851
|
Yamaguchi T, Wing JB, Sakaguchi S. Two modes of immune suppression by Foxp3(+) regulatory T cells under inflammatory or non-inflammatory conditions. Semin Immunol 2011; 23:424-30. [PMID: 22055883 DOI: 10.1016/j.smim.2011.10.002] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 10/11/2011] [Indexed: 10/15/2022]
Abstract
Foxp3-expressing regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance and homeostasis. One of the key issues for understanding Treg immunobiology is to determine how they suppress excessive or aberrant immune responses. Although a number of molecules have been reported to contribute to Treg suppressive function, the importance and precise role of each molecule is not clear. In this review, we propose and discuss that two modes of suppression can be distinguished. In the physiological and steady state, activation of naïve T cells can be suppressed by natural Tregs via deprivation of activation signals including CD28 signal and IL-2 from antigen-reactive T cells, keeping the latter in a naïve state in lymphoid tissues. These deprivation mechanisms are transiently abrogated in inflammatory conditions, allowing T cells to respond to antigen. In contrast, in highly inflammatory environments, for example, in microbial infection, activated Tregs acquire the capacity to kill or inactivate effector T cells and antigen-presenting cells, for example, via granzyme/perforin formation and IL-10 secretion, thereby actively damping excessive immune responses. Understanding these processes will help effectively controlling physiological and pathological immune responses via Tregs.
Collapse
Affiliation(s)
- Tomoyuki Yamaguchi
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | | | | |
Collapse
|
852
|
Ferraro A, Socci C, Stabilini A, Valle A, Monti P, Piemonti L, Nano R, Olek S, Maffi P, Scavini M, Secchi A, Staudacher C, Bonifacio E, Battaglia M. Expansion of Th17 cells and functional defects in T regulatory cells are key features of the pancreatic lymph nodes in patients with type 1 diabetes. Diabetes 2011; 60:2903-13. [PMID: 21896932 PMCID: PMC3198077 DOI: 10.2337/db11-0090] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Autoimmune diseases, including type 1 diabetes, are thought to have a Th17-cell bias and/or a T-regulatory cell (Treg) defect. Understanding whether this is a hallmark of patients with type 1 diabetes is a crucial question that is still unsolved, largely due to the difficulties of accessing tissues targeted by the disease. RESEARCH DESIGN AND METHODS We phenotypically and functionally characterized Th17 cells and Tregs residing in the pancreatic-draining lymph nodes (PLNs) of 19 patients with type 1 diabetes and 63 nondiabetic donors and those circulating in the peripheral blood of 14 type 1 diabetic patients and 11 healthy subjects. RESULTS We found upregulation of Th17 immunity and functional defects in CD4(+)CD25(bright) Tregs in the PLNs of type 1 diabetic subjects but not in their peripheral blood. In addition, the proinsulin-specific Treg-mediated control was altered in the PLNs of diabetic patients. The dysfunctional Tregs isolated from diabetic subjects did not contain contaminant effector T cells and were all epigenetically imprinted to be suppressive, as defined by analysis of the Treg-specific demethylated region within the forkhead box P3 (FOXP3) locus. CONCLUSIONS These data provide evidence for an unbalanced immune status in the PLNs of type 1 diabetic subjects, and treatments restoring the immune homeostasis in the target organ of these patients represent a potential therapeutic strategy.
Collapse
Affiliation(s)
- Alessandra Ferraro
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Socci
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Angela Stabilini
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Valle
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Monti
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Lorenzo Piemonti
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Rita Nano
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | | | - Paola Maffi
- Department of Transplantation Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Secchi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Transplantation Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Staudacher
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Manuela Battaglia
- Diabetes Research Institute, San Raffaele Scientific Institute, Milan, Italy
- Corresponding author: Manuela Battaglia,
| |
Collapse
|
853
|
Mortha A, Diefenbach A. Natural killer cell receptor-expressing innate lymphocytes: more than just NK cells. Cell Mol Life Sci 2011; 68:3541-55. [PMID: 21904914 PMCID: PMC11114688 DOI: 10.1007/s00018-011-0803-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 08/08/2011] [Accepted: 08/08/2011] [Indexed: 12/17/2022]
Abstract
Recently, additional subsets that extend the family of innate lymphocytes have been discovered. Among these newly identified innate lymphoid cells is a subset sharing phenotypic characteristics of natural killer cells and lymphoid tissue inducer cells. These cells co-express the transcription factor RORγt and activating NK cell receptors (NKR), but their lineage and functional qualities remain poorly defined. Here, we discuss recent proposals to place these NKR(+)RORγt(+) innate lymphocytes on hematopoietic lineage maps. An overview of the transcriptional circuitry determining fate decisions of innate lymphocytes and a summary of current concepts concerning plasticity and stability of innate lymphocyte effector fates are provided. We will conclude by discussing the function of RORγt-expressing innate lymphocytes during inflammatory bowel diseases and in the immune response to tumors.
Collapse
Affiliation(s)
- Arthur Mortha
- IMMH, Institute of Medical Microbiology and Hygiene, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, 79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, 79104 Freiburg, Germany
| | - Andreas Diefenbach
- IMMH, Institute of Medical Microbiology and Hygiene, University of Freiburg, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, 79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, 79104 Freiburg, Germany
| |
Collapse
|
854
|
Long SA, Buckner JH. CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. THE JOURNAL OF IMMUNOLOGY 2011; 187:2061-6. [PMID: 21856944 DOI: 10.4049/jimmunol.1003224] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Treg) play a dominant role in suppression of autoimmune pathology, as rescue of Treg number and/or function in model systems can both prevent and reverse disease. These findings have generated a series of studies addressing the role of defects in Treg number and function in human autoimmunity. However, demonstrating global defects in Treg of individuals diagnosed with autoimmune diseases has been challenging. These challenges are founded, in part, in the complexity of human autoimmune diseases in which various genetic factors and environmental triggers contribute to disease susceptibility. Moreover, contribution of failed Treg-mediated suppression to pathogenesis can extend to multiple mechanisms. In this article, we discuss what is known with respect to the number and function of CD4(+)FOXP3(+) Treg in human autoimmunity, focusing on representative autoimmunediseases in which there are diverse Treg-mediated defects. We also highlight the need to better understand Treg plasticity and function in the context of autoimmunity.
Collapse
Affiliation(s)
- S Alice Long
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | |
Collapse
|
855
|
Alterations of peripheral CD4+CD25+Foxp3+ T regulatory cells in mice with STZ-induced diabetes. Cell Mol Immunol 2011; 9:75-85. [PMID: 21983870 DOI: 10.1038/cmi.2011.37] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Complications arising from abnormal immune responses are the major causes of mortality and morbidity in diabetic patients. CD4+CD25+T regulatory cells (Tregs) play pivotal roles in controlling immune homeostasis, immunity and tolerance. The effect of hyperglycemia on CD4+CD25+Tregs has not yet been addressed. Here we used streptozotocin (STZ)-induced diabetic mice to study the effects of long-term hyperglycemia on CD4+CD25+Tregs in vivo. Four months after the onset of diabetes, the frequency of CD4+CD25+Foxp3+ T regulatory cells was significantly elevated in the spleen, peripheral blood lymphocytes (PBLs), peripheral lymph nodes (pLNs) and mesenteric LNs (mLNs). CD4+CD25+Tregs obtained from mice with diabetes displayed defective immunosuppressive functions and an activated/memory phenotype. Insulin administration rescued these changes in the CD4+CD25+ Tregs of diabetic mice. The percentage of thymic CD4+CD25+ naturally occurring Tregs (nTregs) and peripheral CD4+Helios+Foxp3+ nTregs were markedly enhanced in diabetic mice, indicating that thymic output contributed to the increased frequency of peripheral CD4+CD25+Tregs in diabetic mice. In an in vitro assay in which Tregs were induced from CD4+CD25- T cells by transforming growth factor (TGF)-β, high glucose enhanced the efficiency of CD4+CD25+Foxp3+ inducible Tregs (iTregs) induction. In addition, CD4+CD25- T cells from diabetic mice were more susceptible to CD4+CD25+Foxp3+ iTreg differentiation than those cells from control mice. These data, together with the enhanced frequency of CD4+Helios-Foxp3+ iTregs in the periphery of mice with diabetes, indicate that enhanced CD4+CD25+Foxp3+ iTreg induction also contributes to a peripheral increase iCD4+CD25+Tregs in diabetic mice. Our data show that hyperglycemia may alter the frequency of CD4+CD25+Foxp3+ Tregs in mice, which may result in late-state immune dysfunction in patients with diabetes.
Collapse
|
856
|
Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, Hall JA, Yagi R, Naik S, Bhairavabhotla R, Paul WE, Bosselut R, Wei G, Zhao K, Oukka M, Zhu J, Belkaid Y. GATA3 controls Foxp3⁺ regulatory T cell fate during inflammation in mice. J Clin Invest 2011; 121:4503-15. [PMID: 21965331 DOI: 10.1172/jci57456] [Citation(s) in RCA: 431] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/24/2011] [Indexed: 12/17/2022] Open
Abstract
Tregs not only keep immune responses to autoantigens in check, but also restrain those directed toward pathogens and the commensal microbiota. Control of peripheral immune homeostasis by Tregs relies on their capacity to accumulate at inflamed sites and appropriately adapt to their local environment. To date, the factors involved in the control of these aspects of Treg physiology remain poorly understood. Here, we show that the canonical Th2 transcription factor GATA3 is selectively expressed in Tregs residing in barrier sites including the gastrointestinal tract and the skin. GATA3 expression in both murine and human Tregs was induced upon TCR and IL-2 stimulation. Although GATA3 was not required to sustain Treg homeostasis and function at steady state, GATA3 played a cardinal role in Treg physiology during inflammation. Indeed, the intrinsic expression of GATA3 by Tregs was required for their ability to accumulate at inflamed sites and to maintain high levels of Foxp3 expression in various polarized or inflammatory settings. Furthermore, our data indicate that GATA3 limits Treg polarization toward an effector T cell phenotype and acquisition of effector cytokines in inflamed tissues. Overall, our work reveals what we believe to be a new facet in the complex role of GATA3 in T cells and highlights what may be a fundamental role in controlling Treg physiology during inflammation.
Collapse
Affiliation(s)
- Elizabeth A Wohlfert
- Mucosal Immunology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
857
|
Culina S, Mallone R. Pathogenic and regulatory T cells in type 1 diabetes: losing self-control, restoring it, and how to take the temperature. Curr Diab Rep 2011; 11:426-33. [PMID: 21732231 DOI: 10.1007/s11892-011-0209-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The central role of T cells in type 1 diabetes pathogenesis is well established, but these cells continue to pose numerous challenges in understanding their dynamics and in following their modifications. Important progress has been recently made in pinpointing some novel antigens targeted by pathogenic T cells and the epitope sequences recognized. Studies on the interplay between effector T cells, their regulatory counterparts, and cells of the innate immune system have unraveled novel pathways and may inspire new therapeutic approaches. At the same time, the appreciation of the plasticity of regulatory T cells has raised important caveats on their use for cell-based therapies. Continuous development of T-cell assays exploring both pathogenic and regulatory players will be critical to "take the temperature" of undergoing disease progression and reversal.
Collapse
Affiliation(s)
- Slobodan Culina
- INSERM U986, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 75674, Paris, Cedex 14, France.
| | | |
Collapse
|
858
|
Fan H, Cao P, Game DS, Dazzi F, Liu Z, Jiang S. Regulatory T cell therapy for the induction of clinical organ transplantation tolerance. Semin Immunol 2011; 23:453-61. [PMID: 21920772 DOI: 10.1016/j.smim.2011.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The pursuit of transplantation tolerance is the holygrail in clinical organ transplantation. It has been established that regulatory T cells (Tregs) can confer donor-specific tolerance in mouse models of transplantation. However, this is crucially dependent on the strain combination, the organ transplanted and most importantly, the ratio of Tregs to alloreactive effector T cells. The ex vivo expansion of Tregs is one solution to increase the number of alloantigen specific cells capable of suppressing the alloresponse. Indeed, ex vivo expanded, alloantigen specific murine Tregs are shown to preferentially migrate to, and proliferate in, the graft and draining lymph node. In human transplantation it has been proposed that depletion of the majority of direct pathway alloreactive T cells will be required to tip the balance in favour of regulation. Ex vivo expansion of alloantigen specific, indirect pathway human Tregs, which can cross regulate the residual direct pathway has been established. Rapid expansion of these cells is possible, whilst they retain antigen specificity, suppressive properties and favourable homing markers. Furthermore, considerable progress has been made to define which immunosuppressive drugs favour the expansion and function of Tregs. Currently a series of clinical trials of adoptive Treg therapy in combination with depletion of alloreactive T cells and short term immunosuppression are underway for human transplantation with the aim of minimizing immunosuppressive drugs and completely withdrawal.
Collapse
Affiliation(s)
- Huimin Fan
- Shanghai East Hospital of Tongji University, Shanghai 200120, China
| | | | | | | | | | | |
Collapse
|
859
|
Liu Y, Zhu T, Cai G, Qin Y, Wang W, Tang G, Zhao D, Shen Q. Elevated circulating CD4+ ICOS+ Foxp3+ T cells contribute to overproduction of IL-10 and are correlated with disease severity in patients with systemic lupus erythematosus. Lupus 2011; 20:620-7. [PMID: 21558138 DOI: 10.1177/0961203310392431] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this work, we aimed to investigate the frequency, possible categories and clinical significance of circulating CD4+ ICOS+ FoxP3+ T cells in patients with systemic lupus erythematosus (SLE). The frequency of circulating CD4+ ICOS+ FoxP3+ T cells was analysed by flow-cytometric analysis in 32 SLE patients, 10 rheumatoid arthritis patients and 32 healthy controls. Production of IL-10 and mTGF-β by different CD4+ T-cell populations was determined by intracellular cytokine staining. Plasma levels of IL-10 and TGF-β were determined by enzyme-linked immunosorbent assay (ELISA). The frequency of circulating CD4+ ICOS+ FoxP3+ T cells was significantly increased in SLE patients as compared with control groups. The elevated frequency of CD4+ ICOS+ FoxP3+ T cells had a positive correlation with SLE Disease Activity Index (SLEDAI) scores and serum anti-dsDNA but a negative correlation with serum C3. Additionally, the CD4+ ICOS+ Foxp3+ T cells contained significantly higher percentages of IL-10-producing cells than CD4+ ICOS- Foxp3+ T cells. A significant positive correlation was also observed between the frequency of CD4+ ICOS+ Foxp3+ T cells and the plasma level of IL-10 in SLE patients. In conclusion, an increased frequency of circulating CD4+ ICOS+ Foxp3+ T cells was observed in patients with SLE, suggesting its potential importance in the immunopathogenesis of SLE. Analysis of the CD4+ ICOS+ FoxP3+ T-cell population may be useful for the evaluation of lupus disease severity.
Collapse
Affiliation(s)
- Y Liu
- Department of Laboratory Diagnosis, Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
860
|
B cell-derived IL-10 suppresses inflammatory disease in Lyn-deficient mice. Proc Natl Acad Sci U S A 2011; 108:E823-32. [PMID: 21911371 DOI: 10.1073/pnas.1107913108] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lyn kinase deficient mice represent a well established genetic model of autoimmune/autoinflammatory disease that resembles systemic lupus erythematosus. We report that IL-10 plays a crucial immunosuppressive role in this model, modulating the inflammatory component of the disease caused by myeloid and T-cell activation. Double-mutant lyn(-/-)IL-10(-/-) mice manifested severe splenomegaly and lymphadenopathy, dramatically increased proinflammatory cytokine production, and severe tissue inflammation. Single-mutant lyn(-/-)mice showed expansion of IL-10-producing B cells. Interestingly, WT B cells adoptively transferred into lyn(-/-) mice showed increased differentiation into IL-10-producing B cells that assumed a similar phenotype to endogenous lyn(-/-) IL-10-producing B cells, suggesting that the inflammatory environment present in lyn(-/-) mice induces IL-10-producing B-cell differentiation. B cells, but not T or myeloid cells, were the critical source of IL-10 able to reduce inflammation and autoimmunity in double mutant lyn(-/-)IL-10(-/-) mice. IL-10 secretion by B cells was also crucial to sustain transcription factor Forkhead Box P3 (Foxp3) expression in regulatory T cells during disease development. These data reveal a dominant immunosuppressive function of B-cell-derived IL-10 in the Lyn-deficient model of autoimmunity, extending our current understanding of the role of IL-10 and IL-10-producing B cells in systemic lupus erythematosus.
Collapse
|
861
|
Takahashi R, Nishimoto S, Muto G, Sekiya T, Tamiya T, Kimura A, Morita R, Asakawa M, Chinen T, Yoshimura A. SOCS1 is essential for regulatory T cell functions by preventing loss of Foxp3 expression as well as IFN-{gamma} and IL-17A production. ACTA ACUST UNITED AC 2011; 208:2055-67. [PMID: 21893603 PMCID: PMC3182063 DOI: 10.1084/jem.20110428] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
SOCS1 is required to restrict IFN-γ and IL-17 expression and maintain Foxp3 expression in and function of regulatory T cells. Regulatory T cells (Treg cells) maintain immune homeostasis by limiting inflammatory responses. SOCS1 (suppressor of cytokine signaling 1), a negative regulator of cytokine signaling, is necessary for the suppressor functions of Treg cells in vivo, yet detailed mechanisms remain to be clarified. We found that Socs1−/− Treg cells produced high levels of IFN-γ and rapidly lost Foxp3 when transferred into Rag2−/− mice or cultured in vitro, even though the CNS2 (conserved noncoding DNA sequence 2) in the Foxp3 enhancer region was fully demethylated. Socs1−/− Treg cells showed hyperactivation of STAT1 and STAT3. Because Foxp3 expression was stable and STAT1 activation was at normal levels in Ifnγ−/−Socs1−/− Treg cells, the restriction of IFN-γ–STAT1 signaling by SOCS1 is suggested to be necessary for stable Foxp3 expression. However, Ifnγ−/−Socs1−/− Treg cells had hyperactivated STAT3 and higher IL-17A (IL-17) production compared with Ifnγ−/−Socs1+/+ Treg cells and could not suppress colitis induced by naive T cells in Rag2−/− mice. In vitro experiments suggested that cytokines produced by Socs1−/− Treg cells and Ifnγ−/−Socs1−/− Treg cells modulated antigen-presenting cells for preferential Th1 and Th17 induction, respectively. We propose that SOCS1 plays important roles in Treg cell integrity and function by maintaining Foxp3 expression and by suppressing IFN-γ and IL-17 production driven by STAT1 and STAT3, respectively.
Collapse
Affiliation(s)
- Reiko Takahashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjyuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
862
|
Araya N, Sato T, Yagishita N, Ando H, Utsunomiya A, Jacobson S, Yamano Y. Human T-lymphotropic virus type 1 (HTLV-1) and regulatory T cells in HTLV-1-associated neuroinflammatory disease. Viruses 2011; 3:1532-48. [PMID: 21994794 PMCID: PMC3187691 DOI: 10.3390/v3091532] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 08/13/2011] [Accepted: 08/16/2011] [Indexed: 01/12/2023] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is a retrovirus that is the causative agent of adult T cell leukemia/lymphoma (ATL) and associated with multiorgan inflammatory disorders, including HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and uveitis. HTLV-1-infected T cells have been hypothesized to contribute to the development of these disorders, although the precise mechanisms are not well understood. HTLV-1 primarily infects CD4(+) T helper (Th) cells that play a central role in adaptive immune responses. Based on their functions, patterns of cytokine secretion, and expression of specific transcription factors and chemokine receptors, Th cells that are differentiated from naïve CD4(+) T cells are classified into four major lineages: Th1, Th2, Th17, and T regulatory (Treg) cells. The CD4(+)CD25(+)CCR4(+) T cell population, which consists primarily of suppressive T cell subsets, such as the Treg and Th2 subsets in healthy individuals, is the predominant viral reservoir of HTLV-1 in both ATL and HAM/TSP patients. Interestingly, CD4(+)CD25(+)CCR4(+) T cells become Th1-like cells in HAM/TSP patients, as evidenced by their overproduction of IFN-γ, suggesting that HTLV-1 may intracellularly induce T cell plasticity from Treg to IFN-γ(+) T cells. This review examines the recent research into the association between HTLV-1 and Treg cells that has greatly enhanced understanding of the pathogenic mechanisms underlying immune dysregulation in HTLV-1-associated neuroinflammatory disease.
Collapse
Affiliation(s)
- Natsumi Araya
- Department of Rare Diseases Research, Institute of Medical Science, School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan; E-Mails: (N.A.); (T.S.); (N.Y.); (H.A.)
| | - Tomoo Sato
- Department of Rare Diseases Research, Institute of Medical Science, School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan; E-Mails: (N.A.); (T.S.); (N.Y.); (H.A.)
| | - Naoko Yagishita
- Department of Rare Diseases Research, Institute of Medical Science, School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan; E-Mails: (N.A.); (T.S.); (N.Y.); (H.A.)
| | - Hitoshi Ando
- Department of Rare Diseases Research, Institute of Medical Science, School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan; E-Mails: (N.A.); (T.S.); (N.Y.); (H.A.)
| | - Atae Utsunomiya
- Department of Hematology, Imamura Bun-in Hospital, Kagoshima 890-0064, Japan; E-Mail:
| | - Steven Jacobson
- Viral Immunology Section, Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; E-Mail:
| | - Yoshihisa Yamano
- Department of Rare Diseases Research, Institute of Medical Science, School of Medicine, St. Marianna University, Kawasaki 216-8511, Japan; E-Mails: (N.A.); (T.S.); (N.Y.); (H.A.)
| |
Collapse
|
863
|
Abstract
The microenviroment of acute myelogenous leukemia (AML) is suppressive for immune effector cells. Regulatory T cells (Tregs) have been recognized as a contributor factor and may be recruited and exploited by leukemic cells to evade immunesurveillance. Studies have shown that the frequencies of marrow and blood Tregs are greater in patients with AML than in control patients. Although increased Tregs have been associated with a decreased risk of GVHD after allogeneic HCT and hence may impede the graft-versus-tumor effect, recent findings indicate that that this may not be the case. Because there is a need to improve outcomes of standard treatment (chemotherapy with or without allogeneic HCT) in AML, targeting Tregs present an outstanding opportunity in AML because discoveries may apply throughout its treatment. Here, we review data on the roles of Tregs in mediating immune system-AML interactions. We focused on in vitro, animal, and observational human studies of Tregs in AML biology, development, prognosis, and therapy in different settings (eg, vaccination and HCT). Manipulation of Tregs or other types of immunomodulation may become a part of AML treatment in the future.
Collapse
|
864
|
Beyer M, Thabet Y, Müller RU, Sadlon T, Classen S, Lahl K, Basu S, Zhou X, Bailey-Bucktrout SL, Krebs W, Schönfeld EA, Böttcher J, Golovina T, Mayer CT, Hofmann A, Sommer D, Debey-Pascher S, Endl E, Limmer A, Hippen KL, Blazar BR, Balderas R, Quast T, Waha A, Mayer G, Famulok M, Knolle PA, Wickenhauser C, Kolanus W, Schermer B, Bluestone JA, Barry SC, Sparwasser T, Riley JL, Schultze JL. Repression of the genome organizer SATB1 in regulatory T cells is required for suppressive function and inhibition of effector differentiation. Nat Immunol 2011; 12:898-907. [PMID: 21841785 PMCID: PMC3669688 DOI: 10.1038/ni.2084] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 07/07/2011] [Indexed: 12/12/2022]
Abstract
Regulatory T cells (T(reg) cells) are essential for self-tolerance and immune homeostasis. Lack of effector T cell (T(eff) cell) function and gain of suppressive activity by T(reg) cells are dependent on the transcriptional program induced by Foxp3. Here we report that repression of SATB1, a genome organizer that regulates chromatin structure and gene expression, was crucial for the phenotype and function of T(reg) cells. Foxp3, acting as a transcriptional repressor, directly suppressed the SATB1 locus and indirectly suppressed it through the induction of microRNAs that bound the SATB1 3' untranslated region. Release of SATB1 from the control of Foxp3 in T(reg) cells caused loss of suppressive function, establishment of transcriptional T(eff) cell programs and induction of T(eff) cell cytokines. Our data support the proposal that inhibition of SATB1-mediated modulation of global chromatin remodeling is pivotal for maintaining T(reg) cell functionality.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- 3' Untranslated Regions/immunology
- Animals
- Cell Differentiation/drug effects
- Chromatin Assembly and Disassembly/drug effects
- Chromatin Assembly and Disassembly/immunology
- Flow Cytometry
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Gene Expression Profiling
- Gene Expression Regulation
- Genome, Human
- Genome-Wide Association Study
- Humans
- Lentivirus
- Lymphocyte Activation/drug effects
- Matrix Attachment Region Binding Proteins/genetics
- Matrix Attachment Region Binding Proteins/immunology
- Matrix Attachment Region Binding Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/immunology
- MicroRNAs/metabolism
- MicroRNAs/pharmacology
- RNA Interference
- RNA, Small Interfering/immunology
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Self Tolerance/drug effects
- Self Tolerance/genetics
- Self Tolerance/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- Marc Beyer
- Life and Medical Sciences Institute, Laboratory for Genomics and Immunoregulation, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
865
|
Regateiro FS, Howie D, Cobbold SP, Waldmann H. TGF-β in transplantation tolerance. Curr Opin Immunol 2011; 23:660-9. [PMID: 21839624 DOI: 10.1016/j.coi.2011.07.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 07/05/2011] [Indexed: 12/16/2022]
Abstract
TGF-β is a cytokine required for the induction and maintenance of transplantation tolerance in animal models. TGF-β mediates anti-inflammatory effects by acting on many immune cell-types. Central for transplantation tolerance is the role for TGF-β in the induction of Foxp3 and regulatory capacity in CD4(+) T cells. Recently, however, the general anti-inflammatory role of TGF-β in CD4(+) T cell polarization was questioned by the discovery that, in the presence of inflammatory cytokines such as IL-6 or IL-1, TGF-β drives the differentiation of Th17 cells associated with transplant rejection. A better understanding of the factors determining TGF-β production and activation, Foxp3 induction and Treg stability is vital for the development of tolerogenic strategies in transplantation.
Collapse
Affiliation(s)
- Frederico S Regateiro
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | | | | | |
Collapse
|
866
|
Battaglia M, Roncarolo MG. Immune intervention with T regulatory cells: past lessons and future perspectives for type 1 diabetes. Semin Immunol 2011; 23:182-94. [PMID: 21831659 DOI: 10.1016/j.smim.2011.07.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 07/10/2011] [Indexed: 01/11/2023]
Abstract
In type 1 diabetes (T1D), insulin-producing pancreatic β-cells are attacked and destroyed by the immune system. Although man-made insulin is life-saving, it is not a cure and it cannot prevent long-term complications. In addition, most T1D patients would do almost anything to achieve release from the burden of daily glucose monitoring and insulin injection. Despite the formation of very large and promising clinical trials, a means to prevent/cure T1D in humans remains elusive. This has led to an increasing interest in the possibility of using T cells with regulatory properties (Treg cells) as a biological therapy to preserve and restore tolerance to self-antigens. In the present review we will attempt to consolidate learning from the past and to describe what we now believe could in the future become a successful Treg-cell based immune intervention in T1D.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, via Olgettina 58, 20132 Milan, Italy.
| | | |
Collapse
|
867
|
Abstract
Regulatory T (Treg) cells play central role in regulation of immune responses to self-antigens, allergens, and commensal microbiota as well as immune responses to infectious agents and tumors. Transcriptional factor Foxp3 serves as a lineage specification factor of Treg cells. Paucity of Treg cells due to loss-of-function mutations of the Foxp3 gene is responsible for highly aggressive, fatal, systemic immune-mediated inflammatory lesions in mice and humans. Recent studies of Foxp3 expression and function provided critical novel insights into biology of Treg cells and into cellular mechanisms of the immune homeostasis.
Collapse
Affiliation(s)
- Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
868
|
Mellor AL, Munn DH. Physiologic control of the functional status of Foxp3+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:4535-40. [PMID: 21464094 DOI: 10.4049/jimmunol.1002937] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Foxp3-lineage CD4 regulatory T cells (Tregs) were named for their ability to maintain self tolerance and suppress T cell immunity. However, resting Tregs from noninflamed tissues exhibit little suppressor activity, and must be stimulated to acquire such function. Conversely, under certain inflammatory conditions, Tregs may undergo rapid reprogramming to acquire helper/effector functions. In this Brief Review, we describe recent progress in elucidating physiologic processes that control the functional status of Foxp3-lineage Tregs. Emerging evidence suggests the surprising possibility that reprogrammed Tregs can be an indispensable source of helper activity in some physiologic settings, such as priming CD8(+) T cell responses. This suggests a novel paradigm in which Foxp3(+) Tregs intrinsically possess bifunctional potential, acting as a preformed pool of first-responder cells at sites of local inflammation that can either provide classical regulatory/suppressor activity, or rapidly reprogram to supply helper/effector activity, contingent on signals that manifest in local physiologic settings.
Collapse
Affiliation(s)
- Andrew L Mellor
- Department of Medicine, Immunotherapy and Cancer Centers, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
869
|
McMurchy AN, Bushell A, Levings MK, Wood KJ. Moving to tolerance: clinical application of T regulatory cells. Semin Immunol 2011; 23:304-13. [PMID: 21620722 PMCID: PMC3836227 DOI: 10.1016/j.smim.2011.04.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 04/18/2011] [Indexed: 12/13/2022]
Abstract
Decreasing the incidence of chronic rejection and reducing the need for life-long immunosuppression remain important goals in clinical transplantation. In this article, we will review how regulatory T cells (Treg) came to be recognized as an attractive way to prevent or treat allograft rejection, the ways in which Treg can be manipulated or expanded in vivo, and the potential of in vitro expanded/generated Treg for cellular therapy. We will describe the first regulatory T cell therapies that have been or are in the process of being conducted in the clinic as well as the safety concerns of such therapies and how outcomes may be measured.
Collapse
|
870
|
|
871
|
Edinger M, Hoffmann P. Regulatory T cells in stem cell transplantation: strategies and first clinical experiences. Curr Opin Immunol 2011; 23:679-84. [PMID: 21802270 DOI: 10.1016/j.coi.2011.06.006] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 06/30/2011] [Indexed: 01/30/2023]
Abstract
The adoptive transfer of donor-type CD4(+)CD25(+)FOXP3(+) regulatory T cells (Treg) protects from graft-versus-host disease in murine bone marrow transplantation models. Results from first clinical trials exploring such strategies have recently been presented and seem to confirm the efficacy of Treg for the prevention of this severe complication after allogeneic stem cell transplantation. Further improvements in Treg isolation and in vitro expansion technologies will facilitate the broader exploration of Treg therapies, for example, for the treatment of ongoing graft-versus-host disease or the prevention of graft rejection after solid organ transplantation.
Collapse
Affiliation(s)
- Matthias Edinger
- Department of Hematology and Oncology, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany.
| | | |
Collapse
|
872
|
Abstract
Lineage-committed effector CD4(+) T cells are generated at the peak of the primary response and are followed by heterogeneous populations of central and effector memory cells. Here we review the evidence that T helper type 1 (T(H)1) effector cells survive the contraction phase of the primary response and become effector memory cells. We discuss the applicability of this idea to the T(H)2 cell, T(H)17 helper T cell, follicular helper T cell (T(FH) cell) and induced regulatory T cell lineages. We also discuss how central memory cells are formed, with an emphasis on the role of B cells in this process.
Collapse
|
873
|
Li CR, Deiro MF, Godebu E, Bradley LM. IL-7 uniquely maintains FoxP3(+) adaptive Treg cells that reverse diabetes in NOD mice via integrin-β7-dependent localization. J Autoimmun 2011; 37:217-27. [PMID: 21745722 DOI: 10.1016/j.jaut.2011.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 05/04/2011] [Accepted: 06/06/2011] [Indexed: 11/29/2022]
Abstract
Type 1 diabetes (T1D) develops as a consequence of a progressive autoimmune response that destroys insulin-producing β-cells in pancreatic islets. Because of their role(s) in controlling immune responses, considerable effort has been directed toward resolving whether regulatory T cells (Tregs) offer a clinical treatment to restore tolerance in T1D. We previously reported that in vitro-induced adaptive Treg cells (aTregs) can reverse T1D and persist as protective memory cells in the NOD mouse model. In the current study, we investigated mechanisms that regulate aTregs. We found that these FoxP3(+) aTregs expressed high levels of the IL-7 receptor, IL-7Rα, without the high affinity receptor for IL-2, CD25, which is found on natural Treg cells (nTregs). IL-7Rα expression was mirrored by the dependency of aTregs on IL-7 for persistence. IL-10 and TGF-β, effector cytokines of aTregs, were not essential for their maintenance at the level of systemic antibody blocking. Nevertheless, IL-10 modulated cytokine production by aTregs and TGF-β was critical for protection. aTregs were found to infiltrate islets and the expression of integrin-β7 was required for their localization in the pancreas. Furthermore, blocking aTreg entry into the pancreas prevented their control of diabetogenic effector T cells, implying the need for local control of the autoimmune response. The distinct homeostatic regulation of aTregs independently of a response to IL-2, which is defective in T1D patients, suggests that these cells represent a translatable candidate to control the autoimmune response.
Collapse
Affiliation(s)
- Cheng-Rui Li
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
874
|
Nguyen TLM, Sullivan NL, Ebel M, Teague RM, DiPaolo RJ. Antigen-specific TGF-β-induced regulatory T cells secrete chemokines, regulate T cell trafficking, and suppress ongoing autoimmunity. THE JOURNAL OF IMMUNOLOGY 2011; 187:1745-53. [PMID: 21746962 DOI: 10.4049/jimmunol.1004112] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ability to regulate ongoing inflammation using regulatory T cells (Tregs) is under intense investigation. Strategies to induce and expand Ag-specific Tregs are being developed, and whether various types of Tregs are suppressive in the inflammatory conditions associated with ongoing disease needs to be determined. In this study, we report that TGF-β-induced Tregs (iTregs) and expanded Tregs specific for a major self-Ag in autoimmune gastritis suppress inflammation and associated pathology when administered late in the process of ongoing disease. Transferred iTregs localized to the stomach, maintained Foxp3 and suppressor functions, and engaged several distinct mechanisms to alleviate disease progression. In addition to suppressing the production of inflammatory cytokines in the stomach and preventing the destruction of parietal cells, we show that iTregs secrete numerous chemokines and regulate both iTreg and effector T cell trafficking into the stomach. These data support efforts to use iTregs in therapies to treat autoimmunity and inflammatory diseases and provide novel insight into the biological mechanisms of iTreg-mediated immune suppression.
Collapse
Affiliation(s)
- Thanh-Long M Nguyen
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | | | | | | | | |
Collapse
|
875
|
Weiss EM, Schmidt A, Vobis D, Garbi N, Lahl K, Mayer CT, Sparwasser T, Ludwig A, Suri-Payer E, Oberle N, Krammer PH. Foxp3-Mediated Suppression of CD95L Expression Confers Resistance to Activation-Induced Cell Death in Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:1684-91. [DOI: 10.4049/jimmunol.1002321] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
876
|
Penaranda C, Tang Q, Bluestone JA. Anti-CD3 therapy promotes tolerance by selectively depleting pathogenic cells while preserving regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:2015-22. [PMID: 21742976 DOI: 10.4049/jimmunol.1100713] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Monoclonal anti-CD3 Abs have been used clinically for two decades to reverse steroid-resistant acute graft rejection. In autoimmune diabetes, short course treatment with FcR-nonbinding (FNB) anti-CD3 mAb in mice with recent onset of diabetes induces long-term disease remission. Induction of tolerogenic regulatory T cells (Tregs) has been implicated to be one of the mechanisms of action by FNB anti-CD3 mAb in these settings. In this study, we examined the effect of FNB anti-CD3 mAb treatment on the homeostasis of naive, effector, and regulatory T cells in vivo. Anti-CD3 treatment induced a transient systemic rise in the percentage but not absolute number of CD4(+)Foxp3(+) Tregs due to selective depletion of CD4(+)Foxp3(-) conventional T cells. T cell depletion induced by FNB anti-CD3 mAb was independent of the proapoptotic proteins Fas, caspase-3, and Bim and was not inhibited by overexpression of the anti-apoptotic protein, Bcl-2. Tregs were not preferentially expanded and we found no evidence of conversion of conventional T cells into Tregs, suggesting that the pre-existing Tregs are resistant to anti-CD3-induced cell death. Interestingly, expression of the transcription factor Helios, which is expressed by thymus-derived natural Tregs, was increased in Tregs after FNB anti-CD3 mAb treatment, suggesting that the anti-CD3 treatment can alter, and potentially stabilize, Treg function. Taken together, the results suggest that FNB anti-CD3 therapy promotes tolerance by restoring the balance between pathogenic and regulatory T cells.
Collapse
Affiliation(s)
- Cristina Penaranda
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
877
|
Hori S. Regulatory T cell plasticity: beyond the controversies. Trends Immunol 2011; 32:295-300. [DOI: 10.1016/j.it.2011.04.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/28/2011] [Accepted: 04/03/2011] [Indexed: 12/22/2022]
|
878
|
Teshima T, Maeda Y, Ozaki K. Regulatory T cells and IL-17-producing cells in graft-versus-host disease. Immunotherapy 2011; 3:833-52. [DOI: 10.2217/imt.11.51] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Graft-versus-host disease (GvHD), a major complication following allogeneic hematopoietic stem cell transplantation, is mediated by donor-derived T cells. On activation with alloantigens expressed on host antigen-presenting cells, naive CD4+ T cells differentiate into T-helper cell subsets of effector T cells expressing distinct sets of transcriptional factors and cytokines. Classically, acute GvHD was suggested to be predominantly related to Th1 responses. However, we now face a completely different and complex scenario involving possible roles of newly identified Th17 cells as well as Tregs in GvHD. Accumulating data from experimental and clinical studies suggest that the fine balance between Th1, Th2, Th17 and Tregs after transplantation may be an important determinant of the severity, manifestation and tissue distribution of GvHD. Understanding the dynamic process of reciprocal differentiation of regulatory and T-helper cell subsets as well as their interactions will be important in establishing novel strategies for preventing and treating GvHD.
Collapse
Affiliation(s)
- Takanori Teshima
- Center for Cellular & Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshinobu Maeda
- Biopathological Science, Okayama University Graduate School of Medicine & Dentistry, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Katsutoshi Ozaki
- Division of Hematology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329–0498, Japan
| |
Collapse
|
879
|
Bailey-Bucktrout SL, Bluestone JA. Regulatory T cells: stability revisited. Trends Immunol 2011; 32:301-6. [PMID: 21620768 PMCID: PMC3129467 DOI: 10.1016/j.it.2011.04.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 03/28/2011] [Accepted: 04/03/2011] [Indexed: 12/27/2022]
Abstract
Breakdown in self-tolerance is caused, in part, by loss of regulatory T (Treg) cells. Recently, a controversy has surfaced about whether Treg cells are overwhelmingly stable, or if they can be reprogrammed in inflammatory and autoimmune environments. Those in the instability camp have shown that a fraction of Treg cells lose forkhead box P3 protein and acquire effector arm activities. Instability is coupled with interleukin-2 insufficiency and the inflammatory milieu that promotes reprogramming. Here, we highlight the basic tenets of each viewpoint and discuss technical, biological and environmental differences in the models that might help yield a unifying hypothesis. Also considered is how Treg cell instability could link to development of autoimmune disease and the implications for trials of Treg cell-based therapy.
Collapse
Affiliation(s)
- Samantha L Bailey-Bucktrout
- Diabetes Center, University of California San Francisco, HSW 1102, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | | |
Collapse
|
880
|
Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B, Ziegelbauer J, Yassai M, Li SH, Relland LM, Wise PM, Chen A, Zheng YQ, Simpson PM, Gorski J, Salzman NH, Hessner MJ, Chatila TA, Williams CB. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity 2011; 35:109-22. [PMID: 21723159 DOI: 10.1016/j.immuni.2011.03.029] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 02/10/2011] [Accepted: 03/30/2011] [Indexed: 12/16/2022]
Abstract
Although both natural and induced regulatory T (nTreg and iTreg) cells can enforce tolerance, the mechanisms underlying their synergistic actions have not been established. We examined the functions of nTreg and iTreg cells by adoptive transfer immunotherapy of newborn Foxp3-deficient mice. As monotherapy, only nTreg cells prevented disease lethality, but did not suppress chronic inflammation and autoimmunity. Provision of Foxp3-sufficient conventional T cells with nTreg cells reconstituted the iTreg pool and established tolerance. In turn, acute depletion of iTreg cells in rescued mice resulted in weight loss and inflammation. Whereas the transcriptional signatures of nTreg and in vivo-derived iTreg cells were closely matched, there was minimal overlap in their T cell receptor (TCR) repertoires. Thus, iTreg cells are an essential nonredundant regulatory subset that supplements nTreg cells, in part by expanding TCR diversity within regulatory responses.
Collapse
Affiliation(s)
- Dipica Haribhai
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
881
|
Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, Jack RS, Wunderlich FT, Brüning JC, Müller W, Rudensky AY. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 2011; 34:566-78. [PMID: 21511185 DOI: 10.1016/j.immuni.2011.03.018] [Citation(s) in RCA: 699] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 02/26/2011] [Accepted: 03/16/2011] [Indexed: 12/11/2022]
Abstract
Effector CD4+ T cell subsets, whose differentiation is facilitated by distinct cytokine cues, amplify the corresponding type of inflammatory response. Regulatory T (Treg) cells integrate environmental cues to suppress particular types of inflammation. In this regard, STAT3, a transcription factor essential for T helper 17 (Th17) cell differentiation, is necessary for Treg cell-mediated control of Th17 cell responses. Here, we showed that anti-inflammatory interleukin-10 (IL-10), and not proinflammatory IL-6 and IL-23 cytokine signaling, endowed Treg cells with the ability to suppress pathogenic Th17 cell responses. Ablation of the IL-10 receptor in Treg cells resulted in selective dysregulation of Th17 cell responses and colitis similar to that observed in mice harboring STAT3-deficient Treg cells. Thus, Treg cells limit Th17 cell inflammation by serving as principal amplifiers of negative regulatory circuits operating in immune effector cells.
Collapse
Affiliation(s)
- Ashutosh Chaudhry
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
882
|
Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J Invest Dermatol 2011; 131:1853-60. [PMID: 21654831 DOI: 10.1038/jid.2011.139] [Citation(s) in RCA: 316] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Psoriasis is an autoimmune-related chronic inflammatory skin disease that is strongly associated with IL-23 and T helper-17 (Th17) effector cytokines. In addition, CD4+CD25(high) regulatory T-cell (Treg) function appeared to be impaired in psoriasis. CD4+CD25(high)Foxp3+ Tregs are typically considered inhibitors of autoimmune responses. However, under proinflammatory conditions, Tregs can differentiate into inflammation-associated Th17 cells--a paradigm shift, with as yet largely unknown consequences for human disease initiation or progression. Th17 cells are highly proinflammatory T cells that are characterized by IL-17A and IL-22 production and expression of the transcription factor retinoic acid-related orphan receptor γt (RORγt). We here show that Tregs of patients with severe psoriasis, as compared with those of healthy controls, have an enhanced propensity to differentiate into IL-17A-producing cells on ex vivo stimulation. This enhanced Treg differentiation was linked to unexpectedly high RORγt levels and enhanced loss of Foxp3. Notably, IL-23 boosted this Treg differentiation process particularly in patients with psoriasis but less so in controls. IL-23 further reduced Foxp3 expression while leaving the high RORγt levels unaffected. The histone/protein deacetylase inhibitor, Trichostatin-A, prevented Th17 differentiation of Tregs in psoriasis patients. Importantly, IL-17A+/Foxp3+/CD4+ triple-positive cells were present in skin lesions of patients with severe psoriasis. These data stress the clinical relevance of Treg differentiation for the perpetuation of chronic inflammatory disease and may pave novel ways for immunotherapy.
Collapse
|
883
|
Hippen K, Merkel S, Schirm D, Nelson C, Tennis N, Riley J, June C, Miller J, Wagner J, Blazar B. Generation and large-scale expansion of human inducible regulatory T cells that suppress graft-versus-host disease. Am J Transplant 2011; 11:1148-57. [PMID: 21564534 PMCID: PMC3552455 DOI: 10.1111/j.1600-6143.2011.03558.x] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Adoptive transfer of thymus-derived natural regulatory T cells (nTregs) effectively suppresses disease in murine models of autoimmunity and graft-versus-host disease (GVHD). TGFß induces Foxp3 expression and suppressive function in stimulated murine CD4+25- T cells, and these induced Treg (iTregs), like nTreg, suppress auto- and allo-reactivity in vivo. However, while TGFß induces Foxp3 expression in stimulated human T cells, the expanded cells lack suppressor cell function. Here we show that Rapamycin (Rapa) enhances TGFß-dependent Foxp3 expression and induces a potent suppressor function in naive (CD4+ 25-45RA+) T cells. Rapa/TGFß iTregs are anergic, express CD25 at levels higher than expanded nTregs and few cells secrete IL-2, IFNγ or IL-17 even after PMA and Ionomycin stimulation in vitro. Unlike other published methods of inducing Treg function, Rapa/TGFß induces suppressive function even in the presence of memory CD4+ T cells. A single apheresis unit of blood yields an average ~240 × 10⁹ (range ~ 70-560 × 10⁹) iTregs from CD4+25- T cells in ≤ 2 weeks of culture. Most importantly, Rapa/TGFß iTregs suppress disease in a xenogeneic model of GVHD. This study opens the door for iTreg cellular therapy for human diseases.
Collapse
Affiliation(s)
- K.L. Hippen
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| | - S.C. Merkel
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| | - D.K. Schirm
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| | - C. Nelson
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| | | | - J.L. Riley
- Abramson Family Cancer Center Research Institute, University of Pennsylvania Cancer Center, Philadelphia, PA 19104
| | - C.H. June
- Abramson Family Cancer Center Research Institute, University of Pennsylvania Cancer Center, Philadelphia, PA 19104
| | - J.S. Miller
- Molecular and Cellular Therapeutics Facility, University of Minnesota Cancer Center, Minneapolis, MN USA 55455,Department of Medicine, Division of Hematology/Oncology and Transplantation, Minneapolis, MN USA 55455
| | - J.E. Wagner
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455,Molecular and Cellular Therapeutics Facility, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| | - B.R. Blazar
- Department of Pediatrics, Division of Hematology/Oncology and Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN USA 55455
| |
Collapse
|
884
|
Feng T, Cao AT, Weaver CT, Elson CO, Cong Y. Interleukin-12 converts Foxp3+ regulatory T cells to interferon-γ-producing Foxp3+ T cells that inhibit colitis. Gastroenterology 2011; 140:2031-43. [PMID: 21419767 PMCID: PMC3109200 DOI: 10.1053/j.gastro.2011.03.009] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 02/10/2011] [Accepted: 03/04/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Regulatory T (Treg) cells are plastic, but the in vivo mechanisms by which they are converted into foxhead box p3 (Foxp3+) interferon (IFN)-γ+ T cells and whether these converted cells retain the ability to inhibit colitis are not clear. METHODS Foxp3+ Treg cells were generated by culture of naïve CD4+ T cells from Foxp3GFP CBir1 T-cell receptor (TCR) transgenic (Tg) (CBir1-Tg) mice, which are specific for CBir1 flagellin (an immunodominant microbiota antigen), with transforming growth factor-β. Foxp3GFP+ CBir1-Tg Treg cells were isolated by fluorescence-activated cell sorting and transferred into TCRβxδ-/- mice. Colitis was induced by transfer of naïve CBir1-Tg CD4+ T cells into immunodeficient mice. RESULTS Microbiota antigen-specific Foxp3+ Treg cells were converted, in the intestine, to IFN-γ+ T-helper (Th)1 cells, interleukin (IL)-17+ Th17 cells, and Foxp3+ T cells that coexpress IFN-γ and/or IL-17. Conversion of Treg cells into IFN-γ-producing Th1 cells and Foxp3+IFN-γ+ T cells required innate cell production of IL-12 in the intestine; blocking IL-12 with an antibody inhibited their conversion to Th1 and Foxp3+IFN-γ+ T cells in the intestines of mice that were recipients of Treg cells. Addition of IL-12, but not IL-23, promoted conversion of Treg cells into Th1 and Foxp3+IFN-γ+ T cells, in vitro. Foxp3+IFN-γ+ T cells had regulatory activity because they suppressed proliferation of naïve T cells, in vitro, and inhibited induction of colitis by microbiota antigen-specific T cells. IFN-γ+ Th1 cells were not converted into Treg cells; Foxp3+IFN-γ+ T cells differentiated into IFN-γ+ but not Foxp3+ T cells. CONCLUSIONS IL-12 promotes conversion of Treg cells into IFN-γ-expressing cells; Foxp3+IFN-γ+ T cells retain their regulatory functions and develop during the transition of Foxp3+ Treg cells into IFN-γ+ Th1 cells.
Collapse
Affiliation(s)
- Ting Feng
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Microbiology/Immunology, and Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Anthony T. Cao
- Department of Microbiology/Immunology, and Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Casey T. Weaver
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Charles O. Elson
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yingzi Cong
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294,Department of Microbiology/Immunology, and Pathology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
885
|
Saleh M, Elson CO. Experimental inflammatory bowel disease: insights into the host-microbiota dialog. Immunity 2011; 34:293-302. [PMID: 21435584 DOI: 10.1016/j.immuni.2011.03.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease appears to result from an abnormal host immune response to the intestinal microbiota. Experimental models have allowed the dissection of the complex dialog between the host and its microbiota. Through genetic manipulation of the host genome the immune compartments, cells, molecules, and genes that are critical for maintenance of intestinal homeostasis are being identified. Genetic association studies in humans have identified over 100 susceptibility loci. Although there is remarkable coherence between the experimental model and the human genetic data, a full understanding of the mechanisms involved in genetic susceptibility to IBD and of gene-gene and gene-environmental interactions will require a "next generation" of experimental models.
Collapse
Affiliation(s)
- Maya Saleh
- Department of Medicine, McGill University, Montreal, Quebec, Canada H3G 0B1.
| | | |
Collapse
|
886
|
Sharma R, Sung SSJ, Ju CYA, Deshmukh US, Fu SM, Ju ST. Regulatory T-Cell (Treg) hybridoma as a novel tool to study Foxp3 regulation and Treg fate. J Autoimmun 2011; 37:113-21. [PMID: 21621978 DOI: 10.1016/j.jaut.2011.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 05/02/2011] [Indexed: 11/19/2022]
Abstract
The CD25(+)Foxp3(+) regulatory T-cells (Treg) that had lost CD25 and Foxp3 in vivo (ex-Treg) exist but are difficult to study. We generated antigen (Ag)-specific Treg hybridomas from iTreg clones (iTreg-hyb) using iTreg of DO11.10.Foxp3-GFP mice and presented evidence that they behave like ex-Treg. The iTreg-hyb displayed little CD25 and Foxp3-GFP but strong expression could be induced with OVA(323-339) in the presence of Ag-presenting cells, rIL-2 and rTGF-β1. They displayed all of the iTreg-associated markers examined except CTLA-4, the latter was also absent in the ex-Treg. They lacked the Helios transcription factor, suggesting they were derived from iTreg. Similar to ex-Treg, the iTreg-hyb produced high level of IL-2 and Foxp3 under specific activation conditions. Two unusual properties were observed. First, the ability to induce Foxp3-GFP upon activation is progressively lost in culture over a period of 2-4 weeks. Second, Rag2(-/-) spleen cells alone selectively induced Foxp3-GFP expression albeit 30 times less efficient than Ag-specific activation. We identified cell-free supernatant, IL-6, IL-9, and IL-27 as Foxp3-inducing factors. Our study has significant implications to the stability, plasticity and fate of Treg. The usefulness and limitation of iTreg-hyb as a novel tool to study Foxp3 regulation and the fate of specific Treg subsets are discussed.
Collapse
Affiliation(s)
- Rahul Sharma
- Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908-0412, USA
| | | | | | | | | | | |
Collapse
|
887
|
Raymond M, Van VQ, Wakahara K, Rubio M, Sarfati M. Lung dendritic cells induce T(H)17 cells that produce T(H)2 cytokines, express GATA-3, and promote airway inflammation. J Allergy Clin Immunol 2011; 128:192-201.e6. [PMID: 21601259 DOI: 10.1016/j.jaci.2011.04.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 03/11/2011] [Accepted: 04/14/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dendritic cells (DCs) are crucial to shape the adaptive immune response. Extensive in vitro manipulation reprograms T(H)2 and T(H)17 cell lines into T(H)1 cells, leading to the concept of CD4(+) T(H) cell subset plasticity. The conversion of memory T(H)17 cells into T(H)2 cells or vice versa remains to be clarified. OBJECTIVE We examined the localization of T(H)17/T(H)2 cells in vivo, their cellular origin (T(H)2 vs T(H)17), and the underlying mechanisms that drive the generation of these double T(H) producers. METHODS Antigen-loaded bone marrow-derived DCs (ovalbumin-DCs) were repeatedly administered locally (intratracheally) or systemically (intravenously) to naive mice to elicit chronic airway inflammation. Inflamed lungs and mediastinal lymph nodes were examined for the presence of IL-17(+)IL-13(+)IL-4(+)CD4(+) T cells that coexpressed retinoic acid receptor-related orphan receptor γt and GATA-3 (T(H)17/T(H)2). RESULTS We show that repetitive administration of inflammatory ovalbumin-DCs, locally or systemically, promoted the development of antigen-specific T(H)17/T(H)2 cells in lungs and mediastinal lymph nodes. Immunized mice had IgE-independent and steroid-resistant airway inflammation with a mixed neutrophil and eosinophil infiltration of the bronchoalveolar lavage fluid. Airway inflammatory signal regulatory protein α-positive DCs reprogrammed in vitro-generated T(H)17 but not T(H)2 cells, as well as lung effector T(H) cells, into T(H)17/T(H)2 cells. CONCLUSION We demonstrate the existence of T(H)17/T(H)2 cells that express GATA-3 in inflamed tissues and their T(H)17 origin. We further propose that repeated immunization with inflammatory DCs prevails on the route of DC administration to drive T(H)17/T(H)2-associated chronic lung inflammation.
Collapse
Affiliation(s)
- Marianne Raymond
- Immunoregulation Laboratory, Centre Hospitalier de l'Université de Montréal, Research Center (CRCHUM), Notre-Dame Hospital, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
888
|
Genomic views of STAT function in CD4+ T helper cell differentiation. Nat Rev Immunol 2011; 11:239-50. [PMID: 21436836 DOI: 10.1038/nri2958] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Signal transducer and activator of transcription (STAT) proteins are well known for their essential roles in transmitting cytokine-mediated signals and specifying T helper (T(H)) cell differentiation. Recent technological advances have revealed that STAT proteins have broad and complex roles in gene regulation and epigenetic control, including important roles as functional repressors. However, the challenge of how to link signal transduction, nucleosome biology and gene regulation remains. The relevance of tackling this problem is highlighted by genome-wide association studies that link cytokine signalling and STATs to various autoimmune or immune deficiency disorders. Defining exactly how extrinsic signals control the specification and plasticity of T(H) cells will provide important insights and perhaps therapeutic opportunities in these diseases.
Collapse
|
889
|
Affiliation(s)
- Xuehao Wang
- Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China.
| | | | | |
Collapse
|
890
|
Miyara M, Gorochov G, Ehrenstein M, Musset L, Sakaguchi S, Amoura Z. Human FoxP3+ regulatory T cells in systemic autoimmune diseases. Autoimmun Rev 2011; 10:744-55. [PMID: 21621000 DOI: 10.1016/j.autrev.2011.05.004] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2011] [Indexed: 12/16/2022]
Abstract
Since the characterization of CD4(+)CD25(+) regulatory T (Treg) cells in mice, significant progress has been made in the definitions of the phenotype and the function of human Treg cells in health and in pathological conditions. Recent advances in the field leading to a better molecular definition of Treg subsets in humans and the description of the dynamics of differentiation of Treg cells should bring new insights in the understanding of human chronic systemic autoimmune diseases. How Treg cells are compromised in these diseases is a challenging issue because the elucidation of the mechanisms leading to such anomaly might lead to promising novel therapeutic approaches.
Collapse
Affiliation(s)
- Makoto Miyara
- Internal Medicine Department, French national Reference center for SLE and antiphospholipid syndrome AP-HP Hôpital Pitié-Salpêtrière, 75013 Paris, France.
| | | | | | | | | | | |
Collapse
|
891
|
d'Hennezel E, Yurchenko E, Sgouroudis E, Hay V, Piccirillo CA. Single-cell analysis of the human T regulatory population uncovers functional heterogeneity and instability within FOXP3+ cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:6788-97. [PMID: 21576508 DOI: 10.4049/jimmunol.1100269] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Natural FOXP3(+)CD4(+)CD25(High) regulatory T cells are critical in immunological self-tolerance. Their characterization in humans is hindered by the failure to discriminate these cells from activated effector T cells in inflammation. To explore the relationship between FOXP3 expression and regulatory function at the clonal level, we used a single-cell cloning strategy of CD25-expressing CD4(+) T cell subsets from healthy human donors. Our approach unveils a functional heterogeneity nested within CD4(+)CD25(High)FOXP3(+) T cells, and typically not revealed by conventional bulk assays. Whereas most cells display the canonical regulatory T (T(reg)) cell characteristics, a significant proportion of FOXP3(+) T cells is compromised in its suppressive function, despite the maintenance of other phenotypic and functional regulatory T hallmark features. In addition, these nonsuppressive FOXP3(+) T cells preferentially emerge from the CD45RO(+) memory pool, and arise as a consequence of a rapid downregulation of FOXP3 expression upon T cell reactivation. Surprisingly, these dysfunctional T(reg) cells with unstable FOXP3 expression do not manifest overt plasticity in terms of inflammatory cytokine secretion. These results open a path to an extensive study of the functional heterogeneity of CD4(+)CD25(High)FOXP3(+) T(reg) cells and warrant caution in the sole use of FOXP3 as a clinical marker for monitoring of immune regulation in humans.
Collapse
Affiliation(s)
- Eva d'Hennezel
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
892
|
Culina S, Boitard C, Mallone R. Antigen-based immune therapeutics for type 1 diabetes: magic bullets or ordinary blanks? Clin Dev Immunol 2011; 2011:286248. [PMID: 21647401 PMCID: PMC3102326 DOI: 10.1155/2011/286248] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 03/07/2011] [Accepted: 03/08/2011] [Indexed: 12/03/2022]
Abstract
The ideal drug of modern medicine is the one that achieves its therapeutic target with minimal adverse effects. Immune therapy of Type 1 diabetes (T1D) is no exception, and knowledge of the antigens targeted by pathogenic T cells offers a unique opportunity towards this goal. Different antigen formulations are being considered, such as proteins or peptides, either in their native form or modified ad hoc, DNA plasmids, and cell-based agents. Translation from mouse to human should take into account important differences, particularly in the time scale of autoimmune progression, and intervention. Critical parameters such as administration route, dosing and interval remain largely empirical and need to be further dissected. T1D staging through immune surrogate markers before and after treatment will be key in understanding therapeutic actions and to finally turn ordinary blanks into magic bullets.
Collapse
Affiliation(s)
- Slobodan Culina
- INSERM, U986, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 avenue Denfert Rochereau, 75674 Paris Cedex 14, France
- Université Paris-Descartes, 75006 Paris, France
| | - Christian Boitard
- INSERM, U986, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 avenue Denfert Rochereau, 75674 Paris Cedex 14, France
- Université Paris-Descartes, 75006 Paris, France
- Assistance Publique Hôpitaux de Paris, Hôtel Dieu, Service de Diabétologie, 75181 Paris, France
| | - Roberto Mallone
- INSERM, U986, DeAR Lab Avenir, Saint Vincent de Paul Hospital, 82 avenue Denfert Rochereau, 75674 Paris Cedex 14, France
- Université Paris-Descartes, 75006 Paris, France
- Assistance Publique Hôpitaux de Paris, Hôtel Dieu, Service de Diabétologie, 75181 Paris, France
| |
Collapse
|
893
|
Housley WJ, Adams CO, Nichols FC, Puddington L, Lingenheld EG, Zhu L, Rajan TV, Clark RB. Natural but Not Inducible Regulatory T Cells Require TNF-α Signaling for In Vivo Function. THE JOURNAL OF IMMUNOLOGY 2011; 186:6779-87. [DOI: 10.4049/jimmunol.1003868] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
894
|
Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev 2011; 238:247-62. [PMID: 20969597 DOI: 10.1111/j.1600-065x.2010.00951.x] [Citation(s) in RCA: 418] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD4(+) T cells, also known as T-helper (Th) cells, play an important role in orchestrating adaptive immune responses to various infectious agents. They are also involved in the induction of autoimmune and allergic diseases. Upon T-cell receptor (TCR)-mediated cell activation, naive CD4(+) T cells can differentiate into at least four major lineages, Th1, Th2, Th17, and iTreg cells, that participate in different types of immune responses. Networks of cytokines and transcription factors are critical for determining CD4(+) T-cell fates and effector cytokine production. Here, we review collaboration and cross-regulation between various essential cytokines in the activation/induction of key transcription factors during the process of Th cell differentiation towards these distinct lineages. We also discuss the interactions of key transcription factors at both genetic and protein levels and the function of the resulting network(s) in regulating the expression of effector cytokines.
Collapse
Affiliation(s)
- Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA.
| | | |
Collapse
|
895
|
Balasubramani A, Mukasa R, Hatton RD, Weaver CT. Regulation of the Ifng locus in the context of T-lineage specification and plasticity. Immunol Rev 2011; 238:216-32. [PMID: 20969595 DOI: 10.1111/j.1600-065x.2010.00961.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Study of the development of distinct CD4(+) T-cell subsets from naive precursors continues to provide excellent opportunities for dissection of mechanisms that control lineage-specific gene expression or repression. Whereas it had been thought that the induction of transcription networks that control T-lineage commitment were highly stable, reinforced by epigenetic processes that confer heritability of functional phenotypes by the progeny of mature T cells, recent findings support a more dynamic view of T-lineage commitment. Here, we highlight advances in the mapping and functional characterization of cis elements in the Ifng locus that have provided new insights into the control of the chromatin structure and transcriptional activity of this signature T-helper 1 cell gene. We also examine epigenetic features of the Ifng locus that have evolved to enable its reprogramming for expression by other T-cell subsets, particularly T-helper 17 cells, and contrast features of the Ifng locus with those of the Il17a-Il17f locus, which appears less promiscuous.
Collapse
Affiliation(s)
- Anand Balasubramani
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | |
Collapse
|
896
|
Beres A, Komorowski R, Mihara M, Drobyski WR. Instability of Foxp3 expression limits the ability of induced regulatory T cells to mitigate graft versus host disease. Clin Cancer Res 2011; 17:3969-83. [PMID: 21558402 DOI: 10.1158/1078-0432.ccr-10-3347] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Graft versus host disease (GVHD) is the major complication of allogeneic bone marrow transplantation (BMT) and limits the therapeutic efficacy of this modality. Although the role of natural T-regulatory cells (nTreg) in attenuating GVHD has been extensively examined, the ability of induced T-regulatory cells (iTreg) to mitigate GVHD is unknown. The purpose of this study was to examine the ability of in vitro and in vivo iTregs to abrogate GVHD. EXPERIMENTAL DESIGN We examined the ability of in vitro differentiated and in vivo iTregs to reduce the severity of GVHD in a clinically relevant mouse model of BMT. The effect of blockade of interleukin (IL) 6 signaling on the efficacy of these Treg populations was also studied. RESULTS In vitro differentiated iTregs fail to protect mice from lethal GVHD even when administered at high Treg:effector T-cell ratios. Lack of GVHD protection was associated with loss of Foxp3 expression and in vivo reversion of these cells to a proinflammatory phenotype characterized by secretion of IFN-γ. Phenotypic reversion could not be abrogated by blockade of IL-6 signaling or by in vitro exposure of iTregs to all-trans retinoic acid. In contrast, the in vivo induction of iTregs was significantly augmented by IL-6 blockade and this resulted in reduced GVHD. CONCLUSION Instability of Foxp3 expression limits the utility of adoptively transferred iTregs as a source of cellular therapy for the abrogation of GVHD. Blockade of IL-6 signaling augments the ability of in vivo iTregs to prevent GVHD but has no effect on in vitro differentiated iTregs.
Collapse
Affiliation(s)
- Amy Beres
- Bone Marrow Transplant Program and the Departments of Microbiology, Pathology, and Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | |
Collapse
|
897
|
Geng S, Yu Y, Kang Y, Pavlakis G, Jin H, Li J, Hu Y, Hu W, Wang S, Wang B. Efficient induction of CD25- iTreg by co-immunization requires strongly antigenic epitopes for T cells. BMC Immunol 2011; 12:27. [PMID: 21542943 PMCID: PMC3107816 DOI: 10.1186/1471-2172-12-27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 05/05/2011] [Indexed: 01/08/2023] Open
Abstract
Background We previously showed that co-immunization with a protein antigen and a DNA vaccine coding for the same antigen induces CD40low IL-10high tolerogenic DCs, which in turn stimulates the expansion of antigen-specific CD4+CD25-Foxp3+ regulatory T cells (CD25- iTreg). However, it was unclear how to choose the antigen sequence to maximize tolerogenic antigen presentation and, consequently, CD25- iTreg induction. Results In the present study, we demonstrated the requirement of highly antigenic epitopes for CD25- iTreg induction. Firstly, we showed that the induction of CD25- iTreg by tolerogenic DC can be blocked by anti-MHC-II antibody. Next, both the number and the suppressive activity of CD25- iTreg correlated positively with the overt antigenicity of an epitope to activate T cells. Finally, in a mouse model of dermatitis, highly antigenic epitopes derived from a flea allergen not only induced more CD25- iTreg, but also more effectively prevented allergenic reaction to the allergen than did weakly antigenic epitopes. Conclusions Our data thus indicate that efficient induction of CD25- iTreg requires highly antigenic peptide epitopes. This finding suggests that highly antigenic epitopes should be used for efficient induction of CD25- iTreg for clinical applications such as flea allergic dermatitis.
Collapse
Affiliation(s)
- Shuang Geng
- State Key Laboratory for Agro-Biotechnology China Agricultural University, Beijing 100193, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
898
|
Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat Med 2011; 17:673-5. [PMID: 21540856 DOI: 10.1038/nm.2389] [Citation(s) in RCA: 387] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 04/28/2010] [Indexed: 12/13/2022]
Abstract
CD4(+)CD25(high)CD127(low/-) forkhead box p3 (Foxp3)(+) regulatory T cells (T(reg) cells) possess functional plasticity. Here we describe a higher frequency of T helper type 1 (T(H)1)-like, interferon-γ (IFN-γ)-secreting Foxp3(+) T cells in untreated subjects with relapsing remitting multiple sclerosis (RRMS) as compared to healthy control individuals. In subjects treated with IFN-β, the frequency of IFN-γ(+)Foxp3(+) T cells is similar to that in healthy control subjects. In vitro, human T(reg) cells from healthy subjects acquire a T(H)1-like phenotype when cultured in the presence of interleukin-12 (IL-12). T(H)1-like T(reg) cells show reduced suppressive activity in vitro, which can partially be reversed by IFN-γ-specific antibodies or by removal of IL-12.
Collapse
|
899
|
Quezada SA, Peggs KS, Simpson TR, Allison JP. Shifting the equilibrium in cancer immunoediting: from tumor tolerance to eradication. Immunol Rev 2011; 241:104-18. [PMID: 21488893 PMCID: PMC3727276 DOI: 10.1111/j.1600-065x.2011.01007.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The continual interaction of the immune system with a developing tumor is thought to result in the establishment of a dynamic state of equilibrium. This equilibrium depends on the balance between effector and regulatory T-cell compartments. Whereas regulatory T cells can infiltrate and accumulate within tumors, effector T cells fail to efficiently do so. Furthermore, effector T cells that do infiltrate the tumor become tightly controlled by different regulatory cellular subsets and inhibitory molecules. The outcome of this balance is critical to survival, and whereas in some cases the equilibrium can rapidly result in the elimination of the transformed cells by the immune system, in many other cases the tumor manages to escape immune control. In this review, we discuss relevant work focusing on the establishment of the intratumor balance, the dynamic changes in the populations of effector and regulatory T cells within the tumor, and the role of the tumor vasculature and its activation state in the recruitment of different T-cell subsets. Finally, we also discuss work associated to the manipulation of the immune response to tumors and its impact on the infiltration, accumulation, and function of tumor-reactive lymphocytes within the tumor microenvironment.
Collapse
Affiliation(s)
- Sergio A. Quezada
- Ludwig Center for Cancer Immunotherapy, Howard Hughes Medical Institute, and Department of Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10021, USA
| | - Karl S. Peggs
- Department of Haematology, UCL Cancer Institute, Paul O’Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK
| | - Tyler R. Simpson
- Ludwig Center for Cancer Immunotherapy, Howard Hughes Medical Institute, and Department of Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10021, USA
| | - James P. Allison
- Ludwig Center for Cancer Immunotherapy, Howard Hughes Medical Institute, and Department of Immunology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10021, USA
| |
Collapse
|
900
|
Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family. Immunol Rev 2011; 241:180-205. [PMID: 21488898 PMCID: PMC3077803 DOI: 10.1111/j.1600-065x.2011.01011.x] [Citation(s) in RCA: 300] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Positive and negative costimulation by members of the CD28 family is critical for the development of productive immune responses against foreign pathogens and their proper termination to prevent inflammation-induced tissue damage. In addition, costimulatory signals are critical for the establishment and maintenance of peripheral tolerance. This paradigm has been established in many animal models and has led to the development of immunotherapies targeting costimulation pathways for the treatment of cancer, autoimmune disease, and allograft rejection. During the last decade, the complexity of the biology of costimulatory pathways has greatly increased due to the realization that costimulation does not affect only effector T cells but also influences regulatory T cells and antigen-presenting cells. Thus, costimulation controls T-cell tolerance through both intrinsic and extrinsic pathways. In this review, we discuss the influence of costimulation on intrinsic and extrinsic pathways of peripheral tolerance, with emphasis on members of the CD28 family, CD28, cytotoxic T-lymphocyte antigen-4 (CTLA-4), and programmed death-1 (PD-1), as well as the downstream cytokine interleukin-1 (IL-2).
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA 94143-0400, USA
| | | | | | | | | | | |
Collapse
|