1
|
Liu SL, Zhao P, Zhou YM, Peng ZG, Guo N, Sun HB, Cui XQ. IL-10 alleviates aTCMR by inhibiting NFATc1 signaling pathway of T cells after kidney transplantation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167857. [PMID: 40268206 DOI: 10.1016/j.bbadis.2025.167857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/28/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
The cause of acute T cell-mediated rejection (aTCMR) is believed to be immune hyperfunction of T cells after kidney transplantation. Nowadays, calcineurin inhibitors are widely used to inhibit the proliferation of T cells when aTCMR occurs. However, the therapeutic dose window of these drugs is relatively narrow and long time use of these drugs may lead to serious side effects. Besides, whether IL-10, a new immune tolerance mediator, playing a therapeutic role on aTCMR remains unclear. The level of IL-10 decreased in patients with aTCMR, suggesting that IL-10 may be involved in the progression of aTCMR. IL-10 could inhibit the proliferation and metabolism of T cells in vitro and in vivo, accompanied by reducing the levels of IL-2, IFN-γ, and TNF-α. Moreover, we confirmed that IL-10 exerts immunosuppressive effects by inhibiting the NFATc1 signaling pathway of T cells. This viewpoint may provide a new therapeutic idea for clinical application.
Collapse
Affiliation(s)
- Sheng-Li Liu
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China
| | - Peng Zhao
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China
| | - Yan-Man Zhou
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University. No.324, Jingwu Weiqi Road, Jinan 250021, Shandong, China
| | - Zhi-Guo Peng
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China
| | - Ning Guo
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China
| | - Huai-Bin Sun
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China.
| | - Xian-Quan Cui
- Department of organ transplantation, Qilu Hospital of Shandong University, No.107, Wenhua West Road, Jinan 250012, Shandong, China.
| |
Collapse
|
2
|
Udemgba C, Pillay B, Shafer S, Alberstadt A, Abers M, Gilliaux O, Chen K, Rae W, Hanitsch L, Von Bernuth H, Neves JF, Raje N, Moens L, van Hagen PM, Bergerson J, Hartog N, Niehues T, Dückers G, Falcone E, Keller M, Hsu A, Meyts I, Holland SM. IRF2BP2 deficiency: An important form of common variable immunodeficiency with inflammation. J Allergy Clin Immunol 2025:S0091-6749(25)00275-1. [PMID: 40090425 DOI: 10.1016/j.jaci.2025.02.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND IRF2BP2 is a transcription factor that plays an important role in regulating immune pathways, angiogenesis, apoptosis, and cell differentiation. Defects in this gene have been implicated in immunodeficiency. OBJECTIVES To deepen the understanding of the clinical implications of IRF2BP2 variants, we sought to clinically characterize and functionally test 34 individuals with IRF2BP2 variants. METHODS We collected 34 subjects across 18 families with mutations in IRF2BP2. Records were abstracted for clinical phenotypes. Functional testing was performed on PBMCs. NFAT luciferase gene reporter constructs and quantitative cDNA determinations were used to evaluate repressor activity associated with ectopic expression of various IRF2BP2 mutant constructs in Jurkat cells. RESULTS Most subjects had immunodeficiency (91%, n = 30 of 33) with variable gastrointestinal (65%, n = 20 of 31) and inflammatory or autoimmune features (57%, n = 17 of 30), including chronic abdominal pain, colitis, diarrhea, constipation, vitiligo, alopecia, and migratory rashes. There was a reduced frequency of memory B cells with poor immunoglobulin production and reduced calcium flux in response to B-cell receptor stimuli. PBMCs had increased apoptosis in vitro compared to healthy controls. Impaired IRF2BP2 repression of NFAT activation was observed using patient-derived mutant IRF2BP2 constructs compared to wild-type constructs. Similarly, TNF-α transcript levels were higher using patient-derived mutations compared to wild-type IRF2BP2 constructs. CONCLUSIONS IRF2BP2 deficiency causes a complex immunodeficiency including gastrointestinal and inflammatory disorders as well as impaired B-cell maturation. Impaired repression of the NFAT pathway appears to enhance proinflammatory signaling through proinflammatory cytokine expression.
Collapse
Affiliation(s)
- Chioma Udemgba
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Bethany Pillay
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Samantha Shafer
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Angelika Alberstadt
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael Abers
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Olivier Gilliaux
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Karin Chen
- Division of Immunology, Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Wash
| | - William Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Leif Hanitsch
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin Institute of Health at Charité, Berlin, Germany
| | - Horst Von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Joao Farela Neves
- Primary Immunodeficiencies Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal
| | - Nikita Raje
- Division of Allergy, Immunology, Pulmonology, and Sleep Medicine, Department of Pediatrics, Children's Mercy Kansas City, University of Missouri Kansas City, Washington, DC
| | - Leen Moens
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - P Martin van Hagen
- Division of Clinical Immunology, Department of Internal Medicine, Rotterdam, The Netherlands; Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jenna Bergerson
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Nicholas Hartog
- Michigan State University College of Human Medicine Corewell Health and Helen DeVos Children's Hospital, Grand Rapids, Mich
| | - Tim Niehues
- Centre for Child and Adolescent Health, Helios Klinikum Krefeld, affiliated with Rheinisch-Westfälische Technische Hochschule University Aachen, Aachen, Germany
| | - Gregor Dückers
- Centre for Child and Adolescent Health, Helios Klinikum Krefeld, affiliated with Rheinisch-Westfälische Technische Hochschule University Aachen, Aachen, Germany
| | - Emilia Falcone
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Michael Keller
- Division of Allergy and Immunology, Children's National Hospital, Washington, DC
| | - Amy Hsu
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium; Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Steven M Holland
- National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
3
|
Anft M, Zgoura P, Skrzypczyk S, Dürr M, Viebahn R, Westhoff TH, Stervbo U, Babel N. Effects of switching from twice-daily tacrolimus to once-daily extended-release meltdose tacrolimus on cellular immune response. FRONTIERS IN TRANSPLANTATION 2024; 3:1405070. [PMID: 39386200 PMCID: PMC11461451 DOI: 10.3389/frtra.2024.1405070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
Background LCP-Tacro [LCPT], a novel once-daily, extended-release formulation of tacrolimus, has a reduced Cmax with comparable AUC exposure, requiring a ∼30% dose reduction in contrast to immediate-release tacrolimus (IR-Tac). Once-daily LCPT in de novo kidney transplantation has a comparable efficacy and safety profile to that of IR-Tac with advantages in bioavailability and absorption. The present investigation intends to analyze the effects of conversion from IR-Tac to LCPT on phenotype and function of T-cells and B-cells. Methods 16 kidney transplant patients treated by triple standard immunosuppression with a stable graft function undergoing a switch from IR-Tac to LCPT were included in this observational prospective study. We measured the main immune cell types and performed an in-depth characterization of B cell, dendritic cells and T cells including regulatory T cells of the patients before, 4 and 8 weeks after IR-Tac to LCPT conversion using multi-parameter fl ow cytometry. Additionally, we analyzed T cells by assessing third-party antigens (Tetanus Diphtheria, TD)-reactive T cells, which could be analyzed by restimulation with tetanus vaccine. Results Overall, we found no significant alterations following LCPT conversion for the most immune cell populations with a few cell populations showing transient quantitative increase. Thus, 4 weeks after conversion, more regulatory T cells could be measured in the patients with a significant shift from memory to naïve Tregs. Furthermore, we found a transient B cell expansion 4 weeks after conversion from IR-Tac to LCPT. There were no changes in the percentage of other basic immune cell types and the antigen-reactive T cells were also not altered after changing the medication to LCP-tacrolimus. Conclusion Here, we demonstrate first insights into the immune system changes occurred under IR-Tac to LCPT conversion therapy in kidney transplant patients. While phenotypic and functional characteristics of the most immune cell populations did not change, we could observe an a transient expansion of regulatory T cells in peripheral blood following IR-Tac to LCTP conversion, which might additionally contribute to the overall immunosuppressive effect.
Collapse
Affiliation(s)
- Moritz Anft
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Panagiota Zgoura
- Clinic for Internal Medicine, St. Anna Hospital Herne, Herne, Germany
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Sarah Skrzypczyk
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Michael Dürr
- Clinic for Internal Medicine, St. Anna Hospital Herne, Herne, Germany
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
- Berlin Institute of Health, Berlin-BrandenburgCenter for Regenerative Therapies, and Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Augustenburger Platz, Berlin, Germany
| | - Richard Viebahn
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Timm H. Westhoff
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
- Berlin Institute of Health, Berlin-BrandenburgCenter for Regenerative Therapies, and Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Augustenburger Platz, Berlin, Germany
| |
Collapse
|
4
|
Venkatasubramanian D, Senevirathne G, Capellini TD, Craft AM. Leveraging single cell multiomic analyses to identify factors that drive human chondrocyte cell fate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598666. [PMID: 38915712 PMCID: PMC11195167 DOI: 10.1101/2024.06.12.598666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Cartilage plays a crucial role in skeletal development and function, and abnormal development contributes to genetic and age-related skeletal disease. To better understand how human cartilage develops in vivo , we jointly profiled the transcriptome and open chromatin regions in individual nuclei recovered from distal femurs at 2 fetal timepoints. We used these multiomic data to identify transcription factors expressed in distinct chondrocyte subtypes, link accessible regulatory elements with gene expression, and predict transcription factor-based regulatory networks that are important for growth plate or epiphyseal chondrocyte differentiation. We developed a human pluripotent stem cell platform for interrogating the function of predicted transcription factors during chondrocyte differentiation and used it to test NFATC2 . We expect new regulatory networks we uncovered using multiomic data to be important for promoting cartilage health and treating disease, and our platform to be a useful tool for studying cartilage development in vitro . Statement of Significance The identity and integrity of the articular cartilage lining our joints are crucial to pain-free activities of daily living. Here we identified a gene regulatory landscape of human chondrogenesis at single cell resolution, which is expected to open new avenues of research aimed at mitigating cartilage diseases that affect hundreds of millions of individuals world-wide.
Collapse
|
5
|
Yamasaki N, Miura K, Ogata S, Miura S, Uchimura A, Satoh Y, Toshishige M, Hosomi N, Gamboa M, Kitamura N, Kaminuma O. Generation of reporter mice for detecting the transcriptional activity of nuclear factor of activated T cells. Exp Anim 2023; 72:454-459. [PMID: 37100620 PMCID: PMC10658084 DOI: 10.1538/expanim.23-0043] [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: 03/23/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a transcription factor essential for immunological and other biological responses. To develop analyzing system for NFAT activity in vitro and in vivo, we generated reporter mouse lines introduced with NFAT-driven enhanced green fluorescent protein (EGFP) expressing gene construct. Six tandem repeats of -286 to -265 of the human IL2 gene to which NFAT binds in association with its co-transcription factor, activator protein (AP)-1, was conjunct with thymidine kinase minimum promoter and following EGFP coding sequence. Upon introduction of the resulting reporter cassette into C57BL/6 fertilized eggs, the transgenic mice were obtained. Among 7 transgene-positive mice in 110 mice bone, 2 mice showed the designated reporter mouse character. Thus, the EGFP fluorescence of CD4+ and CD8+ T cells in these mice was enhanced by stimulation through CD3 and CD28. Each of phorbol 12-myristate 13-acetate (PMA) and ionomycin (IOM) stimulation weakly but their combined stimulation strongly enhanced EGFP expression. The stimulation-induced EGFP upregulation was also observed following T cell subset differentiation in a different manner. The EGFP induction by PMA + IOM stimulation was more potent than that by CD3/CD28 stimulation in helper T (Th)1, Th2, Th9, and regulatory T cells, while both stimulation conditions displayed the equivalent EGFP induction in Th17 cells. Our NFAT reporter mouse lines are useful for analyzing stimulation-induced transcriptional activation mediated by NFAT in cooperation with AP-1 in T cells.
Collapse
Affiliation(s)
- Norimasa Yamasaki
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kento Miura
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Sawako Ogata
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Shuka Miura
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Arikuni Uchimura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima 732-0815, Japan
| | - Yasunari Satoh
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima 732-0815, Japan
| | - Masaaki Toshishige
- Department of Molecular Biosciences, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami-ku, Hiroshima 732-0815, Japan
| | - Naohisa Hosomi
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Maribet Gamboa
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
- Department of Ecology, Faculty of Sciences, Universidad Católica de la Santísima Concepción, Concepción, Avenue. Alonso de Ribera 2850, Concepción, Bío Bío 4090541, Chile
| | - Noriko Kitamura
- Neurovirology Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Osamu Kaminuma
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
6
|
Son GY, Tu NH, Santi MD, Lopez SL, Souza Bomfim GH, Vinu M, Zhou F, Chaloemtoem A, Alhariri R, Idaghdour Y, Khanna R, Ye Y, Lacruz RS. The Ca 2+ channel ORAI1 is a regulator of oral cancer growth and nociceptive pain. Sci Signal 2023; 16:eadf9535. [PMID: 37669398 PMCID: PMC10747475 DOI: 10.1126/scisignal.adf9535] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/15/2023] [Indexed: 09/07/2023]
Abstract
Oral cancer causes pain associated with cancer progression. We report here that the function of the Ca2+ channel ORAI1 is an important regulator of oral cancer pain. ORAI1 was highly expressed in tumor samples from patients with oral cancer, and ORAI1 activation caused sustained Ca2+ influx in human oral cancer cells. RNA-seq analysis showed that ORAI1 regulated many genes encoding oral cancer markers such as metalloproteases (MMPs) and pain modulators. Compared with control cells, oral cancer cells lacking ORAI1 formed smaller tumors that elicited decreased allodynia when inoculated into mouse paws. Exposure of trigeminal ganglia neurons to MMP1 evoked an increase in action potentials. These data demonstrate an important role of ORAI1 in oral cancer progression and pain, potentially by controlling MMP1 abundance.
Collapse
Affiliation(s)
- Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
| | - Nguyen Huu Tu
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
| | - Maria Daniela Santi
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
| | - Santiago Loya Lopez
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | | | - Manikandan Vinu
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Fang Zhou
- Department of Pathology, New York University Langone Health, New York, NY 10010
| | - Ariya Chaloemtoem
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Rama Alhariri
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | - Yi Ye
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
| |
Collapse
|
7
|
Lainšček D, Golob-Urbanc A, Mikolič V, Pantović-Žalig J, Malenšek Š, Jerala R. Regulation of CD19 CAR-T cell activation based on an engineered downstream transcription factor. Mol Ther Oncolytics 2023; 29:77-90. [PMID: 37223115 PMCID: PMC10200817 DOI: 10.1016/j.omto.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/24/2023] [Indexed: 05/25/2023] Open
Abstract
CAR-T cells present a highly effective therapeutic option for several malignant diseases, based on their ability to recognize the selected tumor surface marker in an MHC-independent manner. This triggers cell activation and cytokine production, resulting in the killing of the cancerous cell presenting markers recognized by the chimeric antigen receptor. CAR-T cells are highly potent serial killers that may cause serious side effects, so their activity needs to be carefully controlled. Here we designed a system to control the proliferation and activation state of CARs based on downstream NFAT transcription factors, whose activity can be regulated via chemically induced heterodimerization systems. Chemical regulators were used to either transiently trigger engineered T cell proliferation or suppress CAR-mediated activation when desired or to enhance activation of CAR-T cells upon engagement of cancer cells, shown also in vivo. Additionally, an efficient sensor to monitor activated CD19 CAR-T cells in vivo was introduced. This implementation in CAR-T cell regulation offers an efficient way for on-demand external control of CAR-T cell activity to improve their safety.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| | - Anja Golob-Urbanc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
| | - Veronika Mikolič
- Department of Hematology, Division of Internal Medicine, University Medical Center Ljubljana, Zaloška 7, Ljubljana 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Jelica Pantović-Žalig
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| |
Collapse
|
8
|
Kim W, Kim HJ, Trinh NT, Yeon HR, Kim JH, Choi IA, Kim HA, Jung JY, Lee KE. Association between nuclear factor of activated T cells C2 polymorphisms and treatment response in rheumatoid arthritis patients receiving tumor necrosis factor-alpha inhibitors. Pharmacogenet Genomics 2022; 32:10-15. [PMID: 34320607 DOI: 10.1097/fpc.0000000000000446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Nuclear factor of activated T cells C2 (NFATC2) is known as a member of the transcription family and enhances tumor necrosis factor-alpha (TNF-α) synthesis in human T cells at the gene transcription level. Although NFATC2 has a potential role in rheumatoid arthritis (RA) progression and treatment, no study has investigated the association between NFATC2 gene polymorphisms and response status in RA patients receiving TNF-α inhibitors. This study aimed to examine the effects of polymorphisms in NFATC2, a TNF-α transcription factor, on response to TNF-α inhibitors. METHODS This prospective observational study was performed in two centers. Seven single nucleotide polymorphisms (SNPs) were investigated. Good responders were defined as patients with disease activity score (DAS)28 ≤3.2 after 6 months of treatment. Logistic regression analyses were used to investigate the association between genetic polymorphisms and response to the treatment. To test the model's goodness of fit, a Hosmer-Lemeshow test was performed. RESULTS This study included 98 patients, among whom 46 showed favorable responses to the treatment. Patients with hypertension revealed an approximately three-fold lower response to TNF-α inhibitors compared to those without hypertension (23.5 vs. 76.5%; P = 0.049). After adjusting for covariates, C allele carriers of NFATC2 rs3787186 exhibited approximately three-fold lower rates of treatment response compared to those with TT genotype (P = 0.037). The Hosmer-Lemeshow test showed that the fitness of the multivariable analysis model was satisfactory (χ2 = 9.745; 8 degrees of freedom; P = 0.283). CONCLUSION This study suggested an association between the C allele of rs3787186 and treatment response in RA patients receiving TNF-α inhibitors.
Collapse
Affiliation(s)
- Woorim Kim
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Hyun Jeong Kim
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Nga Thi Trinh
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Ha Rim Yeon
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Joo Hee Kim
- College of Pharmacy, Ajou University, Worldcup-ro, Yeongtong-gu, Suwon
| | - In Ah Choi
- Division of Rheumatology, Department of Internal Medicine, Chungbuk National University Hospital, 1sunhwan-ro, Seowon-gu, Cheongju
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| | - Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| | - Kyung Eun Lee
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| |
Collapse
|
9
|
Kubota H, Masuda T, Noura M, Furuichi K, Matsuo H, Hirata M, Kataoka TR, Hiramatsu H, Yasumi T, Nakahata T, Imai Y, Takita J, Adachi S, Sugiyama H, Kamikubo Y. RUNX inhibitor suppresses graft-versus-host disease through targeting RUNX-NFATC2 axis. EJHAEM 2021; 2:449-458. [PMID: 35844683 PMCID: PMC9175814 DOI: 10.1002/jha2.230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022]
Abstract
Patients with refractory graft-versus-host disease (GVHD) have a dismal prognosis. Therefore, novel therapeutic targets are still needed to be identified. Runt-related transcriptional factor (RUNX) family transcription factors are essential transcription factors that mediate the essential roles in effector T cells. However, whether RUNX targeting can suppress, and GVHD is yet unknown. Here, we showed that RUNX family members have a redundant role in directly transactivating NFATC2 expression in T cells. We also found that our novel RUNX inhibitor, Chb-M', which is the inhibitor that switches off the entire RUNX family by alkylating agent-conjugated pyrrole-imidazole (PI) polyamides, inhibited T-cell receptor mediated T cell proliferation and allogenic T cell response. These were designed to specifically bind to consensus RUNX-binding sequences (TGTGGT). Chb-M' also suppressed the expression of NFATC2 and pro-inflammatory cytokine genes in vitro. Using xenogeneic GVHD model, mice injected by Chb-M' showed almost no sign of GVHD. Especially, the CD4 T cell was decreased and GVHD-associated cytokines including tissue necrosis factor-α and granulocyte-macrophage colony-stimulating factor were reduced in the peripheral blood of Chb-M' injected mice. Taken together, our data demonstrates that RUNX family transcriptionally upregulates NFATC2 in T cells, and RUNX-NFATC2 axis can be a novel therapeutic target against GVHD.
Collapse
Affiliation(s)
- Hirohito Kubota
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Tatsuya Masuda
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Mina Noura
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Kana Furuichi
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Hidemasa Matsuo
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Masahiro Hirata
- Department of Diagnostic PathologyKyoto University HospitalSakyo‐kuKyotoJapan
| | - Tatsuki R. Kataoka
- Department of Diagnostic PathologyKyoto University HospitalSakyo‐kuKyotoJapan
| | - Hidefumi Hiramatsu
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Takahiro Yasumi
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Tatsutoshi Nakahata
- Drug Discovery Technology Development OfficeCenter for iPS cell research and application (CiRA)Kyoto UniversitySakyo‐kuKyotoJapan
| | - Yoichi Imai
- Department of Hematology/OncologyIMSUT HospitalThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Junko Takita
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Souichi Adachi
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Hiroshi Sugiyama
- Department of ChemistryGraduate School of ScienceKyoto UniversitySakyo‐kuKyotoJapan
| | - Yasuhiko Kamikubo
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| |
Collapse
|
10
|
Kitamura N, Kaminuma O. Isoform-Selective NFAT Inhibitor: Potential Usefulness and Development. Int J Mol Sci 2021; 22:2725. [PMID: 33800389 PMCID: PMC7962815 DOI: 10.3390/ijms22052725] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT), which is the pharmacological target of immunosuppressants cyclosporine and tacrolimus, has been shown to play an important role not only in T cells (immune system), from which their name is derived, but also in many biological events. Therefore, functional and/or structural abnormalities of NFAT are linked to the pathogenesis of diseases in various organs. The NFAT protein family consists of five isoforms, and each isoform performs diverse functions and has unique expression patterns in the target tissues. This diversity has made it difficult to obtain ideal pharmacological output for immunosuppressants that inhibit the activity of almost all NFAT family members, causing serious and wide-ranging side effects. Moreover, it remains unclear whether isoform-selective NFAT regulation can be achieved by targeting the structural differences among NFAT isoforms and whether this strategy can lead to the development of better drugs than the existing ones. This review summarizes the role of the NFAT family members in biological events, including the development of various diseases, as well as the usefulness of and problems associated with NFAT-targeting therapies, including those dependent on current immunosuppressants. Finally, we propose a novel therapeutic strategy based on the molecular mechanisms that enable selective regulation of specific NFAT isoforms.
Collapse
Affiliation(s)
- Noriko Kitamura
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Osamu Kaminuma
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
11
|
Cai SY, Yu D, Soroka CJ, Wang J, Boyer JL. Hepatic NFAT signaling regulates the expression of inflammatory cytokines in cholestasis. J Hepatol 2021; 74:550-559. [PMID: 33039404 PMCID: PMC7897288 DOI: 10.1016/j.jhep.2020.09.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS The nuclear factor of activated T-cells (NFAT) plays an important role in immune responses by regulating the expression of inflammatory genes. However, it is not known whether NFAT plays any role in the bile acid (BA)-induced hepatic inflammatory response. Thus, we aimed to examine the functional role of NFATc3 in cholestatic liver injury in mice and humans. METHODS Gene and protein expression and cellular localization were assessed in primary hepatocyte cultures (mouse and human) and cholestatic liver tissues (murine models and patients with primary biliary cholangitis [PBC] or primary sclerosing cholangitis [PSC]) by quantitative PCR, western blot and immunohistochemistry. Specific NFAT inhibitors were used in vivo and in vitro. Gene reporter assays and ChIP-PCR were used to determine promoter activity. RESULTS NFAT isoforms c1 and c3 were expressed in human and mouse hepatocytes. When treated with cholestatic levels of BAs, nuclear translocation of NFATc3 was increased in both human and mouse hepatocytes and was associated with elevated mRNA levels of IL-8, CXCL2, and CXCL10 in these cells. Blocking NFAT activation with pathway-specific inhibitors or knocking down Nfatc3 expression significantly decreased BA-driven induction of these cytokines in mouse hepatocytes. Nuclear expression of NFATc3/Nfatc3 protein was increased in cholestatic livers, both in mouse models (bile duct ligation or Abcb4-/- mice) and in patients with PBC and PSC in association with elevated tissue levels of Cxcl2 (mice) or IL-8 (humans). Gene reporter assays and ChIP-PCR demonstrated that the NFAT response element in the IL-8 promoter played a key role in BA-induced human IL-8 expression. Finally, blocking NFAT activation in vivo in Abcb4-/- mice reduced cholestatic liver injury. CONCLUSIONS NFAT plays an important role in BA-stimulated hepatic cytokine expression in cholestasis. Blocking hepatic NFAT activation may reduce cholestatic liver injury in humans. LAY SUMMARY Bile acid induces liver injury by stimulating the expression of inflammatory genes in hepatocytes through activation of the transcription factor NFAT. Blocking this activation in vitro (in hepatocyte cultures) and in vivo (in cholestatic mice) decreased the expression of inflammatory genes and reduced liver injury.
Collapse
Affiliation(s)
- Shi-Ying Cai
- Department of Internal Medicine, Liver Center, Yale University School of Medicine, New Haven, CT 06520.
| | - Dongke Yu
- Department of Internal Medicine, Liver Center, Yale University School of Medicine, New Haven, CT 06520
| | - Carol J Soroka
- Department of Internal Medicine, Liver Center, Yale University School of Medicine, New Haven, CT 06520
| | - Jing Wang
- Department of Internal Medicine, Liver Center, Yale University School of Medicine, New Haven, CT 06520
| | - James L Boyer
- Department of Internal Medicine, Liver Center, Yale University School of Medicine, New Haven, CT 06520.
| |
Collapse
|
12
|
Cho J, Park K, Choi SM, Lee J, Lee CH, Lee JK, Heo EY, Kim DK, Lee YJ, Park JS, Cho YJ, Yoon HI, Lee JH, Lee CT, Kim N, Choi KY, Lee KH, Sung J, Won S, Yim JJ. Genome-wide association study of non-tuberculous mycobacterial pulmonary disease. Thorax 2020; 76:169-177. [PMID: 33115937 DOI: 10.1136/thoraxjnl-2019-214430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 08/25/2020] [Accepted: 09/23/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND The prevalence of non-tuberculous mycobacterial pulmonary disease (NTM-PD) is increasing in South Korea and many parts of the world. However, the genetic factors underlying susceptibility to this disease remain elusive. METHODS To identify genetic variants in patients with NTM-PD, we performed a genome-wide association study with 403 Korean patients with NTM-PD and 306 healthy controls from the Healthy Twin Study, Korea cohort. Candidate variants from the discovery cohort were subsequently validated in an independent cohort. The Genotype-Tissue Expression (GTEx) database was used to identify expression quantitative trait loci (eQTL) and to conduct Mendelian randomisation (MR). RESULTS We identified a putatively significant locus on chromosome 7p13, rs849177 (OR, 2.34; 95% CI, 1.71 to 3.21; p=1.36×10-7), as the candidate genetic variant associated with NTM-PD susceptibility. Its association was subsequently replicated and the combined p value was 4.92×10-8. The eQTL analysis showed that a risk allele at rs849177 was associated with lower expression levels of STK17A, a proapoptotic gene. In the MR analysis, a causal effect of STK17A on NTM-PD development was identified (β, -4.627; 95% CI, -8.768 to -0.486; p=0.029). CONCLUSIONS The 7p13 genetic variant might be associated with susceptibility to NTM-PD in the Korean population by altering the expression level of STK17A.
Collapse
Affiliation(s)
- Jaeyoung Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of)
| | - Kyungtaek Park
- Interdisciplinary Program of Bioinformatics, Seoul National University College of Natural Sciences, Seoul, Korea (the Republic of)
| | - Sun Mi Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of)
| | - Jinwoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of).,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Chang-Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of)
| | - Jung-Kyu Lee
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Eun Young Heo
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Deog Kyeom Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Yeon Joo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Jong Sun Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Ho Il Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Jae Ho Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Choon-Taek Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of).,Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea (the Republic of)
| | - Kyu Yeong Choi
- Gwangju Alzheimer's disease and Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea (the Republic of)
| | - Kun Ho Lee
- Gwangju Alzheimer's disease and Related Dementia Cohort Research Center, Chosun University, Gwangju, Korea (the Republic of).,Department of Biomedical Science, Chosun University, Gwangju, Korea (the Republic of).,Aging Neuroscience Research Group, Korea Brain Research Institute, Daegu, Korea (the Republic of)
| | - Joohon Sung
- Department of Public Health Sciences, Seoul National University Graduate School of Public Health, Seoul, Korea (the Republic of).,Seoul National University Institute of Health and Environment, Seoul, Korea (the Republic of)
| | - Sungho Won
- Interdisciplinary Program of Bioinformatics, Seoul National University College of Natural Sciences, Seoul, Korea (the Republic of) .,Department of Public Health Sciences, Seoul National University Graduate School of Public Health, Seoul, Korea (the Republic of).,Seoul National University Institute of Health and Environment, Seoul, Korea (the Republic of)
| | - Jae-Joon Yim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of) .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
13
|
Zhou CJ, Mo LH, Luo XQ, Yang G, Diao WL, Hu SP, Yang PC. Identification of antigen-specific neutrophils in the tonsils with recurrent acute inflammation. Autoimmunity 2020; 53:237-244. [PMID: 32525408 DOI: 10.1080/08916934.2020.1777284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Cai-Jie Zhou
- Beijing University of Chinese Medical Shenzhen Hospital, Shenzhen, China
| | - Li-Hua Mo
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiang-Qian Luo
- Department of Pediatric Otolaryngology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Wei-Liang Diao
- Beijing University of Chinese Medical Shenzhen Hospital, Shenzhen, China
| | - Shi-Ping Hu
- Beijing University of Chinese Medical Shenzhen Hospital, Shenzhen, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
| |
Collapse
|
14
|
Kitamura N, Shindo M, Ohtsuka J, Nakamura A, Tanokura M, Hiroi T, Kaminuma O. Identification of novel interacting regions involving calcineurin and nuclear factor of activated T cells. FASEB J 2020; 34:3197-3208. [PMID: 31909857 DOI: 10.1096/fj.201902229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
Nuclear factor of activated T cells (NFAT) leads to the transcription of diverse inducible genes involved in many biological processes; therefore, aberrant NFAT expression is responsible for the development and exacerbation of various disorders. Since five isoforms of NFAT (NFATc1-c4, NFAT5) exhibit distinct and overlapping functions, selective control of a part, but not all, of NFAT family members is desirable. By comparing the binding activity of each NFATc1-c4 with its regulatory enzyme, calcineurin (CN), using a quantitative immunoprecipitation assay, we found a new CN-binding region (CNBR) selectively functioning in NFATc1 and NFATc4. This region, termed CNBR3, is located between two preexisting CNBR1 and CNBR2, within the Ca2+ regulatory domain. The nuclear translocation of NFATc1 but not NFATc2 in T cells was suppressed by ectopic expression of CNBR3 and, accordingly, NFATc1-dependent cytokine expression was downregulated. Through competition assays using NFATc1-derived partial peptides and mass spectrometry with photoaffinity technology, we identified 18 amino acids in NFATc1 (Arg258 to Pro275 ) and 13 amino acids in CN catalytic subunit (CNA) (Asn77 to Gly89 ) responsible for CNA/CNBR3 binding in which Cys263 and Asp82 , respectively, played crucial roles. The possible selective regulation of NFAT-mediated biological processes by targeting this new CN/NFAT-binding region is suggested.
Collapse
Affiliation(s)
- Noriko Kitamura
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mayumi Shindo
- Center for Basic Technology Research, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jun Ohtsuka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Akira Nakamura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.,Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Osamu Kaminuma
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
15
|
Cowman SA, Jacob J, Hansell DM, Kelleher P, Wilson R, Cookson WOC, Moffatt MF, Loebinger MR. Whole-Blood Gene Expression in Pulmonary Nontuberculous Mycobacterial Infection. Am J Respir Cell Mol Biol 2019; 58:510-518. [PMID: 29206475 DOI: 10.1165/rcmb.2017-0230oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The factors predisposing toward the development of pulmonary nontuberculous mycobacterial (pNTM) disease and influencing disease progression remain unclear. Impaired immune responses have been reported in individuals with pNTM disease, but data are limited and inconsistent. In this study, we sought to use gene expression profiling to examine the host response to pNTM disease. Microarray analysis of whole-blood gene expression was performed on 25 subjects with pNTM disease and 27 uninfected control subjects with respiratory disease. Gene expression results were compared with phenotypic variables and survival data. Compared with uninfected control subjects, pNTM disease was associated with downregulation of 213 transcripts enriched for terms related to T cell signaling, including IFNG. Reduced IFNG expression was associated with more severe computed tomography changes and impaired lung function. Mortality was associated with the expression of transcripts related to the innate immune response and inflammation, whereas transcripts related to T and B cell function were associated with improved survival. These findings suggest that pNTM disease is associated with an aberrant immune response, which may reflect an underlying propensity to infection or result from NTM infection itself. There were important differences in the immune response associated with survival and mortality in pNTM disease.
Collapse
Affiliation(s)
- Steven A Cowman
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Joseph Jacob
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - David M Hansell
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,3 Department of Radiology, Royal Brompton Hospital, London, United Kingdom
| | - Peter Kelleher
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - Robert Wilson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| | - William O C Cookson
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Miriam F Moffatt
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Michael R Loebinger
- 1 National Heart and Lung Institute, Imperial College London, London, United Kingdom.,2 Host Defence Unit and
| |
Collapse
|
16
|
Malsy M, Graf B, Almstedt K. The active role of the transcription factor Sp1 in NFATc2-mediated gene regulation in pancreatic cancer. BMC BIOCHEMISTRY 2019; 20:2. [PMID: 30696421 PMCID: PMC6352339 DOI: 10.1186/s12858-019-0105-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 01/09/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adenocarcinoma of the pancreas is one of the most aggressive tumor diseases affecting the human body. The oncogenic potential of pancreatic cancer is mainly characterized by extremely rapid growth triggered by the activation of oncogenic signaling cascades, which suggests a change in the regulation of important transcription factors. Amongst others, NFAT transcription factors are assumed to play a central role in the carcinogenesis of pancreatic cancer. Recent research has shown the importance of the transcription factor Sp1 in the transcriptional activity of NFATc2 in pancreatic cancer. However, the role of the interaction between these two binding partners remains unclear. The current study investigated the role of Sp1 proteins in the expression of NFATc2 target genes and identified new target genes and their function in cells. A further objective was the domain of the Sp1 protein that mediates interaction with NFATc2. The involvement of Sp1 proteins in NFATc2 target genes was shown by means of a gene expression profile analysis, and the results were confirmed by quantitative RT-PCR. The functional impact of this interaction was shown in a thymidine incorporation assay. A second objective was the physical interaction between NFATc2 and different Sp1 deletion mutants that was investigated by means of immunoprecipitation. RESULTS In pancreatic cancer, the proto-oncogene c-Fos, the tumor necrosis factor TNF-alpha, and the adhesion molecule integrin beta-3 are target genes of the interaction between Sp1 and NFATc2. Loss of just one transcription factor inhibits oncogenic complex formation and expression of cell cycle-regulating genes, thus verifiably decreasing the carcinogenic effect. The current study also showed the interaction between the transcription factor NFATc2 and the N-terminal domain of Sp1 in pancreatic cancer cells. Sp1 increases the activity of NFATc2 in the NFAT-responsive promoter. CONCLUSIONS The regulation of gene promotors during transcription is a rather complex process because of the involvement of many proteins that - as transcription factors or co-factors - regulate promotor activity as required and control cell function. NFATc2 and Sp1 seem to play a key role in the progression of pancreatic cancer.
Collapse
Affiliation(s)
- Manuela Malsy
- Department of Anesthesiology, University Medical Center, Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | - Bernhard Graf
- Department of Anesthesiology, University Medical Center, Regensburg, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | - Katrin Almstedt
- Department of Obstetrics and Gynecology, University Hospital, Mainz, Mainz, Germany
| |
Collapse
|
17
|
Veytia-Bucheli JI, Jiménez-Vargas JM, Melchy-Pérez EI, Sandoval-Hernández MA, Possani LD, Rosenstein Y. K v1.3 channel blockade with the Vm24 scorpion toxin attenuates the CD4 + effector memory T cell response to TCR stimulation. Cell Commun Signal 2018; 16:45. [PMID: 30107837 PMCID: PMC6092819 DOI: 10.1186/s12964-018-0257-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/02/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND In T cells, the Kv1.3 and the KCa3.1 potassium channels regulate the membrane potential and calcium homeostasis. Notably, during TEM cell activation, the number of Kv1.3 channels on the cell membrane dramatically increases. Kv1.3 blockade results in inhibition of Ca2+ signaling in TEM cells, thus eliciting an immunomodulatory effect. Among the naturally occurring peptides, the Vm24 toxin from the Mexican scorpion Vaejovis mexicanus is the most potent and selective Kv1.3 channel blocker known, which makes it a promissory candidate for its use in the clinic. We have shown that addition of Vm24 to TCR-activated human T cells inhibits CD25 expression, cell proliferation and reduces delayed-type hypersensitivity reactions in a chronic inflammation model. Here, we used the Vm24 toxin as a tool to investigate the molecular events that follow Kv1.3 blockade specifically on human CD4+ TEM cells as they are actively involved in inflammation and are key mediators of autoimmune diseases. METHODS We combined cell viability, activation, and multiplex cytokine assays with a proteomic analysis to identify the biological processes affected by Kv1.3 blockade on healthy donors CD4+ TEM cells, following TCR activation in the presence or absence of the Vm24 toxin. RESULTS The peptide completely blocked Kv1.3 channels currents without impairing TEM cell viability, and in response to TCR stimulation, it inhibited the expression of the activation markers CD25 and CD40L (but not that of CD69), as well as the secretion of the pro-inflammatory cytokines IFN-γ and TNF and the anti-inflammatory cytokines IL-4, IL-5, IL-9, IL-10, and IL-13. These results, in combination with data from the proteomic analysis, indicate that the biological processes most affected by the blockade of Kv1.3 channels in a T cell activation context were cytokine-cytokine receptor interaction, mRNA processing via spliceosome, response to unfolded proteins and intracellular vesicle transport, targeting the cell protein synthesis machinery. CONCLUSIONS The Vm24 toxin, a highly specific inhibitor of Kv1.3 channels allowed us to define downstream functions of the Kv1.3 channels in human CD4+ TEM lymphocytes. Blocking Kv1.3 channels profoundly affects the mRNA synthesis machinery, the unfolded protein response and the intracellular vesicle transport, impairing the synthesis and secretion of cytokines in response to TCR engagement, underscoring the role of Kv1.3 channels in regulating TEM lymphocyte function.
Collapse
Affiliation(s)
- José Ignacio Veytia-Bucheli
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
- Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juana María Jiménez-Vargas
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
| | - Erika Isabel Melchy-Pérez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
| | - Monserrat Alba Sandoval-Hernández
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
- Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourival Domingos Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Av. Universidad 2001, Col. Chamilpa, 62210 Cuernavaca, Morelos Mexico
| |
Collapse
|
18
|
Greenland JR, Chong T, Wang AS, Martinez E, Shrestha P, Kukreja J, Hays SR, Golden JA, Singer JP, Tang Q. Suppressed calcineurin-dependent gene expression identifies lung allograft recipients at increased risk of infection. Am J Transplant 2018; 18:2043-2049. [PMID: 29673076 PMCID: PMC6699504 DOI: 10.1111/ajt.14886] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/21/2018] [Accepted: 04/08/2018] [Indexed: 01/25/2023]
Abstract
Lung transplant immunosuppression regimens generally include the calcineurin inhibitor tacrolimus. We hypothesized that mean residual expression (MRE) of calcineurin-dependent genes assesses rejection and infection risk better than does tacrolimus trough. We prospectively followed 44 lung allograft recipients at 2 to 18 months posttransplant and measured changes in whole blood interleukin-2, interferon-γ, and granulocyte-macrophage colony-stimulating factor gene expression following a tacrolimus dose. Posttransplant duration, immunosuppressive medication levels, and bronchoscopic rejection and infection assessments were compared with MRE by using generalized-estimating equation-adjusted models. Prednisolone effect on MRE was assessed ex vivo in blood samples from nontransplanted controls. Tacrolimus concentration inhibiting 50% of cytokine production (IC50 ) was measured in a pretransplant subset. Results showed that MRE did not change with diagnosis of rejection but that airway infection was associated with a 20% absolute decrease (95% confidence interval 11%-29%). MRE increased with time after transplant but was not associated with tacrolimus trough. Interestingly, MRE correlated inversely with corticosteroid dose in the study cohort and ex vivo. Pretransplant tacrolimus IC50 depended on the cytokine measured and varied between individuals, suggesting a range in baseline responses to tacrolimus. We conclude that MRE identifies infection risk in lung allograft recipients, potentially integrating calcineurin inhibitor and steroid effects on lymphocyte effector function.
Collapse
Affiliation(s)
- John R Greenland
- Medical Service, Veterans Affairs Health Care System, San Francisco CA, 94121,Department of Medicine, University of California, San Francisco CA, 94143,Corresponding author:
| | - Tiffany Chong
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Angelia S Wang
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Emily Martinez
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Pavan Shrestha
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco CA, 94143
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Jeffrey A Golden
- Department of Medicine, University of California, San Francisco CA, 94143,Department of Surgery, University of California, San Francisco CA, 94143
| | - Jonathan P Singer
- Department of Medicine, University of California, San Francisco CA, 94143
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco CA, 94143
| |
Collapse
|
19
|
Alam MS, Gaida MM, Debnath S, Tagad HD, Miller Jenkins LM, Appella E, Rahman MJ, Ashwell JD. Unique properties of TCR-activated p38 are necessary for NFAT-dependent T-cell activation. PLoS Biol 2018; 16:e2004111. [PMID: 29357353 PMCID: PMC5794172 DOI: 10.1371/journal.pbio.2004111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/01/2018] [Accepted: 01/08/2018] [Indexed: 01/10/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) transcription factors are required for induction of T-cell cytokine production and effector function. Although it is known that activation via the T-cell antigen receptor (TCR) results in 2 critical steps, calcineurin-mediated NFAT1 dephosphorylation and NFAT2 up-regulation, the molecular mechanisms underlying each are poorly understood. Here we find that T cell p38, which is activated by an alternative pathway independent of the mitogen-activated protein (MAP) kinase cascade and with different substrate specificities, directly controls these events. First, alternatively (but not classically) activated p38 was required to induce the expression of the AP-1 component c-Fos, which was necessary for NFAT2 expression and cytokine production. Second, alternatively (but not classically) activated p38 phosphorylated NFAT1 on a heretofore unidentified site, S79, and in its absence NFAT1 was unable to interact with calcineurin or migrate to the nucleus. These results demonstrate that the acquisition of unique specificities by TCR-activated p38 orchestrates NFAT-dependent T-cell functions. The p38 MAP kinase, which is required for a large number of important biological responses, is activated by an enzymatic cascade that results in its dual phosphorylation on p38T180Y182. T cells have evolved a unique pathway in which T-cell antigen receptor (TCR) ligation results in phosphorylation of p38Y323 (the alternative pathway). Why T cells acquired this pathway is the subject of conjecture. In this study, we examine the activation of 2 members of the nuclear factor of activated T cells (NFAT) family, which, when dephosphorylated by calcineurin, migrate from the cytoplasm to the nucleus. In T cells with the alternative pathway ablated by a single amino acid substitution (p38Y323F), NFAT1 remained in the cytoplasm after stimulation via the TCR. Studies identified NFAT1S79 as a target for alternatively (but not classically) activated p38, and phosphorylation of this residue was required for binding calcineurin and nuclear translocation. Furthermore, although classically activated p38 induced NFAT1 translocation in the absence of NFAT1S79 phosphorylation, unlike alternatively activated p38 it did not cause NFAT2 up-regulation. This paradox was resolved by the finding that only the latter induces c-Fos, which binds to the NFAT2 promoter and participates in its up-regulation. These T-cell-specific p38 activities provide a strong rationale for the acquisition of the alternative mechanism for activating p38.
Collapse
Affiliation(s)
- Muhammad S. Alam
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthias M. Gaida
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Subrata Debnath
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harichandra D. Tagad
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lisa M. Miller Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - M. Jubayer Rahman
- Laboratory of Molecular Immunology at the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
20
|
Kaminuma O, Kitamura N, Nishito Y, Nemoto S, Tatsumi H, Mori A, Hiroi T. Downregulation of NFAT3 Due to Lack of T-Box Transcription Factor TBX5 Is Crucial for Cytokine Expression in T Cells. THE JOURNAL OF IMMUNOLOGY 2017; 200:92-100. [PMID: 29180489 DOI: 10.4049/jimmunol.1602113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 10/19/2017] [Indexed: 11/19/2022]
Abstract
The NFAT family transcription factors play crucial roles in immunological and other biological activities. NFAT3 is rarely expressed in T cells, and the mechanisms and significance of the specific NFAT3 downregulation in T cells have been unknown. In human CD4+ T cells, overexpression of NFAT1 and NFAT3 enhanced and suppressed IL-2 expression, respectively. NFAT3 downregulation in Jurkat cells using RNA interference technology augmented IL-2 expression, whereas a knockdown of NFAT1, NFAT2, and NFAT4 suppressed it. The promoter/enhancer activity of the NFAT-binding site in the IL-2 gene was upregulated and downregulated by NFAT1 and NFAT3, respectively. A study employing NFAT1/NFAT3 chimeric molecules revealed that the region in NFAT3 responsible for NFAT promoter activity inhibition was located within its N-terminal transactivation domain, Ca2+-regulatory domain, and DNA-binding domain. Downregulation of NFAT3 expression in T cells is mediated by lower chromatin accessibility and enhancer activity in its promoter in comparison with aortic smooth muscle cells expressing endogenous NFAT3. The binding sites of T-box transcription factor TBX5 and NK-2 transcription factor-related locus 5 Nkx2.5, which were expressed at higher levels in aortic smooth muscle cells than in T cells, were located within the -387 to +97 NFAT3 promoter region, exhibiting the maximum enhancer activity. Mutating the binding site of TBX5 but not Nkx2.5 diminished the NFAT3 promoter activity, whereas the overexpression of TBX5 enhanced it. Introduction of TBX5 into CD4+ T cells enhanced the expression of NFAT3 and suppressed that of IL-2. TBX5 deficiency-mediated downregulation of NFAT3 is crucial for the high cytokine-producing activity of T cells.
Collapse
Affiliation(s)
- Osamu Kaminuma
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan; .,Center for Life Science Research, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Noriko Kitamura
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Soichi Nemoto
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Hideki Tatsumi
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Akio Mori
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| |
Collapse
|
21
|
Rasooly R, Do PM, Hernlem BJ. Rapid Cell-Based Assay for Detection and Quantification of Active Staphylococcal Enterotoxin Type D. J Food Sci 2017; 82:718-723. [PMID: 28135403 DOI: 10.1111/1750-3841.13634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/28/2016] [Accepted: 12/29/2016] [Indexed: 11/29/2022]
Abstract
Food poisoning by Staphylococcus aureus is a result of ingestion of Staphylococcal enterotoxins (SEs) produced by this bacterium and is a major source of foodborne illness. Staphylococcal enterotoxin D (SED) is one of the predominant enterotoxins recovered in Staphylococcal food poisoning incidences, including a recent outbreak in Guam affecting 300 children. Current immunology methods for SED detection cannot distinguish between the biologically active form of the toxin, which poses a threat, from the inactive form, which poses no threat. In vivo bioassays that measure emetic activity in kitten and monkeys have been used, but these methods rely upon expensive procedures using live animals and raising ethical concerns. A rapid (5 h) quantitative bioluminescence assay, using a genetically engineered T-cell Jurkat cell line expressing luciferase under regulation of nuclear factor of activated T cells response elements, in combination with the lymphoblastoid B-cell line Raji for antigen presentation, was developed. In this assay, the detection limit of biologically active SED is 100 ng/mL, which is 10 times more sensitive than the splenocyte proliferation assay, and 105 times more sensitive than monkey or kitten bioassay. Pasteurization or repeated freeze-thaw cycles had no effect on SED activity, but reduction in SED activity was shown with heat treatment at 100°C for 5 min. It was also shown that milk exhibits a protective effect on SED. This bioluminescence assay may also be used to rapidly evaluate antibodies to SED for potential therapeutic application as a measurement of neutralizing biological effects of SED.
Collapse
Affiliation(s)
- Reuven Rasooly
- Western Regional Research Center, Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, USDA, Albany, CA, U.S.A
| | - Paula M Do
- Western Regional Research Center, Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, USDA, Albany, CA, U.S.A
| | - Bradley J Hernlem
- Western Regional Research Center, Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, USDA, Albany, CA, U.S.A
| |
Collapse
|
22
|
Rasooly R, Do P, Hernlem B. Sensitive, Rapid, Quantitative and in Vitro Method for the Detection of Biologically Active Staphylococcal Enterotoxin Type E. Toxins (Basel) 2016; 8:toxins8050150. [PMID: 27187474 PMCID: PMC4885065 DOI: 10.3390/toxins8050150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus aureus is a major bacterial cause of clinical infections and foodborne illnesses through its production of a group of enterotoxins (SEs) which cause gastroenteritis and also function as superantigens to massively activate T cells. In the present study, we tested Staphylococcal enterotoxin type E (SEE), which was detected in 17 of the 38 suspected staphylococcal food poisoning incidents in a British study and was the causative agent in outbreaks in France, UK and USA. The current method for detection of enterotoxin activity is an in vivo monkey or kitten bioassay; however, this expensive procedure has low sensitivity and poor reproducibility, requires many animals, is impractical to test on a large number of samples, and raises ethical concerns with regard to the use of experimental animals. The purpose of this study is to develop rapid sensitive and quantitative bioassays for detection of active SEE. We apply a genetically engineered T cell-line expressing the luciferase reporter gene under the regulation of nuclear factor of activated T-cells response element (NFAT-RE), combined with a Raji B-cell line that presents the SEE-MHC (major histocompatibility complex) class II to the engineered T cell line. Exposure of the above mixed culture to SEE induces differential expression of the luciferase gene and bioluminescence is read out in a dose dependent manner over a 6-log range. The limit of detection of biologically active SEE is 1 fg/mL which is 109 times more sensitive than the monkey and kitten bioassay.
Collapse
Affiliation(s)
- Reuven Rasooly
- Western Regional Research Center, Foodborne Toxin Detection & Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| | - Paula Do
- Western Regional Research Center, Foodborne Toxin Detection & Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| | - Bradley Hernlem
- Western Regional Research Center, Foodborne Toxin Detection & Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| |
Collapse
|
23
|
Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis 2016; 7:e2199. [PMID: 27100893 PMCID: PMC4855676 DOI: 10.1038/cddis.2016.97] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors consists of four Ca2+-regulated members (NFAT1–NFAT4), which were first described in T lymphocytes. In addition to their well-documented role in T lymphocytes, where they control gene expression during cell activation and differentiation, NFAT proteins are also expressed in a wide range of cells and tissue types and regulate genes involved in cell cycle, apoptosis, angiogenesis and metastasis. The NFAT proteins share a highly conserved DNA-binding domain (DBD), which allows all NFAT members to bind to the same DNA sequence in enhancers or promoter regions. The same DNA-binding specificity suggests redundant roles for the NFAT proteins, which is true during the regulation of some genes such as IL-2 and p21. However, it has become increasingly clear that different NFAT proteins and even isoforms can have unique functions. In this review, we address the possible reasons for these distinct roles, particularly regarding N- and C-terminal transactivation regions (TADs) and the partner proteins that interact with these TADs. We also discuss the genes regulated by NFAT during cell cycle regulation and apoptosis and the role of NFAT during tumorigenesis.
Collapse
|
24
|
NFAT2 Isoforms Differentially Regulate Gene Expression, Cell Death, and Transformation through Alternative N-Terminal Domains. Mol Cell Biol 2015; 36:119-31. [PMID: 26483414 DOI: 10.1128/mcb.00501-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors is composed of four calcium-responsive proteins (NFAT1 to -4). The NFAT2 (also called NFATc1) gene encodes the isoforms NFAT2α and NFAT2β that result mainly from alternative initiation exons that provide two different N-terminal transactivation domains. However, the specific roles of the NFAT2 isoforms in cell physiology remain unclear. Because previous studies have shown oncogenic potential for NFAT2, this study emphasized the role of the NFAT2 isoforms in cell transformation. Here, we show that a constitutively active form of NFAT2α (CA-NFAT2α) and CA-NFAT2β distinctly control death and transformation in NIH 3T3 cells. While CA-NFAT2α strongly induces cell transformation, CA-NFAT2β leads to reduced cell proliferation and intense cell death through the upregulation of tumor necrosis factor alpha (TNF-α). CA-NFAT2β also increases cell death and upregulates Fas ligand (FasL) and TNF-α in CD4(+) T cells. Furthermore, we demonstrate that differential roles of NFAT2 isoforms in NIH 3T3 cells depend on the N-terminal domain, where the NFAT2β-specific N-terminal acidic motif is necessary to induce cell death. Interestingly, the NFAT2α isoform is upregulated in Burkitt lymphomas, suggesting an isoform-specific involvement of NFAT2 in cancer development. Finally, our data suggest that alternative N-terminal domains of NFAT2 could provide differential mechanisms for the control of cellular functions.
Collapse
|
25
|
Perotti V, Baldassari P, Molla A, Vegetti C, Bersani I, Maurichi A, Santinami M, Anichini A, Mortarini R. NFATc2 is an intrinsic regulator of melanoma dedifferentiation. Oncogene 2015; 35:2862-72. [DOI: 10.1038/onc.2015.355] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/04/2015] [Indexed: 12/20/2022]
|
26
|
Silva O, Crocetti J, Humphries LA, Burkhardt JK, Miceli MC. Discs Large Homolog 1 Splice Variants Regulate p38-Dependent and -Independent Effector Functions in CD8+ T Cells. PLoS One 2015; 10:e0133353. [PMID: 26186728 PMCID: PMC4505885 DOI: 10.1371/journal.pone.0133353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 11/22/2022] Open
Abstract
Functionally diverse CD8+ T cells develop in response to antigenic stimulation with differing capacities to couple TCR engagement to downstream signals and functions. However, mechanisms of diversifying TCR signaling are largely uncharacterized. Here we identified two alternative splice variants of scaffold protein Dlg1, Dlg1AB and Dlg1B, that diversify signaling to regulate p38 –dependent and –independent effector functions in CD8+ T cells. Dlg1AB, but not Dlg1B associated with Lck, coupling TCR stimulation to p38 activation and proinflammatory cytokine production. Conversely, both Dlg1AB and Dlg1B mediated p38-independent degranulation. Degranulation depended on a Dlg1 fragment containing an intact Dlg1SH3-domain and required the SH3-ligand WASp. Further, Dlg1 controlled WASp activation by promoting TCR-triggered conformational opening of WASp. Collectively, our data support a model where Dlg1 regulates p38-dependent proinflammatory cytokine production and p38-independent cytotoxic granule release through the utilization of alternative splice variants, providing a mechanism whereby TCR engagement couples downstream signals to unique effector functions in CD8+ T cells.
Collapse
Affiliation(s)
- Oscar Silva
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jillian Crocetti
- Molecular Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, California, United States of America
| | - Lisa A. Humphries
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Janis K. Burkhardt
- Department of Laboratory Medicine, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - M. Carrie Miceli
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Savage SR, Bretz CA, Penn JS. RNA-Seq reveals a role for NFAT-signaling in human retinal microvascular endothelial cells treated with TNFα. PLoS One 2015; 10:e0116941. [PMID: 25617622 PMCID: PMC4305319 DOI: 10.1371/journal.pone.0116941] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/17/2014] [Indexed: 01/10/2023] Open
Abstract
TNFα has been identified as playing an important role in pathologic complications associated with diabetic retinopathy and retinal inflammation, such as retinal leukostasis. However, the transcriptional effects of TNFα on retinal microvascular endothelial cells and the different signaling pathways involved are not yet fully understood. In the present study, RNA-seq was used to profile the transcriptome of human retinal microvascular endothelial cells (HRMEC) treated for 4 hours with TNFα in the presence or absence of the NFAT-specific inhibitor INCA-6, in order to gain insight into the specific effects of TNFα on RMEC and identify any involvement of NFAT signaling. Differential expression analysis revealed that TNFα treatment significantly upregulated the expression of 579 genes when compared to vehicle-treated controls, and subsequent pathway analysis revealed a TNFα-induced enrichment of transcripts associated with cytokine-cytokine receptor interactions, cell adhesion molecules, and leukocyte transendothelial migration. Differential expression analysis comparing TNFα-treated cells to those co-treated with INCA-6 revealed 10 genes whose expression was significantly reduced by the NFAT inhibitor, including those encoding the proteins VCAM1 and CX3CL1 and cytokines CXCL10 and CXCL11. This study identifies the transcriptional effects of TNFα on HRMEC, highlighting its involvement in multiple pathways that contribute to retinal leukostasis, and identifying a previously unknown role for NFAT-signaling downstream of TNFα.
Collapse
Affiliation(s)
- Sara R. Savage
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Colin A. Bretz
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - John S. Penn
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
28
|
Mognol GP, de Araujo-Souza PS, Robbs BK, Teixeira LK, Viola JP. Transcriptional regulation of thec-Mycpromoter by NFAT1 involves negative and positive NFAT-responsive elements. Cell Cycle 2014; 11:1014-28. [DOI: 10.4161/cc.11.5.19518] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
29
|
Crocetti J, Silva O, Humphries LA, Tibbs MD, Miceli MC. Selective phosphorylation of the Dlg1AB variant is critical for TCR-induced p38 activation and induction of proinflammatory cytokines in CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:2651-60. [PMID: 25098293 DOI: 10.4049/jimmunol.1401196] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CD8(+) T cells respond to TCR stimulation by producing proinflammatory cytokines, and destroying infected or malignant cells through the production and release of cytotoxic granules. Scaffold protein Discs large homolog 1 (Dlg1) specifies TCR-dependent functions by channeling proximal signals toward the activation of p38-dependent proinflammatory cytokine gene expression and/or p38-independent cytotoxic granule release. Two Dlg1 variants are expressed in CD8(+) T cells via alternative splicing, Dlg1AB and Dlg1B, which have differing abilities coordinate TCR-dependent functions. Although both variants facilitate p38-independent cytotoxicity, only Dlg1AB coordinates p38-dependent proinflammatory cytokine expression. In this study, we identify TCR-induced Dlg1 tyrosine phosphorylation as a key regulatory step required for Dlg1AB-mediated p38-dependent functions, including proinflammatory cytokine expression. We find that Dlg1AB but not Dlg1B is tyrosine phosphorylated by proximal tyrosine kinase Lck in response to TCR stimulation. Furthermore, we identify Dlg1 tyrosine 222 (Y222) as a major site of Dlg1 phosphorylation required for TCR-triggered p38 activation and NFAT-dependent expression of proinflammatory cytokines, but not for p38-independent cytotoxicity. Taken together, our data support a model where TCR-induced phosphorylation of Dlg1 Y222 is a key point of control that endows Dlg1AB with the ability to coordinate p38 activation and proinflammatory cytokine production. We propose blocking Dlg1AB phosphorylation as a novel therapeutic target to specifically block proinflammatory cytokine production but not cytotoxicity.
Collapse
Affiliation(s)
- Jillian Crocetti
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Oscar Silva
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Lisa A Humphries
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095; Amgen, Thousand Oaks, CA 91320; and
| | - Michelle D Tibbs
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA 90095
| | - M Carrie Miceli
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
30
|
Alam MS, Gaida MM, Ogawa Y, Kolios AGA, Lasitschka F, Ashwell JD. Counter-regulation of T cell effector function by differentially activated p38. ACTA ACUST UNITED AC 2014; 211:1257-70. [PMID: 24863062 PMCID: PMC4042639 DOI: 10.1084/jem.20131917] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Unlike the MAP kinase (MAPK) cascade that phosphorylates p38 on the activation loop, T cell receptor (TCR) signaling results in phosphorylation on Tyr-323 (pY323, alternative pathway). Using mice expressing p38α and p38β with Y323F substitutions, we show that alternatively but not MAPK cascade-activated p38 up-regulates the transcription factors NFATc1 and IRF4, which are required for proliferation and cytokine production. Conversely, activation of p38 with UV or osmotic shock mitigated TCR-mediated activation by phosphorylation and cytoplasmic retention of NFATc1. Notably, UVB treatment of human psoriatic lesions reduced skin-infiltrating p38 pY323(+) T cell IRF4 and IL-17 production. Thus, distinct mechanisms of p38 activation converge on NFATc1 with opposing effects on T cell immunity, which may underlie the beneficial effect of phototherapy on psoriasis.
Collapse
Affiliation(s)
- Muhammad S Alam
- Laboratory of Immune Cell Biology, Center for Cancer Research; Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Matthias M Gaida
- Laboratory of Immune Cell Biology, Center for Cancer Research; Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Youichi Ogawa
- Laboratory of Immune Cell Biology, Center for Cancer Research; Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Antonios G A Kolios
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland Laboratory of Applied Immunobiology, University of Zurich, 8006 Zurich, Switzerland
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research; Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
31
|
Liu H, Zhao Q, Song Q, Zhou FH, Kang HJ, Pan L, Yao YM. Release of High Mobility Protein Box-1 is Greatly Regulated by Nuclear Factor of Activated T Cell-2 in Human Monocytes. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Close talk between inflammatory mediators and immunological cytokines has been discovered and reported. In this study, the role of nuclear factor of activated T cell-2 (NFAT2) in regulation of high mobility group box-1 (HMGB1) release was investigated. THP-1 cell and HEK293T cell were incubated and stimulated by lipopolysaccharide (LPS). Firstly, binding site between HMGB1 and NFAT2 was identified by co-immunoprecipitation (IP). Box A, Box B and CT domain of HMGB1 were constructed, as well as Rel-homology-domain (RHD), pre-RHD and pro-RHD of NFAT2. THP-1 cell was harvested, cell lysate and culture medium were collected at appointed times. Binding between HMGB1 and NFAT2 was measured, HMGB1 protein level in culture medium was analyzed at the same time. Secondly, the role of NFAT2 in regulating HMGB1 release was investigated. When THP-1 cell was cultured for 24 h, HMGB1 protein level was measured at appointed times with or without siRNA to inhibit NFAT2 expression. Our data show that HMGB1 bound to NFAT2 in THP-1 cell cytoplasm. Further experiments showed that box B domain of HMGB1 could bind to pre-RHD of NFAT2. After stimulation by LPS, interaction between HMGB1 and NFAT2 was discovered decreasing gradually. However, HMGB1 protein level increased in culture medium at the same time. Furthermore, HMGB1 release could be enhanced by NFAT2 inhibition. Taken together, release of HMGB1 could be regulated by NFAT2 in human monocytes.
Collapse
Affiliation(s)
- H. Liu
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Zhao
- Gastroenterology Department of Nanlou, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Song
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - F-H. Zhou
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - H-J. Kang
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - L. Pan
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Y-M. Yao
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital (formerly 304th Hospital), Beijing, People's Republic of China
| |
Collapse
|
32
|
Liu FX, Wu CL, Zhu ZA, Li MQ, Mao YQ, Liu M, Wang XQ, Yu DG, Tang TT. Calcineurin/NFAT pathway mediates wear particle-induced TNF-α release and osteoclastogenesis from mice bone marrow macrophages in vitro. Acta Pharmacol Sin 2013; 34:1457-66. [PMID: 24056707 DOI: 10.1038/aps.2013.99] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 07/08/2013] [Indexed: 12/29/2022]
Abstract
AIM To investigate the roles of the calcineurin/nuclear factor of activated T cells (NFAT) pathway in regulation of wear particles-induced cytokine release and osteoclastogenesis from mouse bone marrow macrophages in vitro. METHODS Osteoclasts were induced from mouse bone marrow macrophages (BMMs) in the presence of 100 ng/mL receptor activator of NF-κB ligand (RANKL). Acridine orange staining and MTT assay were used to detect the cell viability. Osteoclastogenesis was determined using TRAP staining and RT-PCR. Bone pit resorption assay was used to examine osteoclast phenotype. The expression and cellular localization of NFATc1 were examined using RT-PCR and immunofluorescent staining. The production of TNFα was analyzed with ELISA. RESULTS Titanium (Ti) or polymethylmethacrylate (PMMA) particles (0.1 mg/mL) did not significantly change the viability of BMMs, but twice increased the differentiation of BMMs into mature osteoclasts, and markedly increased TNF-α production. The TNF-α level in the PMMA group was significantly higher than in the Ti group (96 h). The expression of NFATc1 was found in BMMs in the presence of the wear particles and RANKL. In bone pit resorption assay, the wear particles significantly increased the resorption area and total number of resorption pits in BMMs-seeded ivory slices. Addition of 11R-VIVIT peptide (a specific inhibitor of calcineurin-mediated NFAT activation, 2.0 μmol/L) did not significantly affect the viability of BMMs, but abolished almost all the wear particle-induced alterations in BMMs. Furthermore, VIVIT reduced TNF-α production much more efficiently in the PMMA group than in the Ti group (96 h). CONCLUSION Calcineurin/NFAT pathway mediates wear particles-induced TNF-α release and osteoclastogenesis from BMMs. Blockade of this signaling pathway with VIVIT may provide a promising therapeutic modality for the treatment of periprosthetic osteolysis.
Collapse
|
33
|
Shebzukhov YV, Horn K, Brazhnik KI, Drutskaya MS, Kuchmiy AA, Kuprash DV, Nedospasov SA. Dynamic changes in chromatin conformation at the TNF transcription start site in T helper lymphocyte subsets. Eur J Immunol 2013; 44:251-64. [PMID: 24009130 DOI: 10.1002/eji.201243297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 07/23/2013] [Accepted: 09/02/2013] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor (TNF) is one of the key primary response genes in the immune system that can be activated by a variety of stimuli. Previous analysis of chromatin accessibility to DNaseI demonstrated open chromatin conformation of the TNF proximal promoter in T cells. Here, using chromatin probing with restriction enzyme EcoNI and micrococcal nuclease we show that in contrast to the proximal promoter, the TNF transcription start site remains in a closed chromatin configuration in primary T helper (Th) cells, but acquires an open state after activation or polarization under Th1 and Th17 conditions. We further demonstrate that transcription factor c-Jun plays a pivotal role in the maintenance of open chromatin conformation at the transcription start site of the TNF gene.
Collapse
Affiliation(s)
- Yury V Shebzukhov
- German Rheumatism Research Center, a Leibniz Institute, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Bachu M, Yalla S, Asokan M, Verma A, Neogi U, Sharma S, Murali RV, Mukthey AB, Bhatt R, Chatterjee S, Rajan RE, Cheedarla N, Yadavalli VS, Mahadevan A, Shankar SK, Rajagopalan N, Shet A, Saravanan S, Balakrishnan P, Solomon S, Vajpayee M, Satish KS, Kundu TK, Jeang KT, Ranga U. Multiple NF-κB sites in HIV-1 subtype C long terminal repeat confer superior magnitude of transcription and thereby the enhanced viral predominance. J Biol Chem 2012; 287:44714-35. [PMID: 23132857 DOI: 10.1074/jbc.m112.397158] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We demonstrate that at least three different promoter variant strains of HIV-1 subtype C have been gradually expanding and replacing the standard subtype C viruses in India, and possibly in South Africa and other global regions, over the past decade. The new viral strains contain an additional NF-κB, NF-κB-like, or RBEIII site in the viral promoter. Although the acquisition of an additional RBEIII site is a property shared by all the HIV-1 subtypes, acquiring an additional NF-κB site remains an exclusive property of subtype C. The acquired κB site is genetically distinct, binds the p50-p65 heterodimer, and strengthens the viral promoter at the levels of transcription initiation and elongation. The 4-κB viruses dominate the 3-κB "isogenic" viral strains in pairwise competition assays in T-cell lines, primary cells, and the ecotropic human immunodeficiency virus mouse model. The dominance of the 4-κB viral strains is also evident in the natural context when the subjects are coinfected with κB-variant viral strains. The mean plasma viral loads, but not CD4 counts, are significantly different in 4-κB infection suggesting that these newly emerging strains are probably more infectious. It is possible that higher plasma viral loads underlie selective transmission of the 4-κB viral strains. Several publications previously reported duplication or deletion of diverse transcription factor-binding sites in the viral promoter. Unlike previous reports, our study provides experimental evidence that the new viral strains gained a potential selective advantage as a consequence of the acquired transcription factor-binding sites and importantly that these strains have been expanding at the population level.
Collapse
Affiliation(s)
- Mahesh Bachu
- HIV-AIDS Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru 560064, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
NFAT1 C-terminal domains are necessary but not sufficient for inducing cell death. PLoS One 2012; 7:e47868. [PMID: 23110116 PMCID: PMC3482241 DOI: 10.1371/journal.pone.0047868] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022] Open
Abstract
The proteins belonging to the nuclear factor of activated T cells (NFAT) family of transcription factors are expressed in several cell types and regulate genes involved in differentiation, cell cycle and apoptosis. NFAT proteins share two conserved domains, the NFAT-homology region (NHR) and a DNA-binding domain (DBD). The N- and C-termini display two transactivation domains (TAD-N and TAD-C) that have low sequence similarity. Due to the high sequence conservation in the NHR and DBD, NFAT members have some overlapping roles in gene regulation. However, several studies have shown distinct roles for NFAT proteins in the regulation of cell death. The TAD-C shows low sequence similarity among NFAT family members, but its contribution to specific NFAT1-induced phenotypes is poorly understood. Here, we described at least two regions of NFAT1 TAD-C that confer pro-apoptotic activity to NFAT1. These regions extend from amino acids 699 to 734 and 819 to 850 of NFAT1. We also showed that the NFAT1 TAD-C is unable to induce apoptosis by itself and requires a functional DBD. Furthermore, we showed that when fused to NFAT1 TAD-C, NFAT2, which is associated with cell transformation, induces apoptosis in fibroblasts. Together, these results suggest that the NFAT1 TAD-C includes NFAT death domains that confer to different NFAT members the ability to induce apoptosis.
Collapse
|
36
|
Yamaoka K, Kaminuma O, Kitamura N, Mori A, Tatsumi H, Nemoto S, Hiroi T. Protein phosphatase 1 is involved in IL-2-induced IL-5 and IL-13 expression in human T cells. Genes Cells 2012; 17:611-8. [PMID: 22646506 DOI: 10.1111/j.1365-2443.2012.01610.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/29/2012] [Indexed: 11/29/2022]
Abstract
IL-2 plays an important role in immunological and other biological functions. This cytokine directly induces the production of several cytokines, such as IL-5 and IL-13. The mechanisms of IL-2-mediated cytokine synthesis are mostly unclear; however, the involvement of IL-2 receptor (IL-2R)β has been suggested. In this study, the signaling molecule downstream of IL-2Rβ was investigated, employing a proteomic approach. Full-length IL-2Rβ and its mutant in which the intracellular component was truncated were introduced in an IL-2Rα- and IL-2Rγ-stably transfected T cell hybridoma, S1. The differential phosphorylation profiles of protein tyrosine residues in these cells upon IL-2 stimulation were examined by two-dimensional gel electrophoresis. The candidate phosphoproteins of interest were re-covered, in-gel digested and mass spectrometry fingerprinted. Among proteins specifically phosphorylated in full-length IL-2Rβ-expressing cells in response to IL-2 stimulation, protein phosphatase (PP)1β and FK506-binding protein 4 were identified. Particularly, PP1β augmented IL-5 and IL-13 expression stimulated by IL-2 but not by anti-CD3 antibody in human peripheral CD4+ T cells upon ectopic expression. IL-2-induced cytokine expression was suppressed by overexpression of PP1 regulatory subunit 2. A PP1 inhibitor, tautomycin, but not a PP2A inhibitor, okadaic acid, also inhibited the IL-2R-mediated responses. It was conclusively shown that PP1 is crucially involved in IL-2-mediated IL-5 and IL-13 synthesis in human T cells.
Collapse
Affiliation(s)
- Kazuko Yamaoka
- Department of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, 156-8506, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Reichenberger EJ, Levine MA, Olsen BR, Papadaki ME, Lietman SA. The role of SH3BP2 in the pathophysiology of cherubism. Orphanet J Rare Dis 2012; 7 Suppl 1:S5. [PMID: 22640988 PMCID: PMC3359958 DOI: 10.1186/1750-1172-7-s1-s5] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cherubism is a rare bone dysplasia that is characterized by symmetrical bone resorption limited to the jaws. Bone lesions are filled with soft fibrous giant cell-rich tissue that can expand and cause severe facial deformity. The disorder typically begins in children at ages of 2-5 years and the bone resorption and facial swelling continues until puberty; in most cases the lesions regress spontaneously thereafter. Most patients with cherubism have germline mutations in the gene encoding SH3BP2, an adapter protein involved in adaptive and innate immune response signaling. A mouse model carrying a Pro416Arg mutation in SH3BP2 develops osteopenia and expansile lytic lesions in bone and some soft tissue organs. In this review we discuss the genetics of cherubism, the biological functions of SH3BP2 and the analysis of the mouse model. The data suggest that the underlying cause for cherubism is a systemic autoinflammatory response to physiologic challenges despite the localized appearance of bone resorption and fibrous expansion to the jaws in humans.
Collapse
Affiliation(s)
- Ernst J Reichenberger
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, CT, USA.
| | | | | | | | | |
Collapse
|
38
|
Ishihara S, Schwartz RH. Two-step binding of transcription factors causes sequential chromatin structural changes at the activated IL-2 promoter. THE JOURNAL OF IMMUNOLOGY 2011; 187:3292-9. [PMID: 21832163 DOI: 10.4049/jimmunol.1003173] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Most gene promoters have multiple binding sequences for many transcription factors, but the contribution of each of these factors to chromatin remodeling is still unclear. Although we previously found a dynamic change in the arrangement of nucleosome arrays at the Il2 promoter during T cell activation, its timing preceded that of a decrease in nucleosome occupancy at the promoter. In this article, we show that the initial nucleosome rearrangement was temporally correlated with the binding of NFAT1 and AP-1 (Fos/Jun), whereas the second step occurred in parallel with the recruitment of other transcription factors and RNA polymerase II. Pharmacologic inhibitors for activation of NFAT1 or induction of Fos blocked the initial phase in the sequential changes. This step was not affected, however, by inhibition of c-Jun phosphorylation, which instead blocked the binding of the late transcription factors, the recruitment of CREB-binding protein, and the acetylation of histone H3 at lysine 27. Thus, the sequential recruitment of transcription factors appears to facilitate two separate steps in chromatin remodeling at the Il2 locus.
Collapse
Affiliation(s)
- Satoru Ishihara
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
39
|
NFATc3 regulates the transcription of genes involved in T-cell activation and angiogenesis. Blood 2011; 118:795-803. [PMID: 21642596 DOI: 10.1182/blood-2010-12-322701] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The nuclear factor of activated T cells (NFAT) family of transcription factors plays important roles in many biologic processes, including the development and function of the immune and vascular systems. Cells usually express more than one NFAT member, raising the question of whether NFATs play overlapping roles or if each member has selective functions. Using mRNA knock-down, we show that NFATc3 is specifically required for IL2 and cyclooxygenase-2 (COX2) gene expression in transformed and primary T cells and for T-cell proliferation. We also show that NFATc3 regulates COX2 in endothelial cells, where it is required for COX2, dependent migration and angiogenesis in vivo. These results indicate that individual NFAT members mediate specific functions through the differential regulation of the transcription of target genes. These effects, observed on short-term suppression by mRNA knock-down, are likely to have been masked by compensatory effects in gene-knockout studies.
Collapse
|
40
|
Mueller C, Braag SA, Keeler A, Hodges C, Drumm M, Flotte TR. Lack of cystic fibrosis transmembrane conductance regulator in CD3+ lymphocytes leads to aberrant cytokine secretion and hyperinflammatory adaptive immune responses. Am J Respir Cell Mol Biol 2011; 44:922-9. [PMID: 20724552 PMCID: PMC3135852 DOI: 10.1165/rcmb.2010-0224oc] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 07/26/2010] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis (CF), the most common fatal monogenic disease in the United States, results from mutations in CF transmembrane conductance regulator (CFTR), a chloride channel. The mechanisms by which CFTR mutations cause lung disease in CF are not fully defined but may include altered ion and water transport across the airway epithelium and aberrant inflammatory and immune responses to pathogens within the airways. We have shown that Cftr(-/-) mice mount an exaggerated IgE response toward Aspergillus fumigatus, with higher levels of IL-13 and IL-4, mimicking both the T helper cell type 2-biased immune responses seen in patients with CF. Herein, we demonstrate that these aberrations are primarily due to Cftr deficiency in lymphocytes rather than in the epithelium. Adoptive transfer experiments with CF splenocytes confer a higher IgE response to Aspergillus fumigatus compared with hosts receiving wild-type splenocytes. The predilection of Cftr-deficient lymphocytes to mount T helper cell type 2 responses with high IL-13 and IL-4 was confirmed by in vitro antigen recall experiments. Conclusive data on this phenomenon were obtained with conditional Cftr knockout mice, where mice lacking Cftr in T cell lineages developed higher IgE than their wild-type control littermates. Further analysis of Cftr-deficient lymphocytes revealed an enhanced intracellular Ca(2+) flux in response to T cell receptor activation. This was accompanied by an increase in nuclear localization of the calcium-sensitive transcription factor, nuclear factor of activated T cell, which could drive the IL-13 response. In summary, our data identified that CFTR dysfunction in T cells can lead directly to aberrant immune responses. These findings implicate the lymphocyte population as a potentially important target for CF therapeutics.
Collapse
Affiliation(s)
- Christian Mueller
- University of Massachusetts Medical School Department of Pediatrics and Gene Therapy Center, Worcester, Massachusetts 01605, USA.
| | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Ohoka Y, Yokota A, Takeuchi H, Maeda N, Iwata M. Retinoic acid-induced CCR9 expression requires transient TCR stimulation and cooperativity between NFATc2 and the retinoic acid receptor/retinoid X receptor complex. THE JOURNAL OF IMMUNOLOGY 2010; 186:733-44. [PMID: 21148038 DOI: 10.4049/jimmunol.1000913] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Retinoic acid (RA) imprints gut-homing specificity on T cells upon activation by inducing the expression of chemokine receptor CCR9 and integrin α4β7. CCR9 expression seemed to be more highly dependent on RA than was the α4β7 expression, but its molecular mechanism remained unclear. In this article, we show that NFAT isoforms NFATc1 and NFATc2 directly interact with RA receptor (RAR) and retinoid X receptor (RXR) but play differential roles in RA-induced CCR9 expression on murine naive CD4(+) T cells. TCR stimulation for 6-24 h was required for the acquisition of responsiveness to RA and induced activation of NFATc1 and NFATc2. However, RA failed to induce CCR9 expression as long as TCR stimulation continued. After terminating TCR stimulation or adding cyclosporin A to the culture, Ccr9 gene transcription was induced, accompanied by inactivation of NFATc1 and sustained activation of NFATc2. Reporter and DNA-affinity precipitation assays demonstrated that the binding of NFATc2 to two NFAT-binding sites and that of the RAR/RXR complex to an RA response element half-site in the 5'-flanking region of the mouse Ccr9 gene were critical for RA-induced promoter activity. NFATc2 directly bound to RARα and RXRα, and it enhanced the binding of RARα to the RA response element half-site. NFATc1 also bound to the NFAT-binding sites and directly to RARα and RXRα, but it inhibited the NFATc2-dependent promoter activity. These results suggest that the cooperativity between NFATc2 and the RAR/RXR complex is essential for CCR9 expression on T cells and that NFATc1 interferes with the action of NFATc2.
Collapse
Affiliation(s)
- Yoshiharu Ohoka
- Laboratory of Biodefense Research, Faculty of Pharmaceutical Sciences at Kagawa Campus, Tokushima Bunri University, Kagawa 769-2193, Japan.
| | | | | | | | | |
Collapse
|
43
|
Falvo JV, Tsytsykova AV, Goldfeld AE. Transcriptional control of the TNF gene. ACTA ACUST UNITED AC 2010; 11:27-60. [PMID: 20173386 DOI: 10.1159/000289196] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cytokine TNF is a critical mediator of immune and inflammatory responses. The TNF gene is an immediate early gene, rapidly transcribed in a variety of cell types following exposure to a broad range of pathogens and signals of inflammation and stress. Regulation of TNF gene expression at the transcriptional level is cell type- and stimulus-specific, involving the recruitment of distinct sets of transcription factors to a compact and modular promoter region. In this review, we describe our current understanding of the mechanisms through which TNF transcription is specifically activated by a variety of extracellular stimuli in multiple cell types, including T cells, B cells, macrophages, mast cells, dendritic cells, and fibroblasts. We discuss the role of nuclear factor of activated T cells and other transcription factors and coactivators in enhanceosome formation, as well as the contradictory evidence for a role for nuclear factor kappaB as a classical activator of the TNF gene. We describe the impact of evolutionarily conserved cis-regulatory DNA motifs in the TNF locus upon TNF gene transcription, in contrast to the neutral effect of single nucleotide polymorphisms. We also assess the regulatory role of chromatin organization, epigenetic modifications, and long-range chromosomal interactions at the TNF locus.
Collapse
Affiliation(s)
- James V Falvo
- Immune Disease Institute and Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | | | | |
Collapse
|
44
|
Ding Y, Huang Y, Song N, Gao X, Yuan S, Wang X, Cai H, Fu Y, Luo Y. NFAT1 mediates placental growth factor-induced myelomonocytic cell recruitment via the induction of TNF-alpha. THE JOURNAL OF IMMUNOLOGY 2010; 184:2593-601. [PMID: 20097868 DOI: 10.4049/jimmunol.0902378] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recruitment of bone marrow-derived myelomonocytic cells plays a fundamental role in tumor angiogenesis and metastasis. Placental growth factor (PlGF) is a potent cytokine that can attract myelomonocytic cells to the tumor. However, the underlying mechanism remains obscure. In this study, we demonstrate that tumor-derived PlGF activates NFAT1 via vascular endothelial growth factor receptor 1 in both murine and human myelomonocytic cells. Activation of NFAT1 is crucial for PlGF-induced myelomonocytic cell recruitment as shown by the in vitro transwell migration assay, transendothelial migration assay, and PlGF-overexpressing tumor models in mice, respectively. TNF-alpha is upregulated by PlGF in myelomonocytic cells in an NFAT1-dependent manner, which in turn contributes to PlGF-induced myelomonocytic cell recruitment. Blockade of TNF-alpha expression by RNA interference or neutralization of secreted TNF-alpha with its Ab attenuates PlGF-induced myelomonocytic cell migration and transendothelial migration. Furthermore, the inhibitory effect of NFAT1 RNA interference on PlGF function is rescued by exogenously added TNF-alpha. Taken together, we demonstrate that NFAT1 mediates PlGF-induced myelomonocytic cell recruitment via the induction of TNF-alpha. Our present studies discover a novel role of the NFAT1-TNF-alpha pathway in tumor inflammation, which may provide potential targets to diversify current cancer therapy.
Collapse
Affiliation(s)
- Yanping Ding
- National Engineering Laboratory for Antitumor Protein Therapeutics, Beijing Key Laboratory for Protein Therapeutics, and Cancer Biology Laboratory, Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Osteoporosis and arthritis are highly prevalent diseases and a significant cause of morbidity and mortality worldwide. These diseases result from aberrant tissue remodeling leading to weak, fracture-prone bones or painful, dysfunctional joints. The nuclear factor of activated T cells (NFAT) transcription factor family controls diverse biologic processes in vertebrates. Here, we review the scientific evidence that links NFAT-regulated gene transcription to bone and joint pathology. A particular emphasis is placed on the role of NFATs in bone resorption and formation by osteoclasts and osteoblasts, respectively. In addition, emerging data that connect NFATs with cartilage biology, angiogenesis, nociception, and neurogenic inflammation are explored. The goal of this article is to highlight the importance of tissue remodeling in musculoskeletal disease and situate NFAT-driven cellular responses within this context to inspire future research endeavors.
Collapse
Affiliation(s)
- Despina Sitara
- Department of Infectious Diseases and Immunology, Harvard School of Public Health, Boston, MA 02115, USA
| | | |
Collapse
|
46
|
Abstract
Exposing rodents to brief episodes of hypoxia mimics the hypoxemia and the cardiovascular and metabolic effects observed in patients with obstructive sleep apnoea (OSA), a condition that affects between 5% and 20% of the population. Apart from daytime sleepiness, OSA is associated with a high incidence of systemic and pulmonary hypertension, peripheral vascular disease, stroke and sudden cardiac death. The development of animal models to study sleep apnoea has provided convincing evidence that recurrent exposure to intermittent hypoxia (IH) has significant vascular and haemodynamic impact that explain much of the cardiovascular morbidity and mortality observed in patients with sleep apnoea. However, the molecular and cellular mechanisms of how IH causes these changes is unclear and under investigation. This review focuses on the most recent findings addressing these mechanisms. It includes a discussion of the contribution of the nervous system, circulating and vascular factors, inflammatory mediators and transcription factors to IH-induced cardiovascular disease. It also highlights the importance of reactive oxygen species as a primary mediator of the systemic and pulmonary hypertension that develops in response to exposure to IH.
Collapse
Affiliation(s)
- Laura V González Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, USA.
| | | | | | | |
Collapse
|
47
|
Abstract
The intestinal ecosystem contains a normal microbiota, non-immune cells and immune cells associated with the intestinal mucosa. The mechanisms involved in the modulation of the gut immune system by probiotics are not yet completely understood. The present work studies the effect of a fermented milk containing probiotic bacteriumLactobacillus(Lb.)caseiDN114001 on different parameters of the gut immune system involved with the nonspecific, innate and adaptive response. BALB/c mice received the probiotic bacteriumLb. caseiDN114001 or the probiotic fermented milk (PFM). The interaction of the probiotic bacteria with the intestine was studied by electron and fluorescence microscopy. The immunological parameters were studied in the intestinal tissue and in the supernatant of intestinal cells (IC). Results showed that the probiotic bacterium interact with the IC. The whole bacterium or its fragments make contact with the gut associated immune cells. The PFM stimulated the IC with IL-6 release, as well as cells related to the nonspecific barrier and with the immune cells associated with the gut. This last activity was observed through the increase in the population of different immune cells: T lymphocytes and IgA+ B lymphocytes, and by the expression of cell markers related to both innate and adaptive response (macrophages). PFM was also able to activate the enzyme calcineurine responsible for the activation of the transcriptional factor NFAT. PFM induced mucosal immune stimulation reinforcing the non-specific barrier and modulating the innate immune response in the gut, maintaining the intestinal homeostasis.
Collapse
|
48
|
NFAT isoforms control activity-dependent muscle fiber type specification. Proc Natl Acad Sci U S A 2009; 106:13335-40. [PMID: 19633193 DOI: 10.1073/pnas.0812911106] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The intracellular signals that convert fast and slow motor neuron activity into muscle fiber type specific transcriptional programs have only been partially defined. The calcium/calmodulin-dependent phosphatase calcineurin (Cn) has been shown to mediate the transcriptional effects of motor neuron activity, but precisely how 4 distinct muscle fiber types are composed and maintained in response to activity is largely unknown. Here, we show that 4 nuclear factor of activated T cell (NFAT) family members act coordinately downstream of Cn in the specification of muscle fiber types. We analyzed the role of NFAT family members in vivo by transient transfection in skeletal muscle using a loss-of-function approach by RNAi. Our results show that, depending on the applied activity pattern, different combinations of NFAT family members translocate to the nucleus contributing to the transcription of fiber type specific genes. We provide evidence that the transcription of slow and fast myosin heavy chain (MyHC) genes uses different combinations of NFAT family members, ranging from MyHC-slow, which uses all 4 NFAT isoforms, to MyHC-2B, which only uses NFATc4. Our data contribute to the elucidation of the mechanisms whereby activity can modulate the phenotype and performance of skeletal muscle.
Collapse
|
49
|
Transcription factor C/EBPbeta isoform ratio regulates osteoclastogenesis through MafB. EMBO J 2009; 28:1769-81. [PMID: 19440205 PMCID: PMC2685610 DOI: 10.1038/emboj.2009.127] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Accepted: 04/03/2009] [Indexed: 01/06/2023] Open
Abstract
Disequilibrium between bone-forming osteoblasts and bone-resorbing osteoclasts is central to many bone diseases. Here, we show that dysregulated expression of translationally controlled isoforms of CCAAT/enhancer-binding protein β (C/EBPβ) differentially affect bone mass. Alternative translation initiation that is controlled by the mammalian target of rapamycin (mTOR) pathway generates long transactivating (LAP*, LAP) and a short repressive (LIP) isoforms from a single C/EBPβ transcript. Rapamycin, an inhibitor of mTOR signalling increases the ratio of LAP over LIP and inhibits osteoclastogenesis in wild type (WT) but not in C/EBPβ null (c/ebpβ−/−) or in LIP knock-in (L/L) osteoclast precursors. C/EBPβ mutant mouse strains exhibit increased bone resorption and attenuated expression of MafB, a negative regulator of osteoclastogenesis. Ectopic expression of LAP and LIP in monocytes differentially affect the MafB promoter activity, MafB gene expression and dramatically affect osteoclastogenesis. These data show that mTOR regulates osteoclast formation by modulating the C/EBPβ isoform ratio, which in turn affects osteoclastogenesis by regulating MafB expression.
Collapse
|
50
|
Maxeiner JH, Karwot R, Sauer K, Scholtes P, Boross I, Koslowski M, Türeci O, Wiewrodt R, Neurath MF, Lehr HA, Finotto S. A key regulatory role of the transcription factor NFATc2 in bronchial adenocarcinoma via CD8+ T lymphocytes. Cancer Res 2009; 69:3069-76. [PMID: 19318584 DOI: 10.1158/0008-5472.can-08-1678] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Ca(2+)-regulated calcineurin/nuclear factor of activated T cells (NFAT) cascade controls alternative pathways of T-cell activation and peripheral tolerance. Here, we describe reduction of NFATc2 mRNA expression in the lungs of patients with bronchial adenocarcinoma. In a murine model of bronchoalveolar adenocarcinoma, mice lacking NFATc2 developed more and larger solid tumors than wild-type littermates. The extent of central tumor necrosis was decreased in the tumors in NFATc2((-/-)) mice, and this finding was associated with reduced tumor necrosis factor-alpha and interleukin-2 (IL-2) production by CD8(+) T cells. Adoptive transfer of CD8(+) T cells of NFATc2((-/-)) mice induced transforming growth factor-beta(1) in the airways of recipient mice, thus supporting CD4(+)CD25(+)Foxp-3(+)glucocorticoid-induced tumor necrosis factor receptor (GITR)(+) regulatory T (T(reg)) cell survival. Finally, engagement of GITR in NFATc2((-/-)) mice induced IFN-gamma levels in the airways, reversed the suppression by T(reg) cells, and costimulated effector CD4(+)CD25(+) (IL-2Ralpha) and memory CD4(+)CD127(+) (IL-7Ralpha) T cells, resulting in abrogation of carcinoma progression. Agonistic signaling through GITR, in the absence of NFATc2, thus emerges as a novel possible strategy for the treatment of human bronchial adenocarcinoma in the absence of NFATc2 by enhancing IL-2Ralpha(+) effector and IL-7Ralpha(+) memory-expressing T cells.
Collapse
Affiliation(s)
- Joachim H Maxeiner
- Laboratory of Cellular and Molecular Immunology of the Lung, I. Medical Clinic, University of Mainz, Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|