101
|
NAD(+) regulates Treg cell fate and promotes allograft survival via a systemic IL-10 production that is CD4(+) CD25(+) Foxp3(+) T cells independent. Sci Rep 2016; 6:22325. [PMID: 26928119 PMCID: PMC4772111 DOI: 10.1038/srep22325] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/26/2016] [Indexed: 12/13/2022] Open
Abstract
CD4+ CD25+ Foxp3+ Tregs have been shown to play a central role in immune homeostasis while preventing from fatal inflammatory responses, while Th17 cells have traditionally been recognized as pro-inflammatory mediators implicated in a myriad of diseases. Studies have shown the potential of Tregs to convert into Th17 cells, and Th17 cells into Tregs. Increasing evidence have pointed out CD25 as a key molecule during this transdifferentiation process, however molecules that allow such development remain unknown. Here, we investigated the impact of NAD+ on the fate of CD4+ CD25+ Foxp3+ Tregs in-depth, dissected their transcriptional signature profile and explored mechanisms underlying their conversion into IL-17A producing cells. Our results demonstrate that NAD+ promotes Treg conversion into Th17 cells in vitro and in vivo via CD25 cell surface marker. Despite the reduced number of Tregs, known to promote homeostasis, and an increased number of pro-inflammatory Th17 cells, NAD+ was able to promote an impressive allograft survival through a robust systemic IL-10 production that was CD4+ CD25+ Foxp3+ independent. Collectively, our study unravels a novel immunoregulatory mechanism of NAD+ that regulates Tregs fate while promoting allograft survival that may have clinical applications in alloimmunity and in a wide spectrum of inflammatory conditions.
Collapse
|
102
|
Glycogen synthase kinase 3β inhibition promotes human iTreg differentiation and suppressive function. Immunol Res 2016; 62:60-70. [PMID: 25752456 DOI: 10.1007/s12026-015-8635-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced regulatory T cells (iTregs) are essential to maintain immunological tolerance, immune homeostasis and prevention of autoimmunity. Some studies suggest that glycogen synthase kinase 3β (GSK3β) is involved in the mouse iTreg differentiation; however, whether GSK3β inhibits or enhances iTreg differentiation is still a matter of controversy. To address this issue, we have utilized human naïve CD4(+) T cells and investigated whether GSK3 activity changes during iTreg differentiation and whether altering GSK3 activity influences the development of iTregs and its suppressive function. As a constitutively activated kinase, during iTreg differentiation GSK3β became quickly deactivated (phosphorylated at serine 9), which is dependent on MAPK pathway rather than PI3-kinase/Akt pathway. Our results indicated that inhibition of GSK3β by specific inhibitors, SB216763 or TDZD-8, promoted the differentiation of iTreg and increased their suppressive activity. In contrast, overexpression of GSK3β significantly inhibited iTreg differentiation. Furthermore, GSK3β inhibition enhanced iTreg differentiation through the TGF-β/Smad3 pathway. Taken together, this study demonstrates that inhibition of GSK3β enhances human iTreg differentiation and its suppressive activity, and provides a rationale to target GSK3β as a novel immunotherapeutic strategy.
Collapse
|
103
|
Harnessing the plasticity of CD4(+) T cells to treat immune-mediated disease. Nat Rev Immunol 2016; 16:149-63. [PMID: 26875830 DOI: 10.1038/nri.2015.18] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD4(+) T cells differentiate and acquire distinct functions to combat specific pathogens but can also adapt their functions in response to changing circumstances. Although this phenotypic plasticity can be potentially deleterious, driving immune pathology, it also provides important benefits that have led to its evolutionary preservation. Here, we review CD4(+) T cell plasticity by examining the molecular mechanisms that regulate it - from the extracellular cues that initiate and drive cells towards varying phenotypes, to the cytosolic signalling cascades that decipher these cues and transmit them into the cell and to the nucleus, where these signals imprint specific gene expression programmes. By understanding how this functional flexibility is achieved, we may open doors to new therapeutic approaches that harness this property of T cells.
Collapse
|
104
|
Pedros C, Duguet F, Saoudi A, Chabod M. Disrupted regulatory T cell homeostasis in inflammatory bowel diseases. World J Gastroenterol 2016; 22:974-995. [PMID: 26811641 PMCID: PMC4716049 DOI: 10.3748/wjg.v22.i3.974] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/02/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
In the gut, where billions of non-self-antigens from the food and the microbiota are present, the immune response must be tightly regulated to ensure both host protection against pathogenic microorganisms and the absence of immune-related pathologies. It has been well documented that regulatory T cells (Tregs) play a pivotal role in this context. Indeed, Tregs are able to prevent excessive inflammation, which can lead to the rupture of intestinal homeostasis observed in inflammatory bowel diseases (IBDs). Both the worldwide incidence and prevalence of such diseases have increased throughout the latter part of the 20th century. Therefore, it is crucial to understand how Tregs suppress the colitogenic immune cells to establish new treatments for patients suffering from IBDs. In this review, we will first summarize the results obtained in animal model studies that highlight the importance of Tregs in maintaining intestinal homeostasis and describe the specific suppressive mechanisms involved. Next, our current knowledge about Tregs contribution to human IBDs will be reviewed, as well as the current therapeutic perspective on using Tregs for clinical IBD treatment and the challenges that remain to be resolved to ensure both the safety and effectiveness of these therapies in targeting this critical immune-regulatory cell population.
Collapse
|
105
|
Zhang Y, Collier F, Naselli G, Saffery R, Tang MLK, Allen KJ, Ponsonby AL, Harrison LC, Vuillermin P. Cord blood monocyte–derived inflammatory cytokines suppress IL-2 and induce nonclassic “T
H
2-type” immunity associated with development of food allergy. Sci Transl Med 2016; 8. [DOI: 10.1126/scitranslmed.aad4322] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Infants who develop food allergy display hyperresponsive innate immunity at birth that promotes nonclassical T
H
2 differentiation.
Collapse
Affiliation(s)
- Yuxia Zhang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Fiona Collier
- Barwon Health, Geelong, Victoria 3220, Australia
- Deakin University, Geelong, Victoria 3216, Australia
| | - Gaetano Naselli
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Richard Saffery
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Murdoch Childrens Research Institute, Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Mimi LK. Tang
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Murdoch Childrens Research Institute, Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Katrina J. Allen
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Murdoch Childrens Research Institute, Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Anne-Louise Ponsonby
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Murdoch Childrens Research Institute, Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | - Leonard C. Harrison
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter Vuillermin
- The University of Melbourne, Parkville, Victoria 3010, Australia
- Barwon Health, Geelong, Victoria 3220, Australia
- Deakin University, Geelong, Victoria 3216, Australia
- Murdoch Childrens Research Institute, Royal Children’s Hospital, Parkville, Victoria 3052, Australia
| | | |
Collapse
|
106
|
Xu YJ, Li L, Chen Y, Fu B, Wu DS, Li XL, Zhao XL, Chen FP. Role of HMGB1 in regulation of STAT3 expression in CD4 + T cells from patients with aGVHD after allogeneic hematopoietic stem cell transplantation. Clin Immunol 2015; 161:278-83. [DOI: 10.1016/j.clim.2015.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 12/27/2022]
|
107
|
Nuclear matrix protein SMAR1 control regulatory T-cell fate during inflammatory bowel disease (IBD). Mucosal Immunol 2015; 8:1184-200. [PMID: 25993445 PMCID: PMC4762908 DOI: 10.1038/mi.2015.42] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/31/2015] [Indexed: 02/04/2023]
Abstract
Regulatory T (Treg) cells are essential for self-tolerance and immune homeostasis. Transcription factor Foxp3, a positive regulator of Treg cell differentiation, has been studied to some extent. Signal transducer and activator of transcription factor 3 (STAT3) is known to negatively regulate Foxp3. It is not clear how STAT3 is regulated during Treg differentiation. We show that SMAR1, a known transcription factor and tumor suppressor, is directly involved in maintaining Treg cell fate decision. T-cell-specific conditional knockdown of SMAR1 exhibits increased susceptibility towards inflammatory disorders, such as colitis. The suppressive function of Treg cells is compromised in the absence of SMAR1 leading to increased T helper type 17 (Th17) differentiation and inflammation. Compared with wild-type, the SMAR1(-/-) Treg cells showed increased susceptibility of inflammatory bowel disease in Rag1(-/ -) mice, indicating the role of SMAR1 in compromising Treg cell differentiation resulting in severe colitis. We show that SMAR1 negatively regulate STAT3 expression favoring Foxp3 expression and Treg cell differentiation. SMAR1 binds to the MAR element of STAT3 promoter, present adjacent to interleukin-6 response elements. Thus Foxp3, a major driver of Treg cell differentiation, is regulated by SMAR1 via STAT3 and a fine-tune balance between Treg and Th17 phenotype is maintained.
Collapse
|
108
|
MiR-125a-5p Decreases the Sensitivity of Treg cells Toward IL-6-Mediated Conversion by Inhibiting IL-6R and STAT3 Expression. Sci Rep 2015; 5:14615. [PMID: 26424054 PMCID: PMC4589732 DOI: 10.1038/srep14615] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 09/02/2015] [Indexed: 02/07/2023] Open
Abstract
The transcription factor FOXP3 is essential for the differentiation and function of regulatory T cells (Treg). It is established that the transcription factor GATA-3 is induced in Treg cells under inflammatory conditions. GATA-3 stabilizes FOXP3 levels to avoid the differentiation of Treg cells into inflammatory-like T cells. The IL-6 signal pathway influences the sensitivity of Treg cells towards instability. The mechanism of GATA-3 in regulating FOXP3 and its relation to the IL-6 pathway remains unclear. Here we report how miR-125a-5p plays an important role in regulating the conversion of Treg cells by IL-6. miR-125a-5p expression is low in Treg cells under steady state conditions and can be induced by GATA-3 to inhibit the expression of IL-6R and STAT3. This finding reveals a GATA3/miR-125a-5p/IL-6R and STAT3/FOXP3 regulatory pathway, which determines how Treg cells respond to inflammatory IL-6-rich conditions.
Collapse
|
109
|
O’Connor RA, Anderton SM. Inflammation-associated genes: risks and benefits to Foxp3+ regulatory T-cell function. Immunology 2015; 146:194-205. [PMID: 26190495 PMCID: PMC4582961 DOI: 10.1111/imm.12507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 01/10/2023] Open
Abstract
Foxp3(+) regulatory T (Treg) cells prevent the development of autoimmunity and immunopathology, as well as maintaining homeostasis and tolerance to commensal microorganisms. The suppressive activity of Treg cells is their defining characteristic, generating great interest in their therapeutic potential. However, suppressive and effector functions are not entirely exclusive. Considerable evidence points to the ability of supposedly anti-inflammatory Foxp3-expressing Treg cells to also express transcription factors that have been characterized as cardinal drivers of T effector cell function. We will consider the mounting evidence that Treg cells can function in non-suppressive capacities and review the impetus for this functional change, its relevance to developing immune and autoimmune responses and its significance to the development of Treg-based therapies.
Collapse
Affiliation(s)
- Richard A O’Connor
- MRC Centre for Inflammation Research, Centre for Multiple Sclerosis Research and Centre for Immunity Infection and Evolution, University of EdinburghEdinburgh, UK
| | - Stephen M Anderton
- MRC Centre for Inflammation Research, Centre for Multiple Sclerosis Research and Centre for Immunity Infection and Evolution, University of EdinburghEdinburgh, UK
| |
Collapse
|
110
|
Hsu P, Santner-Nanan B, Hu M, Skarratt K, Lee CH, Stormon M, Wong M, Fuller SJ, Nanan R. IL-10 Potentiates Differentiation of Human Induced Regulatory T Cells via STAT3 and Foxo1. THE JOURNAL OF IMMUNOLOGY 2015; 195:3665-74. [PMID: 26363058 DOI: 10.4049/jimmunol.1402898] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 08/05/2015] [Indexed: 12/18/2022]
Abstract
Foxp3(+) regulatory T cells (Tregs) play essential roles in maintaining the immune balance. Although the majority of Tregs are formed in the thymus, increasing evidence suggests that induced Tregs (iTregs) may be generated in the periphery from naive cells. However, unlike in the murine system, significant controversy exists regarding the suppressive capacity of these iTregs in humans, especially those generated in vitro in the presence of TGF-β. Although it is well known that IL-10 is an important mediator of Treg suppression, the action of IL-10 on Tregs themselves is less well characterized. In this article, we show that the presence of IL-10, in addition to TGF-β, leads to increased expansion of Foxp3(+) iTregs with enhanced CTLA-4 expression and suppressive capability, comparable to that of natural Tregs. This process is dependent on IL-10R-mediated STAT3 signaling, as supported by the lack of an IL-10 effect in patients with IL-10R deficiency and dominant-negative STAT3 mutation. Additionally, IL-10-induced inhibition of Akt phosphorylation and subsequent preservation of Foxo1 function are critical. These results highlight a previously unrecognized function of IL-10 in human iTreg generation, with potential therapeutic implications for the treatment of immune diseases, such as autoimmunity and allergy.
Collapse
Affiliation(s)
- Peter Hsu
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia; Department of Allergy and Immunology, Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia; and
| | - Brigitte Santner-Nanan
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia
| | - Mingjing Hu
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia
| | - Kristen Skarratt
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia
| | - Cheng Hiang Lee
- Department of Gastroenterology, Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia
| | - Michael Stormon
- Department of Gastroenterology, Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia
| | - Melanie Wong
- Department of Allergy and Immunology, Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia; and
| | - Stephen J Fuller
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia
| | - Ralph Nanan
- Charles Perkins Centre Nepean, The University of Sydney, Kingswood, New South Wales 2751, Australia;
| |
Collapse
|
111
|
Wang S, Charbonnier LM, Noval Rivas M, Georgiev P, Li N, Gerber G, Bry L, Chatila TA. MyD88 Adaptor-Dependent Microbial Sensing by Regulatory T Cells Promotes Mucosal Tolerance and Enforces Commensalism. Immunity 2015; 43:289-303. [PMID: 26231118 DOI: 10.1016/j.immuni.2015.06.014] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/20/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
Commensal microbiota promote mucosal tolerance in part by engaging regulatory T (Treg) cells via Toll-like receptors (TLRs). We report that Treg-cell-specific deletion of the TLR adaptor MyD88 resulted in deficiency of intestinal Treg cells, a reciprocal increase in T helper 17 (Th17) cells and heightened interleukin-17 (IL-17)-dependent inflammation in experimental colitis. It also precipitated dysbiosis with overgrowth of segmented filamentous bacteria (SFB) and increased microbial loads in deep tissues. The Th17 cell dysregulation and bacterial dysbiosis were linked to impaired anti-microbial intestinal IgA responses, related to defective MyD88 adaptor- and Stat3 transcription factor-dependent T follicular regulatory and helper cell differentiation in the Peyer's patches. These findings establish an essential role for MyD88-dependent microbial sensing by Treg cells in enforcing mucosal tolerance and maintaining commensalism by promoting intestinal Treg cell formation and anti-commensal IgA responses.
Collapse
Affiliation(s)
- Sen Wang
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Louis-Marie Charbonnier
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Magali Noval Rivas
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Georgiev
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ning Li
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Georg Gerber
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lynn Bry
- Center for Clinical and Translational Metagenomics, Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
112
|
Verhagen J, Wegner A, Wraith DC. Extra-thymically induced T regulatory cell subsets: the optimal target for antigen-specific immunotherapy. Immunology 2015; 145:171-81. [PMID: 25716063 DOI: 10.1111/imm.12458] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 12/13/2022] Open
Abstract
Antigen-specific immunotherapy aims to selectively restore tolerance to innocuous antigens in cases of autoimmune or allergic disease, without the need for general immune suppression. Although the principle of antigen-specific immunotherapy was discovered more than a century ago, its clinical application to date is limited, particularly in the control of autoimmunity. This has resulted mainly from a lack of in-depth understanding of the underlying mechanism. More recently, the differentiation of extra-thymically induced T regulatory (Treg) cell subsets has been shown to be instrumental in peripheral tolerance induction. Two main types of inducible Treg cells, interleukin-10-secreting or Foxp3(+) , have now been described, each with distinct characteristics and methods of therapeutic induction. It is crucial, therefore, to identify the suitability of either subset in the control of specific immune disorders. This review explores their natural function, the known mechanisms of therapeutic differentiation of either subset as well as their in vivo functionality and discusses new developments that may aid their use in antigen-specific immunotherapy, with a focus on autoimmune disease.
Collapse
Affiliation(s)
- Johan Verhagen
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | |
Collapse
|
113
|
Carniti C, Gimondi S, Vendramin A, Recordati C, Confalonieri D, Bermema A, Corradini P, Mariotti J. Pharmacologic Inhibition of JAK1/JAK2 Signaling Reduces Experimental Murine Acute GVHD While Preserving GVT Effects. Clin Cancer Res 2015; 21:3740-9. [PMID: 25977345 DOI: 10.1158/1078-0432.ccr-14-2758] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 05/01/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune-mediated graft-versus-tumor (GVT) effects can occur after allogeneic hematopoietic stem cell transplantation (HSCT), but GVT is tightly linked to its main complication, graft-versus-host disease (GVHD). Strategies aimed at modulating GVHD, while maintaining the GVT effect, are needed to improve the cure rate of transplant. Given the emerging role of Janus-activated kinase (JAK) signaling in lymphoproliferative and myeloproliferative diseases and its established function at dictating T-cell differentiation, we postulated that JAKs might be potential therapeutic targets through a pharmacologic approach. EXPERIMENTAL DESIGN We examined the effect of JAK1/JAK2 modulation by ruxolitinib in a mouse model of fully MHC mismatched bone marrow transplant comprising in vivo tumor inoculation. RESULTS JAK1/JAK2 inhibition by ruxolitinib improved both overall survival (P = 0.03) and acute GVHD pathologic score at target organs (P ≤ 0.001) of treated mice. In addition, treatment with ruxolitinib was associated with a preserved GVT effect, as evidenced by reduction of tumor burden (P = 0.001) and increase of survival time (P = 0.01). JAK1/JAK2 inhibition did not impair the in vivo acquisition of donor T-cell alloreactivity; this observation may account, at least in part, to the preserved GVT effect. Rather, JAK1/JAK2 inhibition of GVHD was associated with the modulation of chemokine receptor expression, which may have been one factor in the reduced infiltration of donor T cells in GVHD target organs. CONCLUSIONS These data provide further evidence that JAK inhibition represents a new and potentially clinically relevant approach to GVHD prevention.
Collapse
Affiliation(s)
- Cristiana Carniti
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Gimondi
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonio Vendramin
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Camilla Recordati
- Mouse and Animal Pathology Laboratory, Fondazione Filarete, Milan, Italy
| | - Davide Confalonieri
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Anisa Bermema
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Corradini
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy. Chair of Hematology, Università degli Studi di Milano, Milan, Italy
| | - Jacopo Mariotti
- Department of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
114
|
Pan W, Zhu S, Dai D, Liu Z, Li D, Li B, Gagliani N, Zheng Y, Tang Y, Weirauch MT, Chen X, Zhu W, Wang Y, Chen B, Qian Y, Chen Y, Fang J, Herbst R, Richman L, Jallal B, Harley JB, Flavell RA, Yao Y, Shen N. MiR-125a targets effector programs to stabilize Treg-mediated immune homeostasis. Nat Commun 2015; 6:7096. [PMID: 25963922 DOI: 10.1038/ncomms8096] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 04/02/2015] [Indexed: 12/19/2022] Open
Abstract
Although different autoimmune diseases show discrete clinical features, there are common molecular pathways intimately involved. Here we show that miR-125a is downregulated in peripheral CD4(+) T cells of human autoimmune diseases including systemic lupus erythematosus and Crohn's disease, and relevant autoimmune mouse models. miR-125a stabilizes both the commitment and immunoregulatory capacity of Treg cells. In miR-125a-deficient mice, the balance appears to shift from immune suppression to inflammation, and results in more severe pathogenesis of colitis and experimental autoimmune encephalomyelitis (EAE). The genome-wide target analysis reveals that miR-125a suppresses several effector T-cell factors including Stat3, Ifng and Il13. Using a chemically synthesized miR-125a analogue, we show potential to re-programme the immune homeostasis in EAE models. These findings point to miR-125a as a critical factor that controls autoimmune diseases by stabilizing Treg-mediated immune homeostasis.
Collapse
Affiliation(s)
- Wen Pan
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China [2] Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiaotong University School of Medicine, Shanghai 200025, China [3] Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Shu Zhu
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Dai Dai
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China [2] Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Zheng Liu
- MedImmune LLC, Gaithersburg, Maryland 20878, USA
| | - Dan Li
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200025, China
| | - Bin Li
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200025, China
| | - Nicola Gagliani
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Yunjiang Zheng
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Yuanjia Tang
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China [2] Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology (CAGE) and Divisions of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio 45229, USA
| | - Xiaoting Chen
- School of Electronic and Computing Systems, University of Cincinnati, Cincinnati, Ohio 45221, USA
| | - Wei Zhu
- MedImmune LLC, Gaithersburg, Maryland 20878, USA
| | - Yue Wang
- MedImmune LLC, Gaithersburg, Maryland 20878, USA
| | - Bo Chen
- MedImmune LLC, Gaithersburg, Maryland 20878, USA
| | - Youcun Qian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Yingxuan Chen
- Division of Gastroenterology and Hepathology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepathology, Renji Hospital, Shanghai Institute of Digestive Disease, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | | | | | | | - John B Harley
- 1] Center for Autoimmune Genomics and Etiology (CAGE) and Divisions of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio 45229, USA [2] US Department of Veterans Affairs Medical Center, Cincinnati, Ohio 45220, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Yihong Yao
- MedImmune LLC, Gaithersburg, Maryland 20878, USA
| | - Nan Shen
- 1] State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China [2] Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiaotong University School of Medicine, Shanghai 200025, China [3] Center for Autoimmune Genomics and Etiology (CAGE) and Divisions of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati Ohio 45229, USA [4] Collaborative Innovation Center of Systems Biomedicine, Shanghai Jiaotong University, Shanghai 200240, China
| |
Collapse
|
115
|
Reis BS, Lee K, Fanok MH, Mascaraque C, Amoury M, Cohn LB, Rogoz A, Dallner OS, Moraes-Vieira PM, Domingos AI, Mucida D. Leptin receptor signaling in T cells is required for Th17 differentiation. THE JOURNAL OF IMMUNOLOGY 2015; 194:5253-60. [PMID: 25917102 DOI: 10.4049/jimmunol.1402996] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/23/2015] [Indexed: 12/14/2022]
Abstract
The hormone leptin plays a key role in energy homeostasis, and the absence of either leptin or its receptor (LepR) leads to severe obesity and metabolic disorders. To avoid indirect effects and to address the cell-intrinsic role of leptin signaling in the immune system, we conditionally targeted LepR in T cells. In contrast with pleiotropic immune disorders reported in obese mice with leptin or LepR deficiency, we found that LepR deficiency in CD4(+) T cells resulted in a selective defect in both autoimmune and protective Th17 responses. Reduced capacity for differentiation toward a Th17 phenotype by lepr-deficient T cells was attributed to reduced activation of the STAT3 and its downstream targets. This study establishes cell-intrinsic roles for LepR signaling in the immune system and suggests that leptin signaling during T cell differentiation plays a crucial role in T cell peripheral effector function.
Collapse
Affiliation(s)
- Bernardo S Reis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Kihyun Lee
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Melania H Fanok
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Cristina Mascaraque
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Manal Amoury
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Lillian B Cohn
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Aneta Rogoz
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Olof S Dallner
- Laboratory of Molecular Genetics, The Rockefeller University, New York, NY 10065
| | - Pedro M Moraes-Vieira
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Medical Deaconess Center, Harvard Medical School, Boston, MA 02215; and
| | - Ana I Domingos
- Obesity Laboratory, The Gulbenkian Science Institute, 2780-156 Oeiras, Portugal
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065;
| |
Collapse
|
116
|
The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood 2015; 125:3183-92. [PMID: 25814531 DOI: 10.1182/blood-2014-10-606830] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/11/2015] [Indexed: 12/19/2022] Open
Abstract
Interleukin (IL)-33 binding to the receptor suppression of tumorigenicity 2 (ST2) produces pro-inflammatory and anti-inflammatory effects. Increased levels of soluble ST2 (sST2) are a biomarker for steroid-refractory graft-versus-host disease (GVHD) and mortality. However, whether sST2 has a role as an immune modulator or only as a biomarker during GVHD was unclear. We show increased IL-33 production by nonhematopoietic cells in the gastrointestinal (GI) tract in mice post-conditioning and patients during GVHD. Exogenous IL-33 administration during the peak inflammatory response worsened GVHD. Conversely, GVHD lethality and tumor necrosis factor-α production was significantly reduced in il33(-/-) recipients. ST2 was upregulated on murine and human alloreactive T cells and sST2 increased as experimental GVHD progressed. Concordantly, st2(-/-) vs wild-type (WT) donor T cells had a marked reduction in GVHD lethality and GI histopathology. Alloantigen-induced IL-18 receptor upregulation was lower in st2(-/-) T cells, and linked to reduced interferon-γ production by st2(-/-) vs WT T cells during GVHD. Blockade of IL-33/ST2 interactions during allogeneic-hematopoietic cell transplantation by exogenous ST2-Fc infusions had a marked reduction in GVHD lethality, indicating a role of ST2 as a decoy receptor modulating GVHD. Together, these studies point to the IL-33/ST2 axis as a novel and potent target for GVHD therapy.
Collapse
|
117
|
Sca1(+) mesenchymal stromal cells inhibit graft-versus-host disease in mice after bone marrow transplantation. Int Immunopharmacol 2015; 26:50-7. [PMID: 25773667 DOI: 10.1016/j.intimp.2015.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/16/2015] [Accepted: 03/02/2015] [Indexed: 01/07/2023]
Abstract
Mesenchymal stromal cells (MSCs) have therapeutic potential for the prevention and treatment of graft-versus-host disease (GVHD). However, MSCs comprise several subpopulations, which have not been individually assessed for their role in GVHD suppression. In this study, we assessed the immunosuppressive effect of bone-related Sca1(+) MSCs on acute GVHD in a MHC-mismatched mouse model of allogeneic hematopoietic stem cell transplantation (HCT). Our results showed that Sca1(+) MSCs decreased the severity of acute GVHD (aGVHD) and prolonged the survival period of allogeneic HCT recipients. This effect was exerted through lowered T lymphocyte infiltration in target organs and by inhibition of CD80/86 expression on host dendritic cells. Furthermore, the expression of cytotoxic T-lymphocyte antigen-4 (CTLA-4), a negative regulator of T cells, was elevated in the recipient splenocytes. In conclusion, bone-related Sca1(+) MSCs subpopulation suppressed GVHD and could be a novel treatment for acute GVHD.
Collapse
|
118
|
Persa E, Balogh A, Sáfrány G, Lumniczky K. The effect of ionizing radiation on regulatory T cells in health and disease. Cancer Lett 2015; 368:252-61. [PMID: 25754816 DOI: 10.1016/j.canlet.2015.03.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
Abstract
Treg cells are key elements of the immune system which are responsible for the immune suppressive phenotype of cancer patients. Interaction of Treg cells with conventional anticancer therapies might fundamentally influence cancer therapy response rates. Radiotherapy, apart from its direct tumor cell killing potential, has a contradictory effect on the antitumor immune response: it augments certain immune parameters, while it depresses others. Treg cells are intrinsically radioresistant due to reduced apoptosis and increased proliferation, which leads to their systemic and/or intratumoral enrichment. While physiologically Treg suppression is not enhanced by irradiation, this is not the case in a tumorous environment, where Tregs acquire a highly suppressive phenotype, which is further increased by radiotherapy. This is the reason why the interest for combined radiotherapy and immunotherapy approaches focusing on the abrogation of Treg suppression has increased in cancer therapy in the last few years. Here we summarize the basic mechanisms of Treg radiation response both in healthy and cancerous environments and discuss Treg-targeted pre-clinical and clinical immunotherapy approaches used in combination with radiotherapy. Finally, the discrepant findings regarding the predictive value of Tregs in therapy response are also reviewed.
Collapse
Affiliation(s)
- Eszter Persa
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Andrea Balogh
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Géza Sáfrány
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Katalin Lumniczky
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary.
| |
Collapse
|
119
|
Basu R, Whitley SK, Bhaumik S, Zindl CL, Schoeb TR, Benveniste EN, Pear WS, Hatton RD, Weaver CT. IL-1 signaling modulates activation of STAT transcription factors to antagonize retinoic acid signaling and control the TH17 cell-iTreg cell balance. Nat Immunol 2015; 16:286-95. [PMID: 25642823 PMCID: PMC4790724 DOI: 10.1038/ni.3099] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/07/2015] [Indexed: 02/06/2023]
Abstract
Interleukin 17 (IL-17)-producing helper T cells (TH17 cells) and CD4(+) inducible regulatory T cells (iTreg cells) emerge from an overlapping developmental program. In the intestines, the vitamin A metabolite retinoic acid (RA) is produced at steady state and acts as an important cofactor to induce iTreg cell development while potently inhibiting TH17 cell development. Here we found that IL-1 was needed to fully override RA-mediated expression of the transcription factor Foxp3 and induce protective TH17 cell responses. By repressing expression of the negative regulator SOCS3 dependent on the transcription factor NF-κB, IL-1 increased the amplitude and duration of phosphorylation of the transcription factor STAT3 induced by TH17-polarizing cytokines, which led to an altered balance in the binding of STAT3 and STAT5 to shared consensus sequences in developing T cells. Thus, IL-1 signaling modulated STAT activation downstream of cytokine receptors differently to control the TH17 cell-iTreg cell developmental fate.
Collapse
Affiliation(s)
- Rajatava Basu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sarah K Whitley
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suniti Bhaumik
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Carlene L Zindl
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Trenton R Schoeb
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Warren S Pear
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robin D Hatton
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
120
|
Zheng Y, Wang Z, Deng L, Zhang G, Yuan X, Huang L, Xu W, Shen L. Modulation of STAT3 and STAT5 activity rectifies the imbalance of Th17 and Treg cells in patients with acute coronary syndrome. Clin Immunol 2015; 157:65-77. [DOI: 10.1016/j.clim.2014.12.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/28/2014] [Accepted: 12/19/2014] [Indexed: 01/09/2023]
|
121
|
The interplay of effector and regulatory T cells in cancer. Curr Opin Immunol 2015; 33:101-11. [PMID: 25728990 DOI: 10.1016/j.coi.2015.02.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/19/2015] [Accepted: 02/06/2015] [Indexed: 01/05/2023]
Abstract
Regulatory T (Treg) cells suppress effector T (Teff) cells and prevent immune-mediated rejection of cancer. Much less appreciated are mechanisms by which Teff cells antagonize Treg cells. Herein, we consider how complex reciprocal interactions between Teff and Treg cells shape their population dynamics within tumors. Under states of tolerance, including during tumor escape, suppressed Teff cells support Treg cell populations through antigen-dependent provision of interleukin (IL)-2. During immune activation, Teff cells can lose this supportive capacity and directly antagonize Treg cell populations to neutralize their immunosuppressive function. While this latter state is rarely achieved spontaneously within tumors, we propose that therapeutic induction of immune activation has the potential to stably disrupt immunosuppressive population states resulting in durable cancer regression.
Collapse
|
122
|
Hsiao HW, Hsu TS, Liu WH, Hsieh WC, Chou TF, Wu YJ, Jiang ST, Lai MZ. Deltex1 antagonizes HIF-1α and sustains the stability of regulatory T cells in vivo. Nat Commun 2015; 6:6353. [PMID: 25695215 PMCID: PMC4346631 DOI: 10.1038/ncomms7353] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 01/22/2015] [Indexed: 12/21/2022] Open
Abstract
Application of regulatory T cells (Tregs) in transplantation, autoimmunity and allergy has been extensively explored, but how Foxp3 and Treg stability is regulated in vivo is incompletely understood. Here, we identify a requirement for Deltex1 (DTX1), a contributor to T-cell anergy and Foxp3 protein level maintenance in vivo. Dtx1(-/-) Tregs are as effective as WT Tregs in the inhibition of CD4(+)CD25(-) T-cell activation in vitro. However, the suppressive ability of Dtx1(-/-) Tregs is greatly impaired in vivo. We find that Foxp3 expression is diminished when Dtx1(-/-) Tregs are co-transferred with effector T cells in vivo. DTX1 promotes the degradation of HIF-1α. Knockout of HIF-1α restores the Foxp3 stability and rescues the defective suppressive activity in Dtx1(-/-) Treg cells in vivo. Our results suggest that DTX1 exerts another level of control on Treg stability in vivo by sustaining the expression of Foxp3 protein in Tregs.
Collapse
Affiliation(s)
- Huey-Wen Hsiao
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Tzu-Sheng Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| | - Wen-Hsien Liu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Wan-Chen Hsieh
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Ting-Fang Chou
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
| | - Yu-Jung Wu
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| | - Si-Tse Jiang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan 74147, Taiwan, R.O.C
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan, R.O.C
- Graduate Institute of Immunology, National Taiwan University, Taipei 10057, Taiwan, R.O.C
| |
Collapse
|
123
|
Betts BC, Sagatys EM, Veerapathran A, Lloyd MC, Beato F, Lawrence HR, Yue B, Kim J, Sebti SM, Anasetti C, Pidala J. CD4+ T cell STAT3 phosphorylation precedes acute GVHD, and subsequent Th17 tissue invasion correlates with GVHD severity and therapeutic response. J Leukoc Biol 2015; 97:807-19. [PMID: 25663681 DOI: 10.1189/jlb.5a1114-532rr] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Th17 cells contribute to severe GVHD in murine bone marrow transplantation. Targeted deletion of the RORγt transcription factor or blockade of the JAK2-STAT3 axis suppresses IL-17 production and alloreactivity by Th17 cells. Here, we show that pSTAT3 Y705 is increased significantly in CD4(+) T cells among human recipients of allogeneic HCT before the onset of Grade II-IV acute GVHD. Examination of target-organ tissues at the time of GVHD diagnosis indicates that the amount of RORγt + Th17 cells is significantly higher in severe GVHD. Greater accumulation of tissue-resident Th17 cells also correlates with the use of MTX- compared with Rapa-based GVHD prophylaxis, as well as a poor therapeutic response to glucocorticoids. RORγt is optimally suppressed by concurrent neutralization of TORC1 with Rapa and inhibition of STAT3 activation with S3I-201, supporting that mTOR- and STAT3-dependent pathways converge upon RORγt gene expression. Rapa-resistant T cell proliferation can be totally inhibited by STAT3 blockade during initial allosensitization. We conclude that STAT3 signaling and resultant Th17 tissue accumulation are closely associated with acute GVHD onset, severity, and treatment outcome. Future studies are needed to validate the association of STAT3 activity in acute GVHD. Novel GVHD prevention strategies that incorporate dual STAT3 and mTOR inhibition merit investigation.
Collapse
Affiliation(s)
- Brian C Betts
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Elizabeth M Sagatys
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Anandharaman Veerapathran
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Mark C Lloyd
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Francisca Beato
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Harshani R Lawrence
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Binglin Yue
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Jongphil Kim
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Said M Sebti
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Claudio Anasetti
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Joseph Pidala
- Departments of *Blood and Marrow Transplantation, Immunology, Hematopathology and Laboratory Medicine, Drug Discovery, and Biostatistics, Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
124
|
Rottenberg ME, Carow B. SOCS3 and STAT3, major controllers of the outcome of infection with Mycobacterium tuberculosis. Semin Immunol 2014; 26:518-32. [DOI: 10.1016/j.smim.2014.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 01/04/2023]
|
125
|
Barbi J, Pardoll D, Pan F. Treg functional stability and its responsiveness to the microenvironment. Immunol Rev 2014; 259:115-39. [PMID: 24712463 DOI: 10.1111/imr.12172] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) prevent autoimmunity and tissue damage resulting from excessive or unnecessary immune activation through their suppressive function. While their importance for proper immune control is undeniable, the stability of the Treg lineage has recently become a controversial topic. Many reports have shown dramatic loss of the signature Treg transcription factor Forkhead box protein 3 (Foxp3) and Treg function under various inflammatory conditions. Other recent studies demonstrate that most Tregs are extremely resilient in their expression of Foxp3 and the retention of suppressive function. While this debate is unlikely to be settled in the immediate future, improved understanding of the considerable heterogeneity within the Foxp3(+) Treg population and how Treg subsets respond to ranging environmental cues may be keys to reconciliation. In this review, we discuss the diverse mechanisms responsible for the observed stability or instability of Foxp3(+) Treg identity and function. These include transcriptional and epigenetic programs, transcript targeting, and posttranslational modifications that appear responsive to numerous elements of the microenvironment. These mechanisms for Treg functional modulation add to the discussion of Treg stability.
Collapse
Affiliation(s)
- Joseph Barbi
- Department of Oncology, Immunology and Hematopoiesis Division, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
126
|
Isomäki P, Junttila I, Vidqvist KL, Korpela M, Silvennoinen O. The activity of JAK-STAT pathways in rheumatoid arthritis: constitutive activation of STAT3 correlates with interleukin 6 levels. Rheumatology (Oxford) 2014; 54:1103-13. [PMID: 25406356 DOI: 10.1093/rheumatology/keu430] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Many cytokines involved in RA activate the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathways. Therapeutic drugs that inhibit these pathways are being developed for RA. To investigate disease-related alterations in the activity of JAK-STAT pathways in RA, we studied the expression and activation of STAT1 and STAT3 in unstimulated and cytokine-stimulated cells and determined the levels of circulating cytokines. METHODS The expression of STAT1 and STAT3 mRNA in peripheral blood (PB) and SF T cells and monocytes was studied in RA patients and healthy volunteers by RT-PCR. Basal and cytokine (IFN-γ, IL-6, IL-10)-induced STAT phosphorylation was analysed in PB T cells and monocytes using multicolour flow cytometric analysis. RESULTS STAT3 mRNA levels were up-regulated in both PB and SF T cells and monocytes from RA patients. STAT1 expression was elevated in SF monocytes. The levels of phospho-STAT3 in resting PB T cells and monocytes were significantly higher in patients with RA than in healthy volunteers. IL-6 levels were elevated in RA plasma and correlated with the level of STAT3 phosphorylation in CD4(+) T cells and monocytes. IL-6-mediated STAT3 activation was deregulated in T cells from RA patients. IL-6-induced phosphorylation of STAT3 was decreased in CD4(+) T cells from patients with high plasma IL-6 levels and constitutive STAT3 phosphorylation. CONCLUSION The results suggest that IL-6 induces hyperactivation of STAT3 in circulating immune cells in active RA, and this subsequently desensitizes the IL-6 response in T cells.
Collapse
Affiliation(s)
- Pia Isomäki
- School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Ilkka Junttila
- School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Krista-Liisa Vidqvist
- School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Markku Korpela
- School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Olli Silvennoinen
- School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland School of Medicine, University of Tampere, Department of Internal Medicine, Centre for Rheumatic Diseases, Tampere University Hospital, Fimlab Laboratories and Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
127
|
Zhao K, Zhao D, Huang D, Yin L, Chen C, Pan B, Wu Q, Li Z, Yao Y, Shen E, Zeng L, Xu K. Interleukin-22 aggravates murine acute graft-versus-host disease by expanding effector T cell and reducing regulatory T cell. J Interferon Cytokine Res 2014; 34:707-15. [PMID: 24720737 DOI: 10.1089/jir.2013.0099] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Graft-versus-host disease (GVHD) as a major complication after allogeneic hematopoietic stem cell transplantation is not well prevented now. We have observed that interleukin-22 (IL-22) produced by Th22, Th1, and Th17 cells participated in GVHD development in our previous study. However, the role of IL-22 in GVHD is still ambiguous. The aim of this study was to illuminate the pathological or protective function and the potential mechanism of IL-22 in the GVHD process. In the present study, we found that compared with mice cotransferred with bone marrow and spleen cells (BS mice) without IL-22 administration, more serious tissue damage and higher GVHD clinical score were observed in BS+IL-22 mice. IL-22 administration was a benefit to early recovery of thymus after irradiation-induced injury. Administration of IL-22 could promote Th1 and Tc1 cell expansion in mesenteric lymph nodes but reduce CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cell number. Levels of systemic inflammatory cytokines (IFN-γ and TNF-α) were upregulated, while the level of immunosuppressive cytokine IL-10 was downregulated in recipients with IL-22 injection. In conclusion, IL-22, which exacerbates both local immune responses and systemic inflammation of recipients, plays a pathogenic role in the GVHD process. The potential mechanism of IL-22 in GVHD may attribute to increased alloreactive effector Th1 and Tc1 cells and decreased inhibitory Treg cell.
Collapse
Affiliation(s)
- Kai Zhao
- 1 Laboratory of Transplantation and Immunology, Xuzhou Medical College , Xuzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Combined with TCR stimuli, extracellular cytokine signals initiate the differentiation of naive CD4(+) T cells into specialized effector T-helper (Th) and regulatory T (Treg) cell subsets. The lineage specification and commitment process occurs through the combinatorial action of multiple transcription factors (TFs) and epigenetic mechanisms that drive lineage-specific gene expression programs. In this article, we review recent studies on the transcriptional and epigenetic regulation of distinct Th cell lineages. Moreover, we review current study linking immune disease-associated single-nucleotide polymorphisms with distal regulatory elements and their potential role in the disease etiology.
Collapse
Affiliation(s)
- Subhash K Tripathi
- Turku Centre for Biotechnology, University of Turku and
Åbo Akademi UniversityTurku, Finland
- National Doctoral Programme in Informational and
Structural BiologyTurku, Finland
- Turku Doctoral Programme of Molecular Medicine (TuDMM),
University of TurkuTurku, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and
Åbo Akademi UniversityTurku, Finland
| |
Collapse
|
129
|
Tilahun AY, Chowdhary VR, David CS, Rajagopalan G. Systemic inflammatory response elicited by superantigen destabilizes T regulatory cells, rendering them ineffective during toxic shock syndrome. THE JOURNAL OF IMMUNOLOGY 2014; 193:2919-30. [PMID: 25092888 DOI: 10.4049/jimmunol.1400980] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Life-threatening infections caused by Staphylococcus aureus, particularly the community-acquired methicillin-resistant strains of S. aureus, continue to pose serious problems. Greater virulence and increased pathogenicity of certain S. aureus strains are attributed to higher prevalence of exotoxins. Of these exotoxins, the superantigens (SAg) are likely most pathogenic because of their ability to rapidly and robustly activate the T cells even in extremely small quantities. Therefore, countering SAg-mediated T cell activation using T regulatory cells (Tregs) might be beneficial in diseases such as toxic shock syndrome (TSS). As the normal numbers of endogenous Tregs in a typical host are insufficient, we hypothesized that increasing the Treg numbers by administration of IL-2/anti-IL-2 Ab immune complexes (IL2C) or by adoptive transfer of ex vivo expanded Tregs might be more effective in countering SAg-mediated immune activation. HLA-DR3 transgenic mice that closely recapitulate human TSS were treated with IL2C to increase endogenous Tregs or received ex vivo expanded Tregs. Subsequently, they were challenged with SAg to induce TSS. Analyses of various parameters reflective of TSS (serum cytokine/chemokine levels, multiple organ pathology, and SAg-induced peripheral T cell expansion) indicated that increasing the Tregs failed to mitigate TSS. On the contrary, serum IFN-γ levels were increased in IL2C-treated mice. Exploration into the reasons behind the lack of protective effect of Tregs revealed IL-17 and IFN-γ-dependent loss of Tregs during TSS. In addition, significant upregulation of glucocorticoid-induced TNFR family-related receptor on conventional T cells during TSS could render them resistant to Treg-mediated suppression, contributing to failure of Treg-mediated immune regulation.
Collapse
Affiliation(s)
| | - Vaidehi R Chowdhary
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905; and
| | - Chella S David
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Govindarajan Rajagopalan
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
130
|
SOCS1 and regulation of regulatory T cells plasticity. J Immunol Res 2014; 2014:943149. [PMID: 25133199 PMCID: PMC4123481 DOI: 10.1155/2014/943149] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/10/2014] [Accepted: 06/30/2014] [Indexed: 01/07/2023] Open
Abstract
Several reports have suggested that natural regulatory T cells (Tregs) lose Forkhead box P3 (Foxp3) expression and suppression activity under certain inflammatory conditions. Treg plasticity has been studied because it may be associated with the pathogenesis of autoimmunity. Some studies showed that a minor uncommitted Foxp3+ T cell population, which lacks hypomethylation at Treg-specific demethylation regions (TSDRs), may convert to effector/helper T cells. Suppressor of cytokine signaling 1 (SOCS1), a negative regulator of cytokine signaling, has been reported to play an important role in Treg cell integrity and function by protecting the cells from excessive inflammatory cytokines. In this review, we discuss Treg plasticity and maintenance of suppression functions in both physiological and pathological settings. In addition, we discuss molecular mechanisms of maintaining Treg plasticity by SOCS1 and other molecules. Such information will be useful for therapy of autoimmune diseases and reinforcement of antitumor immunity.
Collapse
|
131
|
Rébé C, Végran F, Berger H, Ghiringhelli F. STAT3 activation: A key factor in tumor immunoescape. JAKSTAT 2014; 2:e23010. [PMID: 24058791 PMCID: PMC3670267 DOI: 10.4161/jkst.23010] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/21/2012] [Accepted: 11/27/2012] [Indexed: 12/28/2022] Open
Abstract
Cancer growth is controlled by cancer cells (cell intrinsic phenomenon), but also by the immune cells in the tumor microenvironment (cell extrinsic phenomenon). Thus cancer progression is mediated by the activation of transcription programs responsible for cancer cell proliferation, but also induced proliferation/activation of immunosuppressive cells such as Th17, Treg or myeloid derived suppressor cells (MDSCs). One of the key transcription factors involved in these pathways is the signal transducer and activator of transcription 3 (STAT3). In this review we will focus on STAT3 activation in immune cells, and how it impacts on tumor progression.
Collapse
Affiliation(s)
- Cédric Rébé
- INSERM, U866; Dijon, France ; Centre Georges François Leclerc; Dijon, France
| | | | | | | |
Collapse
|
132
|
MeCP2 enforces Foxp3 expression to promote regulatory T cells' resilience to inflammation. Proc Natl Acad Sci U S A 2014; 111:E2807-16. [PMID: 24958888 DOI: 10.1073/pnas.1401505111] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Forkhead box P3(+) (Foxp3(+)) regulatory T cells (Tregs) are crucial for peripheral tolerance. During inflammation, steady Foxp3 expression in Tregs is essential for maintaining their lineage identity and suppressive function. However, the molecular machinery governing Tregs' resilience to inflammation-induced Foxp3 destabilization remains elusive. Here, we demonstrate that methyl-CpG binding protein 2 (MeCP2), an eminent epigenetic regulator known primarily as the etiological factor of Rett syndrome, is critical to sustain Foxp3 expression in Tregs during inflammation. In response to inflammatory stimuli, MeCP2 is specifically recruited to the Conserved Non-Coding sequence 2 region of the foxp3 locus, where it collaborates with cAMP responsive element binding protein 1 to promote local histone H3 acetylation, thereby counteracting inflammation-induced epigenetic silencing of foxp3. Consequently, Treg-specific deletion of MeCP2 causes spontaneous immune activation in mice and failure in protection against autoimmunity. Furthermore, we demonstrate that Foxp3 expression in MeCP2-deficient Tregs diminishes with time, resulting in their failure to suppress effector T-cell-mediated colitis. Thus, MeCP2 serves as a critical safeguard that confers Tregs with resilience against inflammation.
Collapse
|
133
|
Next generation treatment of acute graft-versus-host disease. Leukemia 2014; 28:2283-91. [PMID: 24938648 DOI: 10.1038/leu.2014.195] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/23/2014] [Accepted: 06/04/2014] [Indexed: 02/07/2023]
Abstract
Despite rapid increase in the utilization of allogeneic hematopoietic stem cell transplantation, non-relapse mortality and sequela from acute graft-versus-host disease (GVHD) remain principle barriers. GVHD involves complex interactions between innate and adaptive immunity, culminating in tissue damage by inflammatory mediators and cellular effectors. Recently, our understanding of the molecular intricacies of GVHD has grown tremendously. New insights into the roles played by novel cytokines, chemokines, intracellular signaling pathways, epigenetics and post-translational modifications of proteins in GVHD biology provide numerous targets that might be therapeutically exploited. This review highlights recent advances and identifies opportunities for reshaping contemporary GVHD therapeutics.
Collapse
|
134
|
Yan S, Ding Y, Tian Y, Lu Z, Wang Y, Zhang Q, Ye Y, Zhou L, Xie H, Chen H, Zheng M, Zheng S. MHC-mismatched mice liver transplantation promotes tumor growth in liver graft. Cancer Lett 2014; 351:162-71. [PMID: 24880081 DOI: 10.1016/j.canlet.2014.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 04/19/2014] [Accepted: 05/11/2014] [Indexed: 12/18/2022]
Abstract
Liver transplantation is a final therapeutic option for treatment of hepatic malignancies, but local recurrence remains high after surgery. However, the underlying mechanisms of local tumor recurrence are still unknown. We speculated that immunological status of transplanted liver may contribute to the progress of tumor development. CT-26 tumor cells are injected into graft after allogeneic or syngeneic liver transplantation. The growth pattern of tumor and the co-relationship of regulatory T cell and effector T cells in liver graft were observed and investigated at 3d, 6d, 9d and 15d post-transplantation. The Hepatic Replacement Area of tumor in allogeneic grafts was significantly larger than that in syngeneic grafts. The activation of tumor growth in allografts was due to the dysfunction of effector T cells mediated by regulatory T cells in liver graft. Using nude mice model, we further confirmed that regulatory T cells from allograft significantly weaken the function of effector T cells in vivo. Our data has showed that MHC-mismatched mice liver transplantation can promote tumor growth in liver graft. For the first time, we demonstrated that susceptibility to tumor development in liver graft is due to the down-regulation of effector T cells' function mediated by the regulatory T cells.
Collapse
Affiliation(s)
- Sheng Yan
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yuan Ding
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yang Tian
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Zhongjie Lu
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yan Wang
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Qiyi Zhang
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yufu Ye
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Hui Chen
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Minghao Zheng
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Western Australia 6009, Australia.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China; Key Laboratory of Organ Transplantation Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| |
Collapse
|
135
|
Murase K, Kim HT, Bascug ORG, Kawano Y, Ryan J, Matsuoka KI, Davids MS, Koreth J, Ho VT, Cutler C, Armand P, Alyea EP, Blazar BR, Antin JH, Soiffer RJ, Letai A, Ritz J. Increased mitochondrial apoptotic priming of human regulatory T cells after allogeneic hematopoietic stem cell transplantation. Haematologica 2014; 99:1499-508. [PMID: 24859877 DOI: 10.3324/haematol.2014.104166] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
CD4 regulatory T cells play a critical role in establishment of immune tolerance and prevention of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. The recovery and maintenance of regulatory T cells is dependent on homeostatic factors including the generation of naïve regulatory T cells from hematopoietic precursor cells, the proliferation and expansion of mature regulatory T cells, and the survival of regulatory T cells in vivo. In this study, quantitation of mitochondrial apoptotic priming was used to compare susceptibility of regulatory T cells, conventional CD4 T cells and CD8 T cells to intrinsic pathway apoptosis in 57 patients after allogeneic hematopoietic stem cell transplantation and 25 healthy donors. In healthy donors, regulatory T cells are more susceptible to mitochondrial priming than conventional T cells. Mitochondrial priming is increased after hematopoietic stem cell transplantation in all T-cell subsets and particularly in patients with chronic graft-versus-host disease. Regulatory T cells express high levels of CD95 and are also more susceptible than conventional T cells to apoptosis through the extrinsic pathway. However, CD95 expression and extrinsic pathway apoptosis is not increased after hematopoietic stem cell transplantation. Decreased expression of BCL2 and increased expression of BIM, a mitochondrial cell death activator protein, in regulatory T cells contributes to increased mitochondrial priming in this T-cell subset but additional factors likely contribute to increased mitochondrial priming following hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Kazuyuki Murase
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA Harvard School of Public Health, Boston, MA, USA
| | - O R Gregory Bascug
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yutaka Kawano
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Jeremy Ryan
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | | | - Matthew S Davids
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - John Koreth
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Vincent T Ho
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Corey Cutler
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Philippe Armand
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Edwin P Alyea
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Joseph H Antin
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Robert J Soiffer
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Anthony Letai
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | - Jerome Ritz
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Medical School, Boston, MA, USA Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA Harvard Stem Cell Institute, Boston, MA, USA
| |
Collapse
|
136
|
Pan F, Barbi J. Ubiquitous points of control over regulatory T cells. J Mol Med (Berl) 2014; 92:555-69. [PMID: 24777637 DOI: 10.1007/s00109-014-1156-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/04/2014] [Accepted: 04/11/2014] [Indexed: 12/31/2022]
Abstract
Posttranslational modification by ubiquitin tagging is crucial for regulating the stability, activity and cellular localization of many target proteins involved in processes including DNA repair, cell cycle progression, protein quality control, and signal transduction. It has long been appreciated that ubiquitin-mediated events are important for certain signaling pathways leading to leukocyte activation and the stimulation of effector function. Now it is clear that the activities of molecules and pathways central to immune regulation are also modified and controlled by ubiquitin tagging. Among the mechanisms of immune control, regulatory T cells (or Tregs) are themselves particularly sensitive to such regulation. E3 ligases and deubiquitinases both influence Tregs through their effects on the signaling pathways pertinent to these cells or through the direct, posttranslational regulation of Foxp3. In this review, we will summarize and discuss several examples of ubiquitin-mediated control over multiple aspects of Treg biology including the generation, function and phenotypic fidelity of these cells. Fully explored and exploited, these potential opportunities for Treg modulation may lead to novel immunotherapies for both positive and negative fine-tuning of immune restraint.
Collapse
Affiliation(s)
- Fan Pan
- Immunology and Hematopoiesis Division, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA,
| | | |
Collapse
|
137
|
Hori S. Lineage stability and phenotypic plasticity of Foxp3+regulatory T cells. Immunol Rev 2014; 259:159-72. [DOI: 10.1111/imr.12175] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Shohei Hori
- Laboratory for Immune Homeostasis; RCAI; RIKEN Center for Integrative Medical Sciences; Kanagawa Japan
| |
Collapse
|
138
|
Abstract
Graft-versus-host-disease (GVHD) is a severe complication of allogeneic hematopoietic cell transplantation (allo-HCT) characterized by the production of high levels of proinflammatory cytokines. Activated Janus kinases (JAKs) are required for T-effector cell responses in different inflammatory diseases, and their blockade could potently reduce acute GVHD. We observed that inhibition of JAK1/2 signaling resulted in reduced proliferation of effector T cells and suppression of proinflammatory cytokine production in response to alloantigen in mice. In vivo JAK 1/2 inhibition improved survival of mice developing acute GVHD and reduced histopathological GVHD grading, serum levels of proinflammatory cytokines, and expansion of alloreactive luc-transgenic T cells. Mechanistically, we could show that ruxolitinib impaired differentiation of CD4(+) T cells into IFN-γ- and IL17A-producing cells, and that both T-cell phenotypes are linked to GVHD. Conversely, ruxolitinib treatment in allo-HCT recipients increased FoxP3(+) regulatory T cells, which are linked to immunologic tolerance. Based on these results, we treated 6 patients with steroid-refractory GVHD with ruxolitinib. All patients responded with respect to clinical GVHD symptoms and serum levels of proinflammatory cytokines. In summary, ruxolitinib represents a novel targeted approach in GVHD by suppression of proinflammatory signaling that mediates tissue damage and by promotion of tolerogenic Treg cells.
Collapse
|
139
|
Fu J, Heinrichs J, Yu XZ. Helper T-cell differentiation in graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Arch Immunol Ther Exp (Warsz) 2014; 62:277-301. [PMID: 24699629 DOI: 10.1007/s00005-014-0284-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 01/27/2014] [Indexed: 02/07/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective therapeutic option for many malignant diseases. However, the efficacy of allo-HSCT is limited by the occurrence of destructive graft-versus-host disease (GVHD). Since allogeneic T cells are the driving force in the development of GVHD, their activation, proliferation, and differentiation are key factors to understanding GVHD pathogenesis. This review focuses on one critical aspect: the differentiation and function of helper T (Th) cells in acute GVHD. We first summarize well-established subsets including Th1, Th2, Th17, and T-regulatory cells; their flexibility, plasticity, and epigenetic modification; and newly identified subsets including Th9, Th22, and T follicular helper cells. Next, we extensively discuss preclinical findings of Th-cell lineages in GVHD: the networks of transcription factors involved in differentiation, the cytokine and signaling requirements for development, the reciprocal differentiation features, and the regulation of microRNAs on T-cell differentiation. Finally, we briefly summarize the recent findings on the roles of T-cell subsets in clinical GVHD and ongoing strategies to modify T-cell differentiation for controlling GVHD in patients. We believe further exploration and understanding of the immunobiology of T-cell differentiation in GVHD will expand therapeutic options for the continuing success of allo-HSCT.
Collapse
Affiliation(s)
- Jianing Fu
- Cancer Biology PhD Program, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, 33612, USA
| | | | | |
Collapse
|
140
|
Abstract
The ability of adaptive immune system to protect higher vertebrates from pathogens resides in the ability of B and T cells to express different antigen specific receptors and to respond to different threats by activating distinct differentiation and/or activation pathways. In the past 10 years, the major role of epigenetics in controlling molecular mechanisms responsible for these peculiar features and, more in general, for lymphocyte development has become evident. KRAB-ZFPs is the widest family of mammalian transcriptional repressors, which function through the recruitment of the co-factor KRAB-Associated Protein 1 (KAP1) that in turn engages histone modifiers inducing heterochromatin formation. Although most of the studies on KRAB proteins have been performed in embryonic cells, more recent reports highlighted a relevant role for these proteins also in adult tissues. This article will review the role of KRAB-ZFP and KAP1 in the epigenetic control of mouse and human adaptive immune cells.
Collapse
|
141
|
Abstract
Mutations of STAT3 underlie the autosomal dominant form of hyperimmunoglobulin E syndrome (HIES). STAT3 has critical roles in immune cells and thus, hematopoietic stem cell transplantation (HSCT), might be a reasonable therapeutic strategy in this disease. However, STAT3 also has critical functions in nonhematopoietic cells and dissecting the protean roles of STAT3 is limited by the lethality associated with germline deletion of Stat3. Thus, predicting the efficacy of HSCT for HIES is difficult. To begin to dissect the importance of STAT3 in hematopoietic and nonhematopoietic cells as it relates to HIES, we generated a mouse model of this disease. We found that these transgenic mice recapitulate multiple aspects of HIES, including elevated serum IgE and failure to generate Th17 cells. We found that these mice were susceptible to bacterial infection that was partially corrected by HSCT using wild-type bone marrow, emphasizing the role played by the epithelium in the pathophysiology of HIES.
Collapse
|
142
|
MacDonald KPA, Le Texier L, Zhang P, Morris H, Kuns RD, Lineburg KE, Leveque L, Don AL, Markey KA, Vuckovic S, Bagger FO, Boyle GM, Blazar BR, Hill GR. Modification of T cell responses by stem cell mobilization requires direct signaling of the T cell by G-CSF and IL-10. THE JOURNAL OF IMMUNOLOGY 2014; 192:3180-9. [PMID: 24585878 DOI: 10.4049/jimmunol.1302315] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The majority of allogeneic stem cell transplants are currently undertaken using G-CSF mobilized peripheral blood stem cells. G-CSF has diverse biological effects on a broad range of cells and IL-10 is a key regulator of many of these effects. Using mixed radiation chimeras in which the hematopoietic or nonhematopoietic compartments were wild-type, IL-10(-/-), G-CSFR(-/-), or combinations thereof we demonstrated that the attenuation of alloreactive T cell responses after G-CSF mobilization required direct signaling of the T cell by both G-CSF and IL-10. IL-10 was generated principally by radio-resistant tissue, and was not required to be produced by T cells. G-CSF mobilization significantly modulated the transcription profile of CD4(+)CD25(+) regulatory T cells, promoted their expansion in the donor and recipient and their depletion significantly increased graft-versus-host disease (GVHD). In contrast, stem cell mobilization with the CXCR4 antagonist AMD3100 did not alter the donor T cell's ability to induce acute GVHD. These studies provide an explanation for the effects of G-CSF on T cell function and demonstrate that IL-10 is required to license regulatory function but T cell production of IL-10 is not itself required for the attenuation GVHD. Although administration of CXCR4 antagonists is an efficient means of stem cell mobilization, this fails to evoke the immunomodulatory effects seen during G-CSF mobilization. These data provide a compelling rationale for considering the immunological benefits of G-CSF in selecting mobilization protocols for allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Bailey-Bucktrout SL, Martinez-Llordella M, Zhou X, Anthony B, Rosenthal W, Luche H, Fehling HJ, Bluestone JA. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity 2014; 39:949-62. [PMID: 24238343 DOI: 10.1016/j.immuni.2013.10.016] [Citation(s) in RCA: 304] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 10/30/2013] [Indexed: 12/25/2022]
Abstract
Stable Foxp3 expression is crucial for regulatory T (Treg) cell function. We observed that antigen-driven activation and inflammation in the CNS promoted Foxp3 instability selectively in the autoreactive Treg cells that expressed high amounts of Foxp3 before experimental autoimmune encephalitis induction. Treg cells with a demethylated Treg-cell-specific demethylated region in the Foxp3 locus downregulated Foxp3 transcription in the inflamed CNS during the induction phase of the response. Stable Foxp3 expression returned at the population level with the resolution of inflammation or was rescued by IL-2-anti-IL-2 complex treatment during the antigen priming phase. Thus, a subset of fully committed self-antigen-specific Treg cells lost Foxp3 expression during an inflammatory autoimmune response and might be involved in inadequate control of autoimmunity. These results have important implications for Treg cell therapies and give insights into the dynamics of the Treg cell network during autoreactive CD4(+) T cell effector responses in vivo.
Collapse
|
144
|
Bonelli M, Shih HY, Hirahara K, Singelton K, Laurence A, Poholek A, Hand T, Mikami Y, Vahedi G, Kanno Y, O'Shea JJ. Helper T cell plasticity: impact of extrinsic and intrinsic signals on transcriptomes and epigenomes. Curr Top Microbiol Immunol 2014; 381:279-326. [PMID: 24831346 DOI: 10.1007/82_2014_371] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD4(+) helper T cells are crucial for autoimmune and infectious diseases; however, the recognition of the many, diverse fates available continues unabated. Precisely what controls specification of helper T cells and preserves phenotypic commitment is currently intensively investigated. In this review, we will discuss the major factors that impact helper T cell fate choice, ranging from cytokines and the microbiome to metabolic control and epigenetic regulation. We will also discuss the technological advances along with the attendant challenges presented by "big data," which allow the understanding of these processes on comprehensive scales.
Collapse
Affiliation(s)
- Michael Bonelli
- Molecular Immunology and Inflammation Branch, National Institutes of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Liu Y, Xu Y, Sun J, Ma A, Zhang F, Xia S, Xu G, Liu Y. AKT
hyperactivation confers a
T
h1 phenotype in thymic
T
reg cells deficient in
TGF
‐β receptor II signaling. Eur J Immunol 2013; 44:521-32. [DOI: 10.1002/eji.201243291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 09/30/2013] [Accepted: 10/25/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Yun Liu
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Yingqian Xu
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Jiabin Sun
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Aihui Ma
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Feng Zhang
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Suhua Xia
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Guiqin Xu
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiaotong University School of Medicine Shanghai China
| |
Collapse
|
146
|
Zhang P, Tey SK, Koyama M, Kuns RD, Olver SD, Lineburg KE, Lor M, Teal BE, Raffelt NC, Raju J, Leveque L, Markey KA, Varelias A, Clouston AD, Lane SW, MacDonald KPA, Hill GR. Induced regulatory T cells promote tolerance when stabilized by rapamycin and IL-2 in vivo. THE JOURNAL OF IMMUNOLOGY 2013; 191:5291-303. [PMID: 24123683 DOI: 10.4049/jimmunol.1301181] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Natural regulatory T cells (nTregs) play an important role in tolerance; however, the small numbers of cells obtainable potentially limit the feasibility of clinical adoptive transfer. Therefore, we studied the feasibility and efficacy of using murine-induced regulatory T cells (iTregs) for the induction of tolerance after bone marrow transplantation. iTregs could be induced in large numbers from conventional donor CD4 and CD8 T cells within 1 wk and were highly suppressive. During graft-versus-host disease (GVHD), CD4 and CD8 iTregs suppressed the proliferation of effector T cells and the production of proinflammatory cytokines. However, unlike nTregs, both iTreg populations lost Foxp3 expression within 3 wk in vivo, reverted to effector T cells, and exacerbated GVHD. The loss of Foxp3 in iTregs followed homeostatic and/or alloantigen-driven proliferation and was unrelated to GVHD. However, the concurrent administration of rapamycin, with or without IL-2/anti-IL-2 Ab complexes, to the transplant recipients significantly improved Foxp3 stability in CD4 iTregs (and, to a lesser extent, CD8 iTregs), such that they remained detectable 12 wk after transfer. Strikingly, CD4, but not CD8, iTregs could then suppress Teff proliferation and proinflammatory cytokine production and prevent GVHD in an equivalent fashion to nTregs. However, at high numbers and when used as GVHD prophylaxis, Tregs potently suppress graft-versus-leukemia effects and so may be most appropriate as a therapeutic modality to treat GVHD. These data demonstrate that CD4 iTregs can be produced rapidly in large, clinically relevant numbers and, when transferred in the presence of systemic rapamycin and IL-2, induce tolerance in transplant recipients.
Collapse
Affiliation(s)
- Ping Zhang
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Betts BC, Veerapathran A, Pidala J, Yu XZ, Anasetti C. STAT5 polarization promotes iTregs and suppresses human T-cell alloresponses while preserving CTL capacity. J Leukoc Biol 2013; 95:205-13. [PMID: 24068731 DOI: 10.1189/jlb.0313154] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alloreactivity negatively influences outcomes of organ transplantation or HCT from allogeneic donors. Standard pharmacologic immune suppression impairs T-cell function and jeopardizes the beneficial reconstitution of Tregs. Murine transplantation models have shown that STAT3 is highly expressed in alloreactive T cells and may be therapeutically targeted. The influence and effects of STAT3 neutralization in human alloreactivity, however, remain to be elucidated. In this study, S3I-201, a selective small-molecule inhibitor of STAT3, suppressed human DC-allosensitized T-cell proliferation and abrogated Th17 responses. STAT3 blockade significantly enhanced the expansion of potent iTregs and permitted CD8(+) cytolytic effector function. Mechanistically, S3I-201 polarized the ratio of STAT phosphorylation in favor of STAT5 over STAT3 and also achieved a significant degree of Foxp3 demethylation among the iTregs. Conversely, selective impairment of STAT5 phosphorylation with CAS 285986-31-4 markedly reduced iTregs. STAT3 represents a relevant target for achieving control over human alloresponses, where its suppression facilitates STAT5-mediated iTreg growth and function.
Collapse
|
148
|
Amarnath S. c-Rel in GVHD biology: a missing link. Eur J Immunol 2013; 43:2255-8. [PMID: 24037677 PMCID: PMC3860175 DOI: 10.1002/eji.201343924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 07/22/2013] [Accepted: 07/30/2013] [Indexed: 11/10/2022]
Abstract
Graft-versus-host disease (GVHD) is a major complication associated with allogeneic bone marrow transplantation (BMT). Recent advances in the treatment of lymphoid malignancies with BMT include exploring mechanisms that can inhibit GVHD while maintaining graft-versus-leukemic (GVL) effects. In this issue of the European Journal of Immunology, Yu et al. [Eur. J. Immunol. 2013.43: 2327-2337] demonstrate efficient separation of GVHD and GVL by abrogating c-Rel in T cells. Intrinsic c-Rel deficiency in T cells resulted in complete protection against GVHD in both major and minor histocompatibility mismatched murine models of BMT. Protection against GVHD was associated with a decreased presence of Th1 and Th17 cells with a concomitant increase in Treg-cell numbers. Interestingly, an intrinsic defect of c-Rel also resulted in decreased expression of the Th1-associated chemokine receptor CXCR3. Finally, the absence of c-Rel maintained GVL effects with significant tumor clearance in murine recipients. These data suggest that specific targeting of the T-cell-specific transcription factor c-Rel can inhibit GVHD while maintaining GVL effects.
Collapse
Affiliation(s)
- Shoba Amarnath
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
149
|
Fogli LK, Sundrud MS, Goel S, Bajwa S, Jensen K, Derudder E, Sun A, Coffre M, Uyttenhove C, Van Snick J, Schmidt-Supprian M, Rao A, Grunig G, Durbin J, Casola S, Casola SS, Rajewsky K, Koralov SB. T cell-derived IL-17 mediates epithelial changes in the airway and drives pulmonary neutrophilia. THE JOURNAL OF IMMUNOLOGY 2013; 191:3100-11. [PMID: 23966625 DOI: 10.4049/jimmunol.1301360] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Th17 cells are a proinflammatory subset of effector T cells that have been implicated in the pathogenesis of asthma. Their production of the cytokine IL-17 is known to induce local recruitment of neutrophils, but the direct impact of IL-17 on the lung epithelium is poorly understood. In this study, we describe a novel mouse model of spontaneous IL-17-driven lung inflammation that exhibits many similarities to asthma in humans. We have found that STAT3 hyperactivity in T lymphocytes causes an expansion of Th17 cells, which home preferentially to the lungs. IL-17 secretion then leads to neutrophil infiltration and lung epithelial changes, in turn leading to a chronic inflammatory state with increased mucus production and decreased lung function. We used this model to investigate the effects of IL-17 activity on airway epithelium and identified CXCL5 and MIP-2 as important factors in neutrophil recruitment. The neutralization of IL-17 greatly reduces pulmonary neutrophilia, underscoring a key role for IL-17 in promoting chronic airway inflammation. These findings emphasize the role of IL-17 in mediating neutrophil-driven pulmonary inflammation and highlight a new mouse model that may be used for the development of novel therapies targeting Th17 cells in asthma and other chronic pulmonary diseases.
Collapse
Affiliation(s)
- Laura K Fogli
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Mangus CW, Massey PR, Fowler DH, Amarnath S. Rapamycin resistant murine th9 cells have a stable in vivo phenotype and inhibit graft-versus-host reactivity. PLoS One 2013; 8:e72305. [PMID: 23991087 PMCID: PMC3749115 DOI: 10.1371/journal.pone.0072305] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/04/2013] [Indexed: 12/21/2022] Open
Abstract
The cytokine micro-environment can direct murine CD4+ T cells towards various differentiation lineages such as Th1, Th2 and Tregs even in the presence of rapamycin, which results in T cells that mediate increased in vivo effects. Recently, a new lineage of T cells known as Th9 cells that secrete increased IL-9 have been described. However, it is not known whether Th9 differentiation occurs in the presence of rapamycin or whether adoptively transferred donor Th9 cells would augment or restrict alloreactivity after experimental bone marrow transplantation. We found that CD4+ T cells that were co-stimulated and polarized with TGF-β and IL-4 in the presence or absence of rapamycin each yielded effector cells of Th9 phenotype that secreted increased IL-9 and expressed a transcription factor profile characteristic of both Th9 and Th2 cells (high GATA-3/low T-bet). Augmentation of T cell replete allografts with manufactured rapamycin resistant Th9 cells markedly reduced both CD4+ and CD8+ T cell engraftment and strongly inhibited allo-specific T cell secretion of IFN-γ. The potency of Th9 cell inhibition of alloreactivity was similar to that of rapamycin resistant Th2 cells. Importantly, rapamycin resistant Th9 cells persisted and maintained their cytokine phenotype, thereby indicating limited differentiation plasticity of the Th9 subset. As such, Th9 differentiation proceeds in the presence of rapamycin to generate a cell therapy product that maintains high IL-9 expression in vivo while inhibiting IFN-γ driven alloreactivity.
Collapse
Affiliation(s)
- Courtney W. Mangus
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Howard Hughes Medical Institute, National Institute of Health Research Scholars Program, Chevy Chase, Maryland, United States of America
| | - Paul R. Massey
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Daniel H. Fowler
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shoba Amarnath
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|