51
|
Behrendt AK, Meyer-Bahlburg A, Hansen G. CD137 deficiency does not affect development of airway inflammation or respiratory tolerance induction in murine models. Clin Exp Immunol 2012; 168:308-17. [PMID: 22519594 DOI: 10.1111/j.1365-2249.2012.04572.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The co-stimulatory molecule CD137 (4-1BB) plays a crucial role in the development and persistence of asthma, characterized by eosinophilic airway inflammation, mucus hypersecretion, airway hyperreactivity, increased T helper type 2 (Th2) cytokine production and serum immunoglobulin (Ig)E levels. We have shown previously that application of an agonistic CD137 monoclonal antibody (mAb) prevented and even reversed an already established asthma phenotype. In the current study we investigated whether deficiency of the CD137/CD137L pathway affects the development of allergic airway inflammation or the opposite immune reaction of respiratory tolerance. CD137⁻/⁻ and wild-type (WT) mice were sensitized and challenged with the model allergen ovalbumin (OVA) and analysed for the presence of allergic disease parameters (allergy protocol). Some animals were tolerized by mucosal application of OVA prior to transferring the animals to the allergy protocol to analyse the effect of CD137 loss on tolerance induction (tolerance protocol). Eosinophilic airway inflammation, mucus hypersecretion, Th2 cytokine production and elevated allergen-specific serum IgE levels were increased equally in CD137⁻/⁻ and WT mice. Induction of tolerance resulted in comparable protection from the development of an allergic phenotype in both mouse strains. In addition, no significant differences could be identified in CD4⁺, CD8⁺ and forkhead box protein 3 (FoxP3⁺) regulatory T cells, supporting the conclusion that CD137⁻/⁻ mice show equal Th2-mediated immune responses compared to WT mice. Taken together, CD137⁻/⁻ mice and WT mice develop the same phenotype in a murine model of Th2-mediated allergic airway inflammation and respiratory tolerance.
Collapse
Affiliation(s)
- A-K Behrendt
- Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
52
|
Abstract
4-1BB (CD137), a member of the TNF receptor superfamily, is an activation-induced T-cell costimulatory molecule. Signaling via 4-1BB upregulates survival genes, enhances cell division, induces cytokine production, and prevents activation-induced cell death in T cells. The importance of the 4-1BB pathway has been underscored in a number of diseases, including cancer. Growing evidence indicates that anti-4-1BB monoclonal antibodies possess strong antitumor properties, which in turn are the result of their powerful CD8+ T-cell activating, IFN-γ producing, and cytolytic marker-inducing capabilities. In addition, combination therapy of anti-4-1BB with other anticancer agents, such as radiation, has robust tumor-regressing abilities against nonimmunogenic or poorly immunogenic tumors. Furthermore, the adoptive transfer of ex vivo anti-4-1BB-activated CD8+ T cells from previously tumor-treated animals efficiently inhibits progression of tumors in recipient mice that have been inoculated with fresh tumors. In addition, targeting of tumors with variants of 4-1BBL directed against 4-1BB also have potent antitumor effects. Currently, a humanized anti-4-1BB is in clinical trials in patients with solid tumors, including melanoma, renal carcinoma, and ovarian cancer, and so far seems to have a favorable toxicity profile. In this review, we discuss the basis of the therapeutic potential of targeting the 4-1BB-4-1BBL pathway in cancer treatment.
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | |
Collapse
|
53
|
Abstract
The non-obese diabetic (NOD) mouse spontaneously develops type 1 diabetes (T1D) and has thus served as a model for understanding the genetic and immunological basis, and treatment, of T1D. Since its initial description in 1980, however, the field has matured and recognized that prevention of diabetes in NOD mice (i.e., preventing the disease from occurring by an intervention prior to frank diabetes) is relatively easy to achieve and does not correlate well with curing the disease (after the onset of frank hyperglycemia). Hundreds of papers have described the prevention of diabetes in NOD mice but only a handful have described its actual reversal. The paradoxical conclusion is that preventing the disease in NOD mice does not necessarily tell us what caused the disease nor how to reverse it. The NOD mouse model is therefore best used now, with respect to human disease, as a way to understand the genetic and immunologic causes of and as a model for trying to reverse disease once hyperglycemia occurs. We describe how genetic approaches to identifying causative gene variants can be adapted to identify novel therapeutic agents for reversing new-onset T1D.
Collapse
|
54
|
Reverse signaling through the costimulatory ligand CD137L in epithelial cells is essential for natural killer cell-mediated acute tissue inflammation. Proc Natl Acad Sci U S A 2011; 109:E13-22. [PMID: 22160719 DOI: 10.1073/pnas.1112256109] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Renal ischemia-reperfusion injury (IRI) after kidney transplantation is a major cause of delayed graft function. Even though IRI is recognized as a highly coordinated and specific process, the pathways and mechanisms through which the innate response is activated are poorly understood. In this study, we used a mouse model of acute kidney IRI to examine whether the interactions of costimulatory receptor CD137 and its ligand (CD137L) are involved in the early phase of acute kidney inflammation caused by IRI. We report here that the specific expressions of CD137 on natural killer cells and of CD137L on tubular epithelial cells (TECs) are required for acute kidney IRI. Reverse signaling through CD137L in TECs results in their production of the chemokine (C-X-C motif) receptor 2 ligands CXCL1 and CXCL2 and the subsequent induction of neutrophil recruitment, resulting in a cascade of proinflammatory events during kidney IRI. Our findings identify an innate pathogenic pathway for renal IRI involving the natural killer cell-TEC-neutrophil axis, whereby CD137-CD137L interactions provide the causal contribution of epithelial cell dysregulation to renal IRI. The CD137L reverse signaling pathway in epithelial cells therefore may represent a good target for blocking the initial stage of inflammatory diseases, including renal IRI.
Collapse
|
55
|
Singh UP, Singh NP, Singh B, Price RL, Nagarkatti M, Nagarkatti PS. Cannabinoid receptor-2 (CB2) agonist ameliorates colitis in IL-10(-/-) mice by attenuating the activation of T cells and promoting their apoptosis. Toxicol Appl Pharmacol 2011; 258:256-67. [PMID: 22119709 DOI: 10.1016/j.taap.2011.11.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 10/30/2011] [Accepted: 11/09/2011] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammation caused by hyperactivated effector immune cells that produce pro-inflammatory cytokines. Recent studies have shown that the cannabinoid system may play a critical role in mediating protection against intestinal inflammation. However, the effect of cannabinoid receptor induction after chronic colitis progression has not been investigated. Here, we investigate the effect of cannabinoid receptor-2 (CB2) agonist, JWH-133, after chronic colitis in IL-10(-/-) mice. JWH-133 effectively attenuated the overall clinical score, and reversed colitis-associated pathogenesis and decrease in body weight in IL-10(-/-) mice. After JWH-133 treatment, the percentage of CD4(+) T cells, neutrophils, mast cells, natural killer (NK1.1) cells, and activated T cells declined in the intestinal lamina propria (LP) and mesenteric lymph nodes (MLN) of mice with chronic colitis. JWH-133 was also effective in ameliorating dextran sodium sulfate (DSS)-induced colitis. In this model, JWH-133 reduced the number and percentage of macrophages and IFN-γ expressing cells that were induced during colitis progression. Treatment with aminoalkylindole 6-iodo-pravadoline (AM630), a CB2 receptor antagonist, reversed the colitis protection provided by JWH-133 treatment. Also, activated T cells were found to undergo apoptosis following JWH-133 treatment both in-vivo and in-vitro. These findings suggest that JWH-133 mediates its effect through CB2 receptors, and ameliorates chronic colitis by inducing apoptosis in activated T cells, reducing the numbers of activated T cells, and suppressing induction of mast cells, NK cells, and neutrophils at sites of inflammation in the LP. These results support the idea that the CB2 receptor agonists may serve as a therapeutic modality against IBD.
Collapse
Affiliation(s)
- Udai P Singh
- Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | | | | | | | | | | |
Collapse
|
56
|
Kim J, Kim W, Kim HJ, Park S, Kim HA, Jung D, Choi HJ, Park SJ, Mittler RS, Cho HR, Kwon B. Host CD25+CD4+Foxp3+ regulatory T cells primed by anti-CD137 mAbs inhibit graft-versus-host disease. Biol Blood Marrow Transplant 2011; 18:44-54. [PMID: 21958951 DOI: 10.1016/j.bbmt.2011.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/20/2011] [Indexed: 01/01/2023]
Abstract
CD25(+)CD4(+)Foxp3(+) regulatory T cells (Tregs) play a pivotal role in the maintenance of self-tolerance and regulation of immune responses. Previous studies have demonstrated that CD137 signals can promote proliferation and survival of Tregs in vitro. Here, we show that in vivo CD137-induced expansion of Tregs in naive mice was dependent upon IL-2 secreted by memory T cells. Tregs primed by anti-CD137 mAbs had a higher immunosuppressive capacity. Preconditioning with anti-CD137 mAbs significantly inhibited graft-versus-host disease (GVHD) in the C57BL/6 → (C57BL/6 × DBA/2) F1 acute GVHD model. In this disease model, a high proportion of host Tregs remained long-term in the recipient spleen, whereas donor hematopoietic cells replaced other host bone marrow-derived cells. Transient depletion of Tregs before transfer of donor cells completely abrogated the inhibitory effect of anti-CD137 mAbs on GVHD. In addition, adoptive transfer of anti-CD137-primed Tregs ameliorated GVHD. Our results demonstrate that it is possible to enhance the survival and/or the immunosuppressive activity of host Tregs in nonmyeloablative GVHD, and that 1 way of accomplishing this is through the prophylactic use of anti-CD137 mAbs in nonmyeloablative GVHD.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Kim YH, Choi BK, Shin SM, Kim CH, Oh HS, Park SH, Lee DG, Lee MJ, Kim KH, Vinay DS, Kwon BS. 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modulating the balance between Th17 and regulatory T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:1120-8. [PMID: 21715692 DOI: 10.4049/jimmunol.1002681] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Agonistic anti-4-1BB Ab is known to ameliorate experimental autoimmune encephalomyelitis. 4-1BB triggering typically leads to the expansion of CD8(+) T cells, which produce abundant IFN-γ, and this in turn results in IDO-dependent suppression of autoimmune responses. However, because neutralization of IFN-γ or depletion of CD8(+) T cell only partially abrogates the effect of 4-1BB triggering, we sought to identify an additional mechanism of 4-1BB-triggered suppression of autoimmune responses using IFN-γ- or IFN-γR-deficient mice. 4-1BB triggering inhibited the generation of Th17 cells that is responsible for experimental autoimmune encephalomyelitis induction and progression, and increased Foxp3(+)CD4(+) regulatory T (Treg) cells, particularly among CD4(+) T cells. This was not due to a direct effect of 4-1BB signaling on CD4(+) T cell differentiation: 4-1BB signaling not only reduced Th17 cells and increased Treg cells in wild-type mice, which could be due to IFN-γ production by the CD8(+) T cells, but also did so in IFN-γ-deficient mice, in that case by downregulating IL-6 production. These results show that although secondary suppressive mechanisms evoked by 4-1BB triggering are usually masked by the strong effects of IFN-γ, 4-1BB signaling seems to modulate autoimmune responses by a number of mechanisms, and modulation of the Th17 versus Treg cell balance is one of those mechanisms.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- CD4 Lymphocyte Count
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cells, Cultured
- Disease Progression
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Interferon-gamma/deficiency
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/pathology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Young H Kim
- Immune and Cell Therapy Branch, Division of Cancer Biology, National Cancer Center, Goyang-si, Gyeongi-do 410-769, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Choi BK, Kim YH, Choi JH, Kim CH, Kim KS, Sung YC, Lee YM, Moffett JR, Kwon BS. Unified immune modulation by 4-1BB triggering leads to diverse effects on disease progression in vivo. Cytokine 2011; 55:420-8. [PMID: 21700476 DOI: 10.1016/j.cyto.2011.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 04/28/2011] [Accepted: 05/18/2011] [Indexed: 12/11/2022]
Abstract
4-1BB (CD137) is a powerful T-cell costimulatory molecule in the treatment of virus infections and tumors, but recent studies have also uncovered regulatory functions of 4-1BB signaling. Since 4-1BB triggering suppresses autoimmunity by accumulating indoleamine 2,3-dioxygenase (IDO) in dendritic cells (DCs) in an interferon (IFN)-γ-dependent manner, we asked whether similar molecular and cellular changes were induced by 4-1BB triggering in virus-infected mice. 4-1BB triggering increased IFN-γ and IDO, and suppressed CD4(+) T cells, in C57BL/6 mice infected with the type 1 KOS strain of Herpes simplex virus (HSV-1), as it does in an autoimmune disease model. Detailed analysis of the CD4(+) T suppression showed that freshly activated CD62L(high) T cells underwent apoptosis in the early phase of suppression, and CD62L(low) effector/memory T cells in the later phase. Although 4-1BB triggering resulted in similar cellular changes - increased CD8(+) T and decreased CD4(+) T cells, it had different effects on mortality in mice infected with HSV-1 RE, influenza, and Japanese encephalitis virus (JEV); it increased mortality in influenza-infected mice but decreased it in JEV-infected mice. Since the dominant type of immune cell generated to protect the host was different for each virus - CD4(+) T cells and neutrophils in HSV-1 RE infection, both CD4(+) T and CD8(+) T cells in influenza infection, and a crucial role for B cells in JEV infection, 4-1BB triggering resulted in different therapeutic outcomes. We conclude that the therapeutic outcome of 4-1BB triggering is determined by whether the protective immunity generated against the virus was beneficially altered by the 4-1BB triggering.
Collapse
Affiliation(s)
- Beom K Choi
- Immune and Cell Therapy Branch, Division of Cancer Biology, R&D Center for Cancer Therapeutics, National Cancer Center, 809 Madu, Ilsan, Goyang, Kyeonggi-do 411-769, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Zhang J, Chen G, Zhou D, Wang Z. Expression of CD137 in the cerebral artery after experimental subarachnoid hemorrhage in rats: a pilot study. Brain Res 2011; 1386:200-8. [PMID: 21352817 DOI: 10.1016/j.brainres.2011.02.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 02/15/2011] [Accepted: 02/15/2011] [Indexed: 10/18/2022]
Abstract
Inflammation and immunity play a crucial role in the pathogenesis of cerebral vasospasm after subarachnoid hemorrhage (SAH). CD137 is recognized as an independent costimulatory molecule of T cells and activator of monocytes. A growing body of evidence indicates that CD137 is vital for inflammation and immunity. Therefore, this study aimed to investigate the expression of CD137 in the basilar artery in a rat SAH model and to clarify the potential role of CD137 in cerebral vasospasm. A total of 107 rats were randomly divided into four groups: control group; day 3, day 5, and day 7 groups. Day 3, day 5, and day 7 groups were all SAH groups. The animals in SAH groups were subjected to injection of autologous blood into cisterna magna twice on day 0 and day 2 and were sacrificed on days 3, 5, and 7, respectively. Cross-sectional area of basilar artery was measured and the CD137 expression was assessed by quantitative real-time PCR, Western blot and immunohistochemistry. The cross-sectional area of basilar artery was found to be 57,944±5581μm(2) in control group, 26,100±2639μm(2) in day 3, 19,723±2412μm(2) in day 5, and 28,800±2980μm(2) in day 7 group, respectively. The basilar artery exhibited vasospasm after SAH and became more severe on day 5. The elevated mRNA and protein of CD137 were detected after SAH and peaked on day 5. CD137 is increasingly expressed in a parallel time course to the development of cerebral vasospasm in a rat experimental model of SAH. These findings indicate the possible role of CD137 in the pathogenesis of cerebral vasospasm after SAH.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, PR China
| | | | | | | |
Collapse
|
60
|
Vinay DS, Kwon BS. The tumour necrosis factor/TNF receptor superfamily: therapeutic targets in autoimmune diseases. Clin Exp Immunol 2011; 164:145-57. [PMID: 21401577 DOI: 10.1111/j.1365-2249.2011.04375.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Autoimmune diseases are characterized by the body's ability to mount immune attacks on self. This results from recognition of self-proteins and leads to organ damage due to increased production of pathogenic inflammatory molecules and autoantibodies. Over the years, several new potential therapeutic targets have been identified in autoimmune diseases, notable among which are members of the tumour necrosis factor (TNF) superfamily. Here, we review the evidence that certain key members of this superfamily can augment/suppress autoimmune diseases.
Collapse
Affiliation(s)
- D S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, USA
| | | |
Collapse
|
61
|
Kwon B. Intervention with costimulatory pathways as a therapeutic approach for graft-versus-host disease. Exp Mol Med 2011; 42:675-83. [PMID: 20820112 DOI: 10.3858/emm.2010.42.10.071] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Graft-versus-host disease (GVHD) is mediated by mature donor T cells contained in the hematopoietic stem cell graft. During the development of GVHD, signaling through a variety of costimulatory receptors plays an important role in allogeneic T cell responses. Even though delivery of costimulatory signals is a prerequisite for full activation of donor T cells in the phase of their interactions with host APCs, their involvement with GVHD might occur over multiple stages. Like many other aspects of GVHD, promise of therapeutic interventions with costimulatory pathways has been gleaned from preclinical models. In this review, I summarize some of the advances in roles of costimulatory molecules in GVHD pathophysiology and discuss preclinical approaches that warrant further exploration in the clinic, focusing on novel strategies to delete pathogenic T cells.
Collapse
Affiliation(s)
- Byungsuk Kwon
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea.
| |
Collapse
|
62
|
Abstract
Decades of high-titered antibody are sustained due to the persistence of memory B cells and long-lived plasma cells (PCs). The differentiation of each of these subsets is antigen- and T-cell driven and is dependent on signals acquired and integrated during the germinal center response. Inherent in the primary immune response must be the delivery of signals to B cells to create these populations, which have virtual immortality. Differences in biology and chemotactic behavior disperse memory B cells and long-lived PCs to a spectrum of anatomic sites. Each subset must rely on survival factors that can support their longevity. This review focuses on the generation of each of these subsets, their survival, and renewal, which must occur to sustain serological memory. In this context, we discuss the role of antigen, bystander inflammation, and cellular niches. The contribution of BAFF (B-cell activating factor belonging to the tumor necrosis factor family) and APRIL (a proliferation-inducing ligand) to the persistence of memory B cells and PCs are also detailed. Insights that have been provided over the past few years in the regulation of long-lived B-cell responses will have profound impact on vaccine development, the treatment of pre-sensitized patients for organ transplantation, and therapeutic interventions in both antibody- and T-cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Raul Elgueta
- Department of Nephrology and Transplantation, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | | | | |
Collapse
|
63
|
Kim CS, Tu TH, Kawada T, Kim BS, Yu R. The immune signaling molecule 4-1BB stimulation reduces adiposity, insulin resistance, and hepatosteatosis in obese mice. Endocrinology 2010; 151:4725-35. [PMID: 20719857 DOI: 10.1210/en.2010-0346] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Immune cells (e.g. macrophages and T cells) in adipose tissue play a crucial role in the development of obesity-induced inflammation and metabolic disorders. Here we report findings suggesting that the immune signaling molecule 4-1BB/CD137 is a novel target for treatment of obesity and metabolic disorders. 4-1BB stimulation with agonistic antibody reduced body weight and adiposity and markedly improved glucose intolerance and hepatosteatosis in diet-induced obese mice and genetically obese/diabetic mice. Increases in lymphoid T cell expansion/activation and adipose/hepatic CD8+ T cell recruitment were evident in the anti-4-1BB antibody-treated obese mice. Glycolysis, β-oxidation, and oxygen consumption rates also increased in the treated mice. These findings suggest that 4-1BB-stimulation accompanied by CD8+ T cell expansion/activation enhances glucose/lipid metabolism, leading to increased energy expenditure. Manipulation of 4-1BB may provide a unique immunological strategy against obesity and metabolic disorders.
Collapse
Affiliation(s)
- Chu-Sook Kim
- Department of Food Science and Nutrition, University of Ulsan, Mugeo-dong, Nam-ku, Ulsan 680-749, South Korea
| | | | | | | | | |
Collapse
|
64
|
Lee JM, Seo JH, Kim YJ, Kim YS, Ko HJ, Kang CY. Agonistic Anti-CD137 Monoclonal Antibody Treatment Induces CD11bGr-1 Myeloid-derived Suppressor Cells. Immune Netw 2010; 10:104-8. [PMID: 20631881 PMCID: PMC2902672 DOI: 10.4110/in.2010.10.3.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 06/03/2010] [Accepted: 06/07/2010] [Indexed: 12/30/2022] Open
Abstract
CD137 (4-1BB/tnfrsf9) has been shown to co-stimulate T cells. However, agonistic anti-CD137 monoclonal antibody (mAb) treatment can suppress CD4+ T cells, ameliorating autoimmune diseases, whereas it induces activation of CD8+ T cells, resulting in diverse therapeutic activity in cancer, viral infection. To investigate the CD137-mediated T cell suppression mechanism, we examined whether anti-CD137 mAb treatment could affect CD11b+Gr-1+ myeloid-derived suppressor cells (MDSCs). Intriguingly, anti-CD137 mAb injection significantly increased CD11b+Gr-1+ cells, peaking at days 5 to 10 and continuing for at least 25 days. Furthermore, this cell population could suppress both CD8+ T cells and CD4+ T cells. Thus, this study demonstrated that, for the first time, anti-CD137 mAb treatment could induce CD11b+Gr-1+ MDSCs under normal conditions, suggesting a possible relationship between myeloid cell induction and CD137-mediated immune suppression.
Collapse
Affiliation(s)
- Jung-Mi Lee
- Laboratory of Immunology, Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
65
|
Lee SW, Croft M. 4-1BB as a therapeutic target for human disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 647:120-9. [PMID: 19760070 DOI: 10.1007/978-0-387-89520-8_8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
4-1BB (CD137) is being thought of as an attractive target for immunotherapy of many human immune diseases based on encouraging results with 4-1BB agonistic antibody treatment in mouse models of cancer, autoimmune disease, asthma and additionally as a means to improve vaccination. In this review, we will summarize the results of basic research on 4-1BB and 4-1BB immunotherapy of disease and provide some potential mechanistic insights into the many stimulatory and regulatory functions of 4-1BB.
Collapse
Affiliation(s)
- Seung-Woo Lee
- Molecular Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, California, 92037, USA
| | | |
Collapse
|
66
|
Zhang B, Zhang Y, Niu L, Vella AT, Mittler RS. Dendritic cells and Stat3 are essential for CD137-induced CD8 T cell activation-induced cell death. THE JOURNAL OF IMMUNOLOGY 2010; 184:4770-8. [PMID: 20351189 DOI: 10.4049/jimmunol.0902713] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Agonistic anti-CD137 mAbs either positively or negatively regulate T cell function. When administered at the beginning of lymphocytic choriomeningitis virus Armstrong infection anti-CD137 induced immunosuppression and T cell deletion, and in the case of influenza infection led to increased mortality. In contrast, 72 h delay in anti-CD137 treatment led to an enhanced virus-specific CD8 T cell response and rapid viral clearance. Virus-specific CD8 T cells in anti-CD137-injected mice rapidly upregulate Fas expression, and although necessary, was insufficient to induce CD8 T cell deletion. Strikingly, CD137 signaling in T cells was found to be insufficient to induce suppression or deletion. Rather, immunosuppression and T cell deletion was only observed if CD137 signals were provided to T cells and dendritic cells (DCs). In vitro CD137 crosslinking in DCs led to phosphorylation of Stat3, and importantly, anti-CD137 treatment of lymphocytic choriomeningitis virus Armstrong infected Stat3 conditional knock-out mice induced neither immune suppression or T cell deletion. Taken together, these data suggest that CD137 signaling in DCs can regulate CD8 T cell survival through a Stat3 and Fas-mediated pathway.
Collapse
Affiliation(s)
- Benyue Zhang
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | | | | | | | |
Collapse
|
67
|
Kim YM, Kim HK, Kim HJ, Lee HW, Ju SA, Choi BK, Kwon BS, Kim BS, Kim JB, Lim YT, Yoon S. Expression of 4-1BB and 4-1BBL in thymocytes during thymus regeneration. Exp Mol Med 2010; 41:896-911. [PMID: 19745604 DOI: 10.3858/emm.2009.41.12.095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
4-1BB, a member of the tumor necrosis factor receptor (TNFR) superfamily, is a major costimulatory receptor that is rapidly expressed on the surface of CD4(+) and CD8(+) T cells after antigen- or mitogen-induced activation. The interaction of 4-1BB with 4-1BBL regulates immunity and promotes the survival and expansion of activated T cells. In this study, the expression of 4-1BB and 4-1BBL was examined during regeneration of the murine thymus following acute cyclophosphamide- induced involution. Four-color flow cytometry showed that 4-1BB and 4-1BBL were present in the normal thymus and were preferentially expressed in the regenerating thymus, mainly in CD4(+)CD8(+) double-positive (DP) thymocytes. Furthermore, the CD4(lo)CD8(lo), CD4(+)CD8(lo) and CD4(lo)CD8(+) thymocyte subsets, representing stages of thymocyte differentiation intermediate between DP and single-positive (SP) thymocytes, also expressed 4-1BB and 4-1BBL during thymus regeneration but to a lesser degree. Interestingly, the 4-1BB and 4-1BBL positive cells among the CD4(+)CD8(+) DP thymocytes present during thymus regeneration were TCR(hi) and CD69(+) unlike the corresponding controls. Moreover, the 4-1BB and 4-1BBL positive cells among the intermediate subsets present during thymus regeneration also exhibited TCR(hi/int+) and CD69(+/int) phenotypes, indicating that 4-1BB and 4-1BBL are predominantly expressed by the positively selected population of the CD4(+)CD8(+) DP and the intermediate thymocytes during thymus regeneration. RT-PCR and Western blot analyses confirmed the presence and elevated levels of 4-1BB and 4-1BBL mRNA and protein in thymocytes during thymus regeneration. We also found that the interaction of 4-1BB with 4-1BBL promoted thymocyte adhesion to thymic epithelial cells. Our results suggest that 4-1BB and 4-1BBL participate in T lymphopoiesis associated with positive selection during recovery from acute thymic involution.
Collapse
Affiliation(s)
- Young-Mi Kim
- Department of Pediatrics, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Jeon HJ, Choi JH, Jung IH, Park JG, Lee MR, Lee MN, Kim B, Yoo JY, Jeong SJ, Kim DY, Park JE, Park HY, Kwack K, Choi BK, Kwon BS, Oh GT. CD137 (4–1BB) Deficiency Reduces Atherosclerosis in Hyperlipidemic Mice. Circulation 2010; 121:1124-33. [DOI: 10.1161/circulationaha.109.882704] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
The tumor necrosis factor receptor superfamily, which includes CD40, LIGHT, and OX40, plays important roles in atherosclerosis. CD137 (4-1BB), a member of the tumor necrosis factor receptor superfamily, has been reported to be expressed in human atherosclerotic lesions. However, limited information is available on the precise role of CD137 in atherosclerosis and the effects of blocking CD137/CD137 ligand signaling on lesion formation.
Methods and Results—
We generated CD137-deficient apolipoprotein E–knockout mice (
ApoE
−/−
CD137
−/−
) and LDL-receptor–knockout mice (
Ldlr
−/−
CD137
−/−
) to investigate the role of CD137 in atherogenesis. The deficiency of CD137 induced a reduction in atherosclerotic plaque lesions in both atherosclerosis mouse models, which was attributed to the downregulation of cytokines such as interferon-γ, monocyte chemoattractant protein-1, and tumor necrosis factor-α. CD137 signaling promoted the production of inflammatory molecules, including monocyte chemoattractant protein-1, interleukin-6, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1, in endothelial cells. Stimulation of CD137 ligand signaling activated monocytes/macrophages and augmented the production of proinflammatory cytokines in atherosclerotic vessels.
Conclusions—
CD137/CD137 ligand signaling plays multiple roles in the progression of atherosclerosis, and thus, blockade of this pathway is a promising therapeutic target for the disease.
Collapse
Affiliation(s)
- Hyung Jun Jeon
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Jae-Hoon Choi
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - In-Hyuk Jung
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Jong-Gil Park
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Mi-Ran Lee
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Mi-Ni Lee
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Bora Kim
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Ji-Young Yoo
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Se-Jin Jeong
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Dae-Yong Kim
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Jeong Euy Park
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Hyun-Young Park
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - KyuBum Kwack
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Beom Kyu Choi
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Byoung S. Kwon
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| | - Goo Taeg Oh
- From the Division of Life and Pharmaceutical Science (H.J.J., J.-H.C., I.-H.J., J.-G.P., M.-R.L., M.-N.L., B.K., J.-Y.Y., S.-J.J., G.T.O.), Ewha Womans University, Seoul, Korea; Laboratory of Cellular Physiology and Immunology (J.-H.C.), The Rockefeller University, New York, NY; Departments of Veterinary Pathology (I.-H.J., D.-Y.K.) and Biochemistry (J.-G.P.), College of Veterinary Medicine, Seoul National University, Seoul, Korea; Samsung Medical Center (J.E.P.), Sungkyungkwan University School of
| |
Collapse
|
69
|
Ponte JF, Ponath P, Gulati R, Slavonic M, Paglia M, O'Shea A, Tone M, Waldmann H, Vaickus L, Rosenzweig M. Enhancement of humoral and cellular immunity with an anti-glucocorticoid-induced tumour necrosis factor receptor monoclonal antibody. Immunology 2010; 130:231-42. [PMID: 20201988 DOI: 10.1111/j.1365-2567.2009.03228.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adjuvants, including antibodies to tumour necrosis factor receptor superfamily members, augment immune responses. One member of this family, glucocorticoid-induced tumour necrosis factor receptor (GITR), is expressed at low levels on naive/resting T cells, B cells and macrophages, but at higher levels on T regulatory cells. The aim of this study was to determine the ability of a rat anti-mouse GITR monoclonal antibody, 2F8, to stimulate murine humoral and cellular immunity in a prime boost model with particular attention to posology and antigen-specific effects. 2F8 enhanced the humoral immune response to ovalbumin and haemagglutinin (HA) compared with controls and this enhancement was equal to or greater than that obtained in mice dosed with standard adjuvants. 2F8 F(ab')(2) fragments were as effective as intact antibody in boosting humoral immunity, indicating that FcR-mediated cross-linking of 2F8 is not required for efficacy. Moreover, the enhanced response was durable and antigen specific. Administration of 2F8 shifted the immune response towards a T helper type 1 response with significant enhancement of immunoglobulin G2a- and G2b-specific anti-HA antibodies, as well as enhanced cellular immunity as measured by ELISPOT. 2F8-treated mice also generated significantly more neutralizing antibodies to HA than control mice. Our findings show that anti-GITR is a robust, versatile adjuvant that, unlike commonly used adjuvants that primarily enhance humoral immunity, enhances both humoral and cellular immunity. These results support the continued development of anti-GITR for such indications as haematological and solid tumours, chronic viral infections, and as a vaccine adjuvant.
Collapse
Affiliation(s)
- Jose F Ponte
- Tolerx, Inc., 300 Technology Square, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Wang S, Chen L. Immunobiology of cancer therapies targeting CD137 and B7-H1/PD-1 cosignal pathways. Curr Top Microbiol Immunol 2010; 344:245-67. [PMID: 20582531 DOI: 10.1007/82_2010_81] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is finally entering a new era with manipulation of cosignaling pathways as a therapeutic approach, for which the principle was proved nearly two decades ago. In addition to CTLA-4, CD137 and B7-H1/PD-1 pathways are two new targets in the stage. CD137 pathway is costimulatory and its agonistic antibody delivers potent signal to drive T cell growth and activation. On the other hand, blockade of B7-H1/PD-1 pathway with antagonistic antibody has shown to protect ongoing T cell responses from impairment by immune evasion mechanism in cancer microenvironment. With these tools in hand, a mechanism-based design of combined immunotherapy with high efficacy is becoming a reality.
Collapse
Affiliation(s)
- Shengdian Wang
- Center for Infection and Immunity, Institute for Biophysics, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
71
|
Yang HR, Chou HS, Gu X, Wang L, Brown KE, Fung JJ, Lu L, Qian S. Mechanistic insights into immunomodulation by hepatic stellate cells in mice: a critical role of interferon-gamma signaling. Hepatology 2009; 50:1981-91. [PMID: 19821484 PMCID: PMC2896244 DOI: 10.1002/hep.23202] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
UNLABELLED The liver is considered to be an immune-privileged organ that favors the induction of tolerance. The underlying mechanisms are not completely understood. Interestingly, liver transplants are spontaneously accepted in several animal models, but hepatocyte transplants are acutely rejected, suggesting that liver nonparenchymal cells may effectively protect the parenchymal cells from immune attack. We have shown the profound T cell inhibitory activity of hepatic stellate cells (HSCs). Thus, cotransplantation with HSCs effectively protects islet allografts from rejection in mice. In this study, using T cell receptor transgenic and gene knockout approaches, we provided definitive evidence that HSCs protected cotransplanted islet allografts by exerting comprehensive inhibitory effects on T cells, including apoptotic death in graft-infiltrating antigen-specific effector T cells and marked expansion of CD4(+) Forkhead box protein (Foxp)3(+) T regulatory (Treg) cells. All these effects required an intact interferon-gamma (IFN-gamma) signaling in HSCs, demonstrated by using HSCs isolated from IFN-gamma receptor 1 knockout mice. B7-H1 expression on HSCs, a product molecule of IFN-gamma signaling, was responsible for induction of T cells apoptosis, but had no effect on expansion of Treg cells, suggesting that undetermined effector molecules produced by IFN-gamma signaling is involved in this process. CONCLUSION Upon inflammatory stimulation, specific organ stromal cells (such as HSCs in the liver) demonstrate potent immune regulatory activity. Understanding of the mechanisms involved may lead to development of novel strategies for clinical applications in transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Horng-Ren Yang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
- Department of General Surgery, Cleveland Clinic, Cleveland, OH. USA
| | - Hong-Shuie Chou
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
| | - Xiaodong Gu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
| | - Lianfu Wang
- Department of General Surgery, Cleveland Clinic, Cleveland, OH. USA
| | - Kathleen E. Brown
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
| | - John J. Fung
- Department of General Surgery, Cleveland Clinic, Cleveland, OH. USA
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
- Department of General Surgery, Cleveland Clinic, Cleveland, OH. USA
| | - Shiguang Qian
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH. USA
- Department of General Surgery, Cleveland Clinic, Cleveland, OH. USA
| |
Collapse
|
72
|
Adjuvantive effects of anti-4-1BB agonist Ab and 4-1BBL DNA for a HIV-1 Gag DNA vaccine: different effects on cellular and humoral immunity. Vaccine 2009; 28:1300-9. [PMID: 19944789 DOI: 10.1016/j.vaccine.2009.11.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 11/05/2009] [Accepted: 11/06/2009] [Indexed: 12/21/2022]
Abstract
Plasmid DNA immunizations induce low levels but a broad spectrum of cellular and humoral immune responses. Here, we investigate the potential of co-stimulation through 4-1BB as an adjuvant for a HIV-1 DNA vaccine in mice. We designed plasmid DNAs expressing either the membrane bound or soluble form of 4-1BBL, and compared with the agonistic anti-4-1BB Ab for their ability to adjuvant the Gag DNA vaccine. Both, anti-4-1BB agonistic Ab as well as 4-1BBL DNA enhanced the Gag-specific cellular immune responses. However, in complete contrast to the agonistic Ab that suppressed humoral immunity to Gag, 4-1BBL DNA adjuvanted vaccines enhanced Gag-specific IgG responses. Importantly, the expression of Gag and 4-1BBL from the same plasmid was critical for the adjuvant activity. Collectively, our data suggest that for a HIV-1 vaccine where both antigen-specific cellular and humoral immunity are desirable, 4-1BBL expressed by a DNA vaccine is a superior adjuvant than anti-4-1BB agonistic Ab.
Collapse
|
73
|
Wang C, Lin GHY, McPherson AJ, Watts TH. Immune regulation by 4-1BB and 4-1BBL: complexities and challenges. Immunol Rev 2009; 229:192-215. [PMID: 19426223 DOI: 10.1111/j.1600-065x.2009.00765.x] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SUMMARY The tumor necrosis factor receptor family member 4-1BB plays a key role in the survival of activated and memory CD8(+) T cells. Depending on the disease model, 4-1BB can participate at different stages and influence different aspects of the immune response, likely due to the differential expression of receptor and ligand relative to other costimulatory molecules. Studies comparing mild versus severe influenza infection of mice suggest that the immune system uses inducible receptors such as 4-1BB to prolong the immune response when pathogens take longer to clear. The expression of 4-1BB on diverse cell types, evidence for bidirectional as well as receptor-independent signaling by 4-1BBL, the unexpected hyperproliferation of 4-1BB-deficient T cells, and complex effects of agonistic anti-4-1BB therapy have revealed additional roles for the 4-1BB/4-1BBL receptor/ligand pair in the immune system. In this review, we discuss these diverse roles of 4-1BB and its ligand in the immune response, exploring possible mechanisms for the observed complexities and implications for therapeutic applications of 4-1BB/4-1BBL.
Collapse
Affiliation(s)
- Chao Wang
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|
74
|
Induction of lethal graft-versus-host disease by anti-CD137 monoclonal antibody in mice prone to chronic graft-versus-host disease. Biol Blood Marrow Transplant 2009; 15:306-14. [PMID: 19203721 DOI: 10.1016/j.bbmt.2008.11.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 11/30/2008] [Indexed: 11/23/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is an increasingly frequent complication of allogeneic stem cell transplantation. We previously showed that anti-CD137 monoclonal antibody (mAb) can cure advanced cGVHD by inducing activation-induced cell death of donor T cells. In this study, we examined whether administration of anti-CD137 mAb can prevent the development of cGVHD after bone marrow transplantation (BMT) in mice conditioned with total body irradiation (TBI). We used the B10.D2-->Balb/c (H-2(d)) minor histocompatibility antigen-mismatched model, which reflects clinical and pathological symptoms of human cGVHD. A single injection of anti-CD137 mAb was administered immediately after BMT. In contrast to the results obtained from the curing model of cGVHD, anti-CD137 given simultaneously with BMT resulted in lethal GVHD. Histopathologic evaluation revealed inflammation and damage of target organs from acute GVHD (aGVHD) in anti-CD137-treated mice. Anti-CD137-induced lethal aGVHD required host cells, as well as irradiation and mature donor T cells. Apparently, anti-CD137 mAb rapidly induced activation of donor T cells and sustained their activation status under the inflammatory condition triggered by irradiation. When given on day 12 after irradiation and BMT, anti-CD137 mAb could still exacerbate GVHD, but when given on day 30, it could not. Our data demonstrate that anti-CD137 mAb can amplify inflammation induced by host preconditioning, subsequently resulting in lethal aGVHD; thus, alleviating irradiation-induced toxicity is critical to allow the use of anti-CD137 mAb as GVHD prophylaxis.
Collapse
|
75
|
Abstract
The main stream of CD137 studies has been directed to the function of CD137 in CD8+ T-cell immunity, including its anti-tumor activity, and paradoxically the immunosuppressive activity of CD137, which proves to be of a great therapeutic potential for animal models of a variety of autoimmune and inflammatory diseases. Recent studies, however, add complexes to the biology of CD137. Accumulating is evidence supporting that there exists a bidirectional signal transduction pathway for the CD137 receptor and its ligand (CD137L). CD137/CD137L interactions are involved in the network of hematopoietic and nonhematopoietic cells in addition to the well characterized antigen-presenting cell-T cell interactions. Signaling through CD137L plays a critical role in the differentiation of myeloid cells and their cellular activities, suggesting that CD137L signals trigger and sustain inflammation. The overall consequence might be that the amplified inflammation by CD137L enhances the T-cell activity together with CD137 signals by upregulating costimulatory molecules, MHC molecules, cell adhesion molecules, cytokines, and chemokines. Solving this outstanding issue is urgent and will have an important clinical implication.
Collapse
Affiliation(s)
- Byungsuk Kwon
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
| |
Collapse
|
76
|
Lee SW, Salek-Ardakani S, Mittler RS, Croft M. Hypercostimulation through 4-1BB distorts homeostasis of immune cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:6753-62. [PMID: 19454670 DOI: 10.4049/jimmunol.0803241] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The deleterious side effects associated with a recent clinical trial with anti-CD28 superagonist Abs have questioned the use of reagents to costimulatory molecules in human therapy. We now show that sustained signaling from an agonist Ab to 4-1BB, a member of the TNFR superfamily, results in detrimental effects on immune cell homeostasis. Repeated anti-4-1BB treatment during the reconstitution of hematopoietic cells in irradiated mice engrafted with bone marrow, or in mice infected with vaccinia virus, induced abnormal apoptosis of premature and immature B cells in the bone marrow, and led to peripheral B cell depletion. Inhibition of B cell development was indirect and due to costimulation of CD8 T cells and dependent on IFN-gamma. Moreover, anti-4-1BB also suppressed the development of NK and NKT cells, but in this case independently of T cells and IFN-gamma. The altered NK cell homeostasis resulted from activation-induced cell death triggered by anti-4-1BB. These results show that hypercostimulation elicits strong T cell immunity, but it can simultaneously distort immune homeostasis, suggesting that careful attention to activity, dose, and periodicity of treatment will be needed in any immunotherapeutic strategy with agonist Abs to costimulatory molecules.
Collapse
Affiliation(s)
- Seung-Woo Lee
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
77
|
Abstract
SUMMARY Despite the expression of antigens by tumor cells, spontaneous immune-mediated rejection of cancer seems to be a rare event. T-cell receptor engagement by peptide/major histocompatibility complexes constitutes the main signal for the activation of naive T cells but is not sufficient to initiate a productive generation and maintenance of effector cells. Full activation of T cells requires additional signals driven by costimulatory molecules present on activated antigen-presenting cells but rarely on tumors. Following the discovery of B7-1 (CD80), several other costimulatory molecules have been shown to contribute to T-cell activation and have relevance for improving anti-tumor immunity. Moreover, increasing the understanding of coinhibitory receptors has highlighted key additional pathways that can dominantly inhibit anti-tumor T-cell function. Improving positive costimulation, and interfering with negative regulation, continues to represent an attractive immunotherapeutic approach for the treatment of cancer. This review focuses upon those pathways with the highest potential for clinical application in human cancer patients.
Collapse
Affiliation(s)
- Gregory Driessens
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Thomas F. Gajewski
- Department of Pathology, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
78
|
Attenuation of experimental autoimmune myocarditis by blocking T cell activation through 4-1BB pathway. J Mol Cell Cardiol 2009; 46:719-27. [PMID: 19233196 DOI: 10.1016/j.yjmcc.2009.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 02/03/2009] [Accepted: 02/05/2009] [Indexed: 11/21/2022]
Abstract
4-1BB, a member of the tumor necrosis factor receptor (TNFR) family, binds the 4-1BB ligand (4-1BBL), works as a costimulatory molecule, and regulates T cell-mediated immune responses. Although inflammation is an essential pathological feature of myocarditis, the role of 4-1BB in experimental autoimmune myocarditis (EAM) remains unclear. Lewis rats were immunized on day 0 with purified porcine cardiac myosin to establish EAM. 4-1BB-immunoglobulin (4-1BBIg) was administered intraperitoneally (n=6) a total of 9 times (3 times per week). Rats were killed on day 21 to study effects of 4-1BB/4-1BBL pathway blockade. For controls, isotype-matched human IgG was administered in other EAM rats (n=6). Histologic and echocardiographic examination showed development of EAM attenuated by 4-1BBIg. Suppression of mRNA expression for IL-1alpha, IL-1beta, IL-4, IL-6, and TNF-alpha was noted in the heart tissue treated with 4-1BBIg. Treatment with 4-1BBIg reduced production of Th1-type cytokines, and inhibited T cell proliferation in vitro. In the 4-1BB signaling pathway in splenocytes, 4-1BBIg suppressed JNK, p38, and IkappaB activity but not that of ERK1/2. Blockade of T cell activation through the 4-1BB/4-1BBL pathway regulates development of EAM; therefore, 4-1BB may be an effective target for treating myocarditis.
Collapse
|
79
|
Kim YH, Choi BK, Kang WJ, Kim KH, Kang SW, Mellor AL, Munn DH, Kwon BS. IFN-gamma-indoleamine-2,3 dioxygenase acts as a major suppressive factor in 4-1BB-mediated immune suppression in vivo. J Leukoc Biol 2009; 85:817-25. [PMID: 19218483 DOI: 10.1189/jlb.0408246] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
It has been reported that 4-1BB triggering in vivo selectively suppressed the recall response of staphylococcal enterotoxin A (SEA)-specific CD4(+) T cells, in which CD8(+) T-derived TGF-beta was involved. Here, we have examined an alternative mechanism for the 4-1BB-mediated CD4(+) T suppression, as the neutralization of TGF-beta is only effective in rescuing the SEA-specific recall response at high cellular concentrations. We show that this selective suppression of CD4(+) T cells by 4-1BB triggering in vivo is mediated mainly by induction of indoleamine 2,3-dioxygenase (IDO) in an IFN-gamma-dependent manner. SEA-specific CD4(+) T responses were suppressed partly by TGF-beta-expressing CD8(+) T cells, particularly CD11c(+)CD8(+) T cells, but strongly inhibited by dendritic cells (DCs) expressing IDO. IFN-gamma that increased IDO in DCs was produced primarily from CD11c(+)CD8(+) T cells, which were expanded selectively by 4-1BB stimulation. CD4(+), CD8(+), and plasmacytoid DCs exerted a similar suppressive activity toward the SEA-specific CD4(+) T cells. Neutralization of IFN-gamma or IDO activity in vivo largely reversed the 4-1BB-mediated CD4(+) T suppression. Collectively, these data indicate that 4-1BB-dependent suppression of SEA-specific CD4(+) T responses was mediated mainly by IFN-gamma-dependent IDO induction and partially by TGF-beta.
Collapse
Affiliation(s)
- Young H Kim
- Division of Cell and Immunobiology and R&D Center for Cancer Therapeutics, National Cancer Center, Kyonggi-do, Korea
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Liu GZ, Gomes AC, Fang LB, Gao XG, Hjelmstrom P. Decreased 4-1BB expression on CD4+CD25 high regulatory T cells in peripheral blood of patients with multiple sclerosis. Clin Exp Immunol 2008; 154:22-9. [PMID: 18727631 DOI: 10.1111/j.1365-2249.2008.03730.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
As a tumour necrosis factor receptor superfamily member, 4-1BB (CD137) is preferentially expressed in CD4+CD25+ regulatory T cells (Tregs) and has been suggested to play an important role in regulating the generation or function of Tregs. Recent studies of human Tregs have shown that blood CD4+CD25(high) T cells were much closer to Tregs in terms of their functionality. Furthermore, CD4+CD25(high) Tregs have been found to have a decreased effector function in patients with multiple sclerosis (MS). In this study, we examined the expression of 4-1BB and soluble 4-1BB (s4-1BB) protein levels in the peripheral blood of MS patients. Compared with healthy controls, MS patients had decreased 4-1BB expression in their CD4+C25(high) Tregs and increased plasma s4-1BB protein levels. Moreover, the plasma s4-1BB levels of MS patients were shown to be inversely correlated with the 4-1BB surface expression of CD4+CD25(high) Tregs. The down-regulated 4-1BB expression on CD4+CD25(high) Tregs of MS patients may be involved in the impaired immunoactivity of these Tregs. The elevated s4-1BB levels may, at least in part, function as a self-regulatory attempt to inhibit antigen-driven proliferation of Tregs or their immunosuppressive activity.
Collapse
Affiliation(s)
- G-Z Liu
- Department of Neurology, Peking University People's Hospital, Beijing, China.
| | | | | | | | | |
Collapse
|
81
|
Zhou L, Wang H, Zhong X, Jin Y, Mi QS, Sharma A, McIndoe RA, Garge N, Podolsky R, She JX. The IL-10 and IFN-gamma pathways are essential to the potent immunosuppressive activity of cultured CD8+ NKT-like cells. Genome Biol 2008; 9:R119. [PMID: 18664279 PMCID: PMC2530876 DOI: 10.1186/gb-2008-9-7-r119] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 07/29/2008] [Indexed: 01/30/2023] Open
Abstract
Global gene expression profiling of in vitro cultured CD8+ T cells that express natural killer cell markers revealed differential expression of about 3,000 genes between these cells and naïve CD8+ T cells. Background CD8+ NKT-like cells are naturally occurring but rare T cells that express both T cell and natural killer cell markers. These cells may play key roles in establishing tolerance to self-antigens; however, their mechanism of action and molecular profiles are poorly characterized due to their low frequencies. We developed an efficient in vitro protocol to produce CD8+ T cells that express natural killer cell markers (CD8+ NKT-like cells) and extensively characterized their functional and molecular phenotypes using a variety of techniques. Results Large numbers of CD8+ NKT-like cells were obtained through culture of naïve CD8+ T cells using anti-CD3/anti-CD28-coated beads and high dose IL-2. These cells possess potent activity in suppressing the proliferation of naïve responder T cells. Gene expression profiling suggests that the cultured CD8+ NKT-like cells and the naïve CD8+ T cells differ by more than 2-fold for about 3,000 genes, among which 314 are upregulated by more than 5-fold and 113 are upregulated by more than 10-fold in the CD8+ NKT-like cells. A large proportion of the highly upregulated genes are soluble factors or surface markers that have previously been implicated in immune suppression or are likely to possess immunosuppressive properties. Many of these genes are regulated by two key cytokines, IL-10 and IFN-γ. The immunosuppressive activities of cells cultured from IL-10-/- and IFN-γ-/- mice are reduced by about 70% and about 50%, respectively, compared to wild-type mice. Conclusion Immunosuppressive CD8+ NKT-like cells can be efficiently produced and their immunosuppressive activity is related to many surface and soluble molecules regulated by IL-10 and IFN-γ.
Collapse
Affiliation(s)
- Li Zhou
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, 15th Street, Augusta, GA 30912, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Ge X, Brown J, Sykes M, Boussiotis VA. CD134-allodepletion allows selective elimination of alloreactive human T cells without loss of virus-specific and leukemia-specific effectors. Biol Blood Marrow Transplant 2008; 14:518-30. [PMID: 18410894 DOI: 10.1016/j.bbmt.2008.02.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 02/13/2008] [Indexed: 10/22/2022]
Abstract
Graft-versus-host disease (GVHD) remains a frequent and severe complication of allogeneic stem cell transplantation (SCT). One approach to reducing alloreactivity is to deplete the graft of alloreactive T cells. Global T cell depletion results in poor immune reconstitution with high mortality from viral infections and disease relapse. Therefore, an approach to selectively deplete alloreactive T cells without compromising other responses would be highly beneficial. We undertook studies to identify an inducible activation marker expressed on alloreactive effector T cells following culture with HLA-mismatched allostimulators. Compared to other markers, CD134 was superior because of its negative baseline expression and rapid upregulation after activation. Depletion of CD134(+) cells from responder populations dramatically reduced specific alloreactivity as determined by reduction of helper T cell precursor frequencies below the threshold predicting development of clinical GVHD while retaining responses to third-party alloantigens. CD134-allodepleted populations retained effectors specific for the Wilms' tumor (WT1) leukemia antigen as determined by WT1 specific pentamers, and CMV-specific effectors as determined by CMV-specific pentamers and CMV-specific ELISpot. Thus, use of CD134-allodepleted grafts may improve allogeneic SCT by reducing GVHD without loss of pathogen-specific and leukemia-specific immunity.
Collapse
Affiliation(s)
- Xupeng Ge
- Transplantation Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
83
|
Abstract
The burden of neurological diseases in western societies has accentuated the need to develop effective therapies to stop the progression of chronic neurological diseases. Recent discoveries regarding the role of the immune system in brain damage coupled with the development of new technologies to manipulate the immune response make immunotherapies an attractive possibility to treat neurological diseases. The wide repertoire of immune responses and the possibility to engineer such responses, as well as their capacity to promote tissue repair, indicates that immunotherapy might offer benefits in the treatment of neurological diseases, similar to the benefits that are being associated with the treatment of cancer and autoimmune diseases. However, before applying such strategies to patients it is necessary to better understand the pathologies to be targeted, as well as how individual subjects may respond to immunotherapies, either in isolation or in combination. Due to the powerful effects of the immune system, one priority is to avoid tissue damage due to the activity of the immune system, particularly considering that the nervous system does not tolerate even the smallest amount of tissue damage.
Collapse
|
84
|
Abstract
The TNFR family members OX40 (CD134) and 4-1BB (CD137) have been found to play major roles as costimulatory receptors for both CD4 and CD8 T cells. In particular, in many situations, they can control proliferation, survival, and cytokine production, and hence are thought to dictate accumulation of protective T cells during anti-viral and anti-tumor responses and pathogenic T cells during autoimmune reactions. As opposed to simply controlling the activity of naïve, effector, and memory T cells, recent data have suggested that both molecules are also instrumental in controlling the generation and activity of so-called regulatory or suppressor T cells (Treg), perhaps in both positive and negative manners. Part of the action on Treg might function to further promote protective or pathogenic T cells, but alternate activities of OX40 and 4-1BB on Treg are also being described that suggest that there might be control by these molecules at multiple levels that will alter the biological outcome when these receptors are ligated. This review specifically focuses on recent studies of regulatory T cells, and regulatory or suppressive activity, that are modulated by OX40 or 4-1BB.
Collapse
Affiliation(s)
- Takanori So
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
85
|
McNamara JO, Kolonias D, Pastor F, Mittler RS, Chen L, Giangrande PH, Sullenger B, Gilboa E. Multivalent 4-1BB binding aptamers costimulate CD8+ T cells and inhibit tumor growth in mice. J Clin Invest 2008; 118:376-86. [PMID: 18060045 DOI: 10.1172/jci33365] [Citation(s) in RCA: 248] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 10/29/2007] [Indexed: 12/14/2022] Open
Abstract
4-1BB is a major costimulatory receptor that promotes the survival and expansion of activated T cells. Administration of agonistic anti-4-1BB Abs has been previously shown to enhance tumor immunity in mice. Abs are cell-based products posing significant cost, manufacturing, and regulatory challenges. Aptamers are oligonucleotide-based ligands that exhibit specificity and avidity comparable to, or exceeding, that of Abs. To date, various aptamers have been shown to inhibit the function of their cognate target. Here, we have described the development of an aptamer that binds 4-1BB expressed on the surface of activated mouse T cells and shown that multivalent configurations of the aptamer costimulated T cell activation in vitro and mediated tumor rejection in mice. Because aptamers can be chemically synthesized, manufacturing and the regulatory approval process should be substantially simpler and less costly than for Abs. Agonistic aptamers could therefore represent a superior alternative to Abs for the therapeutic manipulation of the immune system.
Collapse
Affiliation(s)
- James O McNamara
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Olofsson PS, Söderström LA, Wågsäter D, Sheikine Y, Ocaya P, Lang F, Rabu C, Chen L, Rudling M, Aukrust P, Hedin U, Paulsson-Berne G, Sirsjö A, Hansson GK. CD137 Is Expressed in Human Atherosclerosis and Promotes Development of Plaque Inflammation in Hypercholesterolemic Mice. Circulation 2008; 117:1292-301. [DOI: 10.1161/circulationaha.107.699173] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background—
Atherosclerosis is a multifactorial disease in which inflammatory processes play an important role. Inflammation underlies lesion evolution at all stages, from establishment to plaque rupture and thrombosis. Costimulatory molecules of the tumor necrosis factor superfamily such as CD40/CD40L and OX40/OX40L have been implicated in atherosclerosis.
Methods and Results—
This study shows that the tumor necrosis factor superfamily members CD137 and CD137 ligand (CD137L), which play a major role in several autoimmune diseases, may constitute a pathogenic pair in atherogenesis. We detected CD137 protein in human atherosclerotic lesions not only on T cells but also on endothelial cells and showed that CD137 in cultured endothelial cells and smooth muscle cells was induced by proinflammatory cytokines implicated in atherosclerosis. Activation of CD137 by CD137L induced adhesion molecule expression on endothelial cells and reduced smooth muscle cell proliferation. In addition, treatment of atherosclerosis-prone apolipoprotein E–deficient mice with a CD137 agonist caused increased inflammation. T-cell infiltration, mainly of CD8
+
cells, and expression of the murine major histocompatibility complex class II molecule I-A
b
increased significantly in atherosclerotic lesions, as did the aortic expression of proinflammatory cytokines.
Conclusions—
Taken together, these observations suggest that CD137-CD137L interactions in the vasculature may contribute to the progression of atherosclerosis via augmented leukocyte recruitment, increased inflammation, and development of a more disease-prone phenotype.
Collapse
Affiliation(s)
- Peder S. Olofsson
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Leif Å. Söderström
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Dick Wågsäter
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Yuri Sheikine
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Pauline Ocaya
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - François Lang
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Catherine Rabu
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Lieping Chen
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Mats Rudling
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Pål Aukrust
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Ulf Hedin
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Gabrielle Paulsson-Berne
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Allan Sirsjö
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| | - Göran K. Hansson
- From the Center for Molecular Medicine (P.S.O., L.A.S., Y.S., U.H., G.P.-B., G.K.H.) and Center for Metabolism and Endocrinology (M.R.), Department of Medicine, and Department of Anesthesiology and Intensive Care Medicine (P.S.O.), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Division of Biomedicine, Department of Clinical Medicine, University of Örebro, Örebro, Sweden (D.W., P.O., A.S.); Division of Nuclear Medicine/PET and Noninvasive Cardiovascular Imaging Program,
| |
Collapse
|
87
|
Augmentation of SIV DNA vaccine-induced cellular immunity by targeting the 4-1BB costimulatory molecule. Vaccine 2008; 26:3121-34. [PMID: 18336959 DOI: 10.1016/j.vaccine.2008.02.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
DNA vaccines are effective at inducing antigen-specific cellular immune responses. Approaches to improve these responses, however, are needed. We examined the effect of stimulating 4-1BB, an activation-inducible T-cell costimulatory receptor, by intravenously co-administering anti-human 4-1BB monoclonal antibody (mAb) in DNA-immunized cynomolgus macaques. Three groups of six cynomolgus macaques were immunized intramuscularly with a DNA vaccine encoding SIV Gag antigen (pSIVgag) at weeks 0, 4 and 8. At days 12, 15, and 19, six macaques received anti-4-1BB 4E9 mAb and six macaques received anti-4-1BB 10C7 mAb. Treatment with 10C7 mAb led to a significant augmentation of SIV Gag-specific IFN-gamma, granzyme B and perforin responses. Treatment with humanized 4E9 mAb also resulted in an enhancement of SIV Gag-specific cellular responses but the magnitude was lower compared to animals receiving 10C7 mAb. These responses persisted up to week 40 and were mostly mediated by CD8(+) T cells. Treatment with anti-4-1BB mAb was more effective in driving the CD8(+) T cells toward a more differentiated CCR7(-)/CD45RA(+) effector state. This study demonstrates that targeting the 4-1BB molecule in vivo results in an enhanced and long-lasting cellular immune response. 4-1BB stimulation may be a promising approach to enhance the effectiveness of DNA vaccines.
Collapse
|
88
|
Kim DH, Chang WS, Lee YS, Lee KA, Kim YK, Kwon BS, Kang CY. 4-1BB Engagement Costimulates NKT Cell Activation and Exacerbates NKT Cell Ligand-Induced Airway Hyperresponsiveness and Inflammation. THE JOURNAL OF IMMUNOLOGY 2008; 180:2062-8. [DOI: 10.4049/jimmunol.180.4.2062] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
89
|
Lee SJ, Rossi RJ, Lee SK, Croft M, Kwon BS, Mittler RS, Vella AT. CD134 Costimulation Couples the CD137 Pathway to Induce Production of Supereffector CD8 T Cells That Become IL-7 Dependent. THE JOURNAL OF IMMUNOLOGY 2007; 179:2203-14. [PMID: 17675480 DOI: 10.4049/jimmunol.179.4.2203] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The TNFR superfamily members 4-1BB (CD137) and OX40 (CD134) are costimulatory molecules that potently boost CD8 and CD4 T cell responses. Concomitant therapeutic administration of agonist anti-CD137 and -CD134 mAbs mediates rejection of established tumors and fosters powerful CD8 T cell responses. To reveal the mechanism, the role of CD137 expression by specific CD8 T cells was determined to be essential for optimal clonal expansion and accumulation of effector cells. Nonetheless, dual costimulation induced production of supereffector CD8 T cells when either the specific T cells or the host alone bore CD137. Perhaps surprisingly, the total absence of CD137 prevented anti-CD134 augmentation of supereffector differentiation demonstrating an unappreciated link between these related pathways. Ultimately, it was reasoned that these powerful dual costimulatory responses involved common gamma family members, and we show substantial increases of CD25 and IL-7Ralpha-chain expression by the specific CD8 T cells. To investigate this further, it was shown that IL-7 mediated T cell accumulation, but importantly, a gradual and preferential effect of survival was directed toward supereffector CD8 T cells. In fact, a clear enhancement of effector differentiation was demonstrated to be proportional to the increasing amount of IL-7Ralpha expression by the specific CD8 T cells. Therefore, dual costimulation through CD137 and CD134 drives production and survival of supereffector CD8 T cells through a distinct IL-7-dependent pathway.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Survival/drug effects
- Cell Survival/genetics
- Cell Survival/immunology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- Interleukin-2 Receptor alpha Subunit/genetics
- Interleukin-2 Receptor alpha Subunit/immunology
- Interleukin-7/genetics
- Interleukin-7/immunology
- Mice
- Mice, Knockout
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/immunology
- Receptors, OX40/agonists
- Receptors, OX40/genetics
- Receptors, OX40/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
Affiliation(s)
- Seung-Joo Lee
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
McCandless EE, Klein RS. Molecular targets for disrupting leukocyte trafficking during multiple sclerosis. Expert Rev Mol Med 2007; 9:1-19. [PMID: 17637110 DOI: 10.1017/s1462399407000397] [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] [Indexed: 01/24/2023]
Abstract
AbstractAutoimmune diseases of the central nervous system (CNS) involve the migration of abnormal numbers of self-directed leukocytes across the blood–brain barrier that normally separates the CNS from the immune system. The cardinal lesion associated with neuroinflammatory diseases is the perivascular infiltrate, which comprises leukocytes that have traversed the endothelium and have congregated in a subendothelial space between the endothelial-cell basement membrane and the glial limitans. The exit of mononuclear cells from this space can be beneficial, as when virus-specific lymphocytes enter the CNS for pathogen clearance, or might induce CNS damage, such as in the autoimmune disease multiple sclerosis when myelin-specific lymphocytes invade and induce demyelinating lesions. The molecular mechanisms involved in the movement of lymphocytes through these compartments involve multiple signalling pathways between these cells and the microvasculature. In this review, we discuss adhesion, costimulatory, cytokine, chemokine and signalling molecules involved in the dialogue between lymphocytes and endothelial cells that leads to inflammatory infiltrates within the CNS, and the targeting of these molecules as therapies for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Erin E McCandless
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
91
|
Kim J, Kim HJ, Park K, Kim J, Choi HJ, Yagita H, Nam SH, Cho HR, Kwon B. Costimulatory molecule-targeted immunotherapy of cutaneous graft-versus-host disease. Blood 2007; 110:776-82. [PMID: 17363737 DOI: 10.1182/blood-2006-08-043612] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Abstract
Chronic graft-versus-host disease (cGVHD) is an increasingly frequent complication of allogeneic stem cell transplantation. Current therapies for cGVHD reduce symptoms but are not cures. The B10.D2→Balb/c (H-2d) minor histocompatibility antigen-mismatched model, which reflects clinical and pathological symptoms of human cGVHD, was used in this study. We demonstrated that a single injection of an agonistic monoclonal antibody (mAb) against CD137, a member of the tumor necrosis factor receptor superfamily, reverses skin fibrosis, ulceration, and alopecia, a dominant feature of cGVHD (cutaneous GVHD), ultimately improving general health conditions. The reversal is associated with markedly reduced CD4+ T-cell cytokines and increased apoptosis of donor CD4+ T cells. The Fas pathway is required for ameliorating cutaneous GVHD by anti-CD137 mAb. Taken together, these data indicate that the anti-CD137 mAb has a therapeutic effect on cutaneous GVHD by removing donor CD4+ T cells that cause cutaneous GVHD. Thus, our study demonstrates an agonistic mAb, specific for a costimulatory molecule, as a possible target for therapeutic intervention in cutaneous GVHD.
Collapse
Affiliation(s)
- Juyang Kim
- Department of Biological Science and Immunomodulation Reseach Center, University of Ulsan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Dubrot J, Azpilikueta A, Alfaro C, Murillo O, Arina A, Berraondo P, Hervás-Stubbs S, Melero I. Absence of surface expression of CD137 (4-1BB) on Myeloid-derived suppressor cells. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/s0213-9626(07)70081-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
93
|
Fuse S, Bellfy S, Yagita H, Usherwood EJ. CD8+ T cell dysfunction and increase in murine gammaherpesvirus latent viral burden in the absence of 4-1BB ligand. THE JOURNAL OF IMMUNOLOGY 2007; 178:5227-36. [PMID: 17404306 PMCID: PMC4402709 DOI: 10.4049/jimmunol.178.8.5227] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Studies of costimulatory receptors belonging to the TNFR family have revealed their diverse roles in affecting different stages of the T cell response. The 4-1BB ligand (4-1BBL)/4-1BB pathway has emerged as a receptor-ligand pair that impacts not the initial priming, but later phases of the T cell response, such as sustaining clonal expansion and survival, maintaining memory CD8(+) T cells, and supporting secondary expansion upon Ag challenge. Although the role of this costimulatory pathway in CD8(+) T cell responses to acute viral infections has been well-studied, its role in controlling chronic viral infections in vivo is not known to date. Using the murine gammaherpesvirus-68 (MHV-68) model, we show that 4-1BBL-deficient mice lack control of MHV-68 during latency and show significantly increased latent viral loads. In contrast to acute influenza infection, the numbers of MHV-68-specific memory CD8(+) T cells were maintained during latency. However, the virus-specific CD8(+) T cells showed defects in function, including decreased cytolytic function and impaired secondary expansion. Thus, 4-1BBL deficiency significantly affects the function, but not the number, of virus-specific CD8(+) T cells during gammaherpesvirus latency, and its absence results in an increased viral burden. Our study suggests that the 4-1BB costimulatory pathway plays an important role in controlling chronic viral infections.
Collapse
Affiliation(s)
- Shinichiro Fuse
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756
| | - Sarah Bellfy
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Edward J. Usherwood
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756
- Address correspondence and reprint requests to Dr. Edward J. Usherwood, Department of Microbiology and Immunology, Dartmouth Medical School, 1 Medical Center Drive, Lebanon, NH 03756.
| |
Collapse
|
94
|
Angyal A, Prechl J, Sármay G. Possible therapeutic applications of single-chain antibodies in systemic autoimmune diseases. Expert Opin Biol Ther 2007; 7:691-704. [PMID: 17477806 DOI: 10.1517/14712598.7.5.691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
B cells participate in the induction and maintenance of systemic autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus, via production of pathogenic autoantibodies, contributing to the formation of immune complexes. Immune complex deposition in the kidney and joints causes inflammation and organ destruction, and chemokine production enhances T cell activation and tissue damage. The development of the disorder depends on several factors, for example, genetic susceptibility, environmental factors or immune dysregulation. Traditional therapies, which aimed at the alleviation of symptoms, are giving way to biological therapies with the potential of disrupting disease progression. This article focuses on antibody therapies, especially on the applications of single-chain antibodies, as new biological agents for the treatment of systemic autoimmune disorders.
Collapse
Affiliation(s)
- Adrienn Angyal
- Eötvös Loránd University, Department of Immunology, H-1117 Budapest, Pázmány Péter s.1/c, Hungary.
| | | | | |
Collapse
|
95
|
Zhu Y, Zhu G, Luo L, Flies AS, Chen L. CD137 stimulation delivers an antigen-independent growth signal for T lymphocytes with memory phenotype. Blood 2007; 109:4882-9. [PMID: 17244673 PMCID: PMC1885532 DOI: 10.1182/blood-2006-10-043463] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD137 has long been recognized as a costimulatory receptor for growth and functional maturation of recently activated T cells in the presence of T-cell receptor signal. In this report, we present the fact that, in the absence of MHC and antigen, triggering of CD137 by an agonist monoclonal antibody induces vigorous growth of both CD8(+) and CD4(+) T cells with memory phenotype, whereas it does not affect naive T cells. Moreover, T cells with memory phenotype accumulate progressively in transgenic mice overexpressing CD137 ligand. CD137-mediated proliferation of memory T cells is directly through CD137 on T cells and does not require IL-15 and IFN-gamma. Our results define a new role of CD137 signal in the growth of memory T cells.
Collapse
Affiliation(s)
- Yuwen Zhu
- Department of Dermatology, Johns Hopkins University School of Mediicne, 1550 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
96
|
Irie J, Wu Y, Kachapati K, Mittler RS, Ridgway WM. Modulating protective and pathogenic CD4+ subsets via CD137 in type 1 diabetes. Diabetes 2007; 56:186-96. [PMID: 17192481 DOI: 10.2337/db06-0793] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD137 (TNFRSF9) is an activation-inducible T-cell costimulatory molecule and a member of the tumor necrosis factor (TNF) receptor superfamily. Cd137 is also a candidate gene (in the Idd9.3 interval) for autoimmune diabetes in NOD mice. Here, we demonstrate that anti-CD137 treatment protects NOD mice from diabetes. Anti-CD137-treated mice are not protected from insulitis and still harbor pathogenic T-cells, as demonstrated by transfer studies. Transfer of CD4(+), but not CD8(+), cells from anti-CD137-treated pre-diabetic NOD mice into NOD-scid mice delayed diabetes onset. Anti-CD137 treatment significantly increased the number of CD4(+)CD25(+) cells, which demonstrated intracellular Foxp3 expression and in vitro suppressive activity. The CD4(+)CD25(+) cell subset from anti-CD137-treated mice transferred complete protection from diabetes, whereas the CD4(+)CD25(-) cell subset offered no significant protection. Anti-CD137 treatment of NOD-scid recipients of diabetic spleen cells, however, hastened the onset of disease, showing that the effect of anti-CD137 treatment depends on the balance of pathogenic and protective cells. These results support a critical role for CD137 acting in the early phase of autoimmune diabetes to enhance regulatory cell production. Disease-associated CD137 alleles are likely ineffectual at stimulating a regulatory T-cell population sufficient to prevent disease.
Collapse
Affiliation(s)
- Junichiro Irie
- Division of Rheumatology and Immunology, University of Pittsburgh School of Medicine, PA, USA
| | | | | | | | | |
Collapse
|
97
|
Abstract
Monoclonal antibodies (MAb) have been shown to be effective in inducing immune tolerance in transplantation and autoimmunity. Several different MAb have tolerogenic properties and their effect has been studied in a range of experimental animal models and, in some cases, in clinical trials. The tolerant state seems to be maintained by CD4+ regulatory T cells (Treg), induced in the periphery, capable of suppressing other T cells specific for the same antigens or antigens presented by the same antigen presenting cells. Furthermore, following the initial induction of Treg cells under MAb treatment, Treg cells themselves can maintain the tolerant state in a dominant way in the absence of the therapeutic MAb or other immunosuppressive agents, and are able to recruit other T cells into the regulatory pool--a process named infectious tolerance.
Collapse
Affiliation(s)
- Ana Agua-Doce
- Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| | | |
Collapse
|
98
|
Harrison JM, Bertram EM, Ramshaw IA. Exploiting 4-1BB Costimulation for Enhancing Antiviral Vaccination. Viral Immunol 2006; 19:593-601. [PMID: 17201654 DOI: 10.1089/vim.2006.19.593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
4-1BB, a member of the tumor necrosis factor receptor (TNFR) superfamily, is emerging as an important costimulatory molecule, particularly in the regulation of CD8(+) T cell responses. Costimulation through 4-1BB, such as by utilizing agonistic anti-4-1BB monoclonal antibodies, has been well studied in various tumor models. However, 4-1BB is also an important regulator of antiviral CD8(+) T cell responses. This review summarizes these findings and describes how 4-1BB is beginning to be exploited in terms of boosting antiviral vaccine responses.
Collapse
Affiliation(s)
- Jodie M Harrison
- Department of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | | | | |
Collapse
|
99
|
Serghides L, Vidric M, Watts TH. Approaches to studying costimulation of human antiviral T cell responses: prospects for immunotherapeutic vaccines. Immunol Res 2006; 35:137-50. [PMID: 17003516 DOI: 10.1385/ir:35:1:137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/19/2022]
Abstract
The generation of strong and specific CD8 T cell responses is important in the control of viral infections. Costimulatory molecules provide signals necessary for the development or maintenance of these responses. A major focus of our laboratory is to investigate the role of costimulatory molecules of the TNFR and CD28 families in antiviral responses. Our aim is to translate information obtained using murine models to the study of these molecules using human cells. We have devised an in vitro system using recombinant replication- deficient adenovirus to deliver costimulatory molecules to antigen-presenting cells that are then used to stimulate autologous T cells from both healthy and HIV-infected individuals. Here we describe our findings and discuss the implications of incorporating costimulatory molecules into viral vector vaccine strategies.
Collapse
Affiliation(s)
- Lena Serghides
- Department of Immunology, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
100
|
Liu GZ, Gomes AC, Putheti P, Karrenbauer V, Kostulas K, Press R, Hillert J, Hjelmström P, Gao XG. Increased Soluble 4-1BB Ligand (4-1BBL) Levels in Peripheral Blood of Patients with Multiple Sclerosis. Scand J Immunol 2006; 64:412-9. [PMID: 16970683 DOI: 10.1111/j.1365-3083.2006.01796.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
4-1BB ligand (4-1BBL; CD137L) is a member of the tumour necrosis factor superfamily expressed primarily on antigen presenting cells such as B cells, macrophages and dendritic cells. Its engagement with the receptor 4-1BB (CD137) has been shown to promote T-cell activation and regulate proliferation and survival of T cells. The role of the costimulatory molecule in multiple sclerosis (MS) remains unclear. In this study, the expression of 4-1BBL and soluble 4-1BBL (s4-1BBL) protein levels were analysed in peripheral blood of MS patients. Compared with healthy controls, MS patients had an increase in both plasma s4-1BBL protein levels and expression of 4-1BBL in CD14(+) monocytes. In contrast, myelin basic protein-reactive T-cell proliferation was not found to be inhibited by the use of an anti-4-1BBL antibody. The elevated s4-1BBL protein levels in the MS patients may function as a self-regulatory mechanism of 4-1BB/4-1BBL interaction and costimulation.
Collapse
Affiliation(s)
- G-Z Liu
- Department of Neurology, Peking University People's Hospital, Beijing, China.
| | | | | | | | | | | | | | | | | |
Collapse
|