51
|
Porchia BFMM, Diniz MO, Cariri FAMO, Santana VC, Amorim JH, Balan A, Braga CJM, Ferreira LCS. Purified herpes simplex type 1 glycoprotein D (gD) genetically fused with the type 16 human papillomavirus E7 oncoprotein enhances antigen-specific CD8+ T cell responses and confers protective antitumor immunity. Mol Pharm 2011; 8:2320-30. [PMID: 21985578 DOI: 10.1021/mp200194s] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Type 1 herpes virus (HSV-1) glycoprotein D (gD) enhances antigen-specific immune responses, particularly CD8(+) T cell responses, in mice immunized with DNA vaccines encoding hybrid proteins genetically fused with the target antigen at a site near the C-terminal end. These effects are attributed to the interaction of gD with the herpes virus entry mediator (HVEM) and the concomitant blockade of a coinhibitory mechanism mediated by the B- and T-lymphocyte attenuator (BTLA). However, questions concerning the requirement for endogenous synthesis of the antigen or the adjuvant/antigen fusion itself have not been addressed so far. In the present study, we investigated these points using purified recombinant gDs, genetically fused or not with type 16 papilloma virus (HPV-16) E7 oncoprotein. Soluble recombinant gDs, but not denatured forms, retained the ability to bind surface-exposed cellular receptors of HVEM-expressing U937 cells. In addition, in vivo administration of the recombinant proteins, particularly gD genetically fused with E7 (gDE7), promoted the activation of dendritic cells (DC) and antigen-specific cytotoxic CD8(+) T cells. More relevantly, mice immunized with the gDE7 protein developed complete preventive and partial therapeutic antitumor protection, as measured in mice following the implantation of TC-1 cells expressing HPV-16 oncoproteins. Collectively, these results demonstrate that the T cell adjuvant effects of the HSV-1 gD protein did not require endogenous synthesis and could be demonstrated in mice immunized with purified recombinant proteins.
Collapse
Affiliation(s)
- Bruna F M M Porchia
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Park JJ, Anand S, Zhao Y, Matsumura Y, Sakoda Y, Kuramasu A, Strome SE, Chen L, Tamada K. Expression of anti-HVEM single-chain antibody on tumor cells induces tumor-specific immunity with long-term memory. Cancer Immunol Immunother 2011; 61:203-214. [PMID: 21877247 DOI: 10.1007/s00262-011-1101-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/16/2011] [Indexed: 01/01/2023]
Abstract
Genetic engineering of tumor cells to express immune-stimulatory molecules, including cytokines and co-stimulatory ligands, is a promising approach to generate highly efficient cancer vaccines. The co-signaling molecule, LIGHT, is particularly well suited for use in vaccine development as it delivers a potent co-stimulatory signal through the Herpes virus entry mediator (HVEM) receptor on T cells and facilitates tumor-specific T cell immunity. However, because LIGHT binds two additional receptors, lymphotoxin β receptor and Decoy receptor 3, there are significant concerns that tumor-associated LIGHT results in both unexpected adverse events and interference with the ability of the vaccine to enhance antitumor immunity. In order to overcome these problems, we generated tumor cells expressing the single-chain variable fragment (scFv) of anti-HVEM agonistic mAb on the cell surface. Tumor cells expressing anti-HVEM scFv induce a potent proliferation and cytokine production of co-cultured T cells. Inoculation of anti-HVEM scFv-expressing tumor results in a spontaneous tumor regression in CD4+ and CD8+ T cell-dependent fashion, associated with the induction of tumor-specific long-term memory. Stimulation of HVEM and 4-1BB co-stimulatory signals by anti-HVEM scFv-expressing tumor vaccine combined with anti-4-1BB mAb shows synergistic effects which achieve regression of pre-established tumor and T cell memory specific to parental tumor. Taken in concert, our data suggest that genetic engineering of tumor cells to selectively potentiate the HVEM signaling pathway is a promising antitumor vaccine therapy.
Collapse
Affiliation(s)
- Jang-June Park
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Sudarshan Anand
- Department of Pathology and Moores UCSD Cancer Center, University of California, San Diego, CA, USA
| | - Yuming Zhao
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Yumiko Matsumura
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Yukimi Sakoda
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA
| | - Atsuo Kuramasu
- Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Scott E Strome
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA.,Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lieping Chen
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - Koji Tamada
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Baltimore, 655 W. Baltimore St. BRB 9-051, Baltimore, MD, 21201, USA. .,Yamaguchi University Graduate School of Medicine, Ube, Japan. .,Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
53
|
Schaer C, Hiltbrunner S, Ernst B, Mueller C, Kurrer M, Kopf M, Harris NL. HVEM signalling promotes colitis. PLoS One 2011; 6:e18495. [PMID: 21533159 PMCID: PMC3078914 DOI: 10.1371/journal.pone.0018495] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 03/08/2011] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Tumor necrosis factor super family (TNFSF) members regulate important processes involved in cell proliferation, survival and differentiation and are therefore crucial for the balance between homeostasis and inflammatory responses. Several members of the TNFSF are closely associated with inflammatory bowel disease (IBD). Thus, they represent interesting new targets for therapeutic treatment of IBD. METHODOLOGY/PRINCIPAL FINDINGS We have used mice deficient in TNFSF member HVEM in experimental models of IBD to investigate its role in the disease process. Two models of IBD were employed: i) chemical-induced colitis primarily mediated by innate immune cells; and ii) colitis initiated by CD4(+)CD45RB(high) T cells following their transfer into immuno-deficient RAG1(-/-) hosts. In both models of disease the absence of HVEM resulted in a significant reduction in colitis and inflammatory cytokine production. CONCLUSIONS These data show that HVEM stimulatory signals promote experimental colitis driven by innate or adaptive immune cells.
Collapse
Affiliation(s)
- Corinne Schaer
- Molecular Biomedicine, Institute of Integrative Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Stefanie Hiltbrunner
- Molecular Biomedicine, Institute of Integrative Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Bettina Ernst
- Molecular Biomedicine, Institute of Integrative Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Michael Kurrer
- Institute of Pathology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Integrative Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Nicola L. Harris
- Swiss Vaccine Research Institute and Global Health Institute, Ecole Polytechnique Fédérale, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
54
|
T-cell activation triggers death receptor-6 expression in a NF-κB and NF-AT dependent manner. Mol Immunol 2011; 48:1439-47. [PMID: 21501873 DOI: 10.1016/j.molimm.2011.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 03/21/2011] [Accepted: 03/28/2011] [Indexed: 11/23/2022]
Abstract
Death receptor-6 (DR6) apparently participates in the regulation of T-cell activation and/or activity as its genetic disruption results in enhanced CD4+ T-cell expansion, the production of Th2 cytokines, and interestingly also the compromised migration of CD4+ T cells to sites of inflammation. However, the mechanism of regulation of DR6 expression in cells of the immune system is not fully understood. In this communication we show that DR6 is not expressed in resting T cells from human peripheral blood or murine lymph nodes but that its expression is significantly upregulated in CD3 crosslinking- or PMA/ionomycin-activated T lymphocytes. DR6 expression is transiently increased in both activated human CD4+ and CD8+ T cells and it is apparently dependent on the activation of NF-κB and NF-AT signaling pathways. In contrast to primary peripheral blood T cells, the widely used model lymphoblastic leukemia T-cell line Jurkat is DR6-positive and unexpectedly, TCR-mediated stimulation of Jurkat cells strongly downregulates DR6 expression via suppression of its transcription.
Collapse
|
55
|
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
|
56
|
Han B, Wu LQ, Ma X, Wang ZH, Li JP, Bi CY, Yong S. Synergistic effect of IFN-γ gene on LIGHT-induced apoptosis in HepG2 cells via down regulation of Bcl-2. ACTA ACUST UNITED AC 2010; 39:228-38. [PMID: 21117871 DOI: 10.3109/10731199.2010.538403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
To detect the expression of anti-apoptotic factor Bcl-2 and Survivin in transferred HepG2 cells and evaluate the synergistic effect of IFN-γ gene on LIGHT-induced apoptosis signal transduction pathways, the full-length ORF of LIGHT and IFN-γ gene were cloned into pcDNA4 and verified by DNA sequencing. After being optimized by EGFP, recombinant LIGHT and IFN-γ were transferred into the HepG2 cells mediated by a cationic liposome in vitro. The expression of LIGHT and IFN-γ was identified in the supernatants by ELISA. The HepG2 cells were divided into three groups: the control, LIGHT gene transfection alone, and simultaneous transfection of LIGHT and IFN-γ genes. The cell apoptosis and expression of Bcl-2 and Survivin in cell lysate were detected through FCM. After transfection, the apoptosis rate of HepG2 cells was increased with the prolonged time, and the apoptosis rate of LIGHT group was higher than the control group, while the LIGHT/IFN-γ group was higher than the LIGHT group P < 0.01). The expression of Bcl-2 and Survivin in LIGHT group and LIGHT/IFN-γ group decreased dramatically compared with the control group. LIGHT gene alone can result in significant inhibition of HepG2 cells proliferation. INF-γ can synergistically precede LIGHT-induced apoptotic processes through down-regulation of Bcl-2 expression, but not survivin expression.
Collapse
Affiliation(s)
- Bing Han
- Department of Hepatobiliary Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao, Shandong, China
| | | | | | | | | | | | | |
Collapse
|
57
|
Abstract
PURPOSE OF REVIEW The nonimmune effects of currently used immunosuppressive drugs result in a high incidence of late graft loss due to nephrotoxicity and death. As an immune-specific alternative to conventional immunosuppressants, new biotechnology tools can be used to block the costimulation signal of T-cell activation. RECENT FINDINGS Many experimental studies, particularly preclinical studies in nonhuman primates, have focused on blocking 'classical' B7/CD28 and CD40/CD40L pathways, which are critical in primary T-cell activation, but also on new B7/CD28 and TNF/TNF-R pathways families of costimulatory molecules that can deliver positive or negative costimulation signals to regulate the alloimmune response. SUMMARY Belatacept is a new fusion protein derived from CTLA4-Ig that can be used to prevent acute rejection in renal transplantation instead of calcineurin inhibitors. Belatacept can also prevent acute rejection efficiently in humans and, more interestingly, can improve renal function and cardiovascular risk factors in this population.
Collapse
|
58
|
Herpes simplex virus glycoprotein D interferes with binding of herpesvirus entry mediator to its ligands through downregulation and direct competition. J Virol 2010; 84:11646-60. [PMID: 20826693 DOI: 10.1128/jvi.01550-10] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To initiate membrane fusion and virus entry, herpes simplex virus (HSV) gD binds to a cellular receptor such as herpesvirus entry mediator (HVEM). HVEM is a tumor necrosis factor (TNF) receptor family member with four natural ligands that either stimulate (LIGHT and LTα) or inhibit (BTLA and CD160) T cell function. We hypothesized that the interaction of gD with HVEM affects the binding of natural ligands, thereby modulating the immune response during infection. Here, we investigated the effect that gD has on the interaction of HVEM with its natural ligands. First, HSV gD on virions or cells downregulates HVEM from the cell surface. Similarly, trans-interaction with BTLA or LIGHT also downregulates HVEM from the cell surface, suggesting that HSV may subvert a natural mechanism for regulating HVEM activity. Second, we showed that wild-type gD had the lowest affinity for HVEM compared with the four natural ligands. Moreover, gD directly competed for binding to HVEM with BTLA but not LTα or LIGHT, indicating the possibility that gD selectively controls HVEM signals. On the other hand, natural ligands influence the use of HVEM by HSV. For instance, soluble BTLA, LTα, and LIGHT inhibited the binding of wild-type gD to HVEM, and soluble BTLA and LTα blocked HSV infection of HVEM-expressing cells. Thus, gD is at the center of the interplay between HVEM and its ligands. It can interfere with HVEM function in two ways, by competing with the natural ligands and by downregulating HVEM from the cell surface.
Collapse
|
59
|
Peña-Cruz V, McDonough SM, Diaz-Griffero F, Crum CP, Carrasco RD, Freeman GJ. PD-1 on immature and PD-1 ligands on migratory human Langerhans cells regulate antigen-presenting cell activity. J Invest Dermatol 2010; 130:2222-30. [PMID: 20445553 PMCID: PMC2927196 DOI: 10.1038/jid.2010.127] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Langerhans cells (LCs) are known as "sentinels" of the immune system that function as professional antigen-presenting cells (APCs) after migration to draining lymph node. LCs are proposed to have a role in tolerance and the resolution of cutaneous immune responses. The Programmed Death-1 (PD-1) receptor and its ligands, PD-L1 and PD-L2, are a co-inhibitory pathway that contributes to the negative regulation of T-lymphocyte activation and peripheral tolerance. Surprisingly, we found PD-1 to be expressed on immature LCs (iLCs) in situ. PD-1 engagement on iLCs reduced IL-6 and macrophage inflammatory protein (MIP)-1alpha cytokine production in response to TLR2 signals but had no effect on LC maturation. PD-L1 and PD-L2 were expressed at very low levels on iLCs. Maturation of LCs upon migration from epidermis led to loss of PD-l expression and gain of high expression of PD-L1 and PD-L2 as well as co-stimulatory molecules. Blockade of PD-L1 and/or PD-L2 on migratory LCs (mLCs) and DDCs enhanced T-cell activation, as has been reported for other APCs. Thus the PD-1 pathway is active in iLCs and inhibits iLC activities, but expression of receptor and ligands reverses upon maturation and PD-L1 and PD-L2 on mLC function to inhibit T-cell responses.
Collapse
Affiliation(s)
- Victor Peña-Cruz
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Sean M. McDonough
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Felipe Diaz-Griffero
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Christopher P. Crum
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ruben D. Carrasco
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
60
|
Serriari NE, Gondois-Rey F, Guillaume Y, Remmerswaal EBM, Pastor S, Messal N, Truneh A, Hirsch I, van Lier RAW, Olive D. B and T lymphocyte attenuator is highly expressed on CMV-specific T cells during infection and regulates their function. THE JOURNAL OF IMMUNOLOGY 2010; 185:3140-8. [PMID: 20693422 DOI: 10.4049/jimmunol.0902487] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B and T lymphocyte attenuator (BTLA), like its relative programmed cell death-1 (PD-1), is a receptor that negatively regulates murine T cell activation. However, its expression and function on human T cells is currently unknown. We report in this study on the expression of BTLA in human T cell subsets as well as its regulation on virus-specific T cells during primary human CMV infection. BTLA is expressed on human CD4(+) T cells during different stages of differentiation, whereas on CD8(+) T cells, it is found on naive T cells and is progressively downregulated in memory and differentiated effector-type cells. During primary CMV infection, BTLA was highly induced on CMV-specific CD8(+) T cells immediately following their differentiation from naive cells. After control of CMV infection, BTLA expression went down on memory CD8(+) cells. Engagement of BTLA by mAbs blocked CD3/CD28-mediated T cell proliferation and Th1 and Th2 cytokine secretion. Finally, in vitro blockade of the BTLA pathway augmented, as efficient as anti-PD-1 mAbs, allogeneic as well as CMV-specific CD8(+) T cell proliferation. Thus, our results suggest that, like PD-1, BTLA provides a potential target for enhancing the functional capacity of CTLs in viral infections.
Collapse
Affiliation(s)
- Nacer-Eddine Serriari
- Institut National de Santé et de Recherche Médicale Unité Mixte de Recherche 891, Institut Paoli Calmettes, Université de Méditerranée, Infrastructures en Biologie Sante et Agronomie Cancer Immunomonitoring Platform, Institut Fédératif de Recherche 137, Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
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
|
62
|
Kovacs SK, Tiwari V, Prandovszky E, Dosa S, Bacsa S, Valyi-Nagy K, Shukla D, Valyi-Nagy T. Expression of herpes virus entry mediator (HVEM) in the cornea and trigeminal ganglia of normal and HSV-1 infected mice. Curr Eye Res 2010; 34:896-904. [PMID: 19895317 DOI: 10.3109/02713680903184250] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Herpes virus entry mediator (HVEM) plays a critical role in the regulation of inflammation through interaction with its natural ligands LIGHT and lymphotoxin alpha and also serves as one of the entry receptors of herpes simplex virus (HSV). The purpose of this study was to better understand the expression of HVEM in the cornea and trigeminal ganglia (TG), which are important targets of HSV infection. MATERIALS AND METHODS Immunohistochemistry was used to define HVEM expression in the cornea and TG of normal and HSV-1 infected mice euthanized 2 to 5 days or 7 months following corneal inoculation of virus. RESULTS We found that HVEM is widely expressed in the normal corneal epithelium and endothelium, is weakly and focally expressed in the corneal stroma, and is expressed in a portion of neurons and non-neuronal cells in the TG. Acute HSV-1 keratitis and ganglionitis were associated with increased HVEM expression in the corneal epithelium and stroma and in neurons and non-neuronal cells of TG, and many inflammatory cells in these tissues also expressed HVEM. TG derived from mice 7 months after virus inoculation demonstrated latent HSV-1 infection that was associated with increased HVEM expression in neurons and non-neuronal cells relative to uninfected control tissues. Latent TG also contained focal infiltrates of mononuclear inflammatory cells, many of which expressed HVEM. Corneas derived from latently infected mice demonstrated chronic keratitis, with no evidence of virus replication or increased HVEM expression in the corneal epithelium, and inflammatory cells present showed only weak HVEM expression. CONCLUSIONS HVEM is expressed in the cornea and TG and therefore may serve as an HSV entry receptor in these tissues. Furthermore, these findings raise the possibility that changes in HVEM expression following ocular HSV-1 infection can modulate HSV spread and infection-induced inflammation in the cornea and TG.
Collapse
Affiliation(s)
- S Krisztian Kovacs
- Department of Pathology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Derré L, Rivals JP, Jandus C, Pastor S, Rimoldi D, Romero P, Michielin O, Olive D, Speiser DE. BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination. J Clin Invest 2009; 120:157-67. [PMID: 20038811 DOI: 10.1172/jci40070] [Citation(s) in RCA: 225] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 10/07/2009] [Indexed: 12/12/2022] Open
Abstract
The function of antigen-specific CD8+ T cells, which may protect against both infectious and malignant diseases, can be impaired by ligation of their inhibitory receptors, which include CTL-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1). Recently, B and T lymphocyte attenuator (BTLA) was identified as a novel inhibitory receptor with structural and functional similarities to CTLA-4 and PD-1. BTLA triggering leads to decreased antimicrobial and autoimmune T cell responses in mice, but its functions in humans are largely unknown. Here we have demonstrated that as human viral antigen-specific CD8+ T cells differentiated from naive to effector cells, their surface expression of BTLA was gradually downregulated. In marked contrast, human melanoma tumor antigen-specific effector CD8+ T cells persistently expressed high levels of BTLA in vivo and remained susceptible to functional inhibition by its ligand herpes virus entry mediator (HVEM). Such persistence of BTLA expression was also found in tumor antigen-specific CD8+ T cells from melanoma patients with spontaneous antitumor immune responses and after conventional peptide vaccination. Remarkably, addition of CpG oligodeoxynucleotides to the vaccine formulation led to progressive downregulation of BTLA in vivo and consequent resistance to BTLA-HVEM-mediated inhibition. Thus, BTLA activation inhibits the function of human CD8+ cancer-specific T cells, and appropriate immunotherapy may partially overcome this inhibition.
Collapse
Affiliation(s)
- Laurent Derré
- Ludwig Institute for Cancer Research, Hôpital Orthopédique, Niveau 5 Est, Av. Pierre-Decker 4, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
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: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
65
|
Krege J, Seth S, Hardtke S, Davalos-Misslitz ACM, Förster R. Antigen-dependent rescue of nose-associated lymphoid tissue (NALT) development independent of LTbetaR and CXCR5 signaling. Eur J Immunol 2009; 39:2765-78. [PMID: 19757439 DOI: 10.1002/eji.200939422] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Nose-associated lymphoid tissue (NALT) in the rodent upper respiratory tract develops postnatally and is considered to be independent of several factors known to be involved in the organogenesis of LN and Peyer's patches (PP). In this study we demonstrate that at least two different pathways result in NALT development. Following NALT anlage formation the intrinsic pathway relies on a signaling cascade including those mediated through the chemokine receptor CXCR5 and the lymphotoxin beta receptor (LTbetaR). This allows for the formation of high endothelial venules and thereby the recruitment of lymphocytes into NALT. Alternatively, high endothelial venule formation and lymphocyte recruitment can be induced in the NALT anlage by environmental signals, which are independent of LT-betaR and chemokine receptor CXCR5 signaling but in part rely on CD40 ligand. Thus, our study identifies a novel mechanism that facilitates the rescue of NALT development at late stages in adult life independent of the canonical LTbetaR-CXCR5 signaling axis.
Collapse
Affiliation(s)
- Janet Krege
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | | | |
Collapse
|
66
|
Morishige T, Yoshioka Y, Tanabe A, Yao X, Mizuguchi H, Tsunoda SI, Tsutsumi Y, Mukai Y, Okada N, Nakagawa S. Comparison of the anti-tumor activity of native, secreted, and membrane-bound LIGHT in mouse tumor models. Int Immunopharmacol 2009; 10:26-33. [PMID: 19800994 DOI: 10.1016/j.intimp.2009.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 09/21/2009] [Accepted: 09/23/2009] [Indexed: 11/30/2022]
Abstract
The TNF superfamily member LIGHT has potent anti-tumor activity through direct cytotoxicity and activation of the immune response, and is a promising candidate for cancer therapy. Natively, LIGHT exists as both a membrane-anchored form and a proteolytically processed, secreted form. However, the strength of the anti-tumor activity of each form of LIGHT has not been well defined. Here, to identify the optimal form of LIGHT for cancer gene therapy, we constructed fiber-mutant adenovirus vectors (AdRGD) encoding native full-length LIGHT (LIGHT/FL), stably membrane-anchored LIGHT (LIGHT/mem), and fully secreted LIGHT (LIGHT/sec). We then compared the anti-tumor effects of the different forms of LIGHT in mice by intratumoral injection of each AdRGD. We demonstrated that intratumoral injection of AdRGD-LIGHT/sec provided greater tumor suppression than AdRGD-LIGHT/FL, although this effect did not reach statistical significance. By comparison, AdRGD-LIGHT/mem had negligible anti-tumor activity. We also demonstrated that more CD4+ and CD8+ T cells accumulated inside tumors treated in vivo with AdRGD-LIGHT/sec than in tumors treated with AdRGD-LIGHT/FL or AdRGD-LIGHT/mem. These results suggest that the secreted form of LIGHT might be the optimal form for cancer gene therapy.
Collapse
Affiliation(s)
- Tomohiro Morishige
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Pasero C, Barbarat B, Just-Landi S, Bernard A, Aurran-Schleinitz T, Rey J, Eldering E, Truneh A, Costello RT, Olive D. A role for HVEM, but not lymphotoxin-beta receptor, in LIGHT-induced tumor cell death and chemokine production. Eur J Immunol 2009; 39:2502-14. [PMID: 19701890 DOI: 10.1002/eji.200939069] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The TNF member LIGHT also known as TL4 or TNFSF14) can play a major role in cancer control via its two receptors; it induces tumor cell death through lymphotoxin-beta receptor (LT-betaR) and ligation to the herpes virus entry mediator (HVEM) amplifies the immune response. By studying the effect of LIGHT in the transcriptional profile of a lymphoid malignancy, we found that HVEM, but not LT-betaR, stimulation induces a significant increase in the expression of chemokine genes such as IL-8, and an unexpected upregulation of apoptotic genes. This had functional consequences, since LIGHT, or HVEM mAb, thus far known to costimulate T- and B-cell activation, induced chronic lymphocytic leukemia cell death. Many of the mediators involved were identified here, with an apoptotic pathway as demonstrated by caspases activation, decrease in mitochondrial membrane potential, upregulation of the pro-apoptotic protein Bax, but also a role of TRAIL. Moreover, HVEM induced endogenous TNF-alpha production and TNF-alpha enhanced HVEM-mediated cell death. HVEM function was mainly dependent on LIGHT, since other ligands like HSV-glycoprotein D and B and T lymphocyte attenuator were essentially ineffective. In conclusion, we describe a novel, as yet unknown killing effect of LIGHT through HVEM on a lymphoid malignancy, and combined with induction of chemokine release this may represent an additional tool to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Christine Pasero
- INSERM UMR891, Centre de Recherche en Cancérologie de Marseille, Université de Méditerranée, Institut de Cancérologie et d'Immunologie de Marseille, Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
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: 30] [Impact Index Per Article: 2.0] [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
|
69
|
O'Rourke RW, Metcalf MD, White AE, Madala A, Winters BR, Maizlin II, Jobe BA, Roberts CT, Slifka MK, Marks DL. Depot-specific differences in inflammatory mediators and a role for NK cells and IFN-gamma in inflammation in human adipose tissue. Int J Obes (Lond) 2009; 33:978-90. [PMID: 19564875 DOI: 10.1038/ijo.2009.133] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Adipose tissue is a primary in vivo site of inflammation in obesity. Excess visceral adipose tissue (VAT), when compared to subcutaneous adipose tissue (SAT), imparts an increased risk of obesity-related comorbidities and mortality, and exhibits differences in inflammation. Defining depot-specific differences in inflammatory function may reveal underlying mechanisms of adipose-tissue-based inflammation. METHODS Stromovascular cell fractions (SVFs) from VAT and SAT from obese humans undergoing bariatric surgery were studied in an in vitro culture system with transcriptional profiling, flow cytometric phenotyping, enzyme-linked immunosorbent assay and intracellular cytokine staining. RESULTS Transcriptional profiling of SVF revealed differences in inflammatory transcript levels in VAT relative to SAT, including elevated interferon-gamma (IFN-gamma) transcript levels. VAT demonstrated a broad leukocytosis relative to SAT that included macrophages, T cells and natural killer (NK) cells. IFN-gamma induced a proinflammatory cytokine expression pattern in SVF and adipose tissue macrophages (ATM). NK cells, which constitutively expressed IFN-gamma, were present at higher frequency in VAT relative to SAT. Both T and NK cells from SVF expressed IFN-gamma on activation, which was associated with tumor necrosis factor-alpha expression in macrophages. CONCLUSION These data suggest involvement of NK cells and IFN-gamma in regulating ATM phenotype and function in human obesity and a potential mechanism for the adverse physiologic effects of VAT.
Collapse
Affiliation(s)
- R W O'Rourke
- Department of Surgery, Oregon Health and Science University, Portland, OR 97239-3098, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Steinberg MW, Shui JW, Ware CF, Kronenberg M. Regulating the mucosal immune system: the contrasting roles of LIGHT, HVEM, and their various partners. Semin Immunopathol 2009; 31:207-21. [PMID: 19495760 DOI: 10.1007/s00281-009-0157-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 05/13/2009] [Indexed: 12/23/2022]
Abstract
LIGHT and herpes virus entry mediator (HVEM) comprise a ligand-receptor pair in the tumor necrosis factor superfamily. These molecules play an important role in regulating immunity, particularly in the intestinal mucosa. LIGHT also binds the lymphotoxin beta receptor, and HVEM can act as a ligand for immunoglobulin family molecules, including B- and T-lymphocyte attenuator, which suppresses immune responses. Complexity in this pivotal system arises from several factors, including the non-monogamous pairing of ligands and receptors, and reverse signaling or the ability of some ligands to serve as receptors. As a result, recognition events in this fascinating network of interacting molecules can have pro- or anti-inflammatory consequences. Despite complexity, experiments we and others are carrying out are establishing rules for understanding when and in what cell types these molecules contribute to intestinal inflammation.
Collapse
Affiliation(s)
- Marcos W Steinberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
71
|
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: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
72
|
Pentcheva-Hoang T, Corse E, Allison JP. Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections. Immunol Rev 2009; 229:67-87. [PMID: 19426215 DOI: 10.1111/j.1600-065x.2009.00763.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The generation of productive adaptive immune responses depends on the antigen-specific activation of T and B cells. The outcome of T-cell receptor engagement is influenced by signals from both positive and negative regulatory molecules that can either activate or inhibit T-cell function. CD28 and cytotoxic T-lymphocyte antigen-4 are the prototypical members of an immunoglobulin domain-containing protein family that play important roles in the control of T-cell responses against infection, cancer, and in autoimmune disease. Although the precise molecular details of their functions are still under active investigation, tumors and chronic pathogens seem to have exploited these pathways to achieve immune evasion. Furthermore, malfunction of the inhibitory arm of the immune response appears responsible for the development of multiple autoimmune pathologies. As a result, the negative regulators of T-cell activation have become attractive targets for therapeutic intervention in cancer, chronic infection, and autoimmune disease. The application of findings from basic research has provided insight into the manipulation of these pathways in the clinic and offers promising strategies for the treatment of disease.
Collapse
Affiliation(s)
- Tsvetelina Pentcheva-Hoang
- Department of Immunology, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | |
Collapse
|
73
|
Vinay DS, Kwon BS. TNF superfamily: costimulation and clinical applications. Cell Biol Int 2009; 33:453-65. [PMID: 19230849 PMCID: PMC2712666 DOI: 10.1016/j.cellbi.2009.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/04/2009] [Indexed: 12/21/2022]
Abstract
The molecules concerned with costimulation belong either to the immunoglobulin (Ig) or tumor necrosis factor (TNF) superfamily. The tumor necrosis superfamily comprises molecules capable of providing both costimulation and cell death. In this review we briefly summarize certain TNF superfamily receptor-ligand pairs that are endowed with costimulatory properties and their importance in health and disease.
Collapse
Affiliation(s)
- Dass S Vinay
- Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Byoung S Kwon
- Department of Ophthalmology, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA, USA
- Cell and Immunobiology and R&D Center for Cancer Therapeutics, National Cancer Center, Ilsan, Gyeonggi-Do, Korea
| |
Collapse
|
74
|
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 DOI: 10.1002/eji.200838250] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [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
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Ishida S, Yamane S, Nakano S, Yanagimoto T, Hanamoto Y, Maeda-Tanimura M, Toyosaki-Maeda T, Ishizaki J, Matsuo Y, Fukui N, Itoh T, Ochi T, Suzuki R. The interaction of monocytes with rheumatoid synovial cells is a key step in LIGHT-mediated inflammatory bone destruction. Immunology 2008; 128:e315-24. [PMID: 19019090 DOI: 10.1111/j.1365-2567.2008.02965.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Formation of osteoclasts and consequent joint destruction are hallmarks of rheumatoid arthritis (RA). Here we show that LIGHT, a member of the tumour necrosis factor (TNF) superfamily, induced the differentiation into tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs) of CD14(+) monocytes cocultured with nurse-like cells isolated from RA synovium, but not of freshly isolated CD14(+) monocytes. Receptor activator of nuclear factor-kappaB ligand (RANKL) enhanced this LIGHT-induced generation of TRAP-positive MNCs. The MNCs showed the phenotypical and functional characteristics of osteoclasts; they showed the expression of osteoclast markers such as cathepsin K, actin-ring formation, and the ability to resorb bone. Moreover, the MNCs expressed both matrix metalloproteinase 9 (MMP-9) and MMP-12, but the latter was not expressed in osteoclasts induced from CD14(+) monocytes by RANKL. Immunohistochemical analysis showed that the MMP-12-producing MNCs were present in the erosive areas of joints in RA, but not in the affected joints of osteoarthritic patients. These findings suggested that LIGHT might be involved in the progression of inflammatory bone destruction in RA, and that osteoclast progenitors might become competent for LIGHT-mediated osteoclastogenesis via interactions with synoviocyte-like nurse-like cells.
Collapse
Affiliation(s)
- Satoru Ishida
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Abstract
Gene expression studies in scleroderma have shown large and consistent changes in the gene expression of end-target tissues. These changes reflect the lymphocyte infiltration and pathway deregulation potentially linked to disease pathogenesis. Gene expression in scleroderma also reflects the clinical heterogeneity in the disease and can be used to categorize patients. Contained within these gene expression signatures are groups of genes that could serve as biomarkers for clinical end points and disease activity. The use of mechanism-derived gene expression signatures in scleroderma will provide a better understanding of the deregulated pathways contributing to disease pathogenesis.
Collapse
|
77
|
Engineering enhancement of the immune response to HBV DNA vaccine in mice by the use of LIGHT gene adjuvant. J Virol Methods 2008; 153:142-8. [PMID: 18722475 DOI: 10.1016/j.jviromet.2008.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Revised: 07/20/2008] [Accepted: 07/23/2008] [Indexed: 02/06/2023]
Abstract
DNA vaccines could induce protective immune responses in several animal models. Many strategies have been employed to improve the effect of nucleic acid vaccines. LIGHT is a member of the TNF superfamily and functions as a co-stimulatory molecule for T cell proliferation. In the study, the immunogenicity in the induction of humoral and cellular immune responses by HBV DNA vaccine and the adjuvant effect of LIGHT were studied in a murine model. The eukaryotic expression plasmid pcDNA-L was constructed by inserting mouse LIGHT gene into the vector pcDNA3.1(+). In vitro expression of LIGHT was detected by RT-PCR and indirect immunofluorescence assay in transfected HeLa cells. MLR assay showed that LIGHT-transfected DCs induced markedly higher allogeneic lymphocyte proliferation than pcDNA-transfected DCs and untreated DCs at all dilutions. After BALB/c mice were immunized by three intramuscular injections of the HBV DNA vaccine plasmids alone or in combination with LIGHT expression plasmids, the different levels of anti-HBV immune responses were measured comparable to the control groups immunized with parent plasmid pcDNA or PBS. The HBsAg-specific splenocytes proliferation and specific cytotoxic activities of splenic CTLs in the coinoculation group were both significantly higher than those in the HBV DNA single inoculation group, and an enhancement of antibody response was also observed in the coinoculation group compared with the single inoculation group. Taken together, coimmunization of HBV DNA vaccine plasmids and LIGHT expression plasmids can elicit stronger humoral and cellular immune responses in mice than HBV DNA vaccine plasmids alone, and LIGHT may be an effective immunological adjuvant in HBV DNA vaccination.
Collapse
|
78
|
Kim WJ, Kang YJ, Suk K, Park JE, Kwon BS, Lee WH. Comparative analysis of the expression patterns of various TNFSF/TNFRSF in atherosclerotic plaques. Immunol Invest 2008; 37:359-73. [PMID: 18569075 DOI: 10.1080/08820130802123139] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Members of the TNFSF/TNFRSF are involved in the immunoregulation of various immune reactions and diseases. Recently, LIGHT/TR2, GITRL/GITR, and TL1A/DR3 have been reported as playing roles in the inflammatory reactions in atherosclerosis, but a comparative analysis of these molecules has not been conducted. In order to compare their expression patterns, immunohistochemical analyses were performed using six human carotid endoarterectomy samples. The expression of these molecules was detected in the various cell types that constitute atherosclerotic plaques. The expression of all analyzed molecules was detected, albeit at various levels, mainly in foamy macrophages in all tested samples. The strong expression of these molecules in endothelial and smooth muscle cells was also detected in 2 and 1 plaque samples, respectively, while others express only some of the tested molecules. Flow cytometry analyses of human monocyte/macrophage cell lines, U937 and THP-1, detected the expression of the tested molecules while a relatively undifferentiated monocytic cell line, TF-1A, failed to express them. These data indicate that activated and differentiated macrophages are the main cell type expressing tested molecules in atherosclerotic plaques while endothelial and smooth muscle cells can express them in limited cases. Pro-inflammatory activities of the tested molecules may contribute to the atherogenesis by stimulating the cells expressing them in atherosclerotic plaques and the successful treatment of atherosclerosis may require cooperative regulation of these activities.
Collapse
Affiliation(s)
- Won-Jung Kim
- Department of Genetic Engineering, School of Life Sciences and Biotechnology School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | |
Collapse
|
79
|
Heo SK, Yun HJ, Park WH, Park SD. NADPH oxidase activation is required for migration by LIGHT in human monocytes. Biochem Biophys Res Commun 2008; 371:834-40. [DOI: 10.1016/j.bbrc.2008.04.184] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Accepted: 04/29/2008] [Indexed: 10/22/2022]
|
80
|
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: 41] [Impact Index Per Article: 2.4] [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
|
81
|
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
|
82
|
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]
|
83
|
Wang XF, Chen YJ, Wang Q, Ge Y, Dai Q, Yang KF, Zhou YH, Hu YM, Mao YX, Zhang XG. Distinct expression and inhibitory function of B and T lymphocyte attenuator on human T cells. ACTA ACUST UNITED AC 2007; 69:145-53. [PMID: 17257317 DOI: 10.1111/j.1399-0039.2006.00710.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
B and T lymphocyte attenuator (BTLA) has been recently identified as a new inhibitory receptor of the CD28 superfamily, with similarities to cytotoxic T lymphocyte activation antigen (CTLA)-4 and programmed death (PD)-1. Engagement of BTLA on T lymphocytes can profoundly reduce the T cell receptor (TCR)-mediated activation. In this study, we generated four monoclonal antibodies (mAbs) against human BTLA. Using the produced mAb 8H9, the BTLA molecule was found to distinctly express on many subgroups of immunocytes and show a regulatory expression, which was in accordance with its unique ligand herpes virus entry mediator (HVEM) in the process of T cell activation. In addition, the expression of BTLA was increased in the CD4(+) and CD8(+) T cells of pleural fluid in lung cancer patients. Furthermore, we showed that the BTLA-induced negative signals could be triggered by mAb 7D7. Cross-linking of BTLA with mAb 7D7 suppressed T lymphocyte proliferation, downregulated the expression of T cell activation marker CD25, and inhibited the production of interferon (IFN)-gamma, interleukin (IL)-2, IL-4, and IL-10.
Collapse
Affiliation(s)
- X-F Wang
- Medical Biotechnology Institute, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Kang YM, Kim SY, Kang JH, Han SW, Nam EJ, Kyung HS, Park JY, Kim IS. LIGHT up-regulated on B lymphocytes and monocytes in rheumatoid arthritis mediates cellular adhesion and metalloproteinase production by synoviocytes. ACTA ACUST UNITED AC 2007; 56:1106-17. [PMID: 17393389 DOI: 10.1002/art.22493] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To study the expression of LIGHT (tumor necrosis factor superfamily 14) and herpesvirus entry mediator (HVEM; tumor necrosis factor receptor superfamily 14) in rheumatoid arthritis (RA) and to determine the regulatory role of LIGHT on the effector functions of fibroblast-like synoviocytes (FLS). METHODS The expression of LIGHT and HVEM was assessed by immunohistochemical staining of synovial tissue and by flow cytometric analysis of mononuclear cells. The presence of HVEM and lymphotoxin beta receptor was measured by reverse transcriptase-polymerase chain reaction and by flow cytometry. The regulation of effector molecules, including matrix metalloproteinases (MMPs) and adhesion molecules, was evaluated. The adhesiveness of FLS was determined by adhesion assay. RESULTS HVEM was detected in most cell types within rheumatoid synovial tissue, while only a few cells were positive for LIGHT. In RA patients, LIGHT expression was significantly up-regulated only in CD20+ B cells and monocytes, whereas the mean fluorescence intensity of HVEM was down-regulated in mononuclear cells. The stimulation of FLS with LIGHT resulted in the production of MMPs and the expression of adhesion molecules, which were efficiently inhibited by dexamethasone. LIGHT-mediated up-regulation of MMPs and intercellular adhesion molecule 1 was blocked by inhibitors of NF-kappaB and JNK, whereas up-regulation of vascular cell adhesion molecule 1 was blocked by inhibitors of phosphatidylinositol 3-kinase, as well as NF-kappaB. CONCLUSION These data suggest that binding of LIGHT with its receptors may play a role in the progression of inflammation within rheumatoid synovium, especially by mediating the interactions between infiltrating inflammatory cells and stromal cells. These findings thus emphasize the relevance of LIGHT as a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Young Mo Kang
- Kyungpook National University School of Medicine, and Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Tamachi T, Watanabe N, Oya Y, Kagami SI, Hirose K, Saito Y, Iwamoto I, Nakajima H. B and T lymphocyte attenuator inhibits antigen-induced eosinophil recruitment into the airways. Int Arch Allergy Immunol 2007; 143 Suppl 1:50-5. [PMID: 17541277 DOI: 10.1159/000101405] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Signaling through CD28 family co-receptors regulates activation of CD4(+) T cells positively and negatively. It has been shown that stimulatory co-receptors such as CD28 and ICOS play critical roles in the induction of allergic airway inflammation. However, the role of B and T lymphocyte attenuator (BTLA), an inhibitory co-receptor expressed preferentially in Th1 cells, in the regulation of allergic airway inflammation remains to be determined. METHODS We examined antigen-induced eosinophil recruitment and cytokine production in the airways in antigen-sensitized BTLA-deficient (BTLA-/-) mice. We also examined antigen-induced cytokine production and cell proliferation of splenic T cells in antigen-sensitized BTLA-/- mice. RESULTS Antigen-induced eosinophil recruitment and IL-5 production in the airways was enhanced in antigen-sensitized BTLA-/- mice. On the other hand, antigen-induced Th1 and Th2 cytokine production as well as T cell proliferation of splenocytes was normal in BTLA-/-mice. CONCLUSION BTLA inhibits antigen-induced eosinophil recruitment into the airways by preventing IL-5 production from Th2 cells.
Collapse
Affiliation(s)
- Tomohiro Tamachi
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba City, Chiba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Summers-DeLuca LE, McCarthy DD, Cosovic B, Ward LA, Lo CC, Scheu S, Pfeffer K, Gommerman JL. Expression of lymphotoxin-alphabeta on antigen-specific T cells is required for DC function. ACTA ACUST UNITED AC 2007; 204:1071-81. [PMID: 17452522 PMCID: PMC2118582 DOI: 10.1084/jem.20061968] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
During an immune response, activated antigen (Ag)-specific T cells condition dendritic cells (DCs) to enhance DC function and survival within the inflamed draining lymph node (LN). It has been difficult to ascertain the role of the tumor necrosis factor (TNF) superfamily member lymphotoxin-αβ (LTαβ) in this process because signaling through the LTβ-receptor (LTβR) controls multiple aspects of lymphoid tissue organization. To resolve this, we have used an in vivo system where the expression of TNF family ligands is manipulated only on the Ag-specific T cells that interact with and condition Ag-bearing DCs. We report that LTαβ is a critical participant required for optimal DC function, independent of its described role in maintaining lymphoid tissue organization. In the absence of LTαβ or CD40L on Ag-specific T cells, DC dysfunction could be rescued in vivo via CD40 or LTβR stimulation, respectively, suggesting that these two pathways cooperate for optimal DC conditioning.
Collapse
|
87
|
Pierer M, Brentano F, Rethage J, Wagner U, Hantzschel H, Gay RE, Gay S, Kyburz D. The TNF superfamily member LIGHT contributes to survival and activation of synovial fibroblasts in rheumatoid arthritis. Rheumatology (Oxford) 2007; 46:1063-70. [PMID: 17426140 DOI: 10.1093/rheumatology/kem063] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES The TNF superfamily member LIGHT has a T-cell co-stimulatory role and has previously been associated with inflammation and autoimmunity. To investigate its role in rheumatoid arthritis (RA), a disease where activated T cells contribute in a prominent way, we have analysed the expression of LIGHT and its receptors in RA and analysed its effects on synovial fibroblasts in vitro. METHODS The expression of LIGHT was measured in synovial tissues and fluids and the receptors of LIGHT were detected on synovial fibroblasts derived from patients with RA and osteoarthritis (OA). The effects of recombinant LIGHT on the production of proinflammatory cytokines and proteases and on the apoptosis of synovial fibroblasts was assessed. RESULTS LIGHT mRNA was present in synovial tissues of patients with RA but not with OA. Correspondingly, soluble LIGHT protein could be detected in RA synovial fluid samples at much higher levels than in synovial fluid from patients with OA. Immunohistochemical detection of LIGHT and analysis of synovial fluid cells by flow cytometry revealed CD4 T cells as the major source of LIGHT in the rheumatoid joint. Synovial fibroblasts from RA patients were found to express the LIGHT receptors HVEM and LTbetaR. Recombinant LIGHT induced RA synovial fibroblasts to upregulate MMP-9 mRNA, CD54 and IL-6 in an NF-kappaB-dependent fashion. In vitro, exposure of cultured synovial fibroblasts to LIGHT reduced FAS-mediated apoptosis significantly, without affecting the rate of spontaneous apoptosis. CONCLUSIONS The results provide evidence for a novel T-cell-dependent activation of synovial fibroblasts by LIGHT in joints of patients with RA, contributing to an inflammatory and destructive phenotype.
Collapse
MESH Headings
- Aged
- Apoptosis/drug effects
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- CD4-Positive T-Lymphocytes/chemistry
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Female
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Flow Cytometry
- Humans
- Immunohistochemistry
- Intercellular Adhesion Molecule-1/analysis
- Interleukin-6/analysis
- Leukotriene B4/analysis
- Leukotriene B4/metabolism
- Male
- Matrix Metalloproteinase 9/analysis
- Middle Aged
- NF-kappa B/analysis
- NF-kappa B/metabolism
- Osteoarthritis/immunology
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- RNA, Messenger/analysis
- Receptors, Tumor Necrosis Factor, Member 14/analysis
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Recombinant Proteins/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Statistics, Nonparametric
- Synovial Fluid/chemistry
- Synovial Fluid/immunology
- Synovial Fluid/metabolism
- Synovial Membrane/immunology
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
- Tumor Necrosis Factor Ligand Superfamily Member 14/analysis
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
Collapse
Affiliation(s)
- M Pierer
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, Gloriastrasse 25, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Wei CY, Chou YH, Ho FM, Hsieh SL, Lin WW. Signaling pathways of LIGHT induced macrophage migration and vascular smooth muscle cell proliferation. J Cell Physiol 2007; 209:735-43. [PMID: 16972254 DOI: 10.1002/jcp.20742] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The biological actions of LIGHT, a member of the tumor necrosis factor superfamily, are mediated by the interaction with lymphotoxin-beta receptor (LTbetaR) and/or herpes virus entry mediator (HVEM). Previous study demonstrated high-level expressions of LIGHT and HVEM receptors in atherosclerotic plaques. To investigate the role of LIGHT in the functioning of macrophages and vascular smooth muscle cells (VSMC) in relation to atherogenesis, we determined the effects of LIGHT on macrophage migration and VSMC proliferation. We found LIGHT through HVEM activation can induce both events. LIGHT-induced macrophage migration was associated with activation of signaling kinases, including MAPKs, PI3K/Akt, NF-kappaB, Src members, and FAK. Proliferation of VSMC was also shown relating to the activation of MAPKs, PI3K/Akt, and NF-kappaB, which consequently led to alter the expression of cell cycle regulatory molecules. Down-regulation of p21, p27, and p53, and inversely up-regulation of cyclin D and RB hyper-phosphorylation were demonstrated. In conclusion, LIGHT acts as a novel mediator for macrophage migration and VSMC proliferation, suggesting its involvement in the atherogenesis.
Collapse
MESH Headings
- Animals
- Atherosclerosis
- Cell Cycle Proteins/metabolism
- Cell Movement/physiology
- Cell Proliferation
- Cells, Cultured
- Enzyme Activation
- Enzyme Inhibitors/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Mice
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/physiology
- NF-kappa B/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Signal Transduction/physiology
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Chun-Yu Wei
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
89
|
Haselmayer P, Tenzer S, Kwon BS, Jung G, Schild H, Radsak MP. Herpes virus entry mediator synergizes with Toll-like receptor mediated neutrophil inflammatory responses. Immunology 2007; 119:404-11. [PMID: 17067315 PMCID: PMC1819577 DOI: 10.1111/j.1365-2567.2006.02449.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In microbial infections polymorphnuclear neutrophils (PMN) constitute a major part of the innate host defence, based upon their ability to rapidly accumulate in inflamed tissues and clear the site of infection from microbial pathogens by their potent effector mechanisms. The recently described transmembrane receptor herpes virus entry mediator (HVEM) is a member of the tumour necrosis factor receptor super family and is expressed on many haematopoietic cells, including T cells, B cells, natural killer cells, monocytes and PMN. Interaction of HVEM with the natural ligand LIGHT on T cells has a costimulatory effect, and increases the bactericidal activity of PMN. To further characterize the function of HVEM on PMN, we evaluated the effect of receptor ligation on human PMN effector functions using an agonistic monoclonal antibody. Here we demonstrate that activation of HVEM causes activation of neutrophil effector functions, including respiratory burst, degranulation and release of interleukin-8 in synergy with ligands for Toll-like receptors or GM-CSF. In addition, stimulation via HVEM enhanced neutrophil phagocytic activity of complement opsonized, but not of non-opsonized, particles. In conclusion, these results indicate a new, as yet unknown, participation of HVEM in the innate immune response and points to a new link between innate and adaptive immunity.
Collapse
Affiliation(s)
| | - Stefan Tenzer
- Institute of Immunology, University of MainzMainz, Germany
| | | | - Gundram Jung
- Institute for Cell Biology, Department Immunology, University of TübingenTübingen, Germany
| | | | - Markus P Radsak
- Institute of Immunology, University of MainzMainz, Germany
- University Medical Hospital, IIIrd Department of Medicine, University of MainzMainz, Germany
| |
Collapse
|
90
|
Krieg C, Boyman O, Fu YX, Kaye J. B and T lymphocyte attenuator regulates CD8+ T cell-intrinsic homeostasis and memory cell generation. Nat Immunol 2007; 8:162-71. [PMID: 17206146 DOI: 10.1038/ni1418] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 11/06/2006] [Indexed: 12/18/2022]
Abstract
B and T lymphocyte attenuator (BTLA) is a negative regulator of T cell activation, but its function in vivo is not well characterized. Here we show that mice deficient in full-length BTLA or its ligand, herpesvirus entry mediator, had increased number of memory CD8(+) T cells. The memory CD8(+) T cell phenotype resulted from a T cell-intrinsic perturbation of the CD8(+) T cell pool. Naive BTLA-deficient CD8(+) T cells were more efficient than wild-type cells at generating memory in a competitive antigen-specific system. This effect was independent of the initial expansion of the responding antigen-specific T cell population. In addition, BTLA negatively regulated antigen-independent homeostatic expansion of CD4(+) and CD8(+) T cells. These results emphasize two central functions of BTLA in limiting T cell activity in vivo.
Collapse
Affiliation(s)
- Carsten Krieg
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
91
|
Xu Y, Tamada K, Chen L. LIGHT-related molecular network in the regulation of innate and adaptive immunity. Immunol Res 2007; 37:17-32. [PMID: 17496344 DOI: 10.1007/bf02686093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
The LIGHT-related molecular network is composed of at least seven interacting receptors and ligands. Recent studies reveal that this network has profound immune regulatory functions for both innate and adaptive immunity. Experimental data support the concept that this network may also play roles in the pathogenesis of human diseases including cancer, infection, transplantation tolerance, and autoimmune diseases. In this review, we attempt to dissect each molecular interaction in detail and assemble them in the context of their roles in the pathogenesis and possible therapeutic potential in human diseases.
Collapse
Affiliation(s)
- Yanhui Xu
- Molecular Biology Graduate Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | |
Collapse
|
92
|
Snanoudj R, de Préneuf H, Créput C, Arzouk N, Deroure B, Beaudreuil S, Durrbach A, Charpentier B. Costimulation blockade and its possible future use in clinical transplantation. Transpl Int 2006; 19:693-704. [PMID: 16918529 DOI: 10.1111/j.1432-2277.2006.00332.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The nonimmune effects of currently used immunosuppressive drugs result in a high incidence of late graft loss due to nephrotoxicity and death of patients. As an immune-specific alternative to conventional immunosuppressants, new biotechnology tools can be used to block the costimulation signals of T-cell activation. Many experimental studies--particularly preclinical studies in nonhuman primates--have focused on blocking the 'classical' B7/CD28 and CD40/CD40L pathways, which are critical in primary T-cell activation. Here, we review the limitations, the recent advances and the first large-scale clinical application of the CTLA4-Ig fusion protein to block the B7/CD28 costimulation pathway. We also focus on new B7/CD28 and tumor necrosis factor (TNF)/TNF-R family costimulatory molecules that can deliver positive or negative costimulation signals regulating the alloimmune response. Strategies that use single agents to block costimulation have often proved to be insufficient. Given the diversity of the different costimulation molecules, future strategies for human transplantation may involve the simultaneous blockade of several selected pathways or the simultaneous use of conventional immunosuppressants.
Collapse
Affiliation(s)
- Renaud Snanoudj
- Service de Néphrologie et Transplantation Rénale, Hôpital du Kremlin Bicêtre, Le Kremlin-Bicêtre, INSERM U542, Villejuif, France.
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Murphy KM, Nelson CA, Sedý JR. Balancing co-stimulation and inhibition with BTLA and HVEM. Nat Rev Immunol 2006; 6:671-81. [PMID: 16932752 DOI: 10.1038/nri1917] [Citation(s) in RCA: 240] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The interaction between B- and T-lymphocyte attenuator (BTLA), an inhibitory receptor whose extracellular domain belongs to the immunoglobulin superfamily, and herpesvirus-entry mediator (HVEM), a co-stimulatory tumour-necrosis factor receptor, is unique in that it is the only receptor-ligand interaction that directly bridges these two families of receptors. This interaction has raised many questions about how receptors from two different families could interact and what downstream signalling events might occur as a result of receptor ligation. As we discuss, recent studies show that engagement of HVEM with its endogenous ligand (LIGHT) from the tumour-necrosis factor family induces a powerful immune response, whereas HVEM interactions with BTLA negatively regulate T-cell responses.
Collapse
MESH Headings
- Animals
- Disease
- Humans
- Ligands
- Protein Binding
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/immunology
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/classification
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/chemistry
- Receptors, Virus/classification
- Receptors, Virus/immunology
- Signal Transduction
Collapse
Affiliation(s)
- Kenneth M Murphy
- Department of Pathology and Centre for Immunology, Howard Hughes Medical Institute, Washington University School of Medicine, 660 S. Euclid, St Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
94
|
Banks TA, Rickert S, Ware CF. Restoring immune defenses via lymphotoxin signaling: lessons from cytomegalovirus. Immunol Res 2006; 34:243-54. [PMID: 16891674 DOI: 10.1385/ir:34:3:243] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
Although primary infection with human cytomegalovirus (HCMV), a beta-herpesvirus, is widespread and acquired early in life, it rarely causes disease in immune-competent individuals. However, in immune-compromised patients HCMV infection or reactivation invariably leads to serious disease, the effective treatment of which remains a difficult clinical problem. Current antiviral therapy is limited not only by toxicity but also by the continual emergence of drugresistant viruses. The limitations of these current therapeutics provides a strong impetus to develop novel approaches that will enhance the host's immune responsiveness while at the same time effectively controlling virus replication. Type I interferon (IFN) plays a critical role in initiating innate antiviral defenses and promoting adaptive responses and lymphotoxin (LT)-alphabeta has recently been identified as an essential effector cytokine regulating the induction of type I IFN during CMV infection. In particular, CMV infection of immune-compromised mice has revealed the immunotherapeutic potential of the lymphotoxin-beta receptor (LTbetaR) signaling pathway to restore immune function and provide protection from CMV mortality. In this review, we discuss the potential benefits and risks associated with LTbetaR-directed immunotherapy for CMV disease and other persistent viral infections.
Collapse
Affiliation(s)
- Theresa A Banks
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, CA 92121, USA
| | | | | |
Collapse
|
95
|
Eldredge J, Berkowitz S, Corin AF, Day ES, Hayes D, Meier W, Strauch K, Zafari M, Tadi M, Farrington GK. Stoichiometry of LTβR Binding to LIGHT. Biochemistry 2006; 45:10117-28. [PMID: 16906770 DOI: 10.1021/bi060210+] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
LTbetaR is a member of the TNF receptor family of proteins. It binds to two different cell surface ligands, LIGHT, a homotypic trimer, and LTalpha1beta2, a heterotypic trimer. We have measured the affinities of the dimeric IgG fusion protein, LTbetaRIgG, and monomeric LTbetaR protein binding to both LIGHT and LTalpha1beta2 using surface plasmon resonance and found values of <0.1 and 38 nM for LIGHT and <0.1 and 48 nM for LTalpha1beta2, respectively. We also determined the stoichiometries of binding for both forms of the receptor LTbetaRIgG and LTbetaR binding to LIGHT. The data obtained from several biophysical methods are consistent with receptor polypeptide to trimeric ligand ratios of 2:1. The determined masses of the complexes using SEC-LS corresponded to a single LTbetaRIgG bound to a LIGHT trimer, or two LTbetaR bound per LIGHT. Sedimentation velocity of varied ratios of LTbetaR to a fixed concentration of LIGHT were analyzed by SEDANAL and were successfully fit with a model with two tight binding sites on LIGHT and one poor affinity site. Isothermal calorimetric titration of LIGHT with either LTbetaR or LTbetaRIgG also demonstrated stoichiometries of 1:2 and 1:1, respectively. The binding of LTbetaR to LIGHT was endothermic and, hence, entropy-driven. TNFR p55 (extracellular domain) complexed with the trimeric ligand, TNFbeta, exhibits a 3:1 receptor/ligand stoichiometry. This complex has been used as the prototypical model setting the receptor-ligand complexation paradigm for the entire TNF family. The LTbetaR/LIGHT binding stoichiometry of 2:1 demonstrated here does not fit the paradigm. This has numerous implications for cell biology including signaling requiring only dimerization of LTbetaR rather than trimerization as expected from the structural paradigm.
Collapse
Affiliation(s)
- John Eldredge
- Biogen Idec, Inc., 12 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Fujita T, Ukyo N, Hori T, Uchiyama T. Functional characterization of OX40 expressed on human CD8+ T cells. Immunol Lett 2006; 106:27-33. [PMID: 16750861 DOI: 10.1016/j.imlet.2006.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 03/27/2006] [Accepted: 04/02/2006] [Indexed: 11/16/2022]
Abstract
In the present study, we investigated the expression of OX40 on human CD8(+) T cells with regard to expression induction, costimulatory function and possible involvement in cytotoxicity. Human CD8(+) T cells were purified from peripheral blood mononuclear cell (PBMC) of healthy donors and cocultured with allogeneic monocyte-derived dendritic cells. Flow cytometric analysis showed that expression of OX40 was induced on CD8(+) T cells within 1 day and increased to the maximum levels on day 3. An addition of anti-OX40 ligand (OX40L) mAb suppressed CD25 expression, proliferation and IFN-gamma production of CD8(+) T cells, suggesting that OX40 functions as a costimulatory molecule not only for CD4(+) T cells but also for CD8(+) T cells. In parallel, coculture of pre-activated CD8(+) T cells with OX40L-transfected murine epithelial cells (MMCE-OX40L) resulted in an increase in CD25 expression, proliferation and IFN-gamma producing cells, compared with that with the mock control (MMCE-mock). Finally, non-specific cytotoxic activity of preactivated CD8(+) T cells was examined using OKT3 hybridoma as target cells after coculture with these transfectants. Coculture with MMCE-OX40L induced slightly higher cytotoxicity of CD8(+) T cells than that with MMCE-mock. These results indicate that OX40 is induced transiently on CD8(+) T cells upon activation and its signals contribute to both clonal expansion and functional reinforcement.
Collapse
Affiliation(s)
- Tomoko Fujita
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Sakyoku, Japan
| | | | | | | |
Collapse
|
97
|
Sedgmen BJ, Dawicki W, Gommerman JL, Pfeffer K, Watts TH. LIGHT is dispensable for CD4+ and CD8+ T cell and antibody responses to influenza A virus in mice. Int Immunol 2006; 18:797-806. [PMID: 16569673 DOI: 10.1093/intimm/dxl016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The tumor necrosis factor family ligands, LIGHT (lymphotoxin like, exhibits inducible expression and competes with HSV glycoprotein D for HVEM, a receptor expressed by T lymphocytes), 4-1BBL and CD70, are found in the same gene cluster on mouse chromosome 17. Although the roles of 4-1BB-4-1BBL and CD27-CD70 interactions in anti-viral T cell responses have been well established, the role of LIGHT in T cell activation/expansion in vivo is less clear. Under conditions that were previously employed to demonstrate a role for 4-1BBL in CD8+ T cell memory, wild-type and LIGHT-/- mice were infected with influenza A virus and primary and memory/recall responses were measured at various time points thereafter. Neither primary expansion nor memory/recall CD8+ T cell responses were affected by the absence of LIGHT, as measured up to 2 months post-infection. CD4+ T cell responses were also unaffected by LIGHT deficiency. Furthermore, we found that LIGHT played no role in the induction of influenza-specific IgG1 and IgG2a serum antibodies. Taken together, these data suggest that LIGHT is dispensable for the acquired immune response to influenza virus in mice with no effect on the induction, maintenance or reactivation of CD8+ T cell memory.
Collapse
Affiliation(s)
- Bradley J Sedgmen
- Department of Immunology, Room 5263, Medical Sciences Building, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | | | | | | |
Collapse
|
98
|
Anand S, Wang P, Yoshimura K, Choi IH, Hilliard A, Chen YH, Wang CR, Schulick R, Flies AS, Flies DB, Zhu G, Xu Y, Pardoll DM, Chen L, Tamada K. Essential role of TNF family molecule LIGHT as a cytokine in the pathogenesis of hepatitis. J Clin Invest 2006; 116:1045-51. [PMID: 16557300 PMCID: PMC1409742 DOI: 10.1172/jci27083] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 01/31/2006] [Indexed: 12/21/2022] Open
Abstract
LIGHT is an important costimulatory molecule for T cell immunity. Recent studies have further implicated its role in innate immunity and inflammatory diseases, but its cellular and molecular mechanisms remain elusive. We report here that LIGHT is upregulated and functions as a proinflammatory cytokine in 2 independent experimental hepatitis models, induced by concanavalin A and Listeria monocytogenes. Molecular mutagenesis studies suggest that soluble LIGHT protein produced by cleavage from the cell membrane plays an important role in this effect through the interaction with the lymphotoxin-beta receptor (LTbetaR) but not herpes virus entry mediator. NK1.1+ T cells contribute to the production, but not the cleavage or effector functions, of soluble LIGHT. Importantly, treatment with a mAb that specifically interferes with the LIGHT-LTbetaR interaction protects mice from lethal hepatitis. Our studies thus identify a what we believe to be a novel function of soluble LIGHT in vivo and offer a potential target for therapeutic interventions in hepatic inflammatory diseases.
Collapse
MESH Headings
- Animals
- Antigens, Ly
- Antigens, Surface/metabolism
- Concanavalin A/metabolism
- Concanavalin A/pharmacology
- Cytokines/metabolism
- Hepatitis/etiology
- Hepatitis/metabolism
- Inflammation/metabolism
- Lectins, C-Type/metabolism
- Listeria monocytogenes/metabolism
- Listeria monocytogenes/pathogenicity
- Lymphotoxin beta Receptor
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- NK Cell Lectin-Like Receptor Subfamily B
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Member 14
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
- Solubility
- Tumor Necrosis Factor Ligand Superfamily Member 14
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Sudarshan Anand
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pu Wang
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kiyoshi Yoshimura
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - In-Hak Choi
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anja Hilliard
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Youhai H. Chen
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chyung-Ru Wang
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard Schulick
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew S. Flies
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dallas B. Flies
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gefeng Zhu
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yanhui Xu
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Drew M. Pardoll
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lieping Chen
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koji Tamada
- Immunology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Department of Microbiology, Inje University College of Medicine, Pusan, Republic of Korea.
Committee on Immunology and Department of Pathology, University of Chicago, Chicago, Illinois, USA.
Department of Surgery and
Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Biochemistry Graduate Program, Mayo Clinic College of Medicine, Rochester, Minnesota, USA.
Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
99
|
So T, Lee SW, Croft M. Tumor Necrosis Factor/Tumor Necrosis Factor Receptor Family Members That Positively Regulate Immunity. Int J Hematol 2006; 83:1-11. [PMID: 16443545 DOI: 10.1532/ijh97.05120] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interactions between members of the tumor necrosis factor (TNF) family and their specific receptors (TNFRs) are influential in controlling cell division, life, and death. Recent evidence suggests that these interactions control the functionality and longevity of many types of cells involved in immune responses. In particular, it has become evident that certain interactions support the clonal expansion and survival of T-cells, B-cells, and dendritic cells and thus are essential for establishing a robust immune response. This review describes select TNF/TNFR family members that principally support activation and survival and prevent excessive cell death of T-cells (OX40L/OX40, 4-1BBL/4-1BB, CD30L/CD30, LIGHT/HVEM, CD70/CD27, and GITRL/GITR), B-cells (BAFF/BAFFR), and dendritic cells (RANKL/RANK). Expression of these ligands and receptors on the cell surface is highly regulated, and communication via them occurs during contact between T-cells and dendritic cells and between T-cells and B-cells. The functional dynamic between these TNF/TNFR members is slowly being unraveled, including whether these molecules act together or sequentially or control different type of immune responses. This review summarizes aspects of these TNF/TNFR interactions that are potentially important to immune responses.
Collapse
Affiliation(s)
- Takanori So
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, San Diego, California, USA
| | | | | |
Collapse
|
100
|
Gavrieli M, Sedy J, Nelson CA, Murphy KM. BTLA and HVEM Cross Talk Regulates Inhibition and Costimulation. Adv Immunol 2006; 92:157-85. [PMID: 17145304 DOI: 10.1016/s0065-2776(06)92004-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently a new inhibitory immunoglobulin domain-containing lymphocyte receptor was identified on the basis of its T helper 1 (T(H)1)-selective expression in murine T cell lines, which was named B and T lymphocyte attenuator (BTLA). Several groups have confirmed the initial characterization of BTLA as an inhibitory receptor, which was initially inferred from the mild increases in several parameters of BTLA-deficient mice. The initial expectation that BTLA would interact with a B7 family ligand, such as the B7x protein, was surprisingly overturned with the functional cloning of the actual BTLA ligand as herpesvirus entry mediator (HVEM). This was unexpected largely due to the fact that this interaction represents the convergence of two very different, although each quite extensive, families of receptors and ligands. The interaction of BTLA, which belongs to the CD28 family of the immunoglobulin superfamily, and HVEM, a costimulatory tumor-necrosis factor (TNF) receptor (TNFR), is quite unique in that it is the only receptor-ligand interaction that directly bridges these two families of receptors. This interaction has raised many questions about how receptors from two different families could interact and which are the signaling events downstream of receptor ligation. As we discuss here and recently demonstrated, HVEM interaction with BTLA serves to negatively regulate T cell responses, in contrast to the strong activation observed when HVEM engages its endogenous ligand from the TNF family. Finally, as studies of BTLA are just now beginning to extend beyond the initial characterizations, it is becoming clear that there are many complex issues remaining to be resolved, particularly potential polymorphisms that may engender disease susceptibility in the human.
Collapse
Affiliation(s)
- Maya Gavrieli
- Department of Pathology and Center for Immunology, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|