1
|
Pant A, Jain A, Chen Y, Patel K, Saleh L, Tzeng S, Nitta RT, Zhao L, Wu CYJ, Bederson M, Wang WL, Bergsneider BHL, Choi J, Medikonda R, Verma R, Cho KB, Kim LH, Kim JE, Yazigi E, Lee SY, Rajendran S, Rajappa P, Mackall CL, Li G, Tyler B, Brem H, Pardoll DM, Lim M, Jackson CM. The CCR6-CCL20 Axis Promotes Regulatory T-cell Glycolysis and Immunosuppression in Tumors. Cancer Immunol Res 2024; 12:1542-1558. [PMID: 39133127 DOI: 10.1158/2326-6066.cir-24-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Regulatory T cells (Treg) are important players in the tumor microenvironment. However, the mechanisms behind their immunosuppressive effects are poorly understood. We found that CCR6-CCL20 activity in tumor-infiltrating Tregs is associated with greater glycolytic activity and ablation of Ccr6 reduced glycolysis and lactic acid production while increasing compensatory glutamine metabolism. Immunosuppressive activity toward CD8+ T cells was abrogated in Ccr6-/- Tregs due to reduction in activation-induced glycolysis. Furthermore, Ccr6-/- mice exhibited improved survival across multiple tumor models compared to wild-type mice and Treg and CD8+ T-cell depletion abrogated the improvement. In addition, Ccr6 ablation further promoted the efficacy of anti-PD-1 therapy in a preclinical glioma model. Follow-up knockdown of Ccl20 with siRNA also demonstrated improvement in antitumor efficacy. Our results unveil CCR6 as a marker and regulator of Treg-induced immunosuppression and identify approaches to target the metabolic determinants of Treg immunosuppressive activity.
Collapse
Affiliation(s)
- Ayush Pant
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aanchal Jain
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yiyun Chen
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | - Kisha Patel
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura Saleh
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephany Tzeng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Ryan T Nitta
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Liang Zhao
- Department of Oncology and Medicine, Bloomberg-Kimmel Institute for Immunotherapy, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Maria Bederson
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - William Lee Wang
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | | | - John Choi
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Ravi Medikonda
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Rohit Verma
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Kwang Bog Cho
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Lily H Kim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Jennifer E Kim
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eli Yazigi
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Si Yeon Lee
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Sakthi Rajendran
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Prajwal Rajappa
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Crystal L Mackall
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, California
| | - Gordon Li
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Betty Tyler
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Drew M Pardoll
- Department of Oncology and Medicine, Bloomberg-Kimmel Institute for Immunotherapy, the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, California
| | - Christopher M Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Jackson CM, Pant A, Dinalankara W, Choi J, Jain A, Nitta R, Yazigi E, Saleh L, Zhao L, Nirschl TR, Kochel CM, Hwa-Lin Bergsneider B, Routkevitch D, Patel K, Cho KB, Tzeng S, Neshat SY, Kim YH, Smith BJ, Ramello MC, Sotillo E, Wang X, Green JJ, Bettegowda C, Li G, Brem H, Mackall CL, Pardoll DM, Drake CG, Marchionni L, Lim M. The cytokine Meteorin-like inhibits anti-tumor CD8 + T cell responses by disrupting mitochondrial function. Immunity 2024; 57:1864-1877.e9. [PMID: 39111315 PMCID: PMC11324406 DOI: 10.1016/j.immuni.2024.07.003] [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: 08/15/2023] [Revised: 03/08/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024]
Abstract
Tumor-infiltrating lymphocyte (TIL) hypofunction contributes to the progression of advanced cancers and is a frequent target of immunotherapy. Emerging evidence indicates that metabolic insufficiency drives T cell hypofunction during tonic stimulation, but the signals that initiate metabolic reprogramming in this context are largely unknown. Here, we found that Meteorin-like (METRNL), a metabolically active cytokine secreted by immune cells in the tumor microenvironment (TME), induced bioenergetic failure of CD8+ T cells. METRNL was secreted by CD8+ T cells during repeated stimulation and acted via both autocrine and paracrine signaling. Mechanistically, METRNL increased E2F-peroxisome proliferator-activated receptor delta (PPARδ) activity, causing mitochondrial depolarization and decreased oxidative phosphorylation, which triggered a compensatory bioenergetic shift to glycolysis. Metrnl ablation or downregulation improved the metabolic fitness of CD8+ T cells and enhanced tumor control in several tumor models, demonstrating the translational potential of targeting the METRNL-E2F-PPARδ pathway to support bioenergetic fitness of CD8+ TILs.
Collapse
Affiliation(s)
- Christopher M Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Ayush Pant
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wikum Dinalankara
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Choi
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Aanchal Jain
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ryan Nitta
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Eli Yazigi
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura Saleh
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Liang Zhao
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas R Nirschl
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christina M Kochel
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Denis Routkevitch
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kisha Patel
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kwang Bog Cho
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Stephany Tzeng
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah Y Neshat
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA
| | - Young-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Barbara J Smith
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria Cecilia Ramello
- Center for Cell Therapy, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Elena Sotillo
- Center for Cell Therapy, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Jordan J Green
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gordon Li
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA
| | - Henry Brem
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Crystal L Mackall
- Center for Cell Therapy, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA; Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Drew M Pardoll
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Drake
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luigi Marchionni
- Bloomberg-Kimmel Institute for Immunotherapy, Departments of Oncology and Medicine, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Ivanova DL, Mundhenke TM, Gigley JP. The IL-12- and IL-23-Dependent NK Cell Response Is Essential for Protective Immunity against Secondary Toxoplasma gondii Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:2944-2958. [PMID: 31604804 DOI: 10.4049/jimmunol.1801525] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 09/17/2019] [Indexed: 12/22/2022]
Abstract
NK cells can develop cell-intrinsic memory-like characteristics. Whether they develop these characteristics during Toxoplasma gondii infection is unknown. We addressed this question and dissected the mechanisms involved in secondary NK cell responses using a vaccine-challenge mouse model of T. gondii infection. NK cells were required for control of and survival after secondary T. gondii infection. NK cells increased in number at the reinfection site and produced IFN-γ. To test if these T. gondii experienced NK cells were intrinsically different from naive NK cells, we performed NK cell adoptive transfer into RAG2/cγ-chain-/- mice, NK cell fate mapping, and RAG1-/- mice vaccine-challenge experiments. Although NK cells contributed to immunity after reinfection, they did not develop cell-intrinsic memory-like characteristics after T. gondii vaccination. The mechanisms required for generating these secondary NK cell responses were investigated. Secondary NK cell responses were CD4+ or CD8+ T cell independent. Although IL-12 alone is required for NK cell IFN-γ production during primary T. gondii infection, in the absence of IL-12 using IL-12p35-/- mice or anti-IL-12p70, secondary NK cell responses were only partially reduced after reinfection. IL-23 depletion with anti-IL-23p19 in vivo also significantly reduced the secondary NK cell response. IL-12 and IL-23 blockade with anti-IL-12p40 treatment completely eliminated secondary NK cell responses. Importantly, blockade of IL-12, IL-23, or both significantly reduced control of parasite reinfection and increased parasite burden. Our results define a previously unknown protective role for NK cells during secondary T. gondii infection that is dependent on IL-12 and IL-23.
Collapse
Affiliation(s)
- Daria L Ivanova
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | | | - Jason P Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| |
Collapse
|
4
|
Page E, Kwun J, Oh B, Knechtle S. Lymphodepletional strategies in transplantation. Cold Spring Harb Perspect Med 2013; 3:3/7/a015511. [PMID: 23818516 DOI: 10.1101/cshperspect.a015511] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Because lymphocytes were shown to mediate transplant rejection, their depletion has been studied as a mechanism of preventing rejection and perhaps inducing immunologic tolerance. Agents that profoundly deplete lymphocytes have included monoclonal antibodies, cytotoxic drugs, and radiation. We have studied several such agents but focused on antibodies that deplete not only peripheral blood lymphocytes, but also lymph node lymphocytes. Depletion of lymph node T lymphocytes appears to permit peripheral tolerance at least for T cells in animal models. Nevertheless, B-cell responses may be resistant to such approaches, and T memory cells are likewise relatively resistant to depleting antibodies. We review the experimental and clinical approaches to depletion strategies and outline some of the pitfalls of depletion, such as limitations of currently available agents, duration of tolerance, infection, and malignancy. It is notable that most tolerogenic strategies that have been attempted experimentally and clinically include depleting agents even when they are not named as the underlying strategy. Thus, there is an implicitly acknowledged role for reducing the precursor frequency of donor antigen-specific lymphocytes when approaching the daunting goal of transplant tolerance.
Collapse
Affiliation(s)
- Eugenia Page
- Department of Surgery, Division of Transplantation, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|
5
|
Côté AL, Byrne KT, Steinberg SM, Zhang P, Turk MJ. Protective CD8 memory T cell responses to mouse melanoma are generated in the absence of CD4 T cell help. PLoS One 2011; 6:e26491. [PMID: 22046294 PMCID: PMC3202545 DOI: 10.1371/journal.pone.0026491] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 09/28/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We have previously demonstrated that temporary depletion of CD4 T cells in mice with progressive B16 melanoma, followed by surgical tumor excision, induces protective memory CD8 T cell responses to melanoma/melanocyte antigens. We also showed that persistence of these CD8 T cells is supported, in an antigen-dependent fashion, by concurrent autoimmune melanocyte destruction. Herein we explore the requirement of CD4 T cell help in priming and maintaining this protective CD8 T cell response to melanoma. METHODOLOGY AND PRINCIPAL FINDINGS To induce melanoma/melanocyte antigen-specific CD8 T cells, B16 tumor bearing mice were depleted of regulatory T cells (T(reg)) by either temporary, or long-term continuous treatment with anti-CD4 (mAb clone GK1.5). Total depletion of CD4 T cells led to significant priming of IFN-γ-producing CD8 T cell responses to TRP-2 and gp100. Surprisingly, treatment with anti-CD25 (mAb clone PC61), to specifically deplete T(reg) cells while leaving help intact, was ineffective at priming CD8 T cells. Thirty to sixty days after primary tumors were surgically excised, mice completely lacking CD4 T cell help developed autoimmune vitiligo, and maintained antigen-specific memory CD8 T cell responses that were highly effective at producing cytokines (IFN-γ, TNF-α, and IL-2). Mice lacking total CD4 T cell help also mounted protection against re-challenge with B16 melanoma sixty days after primary tumor excision. CONCLUSIONS AND SIGNIFICANCE This work establishes that CD4 T cell help is dispensable for the generation of protective memory T cell responses to melanoma. Our findings support further use of CD4 T cell depletion therapy for inducing long-lived immunity to cancer.
Collapse
Affiliation(s)
- Anik L. Côté
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Katelyn T. Byrne
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Shannon M. Steinberg
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Peisheng Zhang
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Mary Jo Turk
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
6
|
Qu Y, Zhang B, Liu S, Zhang A, Wu T, Zhao Y. 2-Gy whole-body irradiation significantly alters the balance of CD4+ CD25- T effector cells and CD4+ CD25+ Foxp3+ T regulatory cells in mice. Cell Mol Immunol 2010; 7:419-27. [PMID: 20871628 PMCID: PMC4002961 DOI: 10.1038/cmi.2010.45] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 07/06/2010] [Accepted: 07/07/2010] [Indexed: 12/25/2022] Open
Abstract
CD4(+)CD25(+) T regulatory (Treg) cells are critical in inducing and maintaining immunological self-tolerance as well as transplant tolerance. The effect of low doses of whole-body irradiation (WBI) on CD4(+)CD25(+)Foxp3(+) Treg cells has not been determined. The proportion, phenotypes and function of CD4(+)CD25(+) Treg cells were investigated 0.5, 5 and 15 days after euthymic, thymectomized or allogeneic bone marrow transplanted C57BL/6 mice received 2-Gy γ-rays of WBI. The 2-Gy WBI significantly enhanced the ratios of CD4(+)CD25(+) Treg cells and CD4(+)CD25(+)Foxp3(+) Treg cells to CD4(+) T cells in peripheral blood, lymph nodes, spleens and thymi of mice. The CD4(+)CD25(+) Treg cells of the WBI-treated mice showed immunosuppressive activities on the immune response of CD4(+)CD25(-) T effector cells to alloantigens or mitogens as efficiently as the control mice. Furthermore, 2-Gy γ-ray WBI significantly increased the percentage of CD4(+)CD25(+)Foxp3(+) Treg cells in the periphery of either thymectomized mice or allogeneic bone marrow transplanted mice. The in vitro assay showed that ionizing irradiation induced less cell death in CD4(+)CD25(+)Foxp3(+) Treg cells than in CD4(+)CD25(-) T cells. Thus, a low dose of WBI could significantly enhance the level of functional CD4(+)CD25(+)Foxp3(+) Treg cells in the periphery of naive or immunized mice. The enhanced proportion of CD4(+)CD25(+)Foxp3(+) Treg cells in the periphery by a low dose of WBI may make hosts more susceptible to immune tolerance induction.
Collapse
Affiliation(s)
- Yanyan Qu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | |
Collapse
|
7
|
Zhang C, Shan J, Feng L, Lu J, Xiao Z, Luo L, Li C, Guo Y, Li Y. The effects of immunosuppressive drugs on CD4(+) CD25(+) regulatory T cells: a systematic review of clinical and basic research. J Evid Based Med 2010; 3:117-29. [PMID: 21349053 DOI: 10.1111/j.1756-5391.2010.01083.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To review the effects of different immunosuppressive drugs on proliferation and function of regulatory T cells (Tregs). METHODS We searched MEDLINE, Embase (from inception to September 2009), and the Cochrane Library (Issue 4, 2009) for clinical and basic research about the effects of various immunosuppressive drugs on Tregs. Data were extracted and methodological quality was assessed by two independent reviewers. Outcome measures for clinical research included blood Tregs levels, acute rejection episodes, and graft function. Outcomes for basic research included percentage of Tregs proliferation, function, Tregs phenotype, and evidence for possible mechanisms. We analyzed data qualitatively. RESULTS Forty-two studies, including 19 clinical trials and 23 basic studies, were included. The immunosuppressive drugs studied were calcineurin inhibitors (CNIs), Rapa, anti-metabolism drugs, IL-2 receptor-blocking antibodies, T-cell depleting antibodies, and co-stimulation blockade antibodies. Most of the studies were on Rapa and CNIs. Eight basic studies on Rapa and CNIs showed that Rapa could promote the proliferation and function of Tregs, while CNIs could not. Five clinical trials involving a total of 158 patients showed that patients taking Rapa had higher blood concentration of Tregs than patients taking CNIs, but no difference was found in graft function (6-42 months follow-up). CONCLUSION There is substantial evidence that Rapa favors Tregs survival and function. However, the higher numbers of blood Tregs in patients treated with Rapa do not show any association with better graft function. Larger clinical studies with longer follow-up are needed to more thoroughly assess the efficacy of immunosuppressive drugs on Tregs, and reveal whether a relationship exists between Tregs and graft function.
Collapse
Affiliation(s)
- Chuntao Zhang
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
The explosion of new discoveries in the field of immunology has provided new insights into mechanisms that promote an immune response directed against a transplanted organ. Central to the allograft response are T lymphocytes. This review summarizes the current literature on allorecognition, costimulation, memory T cells, T cell migration, and their role in both acute and chronic graft destruction. An in depth understanding of the cellular mechanisms that result in both acute and chronic allograft rejection will provide new strategies and targeted therapeutics capable of inducing long-lasting, allograft-specific tolerance.
Collapse
Affiliation(s)
- Elizabeth Ingulli
- Department of Pediatrics, University of California, San Diego, CA 92093, USA.
| |
Collapse
|
9
|
|
10
|
Park YH, Koo SK, Kim Y, Kim HM, Joe IY, Park CS, Kim SC, Han DJ, Lim DG. Effect of in vitroexpanded CD4(+)CD25(+)Foxp3(+) regulatory T cell therapy combined with lymphodepletion in murine skin allotransplantation. Clin Immunol 2009; 135:43-54. [PMID: 20006940 DOI: 10.1016/j.clim.2009.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 11/18/2009] [Accepted: 11/19/2009] [Indexed: 01/27/2023]
Abstract
A promising approach for preventing allograft rejection involves shifting the balance between cytopathic and regulatory T cells to dominance of the latter cell type. Nonspecific lymphodepletion was conducted by administration of depleting anti-CD4 and anti-CD8 antibodies to reduce effector T cells and adoptive transfer of ex vivo-expanded host Treg cells by stimulation with donor dendritic cells to augment the Treg cell compartment. Evaluation of an MHC-mismatched skin allograft model revealed that combined therapy with these two protocols consistently induced modest prolongation of allograft survival, although all skin grafts were eventually rejected. The administration of IL-2/anti-IL-2 complexes significantly improved the efficacy of combination therapy via promoting the expansion of adoptively transferred Treg cells as well as endogenous recipient Treg cells. We conclude that Treg cell therapy combined with lymphodepletion is of practical benefit for the control of allograft rejection, but requires supplementary measures to promote immune tolerance.
Collapse
Affiliation(s)
- Youn-Hee Park
- Asan Institute for Life Sciences, Ulsan University College of Medicine, Seoul 138-736, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
In vivo characterization of rabbit anti-mouse thymocyte globulin: a surrogate for rabbit anti-human thymocyte globulin. Transplantation 2009; 88:170-9. [PMID: 19623011 DOI: 10.1097/tp.0b013e3181abc061] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Polyclonal rabbit anti-human thymocyte globulin (Thymoglobulin) is used clinically for immunosuppression in solid organ transplantation; however, it is difficult to fully characterize the effects of this agent in humans. METHODS A surrogate rabbit anti-murine thymocyte globulin (mATG) was generated analogously to the commercial product Thymoglobulin and in vivo activities were evaluated, including pharmacokinetics, T-cell depletion, dose response and kinetics, depletion/sparing of T-cell subsets or other leukocyte populations, and depletion in different lymphoid organs. RESULTS Within 1 day, T cells are depleted by mATG in the blood, spleen, lymph node, and bone marrow down to doses of 1 mg/kg. Although mATG binds and depletes thymocytes in vitro, there is no thymocyte depletion in vivo at any dose level, suggesting decreased antibody accessibility to the thymus. After two doses of mATG given 3 days apart, T-cell reconstitution begins as early as day 9 and returns to basal levels by day 21 and 29 for CD4 and CD8 T cells, respectively. There is also preferential depletion of naïve T cells that results in increased ratios of regulatory and memory T cells within 1 day after mATG administration. Depletion of natural killer-T cells, natural killer cells, plasma cells, and plasmablasts occurs, but is modest and more transient compared with T cells. B cells, macrophages, dendritic cells, hematopoetic stem cells, and bone marrow stromal cells seem resistant to mATG depletion. CONCLUSIONS These studies characterize the depletive effects of mATG in normal mice and provide insight into mechanisms of action of Thymoglobulin.
Collapse
|
12
|
Current world literature. Curr Opin Organ Transplant 2009; 14:103-11. [PMID: 19337155 DOI: 10.1097/mot.0b013e328323ad31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Zhang B, Zhang A, Qu Y, Liu J, Niu Z, Zhao Y. Development of mouse CD4+CD25+Foxp3+ regulatory T cells in xenogeneic pig thymic grafts. Transpl Immunol 2009; 20:180-5. [DOI: 10.1016/j.trim.2008.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 09/01/2008] [Accepted: 09/10/2008] [Indexed: 02/07/2023]
|
14
|
Fudaba Y, Onoe T, Chittenden M, Shimizu A, Shaffer JM, Bronson R, Sykes M. Abnormal regulatory and effector T cell function predispose to autoimmunity following xenogeneic thymic transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7649-59. [PMID: 19017953 PMCID: PMC2673578 DOI: 10.4049/jimmunol.181.11.7649] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Porcine thymus grafts support robust murine and human thymopoiesis, generating a diverse T cell repertoire that is deleted of donor and host-reactive cells, achieving specific xenograft tolerance. Positive selection is mediated exclusively by the xenogeneic thymic MHC. Although thymectomized, T cell-depleted normal mice usually remain healthy following xenogeneic thymic transplantation, thymus-grafted congenitally athymic mice frequently develop multiorgan autoimmunity. We investigated the etiology of this syndrome by adoptively transferring lymphocyte populations from fetal pig thymus-grafted BALB/c nude mice to secondary BALB/c nude recipients. Fetal pig thymus-grafted nude mice generated normal numbers of CD25(+)Foxp3(+)CD4 T cells, but these cells lacked the capacity to block autoimmunity. Moreover, thymocytes and peripheral CD4(+)CD25(-) cells from fetal pig thymus-grafted nude mice, but not those from normal mice, induced autoimmunity in nude recipients. Injection of thymic epithelial cells from normal BALB/c mice into fetal pig thymus grafts reduced autoimmunity and enhanced regulatory function of splenocytes. Our data implicate abnormalities in postthymic maturation, expansion, and/or survival of T cells positively selected by a xenogeneic MHC, as well as incomplete intrathymic deletion of thymocytes recognizing host tissue-specific Ags, in autoimmune pathogenesis. Regulatory cell function is enhanced and negative selection of host-specific thymocytes may potentially also be improved by coimplantation of recipient thymic epithelial cells in the thymus xenograft.
Collapse
Affiliation(s)
- Yasuhiro Fudaba
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Takashi Onoe
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Meredith Chittenden
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Akira Shimizu
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Juanita M. Shaffer
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Roderick Bronson
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Megan Sykes
- Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Abstract
T-cell depletion strategies are an efficient therapy for the treatment of acute rejection after organ transplantation and have been successfully used as induction regimens. Although eliminating whole T cells blocks alloreactivity, this therapy challenges the development of regulatory mechanisms because it depletes regulatory cells and modifies the profile of T cells after homeostatic repopulation. Targeting T-cell subpopulations or selectively activated T cells, without modifying Treg cells, could constitute a pro-tolerogenic approach. However, the perfect molecular target that would be totally specific probably still needs to be identified. In this study, we have reviewed the biological activities of broad or specific T-cell depletion strategies as these contribute to the induction of regulatory cells and tolerance in organ transplantation.
Collapse
Affiliation(s)
- Thomas Haudebourg
- INSERM, U643, CHU Nantes, Institut de Transplantation et de Recherche en Transplantation, ITERT, Université de Nantes, Faculté de Médecine, Nantes, France
| | | | | |
Collapse
|