin-9 (IL-9) and have been associated with allergic responses. Here, we found that the expression of the transcription factor forkhead box O1 (Foxo1) was induced in Th9 and Foxo1 plays a crucial role in the differentiation of Th9 cells. Pharmacological inhibition of Foxo1 or genetic disruption of Foxo1 in CD4+ T cells caused a reduction in IL-9 expression while upregulating IL-17A and IFNγ production. Furthermore, chromatin immunoprecipitation (ChIP) followed by luciferase assays revealed direct binding of Foxo1 to both the Il9 and Irf4 promoters and induces their transactivation. Lastly, adoptive transfer of Th9 cells into lungs induced asthma-like symptoms that were ameliorated by Foxo1 inhibitor, AS1842856. Together, our findings demonstrate a novel regulator of Th9 cells with a direct implication in allergic inflammation.
Collapse
Affiliation(s)
- Thomas S Buttrick
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Wang
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Christina Yung
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kenneth G Trieu
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kruti Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Xingbin Ai
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Wassim Elyaman
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
48
|
IRF8-dependent molecular complexes control the Th9 transcriptional program. Nat Commun 2017; 8:2085. [PMID: 29233972 PMCID: PMC5727025 DOI: 10.1038/s41467-017-01070-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 08/16/2017] [Indexed: 12/31/2022] Open
Abstract
Interferon regulatory factors (IRF) have critical functions in lymphoid development and in immune response regulation. Although many studies have described the function of IRF4 in CD4+ T cells, few have focused on the IRF4 homologue, IRF8. Here, we show that IRF8 is required for Th9 differentiation in vitro and in vivo. IRF8 functions through a transcription factor complex consisting of IRF8, IRF4, PU.1 and BATF, which binds to DNA and boosts Il9 transcription. By contrast, IRF8 deficiency promotes the expression of other genes such as Il4, as IRF8 dimerises with the transcriptional repressor ETV6 and inhibits Il4 expression. In vivo, IRF8 is essential for the anti-tumour effects of Th9 cells in mouse melanoma models. Our results show that IRF8 complexes boost the Th9 program and repress Il4 expression to modulate Th9 cell differentiation, thereby implicating IRF8 as a potential therapeutic target to affect Th9 responses in cancer therapy. Interferon regulatory factors IRF regulate lymphoid development, but the specific function of IRF8 in helper T-cell polarization is unclear. Here the authors show that IRF8 forms a complex with IRF4, PU.1 and BATF to modulate the Th9 transcription program and expression of IL-4 and IL-9.
Collapse
|
49
|
Ramadan A, Griesenauer B, Adom D, Kapur R, Hanenberg H, Liu C, Kaplan MH, Paczesny S. Specifically differentiated T cell subset promotes tumor immunity over fatal immunity. J Exp Med 2017; 214:3577-3596. [PMID: 29038366 PMCID: PMC5716032 DOI: 10.1084/jem.20170041] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 07/31/2017] [Accepted: 09/08/2017] [Indexed: 01/10/2023] Open
Abstract
Allogeneic immune cells, particularly T cells in donor grafts, recognize and eliminate leukemic cells via graft-versus-leukemia (GVL) reactivity, and transfer of these cells is often used for high-risk hematological malignancies, including acute myeloid leukemia. Unfortunately, these cells also attack host normal tissues through the often fatal graft-versus-host disease (GVHD). Full separation of GVL activity from GVHD has yet to be achieved. Here, we show that, in mice and humans, a population of interleukin-9 (IL-9)-producing T cells activated via the ST2-IL-33 pathway (T9IL-33 cells) increases GVL while decreasing GVHD through two opposing mechanisms: protection from fatal immunity by amphiregulin expression and augmentation of antileukemic activity compared with T9, T1, and unmanipulated T cells through CD8α expression. Thus, adoptive transfer of allogeneic T9IL-33 cells offers an attractive approach for separating GVL activity from GVHD.
Collapse
Affiliation(s)
| | | | | | - Reuben Kapur
- Indiana University School of Medicine, Indianapolis, IN
| | | | - Chen Liu
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Mark H Kaplan
- Indiana University School of Medicine, Indianapolis, IN
| | | |
Collapse
|
50
|
Kasahara H, Kondo T, Nakatsukasa H, Chikuma S, Ito M, Ando M, Kurebayashi Y, Sekiya T, Yamada T, Okamoto S, Yoshimura A. Generation of allo-antigen-specific induced Treg stabilized by vitamin C treatment and its application for prevention of acute graft versus host disease model. Int Immunol 2017; 29:457-469. [DOI: 10.1093/intimm/dxx060] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/28/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Hidenori Kasahara
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Taisuke Kondo
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Takashi Sekiya
- Department of Immune Regulation, Research Institute, National Center for Global Health and Medicine, Japan
| | - Taketo Yamada
- Department of Pathology, Saitama Medical University, Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|