1
|
Fischer MA, Jia L, Edelblum KL. Type I IFN Induces TCR-dependent and -independent Antimicrobial Responses in γδ Intraepithelial Lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1380-1391. [PMID: 39311642 PMCID: PMC11493514 DOI: 10.4049/jimmunol.2400138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
Intraepithelial lymphocytes (IELs) expressing the TCRγδ survey the intestinal epithelium to limit the invasion of microbial pathogens. The production of type I IFN is a central component of an antiviral immune response, yet how these proinflammatory cytokines contribute to γδ IEL effector function remains unclear. Based on the unique activation status of IELs and their ability to bridge innate and adaptive immunity, we investigated the extent to which type I IFN signaling modulates γδ IEL function. Using an ex vivo culture model, we find that type I IFN alone is unable to drive IFN-γ production, yet low-level TCR activation synergizes with type I IFN to induce IFN-γ production in murine γδ IELs. Further investigation into the underlying molecular mechanisms of costimulation revealed that TCRγδ-mediated activation of NFAT and JNK is required for type I IFN to promote IFN-γ expression in a STAT4-dependent manner. Whereas type I IFN rapidly upregulates antiviral gene expression independent of a basal TCRγδ signal, neither tonic TCR triggering nor the presence of a TCR agonist was sufficient to elicit type I IFN-induced IFN-γ production in vivo. However, bypassing proximal TCR signaling events synergized with IFNAR/STAT4 activation to induce γδ IEL IFN-γ production. These findings indicate that γδ IELs contribute to host defense in response to type I IFN by mounting a rapid antimicrobial response independent of TCRγδ signaling, and may produce IFN-γ in a TCR-dependent manner under permissive conditions.
Collapse
Affiliation(s)
- Matthew A Fischer
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Luo Jia
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Karen L Edelblum
- Center for Immunity and Inflammation, Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
2
|
Fischer MA, Jia L, Edelblum KL. Type I interferon induces TCR-dependent and -independent antimicrobial responses in γδ intraepithelial lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584444. [PMID: 38559228 PMCID: PMC10979951 DOI: 10.1101/2024.03.11.584444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Intraepithelial lymphocytes (IEL) expressing the γδ T cell receptor (TCR) survey the intestinal epithelium to limit the invasion of microbial pathogens. The production of type I interferon (IFN) is a central component of an antiviral immune response, yet how these pro-inflammatory cytokines contribute to γδ IEL effector function remains unclear. Based on the unique activation status of IELs, and their ability to bridge innate and adaptive immunity, we investigated the extent to which type I IFN signaling modulates γδ IEL function. Using an ex vivo culture model, we find that type I IFN alone is unable to drive IFNγ production, yet low level TCR activation synergizes with type I IFN to induce IFNγ production in murine γδ IELs. Further investigation into the underlying molecular mechanisms of co-stimulation revealed that TCRγδ-mediated activation of NFAT and JNK is required for type I IFN to promote IFNγ expression in a STAT4- dependent manner. Whereas type I IFN rapidly upregulates antiviral gene expression independent of a basal TCRγδ signal, neither tonic TCR triggering nor the presence of a TCR agonist was sufficient to elicit type I IFN-induced IFNγ production in vivo . However, bypassing proximal TCR signaling events synergized with IFNAR/STAT4 activation to induce γδ IEL IFNγ production. These findings indicate that γδ IELs contribute to host defense in response to type I IFN by mounting a rapid antimicrobial response independent of TCRγδ signaling, and under permissive conditions, produce IFNγ in a TCR-dependent manner.
Collapse
|
3
|
Belizaire R, Wong WJ, Robinette ML, Ebert BL. Clonal haematopoiesis and dysregulation of the immune system. Nat Rev Immunol 2023; 23:595-610. [PMID: 36941354 PMCID: PMC11140722 DOI: 10.1038/s41577-023-00843-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/23/2023]
Abstract
Age-related diseases are frequently linked to pathological immune dysfunction, including excessive inflammation, autoreactivity and immunodeficiency. Recent analyses of human genetic data have revealed that somatic mutations and mosaic chromosomal alterations in blood cells - a condition known as clonal haematopoiesis (CH) - are associated with ageing and pathological immune dysfunction. Indeed, large-scale epidemiological studies and experimental mouse models have demonstrated that CH can promote cardiovascular disease, chronic obstructive pulmonary disease, chronic liver disease, osteoporosis and gout. The genes most frequently mutated in CH, the epigenetic regulators TET2 and DNMT3A, implicate increased chemokine expression and inflammasome hyperactivation in myeloid cells as a possible mechanistic connection between CH and age-related diseases. In addition, TET2 and DNMT3A mutations in lymphoid cells have been shown to drive methylation-dependent alterations in differentiation and function. Here we review the observational and mechanistic studies describing the connection between CH and pathological immune dysfunction, the effects of CH-associated genetic alterations on the function of myeloid and lymphoid cells, and the clinical and therapeutic implications of CH as a target for immunomodulation.
Collapse
Affiliation(s)
- Roger Belizaire
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Waihay J Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle L Robinette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
4
|
Gauthier T, Chen W. IFN-γ and TGF-β, Crucial Players in Immune Responses: A Tribute to Howard Young. J Interferon Cytokine Res 2022; 42:643-654. [PMID: 36516375 PMCID: PMC9917322 DOI: 10.1089/jir.2022.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/18/2022] [Indexed: 12/15/2022] Open
Abstract
Interferon gamma (IFN-γ) and transforming growth factor beta (TGF-β), both pleiotropic cytokines, have been long studied and described as critical mediators of the immune response, notably in T cells. One of the investigators who made seminal and critical discoveries in the field of IFN-γ biology is Dr. Howard Young. In this review, we provide an overview of the biology of IFN-γ as well as its role in cancer and autoimmunity with an emphasis on Dr. Young's critical work in the field. We also describe how Dr. Young's work influenced our own research studying the role of TGF-β in the modulation of immune responses.
Collapse
Affiliation(s)
- Thierry Gauthier
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Marrero MG, Field SL, Skibiel AL, Dado-Senn B, Driver JP, Laporta J. Increasing serotonin bioavailability alters gene expression in peripheral leukocytes and lymphoid tissues of dairy calves. Sci Rep 2020; 10:9712. [PMID: 32546841 PMCID: PMC7297988 DOI: 10.1038/s41598-020-66326-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
Dairy calves are born with a naïve immune system, making the pre-weaning phase a critical window for immune development. In the U.S., 40-60% of dairy farms feed milk replacer to pre-weaned calves, which are devoid of bioactive factors with immunological roles. Serotonin is a bioactive factor with immunoregulatory properties naturally produced by the calf and present in milk. Human and rodent immune cells express the serotonin machinery, but little is known about the role of serotonin in the bovine immune system. Supplementing milk replacer with 5-hydroxytryptophan (serotonin precursor) or fluoxetine (reuptake inhibitor) increases serotonin bioavailability. We hypothesized that increased serotonin bioavailability promotes serotonergic signaling and modulates the expression of immune related genes in peripheral leukocytes and immune-related tissues of dairy calves. The present experiment targeted candidate genes involved in serotonin production, metabolism, transport, signaling and immune regulation. We established that bovine peripheral leukocytes express all known serotonin receptors, and can synthesize, uptake and degrade serotonin due to the expression of serotonin metabolism-related genes. Indeed, we showed that increasing serotonin bioavailability alters gene expression of serotonin receptors and immune-related genes. Further research will determine whether manipulation of the serotonin pathway could be a feasible approach to bolster dairy calves' immune system.
Collapse
Affiliation(s)
- M G Marrero
- Department of Animal Sciences, University of Florida, Florida, USA
| | - S L Field
- Department of Animal Sciences, University of Florida, Florida, USA
| | - A L Skibiel
- Department of Animal Sciences, University of Florida, Florida, USA
- Department of Animal and Veterinary Science, University of Idaho, Idaho, USA
| | - B Dado-Senn
- Department of Animal Sciences, University of Florida, Florida, USA
| | - J P Driver
- Department of Animal Sciences, University of Florida, Florida, USA
| | - J Laporta
- Department of Animal Sciences, University of Florida, Florida, USA.
| |
Collapse
|
6
|
Kulling PM, Olson KC, Hamele CE, Toro MF, Tan SF, Feith DJ, Loughran TP. Dysregulation of the IFN-γ-STAT1 signaling pathway in a cell line model of large granular lymphocyte leukemia. PLoS One 2018; 13:e0193429. [PMID: 29474442 PMCID: PMC5825082 DOI: 10.1371/journal.pone.0193429] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/09/2018] [Indexed: 02/04/2023] Open
Abstract
T cell large granular lymphocyte leukemia (T-LGLL) is a rare incurable disease that is characterized by defective apoptosis of cytotoxic CD8+ T cells. Chronic activation of the Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) pathway is a hallmark of T-LGLL. One manifestation is the constitutive phosphorylation of tyrosine 701 of STAT1 (p-STAT1). T-LGLL patients also exhibit elevated serum levels of the STAT1 activator, interferon-γ (IFN-γ), thus contributing to an inflammatory environment. In normal cells, IFN-γ production is tightly controlled through induction of IFN-γ negative regulators. However, in T-LGLL, IFN-γ signaling lacks this negative feedback mechanism as evidenced by excessive IFN-γ production and decreased levels of suppressors of cytokine signaling 1 (SOCS1), a negative regulator of IFN-γ. Here we characterize the IFN-γ-STAT1 pathway in TL-1 cells, a cell line model of T-LGLL. TL-1 cells exhibited lower IFN-γ receptor protein and mRNA expression compared to an IFN-γ responsive cell line. Furthermore, IFN-γ treatment did not induce JAK2 or STAT1 activation or transcription of IFN-γ-inducible gene targets. However, IFN-β induced p-STAT1 and subsequent STAT1 gene transcription, demonstrating a specific IFN-γ signaling defect in TL-1 cells. We utilized siRNA targeting of STAT1, STAT3, and STAT5b to probe their role in IL-2-mediated IFN-γ regulation. These studies identified STAT5b as a positive regulator of IFN-γ production. We also characterized the relationship between STAT1, STAT3, and STAT5b proteins. Surprisingly, p-STAT1 was positively correlated with STAT3 levels while STAT5b suppressed the activation of both STAT1 and STAT3. Taken together, these results suggest that the dysregulation of the IFN-γ-STAT1 signaling pathway in TL-1 cells likely results from low levels of the IFN-γ receptor. The resulting inability to induce negative feedback regulators explains the observed elevated IL-2 driven IFN-γ production. Future work will elucidate the best way to target this pathway, with the ultimate goal to find a better therapeutic for T-LGLL.
Collapse
Affiliation(s)
- Paige M. Kulling
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
- Department of Pathology, University of Virginia; Charlottesville, VA United States of America
| | - Kristine C. Olson
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Cait E. Hamele
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Mariella F. Toro
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - David J. Feith
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
| | - Thomas P. Loughran
- University of Virginia Cancer Center, University of Virginia; Charlottesville, VA United States of America
- Department of Medicine, Division of Hematology/Oncology, University of Virginia; Charlottesville, VA United States of America
- * E-mail:
| |
Collapse
|
7
|
Sadreev II, Chen MZQ, Umezawa Y, Biktashev VN, Kemper C, Salakhieva DV, Welsh GI, Kotov NV. The competitive nature of signal transducer and activator of transcription complex formation drives phenotype switching of T cells. Immunology 2017; 153:488-501. [PMID: 29030870 DOI: 10.1111/imm.12851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/16/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022] Open
Abstract
Signal transducers and activators of transcription (STATs) are key molecular determinants of T-cell fate and effector function. Several inflammatory diseases are characterized by an altered balance of T-cell phenotypes and cytokine secretion. STATs, therefore, represent viable therapeutic targets in numerous pathologies. However, the underlying mechanisms by which the same STAT proteins regulate both the development of different T-cell phenotypes and their plasticity during changes in extracellular conditions remain unclear. In this study, we investigated the STAT-mediated regulation of T-cell phenotype formation and plasticity using mathematical modelling and experimental data for intracellular STAT signalling proteins. The close fit of our model predictions to the experimental data allows us to propose a potential mechanism for T-cell switching. According to this mechanism, T-cell phenotype switching is the result of the relative redistribution of STAT dimer complexes caused by the extracellular cytokine-dependent STAT competition effects. The developed model predicts that the balance between the intracellular STAT species defines the amount of the produced cytokines and thereby T-cell phenotypes. The model predictions are consistent with the experimentally observed interferon-γ to interleukin-10 switching that regulates human T helper type 1/type 1 regulatory T-cell responses. The proposed model is applicable to a number of STAT signalling circuits.
Collapse
Affiliation(s)
- Ildar I Sadreev
- Centre for Systems, Dynamics and Control, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Michael Z Q Chen
- School of Automation, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Yoshinori Umezawa
- Department of Dermatology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Vadim N Biktashev
- Centre for Systems, Dynamics and Control, College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, USA.,Division of Transplant Immunology and Mucosal Biology, King's College London, London, UK.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Diana V Salakhieva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Gavin I Welsh
- Bristol Renal, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Nikolay V Kotov
- Biophysics & Bionics Laboratory, Institute of Physics, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
8
|
Ni J, Hölsken O, Miller M, Hammer Q, Luetke-Eversloh M, Romagnani C, Cerwenka A. Adoptively transferred natural killer cells maintain long-term antitumor activity by epigenetic imprinting and CD4 + T cell help. Oncoimmunology 2016; 5:e1219009. [PMID: 27757318 DOI: 10.1080/2162402x.2016.1219009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/25/2016] [Accepted: 07/25/2016] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cell infusions can induce remissions in subsets of patients with different types of cancer. The optimal strategies for NK cell activation prior to infusion are still under debate. There is recent evidence that NK cells can acquire long-term functional competence by preactivation with the cytokines IL-12/15/18. The mechanisms supporting the maintenance of long-term NK cell antitumor activity are incompletely under-stood. Here, we show that NK cells preactivated in vitro with IL-12/15/18, but not with IL-15 alone, maintained high antitumor activity even 1 mo after transfer into lymphopenic RAG-2-/-γc-/- mice. The NK cell intrinsic ability for IFNγ production coincided with demethylation of the conserved non-coding sequence (CNS) 1 in the Ifng locus, previously shown to enhance transcription of Ifng. In a xenograft melanoma mouse model, human IL-12/15/18-preactivated NK cells rejected tumors more efficiently. In RAG-2-/-γc-/- mice, co-transfer of CD4+ T cells further improved the long-term competence of NK cells for IFNγ production that was dependent on IL-2. CD4+ T cell activation during homeostatic proliferation required macrophages and further promoted the long-term NK cell antitumor activity. Thus, NK cells can "remember" a previous exposure to cytokines by epigenetic imprinting resulting in a remarkable stability of the IFNγ-producing phenotype after adoptive transfer. In addition, our results support combination of cytokine-preactivated NK cells with CD4+ T cell activation upon lymphopenic conditioning to achieve long-term NK cell effector function for cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Ni
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Oliver Hölsken
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Matthias Miller
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| | - Quirin Hammer
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Merlin Luetke-Eversloh
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum - A Leibniz Institute , Berlin, Germany
| | - Adelheid Cerwenka
- German Cancer Research Center (DKFZ), Research Group Innate Immunity , Heidelberg, Germany
| |
Collapse
|
9
|
Tsurutani N, Mittal P, St Rose MC, Ngoi SM, Svedova J, Menoret A, Treadway FB, Laubenbacher R, Suárez-Ramírez JE, Cauley LS, Adler AJ, Vella AT. Costimulation Endows Immunotherapeutic CD8 T Cells with IL-36 Responsiveness during Aerobic Glycolysis. THE JOURNAL OF IMMUNOLOGY 2015; 196:124-34. [PMID: 26573834 DOI: 10.4049/jimmunol.1501217] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 10/17/2015] [Indexed: 01/07/2023]
Abstract
CD134- and CD137-primed CD8 T cells mount powerful effector responses upon recall, but even without recall these dual-costimulated T cells respond to signal 3 cytokines such as IL-12. We searched for alternative signal 3 receptor pathways and found the IL-1 family member IL-36R. Although IL-36 alone did not stimulate effector CD8 T cells, in combination with IL-12, or more surprisingly IL-2, it induced striking and rapid TCR-independent IFN-γ synthesis. To understand how signal 3 responses functioned in dual-costimulated T cells we showed that IL-2 induced IL-36R gene expression in a JAK/STAT-dependent manner. These data help delineate a sequential stimulation process where IL-2 conditioning must precede IL-36 for IFN-γ synthesis. Importantly, this responsive state was transient and functioned only in effector T cells capable of aerobic glycolysis. Specifically, as the effector T cells metabolized glucose and consumed O2, they also retained potential to respond through IL-36R. This suggests that T cells use innate receptor pathways such as the IL-36R/axis when programmed for aerobic glycolysis. To explore a function for IL-36R in vivo, we showed that dual costimulation therapy reduced B16 melanoma tumor growth while increasing IL-36R gene expression. In summary, cytokine therapy to eliminate tumors may target effector T cells, even outside of TCR specificity, as long as the effectors are in the correct metabolic state.
Collapse
Affiliation(s)
- Naomi Tsurutani
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Payal Mittal
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Soo Mun Ngoi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Julia Svedova
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Antoine Menoret
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Forrest B Treadway
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Reinhard Laubenbacher
- Center for Quantitative Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Jenny E Suárez-Ramírez
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Linda S Cauley
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Adam J Adler
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| | - Anthony T Vella
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030; and
| |
Collapse
|
10
|
Min X, Ungureanu D, Maxwell S, Hammarén H, Thibault S, Hillert EK, Ayres M, Greenfield B, Eksterowicz J, Gabel C, Walker N, Silvennoinen O, Wang Z. Structural and Functional Characterization of the JH2 Pseudokinase Domain of JAK Family Tyrosine Kinase 2 (TYK2). J Biol Chem 2015; 290:27261-27270. [PMID: 26359499 DOI: 10.1074/jbc.m115.672048] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 11/06/2022] Open
Abstract
JAK (Janus family of cytoplasmic tyrosine kinases) family tyrosine kinase 2 (TYK2) participates in signaling through cytokine receptors involved in immune responses and inflammation. JAKs are characterized by dual kinase domain: a tyrosine kinase domain (JH1) that is preceded by a pseudokinase domain (JH2). The majority of disease-associated mutations in JAKs map to JH2, demonstrating its central regulatory function. JH2s were considered catalytically inactive, but JAK2 JH2 was found to have low autoregulatory catalytic activity. Whether the other JAK JH2s share ATP binding and enzymatic activity has been unclear. Here we report the crystal structure of TYK2 JH2 in complex with adenosine 5'-O-(thiotriphosphate) (ATP-γS) and characterize its nucleotide binding by biochemical and biophysical methods. TYK2 JH2 did not show phosphotransfer activity, but it binds ATP and the nucleotide binding stabilizes the protein without inducing major conformational changes. Mutation of the JH2 ATP-binding pocket increased basal TYK2 phosphorylation and downstream signaling. The overall structural characteristics of TYK2 JH2 resemble JAK2 JH2, but distinct stabilizing molecular interactions around helix αAL in the activation loop provide a structural basis for differences in substrate access and catalytic activities among JAK family JH2s. The structural and biochemical data suggest that ATP binding is functionally important for both TYK2 and JAK2 JH2s, whereas the regulatory phosphorylation appears to be a unique property of JAK2. Finally, the co-crystal structure of TYK2 JH2 complexed with a small molecule inhibitor demonstrates that JH2 is accessible to ATP-competitive compounds, which offers novel approaches for targeting cytokine signaling as well as potential therapeutic applications.
Collapse
Affiliation(s)
- Xiaoshan Min
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080
| | - Daniela Ungureanu
- the Institute of Biomedical Technology, University of Tampere, 33014 Tampere, Finland
| | - Sarah Maxwell
- Departments of Inflammation, Amgen Inc., South San Francisco, California 94080
| | - Henrik Hammarén
- the School of Medicine, University of Tampere, 33014 Tampere, Finland
| | - Steve Thibault
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080
| | | | - Merrill Ayres
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080
| | - Brad Greenfield
- Departments of Inflammation, Amgen Inc., South San Francisco, California 94080
| | - John Eksterowicz
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080
| | - Chris Gabel
- Departments of Inflammation, Amgen Inc., South San Francisco, California 94080
| | - Nigel Walker
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080
| | - Olli Silvennoinen
- the School of Medicine, University of Tampere, 33014 Tampere, Finland; Department of Clinical Hematology, Tampere University Hospital, 33520 Tampere, Finland.
| | - Zhulun Wang
- Departments of Therapeutic Discovery, Amgen Inc., South San Francisco, California 94080.
| |
Collapse
|
11
|
Mechanism of Action of IL-7 and Its Potential Applications and Limitations in Cancer Immunotherapy. Int J Mol Sci 2015; 16:10267-80. [PMID: 25955647 PMCID: PMC4463645 DOI: 10.3390/ijms160510267] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/16/2015] [Accepted: 04/29/2015] [Indexed: 01/10/2023] Open
Abstract
Interleukin-7 (IL-7) is a non-hematopoietic cell-derived cytokine with a central role in the adaptive immune system. It promotes lymphocyte development in the thymus and maintains survival of naive and memory T cell homeostasis in the periphery. Moreover, it is important for the organogenesis of lymph nodes (LN) and for the maintenance of activated T cells recruited into the secondary lymphoid organs (SLOs). The immune capacity of cancer patients is suppressed that is characterized by lower T cell counts, less effector immune cells infiltration, higher levels of exhausted effector cells and higher levels of immunosuppressive cytokines, such as transforming growth factor β (TGF-β). Recombinant human IL-7 (rhIL-7) is an ideal solution for the immune reconstitution of lymphopenia patients by promoting peripheral T cell expansion. Furthermore, it can antagonize the immunosuppressive network. In animal models, IL-7 has been proven to prolong the survival of tumor-bearing hosts. In this review, we will focus on the mechanism of action and applications of IL-7 in cancer immunotherapy and the potential restrictions for its usage.
Collapse
|
12
|
Fodil N, Langlais D, Moussa P, Boivin GA, Di Pietrantonio T, Radovanovic I, Dumaine A, Blanchette M, Schurr E, Gros P, Vidal SM. Specific dysregulation of IFNγ production by natural killer cells confers susceptibility to viral infection. PLoS Pathog 2014; 10:e1004511. [PMID: 25473962 PMCID: PMC4256466 DOI: 10.1371/journal.ppat.1004511] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 10/09/2014] [Indexed: 12/11/2022] Open
Abstract
Natural Killer (NK) cells contribute to the control of viral infection by directly killing target cells and mediating cytokine release. In C57BL/6 mice, the Ly49H activating NK cell receptor plays a key role in early resistance to mouse cytomegalovirus (MCMV) infection through specific recognition of the MCMV-encoded MHC class I-like molecule m157 expressed on infected cells. Here we show that transgenic expression of Ly49H failed to provide protection against MCMV infection in the naturally susceptible A/J mouse strain. Characterization of Ly49H+ NK cells from Ly49h-A transgenic animals showed that they were able to mount a robust cytotoxic response and proliferate to high numbers during the course of infection. However, compared to NK cells from C57BL/6 mice, we observed an intrinsic defect in their ability to produce IFNγ when challenged by either m157-expressing target cells, exogenous cytokines or chemical stimulants. This effect was limited to NK cells as T cells from C57BL/6 and Ly49h-A mice produced comparable cytokine levels. Using a panel of recombinant congenic strains derived from A/J and C57BL/6 progenitors, we mapped the genetic basis of defective IFNγ production to a single 6.6 Mb genetic interval overlapping the Ifng gene on chromosome 10. Inspection of the genetic interval failed to reveal molecular differences between A/J and several mouse strains showing normal IFNγ production. The chromosome 10 locus is independent of MAPK signalling or decreased mRNA stability and linked to MCMV susceptibility. This study highlights the existence of a previously uncovered NK cell-specific cis-regulatory mechanism of Ifnγ transcript expression potentially relevant to NK cell function in health and disease. Cytomegalovirus (CMV) is a ubiquitous herpesvirus that largely infects the human population leading to a significant cause of disease and death in the immunocompromised and elderly. The study of CMV in animal models has helped understand the pathogenic consequences of CMV infection and adds substantial understanding of the complex interplay of host and virus in living systems. Natural Killer (NK) cells have emerged as an important player during CMV infection trough their specific recognition of viral particles determinants and subsequent secretion of cytokines and cytolytic granules. In the present study, we have generated different mouse models to specifically investigate quantify viral recognition and cytokine expression by NK cells during CMV infection as a measure of NK cell function. We found that even after proper recognition of infected cells by NK cells, the adequate production of IFNγ is crucial to restrain viral infection. Moreover, we demonstrated that IFNγ production by NK cells is genetically determined and directly linked to the IFNγ locus. Hence, we provide the first evidence for of a unique mechanism of IFNγ production by NK cells which regulates susceptibility to viral infection.
Collapse
Affiliation(s)
- Nassima Fodil
- Department of Human Genetics and Department of Microbiology and Immunology, McGill University, Life Sciences Complex, Montreal, Quebec, Canada
- * E-mail: (NF); (SMV)
| | - David Langlais
- Biochemistry Department, McGill University, Montréal, Québec, Canada
| | - Peter Moussa
- Department of Human Genetics and Department of Microbiology and Immunology, McGill University, Life Sciences Complex, Montreal, Quebec, Canada
| | - Gregory Allan Boivin
- Department of Human Genetics and Department of Microbiology and Immunology, McGill University, Life Sciences Complex, Montreal, Quebec, Canada
| | - Tania Di Pietrantonio
- Research Institute of the McGill University Health Centre, McGill Centre for the Study of Host Resistance, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Irena Radovanovic
- Biochemistry Department, McGill University, Montréal, Québec, Canada
| | - Anne Dumaine
- Department of Human Genetics and Department of Microbiology and Immunology, McGill University, Life Sciences Complex, Montreal, Quebec, Canada
| | - Mathieu Blanchette
- McGill Centre for Bioinformatics and School of Computer Science, McGill University, Montréal, Québec, Canada
| | - Erwin Schurr
- Research Institute of the McGill University Health Centre, McGill Centre for the Study of Host Resistance, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Philippe Gros
- Biochemistry Department, McGill University, Montréal, Québec, Canada
| | - Silvia Marina Vidal
- Department of Human Genetics and Department of Microbiology and Immunology, McGill University, Life Sciences Complex, Montreal, Quebec, Canada
- * E-mail: (NF); (SMV)
| |
Collapse
|
13
|
Stritesky GL, Kaplan MH. Changing the STATus quo in T helper cells. Transcription 2014; 2:179-182. [PMID: 21922060 DOI: 10.4161/trns.2.4.16614] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 05/23/2011] [Accepted: 05/25/2011] [Indexed: 11/19/2022] Open
Abstract
STAT proteins are obligate promoters of T helper cell differentiation and initial studies suggested that activation of a single STAT protein resulted in a particular phenotype. More recent work has supported a more complex paradigm wherein the activation of several STAT proteins is required for differentiation to a single effector lineage.
Collapse
Affiliation(s)
- Gretta L Stritesky
- Department of Lab Medicine and Pathology; Center for Immunology; University of Minnesota; Minneapolis, MN USA
| | | |
Collapse
|
14
|
Herr F, Lemoine R, Gouilleux F, Meley D, Kazma I, Heraud A, Velge-Roussel F, Baron C, Lebranchu Y. IL-2 phosphorylates STAT5 to drive IFN-γ production and activation of human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:5660-70. [PMID: 24829413 DOI: 10.4049/jimmunol.1300422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human dendritic cells (hDCs) produce IL-2 and express IL-2R α-chain (CD25), but the role of IL-2 in DC functions is not well defined. A recent study suggested that the main function of CD25 on hDCs was to transpresent IL-2 to activate T lymphocytes. Our results demonstrate the expression of the three chains of the IL-2R on hDCs and that IL-2 induces STAT5 phosphorylation. Interestingly, use of inhibitors of p-STAT5 revealed that IL-2 increases LPS-induced IFN-γ through STAT5 phosphorylation. Finally, we report that IL-2 increases the ability of hDCs to activate helpless CD8(+) T cells, most likely because of IL-2-triggered IFN-γ synthesis, as we previously described. For the first time, to our knowledge, we disclose that IL-2 induces monocyte-derived hDC's functional maturation and activation through IL-2R binding. Interestingly, our study suggests a direct effect of anti-CD25 mAbs on hDCs that may contribute to their clinical efficacy.
Collapse
Affiliation(s)
- Florence Herr
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France;
| | - Roxane Lemoine
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France
| | - Fabrice Gouilleux
- Unité Mixte de Recherche 7292, Centre National de la Recherche Scientifique, 37032 Tours, France; and
| | - Daniel Meley
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France
| | - Ihab Kazma
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France
| | - Audrey Heraud
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France
| | | | - Christophe Baron
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France; Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Yvon Lebranchu
- L'Équipe d'Accueil 4245, Université François Rabelais, 37032 Tours, France; Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| |
Collapse
|
15
|
Abstract
In higher eukaryotic organisms epigenetic modifications are crucial for proper chromatin folding and thereby proper regulation of gene expression. In the last years the involvement of aberrant epigenetic modifications in inflammatory and autoimmune diseases has been recognized and attracted significant interest. However, the epigenetic mechanisms underlying the different disease phenotypes are still poorly understood. As autoimmune and inflammatory diseases are at least partly T cell mediated, we will provide in this chapter an introduction to the epigenetics of T cell differentiation followed by a summary of the current knowledge on aberrant epigenetic modifications that dysfunctional T cells display in various diseases such as type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, and asthma.
Collapse
|
16
|
Chang HC, Lewis D, Tung CY, Han L, Henriquez SMP, Voiles L, Lupov IP, Pelloso D, Sinn AL, Pollok KE, de Lumen BO, Li F, Blum JS, Srivastava S, Robertson MJ. Soypeptide lunasin in cytokine immunotherapy for lymphoma. Cancer Immunol Immunother 2013; 63:283-95. [PMID: 24363024 PMCID: PMC3928510 DOI: 10.1007/s00262-013-1513-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 12/12/2013] [Indexed: 01/19/2023]
Abstract
Immunostimulatory cytokines can enhance anti-tumor immunity and are part of the therapeutic armamentarium for cancer treatment. We have previously reported that post-transplant lymphoma patients have an acquired deficiency of signal transducer and activator of transcription 4, which results in defective IFNγ production during clinical immunotherapy. With the goal of further improving cytokine-based immunotherapy, we examined the effects of a soybean peptide called lunasin that synergistically works with cytokines on natural killer (NK) cells. Peripheral blood mononuclear cells of healthy donors and post-transplant lymphoma patients were stimulated with or without lunasin in the presence of IL-12 or IL-2. NK activation was evaluated, and its tumoricidal activity was assessed using in vitro and in vivo tumor models. Chromatin immunoprecipitation assay was performed to evaluate the histone modification of gene loci that are regulated by lunasin and cytokine. Adding lunasin to IL-12- or IL-2-stimulated NK cells demonstrated synergistic effects in the induction of IFNG and GZMB involved in cytotoxicity. The combination of lunasin and cytokines (IL-12 plus IL-2) was capable of restoring IFNγ production by NK cells from post-transplant lymphoma patients. In addition, NK cells stimulated with lunasin plus cytokines displayed higher tumoricidal activity than those stimulated with cytokines alone using in vitro and in vivo tumor models. The underlying mechanism responsible for the effects of lunasin on NK cells is likely due to epigenetic modulation on target gene loci. Lunasin represents a different class of immune modulating agent that may augment the therapeutic responses mediated by cytokine-based immunotherapy.
Collapse
Affiliation(s)
- Hua-Chen Chang
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, SL310, Indianapolis, IN, 46202, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sadeghi A, Ullenhag G, Wagenius G, Tötterman TH, Eriksson F. Rapid expansion of T cells: Effects of culture and cryopreservation and importance of short-term cell recovery. Acta Oncol 2013; 52:978-86. [PMID: 23126547 DOI: 10.3109/0284186x.2012.737020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Successful cell therapy relies on the identification and mass expansion of functional cells for infusion. Cryopreservation of cells is an inevitable step in most cell therapies which also entails consequences for the frozen cells. MATERIAL AND METHODS This study assessed the impact of cryopreservation and the widely used protocol for rapid expansion of T lymphocytes. The effects on cell viability, immunocompetence and the impact on apoptotic and immunosuppressive marker expression were analyzed using validated assays. RESULTS AND CONCLUSION Cryopreservation of lymphocytes during the rapid expansion protocol did not affect cell viability. Lymphocytes that underwent mass expansion or culture in high dose IL-2 were unable to respond to PHA stimulation by intracellular ATP production immediately after thawing (ATP = 16 ± 11 ng/ml). However, their reactivity to PHA was regained within 48 hours of recovery (ATP = 356 ± 61 ng/ml). Analysis of mRNA levels revealed downregulation of TGF-β and IL-10 at all time points. Culture in high dose IL-2 led to upregulation of p73 and BCL-2 mRNA levels while FoxP3 expression was elevated after culture in IL-2 and artificial TCR stimuli. FoxP3 levels decreased after short-term recovery without IL-2 or stimulation. Antigen specificity, as determined by IFNγ secretion, was unaffected by cryopreservation but was completely lost after addition of high dose IL-2 and artificial TCR stimuli. In conclusion, allowing short-time recovery of mass expanded and cryopreserved cells before reinfusion could enhance the outcome of adoptive cell therapy as the cells regain immune competence and specificity.
Collapse
Affiliation(s)
- Arian Sadeghi
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory Uppsala University, Uppsala, Sweden
| | | | | | | | | |
Collapse
|
18
|
Fujimura K, Oyamada A, Iwamoto Y, Yoshikai Y, Yamada H. CD4 T cell-intrinsic IL-2 signaling differentially affects Th1 and Th17 development. J Leukoc Biol 2013; 94:271-9. [DOI: 10.1189/jlb.1112581] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
19
|
Cichocki F, Miller JS, Anderson SK, Bryceson YT. Epigenetic regulation of NK cell differentiation and effector functions. Front Immunol 2013; 4:55. [PMID: 23450696 PMCID: PMC3584244 DOI: 10.3389/fimmu.2013.00055] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 02/11/2013] [Indexed: 12/24/2022] Open
Abstract
Upon maturation, natural killer (NK) cells acquire effector functions and regulatory receptors. New insights suggest a considerable functional heterogeneity and dynamic regulation of receptor expression in mature human NK cell subsets based on different developmental axes. Such processes include acquisition of lytic granules as well as regulation of cytokine production in response to exogenous cytokine stimulation or target cell interactions. One axis is regulated by expression of inhibitory receptors for self-MHC class I molecules, whereas other axes are less well defined but likely are driven by different activating receptor engagements or cytokines. Moreover, the recent identification of long-lived NK cell subsets in mice that are able to expand and respond rapidly following a secondary viral challenge suggest previously unappreciated plasticity in the programming of NK cell differentiation. Here, we review advances in our understanding of mature NK cell development and plasticity with regards to regulation of cellular function. Furthermore, we highlight some of the major questions that remain pertaining to the epigenetic changes that underlie the differentiation and functional specialization of NK cells and the regulation of their responses.
Collapse
Affiliation(s)
- Frank Cichocki
- Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Karolinska University Hospital Huddinge Stockholm, Sweden ; Adult Division of Hematology, Oncology and Transplantation, University of Minnesota Cancer Center Minneapolis, MN, USA
| | | | | | | |
Collapse
|
20
|
Kimura MY, Pobezinsky LA, Guinter TI, Thomas J, Adams A, Park JH, Tai X, Singer A. IL-7 signaling must be intermittent, not continuous, during CD8⁺ T cell homeostasis to promote cell survival instead of cell death. Nat Immunol 2012; 14:143-51. [PMID: 23242416 PMCID: PMC3552087 DOI: 10.1038/ni.2494] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 11/14/2012] [Indexed: 12/11/2022]
Abstract
The maintenance of naive CD8(+) T cells is necessary for lifelong immunocompetence but for unknown reasons requires signaling via both interleukin 7 (IL-7) and the T cell antigen receptor (TCR). We now report that naive CD8(+) T cells required IL-7 signaling to be intermittent, not continuous, because prolonged IL-7 signaling induced naive CD8(+) T cells to proliferate, produce interferon-γ (IFN-γ) and undergo IFN-γ-triggered cell death. Homeostatic engagement of the TCR interrupted IL-7 signaling and thereby supported the survival and quiescence of CD8(+) T cells. However, CD8(+) T cells with insufficient TCR affinity for self ligands received prolonged IL-7 signaling and died during homeostasis. In this study we identified regulation of the duration of IL-7 signaling by homeostatic engagement of the TCR as the basis for in vivo CD8(+) T cell homeostasis.
Collapse
Affiliation(s)
- Motoko Y Kimura
- Experimental Immunology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Lisowska KA, Dębska-Ślizień A, Jasiulewicz A, Daca A, Bryl E, Witkowski JM. The influence of recombinant human erythropoietin on apoptosis and cytokine production of CD4+ lymphocytes from hemodialyzed patients. J Clin Immunol 2012. [PMID: 23180360 PMCID: PMC3591526 DOI: 10.1007/s10875-012-9835-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recombinant human erythropoietin (rhEPO) treatment of hemodialyzed (HD) patients normalizes the altered phenotype of CD4+ lymphocytes and restores the balance of Th1/Th2 cytokines. We decided to test how the presence of rhEPO in cell culture modulates cytokine production of CD4+ lymphocytes in HD patients with stable hemoglobin level and expression of activation antigens of stimulated CD4+ lymphocytes similar to those observed in healthy individuals. We also tested whether the presence of rhEPO in cell culture protects stimulated CD4+ lymphocytes of HD patients from apoptosis. Peripheral blood mononuclear cells (PBMC) of HD patients were stimulated with an immobilized anti-CD3 antibody with or without addition of rhEPO. The percentage of apoptotic CD4+ lymphocytes and the level of Th1/Th2 cytokines in culture supernatants were measured with flow cytometry. HD patients showed a decrease in the percentage of apoptotic CD4+ cells after stimulation with the anti-CD3 antibody combined with rhEPO. The level of IFN-γ and IL-10 was increased while the level of TNF-α was decreased in the presence of rhEPO in cell culture from HD patients. These results confirm the role of rhEPO signaling in T lymphocytes of HD patients.
Collapse
Affiliation(s)
- Katarzyna A Lisowska
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland.
| | | | | | | | | | | |
Collapse
|
22
|
IL-36 signaling amplifies Th1 responses by enhancing proliferation and Th1 polarization of naive CD4+ T cells. Blood 2012; 120:3478-87. [PMID: 22968459 DOI: 10.1182/blood-2012-06-439026] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The interleukin-1 (IL-1) superfamily of cytokines comprises a set of pivotal mediators of inflammation. Among them, the action of IL-36 cytokines in immune responses has remained elusive. In a recent study, we demonstrated a direct effect of IL-36 on immune cells. Here we show that, among T cells, the IL-36 receptor is predominantly expressed on naive CD4(+) T cells and that IL-36 cytokines act directly on naive T cells by enhancing both cell proliferation and IL-2 secretion. IL-36β acts in synergy with IL-12 to promote Th1 polarization and IL-36 signaling is also involved in mediating Th1 immune responses to Bacillus Calmette-Guerin infection in vivo. Our findings point toward a critical function of IL-36 in the priming of Th1 cell responses in vitro, and in adaptive immunity in a model of mycobacterial infection in vivo.
Collapse
|
23
|
Collins PL, Henderson MA, Aune TM. Diverse functions of distal regulatory elements at the IFNG locus. THE JOURNAL OF IMMUNOLOGY 2012; 188:1726-33. [PMID: 22246629 DOI: 10.4049/jimmunol.1102879] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Previous studies have identified multiple conserved noncoding sequences (CNS) at the mouse Ifng locus sufficient for enhancer activity in cell-based assays. These studies do not directly address biology of the human IFNG locus in a genomic setting. IFNG enhancers may be functionally redundant or each may be functionally unique. We test the hypothesis that each IFNG enhancer has a unique necessary function using a bacterial artificial chromosome transgenic model. We find that CNS-30, CNS-4, and CNS+20 are required at distinct stages of Th1 differentiation, whereas CNS-16 has a repressive role in Th1 and Th2 cells. CNS+20 is required for IFN-γ expression by memory Th1 cells and NKT cells. CNS-4 is required for IFN-γ expression by effector Th1 cells. In contrast, CNS-16, CNS-4, and CNS+20 are each partially required for human IFN-γ expression by NK cells. Thus, IFNG CNS enhancers have redundant necessary functions in NK cells but unique necessary functions in Th cells. These results also demonstrate that distinct CNSs are required to transcribe IFNG at each stage of the Th1 differentiation pathway.
Collapse
Affiliation(s)
- Patrick L Collins
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
24
|
Qui HZ, Hagymasi AT, Bandyopadhyay S, St Rose MC, Ramanarasimhaiah R, Ménoret A, Mittler RS, Gordon SM, Reiner SL, Vella AT, Adler AJ. CD134 plus CD137 dual costimulation induces Eomesodermin in CD4 T cells to program cytotoxic Th1 differentiation. THE JOURNAL OF IMMUNOLOGY 2011; 187:3555-64. [PMID: 21880986 DOI: 10.4049/jimmunol.1101244] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cytotoxic CD4 Th1 cells are emerging as a therapeutically useful T cell lineage that can effectively target tumors, but until now the pathways that govern their differentiation have been poorly understood. We demonstrate that CD134 (OX40) costimulation programs naive self- and virus-reactive CD4 T cells to undergo in vivo differentiation into cytotoxic Th1 effectors. CD137 (4-1BB) costimulation maximized clonal expansion, and IL-2 was necessary for cytotoxic Th1 differentiation. Importantly, the T-box transcription factor Eomesodermin was critical for inducing the cytotoxic marker granzyme B. CD134 plus CD137 dual costimulation also imprinted a cytotoxic phenotype on bystanding CD4 T cells. Thus, to our knowledge, the current study identifies for the first time a specific costimulatory pathway and an intracellular mechanism relying on Eomesodermin that induces both Ag-specific and bystander cytotoxic CD4 Th1 cells. This mechanism might be therapeutically useful because CD134 plus CD137 dual costimulation induced CD4 T cell-dependent tumoricidal function in a mouse melanoma model.
Collapse
Affiliation(s)
- Harry Z Qui
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat Immunol 2011; 12:551-9. [PMID: 21516110 PMCID: PMC3304099 DOI: 10.1038/ni.2030] [Citation(s) in RCA: 360] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 03/30/2011] [Indexed: 12/14/2022]
Abstract
T helper (TH) cells control host-defense to pathogens. The receptors for IL-12, IL-4, and IL-6 are required for TH1, TH2, and TH17 differentiation, respectively. IL-2 signaling via the transcription factor STAT5 controls TH2 differentiation by regulating the TH2 cytokine gene cluster and Il4ra expression. Here we show that IL-2 regulates TH1 differentiation, inducing STAT5-dependent IL-12Rβ2 and T-bet expression, with impaired human TH1 differentiation when IL-2 was blocked. TH1 differentiation was also impaired in mouse Il2−/− T cells but restored by IL-12Rβ2 expression. Consistent with IL-2’s inhibition of TH17 differentiation, IL-2 decreased Il6ra and Il6st expression, and Il6st augmented TH17 differentiation even when IL-2 was present. Thus, IL-2 influences TH cell differentiation by modulating cytokine receptor expression to help specify and maintain differentiated states.
Collapse
|
26
|
Development of an IL-15-autocrine CD8 T-cell leukemia in IL-15-transgenic mice requires the cis expression of IL-15Rα. Blood 2011; 117:4032-40. [PMID: 21304101 DOI: 10.1182/blood-2010-09-307504] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
IL-15 has growth-promoting effects on select lymphoid subsets, including natural killer (NK) cells, NK T cells, intraepithelial lymphocytes (IELs), CD8 T cells, and γδ-T cells. Constitutive expression of murine IL-15 in IL-15-transgenic mice was reported to cause T-NK leukemia. We investigated whether IL-15 expression is sufficient for leukemic transformation using a human IL-15-transgenic (IL-15Tg) mouse model. We noted that 100% of the mice observed over a 2-year period (n > 150) developed fatal expansions of CD8 T cells with NK markers, and determined that these cells expressed IL-15 receptor alpha (IL-15Rα). The expression of IL-15Rα on CD8 T cells appears to be required for uncontrolled aggressive lymphoproliferation, because none of the IL-15Rα(-/-)-IL-15Tg mice that we followed for more than 2 years developed the fatal disease despite controlled expansion of CD8 T cells. In addition, in contrast to IL-15Tg mice, in which leukemia-like CD8 T cells expressed IL-15Rα persistently, acutely activated normal CD8 T cells only transiently expressed IL-15Rα. Inhibition of DNA methylation enabled sustained IL-15Rα expression induced by activation. We present a scenario for IL-15Tg mice in which CD8 T cells that acquire constitutive persistent IL-15Rα expression are at a selective advantage and become founder cells, outgrow other lymphocytes, and lead to the establishment of a leukemia-like condition.
Collapse
|
27
|
Abstract
CD4(+) T helper (T(H)) cells play a critical role in orchestrating a pleiotropy of immune activities against a large variety of pathogens. It is generally thought that this is achieved through the acquisition of highly specialized functions after activation followed by the differentiation into various functional subsets. The differentiation process of naive precursor T(H) cells into defined effector subsets is controlled by cells of the innate immune system and their complex array of effector molecules such as secreted cytokines and membrane bound costimulatory molecules. These provide a unique quantitative or qualitative signal initiating T(H) development, which is subsequently reinforced via T cell-mediated feedback signals and selective survival and proliferative cues, ultimately resulting in the predominance of a particular T cell subset. In recent years, the number of defined T(H)cell subsets has expanded and the once rigid division of labor among them has been blurred with reports of plasticity among the subsets. In this chapter, we summarize and speculate on the current knowledge of the differentiation requirements of T(H) cell lineages, with particular focus on the T(H)17 subset.
Collapse
|
28
|
Collins PL, Chang S, Henderson M, Soutto M, Davis GM, McLoed AG, Townsend MJ, Glimcher LH, Mortlock DP, Aune TM. Distal regions of the human IFNG locus direct cell type-specific expression. THE JOURNAL OF IMMUNOLOGY 2010; 185:1492-501. [PMID: 20574006 DOI: 10.4049/jimmunol.1000124] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genes, such as IFNG, which are expressed in multiple cell lineages of the immune system, may employ a common set of regulatory elements to direct transcription in multiple cell types or individual regulatory elements to direct expression in individual cell lineages. By employing a bacterial artificial chromosome transgenic system, we demonstrate that IFNG employs unique regulatory elements to achieve lineage-specific transcriptional control. Specifically, a one 1-kb element 30 kb upstream of IFNG activates transcription in T cells and NKT cells but not in NK cells. This distal regulatory element is a Runx3 binding site in Th1 cells and is needed for RNA polymerase II recruitment to IFNG, but it is not absolutely required for histone acetylation of the IFNG locus. These results support a model whereby IFNG uses cis-regulatory elements with cell type-restricted function.
Collapse
Affiliation(s)
- Patrick L Collins
- Division of Rheumatology, Department of Medicine, Medical Center North T3219, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cruickshank MN, Besant P, Ulgiati D. The impact of histone post-translational modifications on developmental gene regulation. Amino Acids 2010; 39:1087-105. [PMID: 20204433 DOI: 10.1007/s00726-010-0530-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 02/12/2010] [Indexed: 02/06/2023]
Abstract
Eukaryotic genomic DNA is orderly compacted to fit into the nucleus and to inhibit accessibility of specific sequences. DNA is manipulated in many different ways by bound RNA and proteins within the composite material known as chromatin. All of the biological processes that require access to genomic DNA (such as replication, recombination and transcription) therefore are dependent on the precise characteristics of chromatin in eukaryotes. This distinction underlies a fundamental property of eukaryotic versus prokaryotic gene regulation such that chromatin structure must be regulated to precisely repress or relieve repression of particular regions of the genome in an appropriate spatio-temporal manner. As well as playing a key role in structuring genomic DNA, histones are subject to site-specific modifications that can influence the organization of chromatin structure. This review examines the molecular processes regulating site-specific histone acetylation, methylation and phosphorylation with an emphasis on how these processes underpin differentiation-regulated transcription.
Collapse
Affiliation(s)
- Mark N Cruickshank
- Biochemistry and Molecular Biology, School of Biomedical, Biomolecular and Chemical Sciences, University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | | | | |
Collapse
|
30
|
Monitoring of the immunomodulatory effect of CP-690,550 by analysis of the JAK/STAT pathway in kidney transplant patients. Transplantation 2010; 88:1002-9. [PMID: 19855246 DOI: 10.1097/tp.0b013e3181b9ced7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND.: The small molecule drug CP-690,550 inhibits Janus kinase 3 at nanomolar concentrations and has recently been shown to prevent allograft rejection in rodents and nonhuman primates. METHODS.: As part of a phase 1 clinical trial, we investigated the effect of CP-690,550 after 29 days of 30 mg twice daily treatment at the cellular level in eight kidney transplant patients by studying ex vivo phosphorylation of STAT5 (P-STAT5), the key substrate of JAK3. RESULTS.: As determined by quantitative fluorescent western blotting, interleukin-2-induced P-STAT5 in YT cells was reduced by a median of 73% (P<0.01) in the presence of serum collected on day 29 compared with pretreatment baseline. When evaluated by phosphospecific flow cytometry, CP-690,550 also reduced interleukin-2-induced P-STAT5 in CD3 (median 20%; P<0.05), CD3CD4 (median 37%; P<0.05), and CD3CD8 (median 34%; P<0.01) populations in patient-derived peripheral blood mononuclear cells. At the functional level, the inhibitory effect of CP-690,550 was confirmed by determining the expression of several STAT5 targets genes. CONCLUSION.: Analysis of P-STAT5 may, therefore, be used to determine the immunomodulatory effect of CP-690,550 at the cellular level in transplant patients.
Collapse
|
31
|
Shaw MH, Reimer T, Sánchez-Valdepeñas C, Warner N, Kim YG, Fresno M, Nuñez G. T cell-intrinsic role of Nod2 in promoting type 1 immunity to Toxoplasma gondii. Nat Immunol 2009; 10:1267-74. [PMID: 19881508 PMCID: PMC2803073 DOI: 10.1038/ni.1816] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 09/24/2009] [Indexed: 02/08/2023]
Abstract
Nod2 belongs to the (NOD)-like receptor (NLR) family of proteins, which function as intracellular pathogen sensors in innate immune cells. Nod2-deficiency results in an impaired immune response against bacterial pathogens. However, our understanding of how this protein promotes host defense against intracellular parasites is unknown. Here we found that Nod2−/− mice showed reduced clearance of Toxoplasma gondii and decreased interferon-γ production. Reconstitution of T-cell deficient mice with Nod2−/− T cells followed by T. gondii infection revealed a T cell-intrinsic defect. Nod2−/− CD4+ T cells displayed poor helper T cell differentiation, which was associated with impaired IL-2 production and nuclear accumulation of c-Rel. These data revealed a T cell-intrinsic role of Nod2 signaling that is critical for host defense against T. gondii.
Collapse
Affiliation(s)
- Michael H Shaw
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Itsumi M, Yoshikai Y, Yamada H. IL-15 is critical for the maintenance and innate functions of self-specific CD8(+) T cells. Eur J Immunol 2009; 39:1784-93. [PMID: 19544306 DOI: 10.1002/eji.200839106] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
IL-15 is a pleiotropic cytokine involved in host defense as well as autoimmunity. IL-15-deficient mice show a decrease of memory phenotype (MP) CD8(+) T cells, which develop naturally in naïve mice and whose origin is unclear. It has been shown that self-specific CD8(+) T cells developed in male H-Y antigen-specific TCR transgenic mice share many similarities with naturally occurring MP CD8(+) T cells in normal mice. In this study, we found that H-Y antigen-specific CD8(+) T cells in male but not female mice decreased when they were crossed with IL-15-deficient mice, mainly due to impaired peripheral maintenance. The self-specific TCR transgenic CD8(+) T cells developed in IL-15-deficient mice showed altered surface phenotypes and reduced effector functions ex vivo. Bystander activation of the self-specific CD8(+) T cells was induced in vivo during infection with Listeria monocytogenes, in which proliferation but not IFN-gamma production was IL-15-dependent. These results indicated important roles for IL-15 in the maintenance and functions of self-specific CD8(+) T cells, which may be included in the naturally occurring MP CD8(+) T-cell population in naïve normal mice and participate in innate host defense responses.
Collapse
Affiliation(s)
- Momoe Itsumi
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | |
Collapse
|
33
|
The effect of the JAK inhibitor CP-690,550 on peripheral immune parameters in stable kidney allograft patients. Transplantation 2009; 87:79-86. [PMID: 19136895 DOI: 10.1097/tp.0b013e31818bbea7] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION CP-690,550 inhibits Janus kinase 3 (JAK3) which mediates signal transduction of receptors of the common gamma-chain cytokines. These cytokines play key roles in lymphocyte function and homeostasis. As part of a phase 1 trial, we evaluated the effect of CP-690,550 on immune parameters. MATERIAL Stable kidney transplant recipients (n=8) receiving mycophenolate mofetil and prednisolone were treated with CP-690,550, 30 mg twice daily orally for 29 days. Blood samples were collected on days 1 (before first dose), 15, 29 (end of treatment), and 57. RESULTS Two patients experienced minor infections (one urinary tract infection and one mild respiratory tract infection). Leukocyte counts remained stable, whereas a mean decrease in hemoglobulin of 8% was measured (P=0.01). CP-690,550 treatment for 29 days resulted in statistically significant changes in the number of circulating CD19+ B cells (P=0.05), CD3- CD16+ CD56+ natural killer-cells (P<0.01), and CD4+ CD25bright+ T cells (P=0.05; one-way analysis of variance). After CP-690,550 treatment on day 15 the number of B cells increased by a mean of 100%, (P=0.04), whereas those of natural killer cells and CD4+ CD25bright+ T cells decreased by 65% (P=0.001) and 38% (P=0.03, t test), respectively, from pretreatment baseline. However, the regulatory capacities of the residual CD4+ CD25bright+ T cells remained unchanged pre- and posttreatment. In addition, in the presence of CP-690,550, the interferon-[gamma] production capacity of peripheral blood mononuclear cells was reduced by 39% (median) compared with predose baseline (P=0.01). CONCLUSIONS These findings demonstrate the role of JAK3 in the homeostasis and function of select lymphocyte subpopulations. JAK3 inhibition may provide a novel mechanism for the modulation of allogeneic responses in patients after transplantation.
Collapse
|
34
|
Chang S, Collins PL, Aune TM. T-bet dependent removal of Sin3A-histone deacetylase complexes at the Ifng locus drives Th1 differentiation. THE JOURNAL OF IMMUNOLOGY 2009; 181:8372-81. [PMID: 19050254 DOI: 10.4049/jimmunol.181.12.8372] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Forming and removing epigenetic histone marks at gene loci are central processes in differentiation. Here, we explored mechanisms establishing long-range H4 acetylation marks at the Ifng locus during Th1 lineage commitment. In Th0 cells, histone deacetylase (HDAC)-Sin3A complexes recruited to the Ifng locus actively prevented accumulation of H4 acetylation marks. Th1 differentiation caused loss of HDAC-Sin3A complexes by T-bet-dependent mechanisms and accumulation of H4 acetylation marks. HDAC-Sin3A complexes were absent from the locus in NOD Th0 cells, obviating the need for Th1 differentiation signals to establish histone marks and Th1 differentiation. Thus, Ifng transcription is actively prevented in Th0 cells via epigenetic mechanisms and epigenetic defects allow unregulated Ifng transcription that may contribute to autoimmunity.
Collapse
Affiliation(s)
- Shaojing Chang
- Department of Medicine, Division of Rheumatology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
35
|
Abstract
Naïve T helper cells differentiate into two subsets, T helper 1 and 2, which either transcribe the Ifng gene and silence the Il4 gene or transcribe the Il4 gene and silence the Ifng gene, respectively. This process is an essential feature of the adaptive immune response to a pathogen and the development of long-lasting immunity. The 'histone code' hypothesis proposes that formation of stable epigenetic histone marks at a gene locus that activate or repress transcription is essential for cell fate determinations, such as T helper 1/T helper 2 cell fate decisions. Activation and silencing of the Ifng gene are achieved through the creation of stable epigenetic histone marks spanning a region of genomic DNA over 20 times greater than the gene itself. Key transcription factors that drive the T helper 1 lineage decision, signal transducer and activator 4 (STAT4) and T-box expressed in T cells (T-bet), play direct roles in the formation of activating histone marks at the Ifng locus. Conversely, STAT6 and GATA binding protein 3, transcription factors essential for the T helper 2 cell lineage decision, establish repressive histone marks at the Ifng locus. Functional studies demonstrate that multiple genomic elements up to 50 kilobases from Ifng play critical roles in its proper transcriptional regulation. Studies of three-dimensional chromatin conformation indicate that these distal regulatory elements may loop towards Ifng to regulate its transcription. We speculate that these complex mechanisms have evolved to tightly control levels of interferon-gamma production, given that too little or too much production would be very deleterious to the host.
Collapse
Affiliation(s)
- Thomas M Aune
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-2068, USA.
| | | | | |
Collapse
|
36
|
Shi M, Lin TH, Appell KC, Berg LJ. Janus-kinase-3-dependent signals induce chromatin remodeling at the Ifng locus during T helper 1 cell differentiation. Immunity 2008; 28:763-73. [PMID: 18549798 DOI: 10.1016/j.immuni.2008.04.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 01/06/2023]
Abstract
Differentiation of naive CD4+ T cells into T helper type 1 (Th1) effector cells requires both T cell receptor (TCR) signaling and cytokines such as interleukin-12 and interferon gamma (IFN-gamma). Here, we report that a third cytokine signal, mediated by the Janus family tyrosine kinase 3 (Jak3) and signal transducer and activator of transcription 5 (STAT5) pathway, is also required for Th1 cell differentiation. In the absence of Jak3-dependent signals, naive CD4+ T cells proliferate robustly but produce little IFN-gamma after Th1 cell polarization in vitro. This defect is not due to reduced activation of STAT1 or STAT4 or to impaired upregulation of the transcription factor T-bet. Instead, we find that T-bet binding to the Ifng promoter is greatly diminished in the absence of Jak3-dependent signals, correlating with a decrease in Ifng promoter accessibility and histone acetylation. These data indicate that Jak3 regulates epigenetic modification and chromatin remodeling of the Ifng locus during Th1 cell differentiation.
Collapse
Affiliation(s)
- Min Shi
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
37
|
Grant LR, Yao ZJ, Hedrich CM, Wang F, Moorthy A, Wilson K, Ranatunga D, Bream JH. Stat4-dependent, T-bet-independent regulation of IL-10 in NK cells. Genes Immun 2008; 9:316-27. [PMID: 18401353 PMCID: PMC2689787 DOI: 10.1038/gene.2008.20] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 02/11/2008] [Accepted: 02/14/2008] [Indexed: 12/22/2022]
Abstract
Interleukin-10 (IL-10) is intensely studied, yet little is known about the mechanisms that control IL-10 expression. We identified striking similarities between IL-10 and interferon-gamma (IFN-gamma) regulation in mouse natural killer (NK) cells. Like IFN-gamma, IL-10 expression is induced by IL-2 and IL-12 and IL-2+IL-12 stimulation is synergistic. Unlike IFN-gamma, neither IL-18 nor Ly-49D cross-linking induced IL-10 expression however. Additionally, the IL-12 homologs IL-23 and IL-27 also do not regulate NK cell-specific IL-10. We determined that a small population of NK cells accounts for IL-10 production. The induction of IL-10 by IL-2+IL-12 treatment in NK cells appears to be biphasic, with an initial burst of expression which diminishes by 12 h but spikes again at 18 h. We determined that much like IFN-gamma, Stat4 is largely required for IL-12-induced IL-10. Conversely, we observed normal induction of IL-10 in T-bet-deficient NK cells. We identified a Stat4-binding element in the fourth intron of the Il10 gene, which is completely conserved between mouse and human. This intronic Stat4 motif is within a conserved noncoding sequence, which is also a target for cytokine-induced histone acetylation. These findings highlight tissue- and receptor-specific IL-10 regulatory mechanisms, which may be part of an early feedback loop.
Collapse
Affiliation(s)
- LR Grant
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Z-J Yao
- Lymphocyte and Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - CM Hedrich
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - F Wang
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - A Moorthy
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - K Wilson
- Lymphocyte and Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Musculoskeletal and Skin Diseases, Bethesda, MD, USA
| | - D Ranatunga
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - JH Bream
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
38
|
Abstract
Interferon-gamma (IFN-gamma) is crucial for immunity against intracellular pathogens and for tumor control. However, aberrant IFN-gamma expression has been associated with a number of autoinflammatory and autoimmune diseases. This cytokine is produced predominantly by natural killer (NK) and natural killer T (NKT) cells as part of the innate immune response, and by Th1 CD4 and CD8 cytotoxic T lymphocyte (CTL) effector T cells once antigen-specific immunity develops. Herein, we briefly review the functions of IFN-gamma, the cells that produce it, the cell extrinsic signals that induce its production and influence the differentiation of naïve T cells into IFN-gamma-producing effector T cells, and the signaling pathways and transcription factors that facilitate, induce, or repress production of this cytokine. We then review and discuss recent insights regarding the molecular regulation of IFN-gamma, focusing on work that has led to the identification and characterization of distal regulatory elements and epigenetic modifications with the IFN-gamma locus (Ifng) that govern its expression. The epigenetic modifications and three-dimensional structure of the Ifng locus in naive CD4 T cells, and the modifications they undergo as these cells differentiate into effector T cells, suggest a model whereby the chromatin architecture of Ifng is poised to facilitate either rapid opening or silencing during Th1 or Th2 differentiation, respectively.
Collapse
|
39
|
Chen Z, Laurence A, O'Shea JJ. Signal transduction pathways and transcriptional regulation in the control of Th17 differentiation. Semin Immunol 2007; 19:400-8. [PMID: 18166487 PMCID: PMC2323678 DOI: 10.1016/j.smim.2007.10.015] [Citation(s) in RCA: 204] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2007] [Revised: 10/30/2007] [Accepted: 10/31/2007] [Indexed: 11/15/2022]
Abstract
The discovery of a new lineage of helper T cells that selectively produces interleukin (IL)-17 has provided exciting new insights into immunoregulation, host defense and the pathogenesis of autoimmune diseases. Additionally, the discovery of this T cell subset has offered a fresh look at how the complexity of selective regulation of cytokine gene expression might relate to lineage commitment, terminal differentiation and immunologic memory. Information continues to accumulate on factors that regulate Th17 differentiation at a rapid pace and a few lessons have emerged. Like other lineages, Th17 cells preferentially express a transcription factor, retinoic acid-related orphan receptor (ROR)gammat, whose expression seems to be necessary for IL-17 production. In addition, signals from the T-cell receptor are a critical aspect of controlling IL-17 production and the transcription factor nuclear factor of activated T cells (NFATs) appears to be another important regulator. IL-6, IL-21 and IL-23 are all cytokines that activate the transcription factor STAT3, which has been established to be necessary for multiple aspects of the biology of Th17 cells. Similarly, TGFbeta-1 is important for the differentiation of murine Th17 cells and inducible regulatory T cells (iTregs), but how it exerts its effect on IL-17 gene transcription is unknown and there are data indicating TGFbeta-1 is not required for human Th17 differentiation. The extent to which Th17 cells represent terminally differentiated cells or whether they retain plasticity and can develop into another lineage such as IFNgamma secreting Th1 cells is also unclear. Precisely how cytokines produced by this lineage are selectively expressed and selectively extinguished through epigenetic modifications is an area of great importance, but considerable uncertainty.
Collapse
Affiliation(s)
- Zhi Chen
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
40
|
Hamalainen-Laanaya HK, Kobie JJ, Chang C, Zeng WP. Temporal and Spatial Changes of Histone 3 K4 Dimethylation at the IFN-γ Gene during Th1 and Th2 Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2007; 179:6410-5. [DOI: 10.4049/jimmunol.179.10.6410] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
41
|
Trotta R, Ciarlariello D, Dal Col J, Allard J, Neviani P, Santhanam R, Mao H, Becknell B, Yu J, Ferketich AK, Thomas B, Modi A, Blaser BW, Perrotti D, Caligiuri MA. The PP2A inhibitor SET regulates natural killer cell IFN-gamma production. ACTA ACUST UNITED AC 2007; 204:2397-405. [PMID: 17875674 PMCID: PMC2118465 DOI: 10.1084/jem.20070419] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Monokines (i.e., interleukin [IL]-12, -18, and -15) induce natural killer (NK) cells to produce interferon-γ (IFN-γ), which is a critical factor for immune surveillance of cancer and monocyte clearance of infection. We show that SET, which is a potent inhibitor of protein phosphatase type 2A (PP2A) activity, is highly expressed in human CD56bright NK cells, which produce more IFN-γ than CD56dim NK cells. SET was up-regulated upon monokine stimulation of primary human NK cells. Furthermore, ectopic overexpression of SET significantly enhanced IFN-γ gene expression in monokine-stimulated NK cells. In contrast, RNAi-mediated suppression of SET expression renders NK cells inefficient in producing high levels of IFN-γ in response to monokine costimulation. Mechanistically, suppression of PP2A activity by SET is important for IFN-γ gene expression in NK cells. In fact, treatment of primary human NK cells with the PP2A activator 1,9-dideoxy-forskolin, as well as administration of the drug to C57BL/6 mice, significantly reduced NK-dependent IFN-γ production in response to monokine treatment. Further, SET knockdown or pharmacologic activation of PP2A diminished extracellular signal-regulated kinase 1/2, p65RelA, signal transducer and activator of transduction 4 (STAT4), and STAT5 activity in monokine-stimulated NK cells, potentially contributing to the reduction in IFN-γ gene expression. Thus, SET expression is essential for suppressing PP2A phosphatase activity that would otherwise limit NK cell antitumoral and/or antiinflammatory functions by impairing NK cell production of IFN-γ.
Collapse
Affiliation(s)
- Rossana Trotta
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bendle GM, Xue SA, Holler A, Stauss HJ. A study of T cell tolerance to the tumor-associated antigen MDM2: cytokines can restore antigen responsiveness, but not high avidity T cell function. PLoS One 2007; 2:e353. [PMID: 17406677 PMCID: PMC1831496 DOI: 10.1371/journal.pone.0000353] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Accepted: 03/13/2007] [Indexed: 01/23/2023] Open
Abstract
Background Most tumor-associated antigens (TAA) currently used for immunotherapy of cancer are also expressed in normal tissues, which may induce tolerance and impair T cell-mediated immunity. However, there is limited information about how physiological expression in normal tissues alters the function of TAA-specific T cells. Methodology/Principal Findings We used a T cell receptor transgenic model to study how MDM2 expression in normal tissues affects the function of T cells specific for this TAA that is found at high levels in many different types of tumors. We found that some MDM2-specific T cells escaped thymic deletion and persisted in the peripheral T cell pool. When stimulated with antigen, these T cells readily initiated cell division but failed to proliferate and expand, which was associated with a high rate of apoptosis. Both IL-2 and IL-15 efficiently rescued T cell survival and antigen-specific T cell proliferation, while IL-7 and IL-21 were ineffective. Antigen-stimulated T cells showed impaired expression of the effector molecules CD43, granzyme-B and IFN-γ, a defect that was completely restored when T cells were stimulated in the presence of IL-2. In contrast, IL-15 and IL-21 only restored the expression of CD43 and granzyme-B, but not IFN-γ production. Finally, peptide titration experiments with IL-2 rescued T cells indicated that they were of lower avidity than non-tolerant control T cells expressing the same TCR. Conclusions/Significance These data indicate that cytokines can rescue the antigen-specific proliferation and effector function of MDM2-specific T cells, although this does not lead to the recovery of high avidity T cell function. This study sheds light on possible limitations of immunotherapy approaches that target widely expressed TAA, such as MDM2.
Collapse
|
43
|
USUI T. Transcription Factors That Regulate Helper T Cell Differentiation. ACTA ACUST UNITED AC 2007; 30:419-27. [DOI: 10.2177/jsci.30.419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Takashi USUI
- Department of Rheumatology and Clinical Immunology, Kyoto University, Graduate School of Medicine
| |
Collapse
|
44
|
Becknell B, Hughes TL, Freud AG, Blaser BW, Yu J, Trotta R, Mao HC, Caligiuri de Jesús ML, Alghothani M, Benson DM, Lehman A, Jarjoura D, Perrotti D, Bates MD, Caligiuri MA. Hlx homeobox transcription factor negatively regulates interferon-gamma production in monokine-activated natural killer cells. Blood 2006; 109:2481-7. [PMID: 17110450 PMCID: PMC1852195 DOI: 10.1182/blood-2006-10-050096] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells contribute to host immunity, including tumor surveillance, through the production of interferon gamma (IFN-gamma). Although there is some knowledge about molecular mechanisms that induce IFN-gamma in NK cells, considerably less is known about the mechanisms that reduce its expression. Here, we investigate the role of the Hlx transcription factor in IFN-gamma production by NK cells. Hlx expression is induced in monokine-activated NK cells, but with delayed kinetics compared to IFN-gamma. Ectopic Hlx expression decreases IFN-gamma synthesis in primary human NK cells and IFN-gamma promoter activity in an NK-like cell line. Hlx protein levels inversely correlate with those of STAT4, a requisite factor for optimal IFN-gamma transcription. Mechanistically, we provide evidence indicating that Hlx overexpression accelerates dephosphorylation and proteasome-dependent degradation of the active Y693-phosphorylated form of STAT4. Thus, Hlx expression in activated NK cells temporally controls and limits the monokine-induced production of IFN-gamma, in part through the targeted depletion of STAT4.
Collapse
Affiliation(s)
- Brian Becknell
- Medical Scientist Program, Department of Internal Medicine, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Butz NV, Gronostajski RM, Campbell CE. T-box proteins differentially activate the expression of the endogenous interferon γ gene versus transfected reporter genes in non-immune cells. Gene 2006; 377:130-9. [PMID: 16737784 DOI: 10.1016/j.gene.2006.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 03/17/2006] [Accepted: 03/25/2006] [Indexed: 02/02/2023]
Abstract
The T-box transcription factor T-bet is expressed in a number of hematopoetic cell types and plays an essential role in the lineage determination of Th1 T-helper cells. In the absence of T-bet, CD4(+) T-cells fail to induce IFNgamma, the cytokine whose expression characterizes Th1 cells. Here we show that, surprisingly, T-bet induces the expression of endogenous IFNgamma in non-immune human cells, including 293 and other cell lines. Thus T-bet can induce IFNgamma expression independently of its role in T-cell lineage determination. In addition, mutations in T-bet, and chimeras of T-bet with other transcription factors including the T-box transcription factor, TBX2, differentially affect the ability of T-bet to activate expression of endogenous IFNgamma versus a T-site regulated reporter gene. A truncated T-betVp16 fusion protein strongly activates the T-site reporter but fails to activate endogenous IFNgamma. Conversely, native T-bet strongly activates endogenous IFNgamma expression but only weakly activates the reporter gene. Fusion of the Vp16 activation domain to full-length T-bet greatly increases its activation of both endogenous IFNgamma and transfected T-site reporter gene expression. In contrast, TBX2Vp16 potently activates the T-site reporter but has a negligible effect on endogenous IFNgamma expression. Butyrate treatment of T-bet expressing cells potentiates the expression of endogenous IFNgamma but weakly represses expression of the T-site reporter gene. These data indicate that induction of endogenous IFNgamma can be uncoupled from differentiation into the Th1 lineage and that the expression of endogenous IFNgamma versus a T-site reporter gene is differentially regulated by T-bet and other T-box proteins.
Collapse
Affiliation(s)
- Nataliya V Butz
- Department of Biochemistry, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, NY 14214, USA
| | | | | |
Collapse
|
46
|
Nelson EA, Walker SR, Li W, Liu XS, Frank DA. Identification of human STAT5-dependent gene regulatory elements based on interspecies homology. J Biol Chem 2006; 281:26216-24. [PMID: 16840779 PMCID: PMC3427032 DOI: 10.1074/jbc.m605001200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
STAT5 is a transcription factor essential for hematopoietic physiology. STAT5 functions to transduce signals from cytokines to the nucleus where it regulates gene expression. Although several important transcriptional targets of STAT5 are known, most remain unidentified. To identify novel STAT5 targets, we searched chromosomes 21 and 22 for clusters of STAT5 binding sites contained within regions of interspecies homology. We identified four such regions, including one with tandem STAT5 binding sites in the first intron of the NCAM2 gene. Unlike known STAT5 binding sites, this site is found within a very large intron and resides approximately 200 kb from the first coding exon of NCAM2. We demonstrate that this region confers STAT5-dependent transcriptional activity. We show that STAT5 binds in vivo to the NCAM2 intron in the NKL natural killer cell line and that this binding is induced by cytokines that activate STAT5. Neither STAT1 nor STAT3 bind to this region, despite sharing a consensus binding sequence with STAT5. Activation of STAT4 and STAT5 causes the accumulation of both of these STATs to the NCAM2 regulatory region. Therefore, using an informatics based approach to identify STAT5 targets, we have identified NCAM2 as both a STAT4- and STAT5-regulated gene, and we show that its expression is regulated by cytokines essential for natural killer cell survival and differentiation. This strategy may be an effective way to identify functional binding regions for transcription factors with known cognate binding sites anywhere in the genome.
Collapse
Affiliation(s)
- Erik A. Nelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115
- Departments of Medicine, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts 02115
| | - Sarah R. Walker
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115
- Departments of Medicine, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts 02115
| | - Wei Li
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115
- Harvard School of Public Health, Harvard Medical School, Boston, Massachusetts 02115
| | - X. Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115
- Harvard School of Public Health, Harvard Medical School, Boston, Massachusetts 02115
| | - David A. Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115
- Departments of Medicine, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts 02115
- To whom correspondence should be addressed: Dept. of Medical Oncology, Dana-Farber Cancer Institute, 44 Binney St., Boston, MA 02115. Tel.: 617-632-4714; Fax: 617-632-6356;
| |
Collapse
|
47
|
Hashimoto T, Kobayashi N, Kajiyama Y, Kaminuma O, Suko M, Mori A. IL-2-induced IL-13 production by allergen-specific human helper T cell clones. Int Arch Allergy Immunol 2006; 140 Suppl 1:51-4. [PMID: 16772727 DOI: 10.1159/000092711] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Interleukin (IL)-5 and IL-13 are important cytokines in allergic diseases such as asthma and atopic dermatitis. We have reported that the production of IL-5 and IL-13 by mite-responsive T helper cells (Th) is controlled under similar signal requirements, but precise mechanisms are not yet well characterized. METHODS Allergen-specific Th clones were established from peripheral blood lymphocytes of atopic asthmatics, and cytokine synthesis in response to various stimuli was determined by specific ELISAs. IL-13 gene expression was enumerated by quantitative real-time PCR. RESULTS IL-13 production was clearly induced by IL-2. IL-13 mRNA expression was also induced. CONCLUSION The IL-2 signal by itself causes IL-13 synthesis independent of T cell receptor stimulation.
Collapse
Affiliation(s)
- Tomomi Hashimoto
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Usui T, Preiss JC, Kanno Y, Yao ZJ, Bream JH, O'Shea JJ, Strober W. T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription. ACTA ACUST UNITED AC 2006; 203:755-66. [PMID: 16520391 PMCID: PMC2118252 DOI: 10.1084/jem.20052165] [Citation(s) in RCA: 247] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
T helper type 1 (Th1) development is facilitated by interrelated changes in key intracellular factors, particularly signal transducer and activator of transcription (STAT)4, T-bet, and GATA-3. Here we show that CD4+ cells from T-bet−/− mice are skewed toward Th2 differentiation by high endogenous GATA-3 levels but exhibit virtually normal Th1 differentiation provided that GATA-3 levels are regulated at an early stage by anti–interleukin (IL)-4 blockade of IL-4 receptor (R) signaling. In addition, under these conditions, Th1 cells from T-bet−/− mice manifest IFNG promotor accessibility as detected by histone acetylation and deoxyribonuclease I hypersensitivity. In related studies, we show that the negative effect of GATA-3 on Th1 differentiation in T-bet−/− cells arises from its ability to suppress STAT4 levels, because if this is prevented by a STAT4-expressing retrovirus, normal Th1 differentiation is observed. Finally, we show that retroviral T-bet expression in developing and established Th2 cells leads to down-regulation of GATA-3 levels. These findings lead to a model of T cell differentiation that holds that naive T cells tend toward Th2 differentiation through induction of GATA-3 and subsequent down-regulation of STAT4/IL-12Rβ2 chain unless GATA-3 levels or function is regulated by T-bet. Thus, the principal function of T-bet in developing Th1 cells is to negatively regulate GATA-3 rather than to positively regulate the IFNG gene.
Collapse
MESH Headings
- Acetylation
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Down-Regulation/genetics
- Down-Regulation/immunology
- GATA3 Transcription Factor/genetics
- GATA3 Transcription Factor/immunology
- GATA4 Transcription Factor/genetics
- GATA4 Transcription Factor/immunology
- Histones/genetics
- Histones/immunology
- Humans
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Mice
- Mice, Knockout
- Protein Processing, Post-Translational/genetics
- Protein Processing, Post-Translational/immunology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/immunology
- Receptors, Interleukin-12
- Retroviridae
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Box Domain Proteins
- Th1 Cells/immunology
- Th2 Cells/immunology
- Transcription Factors/deficiency
- Transcription Factors/immunology
- Transcription, Genetic/genetics
- Transcription, Genetic/immunology
- Transduction, Genetic/methods
Collapse
Affiliation(s)
- Takashi Usui
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Tato CM, Mason N, Artis D, Shapira S, Caamano JC, Bream JH, Liou HC, Hunter CA. Opposing roles of NF-kappaB family members in the regulation of NK cell proliferation and production of IFN-gamma. Int Immunol 2006; 18:505-13. [PMID: 16481345 PMCID: PMC1800429 DOI: 10.1093/intimm/dxh391] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is well established that the nuclear factor-kappaB (NF-kappaB) family of transcription factors participates in the regulation of many aspects of innate and adaptive immunity. The majority of these reports have focused on the role of NF-kappaB in accessory cell and T or B cell function, but less is known about the role of NF-kappaB in NK cells. However, several studies have demonstrated that these transcription factors are required for NK cell production of IFN-gamma and proliferation. The studies presented here examine the role of two NF-kappaB members, c-Rel and p50, in NK cell function. In vitro data revealed that in the absence of c-Rel, NK cells have a defect in their ability to secrete IFN-gamma, but remain unaffected in their capacity to proliferate. In contrast, p50-/- NK cells have enhanced proliferative and IFN-gamma responses compared with wild-type NK cells. The latter findings suggest a role for p50 as a negative regulator of NK cell production of IFN-gamma and chromatin immunoprecipitation assays demonstrated the association of p50 with the IFN-gamma promoter of resting NK cells. Consistent with the in vitro studies, in vivo studies with NF-kappaB gene-deficient mice infected with Toxoplasma gondii revealed that the absence of p50 leads to enhanced NK cell proliferation and production of IFN-gamma. Together, these studies define distinct roles for c-Rel and p50 in the function of NK cells.
Collapse
Affiliation(s)
- Cristina M Tato
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104-6008, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wilson CB, Merkenschlager M. Chromatin structure and gene regulation in T cell development and function. Curr Opin Immunol 2006; 18:143-51. [PMID: 16472999 PMCID: PMC1820769 DOI: 10.1016/j.coi.2006.01.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2005] [Accepted: 01/31/2006] [Indexed: 01/07/2023]
Abstract
Transcription factors control gene expression programs in the context of the chromatin structure of their target genes. DNA methylation, post-translational histone modifications such as acetylation and methylation, and higher order chromatin organization allow the maintenance of gene expression patterns through mitosis, but how do they accommodate developmentally regulated changes in gene expression programs? Although histone acetylation and deacetylation are in dynamic equilibrium and mechanisms for the removal of methyl groups from histones are emerging, the extent to which there is active demethylation of DNA remains controversial. Looking at chromatin in the three-dimensional space of the nucleus, recent work demonstrates that gene regulation involves contacts between regulatory elements within genes or gene clusters on the same chromosome (in cis) and between different chromosomes (in trans). Finally, non-coding RNAs make a significant contribution to transcriptional and post-transcriptional gene silencing. Together, these advances contribute to an understanding of how gene expression programs are established, maintained and modified during development.
Collapse
|