51
|
Abstract
Regulatory T cells (Tregs) and the PD-1: PD-ligand (PD-L) pathway are both critical to terminating immune responses. Elimination of either can result in the breakdown of tolerance and the development of autoimmunity. The PD-1: PD-L pathway can thwart self-reactive T cells and protect against autoimmunity in many ways. In this review, we highlight how PD-1 and its ligands defend against potentially pathogenic self-reactive effector T cells by simultaneously harnessing two mechanisms of peripheral tolerance: (i) the promotion of Treg development and function and (ii) the direct inhibition of potentially pathogenic self-reactive T cells that have escaped into the periphery. Treg cells induced by the PD-1 pathway may also assist in maintaining immune homeostasis, keeping the threshold for T-cell activation high enough to safeguard against autoimmunity. PD-L1 expression on non-hematopoietic cells as well as hematopoietic cells endows PD-L1 with the capacity to promote Treg development and enhance Treg function in lymphoid organs and tissues that are targets of autoimmune attack. At sites where transforming growth factor-beta is present (e.g. sites of immune privilege or inflammation), PD-L1 may promote the de novo generation of Tregs. When considering the consequences of uncontrolled immunity, it would be therapeutically advantageous to manipulate Treg development and sustain Treg function. Thus, this review also discusses how the PD-1 pathway regulates a number of autoimmune diseases and the therapeutic potential of PD-1: PD-L modulation.
Collapse
Affiliation(s)
- Loise M. Francisco
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
- Brigham & Women’s Hospital, Boston, MA, USA
| | - Peter T. Sage
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
| | - Arlene H. Sharpe
- Departments of Pathology, Harvard Medical School, Boston, MA, USA
- Brigham & Women’s Hospital, Boston, MA, USA
| |
Collapse
|
52
|
Leung S, Liu X, Fang L, Chen X, Guo T, Zhang J. The cytokine milieu in the interplay of pathogenic Th1/Th17 cells and regulatory T cells in autoimmune disease. Cell Mol Immunol 2010; 7:182-9. [PMID: 20383174 PMCID: PMC4002916 DOI: 10.1038/cmi.2010.22] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 12/12/2022] Open
Abstract
The propagation and regulation of an immune response is driven by a network of effector and regulatory T (Treg) cells. The interplay of effector T and Treg cells determines the direction of the immune response towards inflammation or its resolution in an autoimmune disease setting. In autoimmune diseases, this interplay shifts the balance in favor of the development of autoreactive effector T cells, resulting in inflammatory pathology. The objective of an effective therapeutic approach for autoimmune disease is to restore this balance. In this review, we describe the characteristics and development of pathogenic T helper 1 (Th1) and Th17 cells and the beneficial Treg cells in autoimmune diseases and the crucial roles of the cytokine milieu in influencing the balance of these T-cell subsets. Given the importance of cytokines, we discuss current immunotherapeutic strategies using cytokine or cytokine receptor antibodies for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Stewart Leung
- Department of Neuroimmunology, GlaxoSmithKline Research and Development Center, Zhangjiang Pudong, Shanghai, China
| | | | | | | | | | | |
Collapse
|
53
|
Nizar S, Meyer B, Galustian C, Kumar D, Dalgleish A. T regulatory cells, the evolution of targeted immunotherapy. Biochim Biophys Acta Rev Cancer 2010; 1806:7-17. [PMID: 20188145 DOI: 10.1016/j.bbcan.2010.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 02/15/2010] [Accepted: 02/17/2010] [Indexed: 01/09/2023]
Abstract
T regulatory cells are able to suppress anti-tumour immunity in pre-clinical models and in patients. This review highlights the important discoveries in Treg immunology critical to the evolution of targeted immunotherapy. We also describe the therapeutic applications that are currently being assessed and their future potential.
Collapse
Affiliation(s)
- S Nizar
- Department of Cellular and Molecular Medicine, St George's University of London, UK
| | | | | | | | | |
Collapse
|
54
|
HoChang S, Jung EJ, Park YH, Lim DG, Ko NY, Choi WS, Her E, Kim SH, Choi KD, Bae JH, Kim SH, Kang CD, Han DJ, Kim SC. Anti-inflammatory effects of Artemisia princeps in antigen-stimulated T cells and regulatory T cells. J Pharm Pharmacol 2010. [DOI: 10.1211/jpp.61.08.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
The aim was to investigate the anti-inflammatory effects of Artemisia princeps extract on the activity of anti-CD3/CD28-stimulated CD4+CD25- T cells and antigen-expanded regulatory T cells.
Methods
CD4+CD25- T cells were activated with coated anti-CD3 and anti-CD28 and cultured in the presence or absence of various concentrations of A. princeps extract. The cultures were pulsed on Day 6 with [3H]thymidine and, after harvesting the cells, [3H] thymidine incorporation was measured. For analysis of interleukin-2 and interferon-γ secreted from CD4+CD25- T cells, culture supernatants were collected on Days 2 and 6. For the analysis of interleukin-10 secreted from the CD4+CD25- T cells and expanded regulatory T cells, supernatants were collected after 2 and 7 days, respectively. Cytokine levels were determined using an enzyme-linked immunosorbent assay. Potential medicinal components of the A. princeps extract were determined using gas chromatography–mass spectrometry.
Key findings
A. princeps (30 μg/ml) effectively suppressed proliferation of CD4+CD25- T cells that were stimulated with anti-CD3/CD28 without causing cytotoxicity in spleen cells incubated under conditions lacking antigen stimulation. A. princeps inhibited production of the pro-inflammatory cytokines interleukin-2 and interferon-γ in anti-CD3/CD28-stimulated CD4+CD25- T cells. Also, the extract slightly increased production of the anti-inflammatory cytokine interleukin-10 in these cells. In regulatory T cells expanded by anti-CD3/CD28, A. princeps increased production of interleukin-10 and Foxp3.
Conclusions
The results suggest that A. princeps may be useful in the treatment of autoimmune diseases and organ transplantation rejection by inhibiting proliferation of inflammatory T cells, suppressing inflammatory processes in antigen-stimulated CD4+CD25- T cells and increasing activity of expanded regulatory T cells.
Collapse
Affiliation(s)
- Sung HoChang
- BK21 Medical Science Education Center and Department of Biochemistry, Pusan National University School of Medicine, Yangsan-city, Gyeongsangnam-do, Korea
| | - Eun Jung Jung
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Songpa-gu, Seoul, Korea
| | - Youn Hee Park
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Songpa-gu, Seoul, Korea
| | - Dong Gyun Lim
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Songpa-gu, Seoul, Korea
| | - Na Young Ko
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Korea
| | - Erk Her
- Department of Immunology, College of Medicine, Konkuk University, Chungju, Korea
| | - Soo Hyun Kim
- Laboratory of Cytokine Immunology, Institute of Biomedical Science and Technology, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Kang Duk Choi
- Gyeong-gi Regional Research Center, Hankyong National University, Ansung-city, Gyeonggi-do, Korea
| | - Jae Ho Bae
- BK21 Medical Science Education Center and Department of Biochemistry, Pusan National University School of Medicine, Yangsan-city, Gyeongsangnam-do, Korea
| | - Sun Hee Kim
- BK21 Medical Science Education Center and Department of Biochemistry, Pusan National University School of Medicine, Yangsan-city, Gyeongsangnam-do, Korea
| | - Chi Dug Kang
- BK21 Medical Science Education Center and Department of Biochemistry, Pusan National University School of Medicine, Yangsan-city, Gyeongsangnam-do, Korea
| | - Duck Jong Han
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Songpa-gu, Seoul, Korea
| | - Song Cheol Kim
- Department of Surgery and Asan Institute for Life Sciences, Ulsan University College of Medicine and Asan Medical Center, Songpa-gu, Seoul, Korea
| |
Collapse
|
55
|
Sakaguchi S, Wing K, Yamaguchi T. Dynamics of peripheral tolerance and immune regulation mediated by Treg. Eur J Immunol 2009; 39:2331-6. [PMID: 19662638 DOI: 10.1002/eji.200939688] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Peripheral self-tolerance and immune homeostasis are maintained, at least in part, by the balance between Treg and effector T cells. Naturally, arising CD25(+)CD4(+) Treg, which express the transcription factor Foxp3, suppress the activation and proliferation of other lymphocytes in multiple ways. A CTLA-4-dependent suppressive mechanism is shared by every Foxp3(+) Treg at any location and its disruption breaches self-tolerance and immune homeostasis, suggesting that it is a core mechanism of suppression. Depending on the environment, Foxp3(+) Treg also differentiate to exhibit additional suppressive mechanisms, including the secretion of immunosuppressive cytokines. Naïve T cells acquire Foxp3 expression and suppressive activity under certain in vivo and in vitro conditions, whereas some Foxp3(+) T cells may lose Foxp3 and suppressive activity following proliferation in an IL-2-deficient environment. Moreover, activated effector T cells frequently secrete suppressive cytokines, such as IL-10, in a negative feedback fashion. These findings, when taken together, indicate that peripheral immune tolerance and homeostasis are dynamically maintained by functional differentiation within the Foxp3(+) population, occasional conversion between Treg and non-Treg cells, and the interactions among them. These dynamics provide ample opportunities for immune intervention for the benefit of the host.
Collapse
Affiliation(s)
- Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.
| | | | | |
Collapse
|
56
|
Sakaguchi S, Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T. Regulatory T cells: how do they suppress immune responses? Int Immunol 2009; 21:1105-11. [PMID: 19737784 DOI: 10.1093/intimm/dxp095] [Citation(s) in RCA: 650] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells (Tregs), either natural or induced, suppress a variety of physiological and pathological immune responses. One of the key issues for understanding Treg function is to determine how they suppress other lymphocytes at the molecular level in vivo and in vitro. Here we propose that there may be a key suppressive mechanism that is shared by every forkhead box p3 (Foxp3)(+) Treg in vivo and in vitro in mice and humans. When this central mechanism is abrogated, it causes a breach in self-tolerance and immune homeostasis. Other suppressive mechanisms may synergistically operate with this common mechanism depending on the environment and the type of an immune response. Further, Treg-mediated suppression is a multi-step process and impairment or augmentation of each step can alter the ultimate effectiveness of Treg-mediated suppression. These findings will help to design effective ways for controlling immune responses by targeting Treg suppressive functions.
Collapse
Affiliation(s)
- Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
57
|
Field J, Biondo MA, Murphy K, Alderuccio F, Toh BH. Experimental Autoimmune Gastritis: Mouse Models Of Human Organ-specific Autoimmune Disease. Int Rev Immunol 2009; 24:93-110. [PMID: 15763991 DOI: 10.1080/08830180590884585] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Experimental autoimmune gastritis (EAG) is an excellent model of human autoimmune gastritis, the underlying cause of pernicious anaemia. Murine autoimmune gastritis replicates human gastritis in being characterized by a chronic inflammatory mononuclear cell infiltrate in the gastric mucosa, destruction of parietal and zymogenic cells, and autoantibodies to the alpha-and beta-subunits of the gastric H+/K+ ATPase. Disease is induced strain specifically in gastritis-susceptible BALB/c mice by methods with a greater variety than those for most other experimental autoimmune diseases. The disease is induced in the regional gastric lymph node in which pathogenic CD4+ T cells are recruited. The model provides an excellent illustration of regulation by CD4+CD25+T cells, and, indeed, the removal of such regulatory cells, e.g., by neonatal thymectomy, is thought to be a major mechanism by which disease can develop. The culprit T helper type 1 (Th1) CD4+ T cells recognize either the alpha- or beta-subunits of the gastric H+/K+ ATPase, but the beta-subunit appears to be the initiating autoantigen, while the alpha-subunit may have a role in perpetuating disease. Since no specific environmental modifiers are identifiable, the origins of the disease are intrinsic; this is illustrated by the capacity of a cytokine (GM-CSF)-dependent inflammatory stimulus in the stomach to initiate EAG, according to a transgenic model in which thymectomy is dispensible. Thus, EAG is an exquisite model for a reductionist analysis of the multiple elements that in combination induce autoimmunity in humans.
Collapse
Affiliation(s)
- J Field
- Department of Pathology and Immunology, Monash University Medical School, Prahran, Victoria, Australia
| | | | | | | | | |
Collapse
|
58
|
Collison LW, Pillai MR, Chaturvedi V, Vignali DAA. Regulatory T cell suppression is potentiated by target T cells in a cell contact, IL-35- and IL-10-dependent manner. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:6121-8. [PMID: 19414764 PMCID: PMC2698997 DOI: 10.4049/jimmunol.0803646] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regulatory T cells (T(reg)) are believed to suppress conventional T cell (T(conv)) proliferation in vitro in a contact-dependent, cytokine-independent manner, based in part on experiments in which T(reg) and T(conv) are separated by a permeable membrane. We show that the production of IL-35, a novel inhibitory cytokine expressed by natural T(reg), increases substantially following contact with T(conv). Surprisingly, T(reg) were able to mediate potent suppression of T(conv) across a permeable membrane when placed in direct contact with T(conv) in the upper chamber of a Transwell plate. Suppression was IL-35 and IL-10 dependent, and T(conv) activation was required for maximal potentiation of T(reg) suppression. These data suggest that it is the induction of suppression, rather than the function of T(reg) that is obligatorily contact dependent.
Collapse
Affiliation(s)
- Lauren W. Collison
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Meenu R. Pillai
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Vandana Chaturvedi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Dario A. A. Vignali
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
59
|
Kelchtermans H, Geboes L, Mitera T, Huskens D, Leclercq G, Matthys P. Activated CD4+CD25+ regulatory T cells inhibit osteoclastogenesis and collagen-induced arthritis. Ann Rheum Dis 2009; 68:744-50. [PMID: 18480308 DOI: 10.1136/ard.2007.086066] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Patients with rheumatoid arthritis (RA) have defective CD4(+)CD25(+) regulatory T (T(reg)) cells and increased osteoclastogenesis. A similar situation has been described in collagen-induced arthritis (CIA). In this study, it was investigated whether a single transfer of polyclonally activated T(reg) cells inhibits CIA and osteoclastogenesis. METHODS Purified T(reg) cells were expanded in vitro with anti-CD3 and anti-CD28 antibody-coated beads and injected into DBA/1 mice. Mice were immunised with collagen type II (CII) in complete Freund adjuvant (CFA) and scores of arthritis were recorded. In vitro osteoclastogenesis assays were performed on splenocytes by stimulation with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor (NF)kappaB ligand (RANKL). Levels of anti-CII antibody and cytokines were determined in the supernatant using ELISA and Bio-Plex protein array system. RESULTS It was found that 10(6) activated T(reg) cells significantly counteracted the development of CIA, which was accompanied by decreased serum levels of TNFalpha and IL6, but not by inhibition of autoimmune antibody responses. The differentiation of osteoclasts in splenocyte cultures was significantly reduced in the presence of prestimulated T(reg) cells. Expression of cytokines that are described to inhibit osteoclastogenesis, including granulocyte macrophage colony-stimulating factor (GM-CSF), interferon (IFN)gamma, interleukin (IL)5 and IL10, were dramatically increased upon addition of T(reg) cells. Furthermore, splenocytes from mice that had been treated with T(reg) cells displayed an impaired capacity to develop into mature osteoclasts, suggesting that T(reg) cells abrogated osteoclastogenesis in vivo. CONCLUSIONS Activated CD4(+)CD25(+) T(reg) cells improve clinical symptoms of CIA, regulate cytokine production and inhibit osteoclastogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- H Kelchtermans
- Laboratory of Immunobiology, Rega Institute, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
60
|
CD4+Foxp3+ regulatory T cells in the control of autoimmunity: in vivo veritas. Curr Opin Immunol 2008; 20:655-62. [DOI: 10.1016/j.coi.2008.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 09/25/2008] [Accepted: 09/29/2008] [Indexed: 01/11/2023]
|
61
|
Hahn YS, Ji XY, Woo SI, Choi YK, Song MS, Shin KS, Jin N, O'Brien RL, Born WK. Vγ1+ γδ T cells reduce IL-10-producing CD4+CD25+ T cells in the lung of ovalbumin-sensitized and challenged mice. Immunol Lett 2008; 121:87-92. [PMID: 18840468 DOI: 10.1016/j.imlet.2008.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Revised: 08/16/2008] [Accepted: 09/05/2008] [Indexed: 10/21/2022]
Abstract
In OVA-sensitized and challenged mice, gammadelta T cells expressing Vgamma1 enhance airway hyperresponsiveness (AHR) but the underlying mechanism is unclear. These cells also reduce IL-10 levels in the airways, suggesting that they might function by inhibiting CD4(+)CD25(+) regulatory T cells (T(reg)) or other CD4(+) T cells capable of producing IL-10 and suppressing AHR. Indeed, sensitization and challenge with OVA combined with inactivation of Vgamma1(+) cells increased CD4(+)CD25(+) cells in the lung, and markedly those capable of producing IL-10. The cellular change was associated with increased IL-10 and TGF-beta levels in the airways, and a decrease of IL-13. T(reg) include naturally occurring Foxp3(+) T(reg), inducible Foxp3(-) T(reg), and antigen-specific T(reg) many of which express folate receptor 4 (FR4). Although Foxp3 gene expression in the lung was also increased pulmonary CD4(+) T cells, expressing Foxp3-protein or FR4 remained stable. Therefore, the inhibition by Vgamma1(+) gammadelta T cells might not be targeting Foxp3(+) T(reg) but rather CD4(+) T cells destined to produce IL-10.
Collapse
Affiliation(s)
- Youn-Soo Hahn
- Department of Pediatrics, College of Medicine and Medical Research Institute, Chungbuk National University, 62 Kaeshin-dong, Hungduk-gu, Cheongju 361-711, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Kearley J, Robinson DS, Lloyd CM. CD4+CD25+ regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling. J Allergy Clin Immunol 2008; 122:617-24.e6. [PMID: 18672278 PMCID: PMC3389733 DOI: 10.1016/j.jaci.2008.05.048] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 04/25/2008] [Accepted: 05/28/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND CD4(+)CD25(+) regulatory T cells can inhibit excessive T-cell responses in vivo. We have previously demonstrated that prophylactic administration of CD4(+)CD25(+) regulatory T cells suppresses the development of acute allergen-induced airway inflammation in vivo. OBJECTIVE We sought to determine the effect of therapeutic transfer of CD4(+)CD25(+) regulatory T cells on established pulmonary inflammation and the subsequent development of airway remodeling. METHODS CD4(+)CD25(+) cells were transferred after the onset of allergic inflammation, and airway challenges were continued to induce chronic inflammation and airway remodeling. RESULTS Administration of CD4(+)CD25(+) regulatory T cells reduced established lung eosinophilia, T(H)2 infiltration, and expression of IL-5, IL-13, and TGF-beta. Moreover, subsequent mucus hypersecretion and peribronchial collagen deposition were reduced after prolonged challenge. In contrast, transfer of CD4(+)CD25(+) regulatory T cells had no effect on established airway hyperreactivity either 7 days or 4 weeks after transfer. CONCLUSIONS In this study we demonstrate for the first time that therapeutic transfer of CD4(+)CD25(+) regulatory T cells can resolve features of chronic allergen-induced inflammation and prevent development of airway remodeling.
Collapse
Affiliation(s)
- Jennifer Kearley
- Leukocyte Biology Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Douglas S. Robinson
- Leukocyte Biology Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, United Kingdom
- Allergy and Clinical Immunology, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Clare M. Lloyd
- Leukocyte Biology Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, United Kingdom
| |
Collapse
|
63
|
Fragale A, Gabriele L, Stellacci E, Borghi P, Perrotti E, Ilari R, Lanciotti A, Remoli AL, Venditti M, Belardelli F, Battistini A. IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression. THE JOURNAL OF IMMUNOLOGY 2008; 181:1673-82. [PMID: 18641303 DOI: 10.4049/jimmunol.181.3.1673] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Regulatory T (Treg) cells are critical in inducing and maintaining tolerance. Despite progress in understanding the basis of immune tolerance, mechanisms and molecules involved in the generation of Treg cells remain poorly understood. IFN regulatory factor (IRF)-1 is a pleiotropic transcription factor implicated in the regulation of various immune processes. In this study, we report that IRF-1 negatively regulates CD4(+)CD25(+) Treg cell development and function by specifically repressing Foxp3 expression. IRF-1-deficient (IRF-1(-/-)) mice showed a selective and marked increase of highly activated and differentiated CD4(+)CD25(+)Foxp3(+) Treg cells in thymus and in all peripheral lymphoid organs. Furthermore, IRF-1(-/-) CD4(+)CD25(-) T cells showed extremely high bent to differentiate into CD4(+)CD25(+)Foxp3(+) Treg cells, whereas restoring IRF-1 expression in IRF-1(-/-) CD4(+)CD25(-) T cells impaired their differentiation into CD25(+)Foxp3(+) cells. Functionally, both isolated and TGF-beta-induced CD4(+)CD25(+) Treg cells from IRF-1(-/-) mice exhibited more increased suppressive activity than wild-type Treg cells. Such phenotype and functional characteristics were explained at a mechanistic level by the finding that IRF-1 binds a highly conserved IRF consensus element sequence (IRF-E) in the foxp3 gene promoter in vivo and negatively regulates its transcriptional activity. We conclude that IRF-1 is a key negative regulator of CD4(+)CD25(+) Treg cells through direct repression of Foxp3 expression.
Collapse
Affiliation(s)
- Alessandra Fragale
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Gotsch F, Romero R, Kusanovic JP, Erez O, Espinoza J, Kim CJ, Vaisbuch E, Than NG, Mazaki-Tovi S, Chaiworapongsa T, Mazor M, Yoon BH, Edwin S, Gomez R, Mittal P, Hassan SS, Sharma S. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med 2008; 21:529-47. [PMID: 18609361 PMCID: PMC6333088 DOI: 10.1080/14767050802127349] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The anti-inflammatory limb of the immune response is crucial for dampening inflammation. Spontaneous parturition at term and preterm labor (PTL) are mediated by inflammation in the cervix, membranes, and myometrium. This study focuses on the changes in the amniotic fluid concentrations of the anti-inflammatory cytokine interleukin (IL)- 10. The objectives of this study were to determine whether there is a relationship between amniotic fluid concentrations of IL-10 and gestational age, parturition (at term and preterm), and intra-amniotic infection/inflammation (IAI). STUDY DESIGN A cross-sectional study was conducted including 301 pregnant women in the following groups: (1) mid-trimester of pregnancy who delivered at term (n = 112); (2) mid-trimester who delivered preterm neonates (n = 30); (3) term not in labor without IAI (n = 40); (4) term in labor without IAI (n = 24); (5) term in labor with IAI (n = 20); (6) PTL without IAI who delivered at term (n = 31); (7) PTL without IAI who delivered preterm (n = 30); (8) PTL with IAI who delivered preterm (n = 14). IL-10 concentrations in amniotic fluid were determined by a specific and sensitive immunoassay. Non-parametric statistics were used for analysis. RESULTS (1) IL-10 was detectable in amniotic fluid and its median concentration did not change with gestational age from mid-trimester to term. (2) Patients in labor at term had a significantly higher median amniotic fluid IL-10 concentration than that of patients at term not in labor (p = 0.04). (3) Women at term in labor with IAI had a significantly higher median amniotic fluid IL-10 concentration than that of patients at term in labor without IAI (p = 0.02). (4) Women with PTL and IAI who delivered preterm had a significantly higher median amniotic fluid concentration of IL-10 than those without IAI who delivered preterm and than those who delivered at term (p = 0.009 and p < 0.001, respectively). (5) Among patients with preterm labor without IAI, those who delivered preterm had a significantly higher median amniotic fluid IL-10 concentration than those who delivered at term (p = 0.03). CONCLUSIONS The anti-inflammatory cytokine IL-10 is detectable in the amniotic fluid of normal pregnant women. Spontaneous parturition at term and in preterm gestation is associated with increased amniotic fluid concentrations of IL-10. IAI (preterm and at term) is also associated with increased amniotic fluid concentrations of IL-10. We propose that IL-10 has a role in the regulation of the immune response in vivo by initiating actions that dampen inflammation.
Collapse
Affiliation(s)
- Francesca Gotsch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Jimmy Espinoza
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University, Detroit, Michigan, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Moshe Mazor
- Department of Obstetrics and Gynecology, Soroka Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Bo Hyun Yoon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Samuel Edwin
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Ricardo Gomez
- Center for Perinatal Diagnosis and Research (CEDIP), Hospital Sotero del Rio, Universidad Catolica de Cile, Puente Alto, Chile
| | - Pooja Mittal
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University/Hutzel Women’s Hospital, Detroit, Michigan, USA
| | - Surendra Sharma
- Department of Pediatrics and Pathology, Brown University, Providence, RI, USA
| |
Collapse
|
65
|
Stummvoll GH, DiPaolo RJ, Huter EN, Davidson TS, Glass D, Ward JM, Shevach EM. Th1, Th2, and Th17 effector T cell-induced autoimmune gastritis differs in pathological pattern and in susceptibility to suppression by regulatory T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1908-16. [PMID: 18641328 PMCID: PMC2561289 DOI: 10.4049/jimmunol.181.3.1908] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Th cells can be subdivided into IFN-gamma-secreting Th1, IL-4/IL-5-secreting Th2, and IL-17-secreting Th17 cells. We have evaluated the capacity of fully differentiated Th1, Th2, and Th17 cells derived from a mouse bearing a transgenic TCR specific for the gastric parietal cell antigen, H(+)K(+)-ATPase, to induce autoimmune gastritis after transfer to immunodeficient recipients. We have also determined the susceptibility of the disease induced by each of the effector T cell types to suppression by polyclonal regulatory T cells (Treg) in vivo. Each type of effector cell induced autoimmune gastritis with distinct histological patterns. Th17 cells induced the most destructive disease with cellular infiltrates composed primarily of eosinophils accompanied by high levels of serum IgE. Polyclonal Treg could suppress the capacity of Th1 cells, could moderately suppress Th2 cells, but could suppress Th17-induced disease only at early time points. The major effect of the Treg was to inhibit the expansion of the effector T cells. However, effector cells isolated from protected animals were not anergic and were fully competent to proliferate and produce effector cytokines ex vivo. The strong inhibitory effect of polyclonal Treg on the capacity of some types of differentiated effector cells to induce disease provides an experimental basis for the clinical use of polyclonal Treg in the treatment of autoimmune disease in humans.
Collapse
Affiliation(s)
- Georg H. Stummvoll
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Richard J. DiPaolo
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Eva N. Huter
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Todd S. Davidson
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Deborah Glass
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Jerrold M. Ward
- Comparative Medicine Branch, Infectious Disease Pathogenesis Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| | - Ethan M. Shevach
- Laboratory of Immunology, Cellular Immunology Section, National Institutes of Allergy and Infectious Diseaes, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
66
|
In Vitro Expanded Human CD4+CD25+ Regulatory T Cells are Potent Suppressors of T-Cell-Mediated Xenogeneic Responses. Transplantation 2008; 85:1841-8. [DOI: 10.1097/tp.0b013e3181734793] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
67
|
Sydora BC, MacFarlane SM, Tavernini MM, Doyle JSG, Fedorak RN. CD4+CD25+ regulatory T cells have divergent effects on intestinal inflammation in IL-10 gene-deficient mice. Dig Dis Sci 2008; 53:1544-52. [PMID: 17990114 DOI: 10.1007/s10620-007-0064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 10/04/2007] [Indexed: 12/09/2022]
Abstract
The regulatory effect of murine CD4+CD25+ T-cells in vivo appears to be dependent on the secretion of IL-10. The lack of IL-10 in the IL-10 gene-deficient mouse has a profoundly negative effect on the mouse's regulation of the response to intestinal bacteria, resulting in severe enterocolitis. We investigated the effect of neonatal injection with wild-type CD4+CD25+ T-cells on the intestinal immune response in IL-10 gene-deficient mice. At the time of analysis, 8-15 weeks later, all mice demonstrated an increased, antigen-stimulated systemic response. However, the intestinal response was divergent with about half of the mice developing an intestinal inflammation with a high injury score, the other half demonstrating a remarkable reduction in injury score with a marked decrease in intestinal IFNgamma release. Our data demonstrate that CD4+CD25+ T-cells can be activated in IL-10 gene-deficient mice and that this stimulation under stringent conditions has the potential to reduce intestinal inflammation.
Collapse
Affiliation(s)
- Beate C Sydora
- Department of Medicine, Division of Gastroenterology, Center of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Zeidler Ledcor Building, Edmonton, Alberta, Canada.
| | | | | | | | | |
Collapse
|
68
|
Protective role of interleukin-10-producing regulatory dendritic cells against murine autoimmune gastritis. J Gastroenterol 2008; 43:100-7. [PMID: 18306983 DOI: 10.1007/s00535-007-2133-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Accepted: 10/11/2007] [Indexed: 02/04/2023]
Abstract
BACKGROUND Regulatory dendritic cells (Reg-DCs), which induce regulatory T cells and interleukin (IL)-10 in vitro, are capable of inducing immunogenic tolerance in vivo. In this study, we assessed whether Reg-DCs modulate the course of autoimmune processes in a murine model of autoimmune gastritis (AIG). METHODS AIG mice were produced by neonatal thymectomy of 3-day old BALB/c mice followed by administration of polyinosinic:polycytidylic acid (poly I:C). Reg-DCs were produced by culturing bone marrow DCs with IL-10, lipopolysaccharide, and parietal cell (PC) antigen for 2 days. In the course of development of AIG, BALB/c mice were administered either Reg-DCs, mature DCs, or phosphate-buffered saline, intraperitoneally, four times. The levels of gastritis and autoantibody to PC antigen were assessed serially in these mice. RESULTS The stages of gastritis and the titers of autoantibody to PC antigen were significantly lower in Reg-DC-treated mice than in mature DC-treated mice (P<0.05). Spleen cells from Reg-DC-treated mice produced increased levels of IL-10 and decreased levels of IL-12p70 and interferon-gamma (P<0.05). Also, frequencies of IL-10-producing CD4(+)CD25(+) T cells in the spleen and Foxp3(+)CD4(+)CD25(+) T cells in the peripheral blood were significantly higher in Reg-DC-treated mice than in mature DC-treated mice (P<0.05). CONCLUSIONS Taken together, these results suggest that increased induction of CD4(+)CD25(+) regulatory T cells by Reg-DCs might contribute to downregulation of inflammatory processes and autoantibody production during AIG development in mice.
Collapse
|
69
|
Luo X, Zhang Q, Liu V, Xia Z, Pothoven KL, Lee C. Cutting edge: TGF-beta-induced expression of Foxp3 in T cells is mediated through inactivation of ERK. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:2757-61. [PMID: 18292494 PMCID: PMC4289405 DOI: 10.4049/jimmunol.180.5.2757] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The peripheral induction of T regulatory cells can be accomplished by TGF-beta through an epigenetic regulation leading to the expression of Foxp3. However, the exact mechanism of such a TGF-beta-mediated action remains unclear. In the current study, we found that TGF-beta treatment of CD4+CD25- T cells during T cell activation led to a transient inhibition of the phosphorylation of ERK followed by the induction of Foxp3 expression in these cells. Direct treatment with a specific ERK inhibitor, UO126, during CD4+CD25- T cell activation also induced Foxp3 expression and conferred a suppressive function to the induced Foxp3+ T cells. Furthermore, treatment of T cells with either TGF-beta or UO126 significantly down-regulated the expression of DNMTs, a reaction normally elicited by demethylation agents, such as 5-Aza-2'-deoxycytidine. These results indicate that the epigenetic regulation of TGF-beta-induced expression of Foxp3 may be mediated through the inactivation of ERK.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors
- DNA (Cytosine-5-)-Methyltransferases/biosynthesis
- DNA Methylation
- DNA Methyltransferase 3A
- Down-Regulation/genetics
- Down-Regulation/immunology
- Enzyme Activation/immunology
- Forkhead Transcription Factors/biosynthesis
- Gene Expression Regulation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Transgenic
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/metabolism
- Phosphorylation
- Receptors, Antigen, T-Cell/physiology
- Resting Phase, Cell Cycle/immunology
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transforming Growth Factor beta1/physiology
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Qiang Zhang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Victoria Liu
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Zhenbiao Xia
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kathryn L. Pothoven
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Chung Lee
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
70
|
Abstract
The function of regulatory T cells (T(reg) cells) has been attributed to a growing number of diverse pathways, molecules and processes. Seemingly contradictory conclusions regarding the mechanisms underlying T(reg) cell suppressive activity have revitalized skeptics in the field who challenge the core validity of the idea of T(reg) cells as central immune regulators. However, we note that a consensus may be emerging from the data: that multiple T(reg) cell functions act either directly or indirectly at the site of antigen presentation to create a regulatory milieu that promotes bystander suppression and infectious tolerance. Thus, the versatility and adaptability of the Foxp3+ T(reg) cells may in fact be the best argument that these cells are 'multitalented masters of immune regulation'.
Collapse
Affiliation(s)
- Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | | |
Collapse
|
71
|
Laske C, Zank M, Klein R, Stransky E, Batra A, Buchkremer G, Schott K. Autoantibody reactivity in serum of patients with major depression, schizophrenia and healthy controls. Psychiatry Res 2008; 158:83-6. [PMID: 18096244 DOI: 10.1016/j.psychres.2006.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2004] [Revised: 01/16/2006] [Accepted: 04/24/2006] [Indexed: 10/22/2022]
Abstract
The present study assessed 25 patients with unipolar major depression and 34 patients with schizophrenia along with 50 healthy, non-psychiatric controls for the presence of serum antinuclear (ANA), smooth muscle (SMA), anti-endothelial (AEA), anti-sarcolemma (ASA), thyroid gland (TGA) and parietal cell (PCA) antibodies. In the group of patients with major depression, the frequency of elevated ANA, TGA and PCA was significantly higher than in the control group. In addition, the group of patients with schizophrenia significantly more often showed increased levels of ANA and SMA than the control group of healthy volunteers. When the two psychiatric groups were compared, PCA serum titers in major depression and SMA values in schizophrenia were significantly more frequently elevated, whereas values of AEA and ASA showed no difference. These results point towards the existence of an unspecific (auto) immune disposition or reaction in at least a subgroup of patients with major depression and schizophrenia.
Collapse
Affiliation(s)
- Christoph Laske
- Department of Psychiatry and Psychotherapy, University of Tübingen, Osianderstr. 24, Tübingen D-72076, Germany.
| | | | | | | | | | | | | |
Collapse
|
72
|
Kashyap M, Thornton AM, Norton SK, Barnstein B, Macey M, Brenzovich J, Shevach E, Leonard WJ, Ryan JJ. Cutting Edge: CD4 T Cell-Mast Cell Interactions Alter IgE Receptor Expression and Signaling. THE JOURNAL OF IMMUNOLOGY 2008; 180:2039-43. [DOI: 10.4049/jimmunol.180.4.2039] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
73
|
van Driel IR, Ang DKY, Konz RF, Senices M, Young DA, Grusby MJ, Collins M, Dunussi-Joannopoulos K. Role of regulatory T cells in gastrointestinal inflammatory disease. J Gastroenterol Hepatol 2008; 23:171-7. [PMID: 18289352 DOI: 10.1111/j.1440-1746.2007.05278.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Regulatory T cells curb unwanted immune responses and regulate responses to microflora and it is now clear that regulatory T cells play an important role in a number of chronic inflammatory diseases of the gut. First, regulatory T cells are crucial in controlling immune responses to gastric autoantigens and thus preventing autoimmune gastritis and pernicious anemia. Second, regulatory T cells may modulate the response to Helicobacter pylori, thus affecting the ability of the immune system to clear the pathogen and mediate damage to the gastric mucosa. Finally, regulatory T cells play an important role in preventing damaging inflammatory responses to commensal organisms in the lower gut, thus guarding against inflammatory bowel diseases. In the present review, we examine the actions of regulatory T cells in the gut and conclude that further understanding of regulatory T cell biology may lead to new therapeutic approaches to chronic gastrointestinal disease.
Collapse
Affiliation(s)
- Ian R van Driel
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Hall BM, Robinson CM, Plain KM, Verma ND, Carter N, Boyd RA, Tran GT, Hodgkinson SJ. Studies on naïve CD4+CD25+T cells inhibition of naïve CD4+CD25−T cells in mixed lymphocyte cultures. Transpl Immunol 2008; 18:291-301. [DOI: 10.1016/j.trim.2007.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Revised: 08/24/2007] [Accepted: 09/12/2007] [Indexed: 11/28/2022]
|
75
|
Kristensen NN, Olsen J, Gad M, Claesson MH. Genome-wide expression profiling during protection from colitis by regulatory T cells. Inflamm Bowel Dis 2008; 14:75-87. [PMID: 17924563 DOI: 10.1002/ibd.20277] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND In the adoptive transfer model of colitis it has been shown that regulatory T cells (Treg) can hinder disease development and cure already existing mild colitis. The mechanisms underlying this regulatory effect of CD4(+)CD25(+) Tregs are not well understood. METHODS To identify pathways of importance for immune regulation in protected mice we studied the genome-wide expression profile in the inflamed rectum of SCID mice with CD4(+) T cell transfer colitis and in the uninflamed rectum of mice protected from colitis by Treg cells. We used DNA microarray technology (Affymetrix GeneChip Mouse Genome 430 2.0 Array), which enabled an analysis of a complete set of RNA transcript levels in each sample. Array results were confirmed by real-time reverse-transcriptase polymerase chain reaction (RT-PCR). RESULTS Data were analyzed using combined projections to latent structures and functional annotation analysis. The colitic samples were clearly distinguishable from samples from normal mice by a vast number of inflammation- and growth factor-related transcripts. In contrast, the Treg-protected animals could not be distinguished from either the normal BALB/c mice or the normal SCID mice. mRNA expression profiles of cytokine, chemokine, and growth factor genes were significantly altered in colitic as opposed to noncolitic mice. In particular, the transcription factors STAT3, GATA2, and NFkappaB, the cytokine IL1beta, and the chemokine receptors CXCR3 and CCR1 as well as their ligands all seemingly play central roles in the inflammatory processes. CONCLUSIONS We suggest that these molecules alone or in combination could be future therapeutic targets.
Collapse
Affiliation(s)
- Nanna Ny Kristensen
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Panum Institute, Denmark.
| | | | | | | |
Collapse
|
76
|
Abstract
In humans, limited T-cell receptor repertoire and lymphopenia are associated with severe eosinophilic inflammatory disease. A model of lymphopenia and reduced T-cell repertoire was created; C57BL/6 Rag2-/- mice received limited (30,000) or large (2 million) numbers of CD4 T-cells. Three to five months post-transfer, mice that had received 30,000 T-cells, but not those that received 2 million, developed fulminant macrophage pneumonia with eosinophilia, Ym1 deposition. methacholine-induced airway hyperresponsiveness, eosinophilic gastritis and esophagitis. These mice had strikingly elevated serum IgE (in CD3epsilon-/- hosts) and donor-cells were enriched for IL-4, IL-5 and IL-13 producers. Th2 pathology and serum IgE were enhanced when transferred populations were depleted of CD25+ CD4 Tregs, but was more severe when the effector population was derived from limited as compared to the large effector population. Pretreatment of Rag2-/- mice with 300,000 CD25+ CD4 Tregs prior to effector cell transfer prevented disease while pretreatment with 30,000 did not, despite the fact that there were equal numbers of Tregs in the hosts at the time of transfer of effector cells. Limited repertoire complexity of Tregs may lead to a failure to control immunopathologic responses and limited repertoire complexity of conventional cells may be responsible for the Th2 phenotype.
Collapse
Affiliation(s)
- Joshua Milner
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jerrold Ward
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Andrea Keane-Myers
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195
| | - William E. Paul
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
77
|
Bergin PJ, Raghavan S, Svensson H, Starckx S, Van Aelst I, Gjertsson I, Opdenakker G, Quiding-Järbrink M. Gastric gelatinase B/matrix metalloproteinase-9 is rapidly increased in Helicobacter felis-induced gastritis. ACTA ACUST UNITED AC 2007; 52:88-98. [PMID: 17995959 DOI: 10.1111/j.1574-695x.2007.00349.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It has previously been shown that matrix metalloproteinase-9 (MMP-9) levels, originating from macrophages, are considerably increased in human Helicobacter pylori-associated gastritis. Here, the early kinetics of the MMP-9 response resulting from Helicobacter infection in C57BL/6 and MMP-9 knock-out mice using the murine Helicobacter felis model were examined. H. felis infection induced severe gastritis in the murine stomach at just 2 weeks after infection. Before gastritis, an increase was observed in MMP-9-positive cells detected by immunohistochemistry in the basal lamina propria. This finding was corroborated by gelatin zymography of stomach samples. As the gastritis increased so did the concentration of MMP-9 and the incidence of gastric MMP-9-positive cells, their location corresponding to that of macrophages. In contrast, systemic levels of MMP-9 remained unchanged. When MMP-9-deficient mice were infected with H. felis, no significant difference in gastritis development was detected compared with disease development in wild-type animals. We conclude that MMP-9 production is an early event in the response to gastric Helicobacter infection, a feature that may favor the recruitment of immune cells early during infection. At later stages, however, the increased levels of MMP-9 may damage the integrity of the stomach mucosa.
Collapse
Affiliation(s)
- Philip J Bergin
- Department of Microbiology and Immunology, Göteborg University, Göteborg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
78
|
|
79
|
Keever-Taylor CA, Browning MB, Johnson BD, Truitt RL, Bredeson CN, Behn B, Tsao A. Rapamycin enriches for CD4(+) CD25(+) CD27(+) Foxp3(+) regulatory T cells in ex vivo-expanded CD25-enriched products from healthy donors and patients with multiple sclerosis. Cytotherapy 2007; 9:144-57. [PMID: 17453966 DOI: 10.1080/14653240601145223] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND CD4(+) CD25(bright+) regulatory T cells (Treg) can be expanded to clinical doses using CD3/CD28 Ab-coated beads plus IL-2. However, this method requires high purity of the starting population to prevent overgrowth by non-regulatory T cells. Rapamycin, an agent that inhibits T-cell proliferation but selectively spares Treg, may be a means to expand Treg from less pure CD25-enriched cells. METHODS CD25-enriched cells were prepared by a single-step immunomagnetic-selection using anti-CD25 microbeads. The cells were activated with a single addition of anti-CD3/CD28 beads and expanded in ex vivo 15-5% HS and autologous CD4(+) CD25(-) feeder cells,+/-rapamycin (0.01-20 ng/mL). IL-2 was added on day 3. Cells were rested for 2 days in ex vivo 15-5% HS and tested for phenotype, intracellular Foxp3 protein and suppressor activity. RESULTS In the absence of rapamycin, CD25-enriched fractions expanded >17 000-fold by 21 days. Although suppressor activity was detected to day 14, it declined significantly by 21 days as non-regulatory cells expanded. The addition of rapamycin inhibited expansion of non-regulatory T cells at doses > or =1 ng/mL while increasing suppressor activity and the percentage of CD4(+) CD25(+) CD27(+) Foxp3(+) cells. Rapamycin did not enrich for Foxp3(+) cells in expanded cultures of CD4(+) CD25(-) cells. Treg were also readily expanded in cultures of CD25-enriched cells obtained from patients with multiple sclerosis in the presence of rapamycin. DISCUSSION The addition of 1-20 ng/mL rapamycin to CD25-enriched cultures increased the purity of cells with the phenotype and function of Treg. This approach may alleviate the need for rigorous enrichment of Treg prior to activation and expansion for potential clinical use.
Collapse
Affiliation(s)
- C A Keever-Taylor
- Department of Medicine/Division of Neoplastic Diseases and Related Disorders, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | | | | | | | |
Collapse
|
80
|
Sun X, Yamada H, Yoshihara K, Awaya A, Yoshikai Y. In vivo treatment with a nonapeptide thymic hormone, facteur thymique serique (FTS), ameliorates chronic colitis induced by dextran sulphate sodium in mice. Int Immunopharmacol 2007; 7:928-36. [PMID: 17499195 DOI: 10.1016/j.intimp.2007.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2006] [Revised: 02/26/2007] [Accepted: 02/27/2007] [Indexed: 01/12/2023]
Abstract
Facteur thymique serique (FTS), a thymic hormone with nonapeptide is involved in T cell differentiation in intestine. Here we investigated the effect of FTS on dextran sulphate sodium (DSS)-induced colitis. BALB/c mice were subcutaneously treated with 1 mug/mouse/day of FTS daily. FTS did not affect the course of acute colitis induced by DSS as assessed by survival rate, clinical activity of diseases, extent of tissue damage of colons. On the other hand, FTS significantly ameliorated chronic colitis induced by multiple cycles of DSS as reflected by lower lethality, weight loss, clinical scores and histological scores. The levels of interferon (IFN)-gamma, interleukin 1(IL-1)-beta, and IL-12p40 in the culture supernatants of lamina propria (LP) cells of colon without any stimulation and IFN-gamma by T cells in the LP T cells under T cell receptor (TCR) triggering were reduced in FTS-treated mice, whereas the levels of IL-10 by LP cells and LPT cells were higher in FTS-treated mice. Thus, FTS may serve to suppress inflammation in DSS-induced chronic colitis accompanied by increased IL-10 production.
Collapse
Affiliation(s)
- Xun Sun
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | | | | | | | | |
Collapse
|
81
|
Lee CW, Rao VP, Rogers AB, Ge Z, Erdman SE, Whary MT, Fox JG. Wild-type and interleukin-10-deficient regulatory T cells reduce effector T-cell-mediated gastroduodenitis in Rag2-/- mice, but only wild-type regulatory T cells suppress Helicobacter pylori gastritis. Infect Immun 2007; 75:2699-707. [PMID: 17353283 PMCID: PMC1932875 DOI: 10.1128/iai.01788-06] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 12/14/2006] [Accepted: 02/22/2007] [Indexed: 12/25/2022] Open
Abstract
CD4(+) CD45RB(hi) CD25(-) effector T cells (T(E)) promote Helicobacter pylori gastritis in mice, and CD4(+) CD45RB(lo) CD25(+) regulatory T cells (T(R)) are anti-inflammatory. Using adoptive transfer into H. pylori-infected Rag2(-/-) mice, we evaluated effects of wild-type (wt) C57BL/6 or congenic interleukin-10-deficient (IL-10(-/-)) T(R) cells on gastritis, gastric cytokines, and H. pylori colonization. Infected Rag2(-/-) mice colonized in the corpus and antrum with 10(5) to 10(6) H. pylori CFU/gram without associated gastritis. T(E) cell transfer caused morbidity and an H. pylori-independent pangastritis and duodenitis (gastroduodenitis) associated with increased expression of gamma interferon (IFN-gamma) and tumor necrosis factor alpha. T(E) cell transfer to H. pylori-infected mice led to additive corpus gastritis associated with inflammatory cytokine expression and reduced colonization. wt T(R) cells reduced morbidity, H. pylori corpus gastritis, gastroduodenitis, and inflammatory cytokine expression and reversed the decline in H. pylori colonization attributable to T(E) cells. Although less effective than wt T(R) cells, IL-10(-/-) T(R) cells also reduced morbidity and gastroduodenitis but did not reduce H. pylori corpus gastritis or impact T(E) cell inhibition of colonization. Gastric tissues from mice receiving wt T(R) cells expressed higher levels of Foxp3 compared to recipients of IL-10(-/-) T(R) cells, consistent with lower regulatory activity of IL-10(-/-) T(R) cells. These results demonstrate that wt T(R) cells suppressed T(E)-cell-mediated H. pylori-independent gastroduodenitis and H. pylori-dependent corpus gastritis more effectively than IL-10(-/-) T(R) cells. Compartmental differences in T(E)-cell- and H. pylori-mediated inflammation and in regulatory effects between wt T(R) and IL-10(-/-) T(R) cells suggest that IL-10 expression by wt T(R) cells is important to regulatory suppression of gastric inflammation.
Collapse
Affiliation(s)
- Chung-Wei Lee
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
82
|
van Wijk F, Wehrens EJM, Nierkens S, Boon L, Kasran A, Pieters R, Knippels LMJ. CD4+CD25+T cells regulate the intensity of hypersensitivity responses to peanut, but are not decisive in the induction of oral sensitization. Clin Exp Allergy 2007; 37:572-81. [PMID: 17430355 DOI: 10.1111/j.1365-2222.2007.02681.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Naturally occurring CD4+CD25+ regulatory T cells (Tregs) play a critical role in the maintenance of self-tolerance and it has been suggested that these Tregs may also be involved in preventing allergic disease. OBJECTIVE The precise role of CD4+CD25+ T cells in the regulation of allergic responses to mucosal antigens remains to be elucidated. In the present study, it was investigated whether CD4+CD25+ T cells are involved in the induction of oral tolerance and whether they play a role in controlling hypersensitivity responses to food proteins. METHODS CD4+CD25+ T cells were depleted with PC61 mAb before the induction of low dose oral tolerance to peanut extract (PE). In addition, CD4+CD25+ T cell depletion was performed during sensitization or before oral challenge, using a C3H/HeOuJ mouse model of allergic sensitization to peanut. RESULTS Oral tolerance to PE could not be induced in CD4+CD25+ T cell-depleted mice. However, CD4+CD25+ T cell depletion during long-term exposure to PE alone did not result in allergic sensitization. In sensitized mice, anti-CD25 treatment during oral exposure resulted in higher levels of PE-specific IgE and increased mast cell degranulation upon an oral challenge. In contrast, anti-CD25 treatment of PE-sensitized mice before oral challenges did not affect the level of mast cell degranulation. CONCLUSION These results indicate that CD4+CD25+ Tregs are involved in maintaining tolerance to oral antigens and regulate the intensity of an IgE-mediated food hypersensitivity response, but are not crucial in preventing sensitization. Accordingly, CD4+CD25+ Tregs may represent a potential tool for the treatment of food allergic disorders.
Collapse
Affiliation(s)
- F van Wijk
- Wilhelmina Children's Hospital, Department of Pediatric Immunology, and Institute for Risk Assessment Sciences, Department of Immunotoxicology, Utrecht University, Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
83
|
Durakovic N, Radojcic V, Powell J, Luznik L. Rapamycin Promotes Emergence of IL-10-Secreting Donor Lymphocyte Infusion-Derived T Cells Without Compromising Their Graft-Versus-Leukemia Reactivity. Transplantation 2007; 83:631-40. [PMID: 17353785 DOI: 10.1097/01.tp.0000256158.84418.ff] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There are limited data examining the effects of pharmacological immunosuppression on the in vivo fate of donor lymphocyte infusions (DLI)-derived T cells, their function, and their antitumor efficacy. METHODS We addressed this question in a murine model in which DLI is given to stable mixed chimeras resulting in lymphohematopoietic graft-versus-host (LH-GVH) response. In this model, LH-GVH potency can be directly measured as the kinetics of conversion to full donor chimerism and can be correlated with associated graft-versus-leukemia (GVL) reactivity. RESULTS We found discordance in DLI-mediated LH-GVH reactivity depending on the timing of rapamycin (RAPA) administration. Delayed administration of RAPA in contrast to its early administration at the time of adoptive transfer did not interfere with conversion to full donor chimerism. Moreover, delayed administration of RAPA preserved the GVL reactivity of DLI. Analysis of the long-term chimeras showed that regardless of RAPA administration, adoptively transferred T cells mediating the LH-GVH response contribute minimally to the reconstitution of the peripheral T-cell compartment and exhibit profound hyporesponsiveness and decreased production of interleukin (IL)-2 on restimulation in vitro. However, we observed only in the RAPA-treated chimeras that the remaining hyporesponsive DLI-derived CD4+ T cells secrete large amounts of IL-10, a known immunoregulatory cytokine. CONCLUSIONS We conclude that delayed administration of RAPA after DLI does not interfere with their LH-GVH reactivity but promotes the emergence of IL-10-secreting DLI-derived CD4+ T cells that might contribute to the drug's known ability to promote bilateral donor host tolerance without interfering with GVL reactivity.
Collapse
Affiliation(s)
- Nadira Durakovic
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | | | | |
Collapse
|
84
|
Liu VC, Wong LY, Jang T, Shah AH, Park I, Yang X, Zhang Q, Lonning S, Teicher BA, Lee C. Tumor Evasion of the Immune System by Converting CD4+CD25− T Cells into CD4+CD25+ T Regulatory Cells: Role of Tumor-Derived TGF-β. THE JOURNAL OF IMMUNOLOGY 2007; 178:2883-92. [PMID: 17312132 DOI: 10.4049/jimmunol.178.5.2883] [Citation(s) in RCA: 354] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4+CD25+ T regulatory (T(reg)) cells were initially described for their ability to suppress autoimmune diseases in animal models. An emerging interest is the potential role of T(reg) cells in cancer development and progression because they have been shown to suppress antitumor immunity. In this study, CD4+CD25- T cells cultured in conditioned medium (CM) derived from tumor cells, RENCA or TRAMP-C2, possess similar characteristics as those of naturally occurring T(reg) cells, including expression of Foxp3, a crucial transcription factor of T(reg) cells, production of low levels of IL-2, high levels of IL-10 and TGF-beta, and the ability to suppress CD4+CD25- T cell proliferation. Further investigation revealed a critical role of tumor-derived TGF-beta in converting CD4+CD25- T cells into T(reg) cells because a neutralizing Ab against TGF-beta, 1D11, completely abrogated the induction of T(reg) cells. CM from a nontumorigenic cell line, NRP-152, or irradiated tumor cells did not convert CD4+CD25- T cells to T(reg) cells because they produce low levels of TGF-beta in CM. Finally, we observed a reduced tumor burden in animals receiving 1D11. The reduction in tumor burden correlated with a decrease in tumor-derived TGF-beta. Treatment of 1D11 also reduced the conversion of CD4+ T cells into T(reg) cells and subsequent T(reg) cell-mediated suppression of antitumor immunity. In summary, we have demonstrated that tumor cells directly convert CD4+CD25- T cells to T(reg) cells through production of high levels of TGF-beta, suggesting a possible mechanism through which tumor cells evade the immune system.
Collapse
Affiliation(s)
- Victoria C Liu
- Department of Urology, Northwestern University Medical School, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Joetham A, Takeda K, Takada K, Taube C, Miyahara N, Matsubara S, Matsubara S, Koya T, Rha YH, Dakhama A, Gelfand EW. Naturally occurring lung CD4(+)CD25(+) T cell regulation of airway allergic responses depends on IL-10 induction of TGF-beta. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:1433-42. [PMID: 17237391 DOI: 10.4049/jimmunol.178.3.1433] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peripheral tolerance to allergens is mediated in large part by the naturally occurring lung CD4(+)CD25(+) T cells, but their effects on allergen-induced airway responsiveness have not been well defined. Intratracheal, but not i.v., administration of naive lung CD4(+)CD25(+) T cells before allergen challenge of sensitized mice, similar to the administration of the combination of rIL-10 and rTGF-beta, resulted in reduced airway hyperresponsiveness (AHR) and inflammation, lower levels of Th2 cytokines, higher levels of IL-10 and TGF-beta, and less severe lung histopathology. Significantly, CD4(+)CD25(+) T cells isolated from IL-10(-/-) mice had no effect on AHR and inflammation, but when incubated with rIL-10 before transfer, suppressed AHR, and inflammation, and was associated with elevated levels of bronchoalveolar lavage TGF-beta levels. By analogy, anti-TGF-beta treatment reduced regulatory T cell activity. These data identify naturally occurring lung CD4(+)CD25(+) T cells as capable of regulating lung allergic responses in an IL-10- and TGF-beta-dependent manner.
Collapse
Affiliation(s)
- Anthony Joetham
- Department of Pediatrics, Division of Cell Biology, National Jewish Medical and Research Center, Denver, CO 80206, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Sukiennicki TL, Fowell DJ. Distinct molecular program imposed on CD4+ T cell targets by CD4+CD25+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2007; 177:6952-61. [PMID: 17082610 DOI: 10.4049/jimmunol.177.10.6952] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CD4+CD25+ regulatory T cells (Tregs) are key modulators of immunity, but their mechanism of action is unclear. To elucidate the molecular consequences of Treg encounter, we analyzed changes in gene expression in CD4+ T cell targets activated in the presence or absence of CD4+CD25+ Tregs. Tregs did not alter the early activation program of CD4+ T cells, but had reversed many of the activation-induced changes by 36 h. It is not known whether Tregs simply induce a set of transcriptional changes common to other nonproliferative states or whether instead Tregs mediate a distinct biological activity. Therefore, we compared the gene profile of T cells following Treg encounter with that of T cells made anergic, TGF-beta-treated, or IL-2-deprived; all possible modes of Treg action. Strikingly, all genes down-regulated in suppressed cells were indeed common to these nonproliferative states. In contrast, Treg encounter led to elevated expression of a unique set of genes in the target T cells. Although different from the nonproliferative states tested, the Treg-imposed gene program is exemplified by expression of many genes associated with growth arrest or inhibition of proliferation. We suggest that Tregs function by the induction of a distinct set of negative regulatory factors that initiate or maintain target T cells in a nonproliferative state.
Collapse
Affiliation(s)
- Teresa L Sukiennicki
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY 14642, USA
| | | |
Collapse
|
87
|
Milner JD, Ward JM, Keane-Myers A, Paul WE. Lymphopenic mice reconstituted with limited repertoire T cells develop severe, multiorgan, Th2-associated inflammatory disease. Proc Natl Acad Sci U S A 2007; 104:576-81. [PMID: 17202252 PMCID: PMC1761908 DOI: 10.1073/pnas.0610289104] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Lymphopenia and restricted T cell repertoires in humans are often associated with severe eosinophilic disease and a T cell Th2 bias. To examine the pathogenesis of this phenomenon, C57BL/6 Rag2-/- mice received limited (3 x 10(4)) or large (2 x 10(6)) numbers of CD4 T cells. Three to 5 months after transfer, mice that had received 3 x 10(4) T cells, but not those that received 2 x 10(6), developed fulminant macrophage pneumonia with eosinophilia, Ym1 deposition, and methacholine-induced airway hyperresponsiveness, as well as eosinophilic gastritis; esophagitis and other organ damage occurred in some cases. Donor cells were enriched for IL-4, IL-5, and IL-13 producers. When 3 x 10(4) cells were transferred into CD3epsilon-/- hosts, the mice developed strikingly elevated serum IgE. Prior transfer of 3 x 10(5) CD25+ CD4 T cells into Rag2-/- recipients prevented disease upon subsequent transfer of CD25- CD4 T cells, whereas 3 x 10(4) regulatory T cells (Tregs) did not, despite the fact that there were equal total numbers of Tregs in the host at the time of transfer of CD25- CD4 T cells. Limited repertoire complexity of Tregs may lead to a failure to control induction of immunopathologic responses, and limitation in repertoire complexity of conventional cells may be responsible for the Th2 phenotype.
Collapse
Affiliation(s)
| | - Jerrold M. Ward
- Infectious Disease Pathogenesis Section, Comparative Medicine Branch, and
| | - Andrea Keane-Myers
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - William E. Paul
- *Laboratory of Immunology
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
88
|
Abstract
Leishmania are digenetic protozoan parasites that are inoculated into the skin by vector sand flies, are taken up by macrophages, and produce a spectrum of chronic diseases in their natural reservoir and susceptible human hosts. During the early establishment of infection in the skin and lymphoid organs, Leishmania produce multiple effects on macrophage and dendritic cell functions that inhibit their innate anti-microbial defenses and impair their capacity to initiate T-helper 1 cell immunity. In addition, the skin is a site preconditioned for early parasite survival by virtue of a high frequency of steady-state, natural CD25+Foxp3+ regulatory T cells (Tregs) that function to suppress the generation of unneeded immune responses to infectious and non-infectious antigens to which the skin is regularly exposed. In murine models of infection, antigen-induced CD25+/-Foxp3-interleukin (IL)-10+ Treg cells act during the effector phase of the immune response to control immunopathology and may also delay or prevent healing. Finally, following resolution of infection in healed mice, CD25+Foxp3+ Tregs function in an IL-10-dependent manner to prevent sterile cure and establish a long-term state of functional immune privilege in the skin.
Collapse
Affiliation(s)
- Nathan Peters
- Laboratory of Parasitic Diseases, NIAID, Bethesda, MD 20892-0425, USA
| | | |
Collapse
|
89
|
Beaudreuil S, Samuel D, Rouas-Freiss N, Durrbach A. New aspect of immunosuppressive treatment in liver transplantation. How could you induce tolerance in liver transplantation? Transpl Immunol 2006; 17:98-107. [PMID: 17306740 DOI: 10.1016/j.trim.2006.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 11/10/2006] [Indexed: 12/13/2022]
Abstract
New immunosuppressive strategies have improved short- and long-term graft survival. The current aim is to decrease the intensity of the immunosuppressive regimen, in an attempt to limit side effects and the direct toxicity of calcineurin inhibitor (CNI) for kidney function. We describe here current experience in liver and liver-kidney transplantation, the mechanism of tolerance and the immunosuppressive strategy used in liver transplantation.
Collapse
Affiliation(s)
- Severine Beaudreuil
- Department of Nephrology, IFRNT University Hospital of Kremlin-Bicetre, 78 avenue du General Leclerc, 94275 Le Kremlin-Bicetre cedex, France
| | | | | | | |
Collapse
|
90
|
Zwar TD, van Driel IR, Gleeson PA. Guarding the immune system: Suppression of autoimmunity by CD4+CD25+immunoregulatory T cells. Immunol Cell Biol 2006; 84:487-501. [PMID: 16956386 DOI: 10.1111/j.1440-1711.2006.01471.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CD4+CD25+Foxp3+ T cells (CD25+ T regulatory [Treg] cells) are a naturally occurring suppressor T-cell population that regulates a wide variety of immune responses. A major function of CD25+ Treg cells is to inhibit the activity of self-reactive T cells that can potentially cause autoimmune disease. This review examines the recent advances in CD25+ Treg cell biology, with particular focus on the thymic and peripheral development of CD25+ Treg cells, the signals that promote their expansion and maintenance in the periphery and the mechanism by which they mediate their suppressor activity in peripheral lymphoid tissues. An understanding of these issues is likely to facilitate the development of CD25+ Treg-cell-based therapies for the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Tricia D Zwar
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
91
|
Abstract
Regulatory T cells (Tregs) are a specialized subpopulation of T cells that act to suppress activation of other immune cells and thereby maintain immune system homeostasis, self-tolerance as well as control excessive response to foreign antigens. The mere concept of Tregs was the subject of significant controversy among immunologists for many years owing to the paucity of reliable markers for defining these cells and the ambiguity of the nature and molecular basis of suppressive phenomena. However, recent advances in the molecular characterization of this cell population have firmly established their existence and their vital role in the vertebrate immune system. Of interest, accumulating evidence from both humans and experimental animal models has implicated the involvement of Tregs in the development of graft-versus-host disease (GVHD). The demonstration that Tregs could separate GVHD from graft-versus-tumor (GVT) activity suggests that their immunosuppressive potential could be manipulated to reduce GVHD without detrimental consequence on GVT effect. Although a variety of T lymphocytes with suppressive capabilities have been reported, the two best-characterized subsets are the naturally arising, intrathymic-generated Tregs (natural Tregs) and the peripherally generated, inducible Tregs (inducible Tregs). This review summarizes our current knowledge of the generation, function and regulation of these two populations of Tregs during an immune response. Their role in the development of GVHD and their therapeutic potential for the prevention and treatment of GVHD will also be described.
Collapse
Affiliation(s)
- N T Le
- Division of Cellular Therapy/Bone Marrow Transplantation, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
92
|
Abstract
Endowed with the ability to actively suppress an immune response, regulatory T cells (Tregs) hold the promise of halting ongoing pathogenic autoimmunity and restoring self-tolerance in patients suffering from autoimmune diseases. Through many in vitro and in vivo studies, we have learned that Tregs can function in the lymph nodes as well as in the peripheral tissues. In vivo, Tregs act through dendritic cells to limit autoreactive T-cell activation, thus preventing their differentiation and acquisition of effector functions. By limiting the supply of activated pathogenic cells, Tregs prevent or slow down the progression of autoimmune diseases. However, this protective mechanism appears insufficient in autoimmune individuals, likely because of a shortage of Tregs cells and/or the development and accumulation of Treg-resistant pathogenic T cells over the long disease course. Thus, restoration of self-tolerance in these patients will likely require purging of pathogenic T cells along with infusion of Tregs with increased ability to control ongoing tissue injury. In this review, we highlight advances in dissecting Treg function in vivo in autoimmune settings and summarize multiple studies that have overcome the limitations of the low abundance of Tregs and their hypoproliferative phenotype to develop Treg-based therapies.
Collapse
Affiliation(s)
- Qizhi Tang
- UCSF Diabetes Center, Department of Medicine and Department of Pathology, University of California, San Francisco, CA 94143-0540, USA
| | | |
Collapse
|
93
|
Izcue A, Coombes JL, Powrie F. Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunol Rev 2006; 212:256-71. [PMID: 16903919 DOI: 10.1111/j.0105-2896.2006.00423.x] [Citation(s) in RCA: 358] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) tract is the main interface where the body encounters exogenous antigens. It is crucial that the local response here is tightly regulated to avoid an immune reaction against dietary antigens and commensal flora while still mounting an efficient defense against pathogens. Faults in establishing intestinal tolerance can lead to disease, inducing local and often also systemic inflammation. Studies in human as well as in animal models suggest a role for regulatory T cells (Tregs) in maintaining intestinal homeostasis. Transfer of Tregs can not only prevent the development of colitis in animal models but also cure established disease, acting both systemically and at the site of inflammation. In this review, we discuss the major regulatory pathways, including transforming growth factor-beta (TGF-beta), interleukin-10 (IL-10), and cytotoxic T-lymphocyte antigen-4 (CTLA-4), and their role in Treg-mediated control of systemic and mucosal responses. In addition, we give an overview of the known mechanisms of lymphocyte migration to the intestine and discuss how CD103 expression can influence the balance between regulatory and effector T cells. Further understanding of the factors that control the activity of Tregs in different immune compartments may facilitate the design of strategies to target regulation in a tissue-specific way.
Collapse
Affiliation(s)
- Ana Izcue
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
94
|
Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. ACTA ACUST UNITED AC 2006; 203:2451-60. [PMID: 17015634 PMCID: PMC2118140 DOI: 10.1084/jem.20060956] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pathogen persistence after clinical cure is a hallmark of many chronic infections. Previously, we showed that naturally occurring CD4+CD25+ regulatory T (nTreg) cells rapidly accumulate within chronic dermal sites of Leishmania major infection where they suppress anti-pathogen CD4+ T cell responses, favor parasite persistence and dermal pathology, and consequently control concomitant immunity. Here, we postulated that chemokines might direct nTreg cell homing in sites of infection and show that CD4+CD25+ nTreg cells, compared with normal CD4+ T cells, preferentially express the CCR5 chemokine receptor, which enables them to migrate in response to CCR5 ligands in vitro. We show that in contrast to their wild-type (WT) counterparts, CCR5-/- CD4+CD25+ nTreg cells resulted in an increased magnitude of parasite-specific, interferon gamma-producing CD4+ T cells within infection sites, dramatically reduced parasite numbers, and potent resistance to infection, a finding consistent with the clinical outcome of infected CCR5-/- mice. Interestingly, this resistance was related to an inefficient migration of CCR5-/- nTreg cells to infected dermal sites compared with WT nTreg cells. Thus, this study shows that CCR5 directs the homing of CD4+CD25+ nTreg cells to L. major-infected dermal sites where they promote the establishment of infection and long-term survival of the parasite in the immune host.
Collapse
MESH Headings
- Animals
- Chemotaxis, Leukocyte/immunology
- Leishmania major/immunology
- Leishmania major/pathogenicity
- Leishmaniasis, Cutaneous/immunology
- Leishmaniasis, Cutaneous/parasitology
- Leishmaniasis, Cutaneous/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, CCR5/biosynthesis
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/physiology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Ekaterina Yurchenko
- Department of Microbiology and Immunology, McGill University, Montreal, H3A 2B4, Canada
| | | | | | | | | | | |
Collapse
|
95
|
Nguyen VH, Zeiser R, Negrin RS. Role of Naturally Arising Regulatory T Cells in Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2006; 12:995-1009. [PMID: 17084366 DOI: 10.1016/j.bbmt.2006.04.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 04/25/2006] [Indexed: 12/18/2022]
Abstract
Naturally arising CD4(+)CD25(+) regulatory T cells (Tregs) have the potential to suppress aberrant immune responses and to regulate peripheral T-cell homeostasis. In murine models of bone marrow transplantation, Tregs promote donor bone marrow engraftment and decrease the incidence and severity of graft-versus-host-disease without abrogating the beneficial graft-versus-tumor immunologic effect. These findings, in concert with observations that Tregs in mice and humans share phenotypic and functional characteristics, have led to active investigations into the use of these cells to decrease complications associated with human hematopoietic cell transplantation. Early human studies suggest that an imbalance of Tregs and effector T cells may contribute to the development of graft-versus-host-disease. However, the mechanisms of immunoregulation, in particular the allorecognition properties of Tregs, their effects on and interaction with other immune cells, and their sites of suppressive activity, are not well understood. In this review, we discuss the current knowledge of Treg biology and the potential therapeutic strategies and barriers of Treg immunotherapy in human hematopoietic cell transplantation.
Collapse
Affiliation(s)
- Vu H Nguyen
- Center for Clinical Science Research, Department of Medicine, Division of Bone Marrow Transplantation, Stanford University, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
96
|
Suri-Payer E, Fritzsching B. Regulatory T cells in experimental autoimmune disease. ACTA ACUST UNITED AC 2006; 28:3-16. [PMID: 16838180 DOI: 10.1007/s00281-006-0021-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 05/17/2006] [Indexed: 02/07/2023]
Abstract
During the past 10 years, CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) have been extensively studied for their function in autoimmune disease. This review summarizes the evidence for a role of Treg in suppression of innate and adaptive immune responses in experimental models of autoimmunity including arthritis, colitis, diabetes, autoimmune encephalomyelitis, lupus, gastritis, oophoritis, prostatitis, and thyroiditis. Antigen-specific activation of Treg, but antigen-independent suppressive function, emerges as a common paradigm derived from several disease models. Treg suppress conventional T cells (Tcon) by direct cell contact in vitro. However, downmodulation of dendritic cell function and secretion of inhibitory cytokines such as IL-10 and TGF-beta might underlie Treg function in vivo. The final outcome of autoimmunity vs tolerance depends on the balance between stimulatory signals (Toll-like receptor engagement, costimulation, and antigen dose) and inhibitory signals from Treg. Whereas most experimental settings analyze the capacity of Treg to prevent onset of autoimmune disease, more recent efforts indicate successful treatment of ongoing disease. Thus, Treg are on the verge of moving from experimental animal models into clinical applications in humans.
Collapse
Affiliation(s)
- Elisabeth Suri-Payer
- Division of Immunogenetics, D030, German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | | |
Collapse
|
97
|
Kristensen NN, Brudzewsky D, Gad M, Claesson MH. Chemokines involved in protection from colitis by CD4+CD25+ regulatory T cells. Inflamm Bowel Dis 2006; 12:612-8. [PMID: 16804399 DOI: 10.1097/01.ibd.0000225342.44850.d5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Chemokines are small proteins involved in the direction of migration of immune cells both during normal homeostasis and inflammation. Chemokines have been implicated in the pathology of many different inflammatory disorders and are therefore appealing therapeutic targets. Using a chemokine/chemokine receptor-specific gene expression profiling system of 67 genes, the authors have determined the expression profile of chemokine and chemokine receptor genes in the rectum of colitic mice and in mice that have been protected fromcolitis by CD4CD25 regulatory T cells. In mice protected from colitis, the authors found down regulation of the mRNA expression of the inflammatory chemokine receptors CCR1 and CXCR3 and their ligands CXCL9, CXCL10, CCL5, and CCL7. Also the transcripts for CCR9, CCL25, CCL17, and CXCL1 are found down regulated in protected compared with colitic animals. In addition, the authors' results suggest that CCL20 is used by CCR6 regulatory T cells in the complex process of controlling colitis because transcripts for this chemokine were expressed to a higher level in protected animals. The chemokine pathways identified in the present study may be of importance for the development of new targets for anti-inflammatory treatment strategies in human inflammatory bowel disease.
Collapse
Affiliation(s)
- Nanna Ny Kristensen
- Department of Medical Anatomy, University of Copenhagen, The Panum Institute, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
98
|
Holm TL, Lundsgaard D, Markholst H. Characteristics of Rat CD4+CD25+ T Cells and Their Ability to Prevent Not Only Diabetes But Also Insulitis in an Adoptive Transfer Model in BB Rats. Scand J Immunol 2006; 64:17-29. [PMID: 16784487 DOI: 10.1111/j.1365-3083.2006.01760.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human and mouse CD4(+)CD25(+) T cells have been intensively studied through the last decade. However, little is known about this subset in other species. This study describes the phenotype of rat CD4(+)CD25(+) Foxp3(+) T cells and the site in which they exert regulation in a transfer-induced autoimmune diabetes model. Several proteins and mRNAs are up-regulated in unstimulated rat CD4(+)CD25(+) T cells compared with CD4(+)CD25(-) T cells, including Foxp3, Lag-3, CD80, interleukin 10 (IL-10) and CTLA-4. To investigate CD4(+)CD25(+) T cells in vivo, we transferred three million diabetogenic T cells either alone or in combination with two million CD4(+)CD25(+) T cells to 30-day-old BB rats. The pancreas and the pancreatic lymph nodes were examined as two potential regulatory sites. Time-course analysis of pancreatic histology following diabetogenic T-cell transfers revealed insulitis from about 14 days after transfer. By contrast, rats receiving both diabetogenic T cells and CD4(+)CD25(+) T cells had no insulitis at any time. Moreover, the frequency of diabetogenic T cells in the pancreatic lymph nodes 2 days after transfer was significantly reduced in rats receiving both subsets. These data indicate that the primary site of T-cell regulation is in the draining lymph nodes and not the pancreas in our model.
Collapse
Affiliation(s)
- T L Holm
- Hagedorn Research Institute, Gentofte, Denmark
| | | | | |
Collapse
|
99
|
Ocklenburg F, Moharregh-Khiabani D, Geffers R, Janke V, Pfoertner S, Garritsen H, Groebe L, Klempnauer J, Dittmar KEJ, Weiss S, Buer J, Probst-Kepper M. UBD, a downstream element of FOXP3, allows the identification of LGALS3, a new marker of human regulatory T cells. J Transl Med 2006; 86:724-37. [PMID: 16702978 DOI: 10.1038/labinvest.3700432] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Here, we report the identification of the ubiquitin-like gene UBD as a downstream element of FOXP3 in human activated regulatory CD4(+)CD25(hi) T cells (T(reg)). Retroviral transduction of UBD in human allo-reactive effector CD4(+) T helper (T(h)) cells upregulates CD25 and mediates downregulation of IL4 and IL5 expression similar to overexpression of FOXP3. Moreover, UBD impairs T(h) cell proliferation without upregulation of FOXP3 and impairs calcium mobilization. In the presence of ionomycin, overexpression of UBD in T(h) cells leads to the induction of IL1R2 that resemble FOXP3-transduced T(h) cells and naturally derived T(reg) cells. A comparison of the transcriptome of FOXP3- and UBD-transduced T(h) cells with T(reg) cells allowed the identification of the gene LGALS3. However, high levels of LGALS3 protein expression were observed only in human CD4(+)CD25(hi) derived T(reg) cells and FOXP3-transduced T(h) cells, whereas little was induced in UBD-transduced T(h) cells. Thus, UBD contributes to the anergic phenotype of human regulatory T cells and acts downstream in FOXP3 induced regulatory signaling pathways, including regulation of LGALS3 expression. High levels of LGALS3 expression represent a FOXP3-signature of human antigen-stimulated CD4(+)CD25(hi) derived regulatory T cells.
Collapse
Affiliation(s)
- Frank Ocklenburg
- Junior Research Group for Xenotransplantation, Department of Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Células T, em particular as células T CD4+, têm sido associadas a muitos aspectos das doenças de pele. A evidência atual sugere, porém, que o papel dos linfócitos T CD4+ no desenvolvimento de inflamação cutânea excede o de ativador pró-inflamatório das células T de ação que dirigem a resposta imune. Subtipos de células T com capacidade reguladora, tais como Tregs CD4+CD25+high, têm sido identificadas. Observações recentes sugerem que em algumas doenças da pele a função dessas células está modificada. Portanto, o desenvolvimento e a função de Tregs na dermatologia são atualmente um tópico atraente devido a sua importância no controle da resposta do sistema imune contra tumores e doenças infecciosas, bem como inibindo o desenvolvimento de auto-imunidade e alergia. Assim, mecanismos reguladores defeituosos podem permitir a quebra da tolerância imune periférica seguida por inflamação crônica e doença. Detalham-se as anormalidades funcionais e a contribuição de diferentes subtipos de células T reguladoras no desenvolvimento de doenças dermatológicas nesta revisão. Acentuam-se os possíveis alvos terapêuticos e as modificações dos T reguladores causados por imunomoduladores usados no campo da dermatologia.
Collapse
|