1
|
Cai W, Tanaka K, Mi X, Rajasekhar VK, Khan JF, Yoo S, de Stanchina E, Rahman J, Mathew S, Abrahimi P, Souness S, Purdon TJ, McDowell JR, Meyerberg J, Fujino T, Healey JH, Abdel-Wahab O, Scheinberg DA, Brentjens RJ, Daniyan AF. Augmenting CAR T-cell Functions with LIGHT. Cancer Immunol Res 2024; 12:1361-1379. [PMID: 38959337 PMCID: PMC11444887 DOI: 10.1158/2326-6066.cir-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 07/05/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has resulted in remarkable clinical success in the treatment of B-cell malignancies. However, its clinical efficacy in solid tumors is limited, primarily by target antigen heterogeneity. To overcome antigen heterogeneity, we developed CAR T cells that overexpress LIGHT, a ligand of both lymphotoxin-β receptor on cancer cells and herpes virus entry mediator on immune cells. LIGHT-expressing CAR T cells displayed both antigen-directed cytotoxicity mediated by the CAR and antigen-independent killing mediated through the interaction of LIGHT with lymphotoxin-β receptor on cancer cells. Moreover, CAR T cells expressing LIGHT had immunostimulatory properties that improved the cells' proliferation and cytolytic profile. These data indicate that LIGHT-expressing CAR T cells may provide a way to eliminate antigen-negative tumor cells to prevent antigen-negative disease relapse.
Collapse
Affiliation(s)
- Winson Cai
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Kento Tanaka
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Xiaoli Mi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jonathan F. Khan
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sarah Yoo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | - Jahan Rahman
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Serena Mathew
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Parwiz Abrahimi
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - Sydney Souness
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | | | | | - Jeremy Meyerberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Takeshi Fujino
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - John H. Healey
- Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA 10065
- Pharmacology Program, Weill Cornell Medical College, New York, NY, USA 10021
| | | | | |
Collapse
|
2
|
Mélique S, Vadel A, Rouquié N, Yang C, Bories C, Cotineau C, Saoudi A, Fazilleau N, Lesourne R. THEMIS promotes T cell development and maintenance by rising the signaling threshold of the inhibitory receptor BTLA. Proc Natl Acad Sci U S A 2024; 121:e2318773121. [PMID: 38713628 PMCID: PMC11098085 DOI: 10.1073/pnas.2318773121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/12/2024] [Indexed: 05/09/2024] Open
Abstract
The current paradigm about the function of T cell immune checkpoints is that these receptors switch on inhibitory signals upon cognate ligand interaction. We here revisit this simple switch model and provide evidence that the T cell lineage protein THEMIS enhances the signaling threshold at which the immune checkpoint BTLA (B- and T-lymphocyte attenuator) represses T cell responses. THEMIS is recruited to the cytoplasmic domain of BTLA and blocks its signaling capacity by promoting/stabilizing the oxidation of the catalytic cysteine of the tyrosine phosphatase SHP-1. In contrast, THEMIS has no detectable effect on signaling pathways regulated by PD-1 (Programmed cell death protein 1), which depend mainly on the tyrosine phosphatase SHP-2. BTLA inhibitory signaling is tuned according to the THEMIS expression level, making CD8+ T cells more resistant to BTLA-mediated inhibition than CD4+ T cells. In the absence of THEMIS, the signaling capacity of BTLA is exacerbated, which results in the attenuation of signals driven by the T cell antigen receptor and by receptors for IL-2 and IL-15, consequently hampering thymocyte positive selection and peripheral CD8+ T cell maintenance. By characterizing the pivotal role of THEMIS in restricting the transmission of BTLA signals, our study suggests that immune checkpoint operability is conditioned by intracellular signal attenuators.
Collapse
Affiliation(s)
- Suzanne Mélique
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Aurélie Vadel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nelly Rouquié
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cui Yang
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Cyrielle Bories
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Coline Cotineau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Abdelhadi Saoudi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Nicolas Fazilleau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| | - Renaud Lesourne
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse31024, France
| |
Collapse
|
3
|
Szymczak M, Ziętkiewicz S, Kuncewicz K, Rodziewicz-Motowidło S, Orlikowska M. Expression, purification, and efficient refolding of the extracellular domain of Escherichia coli-expressed signaling receptor herpesvirus entry mediator. Protein Expr Purif 2019; 164:105450. [PMID: 31299214 DOI: 10.1016/j.pep.2019.105450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 01/21/2023]
Abstract
Herpesvirus entry mediator (HVEM), a member of the TNF-receptor superfamily, plays an important role in the regulation of the immune system. It forms a complex with ligands and can either activate or inhibit the response of the immune system. Furthermore, HVEM can exhibit pro-inflammatory or anti-inflammatory effects in many human diseases. Therefore, understanding the mechanism underlying the interaction of HVEM with other receptors is extremely important to design small therapeutic molecules that can stimulate the response of the immune system. In this study, we attempted to develop the most efficient method for the expression and purification of the extracellular domain of HVEM using Escherichia coli. The soluble fraction constituted only a small portion of the E. coli-expressed protein, whereas majority of the protein was found to be accumulated in the insoluble fraction. Three different protein refolding methods were analyzed: dialysis, dilution, and using chromatographic column. The oligomeric state of the protein was determined by characterizing the obtained fractions using analytical size exclusion chromatography. All the obtained fractions were tested for their ability to form a complex with B- and T-lymphocyte attenuator using enzyme-linked immunosorbent assay. The results of this study provide crucial information regarding the production of HVEM protein in a robust, well-established, and convenient heterologous expression system using E. coli as a host. In addition, it allows for the selection of the most effective method for appropriate refolding of HVEM protein, which gets accumulated in the insoluble fraction.
Collapse
Affiliation(s)
- Marta Szymczak
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Szymon Ziętkiewicz
- Department of Molecular and Cellular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk, 80-822, Gdansk, Kladki 24, Poland
| | - Katarzyna Kuncewicz
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Sylwia Rodziewicz-Motowidło
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland
| | - Marta Orlikowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308, Gdansk, Wita Stwosza 63, Poland.
| |
Collapse
|
4
|
Paluch C, Santos AM, Anzilotti C, Cornall RJ, Davis SJ. Immune Checkpoints as Therapeutic Targets in Autoimmunity. Front Immunol 2018; 9:2306. [PMID: 30349540 PMCID: PMC6186808 DOI: 10.3389/fimmu.2018.02306] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022] Open
Abstract
Antibodies that block the immune checkpoint receptors PD1 and CTLA4 have revolutionized the treatment of melanoma and several other cancers, but in the process, a new class of drug side effect has emerged—immune related adverse events. The observation that therapeutic blockade of these inhibitory receptors is sufficient to break self-tolerance, highlights their crucial role in the physiological modulation of immune responses. Here, we discuss the rationale for targeting immune checkpoint receptors with agonistic agents in autoimmunity, to restore tolerance when it is lost. We review progress that has been made to date, using Fc-fusion proteins, monoclonal antibodies or other novel constructs to induce immunosuppressive signaling through these pathways. Finally, we explore potential mechanisms by which these receptors trigger and modulate immune cell function, and how understanding these processes might shape the design of more effective therapeutic agents in future.
Collapse
Affiliation(s)
- Christopher Paluch
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Ana Mafalda Santos
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Consuelo Anzilotti
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Richard J Cornall
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon J Davis
- MRC Human Immunology Unit, University of Oxford, Oxford, United Kingdom.,Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Maeda T, Suetake H, Odaka T, Miyadai T. Original Ligand for LTβR Is LIGHT: Insight into Evolution of the LT/LTβR System. THE JOURNAL OF IMMUNOLOGY 2018; 201:202-214. [PMID: 29769272 DOI: 10.4049/jimmunol.1700900] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 04/25/2018] [Indexed: 01/23/2023]
Abstract
The lymphotoxin (LT)/LTβ receptor (LTβR) axis is crucial for the regulation of immune responses and development of lymphoid tissues in mammals. Despite the importance of this pathway, the existence and function of LT and LTβR remain obscure for nonmammalian species. In this study, we report a nonmammalian LTβR and its ligand. We demonstrate that TNF-New (TNFN), which has been considered orthologous to mammalian LT, was expressed on the cell surface as a homomer in vitro. This different protein structure indicates that TNFN is not orthologous to mammalian LTα and LTβ. Additionally, we found that LTβR was conserved in teleosts, but the soluble form of recombinant fugu LTβR did not bind to membrane TNFN under the circumstance tested. Conversely, the LTβR recombinant bound to another ligand, LIGHT, similar to that of mammals. These findings indicate that teleost LTβR is originally a LIGHT receptor. In the cytoplasmic region of fugu LTβR, recombinant fugu LTβR bound to the adaptor protein TNFR-associated factor (TRAF) 2, but little to TRAF3. This difference suggests that teleost LTβR could potentially activate the classical NF-κB pathway with a novel binding domain, but would have little ability to activate an alternative one. Collectively, our results suggested that LIGHT was the original ligand for LTβR, and that the teleost immune system lacked the LT/LTβR pathway. Acquisition of the LT ligand and TRAF binding domain after lobe-finned fish may have facilitated the sophistication of the immune system and lymphoid tissues.
Collapse
Affiliation(s)
- Tomoki Maeda
- Graduate School of Biosciences and Biotechnology, Fukui Prefectural University, Fukui 917-0003, Japan.,Japan Society for the Promotion of Science, Tokyo 102-0083, Japan; and
| | - Hiroaki Suetake
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| | - Tomoyuki Odaka
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| | - Toshiaki Miyadai
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan
| |
Collapse
|
6
|
Seeger H, Lindenmeyer MT, Cohen CD, Jaeckel C, Nelson PJ, Chen J, Edenhofer I, Kozakowski N, Regele H, Boehmig G, Brandt S, Wuethrich RP, Heikenwalder M, Fehr T, Segerer S. Lymphotoxin expression in human and murine renal allografts. PLoS One 2018; 13:e0189396. [PMID: 29300739 PMCID: PMC5754061 DOI: 10.1371/journal.pone.0189396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/26/2017] [Indexed: 01/23/2023] Open
Abstract
The kidney is the most frequently transplanted solid organ. Recruitment of inflammatory cells, ranging from diffuse to nodular accumulations with defined microarchitecture, is a hallmark of acute and chronic renal allograft injury. Lymphotoxins (LTs) mediate the communication of lymphocytes and stromal cells and play a pivotal role in chronic inflammation and formation of lymphoid tissue. The aim of this study was to assess the expression of members of the LT system in acute rejection (AR) and chronic renal allograft injury such as transplant glomerulopathy (TG) and interstitial fibrosis/tubular atrophy (IFTA). We investigated differentially regulated components in transcriptomes of human renal allograft biopsies. By microarray analysis, we found the upregulation of LTβ, LIGHT, HVEM and TNF receptors 1 and 2 in AR and IFTA in human renal allograft biopsies. In addition, there was clear evidence for the activation of the NFκB pathway, most likely a consequence of LTβ receptor stimulation. In human renal allograft biopsies with transplant glomerulopathy (TG) two distinct transcriptional patterns of LT activation were revealed. By quantitative RT-PCR robust upregulation of LTα, LTβ and LIGHT was shown in biopsies with borderline lesions and AR. Immunohistochemistry revealed expression of LTβ in tubular epithelial cells and inflammatory infiltrates in transplant biopsies with AR and IFTA. Finally, activation of LT signaling was reproduced in a murine model of renal transplantation with AR. In summary, our results indicate a potential role of the LT system in acute renal allograft rejection and chronic transplant injury. Activation of the LT system in allograft rejection in rodents indicates a species independent mechanism. The functional role of the LT system in acute renal allograft rejection and chronic injury remains to be determined.
Collapse
Affiliation(s)
- Harald Seeger
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
- * E-mail:
| | - Maja T. Lindenmeyer
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Clemens D. Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Carsten Jaeckel
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Peter J. Nelson
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Munich, Germany
| | - Jin Chen
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | - Ilka Edenhofer
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | | | - Heinz Regele
- Clinical Institute of Pathology, University of Vienna, Vienna, Austria
| | - Georg Boehmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Austria
| | - Simone Brandt
- Institute of Surgical Pathology, University Hospital Zuerich, Zurich, Switzerland
| | - Rudolf P. Wuethrich
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Fehr
- Department of Internal Medicine, Kantonsspital Graubuenden, Chur, Switzerland
| | - Stephan Segerer
- Division of Nephrology, University Hospital, Zuerich, Switzerland
- Institute of Physiology and Zuerich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zuerich, Switzerland
- Division of Nephrology, Kantonsspital Aarau, Aarau, Switzerland
| |
Collapse
|
7
|
Associations between HVEM/LIGHT/BTLA/CD160 polymorphisms and the occurrence of antibody-mediate rejection in renal transplant recipients. Oncotarget 2017; 8:100079-100094. [PMID: 29245962 PMCID: PMC5725004 DOI: 10.18632/oncotarget.21941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/19/2017] [Indexed: 12/25/2022] Open
Abstract
Antibody-mediated rejection (ABMR) is a serious complications that can occur following renal transplantation. The production of donor-specific antibodies by the humoral immune response can trigger costimulatory signals, which are crucial in activating immune cells, and therefore, playing a potential role in ABMR. To investigate the role of HVEM/LIGHT/BTLA/CD160 polymorphisms in ABMR, we retrospectively analyzed 200 renal transplant recipients. We adopted next-generation sequencing (NGS) to identify HVEM/LIGHT/BTLA/CD160 single-nucleotide polymorphisms (SNPs) in the genotypes of these patients. We divided the patients into two groups: those with ABMR and those who were stable. We adopted multiple models and performed regression analysis after adjusting for multiple confounding variables, to determine the correlation between the SNPs and ABMR. We obtained 41 high-quality SNPs readouts. However, we did not observe any significant association between these polymorphisms and the pathogenesis of ABMR in any of the models.Nevertheless, since there is evidence suggesting the involvement of costimulatory signals in graft rejection, further research should be conducted to better understand how genetic polymorphisms may be involved in ABMR.
Collapse
|
8
|
Edwards RG, Longnecker R. Herpesvirus Entry Mediator and Ocular Herpesvirus Infection: More than Meets the Eye. J Virol 2017; 91:e00115-17. [PMID: 28404853 PMCID: PMC5469272 DOI: 10.1128/jvi.00115-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As its name suggests, the host receptor herpesvirus entry mediator (HVEM) facilitates herpes simplex virus (HSV) entry through interactions with a viral envelope glycoprotein. HVEM also bridges several signaling networks, binding ligands from both tumor necrosis factor (TNF) and immunoglobulin (Ig) superfamilies with diverse, and often opposing, outcomes. While HVEM was first identified as a viral entry receptor for HSV, it is only recently that HVEM has emerged as an important host factor in immunopathogenesis of ocular HSV type 1 (HSV-1) infection. Surprisingly, HVEM exacerbates disease development in the eye independently of entry. HVEM signaling has been shown to play a variety of roles in modulating immune responses to HSV and other pathogens, and there is increasing evidence that these effects are responsible for HVEM-mediated pathogenesis in the eye. Here, we review the dual branches of HVEM function during HSV infection: entry and immunomodulation. HVEM is broadly expressed; intersects two important immunologic signaling networks; and impacts autoimmunity, infection, and inflammation. We hope that by understanding the complex range of effects mediated by this receptor, we can offer insights applicable to a wide variety of disease states.
Collapse
Affiliation(s)
- Rebecca G Edwards
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Richard Longnecker
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
9
|
Sabokbar A, Mahoney DJ, Hemingway F, Athanasou NA. Non-Canonical (RANKL-Independent) Pathways of Osteoclast Differentiation and Their Role in Musculoskeletal Diseases. Clin Rev Allergy Immunol 2017; 51:16-26. [PMID: 26578261 DOI: 10.1007/s12016-015-8523-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Osteoclasts are multinucleated cells derived from mononuclear phagocyte precursors (monocytes, macrophages); in the canonical pathway of osteoclastogenesis, these cells fuse and differentiate to form specialised bone-resorbing osteoclasts in the presence of receptor activator for nuclear factor kappa B ligand (RANKL). Non-canonical pathways of osteoclastogenesis have been described in which several cytokines and growth factors are able to substitute for RANKL. These humoral factors can generally be divided into those which, like RANKL, are tumour necrosis family (TNF) superfamily members and those which are not; the former include TNFα lymphotoxin exhibiting inducible expression and competing with herpes simplex virus glycoprotein D for herpesvirus entry mediator, a receptor expressed by T lymphocytes (LIGHT), a proliferation inducing ligand (APRIL) and B cell activating factor (BAFF); the latter include transforming growth factor beta (TGF-β), interleukin-6 (IL-6), IL-8, IL-11, nerve growth factor (NGF), insulin-like growth factor-I (IGF-I) and IGF-II. This review summarises the evidence for these RANKL substitutes in inducing osteoclast differentiation from tissue-derived and circulating mononuclear phagocytes. It also assesses the role these factors are likely to play in promoting the pathological bone resorption seen in many inflammatory and neoplastic lesions of bone and joint including rheumatoid arthritis, aseptic implant loosening and primary and secondary tumours of bone.
Collapse
Affiliation(s)
- A Sabokbar
- The Botnar Research Centre, Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Clinical Laboratory Services, Nuffield Orthopaedic Centre, University of Oxford, Oxford, OX3 7LD, UK
| | - D J Mahoney
- The Botnar Research Centre, Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Clinical Laboratory Services, Nuffield Orthopaedic Centre, University of Oxford, Oxford, OX3 7LD, UK
| | - F Hemingway
- The Botnar Research Centre, Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Clinical Laboratory Services, Nuffield Orthopaedic Centre, University of Oxford, Oxford, OX3 7LD, UK
| | - N A Athanasou
- The Botnar Research Centre, Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal and Clinical Laboratory Services, Nuffield Orthopaedic Centre, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
10
|
Luo R, Liu C, Elliott SE, Wang W, Parchim N, Iriyama T, Daugherty PS, Tao L, Eltzschig HK, Blackwell SC, Sibai BM, Kellems RE, Xia Y. Transglutaminase is a Critical Link Between Inflammation and Hypertension. J Am Heart Assoc 2016; 5:JAHA.116.003730. [PMID: 27364991 PMCID: PMC5015405 DOI: 10.1161/jaha.116.003730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background The pathogenesis of essential hypertension is multifactorial with different underlying mechanisms contributing to disease. We have recently shown that TNF superfamily member 14 LIGHT (an acronym for homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator, a receptor expressed by T lymphocytes, also known as TNFSF14) induces hypertension when injected into mice. Research reported here was undertaken to examine the role of transglutaminase (TGase) in LIGHT‐induced hypertension. Methods and Results Initial experiments showed that plasma and kidney TGase activity was induced by LIGHT infusion (13.91±2.92 versus 6.75±1.92 mU/mL and 19.86±3.55 versus 12.00±0.97 mU/10 μg) and was accompanied with hypertension (169±7.16 versus 117.17±11.57 mm Hg at day 14) and renal impairment (proteinuria, 61.33±23.21 versus 20.38±9.01 μg/mg; osmolality, 879.57±93.02 versus 1407.2±308.04 mmol/kg). The increase in renal TGase activity corresponded to an increase in RNA for the tissue TGase isoform, termed TG2. Pharmacologically, we showed that LIGHT‐induced hypertension and renal impairment did not occur in the presence of cystamine, a well‐known competitive inhibitor of TGase activity. Genetically, we showed that LIGHT‐mediated induction of TGase, along with hypertension and renal impairment, was dependent on interleukin‐6 and endothelial hypoxia inducible factor‐1α. We also demonstrated that interleukin‐6, endothelial hypoxia inducible factor‐1α, and TGase are required for LIGHT‐induced production of angiotensin receptor agonistic autoantibodies. Conclusions Thus, LIGHT‐induced hypertension, renal impairment, and production of angiotensin receptor agonistic autoantibodies require TGase, most likely the TG2 isoform. Our findings establish TGase as a critical link between inflammation, hypertension, and autoimmunity.
Collapse
Affiliation(s)
- Renna Luo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chen Liu
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX
| | - Serra E Elliott
- Department of Chemical Engineering, University of California, Santa Barbara, CA
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Nicholas Parchim
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX
| | - Takayuki Iriyama
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX Department of Obstetrics and Gynecology, University of Tokyo, Japan
| | - Patrick S Daugherty
- Department of Chemical Engineering, University of California, Santa Barbara, CA
| | - Lijian Tao
- Department of Nephrology, The First Xiangya Hospital of Central South University, Changsha, Hunan, PRC
| | - Holger K Eltzschig
- Department of Anesthesiology, University of Colorado-Medical School, Denver, CO
| | - Sean C Blackwell
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Texas Health Science Center at Houston, TX
| | - Baha M Sibai
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Texas Health Science Center at Houston, TX
| | - Rodney E Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX The University of Texas Graduate School of Biomedical Sciences at Houston, TX
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, TX The University of Texas Graduate School of Biomedical Sciences at Houston, TX
| |
Collapse
|
11
|
Sakoda Y, Nagai T, Murata S, Mizuno Y, Kurosawa H, Shoda H, Morishige N, Yanai R, Sonoda KH, Tamada K. Pathogenic Function of Herpesvirus Entry Mediator in Experimental Autoimmune Uveitis by Induction of Th1- and Th17-Type T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:2947-54. [PMID: 26912321 DOI: 10.4049/jimmunol.1501742] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/20/2016] [Indexed: 01/23/2023]
Abstract
Herpesvirus entry mediator (HVEM), a member of the TNFR superfamily, serves as a unique molecular switch to mediate both stimulatory and inhibitory cosignals, depending on its functions as a receptor or ligand interacting with multiple binding partners. In this study, we explored the cosignaling functions of HVEM in experimental autoimmune uveitis (EAU), a mouse model resembling human autoimmune uveitis conditions such as ocular sarcoidosis and Behcet disease. Our studies revealed that EAU severity significantly decreased in HVEM-knockout mice compared with wild-type mice, suggesting that stimulatory cosignals from the HVEM receptor are predominant in EAU. Further studies elucidated that the HVEM cosignal plays an important role in the induction of both Th1- and Th17-type pathogenic T cells in EAU, including differentiation of IL-17-producing αβ(+)γδ(-) conventional CD4(+) T cells. Mice lacking lymphotoxin-like, inducible expression, competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes : LIGHT), B- and T-lymphocyte attenuator (BTLA) or both LIGHT and BTLA are also less susceptible to EAU, indicating that LIGHT-HVEM and BTLA-HVEM interactions, two major molecular pathways mediating HVEM functions, are both important in determining EAU pathogenesis. Finally, blocking HVEM cosignals by antagonistic anti-HVEM Abs ameliorated EAU. Taken together, our studies revealed a novel function of the HVEM cosignaling molecule and its ligands in EAU pathogenesis through the induction of Th1- and Th17-type T cell responses and suggested that HVEM-related molecular pathways can be therapeutic targets in autoimmune uveitis.
Collapse
Affiliation(s)
- Yukimi Sakoda
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan; and
| | - Tomohiko Nagai
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan; and Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Sizuka Murata
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Yukari Mizuno
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Hiromi Kurosawa
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan; and
| | - Hiromi Shoda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Naoyuki Morishige
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Ryoji Yanai
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube City, Yamaguchi 755-8505, Japan; and
| |
Collapse
|
12
|
del Rio ML, Fernandez-Renedo C, Chaloin O, Scheu S, Pfeffer K, Shintani Y, Perez-Simon JA, Schneider P, Rodriguez-Barbosa JI. Immunotherapeutic targeting of LIGHT/LTβR/HVEM pathway fully recapitulates the reduced cytotoxic phenotype of LIGHT-deficient T cells. MAbs 2016; 8:478-90. [PMID: 26752542 DOI: 10.1080/19420862.2015.1132130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor (TNF)/TNF receptor (TNFR) superfamily members play essential roles in the development of the different phases of the immune response. Mouse LIGHT (TNFSF14) is a type II transmembrane protein with a C-terminus extracellular TNF homology domain (THD) that assembles in homotrimers and regulates the course of the immune responses by signaling through 2 receptors, the herpes virus entry mediator (HVEM, TNFRSF14) and the lymphotoxin β receptor (LTβR, TNFRSF3). LIGHT is a membrane-bound protein transiently expressed on activated T cells, natural killer (NK) cells and immature dendritic cells that can be proteolytically cleaved by a metalloprotease and released to the extracellular milieu. The immunotherapeutic potential of LIGHT blockade was evaluated in vivo. Administration of an antagonist of LIGHT interaction with its receptors attenuated the course of graft-versus-host reaction and recapitulated the reduced cytotoxic activity of LIGHT-deficient T cells adoptively transferred into non-irradiated semiallogeneic recipients. The lack of LIGHT expression on donor T cells or blockade of LIGHT interaction with its receptors slowed down the rate of T cell proliferation and decreased the frequency of precursor alloreactive T cells, retarding T cell differentiation toward effector T cells. The blockade of LIGHT/LTβR/HVEM pathway was associated with delayed downregulation of interleukin-7Rα and delayed upregulation of inducible costimulatory molecule expression on donor alloreactive CD8 T cells that are typical features of impaired T cell differentiation. These results expose the relevance of LIGHT/LTβR/HVEM interaction for the potential therapeutic control of the allogeneic immune responses mediated by alloreactive CD8 T cells that can contribute to prolong allograft survival.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- a Transplantation Immunobiology Section, Institute of Biomedicine, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital , Leon , Spain
| | - Carlos Fernandez-Renedo
- a Transplantation Immunobiology Section, Institute of Biomedicine, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital , Leon , Spain
| | - Olivier Chaloin
- b CNRS UPR 3572, IBMC, Immunopathologie et Chimie Thérapeutique, 15 rue René Descartes , Strasbourg , France
| | - Stefanie Scheu
- c Institute of Medical Microbiology and Hospital Hygiene, University of Duesseldorf, Universitaetsstr. 1, Geb. 22.21 , Duesseldorf , D-40225 Germany
| | - Klaus Pfeffer
- c Institute of Medical Microbiology and Hospital Hygiene, University of Duesseldorf, Universitaetsstr. 1, Geb. 22.21 , Duesseldorf , D-40225 Germany
| | - Yasushi Shintani
- d Department of International Affairs , Japan Science and Technology Agency, K´s Gobancho 7 , Gobancho Chiyoda-Ku , Tokyo , 102-0076 , Japan
| | - Jose-Antonio Perez-Simon
- e Department of Hematology , University Hospital Virgen del Rocio / Institute of Biomedicine (IBIS / CSIC) , Sevilla , Spain
| | - Pascal Schneider
- f Department of Biochemistry , University of Lausanne , 1066 Epalinges , Switzerland
| | - Jose-Ignacio Rodriguez-Barbosa
- a Transplantation Immunobiology Section, Institute of Biomedicine, University of Leon and Castilla and Leon Regional Transplantation Coordination, Leon University Hospital , Leon , Spain
| |
Collapse
|
13
|
Okwor I, Xu G, Tang H, Liang Y, Fu YX, Uzonna JE. Deficiency of CD40 Reveals an Important Role for LIGHT in Anti-Leishmania Immunity. THE JOURNAL OF IMMUNOLOGY 2015; 195:194-202. [PMID: 26026056 DOI: 10.4049/jimmunol.1401892] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 05/04/2015] [Indexed: 01/19/2023]
Abstract
We previously showed that LIGHT and its receptor herpes virus entry mediator (HVEM) are important for development of optimal CD4(+) Th1 cell immunity and resistance to primary Leishmania major infection in mice. In this study, we further characterized the contributions of this molecule in dendritic cell (DC) maturation, initiation, and maintenance of primary immunity and secondary anti-Leishmania immunity. Flow-cytometric studies showed that CD8α(+) DC subset was mostly affected by HVEM-Ig and lymphotoxin β receptor-Ig treatment. LIGHT signaling is required at both the priming and the maintenance stages of primary anti-Leishmania immunity but is completely dispensable during secondary immunity in wild type mice. However, LIGHT blockade led to impaired IL-12 and IFN-γ responses and loss of resistance in healed CD40-deficient mice after L. major challenge. The protective effect of LIGHT was mediated primarily via its interaction with lymphotoxin β receptor on CD8α(+) DCs. Collectively, our results show that although LIGHT is critical for maintenance of primary Th1 response, it is dispensable during secondary anti-Leishmania immunity in the presence of functional CD40 signaling as seen in wild type mice.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Guilian Xu
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Haidong Tang
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Yong Liang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Jude E Uzonna
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| |
Collapse
|
14
|
Therapeutic blockade of LIGHT interaction with herpesvirus entry mediator and lymphotoxin β receptor attenuates in vivo cytotoxic allogeneic responses. Transplantation 2015; 98:1165-74. [PMID: 25226173 DOI: 10.1097/tp.0000000000000417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Tumor necrosis factor/tumor necrosis factor receptor superfamily members conform a group of molecular interaction pathways of essential relevance during the process of T-cell activation and differentiation toward effector cells and particularly for the maintenance phase of the immune response. Specific blockade of these interacting pathways, such as CD40-CD40L, contributes to modulate the deleterious outcome of allogeneic immune responses. We postulated that antagonizing the interaction of LIGHT expression on activated T cells with its receptors, herpesvirus entry mediator and lymphotoxin β receptor, may decrease T cell-mediated allogeneic responses. METHODS A flow cytometry competition assay was designed to identify anti-LIGHT monoclonal antibodies capable to prevent the interaction of mouse LIGHT with its receptors expressed on transfected cells. An antibody with the desired specificity was evaluated in a short-term in vivo allogeneic cytotoxic assay and tested for its ability to detect endogenous mouse LIGHT. RESULTS We provide evidence for the first time that in mice, as previously described in humans, LIGHT protein is rapidly and transiently expressed after T-cell activation, and this expression was stronger on CD8 T cells than on CD4 T cells. Two anti-LIGHT antibodies prevented interactions of mouse LIGHT with its two known receptors, herpesvirus entry mediator and lymphotoxin β receptor. In vivo administration of anti-LIGHT antibody (clone 10F12) ameliorated host antidonor short-term cytotoxic response in wild type B6 mice, although to a lesser extent than that observed in LIGHT-deficient mice. CONCLUSION The therapeutic targeting of LIGHT may contribute to achieve a better control of cytotoxic responses refractory to current immunosuppressive drugs in transplantation.
Collapse
|
15
|
Figgett WA, Vincent FB, Saulep-Easton D, Mackay F. Roles of ligands from the TNF superfamily in B cell development, function, and regulation. Semin Immunol 2014; 26:191-202. [PMID: 24996229 DOI: 10.1016/j.smim.2014.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/09/2014] [Indexed: 01/01/2023]
Abstract
Most ligands from the tumour necrosis factor (TNF) superfamily play very important roles in the immune system, and particularly so in B lymphocyte biology. TNF ligands are essential to many aspects of normal B cell biology from development in the bone marrow to maturation in the periphery as well as for activation and differentiation into germinal centre, memory or plasma cells. TNF ligands also influence other aspects of B cell biology such as their ability to present antigens or regulate immune responses. Importantly, inadequate regulation of many TNF ligands is associated with B cell disorders including autoimmunity and cancers. As a result, inhibitors of a number of TNF ligands have been tested in the clinic, with some becoming very successful approved treatments alleviating B cell-mediated pathologies.
Collapse
Affiliation(s)
- William A Figgett
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Fabien B Vincent
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Damien Saulep-Easton
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia
| | - Fabienne Mackay
- Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Commercial Road, Melbourne, Victoria 3004, Australia.
| |
Collapse
|
16
|
Goris A, Pauwels I, Dubois B. Progress in multiple sclerosis genetics. Curr Genomics 2013; 13:646-63. [PMID: 23730204 PMCID: PMC3492804 DOI: 10.2174/138920212803759695] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/20/2012] [Accepted: 09/24/2012] [Indexed: 01/06/2023] Open
Abstract
A genetic component in the susceptibility to multiple sclerosis (MS) has long been known, and the first and major genetic risk factor, the HLA region, was identified in the 1970’s. However, only with the advent of genome-wide association studies in the past five years did the list of risk factors for MS grow from 1 to over 50. In this review, we summarize the search for MS risk genes and the latest results. Comparison with data from other autoimmune and neurological diseases and from animal models indicates parallels and differences between diseases. We discuss how these translate into an improved understanding of disease mechanisms, and address current challenges such as genotype-phenotype correlations, functional mechanisms of risk variants and the missing heritability.
Collapse
Affiliation(s)
- An Goris
- Laboratory for Neuroimmunology, Section of Experimental Neurology, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
17
|
Li C, Chen S, Song J, Liu H, Gu W, Ai H, Zhao B, Zhang S. Molecular cloning and characterization of TNFSF14 (LIGHT) and its receptor TNFRSF14 (HVEM) in guinea pig (Cavia porcellus). Gene 2013; 526:374-84. [PMID: 23732292 DOI: 10.1016/j.gene.2013.05.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 05/06/2013] [Accepted: 05/15/2013] [Indexed: 01/05/2023]
Abstract
LIGHT (lymphotoxin-related inducible ligand that competes with herpes simplex virus (HSV) glycoprotein D for herpesvirus entry mediator on T cells) is a member of the tumor necrosis factor (TNF) ligand superfamily, which plays important roles in inflammatory and immune responses. In the present study, the cDNAs of guinea pig (Cavia porcellus) LIGHT (designated as gpLIGHT) and its receptor herpes virus entry mediator (designated as gpHVEM) were amplified from spleen by reverse transcription polymerase chain reaction (RT-PCR). The ORFs of gpLIGHT and gpHVEM cover 726 and 861 bp, encoding predicted proteins with 241 and 286 aas, respectively. The three-dimensional (3D) structure, phylogenetic relationships, and characterization of both genes were also analyzed. We also generated a 3D model to verify interaction between the two proteins. Real-time quantitative PCR (qPCR) analysis revealed that both LIGHT and HVEM are constitutively expressed in guinea pig various tissues. A fusion protein SUMO (Small Ubiquitin-like Modifier)-gpsLIGHT (the soluble mature part of gpLIGHT) was efficiently expressed in Escherichia coli BL21 (DE3) and purified using metal chelate affinity chromatography (Ni-NTA). Laser scanning confocal microscopy (LSCM) showed that gpsLIGHT can bind its receptors on T cells. The LIGHT-HVEM signaling pathway plays an important role in the immune system, and our results might provide a platform for further research into the effects of LIGHT and HVEM.
Collapse
Affiliation(s)
- Chunlan Li
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, Life Sciences College, Nanjing Normal University, Nanjing 210046, China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Maña P, Liñares D, Silva DG, Fordham S, Scheu S, Pfeffer K, Staykova M, Bertram EM. LIGHT (TNFSF14/CD258) Is a Decisive Factor for Recovery from Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:154-63. [DOI: 10.4049/jimmunol.1203016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
del Rio ML, Schneider P, Fernandez-Renedo C, Perez-Simon JA, Rodriguez-Barbosa JI. LIGHT/HVEM/LTβR interaction as a target for the modulation of the allogeneic immune response in transplantation. Am J Transplant 2013; 13:541-51. [PMID: 23356438 DOI: 10.1111/ajt.12089] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/12/2012] [Accepted: 11/30/2012] [Indexed: 01/25/2023]
Abstract
The exchange of information during interactions of T cells with dendritic cells, B cells or other T cells regulates the course of T, B and DC-cell activation and their differentiation into effector cells. The tumor necrosis factor superfamily member LIGHT (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) is transiently expressed upon T cell activation and modulates CD8 T cell-mediated alloreactive responses upon herpes virus entry mediator (HVEM) and lymphotoxin β receptor (LTβR) engagement. LIGHT-deficient mice, or WT mice treated with LIGHT-targeting decoy receptors HVEM-Ig, LTβR-Ig or sDcR3-Ig, exhibit prolonged graft survival compared to untreated controls, suggesting that LIGHT modulates the course and severity of graft rejection. Therefore, targeting the interaction of LIGHT with HVEM and/or LTβR using recombinant soluble decoy receptors or monoclonal antibodies represent an innovative therapeutic strategy for the prevention and treatment of allograft rejection and for the promotion of donor-specific tolerance.
Collapse
Affiliation(s)
- M-L del Rio
- Transplantation Immunobiology Section, Institute of Biomedicine, University of Leon, Leon, Spain
| | | | | | | | | |
Collapse
|
20
|
Bakdash G, Sittig SP, van Dijk T, Figdor CG, de Vries IJM. The nature of activatory and tolerogenic dendritic cell-derived signal II. Front Immunol 2013; 4:53. [PMID: 23450201 PMCID: PMC3584294 DOI: 10.3389/fimmu.2013.00053] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/11/2013] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are central in maintaining the intricate balance between immunity and tolerance by orchestrating adaptive immune responses. Being the most potent antigen presenting cells, DCs are capable of educating naïve T cells into a wide variety of effector cells ranging from immunogenic CD4+ T helper cells and cytotoxic CD8+ T cells to tolerogenic regulatory T cells. This education is based on three fundamental signals. Signal I, which is mediated by antigen/major histocompatibility complexes binding to antigen-specific T cell receptors, guarantees antigen specificity. The co-stimulatory signal II, mediated by B7 family molecules, is crucial for the expansion of the antigen-specific T cells. The final step is T cell polarization by signal III, which is conveyed by DC-derived cytokines and determines the effector functions of the emerging T cell. Although co-stimulation is widely recognized to result from the engagement of T cell-derived CD28 with DC-expressed B7 molecules (CD80/CD86), other co-stimulatory pathways have been identified. These pathways can be divided into two groups based on their impact on primed T cells. Whereas pathways delivering activatory signals to T cells are termed co-stimulatory pathways, pathways delivering tolerogenic signals to T cells are termed co-inhibitory pathways. In this review, we discuss how the nature of DC-derived signal II determines the quality of ensuing T cell responses and eventually promoting either immunity or tolerance. A thorough understanding of this process is instrumental in determining the underlying mechanism of disorders demonstrating distorted immunity/tolerance balance, and would help innovating new therapeutic approaches for such disorders.
Collapse
Affiliation(s)
- Ghaith Bakdash
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
21
|
Hemingway F, Kashima TG, Knowles HJ, Athanasou NA. Investigation of osteoclastogenic signalling of the RANKL substitute LIGHT. Exp Mol Pathol 2013; 94:380-5. [PMID: 23391709 DOI: 10.1016/j.yexmp.2013.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 01/28/2013] [Indexed: 01/07/2023]
Abstract
LIGHT (TNFSF14) is a member of the TNF superfamily and is known to substitute for RANKL to induce osteoclast differentiation. LIGHT binds HVEM and LTβR, but it is not known whether these receptors play a role in osteoclast formation or whether LIGHT acts via RANKL signalling pathways. We found that both RANKL and LIGHT strongly induced phosphorylation of Akt and NFκB but not JNK in mouse osteoclast precursor cells. The addition of an Akt inhibitor showed decreased osteoclast differentiation and resorption mediated by both RANKL and LIGHT. RT-PCR and FACS analysis showed that CD14(+) human osteoclast precursors expressed HVEM and LTβR; expression levels of HVEM increased in the course of osteoclastogenesis and a decrease in LIGHT expression was associated with an increase in HVEM suggesting that there is a feedback loop related to this receptor. Our findings show that LIGHT is not inhibited by the soluble RANKL receptor OPG and that LIGHT is a potent osteoclastogenesis factor that activates the Akt, NFκB and JNK pathways.
Collapse
Affiliation(s)
- F Hemingway
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford OX3 7HE, United Kingdom
| | | | | | | |
Collapse
|
22
|
Abstract
Upon activation, T cells of various subsets are the most important mediators in cell-mediated immune responses. Activated T cells play an important role in immune system related diseases such as chronic inflammatory diseases, viral infections, autoimmune disease, transplant rejection, Crohn disease, diabetes, and many more. Therefore, efforts have been made to both visualize and treat activated T cells specifically. This review summarizes imaging approaches and selective therapeutics for activated T cells and gives an outlook on how tracking and treating can be combined into theragnositc agents for activated T cells.
Collapse
|
23
|
Sirolimus-based regimen promotes inhibitory costimulatory signal of HVEM/BTLA/CD160/LIGHT pathway in allo-renal recipients. Transpl Immunol 2012; 28:38-47. [PMID: 23165214 DOI: 10.1016/j.trim.2012.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 10/24/2012] [Accepted: 11/09/2012] [Indexed: 02/05/2023]
Abstract
HVEM/BTLA/CD160/LIGHT pathway is a very special costimulatory molecule system which can regulate T-cell immune responses by activating both inflammatory and inhibitory signalings. The regulatory effect of Sirolimus on HVME costimulatory system in allo-renal recipients has not been reported. In this study, we analyzed the expression of HVEM, BTLA, CD160 and LIGHT on circulating T cell subgroups and the expression of HVEM on CD4+ Tregs by flow cytometry and also the pre-dose concentration of Sirolimus by automatic analyzer. Both the allo-renal recipients receiving Sirolimus immunosuppressive regimen and health volunteers were included. The expression of both BTLA and CD160 on T cells increased significantly while the expression of LIGHT on T cells decreased significantly in allo-renal recipients receiving Sirolimus regimen (p<0.05). The expression of HVEM on T cells and CD4+ T-cell subgroup decreased (p<0.05) while that on CD8+ T-cell subgroup remained roughly normal (p>0.05).The expression of HVEM on CD4+ Tregs increased significantly (p<0.05) in allo-renal recipients receiving Sirolimus regimen (p<0.05). Though regulating the expression of HVEM/BTLA/CD160/LIGHT costimulatory system, Sirolimus-based regimen promotes inhibitory costimulatory signal in T cells and enhances the function of CD4+ Tregs in allo-renal recipients, which are in benefit of the control of transplant rejection as well as the induction and maintenance of transplant tolerance.
Collapse
|
24
|
Suzuki JI, Ogawa M, Hirata Y, Nagai R, Isobe M. Effects of immunoglobulin to prevent coronary allograft vasculopathy in heart transplantation. Expert Opin Ther Targets 2012; 16:783-9. [PMID: 22762198 DOI: 10.1517/14728222.2012.697555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Although 100,000 cardiac transplants have been performed, coronary allograft vasculopathy (CAV), which is a phenomenon of chronic rejection, is still a serious problem. AREAS COVERED Several adhesion molecules, cytokines, and chemokines play a critical role in the process. Recent investigations have proved some promising methodologies for preventing or treating rejection. Although immunoglobulins are known to be an effective treatment in many diseases, their effect on cardiac transplantation or CAV is to be elucidated. EXPERT OPINION In this review article, we described some promising methodologies that use immunoglobulins to prevent CAV. Immunoglobulins may be used to prevent CAV.
Collapse
Affiliation(s)
- Jun-ichi Suzuki
- University of Tokyo, Graduate School of Medicine, Department of Advanced Clinical Science and Therapeutics, 7-3-1 Hongo, Bunkyo, Tokyo 113-8655, Japan.
| | | | | | | | | |
Collapse
|
25
|
Heo SK, Ju SA, Kim GY, Park SM, Back SH, Park NH, Min YJ, An WG, Nguyen TTH, Kim SM, Kim BS. The presence of high level soluble herpes virus entry mediator in sera of gastric cancer patients. Exp Mol Med 2012; 44:149-58. [PMID: 22113134 PMCID: PMC3296811 DOI: 10.3858/emm.2012.44.2.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development of gastric cancer (GC) is closely related to chronic inflammation caused by Helicobacter pylori infection, and herpes virus entry mediator (HVEM) is a receptor expressed on the surface of leukocytes that mediates potent inflammatory responses in animal models. However, the role of HVEM in human GC has not been studied. Previously, we showed that the interaction of HVEM on human leukocytes with its ligand LIGHT induces intracellular calcium mobilization, which results in inflammatory responses including induction of proinflammatory cytokine production and anti-bacterial activities. In this study, we report that leukocytes from GC patients express lower levels of membrane HVEM (mHVEM) and have lower LIGHT-induced bactericidal activities than those from healthy controls (HC). In contrast, levels of soluble HVEM (sHVEM) in the sera of GC patients were significantly higher than in those of HC. We found that monocyte membrane-bound HVEM is released into the medium when cells are activated by proinflammatory cytokines such as TNF-α and IL-8, which are elevated in the sera of GC patients. mHVEM level dropped in parallel with the release of sHVEM, and release was completely blocked by the metalloprotease inhibitor, GM6001. We also found that the low level of mHVEM on GC patient leukocytes was correlated with low LIGHT-induced bactericidal activities against H. pylori and S. aureus and production of reactive oxygen species. Our results indicate that mHVEM on leukocytes and sHVEM in sera may contribute to the development and/or progression of GC.
Collapse
Affiliation(s)
- Sook-Kyoung Heo
- Department of Biological Sciences University of Ulsan Ulsan 680-749, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
del Rio ML, Jones ND, Buhler L, Norris P, Shintani Y, Ware CF, Rodriguez-Barbosa JI. Selective blockade of herpesvirus entry mediator-B and T lymphocyte attenuator pathway ameliorates acute graft-versus-host reaction. THE JOURNAL OF IMMUNOLOGY 2012; 188:4885-96. [PMID: 22490863 DOI: 10.4049/jimmunol.1103698] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cosignaling network mediated by the herpesvirus entry mediator (HVEM; TNFRSF14) functions as a dual directional system that involves proinflammatory ligand, lymphotoxin that exhibits inducible expression and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes (LIGHT; TNFSF14), and the inhibitory Ig family member B and T lymphocyte attenuator (BTLA). To dissect the differential contributions of HVEM/BTLA and HVEM/LIGHT interactions, topographically-specific, competitive, and nonblocking anti-HVEM Abs that inhibit BTLA binding, but not LIGHT, were developed. We demonstrate that a BTLA-specific competitor attenuated the course of acute graft-versus-host reaction in a murine F(1) transfer semiallogeneic model. Selective HVEM/BTLA blockade did not inhibit donor T cell infiltration into graft-versus-host reaction target organs, but decreased the functional activity of the alloreactive T cells. These results highlight the critical role of HVEM/BTLA pathway in the control of the allogeneic immune response and identify a new therapeutic target for transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Immunobiology Section, Institute of Biomedicine, University of Leon, 24007 Leon, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
The role of costimulatory receptors of the tumour necrosis factor receptor family in atherosclerosis. J Biomed Biotechnol 2011; 2012:464532. [PMID: 22235167 PMCID: PMC3253462 DOI: 10.1155/2012/464532] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 10/11/2011] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that is mediated by both the innate and adaptive immune responses. T lymphocytes, that together with B cells are the cellular effectors of the adaptive immune system, are currently endowed with crucial roles in the development and progression of atherosclerosis. Costimulatory receptors are a class of molecules expressed by T lymphocytes that regulate the activation of T cells and the generation of effector T-cell responses. In this review we present the roles of costimulatory receptors of the tumour necrosis factor receptor (TNFR) superfamily in atherosclerosis and discuss the implications for future therapies that could be used to specifically modulate the immune response of pathogenic T cells in this disease.
Collapse
|
28
|
Steinberg M, Cheung TC, Ware CF. The signaling networks of the herpesvirus entry mediator (TNFRSF14) in immune regulation. Immunol Rev 2011; 244:169-87. [PMID: 22017438 PMCID: PMC3381650 DOI: 10.1111/j.1600-065x.2011.01064.x] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The tumor necrosis factor (TNF) receptor superfamily member herpesvirus entry mediator (HVEM) (TNFRSF14) regulates T-cell immune responses by activating both inflammatory and inhibitory signaling pathways. HVEM acts as both a receptor for the canonical TNF-related ligands, LIGHT [lymphotoxin-like, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed on T lymphocytes] and lymphotoxin-α, and as a ligand for the immunoglobulin superfamily proteins BTLA (B and T lymphocyte attenuator) and CD160, a feature distinguishing HVEM from other immune regulatory molecules. The ability of HVEM to interact with multiple ligands in distinct configurations creates a functionally diverse set of intrinsic and bidirectional signaling pathways that control both inflammatory and inhibitory responses. The HVEM system is integrated into the larger LTβR and TNFR network through extensive shared ligand and receptor usage. Experimental mouse models and human diseases indicate that dysregulation of HVEM network may contribute to autoimmune pathogenesis, making it an attractive target for drug intervention.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Autoimmunity
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/metabolism
- Gene Expression/immunology
- Herpes Simplex/immunology
- Herpes Simplex/metabolism
- Herpes Simplex/virology
- Herpesvirus 1, Human/immunology
- Humans
- Immunity, Innate
- Lymphocyte Activation
- Lymphotoxin beta Receptor/genetics
- Lymphotoxin beta Receptor/immunology
- Lymphotoxin beta Receptor/metabolism
- Lymphotoxin-alpha/genetics
- Lymphotoxin-alpha/immunology
- Lymphotoxin-alpha/metabolism
- Mice
- Mice, Knockout
- Protein Binding/immunology
- Receptor Cross-Talk/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Receptors, Tumor Necrosis Factor, Member 14/immunology
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
Collapse
Affiliation(s)
| | | | - Carl F. Ware
- Laboratory of Molecular Immunology, Center for Infectious and Inflammatory Diseases, Sanford|Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
29
|
Abstract
Secondary, so-called costimulatory, signals are critically required for the process of T cell activation. Since landmark studies defined that T cells receiving a T cell receptor signal without a costimulatory signal, are tolerized in vitro, the investigation of T cell costimulation has attracted intense interest. Early studies demonstrated that interrupting T cell costimulation allows attenuation of the alloresponse, which is particularly difficult to modulate due to the clone size of alloreactive T cells. The understanding of costimulation has since evolved substantially and now encompasses not only positive signals involved in T cell activation but also negative signals inhibiting T cell activation and promoting T cell tolerance. Costimulation blockade has been used effectively for the induction of tolerance in rodent models of transplantation, but turned out to be less potent in large animals and humans. In this overview we will discuss the evolution of the concept of T cell costimulation, the potential of 'classical' and newly identified costimulation pathways as therapeutic targets for organ transplantation as well as progress towards clinical application of the first costimulation blocking compound.
Collapse
Affiliation(s)
- Nina Pilat
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| | - Mohamed H. Sayegh
- Brigham and Women's Hospital & Children's Hospital Boston, Harvard Medical School, Boston, USA
| | - Thomas Wekerle
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Austria
| |
Collapse
|
30
|
Chittasupho C, Siahaan TJ, Vines CM, Berkland C. Autoimmune therapies targeting costimulation and emerging trends in multivalent therapeutics. Ther Deliv 2011; 2:873-89. [PMID: 21984960 PMCID: PMC3186944 DOI: 10.4155/tde.11.60] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Proteins participating in immunological signaling have emerged as important targets for controlling the immune response. A multitude of receptor-ligand pairs that regulate signaling pathways of the immune response have been identified. In the complex milieu of immune signaling, therapeutic agents targeting mediators of cellular signaling often either activate an inflammatory immune response or induce tolerance. This review is primarily focused on therapeutics that inhibit the inflammatory immune response by targeting membrane-bound proteins regulating costimulation or mediating immune-cell adhesion. Many of these signals participate in larger, organized structures such as the immunological synapse. Receptor clustering and arrangement into organized structures is also reviewed and emerging trends implicating a potential role for multivalent therapeutics is posited.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Chemistry, University of Kansas, KS, USA
- Department of Pharmaceutical Technology, Srinakharinwirot University, Nakhonnayok, Thailand
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, University of Kansas, KS, USA
| | - Charlotte M Vines
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, KS, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, KS, USA
- Department of Pharmaceutical Chemistry, Department of Chemical & Petroleum Engineering, 2030 Becker Drive, Lawrence, KS 66047, USA
| |
Collapse
|
31
|
Kim WK, Sul OJ, Kwak JS, Hur HY, Latour AM, Koller BH, Kwon BS, Jeong CS. Nuclear factor of activated T cells negatively regulates expression of the tumor necrosis factor receptor-related 2 gene in T cells. Exp Mol Med 2011; 42:805-10. [PMID: 20948279 DOI: 10.3858/emm.2010.42.12.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Tumor necrosis factor receptor-related 2 (TR2, HVEM or TNFRSF-14) plays an important role in immune responses, however, the mechanisms regulating its expression are unclear. To understand the control of TR2 gene expression, we studied the upstream region of the gene. Gel supershift assays revealed inducible binding of nuclear factor of activated T cells (NFAT) to a putative NFAT site within the TR2 promoter. Furthermore, cotransfection of a dominant negative NFAT construct, or siRNA for NFAT, resulted in increased expression of a TR2 reporter gene. Our findings demonstrate that NFAT negatively regulates TR2 expression in activated T cells.
Collapse
Affiliation(s)
- Woon-Ki Kim
- Department of Biological Science and the Immunomodulation Research Center, University of Ulsan, Ulsan 680-749, Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang M, Howard K, Winters A, Steavenson S, Anderson S, Smelt S, Doellgast G, Sheelo C, Stevens J, Kim H, Hamburger A, Sein A, Caughey DJ, Lee F, Hsu H, Siu G, Byrne FR. Monoclonal antibodies to B and T lymphocyte attenuator (BTLA) have no effect on in vitro B cell proliferation and act to inhibit in vitro T cell proliferation when presented in a cis, but not trans, format relative to the activating stimulus. Clin Exp Immunol 2010; 163:77-87. [PMID: 21078085 DOI: 10.1111/j.1365-2249.2010.04259.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
B and T lymphocyte attenuator (BTLA) is an immunoglobulin superfamily member surface protein expressed on B and T cells. Its ligand, herpesvirus entry mediator (HVEM), is believed to act as a monomeric agonist that signals via the CRD1 of HVEM to inhibit lymphocyte activation: HVEM is also the receptor for lymphotoxin-α and LIGHT, which both bind in the CRD2 and CRD3 domains of the HVEM molecule, and for CD160 which competes with BTLA. We have shown that recombinant HVEM and a panel of different monoclonal antibodies specifically bind murine BTLA on both B and T cells and that some antibodies inhibit anti-CD3ε-induced T cell proliferation in vitro, but only when constrained appropriately with a putatively cross-linking reagent. The antibodies had no significant effect on in vitro T cell proliferation in a mixed lymphocyte reaction (MLR) assay nor on in vitro DO11.10 antigen-induced T cell proliferation. None of these antibodies, nor HVEM-Fc, had any significant effect on in vitro B cell proliferation induced by anti-immunoglobulin M antibodies (±anti-CD40) or lipopolysaccharide. We further elucidated the requirements for inhibition of in vitro T cell proliferation using a beads-based system to demonstrate that the antibodies that inhibited T cell proliferation in vitro were required to be presented to the T cell in a cis, and not trans, format relative to the anti-CD3ε stimulus. We also found that antibodies that inhibited T cell proliferation in vitro had no significant effect on the antibody captured interleukin-2 associated with the in vivo activation of DO11.10 T cells transferred to syngeneic recipient BALB/c mice. These data suggest that there may be specific structural requirements for the BTLA molecule to exert its effect on lymphocyte activation and proliferation.
Collapse
Affiliation(s)
- M Zhang
- Amgen Inc., Thousand Oaks, CA 91320, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
B and T lymphocyte associated (BTLA) is an Ig domain superfamily protein with cytoplasmic immunoreceptor tyrosine-based inhibitory motifs. Its ligand, herpesvirus entry mediator (HVEM), is a tumor necrosis factor receptor superfamily member. The unique interaction between BTLA and HVEM allows for a system of bidirectional signaling that must be appropriately regulated to balance the outcome of the immune response. HVEM engagement of BTLA produces inhibitory signals through SH2 domain-containing protein tyrosine phosphatase 1 (Shp-1) and Shp-2 association, whereas BTLA engagement of HVEM produces proinflammatory signals via activation of NF-kappaB. The BTLA-HVEM interaction is intriguing and quite complex given that HVEM has four other ligands that also influence immune responses, the conventional TNF ligand LIGHT and lymphotoxin alpha, as well as herpes simplex virus glycoprotein D and the glycosylphosphatidylinositol-linked Ig domain protein CD160. BTLA-HVEM interactions have been shown to regulate responses in several pathogen and autoimmune settings, but our understanding of this complex system of interactions is certainly incomplete. Recent findings of spontaneous inflammation in BTLA-deficient mice may provide an important clue.
Collapse
Affiliation(s)
- Theresa L Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
34
|
Adenovirus-mediated LIGHT gene modification in murine B-cell lymphoma elicits a potent antitumor effect. Cell Mol Immunol 2010; 7:296-305. [PMID: 20418899 DOI: 10.1038/cmi.2010.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.
Collapse
|
35
|
del Rio ML, Lucas CL, Buhler L, Rayat G, Rodriguez-Barbosa JI. HVEM/LIGHT/BTLA/CD160 cosignaling pathways as targets for immune regulation. J Leukoc Biol 2009; 87:223-35. [DOI: 10.1189/jlb.0809590] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
36
|
Wang Y, Zhu M, Miller M, Fu YX. Immunoregulation by tumor necrosis factor superfamily member LIGHT. Immunol Rev 2009; 229:232-43. [PMID: 19426225 DOI: 10.1111/j.1600-065x.2009.00762.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
SUMMARY LIGHT (homologous to lymphotoxins, inducible expression, competes with herpesvirus glycoprotein D for herpesvirus entry mediator, a receptor expressed on T lymphocytes) is a member of the tumor necrosis factor superfamily that contributes to the regulation of immune responses. LIGHT can influence T-cell activation both directly and indirectly by engagement of various receptors that are expressed on T cells and on other types of cells. LIGHT, LIGHT receptors, and their related binding partners constitute a complicated molecular network in the regulation of various processes. The molecular cross-talk among LIGHT and its related molecules presents challenges and opportunities for us to study and to understand the full extent of the LIGHT function. Previous research from genetic and functional studies has demonstrated that dysregulation of LIGHT expression can result in the disturbance of T-cell homeostasis and activation, changing the ability of self-tolerance and of the control of infection. Meanwhile, blockade of LIGHT activity can ameliorate the severity of various T-cell-mediated diseases. These observations indicate the importance of LIGHT and its involvement in many physiological and pathological conditions. Understanding LIGHT interactions offers promising new therapeutic strategies that target LIGHT-engaged pathways to fight against cancer and various infectious diseases.
Collapse
Affiliation(s)
- Yugang Wang
- The Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
37
|
Nolte MA, van Olffen RW, van Gisbergen KPJM, van Lier RAW. Timing and tuning of CD27-CD70 interactions: the impact of signal strength in setting the balance between adaptive responses and immunopathology. Immunol Rev 2009; 229:216-31. [PMID: 19426224 DOI: 10.1111/j.1600-065x.2009.00774.x] [Citation(s) in RCA: 245] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SUMMARY After binding its natural ligand cluster of differentiation 70 (CD70), CD27, a tumor necrosis factor receptor (TNFR)-associated factor-binding member of the TNFR family, regulates cellular activity in subsets of T, B, and natural killer cells as well as hematopoietic progenitor cells. In normal immune responses, CD27 signaling appears to be limited predominantly by the restricted expression of CD70, which is only transiently expressed by cells of the immune system upon activation. Studies performed in CD27-deficient and CD70-transgenic mice have defined a non-redundant role of this receptor-ligand pair in shaping adaptive T-cell responses. Moreover, adjuvant properties of CD70 have been exploited for the design of anti-cancer vaccines. However, continuous CD27-CD70 interactions may cause immune dysregulation and immunopathology in conditions of chronic immune activation such as during persistent virus infection and autoimmune disease. We conclude that optimal tuning of CD27-CD70 interaction is crucial for the regulation of the cellular immune response. We provide a detailed comparison of costimulation through CD27 with its closely related family members 4-1BB (CD137), CD30, herpes virus entry mediator, OX40 (CD134), and glucocorticoid-induced TNFR family-related gene, and we argue that these receptors do not have a unique function per se but that rather the timing, context, and intensity of these costimulatory signals determine the functional consequence of their activity.
Collapse
Affiliation(s)
- Martijn A Nolte
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
38
|
Cai G, Wang H, Qin Q, Zhang J, Zhu Z, Liu M, Shen Q. Amelioration of myocarditis by HVEM-overexpressing dendritic cells through induction of IL-10-producing cells. Cardiovasc Res 2009; 84:425-33. [DOI: 10.1093/cvr/cvp219] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
39
|
Cai G, Freeman GJ. The CD160, BTLA, LIGHT/HVEM pathway: a bidirectional switch regulating T-cell activation. Immunol Rev 2009; 229:244-58. [DOI: 10.1111/j.1600-065x.2009.00783.x] [Citation(s) in RCA: 234] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
Oluwole SF, Oluwole OO, Adeyeri AO, DePaz HA. New strategies in immune tolerance induction. Cell Biochem Biophys 2009; 40:27-48. [PMID: 15289641 DOI: 10.1385/cbb:40:3:27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Induction of tolerance in clinical organ transplantation that will obviate the use of chronic immunosuppression and preserve host immune response to other antigens remains the goal of transplant research. The thymus plays a critical role in the ability of the immune system to discriminate between self- and nonself-antigens or harmful and harmless alloantigens. We now know that multiple factors determine how the immune system responds to a self-antigen or foreign antigen. These determinants include developmental stage of the host, stage of T-cell maturity, site of antigen encounter, type and maturity of antigen-presenting cells, and presence and type of costimulatory molecules. Our understanding of the mechanisms of T-cell interactions with peptide/ major histocompatibility complex in peripheral lymphoid organs has led to experiments that translate into peripheral T-cell tolerance. The induction of high-avidity peripheral alloreactive T cells in the early phase of organ transplantation makes it difficult to achieve long-term alloantigen-specific tolerance without the use of transient perioperative immunosuppression. Therefore, protocols that induce robust tolerance in rodent and nonhuman primate models involve the use of donor antigen combined with a short course of perioperative immunosuppression. These studies suggest that the underlying mechanisms of peripheral tolerance include deletion, anergy, immune deviation, and regulatory T cells. This review focuses on recent advances in tolerance induction in experimental animal models and discusses their relevance to the development of protocols for the induction and maintenance of clinical transplant tolerance.
Collapse
Affiliation(s)
- Soji F Oluwole
- Department of Surgery, Columbia University, College of Physicians and Surgeons, New York, NY, USA.
| | | | | | | |
Collapse
|
41
|
Ware CF. Targeting the LIGHT-HVEM Pathway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:146-55. [DOI: 10.1007/978-0-387-89520-8_10] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Kober J, Leitner J, Klauser C, Woitek R, Majdic O, Stöckl J, Herndler-Brandstetter D, Grubeck-Loebenstein B, Reipert BM, Pickl WF, Pfistershammer K, Steinberger P. The capacity of the TNF family members 4-1BBL, OX40L, CD70, GITRL, CD30L and LIGHT to costimulate human T cells. Eur J Immunol 2008; 38:2678-88. [PMID: 18825741 PMCID: PMC2975061 DOI: 10.1002/eji.200838250] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Activating signals generated by members of the tumour necrosis factor receptor superfamily upon interaction with their cognate ligands play important roles in T-cell responses. Members of the tumour necrosis factor family namely 4-1BBL, OX40L, CD70, GITRL, LIGHT and CD30L have been described to function as costimulatory molecules by binding such receptors on T cells. Using our recently described system of T-cell stimulator cells we have performed the first study where all these molecules have been assessed and compared regarding their capacity to costimulate proliferation and cytokine production of human T cells. 4-1BBL, which we found to be the most potent molecule in this group, was able to mediate sustained activation and proliferation of human T cells. OX40L and CD70 were also strong inducers of T-cell proliferation, whereas the costimulatory capacity of human GITRL was significantly lower. Importantly CD30L and LIGHT consistently failed to act costimulatory on human T cells, and we therefore suggest that these molecules might be functionally distinct from the costimulatory members of this family.
Collapse
Affiliation(s)
- Johanna Kober
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Christoph Klauser
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ramona Woitek
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Otto Majdic
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Beatrix Grubeck-Loebenstein
- Immunology Division, Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | - Winfried F. Pickl
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katharina Pfistershammer
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
del Rio ML, Buhler L, Gibbons C, Tian J, Rodriguez-Barbosa JI. PD-1/PD-L1, PD-1/PD-L2, and other co-inhibitory signaling pathways in transplantation. Transpl Int 2008; 21:1015-28. [PMID: 18662368 DOI: 10.1111/j.1432-2277.2008.00726.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Transplantation of cells, tissues and vascularized solid organs is a successful therapeutic intervention for many end-stage chronic diseases. The combination of co-stimulatory blockade with the delivery of negative signals to T cells through co-inhibitory receptors would provide a robust approach to modulating T-cell receptor signaling and improving alloantigen-specific control of transplant rejection. This approach based on fundamental knowledge of APC/T-cell interactions may complement conventional therapies in the near future to reinforce long-term allograft survival, and permit minimal immunosuppression. The focus of this review was primarily on two major co-inhibitory signaling pathways, namely PD-1/PD-L1/PD-L2 and BTLA/CD160/HVEM/LIGHT that have been thoroughly characterized in murine models of transplantation using genetically modified mice, specific monoclonal antibodies and fusion proteins.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Laboratory of Immunobiology, School of Biotechnology and Institute of Biomedicine, University of Leon, Leon, Spain
| | | | | | | | | |
Collapse
|
44
|
Tao R, Wang L, Murphy KM, Fraser CC, Hancock WW. Regulatory T cell expression of herpesvirus entry mediator suppresses the function of B and T lymphocyte attenuator-positive effector T cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:6649-55. [PMID: 18453584 DOI: 10.4049/jimmunol.180.10.6649] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The binding of herpesvirus entry mediator (HVEM) to B and T lymphocyte attenuator (BTLA) is known to activate an inhibitory signaling cascade in effector T (Teff) cells, but we now report that the HVEM-BTLA pathway is also important to the suppressive function of regulatory T cells (Tregs). Although naive T cells up-regulated BTLA upon TCR activation, Treg expression of BTLA remained low, regardless of TCR activation. Moreover, BTLA(-/-) CD4(+)CD25(+) Tregs had normal suppressive activity, whereas BTLA(-/-) Teff cells were more resistant than wild-type Teff cells to suppression by Tregs, suggesting BTLA expression by Teff cells was required for their suppression by Tregs. In contrast to BTLA, HVEM expression was comparable in naive Tregs vs Teff cells, but after stimulation HVEM expression was quickly down-regulated by Teff cells, whereas HVEM was further up-regulated by Tregs. HVEM(-/-) Tregs had decreased suppressive activity as compared with wild-type Tregs, indicating that Treg expression of HVEM was required for optimal suppression. Consistent with this, T cells from Scurfy mice (FoxP3 mutant) lacked HVEM gene expression, and adoptively transferred wild-type but not HVEM(-/-) Tregs were able to control alloresponses in vivo by normal Teff cells. Our data demonstrate that Tregs can exert their effects via up-regulation of the negative costimulatory ligand HVEM, which upon binding to BTLA expressed by Teff cells helps mediate the suppressive functions of Tregs in vitro and in vivo.
Collapse
Affiliation(s)
- Ran Tao
- Department of Pathology and Laboratory Medicine, Joseph Stokes Jr Research Institute and Biesecker Pediatric Liver Center, The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
SUMMARY Cytokines mediate key communication pathways essential for regulation of immune responses. Full activation of antigen-responding lymphocytes requires cooperating signals from the tumor necrosis factor (TNF)-related cytokines and their specific receptors. LIGHT, a lymphotoxin-beta (LTbeta)-related TNF family member, modulates T-cell activation through two receptors, the herpesvirus entry mediator (HVEM) and indirectly through the LT-beta receptor. An unexpected finding revealed a non-canonical binding site on HVEM for the immunoglobulin superfamily member, B and T lymphocyte attenuator (BTLA), and an inhibitory signaling protein suppressing T-cell activation. Thus, HVEM can act as a molecular switch between proinflammatory and inhibitory signaling. The non-canonical HVEM-BTLA pathway also acts to counter LTbetaR signaling that promotes the proliferation of antigen-presenting dendritic cells (DCs) within lymphoid tissue microenvironments. These results indicate LTbeta receptor and HVEM-BTLA pathways form an integrated signaling circuit. Targeting these cytokine pathways with specific antagonists (antibody or decoy receptor) can alter lymphocyte differentiation and activation. Alternately, agonists directed at their cell surface receptors can restore homeostasis and potentially reset immune and inflammatory processes, which may be useful in treating autoimmune and infectious diseases and cancer.
Collapse
Affiliation(s)
- Carl F Ware
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
46
|
Xu G, Liu D, Okwor I, Wang Y, Korner H, Kung SKP, Fu YX, Uzonna JE. LIGHT Is critical for IL-12 production by dendritic cells, optimal CD4+ Th1 cell response, and resistance to Leishmania major. THE JOURNAL OF IMMUNOLOGY 2007; 179:6901-9. [PMID: 17982081 DOI: 10.4049/jimmunol.179.10.6901] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although studies indicate LIGHT (lymphotoxin (LT)-like, exhibits inducible expression and competes with HSV glycoprotein D for herpes virus entry mediator (HVEM), a receptor expressed by T lymphocytes) enhances inflammation and T cell-mediated immunity, the mechanisms involved in this process remain obscure. In this study, we assessed the role of LIGHT in IL-12 production and development of CD4(+) Th cells type one (Th1) in vivo. Bone marrow-derived dendritic cells from LIGHT(-/-) mice were severely impaired in IL-12p40 production following IFN-gamma and LPS stimulation in vitro. Furthermore, blockade of LIGHT in vitro and in vivo with HVEM-Ig and LT beta receptor (LTbetaR)-Ig leads to impaired IL-12 production and defective polyclonal and Ag-specific IFN-gamma production in vivo. In an infection model, injection of HVEM-Ig or LTbetaR-Ig into the usually resistant C57BL/6 mice results in defective IL-12 and IFN-gamma production and severe susceptibility to Leishmania major that was reversed by rIL-12 treatment. This striking susceptibility to L. major in mice injected with HVEM-Ig or LTbetaR-Ig was also reproduced in LIGHT(-/-) --> RAG1(-/-) chimeric mice. In contrast, L. major-infected LTbeta(-/-) mice do not develop acute disease, suggesting that the effect of LTbetaR-Ig is not due to blockade of membrane LT (LTalpha1beta2) signaling. Collectively, our data show that LIGHT plays a critical role for optimal IL-12 production by DC and the development of IFN-gamma-producing CD4(+) Th1 cells and its blockade results in severe susceptibility to Leishmania major.
Collapse
Affiliation(s)
- Guilian Xu
- Department of Immunology, Faculty of Medicine University of Manitoba, Winnipeg, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Sul OJ, Lee HW, Kim WK, Choi JH, Kwak JS, Choi YJ, Latour AM, Koller BH, Jeong CS, Kwon BS. Regulation of the murine TR2/HVEM gene expression by IRF. Biochem Biophys Res Commun 2007; 367:277-83. [PMID: 18155158 DOI: 10.1016/j.bbrc.2007.12.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 12/12/2007] [Indexed: 10/22/2022]
Abstract
TR2 (TNFR-related 2, HVEM, or TNFRSF-14), a member of the TNFR family, is involved in a number of immune responses. While TR2 is expressed on the surface of T cells during the resting state, little is known regarding how expression of the TR2 gene is regulated. To understand the mechanisms regulating the expression of TR2 in T cells, we analyzed the 5' flanking region of TR2. We identified an important region for the activity of the TR2 promoter using site directed mutagenesis. Using EMSA analysis, we found that IRF-2 was bound to the promoter region of the TR2 gene during the resting state of EL-4 T cells. Transfection of IRF-2 expression plasmid and of dominant negative IRF-2 mutant further confirmed our results. Together, these data demonstrate that IRF-2 is involved in the regulation of TR2 expression in EL-4 T cells.
Collapse
Affiliation(s)
- Ok-Ju Sul
- Department of Biological Science and the Immunomodulation Research Center, University of Ulsan, San29 Mugeo-Dong, Nam-Gu, Ulsan 680-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kingsley CI, Nadig SN, Wood KJ. Transplantation tolerance: lessons from experimental rodent models. Transpl Int 2007; 20:828-41. [PMID: 17711408 PMCID: PMC2156188 DOI: 10.1111/j.1432-2277.2007.00533.x] [Citation(s) in RCA: 52] [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: 04/26/2007] [Revised: 05/23/2007] [Accepted: 07/10/2007] [Indexed: 12/30/2022]
Abstract
Immunological tolerance or functional unresponsiveness to a transplant is arguably the only approach that is likely to provide long-term graft survival without the problems associated with life-long global immunosuppression. Over the past 50 years, rodent models have become an invaluable tool for elucidating the mechanisms of tolerance to alloantigens. Importantly, rodent models can be adapted to ensure that they reflect more accurately the immune status of human transplant recipients. More recently, the development of genetically modified mice has enabled specific insights into the cellular and molecular mechanisms that play a key role in both the induction and maintenance of tolerance to be obtained and more complex questions to be addressed. This review highlights strategies designed to induce alloantigen specific immunological unresponsiveness leading to transplantation tolerance that have been developed through the use of experimental models.
Collapse
Affiliation(s)
- Cherry I Kingsley
- Transplantation Research Immunology Group, Nuffield Department of Surgery, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
49
|
Oxelius VA. Personal glimpses of Robert A. Good. Immunol Res 2007. [DOI: 10.1007/s12026-007-0008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Fan K, Wang H, Wei H, Zhou Q, Kou G, Hou S, Qian W, Dai J, Li B, Zhang Y, Zhu T, Guo Y. Blockade of LIGHT/HVEM and B7/CD28 Signaling Facilitates Long-Term Islet Graft Survival With Development of Allospecific Tolerance. Transplantation 2007; 84:746-54. [PMID: 17893608 DOI: 10.1097/01.tp.0000280545.14489.df] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Previous studies have shown that blockade of LIGHT, a T-cell costimulatory molecule belonging to the tumor necrosis factor (TNF) superfamily, by soluble lymphotoxin beta receptor-Ig (LTbetaR-Ig) inhibited the development of graft-versus-host disease. The cardiac allografts were significantly prolonged in LIGHT deficient mice. No data are yet available regarding the role of the LIGHT/HVEM pathway in more stringent fully allogeneic models such as skin and islet transplantation models. METHODS Streptozotocin-induced chemical diabetic BALB/C mice underwent transplantation with allogeneic C57BL/6 islets and were treated with LTbetaR-Ig, CTLA4-Ig or a combination of both in the early peritransplant period. RESULTS Administration of CTLA4-Ig or LTbeta R-Ig alone only increased graft survival to 55 days and 27 days respectively, whereas simultaneous blockade of both pathways significantly prolonged the islet allograft survival for more than 100 days. Long-term survivors were retransplanted with donor-specific (C57BL/6) islets and the grafted islets remained functional for more than 100 days. All of islet allografts were protected against rejection when the mixtures of 1x10(6) CD4+ T cells from tolerant mice and islet allografts were cotransplanted under the renal capsule of the naïve BALB/c recipients. CONCLUSIONS These data indicate that: 1) a synergistic effect for prolonged graft survival can be obtained by simultaneously blocking LIGHT and CD28 signaling in the stringent model of islet allotransplantation; 2) development of donor-specific immunological tolerance is associated with the presence of regulatory T-cell activity; and 3) local cotransplantation of the allografts with the regulatory T cells can effectively prevent allograft rejection and induce donor-specific tolerance in lymphocytes-sufficient recipients.
Collapse
Affiliation(s)
- Kexing Fan
- International Joint Cancer Institute, The Second Military Medical University, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|