801
|
Yurchenko E, Shio MT, Huang TC, Da Silva Martins M, Szyf M, Levings MK, Olivier M, Piccirillo CA. Inflammation-driven reprogramming of CD4+ Foxp3+ regulatory T cells into pathogenic Th1/Th17 T effectors is abrogated by mTOR inhibition in vivo. PLoS One 2012; 7:e35572. [PMID: 22545118 PMCID: PMC3335853 DOI: 10.1371/journal.pone.0035572] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 03/20/2012] [Indexed: 11/19/2022] Open
Abstract
While natural CD4(+)Foxp3(+) regulatory T (nT(REG)) cells have long been viewed as a stable and distinct lineage that is committed to suppressive functions in vivo, recent evidence supporting this notion remains highly controversial. We sought to determine whether Foxp3 expression and the nT(REG) cell phenotype are stable in vivo and modulated by the inflammatory microenvironment. Here, we show that Foxp3(+) nT(REG) cells from thymic or peripheral lymphoid organs reveal extensive functional plasticity in vivo. We show that nT(REG) cells readily lose Foxp3 expression, destabilizing their phenotype, in turn, enabling them to reprogram into Th1 and Th17 effector cells. nT(REG) cell reprogramming is a characteristic of the entire Foxp3(+) nT(REG) population and the stable Foxp3(NEG) T(REG) cell phenotype is associated with a methylated foxp3 promoter. The extent of nT(REG) cell reprogramming is modulated by the presence of effector T cell-mediated signals, and occurs independently of variation in IL-2 production in vivo. Moreover, the gut microenvironment or parasitic infection favours the reprogramming of Foxp3(+) T(REG) cells into effector T cells and promotes host immunity. IL-17 is predominantly produced by reprogrammed Foxp3(+) nT(REG) cells, and precedes Foxp3 down-regulation, a process accentuated in mesenteric sites. Lastly, mTOR inhibition with the immunosuppressive drug, rapamycin, stabilizes Foxp3 expression in T(REG) cells and strongly inhibits IL-17 but not RORγt expression in reprogrammed Foxp3(-) T(REG) cells. Overall, inflammatory signals modulate mTOR signalling and influence the stability of the Foxp3(+) nT(REG) cell phenotype.
Collapse
Affiliation(s)
- Ekaterina Yurchenko
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Marina T. Shio
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Tony C. Huang
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Maria Da Silva Martins
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Megan K. Levings
- Child and Family Research Institute, Department of Surgery, University of British Columbia, Vancouver, British Colombia, Canada
| | - Martin Olivier
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Ciriaco A. Piccirillo
- Departments of Microbiology and Immunology and Medicine, McGill University, Montreal, Quebec, Canada
- FOCIS Center of Excellence, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
802
|
Lindqvist CA, Loskog ASI. T regulatory cells in B-cell malignancy - tumour support or kiss of death? Immunology 2012; 135:255-60. [PMID: 22112044 DOI: 10.1111/j.1365-2567.2011.03539.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is well established that T regulatory (Treg) cells counteract tumour immunity. However, conflicting results describing the role of Treg cells in haematological tumours warrant further investigations to clarify the interactions between Treg cells and the tumour. B-cell malignancy derives from different stages of B-cell development and differentiation in which T cells play a profound role. The transformed B cell may still be in need of T-cell help to thrive but simultaneously they may be recognized and destroyed by cytotoxic lymphocytes. Recent reports demonstrate that Treg cells can suppress and even kill B cells as part of their normal function to rescue the body from autoimmunity. An emerging body of evidence points out that Treg cells not only inhibit tumour-specific T cells but may also have a role in suppressing the progression of the B-cell tumour. In this review, we discuss the origin and function of Treg cells and their role in patients with B-cell tumours.
Collapse
Affiliation(s)
- Camilla A Lindqvist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
803
|
Prinz I, Koenecke C. Therapeutic potential of induced and natural FoxP3(+) regulatory T cells for the treatment of Graft-versus-host disease. Arch Immunol Ther Exp (Warsz) 2012; 60:183-90. [PMID: 22476537 DOI: 10.1007/s00005-012-0172-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/04/2012] [Indexed: 01/30/2023]
Abstract
Graft-versus-host disease (GvHD) remains a major complication after allogeneic hematopoietic stem-cell-transplantation. Present GvHD prophylaxis and treatment is still based on unspecific immunosuppressive drug therapy. Over the last decade, the potential of cell-based therapies involving the infusion of regulatory T cells has emerged as a feasible alternative approach for the treatment and prevention of GvHD. Here we review current efforts to translate data obtained in rodent models into clinical trials. Special emphasis is placed on the variety of strategies to generate sufficient numbers of alloantigen-specific regulatory T cells for adoptive cell therapy. This can be achieved either by expansion or by induction of a regulatory phenotype in naive T cells. Stability of the immunosuppressive phenotype of transferred regulatory T cells even in the highly inflammatory environment of acute GvHD will be thereby a critical parameter for actual therapeutic application.
Collapse
Affiliation(s)
- Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
| | | |
Collapse
|
804
|
Joetham A, Ohnishi H, Okamoto M, Takeda K, Schedel M, Domenico J, Dakhama A, Gelfand EW. Loss of T regulatory cell suppression following signaling through glucocorticoid-induced tumor necrosis receptor (GITR) is dependent on c-Jun N-terminal kinase activation. J Biol Chem 2012; 287:17100-17108. [PMID: 22461627 DOI: 10.1074/jbc.m111.316943] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Naturally occurring Foxp3(+)CD4(+)CD25(+) T regulatory cell (nTreg)-mediated suppression of lung allergic responses is abrogated following ligation of glucocorticoid-induced tumor necrosis receptor (GITR) family-related protein. In vitro stimulation of nTregs with GITR ligand increased phosphorylation of c-Jun N-terminal kinase (JNK) but not extracellular signal-regulated protein kinase (ERK) or p38 MAPK. SP600125, a known JNK inhibitor, prevented GITR-mediated phosphorylation of JNK. Activation of JNK was associated with increases in the upstream mitogen-activated protein kinase kinase 7 (MKK7) and the downstream transcription factor NF-κβ. Phosphorylated c-Jun (p-c-Jun), indicative of the activation of JNK, was detected in the immunoprecipitates of nTregs from wild-type but not JNK- or GITR-deficient mice. Treatment with an inhibitor of JNK phosphorylation resulted in complete reversal of all GITR-induced changes in nTreg phenotype and function, with full restoration of suppression of in vivo lung allergic responses and in vitro proliferation of activated CD4(+)CD25(-) T cells. Thus, regulation of JNK phosphorylation plays a central role in T regulatory cell function with therapeutic implications for the treatment of asthma and autoimmune diseases.
Collapse
Affiliation(s)
- Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Hiroshi Ohnishi
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Masakazu Okamoto
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Joanne Domenico
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Azzeddine Dakhama
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206.
| |
Collapse
|
805
|
Oh S, Aitken M, Simons DM, Basehoar A, Garcia V, Kropf E, Caton AJ. Requirement for diverse TCR specificities determines regulatory T cell activity in a mouse model of autoimmune arthritis. THE JOURNAL OF IMMUNOLOGY 2012; 188:4171-80. [PMID: 22450809 DOI: 10.4049/jimmunol.1103598] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) are required to restrain the immune system from mounting an autoaggressive systemic inflammatory response, but why their activity can prevent (or allow) organ-specific autoimmunity remains poorly understood. We have examined how TCR specificity contributes to Treg activity using a mouse model of spontaneous autoimmune arthritis, in which CD4(+) T cells expressing a clonotypic TCR induce disease by an IL-17-dependent mechanism. Administration of polyclonal Tregs suppressed Th17 cell formation and prevented arthritis development; notably, Tregs expressing the clonotypic TCR did not. These clonotypic Tregs exerted Ag-specific suppression of effector CD4(+) T cells using the clonotypic TCR in vivo, but failed to mediate bystander suppression and did not prevent Th17 cells using nonclonotypic TCRs from accumulating in joint-draining lymph nodes of arthritic mice. These studies indicate that the availability of Tregs with diverse TCR specificities can be crucial to their activity in autoimmune arthritis.
Collapse
Affiliation(s)
- Soyoung Oh
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
806
|
Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of treg-mediated T cell suppression. Front Immunol 2012; 3:51. [PMID: 22566933 PMCID: PMC3341960 DOI: 10.3389/fimmu.2012.00051] [Citation(s) in RCA: 499] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 03/01/2012] [Indexed: 12/22/2022] Open
Abstract
CD4(+)CD25(high)Foxp3(+) regulatory T cells (Tregs) can suppress other immune cells and, thus, are critical mediators of peripheral self-tolerance. On the one hand, Tregs avert autoimmune disease and allergies. On the other hand, Tregs can prevent immune reactions against tumors and pathogens. Despite the importance of Tregs, the molecular mechanisms of suppression remain incompletely understood and controversial. Proliferation and cytokine production of CD4(+)CD25(-) conventional T cells (Tcons) can be inhibited directly by Tregs. In addition, Tregs can indirectly suppress Tcon activation via inhibition of the stimulatory capacity of antigen presenting cells. Direct suppression of Tcons by Tregs can involve immunosuppressive soluble factors or cell contact. Different mechanisms of suppression have been described, so far with no consensus on one universal mechanism. Controversies might be explained by the fact that different mechanisms may operate depending on the site of the immune reaction, on the type and activation state of the suppressed target cell as well as on the Treg activation status. Further, inhibition of T cell effector function can occur independently of suppression of proliferation. In this review, we summarize the described molecular mechanisms of suppression with a particular focus on suppression of Tcons and rapid suppression of T cell receptor-induced calcium (Ca(2+)), NFAT, and NF-κB signaling in Tcons by Tregs.
Collapse
Affiliation(s)
- Angelika Schmidt
- Division of Immunogenetics, Tumorimmunology Program, German Cancer Research Center (DKFZ) Heidelberg, Germany
| | | | | |
Collapse
|
807
|
Abstract
Regulatory T (Treg) cells expressing the transcription factor Foxp3 constitute a unique T-cell lineage committed to suppressive functions and play a central role in maintaining self-tolerance and immune homeostasis. While their differentiation state is remarkably stable in the face of various perturbations from the extracellular environment, recent studies have also revealed their adaptability to the changing environment; in response to extrinsic cues, Treg cells differentiate further into distinct substates to regulate different classes of immune responses effectively. In contrast, some other recent studies have challenged this notion of a committed Treg cell lineage and suggested that Treg cells might lose their identity and be reprogrammed to various effector helper T cells under certain circumstances, although this issue of environment-induced Treg cell reprogramming remains highly controversial. This review will focus on recent advances in our understanding of how the stability and adaptability of Treg cell lineage is regulated and how it might be perturbed in a changing environment.
Collapse
Affiliation(s)
- Shohei Hori
- Research Unit for Immune Homeostasis, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| |
Collapse
|
808
|
Immune modulation of inflammatory conditions: regulatory T cells for treatment of GvHD. Immunol Res 2012; 53:200-12. [DOI: 10.1007/s12026-012-8267-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
809
|
Lahdenperä AI, Hölttä V, Ruohtula T, Salo HM, Orivuori L, Westerholm-Ormio M, Savilahti E, Fälth-Magnusson K, Högberg L, Ludvigsson J, Vaarala O. Up-regulation of small intestinal interleukin-17 immunity in untreated coeliac disease but not in potential coeliac disease or in type 1 diabetes. Clin Exp Immunol 2012; 167:226-34. [PMID: 22235998 DOI: 10.1111/j.1365-2249.2011.04510.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Up-regulation of interleukin (IL)-17 in small intestinal mucosa has been reported in coeliac disease (CD) and in peripheral blood in type 1 diabetes (T1D). We explored mucosal IL-17 immunity in different stages of CD, including transglutaminase antibody (TGA)-positive children with potential CD, children with untreated and gluten-free diet-treated CD and in children with T1D. Immunohistochemistry was used for identification of IL-17 and forkhead box protein 3 (FoxP3)-positive cells and quantitative polymerase chain reaction (qPCR) for IL-17, FoxP3, retinoic acid-related orphan receptor (ROR)c and interferon (IFN)-γ transcripts. IL-1β, IL-6 and IL-17 were studied in supernatants from biopsy cultures. Expression of the apoptotic markers BAX and bcl-2 was evaluated in IL-17-stimulated CaCo-2 cells. The mucosal expression of IL-17 and FoxP3 transcripts were elevated in individuals with untreated CD when compared with the TGA-negative reference children, children with potential CD or gluten-free diet-treated children with CD (P < 0·005 for all IL-17 comparisons and P < 0·01 for all FoxP3 comparisons). The numbers of IL-17-positive cells were higher in lamina propria in children with CD than in children with T1D (P < 0·05). In biopsy specimens from patients with untreated CD, enhanced spontaneous secretion of IL-1β, IL-6 and IL-17 was seen. Activation of anti-apoptotic bcl-2 in IL-17-treated CaCo-2 epithelial cells suggests that IL-17 might be involved in mucosal protection. Up-regulation of IL-17 could, however, serve as a biomarker for the development of villous atrophy and active CD.
Collapse
Affiliation(s)
- A I Lahdenperä
- Division of Paediatrics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
810
|
T-cell proliferation and forkhead box P3 expression in human T cells are dependent on T-cell density: physics of a confined space? Hum Immunol 2012; 73:223-31. [DOI: 10.1016/j.humimm.2011.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 12/02/2011] [Accepted: 12/21/2011] [Indexed: 01/06/2023]
|
811
|
Walline CC, Kanakasabai S, Bright JJ. Dynamic interplay of T helpercell subsets in experimental autoimmune encephalomyelitis. World J Immunol 2012; 2:1-13. [DOI: 10.5411/wji.v2.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the temporal onset and dynamic interplay of CD4+ T helper cell subsets in experimental autoimmune encephalomyelitis (EAE).
METHODS: EAE was induced in C57BL/6 mice by immunization with myelin oligodendrocyte glycoprotein peptide p35-55. The clinical signs were scored and the tissue samples and immune cells isolated for analysis at different phases of EAE. The expression levels of inflammatory cytokines and related transcription factors were detected by quantitative reverse transcription polymerase chain reaction (PCR) and enzyme linked immunosorbant assay (ELISA). The percentages of Th1, Th17, Th2, Treg and memory T cell subsets in EAE were analyzed by immunostaining and flow cytometry. The data were analyzed by statistical techniques.
RESULTS: Quantitative real-time PCR analysis showed that EAE mice express elevated levels of Th1 [interferon gamma (IFNγ), interleukin (IL)-12p40], Th17 [IL-17, related orphan receptor gamma (RORγ), IL-12p40] and Treg [Foxp3, Epstein-Barr virus induced gene 3 (EBI3), IL-10] genes in the central nervous system at the peak of the disease. Whereas, the expression of Th1 (IFNγ, T-bet, IL-12p35, IL-12p40), Th17 (RORγ, IL-12p40), Th2 (IL-4) and Treg (Foxp3, EBI3) response genes was reduced in the spleen during pre-disease but gradually recovered at the later phases of EAE. ELISA and flow cytometry analyses showed an increase in Th17 response in the periphery, while Th1 response remained unchanged at the peak of disease. The mRNA levels of IFNγ, IL-17 and IL-12p40 in the brain were increased by 23 (P < 0.001), 9 (P < 0.05) and 14 (P < 0.01) fold, respectively, on day 21 of EAE. Conversely, the mRNA expression of IL-10 was increased by 2 fold (P < 0.05) in the spleen on day 21. CD4+CD25+Foxp3+Treg response was reduced at pre-disease but recovered to naïve levels by disease onset. The percentage of CD25+Foxp3+ regulatory T cells decreased from 7.7% in the naïve to 3.2% (P < 0.05) on day 7 of EAE, which then increased to 8.4% by day 28. Moreover, the CD4+CD127+CD44high memory T cell response was increased during the onset and recovery phases of EAE. The memory and effector cells showed an inverse relationship in EAE, where the memory T cells increased from 12.3% in naïve to 20% by day 21, and the effector cells decreased from 32% in naïve to 21% (P < 0.01) by day 21. The wild type C57BL/6 mice with EAE showed elevated levels of effector-memory T cells (TEM) with concomitant reduction in central-memory T cells (TCM), but the EAE-resistant IL-7R deficient mice showed elevated TCM with no effect on TEM cells in EAE.
CONCLUSION: Our findings highlight the temporal onset and dynamic interplay of effector, memory and regulatory CD4+ T cell subsets and its significance to clinical outcome in EAE and other autoimmune diseases.
Collapse
|
812
|
Barnaba V, Paroli M, Piconese S. The ambiguity in immunology. Front Immunol 2012; 3:18. [PMID: 22566903 PMCID: PMC3341998 DOI: 10.3389/fimmu.2012.00018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 02/02/2012] [Indexed: 01/07/2023] Open
Abstract
In the present article, we discuss the various ambiguous aspects of the immune system that render this complex biological network so highly flexible and able to defend the host from different external invaders. This ambiguity stems mainly from the property of the immune system to be both protective and harmful. Immunity cannot be fully protective without producing a certain degree of damage (immunopathology) to the host. The balance between protection and tissue damage is, therefore, critical for the establishment of immune homeostasis and protection. In this review, we will consider as ambiguous, various immunological tactics including: (a) the opposing functions driving immune responses, immune-regulation, and contra-regulation, as well as (b) the phenomenon of chronic immune activation as a result of a continuous cross-presentation of apoptotic T cells by dendritic cells. All these plans participate principally to maintain a state of chronic low-level inflammation during persisting infections, and ultimately to favor the species survival.
Collapse
Affiliation(s)
- Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma Rome, Italy
| | | | | |
Collapse
|
813
|
Koguchi Y, Buenafe AC, Thauland TJ, Gardell JL, Bivins-Smith ER, Jacoby DB, Slifka MK, Parker DC. Preformed CD40L is stored in Th1, Th2, Th17, and T follicular helper cells as well as CD4+ 8- thymocytes and invariant NKT cells but not in Treg cells. PLoS One 2012; 7:e31296. [PMID: 22363608 PMCID: PMC3283616 DOI: 10.1371/journal.pone.0031296] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/05/2012] [Indexed: 01/01/2023] Open
Abstract
CD40L is essential for the development of adaptive immune responses. It is generally thought that CD40L expression in CD4+ T cells is regulated transcriptionally and made from new mRNA following antigen recognition. However, imaging studies show that the majority of cognate interactions between effector CD4+ T cells and APCs in vivo are too short to allow de novo CD40L synthesis. We previously showed that Th1 effector and memory cells store preformed CD40L (pCD40L) in lysosomal compartments and mobilize it onto the plasma membrane immediately after antigenic stimulation, suggesting that primed CD4+ T cells may use pCD40L to activate APCs during brief encounters. Indeed, our recent study showed that pCD40L is sufficient to mediate selective activation of cognate B cells and trigger DC activation in vitro. In this study, we show that pCD40L is present in Th1 and follicular helper T cells developed during infection with lymphocytic choriomeningitis virus, Th2 cells in the airway of asthmatic mice, and Th17 cells from the CNS of animals with experimental autoimmune encephalitis (EAE). pCD40L is nearly absent in both natural and induced Treg cells, even in the presence of intense inflammation such as occurs in EAE. We also found pCD40L expression in CD4 single positive thymocytes and invariant NKT cells. Together, these results suggest that pCD40L may function in T cell development as well as an unexpectedly broad spectrum of innate and adaptive immune responses, while its expression in Treg cells is repressed to avoid compromising their suppressive activity.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Abigail C. Buenafe
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Timothy J. Thauland
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jennifer L. Gardell
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Elizabeth R. Bivins-Smith
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - David B. Jacoby
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Mark K. Slifka
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - David C. Parker
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
814
|
Jacobs JFM, Nierkens S, Figdor CG, de Vries IJM, Adema GJ. Regulatory T cells in melanoma: the final hurdle towards effective immunotherapy? Lancet Oncol 2012; 13:e32-42. [PMID: 22225723 DOI: 10.1016/s1470-2045(11)70155-3] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Immunotherapy studies in patients with melanoma have reported success in the expansion of tumour-specific effector T cells in vivo, but even in the presence of substantial numbers of functional T cells circulating in the blood, favourable clinical outcomes are scarce. This failure to induce robust clinical responses might be related to tumour-induced immune evasion, rendering the host tolerant to melanoma antigens. Immunosuppression in the tumour microenvironment mediated by regulatory T cells (Treg) is a dominant mechanism of tumour immune escape and is a major hurdle for tumour immunotherapy. Accumulation of Treg in melanoma is frequently recorded and the ratio of CD8-positive T cells versus Treg in the tumour microenvironment is predictive for survival of patients with melanoma. Hence, depletion of Treg seems to be a promising strategy for the enhancement of melanoma-specific immunity. Indeed, murine studies have shown that Treg depletion greatly increases the efficacy of immunotherapy. But despite the success of some strategies in depletion of Treg in patients, overall clinical efficacy has been disappointing. The lack of Treg specificity of the Treg depleting strategies applied so far imply that well-designed studies into dosage, timing, and administration regimens with more specific agents are urgently needed. Depletion of functional Treg from the tumour microenvironment as part of multifaceted immunotherapeutic treatments is a major challenge to induce clinically relevant immune responses against melanomas.
Collapse
Affiliation(s)
- Joannes F M Jacobs
- Department of Tumour Immunology, Nijmegen Centre for Molecular Life Sciences, Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
815
|
Immunomodulation of antigen presenting cells promotes natural regulatory T cells that prevent autoimmune diabetes in NOD mice. PLoS One 2012; 7:e31153. [PMID: 22355341 PMCID: PMC3280284 DOI: 10.1371/journal.pone.0031153] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 01/03/2012] [Indexed: 01/07/2023] Open
Abstract
Progression towards type 1 diabetes (T1D) in susceptible patients is linked to a progressive decline in the capacity of regulatory T cells (Treg) to maintain tolerance. As such, therapies aimed at redressing the failing Treg compartment have been the subject of intense investigation. Treg dysfunction in T1D has recently been linked to a reduced capacity of antigen presenting cells (APCs) to maintain Treg function rather than Treg intrinsic defects. This suggests that therapies aimed simply at addressing the failing Treg compartment are unlikely to provide long-term protection. Here, we demonstrate that modulation of the inflammatory status of CD11b+CD11c− APCs favors the upregulation of protective Tregs in a mouse model of T1D. We further demonstrate that reduced expression of the costimulatory molecule CD40 plays a role in this increased immunoregulatory capacity. Strikingly, Treg upregulation resulted exclusively from an increase in natural Tregs rather than the peripheral conversion of conventional T cells. This suggests that modulation of CD11b+ CD11c− APCs inflammatory properties favors the establishment of natural Treg responses that, unlike adaptive Treg responses, are likely to maintain tolerance to a broad range of antigens. As such, modulation of this APC subset represents a potential therapeutic avenue to reestablish peripheral tolerance and protect from autoimmune diseases such as T1D.
Collapse
|
816
|
Issazadeh-Navikas S, Teimer R, Bockermann R. Influence of dietary components on regulatory T cells. Mol Med 2012; 18:95-110. [PMID: 22113499 DOI: 10.2119/molmed.2011.00311] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/28/2011] [Indexed: 12/20/2022] Open
Abstract
Common dietary components including vitamins A and D, omega-3 and probiotics are now widely accepted to be essential to protect against many diseases with an inflammatory nature. On the other hand, high-fat diets are documented to exert multiple deleterious effects, including fatty liver diseases. Here we discuss the effect of dietary components on regulatory T cell (Treg) homeostasis, a central element of the immune system to prevent chronic tissue inflammation. Accordingly, evidence on the impact of dietary components on diseases in which Tregs play an influential role will be discussed. We will review chronic tissue-specific autoimmune and inflammatory conditions such as inflammatory bowel disease, type 1 diabetes mellitus, multiple sclerosis, rheumatoid arthritis and allergies among chronic diseases where dietary factors could have a direct influence via modulation of Tregs homeostasis and functions.
Collapse
|
817
|
Miyao T, Floess S, Setoguchi R, Luche H, Fehling HJ, Waldmann H, Huehn J, Hori S. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity 2012; 36:262-75. [PMID: 22326580 DOI: 10.1016/j.immuni.2011.12.012] [Citation(s) in RCA: 465] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 12/08/2011] [Accepted: 12/23/2011] [Indexed: 12/13/2022]
Abstract
The emerging notion of environment-induced reprogramming of Foxp3(+) regulatory T (Treg) cells into helper T (Th) cells remains controversial. By genetic fate mapping or adoptive transfers, we have identified a minor population of nonregulatory Foxp3(+) T cells exhibiting promiscuous and transient Foxp3 expression, which gave rise to Foxp3(-) ("exFoxp3") Th cells and selectively accumulated in inflammatory cytokine milieus or in lymphopenic environments including those in early ontogeny. In contrast, Treg cells did not undergo reprogramming under those conditions irrespective of their thymic or peripheral origins. Moreover, although a few Treg cells transiently lose Foxp3 expression, such "latent" Treg cells retained their memory and robustly re-expressed Foxp3 and suppressive function upon activation. This study establishes that Treg cells constitute a stable cell lineage, whose committed state in a changing environment is ensured by DNA demethylation of the Foxp3 locus irrespectively of ongoing Foxp3 expression.
Collapse
Affiliation(s)
- Takahisa Miyao
- Research Unit for Immune Homeostasis, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | |
Collapse
|
818
|
Hatam LJ, Devoti JA, Rosenthal DW, Lam F, Abramson AL, Steinberg BM, Bonagura VR. Immune suppression in premalignant respiratory papillomas: enriched functional CD4+Foxp3+ regulatory T cells and PD-1/PD-L1/L2 expression. Clin Cancer Res 2012; 18:1925-35. [PMID: 22322668 DOI: 10.1158/1078-0432.ccr-11-2941] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Respiratory papillomas, caused by human papillomaviruses types 6 and 11 (HPV6/11), are premalignant lesions with potential for malignant conversion. The cytokine and chemokine micromilieu of papillomas is T(H)2-like with a marked absence of IFN-γ expression. To illuminate why patients with recurrent respiratory papillomatosis (RRP) fail to effectively control their disease, we further investigated the suppressive cellular microenvironment in papillomas. EXPERIMENTAL DESIGN CD4(+)CD25(+)CD127(low/-)Foxp3(+) regulatory T cells (Treg) and CD4(+)CD25(-)CD127(low/-)Foxp3(-) T cells within papillomas were characterized and isolated. Their suppressor function was measured by inhibition of peripheral blood mononuclear cell (PBMC) proliferation. Expression of PD-1, CD69, and Helios was identified on these T cells. PD-L1, PD-L2, CCL17, and CCL22 mRNA was also identified in papillomas by quantitative PCR. RESULTS Functional Tregs were markedly enriched in papillomas and strongly inhibited anti-CD3 and anti-CD28 antibody activated PBMC proliferation. The natural Treg marker Helios was reduced on Tregs from papillomas, indicating that the majority of Tregs in papillomas are adaptive. The majority of the papilloma-derived CD4(+) T cells expressed the CD4(+)CD25(-)CD127(low/-)Foxp3(-)PD1(+)CD69(+) phenotype and failed to suppress PBMC proliferation, suggesting that they are chronically activated and exhausted. The Treg-attracting chemokine CCL22 was equally expressed by all laryngeal tissues examined. However, CCL17 was robustly expressed by papillomas compared with unaffected laryngeal tissues from RRP patients and individuals without RRP. PD-L1 was elevated in papillomas compared with control laryngeal tissues. CONCLUSIONS Papilloma CD4(+) T cells are enriched with functional Tregs, and the adaptive Helios(-) Treg fraction was increased within the T(H)2-like papilloma micromilieu. CD4(+)CD25(-)CD127(low/-)Foxp3(-) T-cells failed to suppress PBMC proliferation and may be exhausted. The PD-1/PDL-1 pathway may represent an additional immunosuppressive mechanism that contributes to defective HPV6/11 clearance in RRP.
Collapse
Affiliation(s)
- Lynda J Hatam
- The Feinstein Institute for Medical Research, 350 Community Drive, Room 1239, Manhasset, NY 11030, USA
| | | | | | | | | | | | | |
Collapse
|
819
|
Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, Umetsu DT, Rudensky AY. Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature 2012; 482:395-9. [PMID: 22318520 DOI: 10.1038/nature10772] [Citation(s) in RCA: 650] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 12/06/2011] [Indexed: 12/13/2022]
Abstract
A balance between pro- and anti-inflammatory mechanisms at mucosal interfaces, which are sites of constitutive exposure to microbes and non-microbial foreign substances, allows for efficient protection against pathogens yet prevents adverse inflammatory responses associated with allergy, asthma and intestinal inflammation. Regulatory T (T(reg)) cells prevent systemic and tissue-specific autoimmunity and inflammatory lesions at mucosal interfaces. These cells are generated in the thymus (tT(reg) cells) and in the periphery (induced (i)T(reg) cells), and their dual origin implies a division of labour between tT(reg) and iT(reg) cells in immune homeostasis. Here we show that a highly selective blockage in differentiation of iT(reg) cells in mice did not lead to unprovoked multi-organ autoimmunity, exacerbation of induced tissue-specific autoimmune pathology, or increased pro-inflammatory responses of T helper 1 (T(H)1) and T(H)17 cells. However, mice deficient in iT(reg) cells spontaneously developed pronounced T(H)2-type pathologies at mucosal sites--in the gastrointestinal tract and lungs--with hallmarks of allergic inflammation and asthma. Furthermore, iT(reg)-cell deficiency altered gut microbial communities. These results suggest that whereas T(reg) cells generated in the thymus appear sufficient for control of systemic and tissue-specific autoimmunity, extrathymic differentiation of T(reg) cells affects commensal microbiota composition and serves a distinct, essential function in restraint of allergic-type inflammation at mucosal interfaces.
Collapse
Affiliation(s)
- Steven Z Josefowicz
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, New York 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
820
|
Kwon HS, Lim HW, Wu J, Schnölzer M, Verdin E, Ott M. Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2712-21. [PMID: 22312127 DOI: 10.4049/jimmunol.1100903] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The Foxp3 transcription factor is the master regulator of regulatory T cell (Treg) differentiation and function. Its activity is regulated by reversible acetylation. Using mass spectrometry of immunoprecipitated proteins, we identify three novel acetylation sites in murine Foxp3 (K31, K262, and K267) and the corresponding sites in human FOXP3 proteins. Newly raised modification-specific Abs against acetylated K31 and K267 confirm acetylation of these residues in murine Tregs. Mutant Foxp3 proteins carrying arginine substitutions at the three acetylation sites (3KR) accumulate in T cells to higher levels than wild-type Foxp3 and exert better suppressive activity in coculture experiments. Acetylation and stability of wild-type, but not mutant, Foxp3 is enhanced when cells are treated with Ex-527, an inhibitor of the NAD(+)-dependent deacetylase SIRT1. Treatment with Ex-527 promotes Foxp3 expression during induced Treg differentiation, enhances Foxp3 levels in natural Tregs, and prevents loss of Foxp3 expression in adoptively transferred Tregs in mice. Our data identify SIRT1 as a negative regulator of Treg function via deacetylation of three novel target sites in Foxp3. SIRT1 inhibitors strengthen the suppressive activity of Tregs and may be useful in enhancing Treg-based therapeutic approaches to autoimmune diseases or graft rejections.
Collapse
Affiliation(s)
- Hye-Sook Kwon
- Gladstone Institute of Virology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | | | | | | | | |
Collapse
|
821
|
Abstract
Regulatory T cells (Tregs) are long-lived cells that suppress immune responses in vivo in a dominant and antigen-specific manner. Therefore, therapeutic application of Tregs to control unwanted immune responses is an active area of investigation. Tregs can confer long-term protection against auto-inflammatory diseases in mouse models. They have also been shown to be effective in suppressing alloimmunity in models of graft-versus-host disease and organ transplantation. Building on extensive research in Treg biology and preclinical testing of therapeutic efficacy over the past decade, we are now at the point of evaluating the safety and efficacy of Treg therapy in humans. This review focuses on developing therapy for transplantation using CD4(+)Foxp3(+) Tregs, with an emphasis on the studies that have informed clinical approaches that aim to maximize the benefits while overcoming the challenges and risks of Treg cell therapy.
Collapse
Affiliation(s)
- Qizhi Tang
- Division of Transplantation, Department of Surgery, University of California San Francisco, San Francisco, CA 94143-0780, USA.
| | | | | |
Collapse
|
822
|
Yamanishi H, Murakami H, Ikeda Y, Abe M, Kumagi T, Hiasa Y, Matsuura B, Onji M. Regulatory dendritic cells pulsed with carbonic anhydrase I protect mice from colitis induced by CD4+CD25- T cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:2164-72. [PMID: 22291189 DOI: 10.4049/jimmunol.1100559] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inflammatory bowel disease (IBD), which is characterized by a dysregulated intestinal immune response, is postulated to be controlled by intestinal self-antigens and bacterial Ags. Fecal extracts called cecal bacterial Ag (CBA) have been implicated in the pathogenesis of IBD. In this study, we identified a major protein of CBA related to the pathogenesis of IBD and established a therapeutic approach using Ag-pulsed regulatory dendritic cells (Reg-DCs). Using two-dimensional gel electrophoresis and MALDI-TOF mass spectrometry, carbonic anhydrase I (CA I) was identified as a major protein of CBA. Next, we induced colitis by transfer of CD4(+)CD25(-) T cells obtained from BALB/c mice into SCID mice. Mice were treated with CBA- or CA I-pulsed Reg-DCs (Reg-DCs(CBA) or Reg-DCs(CA1)), which expressed CD200 receptor 3 and produced high levels of IL-10. Treatment with Reg-DCs(CBA) and Reg-DCs(CA1) ameliorated colitis. This effect was shown to be Ag-specific based on no clinical response of irrelevant Ag (keyhole limpet hemocyanin)-pulsed Reg-DCs. Foxp3 mRNA expression was higher but RORγt mRNA expression was lower in the mesenteric lymph nodes (MLNs) of the Reg-DCs(CA1)-treated mice compared with those in the MLNs of control mice. In the MLNs, Reg-DCs(CA1)-treated mice had higher mRNA expression of IL-10 and TGF-β1 and lower IL-17 mRNA expression and protein production compared with those of control mice. In addition, Reg-DCs(CBA)-treated mice had higher Foxp3(+)CD4(+)CD25(+) and IL-10-producing regulatory T cell frequencies in MLNs. In conclusion, Reg-DCs(CA1) protected progression of colitis induced by CD4(+)CD25(-) T cell transfer in an Ag-specific manner by inducing the differentiation of regulatory T cells.
Collapse
Affiliation(s)
- Hirofumi Yamanishi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa, Tōon, Ehime 791-0295, Japan
| | | | | | | | | | | | | | | |
Collapse
|
823
|
Issa F, Chandrasekharan D, Wood KJ. Regulatory T cells as modulators of chronic allograft dysfunction. Curr Opin Immunol 2012; 23:648-54. [PMID: 21752619 DOI: 10.1016/j.coi.2011.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 06/20/2011] [Indexed: 12/19/2022]
Abstract
Chronic allograft dysfunction (CAD) in solid organ transplantation is a principal cause of patient morbidity and late allograft loss. The pathogenesis of CAD is largely secondary to chronic damage by the adaptive immune system and long-term immunosuppression. Manipulating these factors may be possible with the use of regulatory T cells (Treg), which have the ability to suppress specific immune responses and therefore potentially remove the need for immunosuppressive drugs. Studies of CAD in experimental models have demonstrated the capacity for both mouse and human Treg cellular therapy to prevent the development of some manifestations of CAD. Furthermore, a role for Treg has been demonstrated in clinically tolerant transplant patients. Certain immunosuppressive therapies are also proving to be 'Treg friendly' and may be helpful in promoting Treg while maintaining other immunosuppressive activity. With this in mind, monitoring for biomarkers of operational tolerance with tailored immunosuppressive therapy or controlled weaning in conjunction with Treg cellular therapy may be a useful strategy to pursue.
Collapse
Affiliation(s)
- Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | | | | |
Collapse
|
824
|
Wang C, Lee JH, Kim CH. Optimal population of FoxP3+ T cells in tumors requires an antigen priming-dependent trafficking receptor switch. PLoS One 2012; 7:e30793. [PMID: 22292042 PMCID: PMC3264621 DOI: 10.1371/journal.pone.0030793] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/29/2011] [Indexed: 01/21/2023] Open
Abstract
FoxP3+ T cells populate tumors and regulate anti-tumor immunity. The requirement for optimal population of FoxP3+ regulatory T cells in tumors remains unclear. We investigated the migration requirement and stability of tumor-associated FoxP3+ T cells. We found that only memory, but not naïve, FoxP3+ T cells are highly enriched in tumors. Almost all of the tumor-infiltrating FoxP3+ T cells express Helios, an antigen associated either with thymus-generated FoxP3+ T cells or activated T cells in the periphery. The tumor-infiltrating FoxP3+ T cells largely lack CD62L and CCR7, two trafficking receptors required for T cell migration into secondary lymphoid tissues. Instead, the tumor infiltrating FoxP3+ T cells highly express memory/tumor-associated CCR8 and CXCR4. Antigen priming is required for induction of this trafficking receptor phenotype in FoxP3+ T cells and only antigen primed, but not antigen-inexperienced naive, FoxP3+ T cells can efficiently migrate into tumors. While the migration of FoxP3+ T cells into tumors was a readily detectable event, generation of induced FoxP3+ T cells within tumors was unexpectedly inefficient. Genetic marking of current and ex-FoxP3+ T cells revealed that tumor-infiltrating FoxP3+ T cells are highly stable and do not readily convert back to FoxP3− T cells. Taken together, our results indicate that population of tumors with thymus-generated FoxP3+ T cells requires an antigen priming-dependent trafficking receptor switch in lymphoid tissues.
Collapse
Affiliation(s)
- Chuanwu Wang
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue Cancer Center, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Jee H. Lee
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue Cancer Center, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
| | - Chang H. Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue Cancer Center, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
825
|
Lobo PI, Bajwa A, Schlegel KH, Vengal J, Lee SJ, Huang L, Ye H, Deshmukh U, Wang T, Pei H, Okusa MD. Natural IgM anti-leukocyte autoantibodies attenuate excess inflammation mediated by innate and adaptive immune mechanisms involving Th-17. THE JOURNAL OF IMMUNOLOGY 2012; 188:1675-85. [PMID: 22262657 DOI: 10.4049/jimmunol.1101762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Little is known about the function of natural IgM autoantibodies, especially that of IgM anti-leukocyte autoantibodies (IgM-ALA). Natural IgM-ALA are present at birth and characteristically increase during inflammatory and infective conditions. Our prior clinical observations and those of other investigators showing fewer rejections in renal and cardiac allografts transplanted into recipients with high levels of IgM-ALA led us to investigate whether IgM-ALA regulate the inflammatory response. In this article, we show that IgM, in physiologic doses, inhibit proinflammatory cells from proliferating and producing IFN-γ and IL-17 in response to alloantigens (MLR), anti-CD3, and the glycolipid α-galactosyl ceramide. We showed in an IgM knockout murine model, with intact B cells and regulatory T cells, that there was more severe inflammation and loss of function in the absence of IgM after renal ischemia reperfusion injury and cardiac allograft rejection. Replenishing IgM in IgM knockout mice or increasing the levels of IgM-ALA in wild-type B6 mice significantly attenuated the inflammation in both of these inflammatory models that involve IFN-γ and IL-17. The protective effect on renal ischemia reperfusion injury was not observed using IgM preadsorbed with leukocytes to remove IgM-ALA. We provide data to show that the anti-inflammatory effect of IgM is mediated, in part, by inhibiting TLR-4-induced NF-κB translocation into the nucleus and inhibiting differentiation of activated T cells into Th-1 and Th-17 cells. These observations highlight the importance of IgM-ALA in regulating excess inflammation mediated by both innate and adaptive immune mechanisms and where the inflammatory response involves Th-17 cells that are not effectively regulated by regulatory T cells.
Collapse
Affiliation(s)
- Peter I Lobo
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
826
|
Abstract
Monogenic autoimmune syndromes provide a rare yet powerful glimpse into the fundamental mechanisms of immunologic tolerance. Such syndromes reveal not only the contribution of an individual breakpoint in tolerance but also patterns in the pathogenesis of autoimmunity. Disturbances in innate immunity, a system built for ubiquitous sensing of danger signals, tend to generate systemic autoimmunity. For example, defects in the clearance of self-antigens and chronic stimulation of type 1 interferons lead to the systemic autoimmunity seen in C1q deficiency, SPENCDI, and AGS. In contrast, disturbances of adaptive immunity, which is built for antigen specificity, tend to produce organ-specific autoimmunity. Thus, the loss of lymphocyte homeostasis, whether through defects in apoptosis, suppression, or negative selection, leads to organ-specific autoimmunity in ALPS, IPEX, and APS1. We discuss the unique mechanisms of disease in these prominent syndromes as well as how they contribute to the spectrum of organ-specific or systemic autoimmunity. The continued study of rare variants in autoimmune disease will inform future investigations and treatments directed at rare and common autoimmune diseases alike.
Collapse
Affiliation(s)
- Mickie H. Cheng
- Diabetes Center; Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Francisco, San Francisco, California 94143;
| | - Mark S. Anderson
- Diabetes Center; Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Francisco, San Francisco, California 94143;
| |
Collapse
|
827
|
Sakaguchi S, Powrie F, Ransohoff RM. Re-establishing immunological self-tolerance in autoimmune disease. Nat Med 2012; 18:54-8. [DOI: 10.1038/nm.2622] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
828
|
González-Navajas JM, Lee J, David M, Raz E. Immunomodulatory functions of type I interferons. Nat Rev Immunol 2012; 12:125-35. [PMID: 22222875 PMCID: PMC3727154 DOI: 10.1038/nri3133] [Citation(s) in RCA: 751] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interferon-α (IFNα) and IFNβ, collectively known as type I IFNs, are the major effector cytokines of the host immune response against viral infections. However, the production of type I IFNs is also induced in response to bacterial ligands of innate immune receptors and/or bacterial infections, indicating a broader physiological role for these cytokines in host defence and homeostasis than was originally assumed. The main focus of this Review is the underappreciated immunomodulatory functions of type I IFNs in health and disease. We discuss their function in the regulation of innate and adaptive immune responses, the response to bacterial ligands, inflammasome activation, intestinal homeostasis and inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- José M González-Navajas
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0663, USA.
| | | | | | | |
Collapse
|
829
|
Abstract
The immune system has evolved to mount an effective defense against pathogens and to minimize deleterious immune-mediated inflammation caused by commensal microorganisms, immune responses against self and environmental antigens, and metabolic inflammatory disorders. Regulatory T (Treg) cell-mediated suppression serves as a vital mechanism of negative regulation of immune-mediated inflammation and features prominently in autoimmune and autoinflammatory disorders, allergy, acute and chronic infections, cancer, and metabolic inflammation. The discovery that Foxp3 is the transcription factor that specifies the Treg cell lineage facilitated recent progress in understanding the biology of regulatory T cells. In this review, we discuss cellular and molecular mechanisms in the differentiation and function of these cells.
Collapse
Affiliation(s)
- Steven Z Josefowicz
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute, New York, NY 10021, USA
| | | | | |
Collapse
|
830
|
Abstract
Type 1 diabetes presents clinically with overt hyperglycemia resulting from progressive immune-mediated destruction of pancreatic β-cells and associated metabolic dysfunction. Combined genetic and immunological studies now highlight deficiencies in both the interleukin-2 (IL-2) receptor and its downstream signaling pathway as a central defect in the pathogenesis of type 1 diabetes. Prior intervention studies in animal models indicate that augmenting IL-2 signaling can prevent and reverse disease, with protection conferred primarily by restoration of regulatory T-cell (Treg) function. In this article, we will focus on studies of type 1 diabetes noting deficient IL-2 signaling and build what we believe forms the molecular framework for their contribution to the disease. This activity results in the identification of a series of potentially novel therapeutic targets that could restore proper immune regulation in type 1 diabetes by augmenting the IL-2 pathway.
Collapse
|
831
|
Psychoneuroimmunology meets neuropsychopharmacology: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology 2012; 37:137-62. [PMID: 21918508 PMCID: PMC3238082 DOI: 10.1038/npp.2011.205] [Citation(s) in RCA: 659] [Impact Index Per Article: 54.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The potential contribution of chronic inflammation to the development of neuropsychiatric disorders such as major depression has received increasing attention. Elevated biomarkers of inflammation, including inflammatory cytokines and acute-phase proteins, have been found in depressed patients, and administration of inflammatory stimuli has been associated with the development of depressive symptoms. Data also have demonstrated that inflammatory cytokines can interact with multiple pathways known to be involved in the development of depression, including monoamine metabolism, neuroendocrine function, synaptic plasticity, and neurocircuits relevant to mood regulation. Further understanding of mechanisms by which cytokines alter behavior have revealed a host of pharmacologic targets that may be unique to the impact of inflammation on behavior and may be especially relevant to the treatment and prevention of depression in patients with evidence of increased inflammation. Such targets include the inflammatory signaling pathways cyclooxygenase, p38 mitogen-activated protein kinase, and nuclear factor-κB, as well as the metabolic enzyme, indoleamine-2,3-dioxygenase, which breaks down tryptophan into kynurenine. Other targets include the cytokines themselves in addition to chemokines, which attract inflammatory cells from the periphery to the brain. Psychosocial stress, diet, obesity, a leaky gut, and an imbalance between regulatory and pro-inflammatory T cells also contribute to inflammation and may serve as a focus for preventative strategies relevant to both the development of depression and its recurrence. Taken together, identification of mechanisms by which cytokines influence behavior may reveal a panoply of personalized treatment options that target the unique contributions of the immune system to depression.
Collapse
|
832
|
Hansmann L, Schmidl C, Kett J, Steger L, Andreesen R, Hoffmann P, Rehli M, Edinger M. Dominant Th2 differentiation of human regulatory T cells upon loss of FOXP3 expression. THE JOURNAL OF IMMUNOLOGY 2011; 188:1275-82. [PMID: 22210907 DOI: 10.4049/jimmunol.1102288] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CD4(+)CD25(+)FOXP3(+) regulatory T cells (Treg) are pivotal for peripheral self-tolerance. They prevent immune responses to auto- and alloantigens and are thus under close scrutiny as cellular therapeutics for autoimmune diseases and the prevention or treatment of alloresponses after organ or stem cell transplantation. We previously showed that human Treg with a memory cell phenotype, but not those with a naive phenotype, rapidly downregulate expression of the lineage-defining transcription factor FOXP3 upon in vitro expansion. We now compared the transcriptomes of stable FOXP3(+) Treg and converted FOXP3(-) ex-Treg by applying a newly developed intranuclear staining protocol that permits the isolation of intact mRNA from fixed, permeabilized, and FACS-purified cell populations. Whole-genome microarray analysis revealed strong and selective upregulation of Th2 signature genes, including GATA-3, IL-4, IL-5, and IL-13, upon downregulation of FOXP3. Th2 differentiation of converted FOXP3(-) ex-Treg occurred even under nonpolarizing conditions and could not be prevented by IL-4 signaling blockade. Thus, our studies identify Th2 differentiation as the default developmental program of human Treg after downregulation of FOXP3.
Collapse
Affiliation(s)
- Leo Hansmann
- Department of Hematology and Oncology, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
833
|
Nature and nurture in Foxp3(+) regulatory T cell development, stability, and function. Hum Immunol 2011; 73:232-9. [PMID: 22240298 DOI: 10.1016/j.humimm.2011.12.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 04/18/2011] [Accepted: 12/19/2011] [Indexed: 02/06/2023]
Abstract
Foxp3(+) regulatory T lymphocytes (Treg) are critical homeostatic regulators of immune and inflammatory responses. Their absence leads to fulminant multiorgan autoimmunity. This review explores recent studies that have altered our emerging view of the development, stability, and plasticity of these cells. Treg appear not to be a single entity, but a family of immunomodulatory cell types with shared capabilities. On a first level, Treg may alternatively form in response to developmental cues in the thymus as a distinct lineage of CD4(+) T cells or adaptively, in response to environmental cues received by mature conventional CD4(+) T lymphocytes. These 2 populations bear distinct specificity, stability, and genetic profiles and are differentially used in immune responses. Secondarily, in a manner analogous to the generation of T helper (Th)-1, Th2, and other T cell subsets, Treg may further specialize, adapting to the needs of their immunologic surroundings. Treg therefore comprise developmentally distinct, functionally overlapping cell populations that are uniquely designed to preserve immunologic homeostasis. They combine an impressive degree of both stability and adaptability.
Collapse
|
834
|
Klein M, Vaeth M, Scheel T, Grabbe S, Baumgrass R, Berberich-Siebelt F, Bopp T, Schmitt E, Becker C. Repression of cyclic adenosine monophosphate upregulation disarms and expands human regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2011; 188:1091-7. [PMID: 22190184 DOI: 10.4049/jimmunol.1102045] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The main molecular mechanism of human regulatory T cell (Treg)-mediated suppression has not been elucidated. We show in this study that cAMP represents a key regulator of human Treg function. Repression of cAMP production by inhibition of adenylate cyclase activity or augmentation of cAMP degradation through ectopic expression of a cAMP-degrading phosphodiesterase greatly reduces the suppressive activity of human Treg in vitro and in a humanized mouse model in vivo. Notably, cAMP repression additionally abrogates the anergic state of human Treg, accompanied by nuclear translocation of NFATc1 and induction of its short isoform NFATc1/αA. Treg expanded under cAMP repression, however, do not convert into effector T cells and regain their anergic state and suppressive activity upon proliferation. Together, these findings reveal the cAMP pathway as an attractive target for clinical intervention with Treg function.
Collapse
Affiliation(s)
- Matthias Klein
- University Medical Center, Institute for Immunology, Johannes Gutenberg-University, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
835
|
Ernerudh J, Berg G, Mjösberg J. Regulatory T helper cells in pregnancy and their roles in systemic versus local immune tolerance. Am J Reprod Immunol 2011; 66 Suppl 1:31-43. [PMID: 21726336 DOI: 10.1111/j.1600-0897.2011.01049.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
PROBLEM During pregnancy, the maternal immune system needs to adapt in order not to reject the semi-allogenic fetus. METHOD In this review, we describe and discuss the role of regulatory T (Treg) cells in fetal tolerance. RESULTS Treg cells constitute a T helper lineage that is derived from thymus (natural Treg cells) or is induced in the periphery (induced Treg cells). Treg cells are enriched at the fetal-maternal interface, showing a suppressive phenotype. In contrast, Treg cells are not increased in the circulation of pregnant women, and the suppressive capacity is similar to that in non-pregnant women. However, aberrations in Treg frequencies and functions, both systemically and in the uterus, may be involved in the complications of pregnancy. CONCLUSION Treg cells seem to have distinguished roles locally versus systemically, based on their distribution and phenotype.
Collapse
Affiliation(s)
- Jan Ernerudh
- Division of Clinical Immunology, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | | | | |
Collapse
|
836
|
Hirahara K, Vahedi G, Ghoreschi K, Yang XP, Nakayamada S, Kanno Y, O'Shea JJ, Laurence A. Helper T-cell differentiation and plasticity: insights from epigenetics. Immunology 2011; 134:235-45. [PMID: 21977994 DOI: 10.1111/j.1365-2567.2011.03483.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CD4(+) T cells have critical roles in orchestrating immune responses to diverse microbial pathogens. This is accomplished through the differentiation of CD4(+) T helper cells to specialized subsets in response to microbial pathogens, which evoke a distinct cytokine milieu. Signal transducer and activator of transcription family transcription factors sense these cytokines and they in turn regulate expression of lineage-defining master regulators that programme selective gene expression, resulting in distinctive phenotypes. However, phenotype and restricted gene expression are determined not only by the action of transcription factors; chromatin accessibility is required for these factors to exert their effect. Technical advances have greatly expanded our understanding of transcription factor action and dynamic changes in the epigenome that accompany cellular differentiation. In this review, we will discuss recent progress in the understanding of how cytokines influence gene expression and epigenetic modifications, and the impact of these findings on our views of helper cell lineage commitment and plasticity.
Collapse
Affiliation(s)
- Kiyoshi Hirahara
- Department of Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1930, USA.
| | | | | | | | | | | | | | | |
Collapse
|
837
|
Tran DQ. TGF-β: the sword, the wand, and the shield of FOXP3(+) regulatory T cells. J Mol Cell Biol 2011; 4:29-37. [PMID: 22158907 DOI: 10.1093/jmcb/mjr033] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Since its rediscovery in the mid-1990s, FOXP3(+) regulatory T cells (Tregs) have climbed the rank to become commander-in-chief of the immune system. They possess diverse power and ability to orchestrate the immune system in time of inflammation and infection as well as in time of harmony and homeostasis. To be the commander-in-chief, they must be equipped with both offensive and defensive weaponry. This review will focus on the function of transforming growth factor-β (TGF-β) as the sword, the wand, and the shield of Tregs. Functioning as a sword, this review will begin with a discussion of the evidence that supports how Tregs utilize TGF-β to paralyze cell activation and differentiation to suppress immune response. It will next provide evidence on how TGF-β from Tregs acts as a wand to convert naïve T cells into iTregs and Th17 to aid in their combat against inflammation and infection. Lastly, the review will present evidence on the role of TGF-β produced by Tregs in providing a shield to protect and maintain Tregs against apoptosis and destabilization when surrounded by inflammation and constant stimulation. This triadic function of TGF-β empowers Tregs with the responsibility and burden to maintain homeostasis, promote immune tolerance, and regulate host defense against foreign pathogens.
Collapse
Affiliation(s)
- Dat Q Tran
- Division of Pediatric Research Center, Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX 77030, USA.
| |
Collapse
|
838
|
Panzer M, Sitte S, Wirth S, Drexler I, Sparwasser T, Voehringer D. Rapid In Vivo Conversion of Effector T Cells into Th2 Cells during Helminth Infection. THE JOURNAL OF IMMUNOLOGY 2011; 188:615-23. [DOI: 10.4049/jimmunol.1101164] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
839
|
Xiao X, Gong W, Demirci G, Liu W, Spoerl S, Chu X, Bishop DK, Turka LA, Li XC. New insights on OX40 in the control of T cell immunity and immune tolerance in vivo. THE JOURNAL OF IMMUNOLOGY 2011; 188:892-901. [PMID: 22147766 DOI: 10.4049/jimmunol.1101373] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OX40 is a T cell costimulatory molecule that belongs to the TNFR superfamily. In the absence of immune activation, OX40 is selectively expressed by Foxp3(+) regulatory T cells (Tregs), but not by resting conventional T cells. The exact role of OX40 in Treg homeostasis and function remains incompletely defined. In this study, we demonstrate that OX40 engagement in vivo in naive mice induces initial expansion of Foxp3(+) Tregs, but the expanded Tregs have poor suppressive function and exhibit features of exhaustion. We also show that OX40 enables the activation of the Akt and Stat5 pathways in Tregs, resulting in transient proliferation of Tregs and reduced levels of Foxp3 expression. This creates a state of relative IL-2 deficiency in naive mice that further impacts Tregs. This exhausted Treg phenotype can be prevented by exogenous IL-2, as both OX40 and IL-2 agonists drive further expansion of Tregs in vivo. Importantly, Tregs expanded by both OX40 and IL-2 agonists are potent suppressor cells, and in a heart transplant model, they promote long-term allograft survival. Our data reveal a novel role for OX40 in promoting immune tolerance and may have important clinical implications.
Collapse
Affiliation(s)
- Xiang Xiao
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
840
|
Quezada SA, Peggs KS. Tumor-reactive CD4+ T cells: plasticity beyond helper and regulatory activities. Immunotherapy 2011; 3:915-7. [PMID: 21843076 DOI: 10.2217/imt.11.83] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
841
|
|
842
|
Li L, Boussiotis VA. Molecular and functional heterogeneity of T regulatory cells. Clin Immunol 2011; 141:244-52. [PMID: 21945485 PMCID: PMC3221919 DOI: 10.1016/j.clim.2011.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Accepted: 08/22/2011] [Indexed: 12/25/2022]
Abstract
Naturally occurring CD4+ T regulatory (Treg) cells are produced during maturation in the thymus and have a mandatory role in maintaining homeostasis and immune quiescence. Development and function of Treg cells depends on the transcription factor forkhead box P3 (Foxp3), which is necessary and sufficient for Treg cell function. Currently emerging evidence indicates Treg cells display molecular and functional heterogeneity and can be categorized into naïve and effector- or memory-like cells, which can produce effector cytokines supporting the idea that Treg cells retain plasticity. The role of Treg cells that acquire these properties remains unclear and is currently under intense investigation. In this review, we summarize recent advances on the differentiation of effector- or memory-like Treg cells, the impact of the cytokine milieu on the molecular and functional heterogeneity of Treg cells, and the clinical implications of the heterogeneity and specialization of Treg cells.
Collapse
Affiliation(s)
- Lequn Li
- Department of Medicine, Division of Hematology-Oncology and Cancer Biology, Harvard Medical School, Boston MA 02215, USA
| | | |
Collapse
|
843
|
de Jong HJ, Cohen Tervaert JW, Saldi SR, Vandebriel RJ, Souverein PC, Meyboom RH, van Loveren H, Klungel OH. Association Between Statin Use and Lupus-Like Syndrome Using Spontaneous Reports. Semin Arthritis Rheum 2011; 41:373-81. [DOI: 10.1016/j.semarthrit.2011.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/23/2011] [Accepted: 06/14/2011] [Indexed: 10/17/2022]
|
844
|
Abstract
PURPOSE OF REVIEW To summarize studies on the development and function of T-regulatory (TR) cells in primary immune deficiencies (PIDs). RECENT FINDINGS PIDs are associated with high rates of autoimmunity. TR cells, which are critical to the control of autoimmunity, appear involved in the pathogenesis of PID-related autoimmunity. A number of PIDs, including Omenn's syndrome and Wiskott-Aldrich syndrome, have been associated with impaired production and/or function of thymus-derived (natural) TR cells. Recently defined primary immunodeficiencies, including Stim1 deficiency, IL-10 receptor deficiency, and xIAP deficiency, have been associated with defects in TR cells. De-novo generated TR cells from peripheral CD4 conventional T cells is impaired in the hyper IgE syndrome. SUMMARY Gene defects underlying PIDs may also compromise the TR cell, leading to breakdown of peripheral tolerance.
Collapse
Affiliation(s)
- James W Verbsky
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | |
Collapse
|
845
|
Li CR, Baaten BJG, Bradley LM. Harnessing memory adaptive regulatory T cells to control autoimmunity in type 1 diabetes. J Mol Cell Biol 2011; 4:38-47. [PMID: 22116888 DOI: 10.1093/jmcb/mjr040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type 1 diabetes (T1D) results from autoimmune destruction of insulin-producing β-cells in the pancreatic islets. There is an immediate need to restore both β-cell function and immune tolerance to control disease progression and ultimately cure T1D. Currently, there is no effective treatment strategy to restore glucose regulation in patients with T1D. FoxP3-expressing CD4(+) regulatory T cells (Tregs) are potential candidates to control autoimmunity because they play a central role in maintaining self-tolerance. However, deficiencies in either naturally occurring Tregs (nTregs) themselves and/or their ability to control pathogenic effector T cells have been associated with T1D. Here, we hypothesize that nTregs can be replaced by FoxP3(+) adaptive Tregs (aTregs), which are uniquely equipped to combat autoreactivity in T1D. Unlike nTregs, aTregs are stable and provide long-lived protection. In this review, we summarize the current understanding of aTregs and their potential for use as an immunological intervention to treat T1D.
Collapse
Affiliation(s)
- Cheng-Rui Li
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
846
|
Pandiyan P, Zheng L, Lenardo MJ. The molecular mechanisms of regulatory T cell immunosuppression. Front Immunol 2011; 2:60. [PMID: 22566849 PMCID: PMC3342245 DOI: 10.3389/fimmu.2011.00060] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 10/19/2011] [Indexed: 12/22/2022] Open
Abstract
CD4⁺CD25⁺Foxp3⁺ T lymphocytes, known as regulatory T cells or T(regs), have been proposed to be a lineage of professional immune suppressive cells that exclusively counteract the effects of the immunoprotective "helper" and "cytotoxic" lineages of T lymphocytes. Here we discuss new concepts on the mechanisms and functions of T(regs). There are several key points we emphasize: 1. Tregs exert suppressive effects both directly on effector T cells and indirectly through antigen-presenting cells; 2. Regulation can occur through a novel mechanism of cytokine consumption to regulate as opposed to the usual mechanism of cytokine/chemokine production; 3. In cases where CD4⁺ effector T cells are directly inhibited by T(regs), it is chiefly through a mechanism of lymphokine withdrawal apoptosis leading to polyclonal deletion; and 4. Contrary to the current view, we discuss new evidence that T(regs), similar to other T-cells lineages, can promote protective immune responses in certain infectious contexts (Chen et al., 2011; Pandiyan et al., 2011). Although these points are at variance to varying degrees with the standard model of T(reg) behavior, we will recount developing findings that support these new concepts.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD, USA.
| | | | | |
Collapse
|
847
|
Sela U, Olds P, Park A, Schlesinger SJ, Steinman RM. Dendritic cells induce antigen-specific regulatory T cells that prevent graft versus host disease and persist in mice. ACTA ACUST UNITED AC 2011; 208:2489-96. [PMID: 22084406 PMCID: PMC3256961 DOI: 10.1084/jem.20110466] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Regulatory T cells generated by allostimulation with dendritic cells, transforming growth factor β, and retinoic acid stably express Foxp3 and can suppress even ongoing GVHD in mice. Foxp3+ regulatory T cells (T reg cells) effectively suppress immunity, but it is not determined if antigen-induced T reg cells (iT reg cells) are able to persist under conditions of inflammation and to stably express the transcription factor Foxp3. We used spleen cells to stimulate the mixed leukocyte reaction (MLR) in the presence of transforming growth factor β (TGF-β) and retinoic acid. We found that the CD11chigh dendritic cell fraction was the most potent at inducing high numbers of alloreactive Foxp3+ cells. The induced CD4+CD25+Foxp3+ cells appeared after extensive proliferation. When purified from the MLR, iT reg cells suppressed both primary and secondary MLR in vitro in an antigen-specific manner. After transfer into allogeneic mice, iT reg cells persisted for 6 mo and prevented graft versus host disease (GVHD) caused by co-transferred CD45RBhi T cells. Similar findings were made when iT reg cells were transferred after onset of GVHD. The CNS2 intronic sequence of the Foxp3 gene in the persisting iT reg cells was as demethylated as the corresponding sequence of naturally occurring T reg cells. These results indicate that induced Foxp3+ T reg cells, after proliferating and differentiating into antigen-specific suppressive T cells, can persist for long periods while suppressing a powerful inflammatory disease.
Collapse
Affiliation(s)
- Uri Sela
- Laboratory of Cellular Physiology and Immunology, Chris Browne Center for Immunology and Immune Disease, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
848
|
Procaccini C, Galgani M, De Rosa V, Matarese G. Intracellular metabolic pathways control immune tolerance. Trends Immunol 2011; 33:1-7. [PMID: 22075206 DOI: 10.1016/j.it.2011.09.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 08/11/2011] [Accepted: 09/06/2011] [Indexed: 11/16/2022]
Abstract
Disorders such as obesity and type 2 diabetes have been linked to immune dysfunction, raising the possibility that metabolic alterations can be induced by or be a consequence of alterations in immunological tolerance. Here, we describe how intracellular metabolic signalling pathways can 'sense' host energy/nutritional status, and in response, modulate regulatory T (Treg) cell function. In particular, we focus on mammalian target of rapamycin (mTOR) signalling, and how stimuli such as nutrients and leptin activate mTOR in an oscillatory manner to determine Treg cell proliferation status. We propose that metabolic changes such as nutritional deprivation or overload could dictate the characteristics of the Treg cell compartment and subsequent downstream immune reactions.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli 80131, Italy
| | | | | | | |
Collapse
|
849
|
Abstract
The discovery of regulatory T cells (Tregs) as a crucial component of peripheral down-regulation of immunity to self and allogeneic antigens has raised legitimate hope for the development of Treg-based clinical protocols for tolerance to allografts. The present review addresses the question of whether therapeutic Tregs are ready to enter the clinical transplantation arena. In light of recent experimental observations, we will revisit some fundamentals of T cell and Treg biology that stress the need for further studies prior to applications and provide conceptual cues for novel therapeutic approaches.
Collapse
Affiliation(s)
- Christian Leguern
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital East, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
850
|
Molecular mechanisms underlying the regulation and functional plasticity of FOXP3(+) regulatory T cells. Genes Immun 2011; 13:1-13. [PMID: 22048454 DOI: 10.1038/gene.2011.77] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CD4(+) CD25(+) regulatory T (Treg) cells engage in the maintenance of immunological self-tolerance and homeostasis by limiting aberrant or excessive inflammation. The transcription factor forkhead box P3 (FOXP3) is critical for the development and function of Treg cells. The differentiation of the Treg cell lineage is not terminal, as developmental and functional plasticity occur through the sensing of inflammatory signals in the periphery. Here, we review the recent progress in our understanding of the molecular mechanisms underlying the regulation and functional plasticity of CD4(+) CD25(+) FOXP3(+) Treg cells, through the perturbation of FOXP3 and its complex at a transcriptional, translational and post-translational level.
Collapse
|