1
|
Wen S, Su X, Guo J, Ou Z, Wang L, Yue Z, Zhao J, Ran L, Hu J, Wang Y, Ran M, He Q, Ji P, Ye L, Chen Z, Xu L, Huang Q. Bcl6 controls the stability and suppressive function of regulatory T cells in head and neck squamous cell carcinoma. Genes Dis 2025; 12:101505. [PMID: 40290124 PMCID: PMC12033904 DOI: 10.1016/j.gendis.2024.101505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/16/2024] [Accepted: 12/02/2024] [Indexed: 04/30/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) ranks as the sixth most common cancer globally. Most studies in HNSCC demonstrated that regulatory T (Treg) cells confine the anti-tumor activity of effector T cells which may contribute to the immune escape and uncontrolled tumor progression. Here, we uncovered that the specific abrogation of Bcl6 in Treg cells resulted in significantly delayed malignant transformation of 4NQO-induced tumorigenesis. Bcl6 deficiency impairs the lineage stability of Treg cells by down-regulating the histone H3K4 trimethylation. Importantly, Bcl6 inhibition repressed the tumor growth of murine HNSCC and exhibited synergistic effects with immune checkpoint blockade therapy. These findings suggest that Bcl6 can be exploited as a promising therapeutic target for HNSCC treatment.
Collapse
Affiliation(s)
- Shuqiong Wen
- Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510055, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Junyi Guo
- Department of Stomatology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhanpeng Ou
- Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Lisha Wang
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Zhengliang Yue
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Ling Ran
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Jianjun Hu
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Yuzhu Wang
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Mengqu Ran
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing 400016, China
| | - Qinyi He
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lifan Xu
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Qizhao Huang
- Institute of Immunological Innovation and Translation, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
2
|
Shouse AN, Villarino AV, Malek TR. Interleukin-2 receptor signaling acts as a checkpoint that influences the distribution of regulatory T cell subsets. iScience 2024; 27:111248. [PMID: 39759017 PMCID: PMC11700635 DOI: 10.1016/j.isci.2024.111248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 01/07/2025] Open
Abstract
Regulatory T cells (Tregs) require IL-2 for survival in the periphery, yet how IL-2 shapes Treg heterogeneity remains poorly defined. Here we show that inhibition of IL-2R signaling in post-thymic Tregs leads to a preferential early loss of circulating Tregs (cTregs). Gene expression of cTregs was more dependent on IL-2R signaling than effector Tregs (eTregs). Unexpectedly, ablation of IL-2R signaling in cTregs resulted in increased proliferation, expression of eTreg genes, and enhanced capacity to develop into eTregs. Thus, IL-2R signaling normally acts as a checkpoint to maintain cTreg homeostasis while restraining their development into eTregs. Loss of IL-2R signaling also alters the distribution of eTreg subsets, with increased IFNγR1+ eTregs and CXCR5+ PD-1+ T follicular regulatory (TFR) cells but decreased intestinal RORγt+ TR17 cells. These changes lower eTreg suppressive function supporting expansion of IFNγ-secreting T effector cells. Thus, IL-2R signaling also safeguards Treg function and licenses differentiation of specialized eTregs.
Collapse
Affiliation(s)
- Acacia N. Shouse
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Alejandro V. Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas R. Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
3
|
Jamison BL, Lawrance M, Wang CJ, DeBerg HA, Ziegler LJ, Sansom DM, Gavin MA, Walker LSK, Campbell DJ. An IL-2 mutein increases regulatory T cell suppression of dendritic cells via IL-10 and CTLA-4 to promote T cell anergy. Cell Rep 2024; 43:114938. [PMID: 39488830 PMCID: PMC11602548 DOI: 10.1016/j.celrep.2024.114938] [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: 12/09/2023] [Revised: 08/15/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
Interleukin-2 (IL-2) variants with increased CD25 dependence that selectively expand Foxp3+ regulatory T (TR) cells are in clinical trials for treating inflammatory diseases. Using an Fc-fused IL-2 mutein (Fc.IL-2 mutein) we developed that prevents diabetes in non-obese diabetic (NOD) mice, we show that Fc.IL-2 mutein induced an activated TR population with elevated proliferation, a transcriptional program associated with Stat5- and T cell receptor-dependent gene modules, and high IL-10 and CTLA-4 expression. Increased IL-10 signaling limited surface major histocompatibility complex class II upregulation during conventional dendritic cell (cDC) maturation, while increased CTLA-4-dependent transendocytosis led to the transfer of CD80 and CD86 co-stimulatory ligands from maturing cDCs to TR cells. In NOD mice, Fc.IL-2 mutein treatment promoted the suppression of cDCs in the inflamed pancreas and pancreatic lymph nodes, resulting in T cell anergy. Thus, IL-2 mutein-expanded TR cells have enhanced functional properties and restrict cDC function, offering promise for targeted immunotherapy use in autoimmune disease.
Collapse
Affiliation(s)
- Braxton L Jamison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Matthew Lawrance
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Chun Jing Wang
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Hannah A DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Lauren J Ziegler
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - David M Sansom
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Marc A Gavin
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98126, USA
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London NW3 2PP, UK
| | - Daniel J Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98126, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 981098, USA.
| |
Collapse
|
4
|
Zhou Z, Xu J, Liu S, Lv Y, Zhang R, Zhou X, Zhang Y, Weng S, Xu H, Ba Y, Zuo A, Han X, Liu Z. Infiltrating treg reprogramming in the tumor immune microenvironment and its optimization for immunotherapy. Biomark Res 2024; 12:97. [PMID: 39227959 PMCID: PMC11373505 DOI: 10.1186/s40364-024-00630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Immunotherapy has shown promising anti-tumor effects across various tumors, yet it encounters challenges from the inhibitory tumor immune microenvironment (TIME). Infiltrating regulatory T cells (Tregs) are important contributors to immunosuppressive TIME, limiting tumor immunosurveillance and blocking effective anti-tumor immune responses. Although depletion or inhibition of systemic Tregs enhances the anti-tumor immunity, autoimmune sequelae have diminished expectations for the approach. Herein, we summarize emerging strategies, specifically targeting tumor-infiltrating (TI)-Tregs, that elevate the capacity of organisms to resist tumors by reprogramming their phenotype. The regulatory mechanisms of Treg reprogramming are also discussed as well as how this knowledge could be utilized to develop novel and effective cancer immunotherapies.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, China
| | - Jiaxin Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Human Anatomy, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yingying Lv
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
5
|
Shouse AN, LaPorte KM, Malek TR. Interleukin-2 signaling in the regulation of T cell biology in autoimmunity and cancer. Immunity 2024; 57:414-428. [PMID: 38479359 PMCID: PMC11126276 DOI: 10.1016/j.immuni.2024.02.001] [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: 11/22/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 05/26/2024]
Abstract
Interleukin-2 (IL-2) is a critical cytokine for T cell peripheral tolerance and immunity. Here, we review how IL-2 interaction with the high-affinity IL-2 receptor (IL-2R) supports the development and homeostasis of regulatory T cells and contributes to the differentiation of helper, cytotoxic, and memory T cells. A critical element for each T cell population is the expression of CD25 (Il2rα), which heightens the receptor affinity for IL-2. Signaling through the high-affinity IL-2R also reinvigorates CD8+ exhausted T (Tex) cells in response to checkpoint blockade. We consider the molecular underpinnings reflecting how IL-2R signaling impacts these various T cell subsets and the implications for enhancing IL-2-dependent immunotherapy of autoimmunity, other inflammatory disorders, and cancer.
Collapse
Affiliation(s)
- Acacia N Shouse
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Kathryn M LaPorte
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
6
|
Paterson CW, Fay KT, Chen CW, Klingensmith NJ, Gutierrez MB, Liang Z, Coopersmith CM, Ford ML. CTLA-4 Checkpoint Inhibition Improves Sepsis Survival in Alcohol-Exposed Mice. Immunohorizons 2024; 8:74-88. [PMID: 38226924 PMCID: PMC10835704 DOI: 10.4049/immunohorizons.2300060] [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: 11/20/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Chronic alcohol use increases morbidity and mortality in the setting of sepsis. Both chronic alcohol use and sepsis are characterized by immune dysregulation, including overexpression of T cell coinhibitory molecules. We sought to characterize the role of CTLA-4 during sepsis in the setting of chronic alcohol exposure using a murine model of chronic alcohol ingestion followed by cecal ligation and puncture. Results indicated that CTLA-4 expression is increased on CD4+ T cells isolated from alcohol-drinking septic mice as compared with either alcohol-drinking sham controls or water-drinking septic mice. Moreover, checkpoint inhibition of CTLA-4 improved sepsis survival in alcohol-drinking septic mice, but not water-drinking septic mice. Interrogation of the T cell compartments in these animals following pharmacologic CTLA-4 blockade, as well as following conditional Ctla4 deletion in CD4+ T cells, revealed that CTLA-4 deficiency promoted the activation and proliferation of effector regulatory T cells and the generation of conventional effector memory CD4+ T cells. These data highlight an important role for CTLA-4 in mediating mortality during sepsis in the setting of chronic alcohol exposure and may inform future approaches to develop targeted therapies for this patient population.
Collapse
Affiliation(s)
- Cameron W. Paterson
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
- Lieutenant, Medical Corps, Naval Reserve Officer Training Corp, United States Navy, Atlanta, GA
| | - Katherine T. Fay
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Ching-Wen Chen
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Nathan J. Klingensmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Melissa B. Gutierrez
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Zhe Liang
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Craig M. Coopersmith
- Department of Surgery, Emory Critical Care Center, Emory University School of Medicine, Atlanta GA
| | - Mandy L. Ford
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta GA
| |
Collapse
|
7
|
Jamison BL, Lawrance M, Wang CJ, DeBerg HA, Sansom DM, Gavin MA, Walker LS, Campbell DJ. An IL-2 mutein increases IL-10 and CTLA-4-dependent suppression of dendritic cells by regulatory T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569613. [PMID: 38106196 PMCID: PMC10723345 DOI: 10.1101/2023.12.01.569613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Interleukin-2 (IL-2) variants with increased CD25 dependence that selectively expand Foxp3+ regulatory T (TR) cells are in clinical trials for treating inflammatory diseases. Using an Fc-fused IL-2 mutein (Fc.IL-2 mutein) we developed that prevents diabetes in non-obese diabetic (NOD) mice, we show that Fc.IL-2 mutein induced an activated TR population with elevated proliferation, a transcriptional program associated with Stat5- and TCR-dependent gene modules, and high IL-10 and CTLA-4 expression. Increased IL-10 signaling limited surface MHC class II upregulation during conventional dendritic cell (cDC) maturation, while increased CTLA-4-dependent transendocytosis led to the transfer of CD80 and CD86 costimulatory ligands from maturing cDCs to TR cells. In NOD mice, Fc.IL-2 mutein treatment promoted the suppression of cDCs in the inflamed pancreas and pancreatic lymph nodes resulting in T cell anergy. Thus, IL-2 mutein-expanded TR cells have enhanced functional properties and restrict cDC function, offering promise for targeted immunotherapy use in autoimmune disease.
Collapse
Affiliation(s)
- Braxton L. Jamison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA
| | | | - Chun Jing Wang
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | | | - David M. Sansom
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | | | - Lucy S.K. Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Daniel J. Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA
- Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
8
|
Herppich S, Hoenicke L, Kern F, Kruse F, Smout J, Greweling-Pils MC, Geffers R, Burton OT, Liston A, Keller A, Floess S, Huehn J. Zfp362 potentiates murine colonic inflammation by constraining Treg cell function rather than promoting Th17 cell differentiation. Eur J Immunol 2023; 53:e2250270. [PMID: 37366299 DOI: 10.1002/eji.202250270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/02/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
Mucosal barrier integrity and pathogen clearance is a complex process influenced by both Th17 and Treg cells. Previously, we had described the DNA methylation profile of Th17 cells and identified Zinc finger protein (Zfp)362 to be uniquely demethylated. Here, we generated Zfp362-/- mice to unravel the role of Zfp362 for Th17 cell biology. Zfp362-/- mice appeared clinically normal, showed no phenotypic alterations in the T-cell compartment, and upon colonization with segmented filamentous bacteria, no effect of Zfp362 deficiency on Th17 cell differentiation was observed. By contrast, Zfp362 deletion resulted in increased frequencies of colonic Foxp3+ Treg cells and IL-10+ and RORγt+ Treg cell subsets in mesenteric lymph nodes. Adoptive transfer of naïve CD4+ T cells from Zfp362-/- mice into Rag2-/- mice resulted in a significantly lower weight loss when compared with controls receiving cells from Zfp362+/+ littermates. However, this attenuated weight loss did not correlate with alterations of Th17 cells but instead was associated with an increase of effector Treg cells in mesenteric lymph nodes. Together, these results suggest that Zfp362 plays an important role in promoting colonic inflammation; however, this function is derived from constraining the effector function of Treg cells rather than directly promoting Th17 cell differentiation.
Collapse
Affiliation(s)
- Susanne Herppich
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lisa Hoenicke
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Fabian Kern
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Saarland University, Saarbrücken, Germany
- Department of Clinical Bioinformatics, Saarland University, Homburg, Germany
| | - Friederike Kruse
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Justine Smout
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Oliver T Burton
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Adrian Liston
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Andreas Keller
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Saarland University, Saarbrücken, Germany
- Department of Clinical Bioinformatics, Saarland University, Homburg, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
9
|
Jovisic M, Mambetsariev N, Singer BD, Morales-Nebreda L. Differential roles of regulatory T cells in acute respiratory infections. J Clin Invest 2023; 133:e170505. [PMID: 37463441 PMCID: PMC10348770 DOI: 10.1172/jci170505] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Acute respiratory infections trigger an inflammatory immune response with the goal of pathogen clearance; however, overexuberant inflammation causes tissue damage and impairs pulmonary function. CD4+FOXP3+ regulatory T cells (Tregs) interact with cells of both the innate and the adaptive immune system to limit acute pulmonary inflammation and promote its resolution. Tregs also provide tissue protection and coordinate lung tissue repair, facilitating a return to homeostatic pulmonary function. Here, we review Treg-mediated modulation of the host response to respiratory pathogens, focusing on mechanisms underlying how Tregs promote resolution of inflammation and repair of acute lung injury. We also discuss potential strategies to harness and optimize Tregs as a cellular therapy for patients with severe acute respiratory infection and discuss open questions in the field.
Collapse
Affiliation(s)
- Milica Jovisic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
| | | | - Benjamin D. Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
- Department of Biochemistry and Molecular Genetics, and
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Simpson Querrey Lung Institute for Translational Science
| |
Collapse
|
10
|
Paterson CW, Gutierrez MB, Coopersmith CM, Ford ML. Impact of chronic alcohol exposure on conventional and regulatory murine T cell subsets. Front Immunol 2023; 14:1142614. [PMID: 37006296 PMCID: PMC10063870 DOI: 10.3389/fimmu.2023.1142614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction Chronic alcohol use poses significant negative consequences to public health and, among its many biologic effects, is associated with significant T cell dysregulation within the adaptive immune system that has yet to be fully characterized. Novel, automated strategies for high dimensional flow cytometric analysis of the immune system are rapidly improving researchers' ability to detect and characterize rare cell types. Methods Using a murine model of chronic alcohol ingestion in conjunction with viSNE and CITRUS analysis tools, we performed a machine-driven, exploratory analysis comparing rare splenic subpopulations within the conventional CD4+, regulatory CD4+ and CD8+ T cell compartments between alcohol- and water-fed animals. Results While there were no differences in the absolute numbers of bulk CD3+ T cells, bulk CD4+ T cells, bulk CD8+ T cells, Foxp3- CD4+ conventional T cells (Tconv) or Foxp3+ CD4+ regulatory T cells (Treg), we identified populations of naïve Helios+ CD4+Tconv and naïve CD103+ CD8+ splenic T cells that were decreased in chronically alcohol exposed mice versus water-fed controls. In addition, we identified increased CD69+ Treg and decreased CD103+ effector regulatory T cell (eTreg) subsets in conjunction with increased frequency of a population that may represent a transitional phenotype between central regulatory T cell (cTreg) and eTreg. Discussion These data provide further resolution into the character of decreased naïve T cell populations known to be present in alcohol exposed mice, as well as describe alterations in effector regulatory T cell phenotypes associated with the pathogenesis of chronic alcohol-induced immune dysfunction.
Collapse
Affiliation(s)
- Cameron W. Paterson
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Medical Corps, United States Navy, Navy Reserve Officer Training Corps (NROTC), Atlanta, GA, United States
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Melissa B. Gutierrez
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
11
|
Li Y, Li X, Geng X, Zhao H. The IL-2A receptor pathway and its role in lymphocyte differentiation and function. Cytokine Growth Factor Rev 2022; 67:66-79. [DOI: 10.1016/j.cytogfr.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/03/2022]
|
12
|
Abstract
The transforming growth factor-β (TGF-β) family includes cytokines controlling cell behavior, differentiation and homeostasis of various tissues including components of the immune system. Despite well recognized importance of TGF-β in controlling T cell functions, the immunomodulatory roles of many other members of the TGF-β cytokine family, especially bone morphogenetic proteins (BMPs), start to emerge. Bone Morphogenic Protein Receptor 1α (BMPR1α) is upregulated by activated effector and Foxp3+ regulatory CD4+ T cells (Treg cells) and modulates functions of both of these cell types. BMPR1α inhibits generation of proinflammatory Th17 cells and sustains peripheral Treg cells. This finding underscores the importance of the BMPs in controlling Treg cell plasticity and transition between Treg and Th cells. BMPR1α deficiency in in vitro induced and peripheral Treg cells led to upregulation of Kdm6b (Jmjd3) demethylase, an antagonist of polycomb repressive complex 2 (PRC2), and cell cycle inhibitor Cdkn1a (p21Cip1) promoting cell senescence. This indicates that BMPs and BMPR1α may represent regulatory modules shaping epigenetic landscape and controlling proinflammatory reprogramming of Th and Treg cells. Revealing functions of other BMP receptors and their crosstalk with receptors for TGF-β will contribute to our understanding of peripheral immunoregulation.
Collapse
Affiliation(s)
- Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| |
Collapse
|
13
|
Induction of Foxp3 and activation of Tregs by HSP gp96 for treatment of autoimmune diseases. iScience 2021; 24:103445. [PMID: 34877502 PMCID: PMC8633978 DOI: 10.1016/j.isci.2021.103445] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/16/2021] [Accepted: 11/10/2021] [Indexed: 01/06/2023] Open
Abstract
Upregulation and stabilization of Foxp3 expression in Tregs are essential for regulating Treg function and immune homeostasis. In this study, gp96 immunization showed obvious therapeutic effects in a Lyn–/– mouse model of systemic lupus erythematosus. Moreover, gp96 alleviated the initiation and progression of MOG-induced experimental autoimmune encephalomyelitis. Immunization of gp96 increased Treg frequency, expansion, and suppressive function. Gene expression profiling identified the NF-κB family member p65 and c-Rel as the key transcription factors for enhanced Foxp3 expression in Treg by gp96. Mutant gp96 within its Toll-like receptor (TLR) binding domain, TLR2 knockout mice, and mice with cell-specific deletion of MyD88, were used to demonstrate that gp96 activated Tregs and induced Foxp3 expression via a TLR2-MyD88-mediated NF-κB signaling pathway. Taken together, these results show that gp96 immunization restricted antibody-induced and Th-induced autoimmune diseases by integrating Treg expansion and activation, indicating its potential clinical usefulness against autoimmune diseases. SLE symptoms in Lyn–/– mice are ameliorated by gp96 immunization Tregs expanded by gp96 provide potential in suppressing Th-mediated EAE Gp96 promotes Treg proliferation, stability, and suppressive function Gp96 binds to and activates Treg in a TLR2-MyD88-NF-кB-Foxp3 pathway
Collapse
|
14
|
Browning LM, Miller C, Kuczma M, Pietrzak M, Jing Y, Rempala G, Muranski P, Ignatowicz L, Kraj P. Bone Morphogenic Proteins Are Immunoregulatory Cytokines Controlling FOXP3 + T reg Cells. Cell Rep 2021; 33:108219. [PMID: 33027660 DOI: 10.1016/j.celrep.2020.108219] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 07/28/2020] [Accepted: 09/10/2020] [Indexed: 12/23/2022] Open
Abstract
Bone morphogenic proteins (BMPs) are members of the transforming growth factor β (TGF-β) cytokine family promoting differentiation, homeostasis, and self-renewal of multiple tissues. We show that signaling through the bone morphogenic protein receptor 1α (BMPR1α) sustains expression of FOXP3 in Treg cells in peripheral lymphoid tissues. BMPR1α signaling promotes molecular circuits supporting acquisition and preservation of Treg cell phenotype and inhibiting differentiation of pro-inflammatory effector Th1/Th17 CD4+ T cell. Mechanistically, increased expression of KDM6B (JMJD3) histone demethylase, an antagonist of the polycomb repressive complex 2, underlies lineage-specific changes of T cell phenotypes associated with abrogation of BMPR1α signaling. These results reveal that BMPs are immunoregulatory cytokines mediating maturation and stability of peripheral FOXP3+ regulatory T cells (Treg cells) and controlling generation of iTreg cells. Thus, we establish that BMPs, a large cytokine family, are an essential link between stromal tissues and the adaptive immune system involved in sustaining tissue homeostasis by promoting immunological tolerance.
Collapse
Affiliation(s)
- Lauren M Browning
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Caroline Miller
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Michal Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Yu Jing
- Center for Bioelectrics, Old Dominion University, Norfolk, VA 23529, USA
| | - Grzegorz Rempala
- College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Pawel Muranski
- Columbia University Medical Center, New York, NY 10032, USA
| | - Leszek Ignatowicz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
15
|
Sun Y, Anyalebechi JC, Sun H, Yumoto T, Xue M, Liu D, Liang Z, Coopersmith CM, Ford ML. Anti-TIGIT differentially affects sepsis survival in immunologically experienced versus previously naive hosts. JCI Insight 2021; 6:141245. [PMID: 33682797 PMCID: PMC8021109 DOI: 10.1172/jci.insight.141245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 01/20/2021] [Indexed: 12/29/2022] Open
Abstract
Mounting evidence suggests that the balance of T cell costimulatory and coinhibitory signals contributes to mortality during sepsis. Here, we identified a critical role of the coinhibitory molecule T cell Ig and ITIM domain (TIGIT) in regulating sepsis mortality. Because TIGIT is significantly upregulated on memory T cells, we developed a "memory mouse" model to study the role of TIGIT during sepsis in a more physiologically relevant context. Mice received sequential pathogen exposure and developed memory T cell frequencies, similar to those observed in adult humans, and were then subjected to sepsis induction via cecal ligation and puncture. Our results show that targeting the TIGIT pathway during sepsis is fundamentally different in previously naive versus memory mice, in that αTIGIT Ab had no effect on survival in previously naive septic mice but sharply worsened survival in memory septic mice. Mechanistically, αTIGIT increased apoptosis of memory T cells, decreased T cell function, and downregulated the costimulatory receptor DNAM on memory CD8+ T cells in memory septic mice, but not in previously naive septic mice. Additionally, αTIGIT diminished Helios expression in Tregs in memory but not previously naive septic mice. These data highlight fundamental differences in the pathophysiological impact of targeting TIGIT in immunologically experienced versus previously naive hosts during sepsis.
Collapse
Affiliation(s)
- Yini Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jerome C. Anyalebechi
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - He Sun
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Hepatobiliary Surgery and Transplantation, The First Affiliated Hospital of China Medical University, China Medical University, Shenyang, China
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Tetsuya Yumoto
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ming Xue
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Danya Liu
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Lightbourn CO, Wolf D, Copsel SN, Wang Y, Pfeiffer BJ, Barreras H, Bader CS, Komanduri KV, Perez VL, Levy RB. Use of Post-transplant Cyclophosphamide Treatment to Build a Tolerance Platform to Prevent Liquid and Solid Organ Allograft Rejection. Front Immunol 2021; 12:636789. [PMID: 33737937 PMCID: PMC7962410 DOI: 10.3389/fimmu.2021.636789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
Corneal transplantation (CT) is the most frequent type of solid organ transplant (SOT) performed worldwide. Unfortunately, immunological rejection is the primary cause of graft failure for CT and therefore advances in immune regulation to induce tolerance remains an unmet medical need. Recently, our work and others in pre-clinical studies found that cyclophosphamide (Cy) administered after (“post-transplant,” PTCy) hematopoietic stem cell transplantation (HSCT), i.e., liquid transplants is effective for graft vs. host disease prophylaxis and enhances overall survival. Importantly, within the past 10 years, PTCy has been widely adopted for clinical HSCT and the results at many centers have been extremely encouraging. The present studies found that Cy can be effectively employed to prolong the survival of SOT, specifically mouse corneal allografts. The results demonstrated that the timing of PTCy administration is critical for these CT and distinct from the kinetics employed following allogeneic HSCT. PTCy was observed to interfere with neovascularization, a process critically associated with immune rejection of corneal tissue that ensues following the loss of ocular “immune privilege.” PTCy has the potential to delete or directly suppress allo-reactive T cells and treatment here was shown to diminish T cell rejection responses. These PTCy doses were observed to spare significant levels of CD4+ FoxP3+ (Tregs) which were found to be functional and could readily receive stimulating signals leading to their in vivo expansion via TNFRSF25 and CD25 agonists. In total, we posit future studies can take advantage of Cy based platforms to generate combinatorial strategies for long-term tolerance induction.
Collapse
Affiliation(s)
- Casey O Lightbourn
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dietlinde Wolf
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sabrina N Copsel
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ying Wang
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brent J Pfeiffer
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Henry Barreras
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Cameron S Bader
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Victor L Perez
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Foster Center for Ocular Immunology at Duke Eye Center, Duke University, Durham, NC, United States
| | - Robert B Levy
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
17
|
Ward NC, Lui JB, Hernandez R, Yu L, Struthers M, Xie J, Santos Savio A, Dwyer CJ, Hsiung S, Yu A, Malek TR. Persistent IL-2 Receptor Signaling by IL-2/CD25 Fusion Protein Controls Diabetes in NOD Mice by Multiple Mechanisms. Diabetes 2020; 69:2400-2413. [PMID: 32843568 PMCID: PMC7576568 DOI: 10.2337/db20-0186] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 08/21/2020] [Indexed: 10/23/2022]
Abstract
Low-dose interleukin-2 (IL-2) represents a new therapeutic approach to regulate immune homeostasis to promote immune tolerance in patients with autoimmune diseases, including type 1 diabetes. We have developed a new IL-2-based biologic, an IL-2/CD25 fusion protein, with greatly improved pharmacokinetics and pharmacodynamics when compared with recombinant IL-2 to enhance this type of immunotherapy. In this study, we show that low-dose mouse IL-2/CD25 (mIL-2/CD25), but not an equivalent amount of IL-2, prevents the onset of diabetes in NOD mice and controls diabetes in hyperglycemic mice. mIL-2/CD25 acts not only to expand regulatory T cells (Tregs) but also to increase their activation and migration into lymphoid tissues and the pancreas. Lower incidence of diabetes is associated with increased serum levels of IL-10, a cytokine readily produced by activated Tregs. These effects likely act in concert to lower islet inflammation while increasing Tregs in the remaining inflamed islets. mIL-2/CD25 treatment is also associated with lower anti-insulin autoantibody levels in part by inhibition of T follicular helper cells. Thus, long-acting mIL-2/CD25 represents an improved IL-2 analog that persistently elevates Tregs to maintain a favorable Treg/effector T cell ratio that limits diabetes by expansion of activated Tregs that readily migrate into lymphoid tissues and the pancreas while inhibiting autoantibodies.
Collapse
Affiliation(s)
- Natasha C Ward
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Jen Bon Lui
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Rosmely Hernandez
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Mary Struthers
- Immunology Discovery, Bristol-Myers Squibb, Princeton, NJ
| | - Jenny Xie
- Immunology Discovery, Bristol-Myers Squibb, Princeton, NJ
| | - Alicia Santos Savio
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Sunnie Hsiung
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Aixin Yu
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
18
|
Ulbar F, Villanova I, Giancola R, Baldoni S, Guardalupi F, Fabi B, Olioso P, Capone A, Sola R, Ciardelli S, Del Papa B, Brattelli A, Ricciardi I, Taricani S, Sabbatinelli G, Iuliani O, Passeri C, Sportoletti P, Santarone S, Pierini A, Calabrese G, Falzetti F, Bonfini T, Accorsi P, Ruggeri L, Martelli MF, Velardi A, Di Ianni M. Clinical-Grade Expanded Regulatory T Cells Are Enriched with Highly Suppressive Cells Producing IL-10, Granzyme B, and IL-35. Biol Blood Marrow Transplant 2020; 26:2204-2210. [PMID: 32961369 DOI: 10.1016/j.bbmt.2020.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
In the setting of T cell-depleted, full-haplotype mismatched transplantation, adoptive immunotherapy with regulatory T cells (Tregs) and conventional T cells (Tcons) can prevent graft-versus-host disease (GVHD) and improve post-transplantation immunologic reconstitution and is associated with a powerful graft-versus-leukemia effect. To improve the purity and the quantity of the infused Tregs, good manufacturing practices (GMP)-compatible expansion protocols are needed. Here we expanded Tregs using an automated, clinical-grade protocol. Cells were extensively characterized in vitro, and their efficiency was tested in vivo in a mouse model. Tregs were selected by CliniMacs (CD4+CD25+, 94.5 ± 6.3%; FoxP3+, 63.7 ± 11.5%; CD127+, 20 ± 3%; suppressive activity, 60 ± 7%), and an aliquot of 100 × 106 was expanded for 14 days using the CliniMACS Prodigy System, obtaining 684 ± 279 × 106 cells (CD4+CD25+, 99.6 ± 0.2%; FoxP3+, 82 ± 8%; CD127+, 1.1 ± 0.8%; suppressive activity, 75 ± 12%). CD39 and CTLA4 expression levels increased from 22.4 ± 12% to 58.1 ± 13.3% (P < .05) and from 20.4 ± 6.7% to 85.4 ± 9.8% (P < .01), respectively. TIM3 levels increased from .4 ± .05% to 29 ± 16% (P < .05). Memory Tregs were the prevalent population, whereas naive Tregs almost disappeared at the end of the culture. mRNA analysis displayed significant increases in CD39, IL-10, granzyme B, and IL-35 levels at the end of culture period (P < .05). Conversely, IFNγ expression decreased significantly by day +14. Expanded Tregs were sorted according to TIM3, CD39, and CD62L expression levels (purity >95%). When sorted populations were analyzed, TIM3+ cells showed significant increases in IL-10 and granzyme B (P < .01) .When expanded Tregs were infused in an NSG murine model, mice that received Tcons only died of GVHD, whereas mice that received both Tcons and Tregs survived without GVHD. GMP grade expanded cells that display phenotypic and functional Treg characteristics can be obtained using a fully automated system. Treg suppression is mediated by multiple overlapping mechanisms (eg, CTLA-4, CD39, IL-10, IL-35, TGF-β, granzyme B). TIM3+ cells emerge as a potentially highly suppressive population. © 2020 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Francesca Ulbar
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Ida Villanova
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | | | - Stefano Baldoni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Francesco Guardalupi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Bianca Fabi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Paola Olioso
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Anita Capone
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Rosaria Sola
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Sara Ciardelli
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | | | - Ilda Ricciardi
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Stefano Taricani
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Giulia Sabbatinelli
- Department of Neurosciences, Imaging and Clinical Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Ornella Iuliani
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Cecilia Passeri
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Paolo Sportoletti
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Stella Santarone
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Antonio Pierini
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Giuseppe Calabrese
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy; Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Franca Falzetti
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Tiziana Bonfini
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Patrizia Accorsi
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Loredana Ruggeri
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Massimo Fabrizio Martelli
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy; Department of Oncology Hematology, Pescara Hospital, Pescara, Italy.
| |
Collapse
|
19
|
Dwyer CJ, Knochelmann HM, Smith AS, Wyatt MM, Rangel Rivera GO, Arhontoulis DC, Bartee E, Li Z, Rubinstein MP, Paulos CM. Fueling Cancer Immunotherapy With Common Gamma Chain Cytokines. Front Immunol 2019; 10:263. [PMID: 30842774 PMCID: PMC6391336 DOI: 10.3389/fimmu.2019.00263] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
Adoptive T cell transfer therapy (ACT) using tumor infiltrating lymphocytes or lymphocytes redirected with antigen receptors (CAR or TCR) has revolutionized the field of cancer immunotherapy. Although CAR T cell therapy mediates robust responses in patients with hematological malignancies, this approach has been less effective for treating patients with solid tumors. Additionally, toxicities post T cell infusion highlight the need for safer ACT protocols. Current protocols traditionally expand T lymphocytes isolated from patient tumors or from peripheral blood to large magnitudes in the presence of high dose IL-2 prior to infusion. Unfortunately, this expansion protocol differentiates T cells to a full effector or terminal phenotype in vitro, consequently reducing their long-term survival and antitumor effectiveness in vivo. Post-infusion, T cells face further obstacles limiting their persistence and function within the suppressive tumor microenvironment. Therapeutic manipulation of T cells with common γ chain cytokines, which are critical growth factors for T cells, may be the key to bypass such immunological hurdles. Herein, we discuss the primary functions of the common γ chain cytokines impacting T cell survival and memory and then elaborate on how these distinct cytokines have been used to augment T cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Guillermo O Rangel Rivera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Dimitrios C Arhontoulis
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Eric Bartee
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
20
|
Copsel SN, Lightbourn CO, Barreras H, Lohse I, Wolf D, Bader CS, Manov J, Kale BJ, Shah D, Brothers SP, Perez VL, Komanduri KV, Wahlestedt C, Levy RB. BET Bromodomain Inhibitors Which Permit Treg Function Enable a Combinatorial Strategy to Suppress GVHD in Pre-clinical Allogeneic HSCT. Front Immunol 2019; 9:3104. [PMID: 30733722 PMCID: PMC6353853 DOI: 10.3389/fimmu.2018.03104] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/17/2018] [Indexed: 01/09/2023] Open
Abstract
A recent approach for limiting production of pro-inflammatory cytokines has been to target bromodomain and extra-terminal (BET) proteins. These epigenetic readers of histone acetylation regulate transcription of genes involved in inflammation, cardiovascular disease, and cancer. Development of BET inhibitors (BETi) has generated enormous interest for their therapeutic potential. Because inflammatory signals and donor T cells promote graft-versus-host disease (GVHD), regulating both pathways could be effective to abrogate this disorder. The objective of the present study was to identify a BETi which did not interfere in vivo with CD4+FoxP3+ regulatory T cell (Treg) expansion and function to utilize together with Tregs following allogeneic hematopoietic stem cell transplantation (aHSCT) to ameliorate GVHD. We have reported that Tregs can be markedly expanded and selectively activated with increased functional capacity by targeting TNFRSF25 and CD25 with TL1A-Ig and low dose IL-2, respectively. Here, mice were treated over 7 days (TL1A-Ig + IL-2) together with BETi. We found that the BETi EP11313 did not decrease frequency/numbers or phenotype of expanded Tregs as well as effector molecules, such as IL-10 and TGF-β. However, BETi JQ1 interfered with Treg expansion and altered subset distribution and phenotype. Notably, in Treg expanded mice, EP11313 diminished tnfa and ifng but not il-2 RNA levels. Remarkably, Treg pSTAT5 expression was not affected by EP11313 supporting the notion that Treg IL-2 signaling remained intact. MHC-mismatched aHSCT (B6 → BALB/c) was performed using in vivo expanded donor Tregs with or without EP11313 short-term treatment in the recipient. Early post-transplant, improvement in the splenic and LN CD4/CD8 ratio along with fewer effector cells and high Treg levels in aHSCT recipients treated with expanded Tregs + EP11313 was detected. Interestingly, this group exhibited a significant diminution of GVHD clinical score with less skin and ocular involvement. Finally, using low numbers of highly purified expanded Tregs, improved clinical GVHD scores were observed in EP11313 treated recipients. In total, we conclude that use of this novel combinatorial strategy can suppress pre-clinical GVHD and posit, in vivo EP11313 treatment might be useful combined with Treg expansion therapy for treatment of diseases involving inflammatory responses.
Collapse
Affiliation(s)
- Sabrina N Copsel
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Casey O Lightbourn
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Henry Barreras
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ines Lohse
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Center for Therapeutic Innovation and Department of Psychiatry and Behavior Sciences, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dietlinde Wolf
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Cameron S Bader
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - John Manov
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brandon J Kale
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Devangi Shah
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Shaun P Brothers
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Center for Therapeutic Innovation and Department of Psychiatry and Behavior Sciences, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Victor L Perez
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Krishna V Komanduri
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Claes Wahlestedt
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Center for Therapeutic Innovation and Department of Psychiatry and Behavior Sciences, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Robert B Levy
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States.,Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
21
|
Seitz C, Liu S, Klocke K, Joly AL, Czarnewski PV, Tibbitt CA, Parigi SM, Westerberg LS, Coquet JM, Villablanca EJ, Wing K, Andersson J. Multi-faceted inhibition of dendritic cell function by CD4 +Foxp3 + regulatory T cells. J Autoimmun 2019; 98:86-94. [PMID: 30616979 DOI: 10.1016/j.jaut.2018.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/22/2022]
Abstract
CTLA-4 is required for CD4+Foxp3+ regulatory T (Treg) cell function, but its mode of action remains incompletely defined. Herein we generated Ctla-4ex2fl/flFoxp3-Cre mice with Treg cells exclusively expressing a naturally occurring, ligand-independent isoform of CTLA-4 (liCTLA-4) that cannot interact with the costimulatory molecules CD80 and CD86. The mice did not exhibit any signs of effector T cell activation early in life, however, at 6 months of age they exhibited excessive T cell activation and inflammation in lungs. In contrast, mice with Treg cells completely lacking CTLA-4 developed lymphoproliferative disease characterized by multi-organ inflammation early in life. In vitro, Treg cells exclusively expressing liCTLA-4 inhibited CD80 and CD86 expression on dendritic cells (DC). Conversely, Treg cells required the extra-cellular part of CTLA-4 to up-regulate expression of the co-inhibitory molecule PD-L2 on DCs. Transcriptomic analysis of suppressed DCs revealed that Treg cells induced a specific immunosuppressive program in DCs.
Collapse
Affiliation(s)
- Christina Seitz
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Sang Liu
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Klocke
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne-Laure Joly
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Christopher A Tibbitt
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sara M Parigi
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Kajsa Wing
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - John Andersson
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
22
|
Copsel S, Wolf D, Kale B, Barreras H, Lightbourn CO, Bader CS, Alperstein W, Altman NH, Komanduri KV, Levy RB. Very Low Numbers of CD4 + FoxP3 + Tregs Expanded in Donors via TL1A-Ig and Low-Dose IL-2 Exhibit a Distinct Activation/Functional Profile and Suppress GVHD in a Preclinical Model. Biol Blood Marrow Transplant 2018; 24:1788-1794. [PMID: 29751114 DOI: 10.1016/j.bbmt.2018.04.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 04/23/2018] [Indexed: 11/25/2022]
Abstract
Regulatory T cells (Tregs) are essential for the maintenance of tolerance and immune homeostasis. In allogeneic hematopoietic stem cell transplantation (aHSCT), transfer of appropriate Treg numbers is a promising therapy for the prevention of graft-versus-host disease (GVHD). We have recently reported a novel approach that induces the marked expansion and selective activation of Tregs in vivo by targeting tumor necrosis factor receptor superfamily 25 (TNFRSF25) and CD25. A potential advance to promote clinical application of Tregs to ameliorate GVHD and other disorders would be the generation of more potent Treg populations. Here we wanted to determine if very low doses of Tregs generated using the "2-pathway" stimulation protocol via TL1A-Ig fusion protein and low-dose IL-2 (targeting TNFRSF25 and CD25, respectively) could be used to regulate preclinical GVHD. Analysis of such 2-pathway expanded Tregs identified higher levels of activation and functional molecules (CD103, ICOS-1, Nrp-1, CD39, CD73, il-10, and tgfb1) versus unexpanded Tregs. Additionally, in vitro assessment of 2-pathway stimulated Tregs indicated enhanced suppressor activity. Notably, transplant of extremely low numbers of these Tregs (1:6 expanded Tregs/conventional T cells) suppressed GVHD after an MHC-mismatched aHSCT. Overall, these results demonstrate that 2-pathway stimulated CD4+ FoxP3+ Tregs were quantitatively and qualitatively more functionally effective than unexpanded Tregs. In total, the findings in this study support the notion that such 2-pathway stimulated Tregs may be useful for prevention of GVHD and ultimately promote more widespread application of aHSCT in the clinic.
Collapse
Affiliation(s)
- Sabrina Copsel
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Dietlinde Wolf
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida
| | - Brandon Kale
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Henry Barreras
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Casey O Lightbourn
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Cameron S Bader
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Warren Alperstein
- Department of Pediatrics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Norman H Altman
- Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Krishna V Komanduri
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida; Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Robert B Levy
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida; Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
23
|
Lee JY, Kim J, Yi J, Kim D, Kim HO, Han D, Sprent J, Lee YJ, Surh CD, Cho JH. Phenotypic and Functional Changes of Peripheral Ly6C + T Regulatory Cells Driven by Conventional Effector T Cells. Front Immunol 2018; 9:437. [PMID: 29616017 PMCID: PMC5864862 DOI: 10.3389/fimmu.2018.00437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 02/19/2018] [Indexed: 01/26/2023] Open
Abstract
A relatively high affinity/avidity of T cell receptor (TCR) recognition for self-peptide bound to major histocompatibility complex II (self-pMHC) ligands is a distinctive feature of CD4 T regulatory (Treg) cells, including their development in the thymus and maintenance of their suppressive functions in the periphery. Despite such high self-reactivity, however, all thymic-derived peripheral Treg populations are neither homogenous in their phenotype nor uniformly immune-suppressive in their function under steady state condition. We show here that based on the previously defined heterogeneity in the phenotype of peripheral Treg populations, Ly6C expression on Treg marks a lower degree of activation, proliferation, and differentiation status as well as functional incompetence. We also demonstrate that Ly6C expression on Treg in a steady state is either up- or downregulated depending on relative amounts of tonic TCR signals derived from its contacts with self-ligands. Interestingly, peripheral appearance and maintenance of these Ly6C-expressing Treg cells largely differed in an age-dependent manner, with their proportion being continuously increased from perinatal to young adult period but then being gradually declined with age. The reduction of Ly6C+ Treg in the aged mice was not due to their augmented cell death but rather resulted from downregulation of Ly6C expression. The Ly6C downregulation was accompanied by proliferation of Ly6C+ Treg cells and subsequent change into Ly6C− effector Treg with concomitant restoration of immune-suppressive activity. Importantly, we found that this phenotypic and functional change of Ly6C+ Treg is largely driven by conventional effector T cell population. Collectively, these findings suggest a potential cross-talk between peripheral Treg subsets and effector T cells and provides better understanding for Treg homeostasis and function on maintaining self-tolerance.
Collapse
Affiliation(s)
- Jun Young Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Juhee Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Jaeu Yi
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Daeun Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Hee-Ok Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Daehee Han
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - You Jeong Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Charles D Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea.,Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Jae-Ho Cho
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, South Korea.,Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| |
Collapse
|
24
|
Toomer KH, Malek TR. Cytokine Signaling in the Development and Homeostasis of Regulatory T cells. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028597. [PMID: 28620098 DOI: 10.1101/cshperspect.a028597] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cytokine signaling is indispensable for regulatory T-cell (Treg) development in the thymus, and also influences the homeostasis, phenotypic diversity, and function of Tregs in the periphery. Because Tregs are required for establishment and maintenance of immunological self-tolerance, investigating the role of cytokines in Treg biology carries therapeutic potential in the context of autoimmune disease. This review discusses the potent and diverse influences of interleukin (IL)-2 signaling on the Treg compartment, an area of knowledge that has led to the use of low-dose IL-2 as a therapy to reregulate autoaggressive immune responses. Evidence suggesting Treg-specific impacts of the cytokines transforming growth factor β (TGF-β), IL-7, thymic stromal lymphopoietin (TSLP), IL-15, and IL-33 is also presented. Finally, we consider the technical challenges and knowledge limitations that must be overcome to bring other cytokine-based, Treg-targeted therapies into clinical use.
Collapse
Affiliation(s)
- Kevin H Toomer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida 33136.,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136
| |
Collapse
|
25
|
Sprouse ML, Shevchenko I, Scavuzzo MA, Joseph F, Lee T, Blum S, Borowiak M, Bettini ML, Bettini M. Cutting Edge: Low-Affinity TCRs Support Regulatory T Cell Function in Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2017; 200:909-914. [PMID: 29282307 DOI: 10.4049/jimmunol.1700156] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
Regulatory T cells (Tregs) use a distinct TCR repertoire and are more self-reactive compared with conventional T cells. However, the extent to which TCR affinity regulates the function of self-reactive Tregs is largely unknown. In this study, we used a two-TCR model to assess the role of TCR affinity in Treg function during autoimmunity. We observed that high- and low-affinity Tregs were recruited to the pancreas and contributed to protection from autoimmune diabetes. Interestingly, high-affinity cells preferentially upregulated the TCR-dependent Treg functional mediators IL-10, TIGIT, GITR, and CTLA4, whereas low-affinity cells displayed increased transcripts for Areg and Ebi3, suggesting distinct functional profiles. The results of this study suggest mechanistically distinct and potentially nonredundant roles for high- and low-affinity Tregs in controlling autoimmunity.
Collapse
Affiliation(s)
- Maran L Sprouse
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Ivan Shevchenko
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030
| | - Faith Joseph
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Thomas Lee
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Samuel Blum
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Malgorzata Borowiak
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030; and.,McNair Medical Institute, Houston, TX 77030
| | - Matthew L Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030.,McNair Medical Institute, Houston, TX 77030
| | - Maria Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030; .,McNair Medical Institute, Houston, TX 77030
| |
Collapse
|
26
|
Dwyer CJ, Bayer AL, Fotino C, Yu L, Cabello-Kindelan C, Ward NC, Toomer KH, Chen Z, Malek TR. Altered homeostasis and development of regulatory T cell subsets represent an IL-2R-dependent risk for diabetes in NOD mice. Sci Signal 2017; 10:10/510/eaam9563. [PMID: 29259102 DOI: 10.1126/scisignal.aam9563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytokine interleukin-2 (IL-2) is critical for the functions of regulatory T cells (Tregs). The contribution of polymorphisms in the gene encoding the IL-2 receptor α subunit (IL2RA), which are associated with type 1 diabetes, is difficult to determine because autoimmunity depends on variations in multiple genes, where the contribution of any one gene product is small. We investigated the mechanisms whereby a modest reduction in IL-2R signaling selectively in T lymphocytes influenced the development of diabetes in the NOD mouse model. The sensitivity of IL-2R signaling was reduced by about two- to threefold in Tregs from mice that coexpressed wild-type IL-2Rβ and a mutant subunit (IL-2RβY3) with reduced signaling (designated NOD-Y3). Male and female NOD-Y3 mice exhibited accelerated diabetes onset due to intrinsic effects on multiple activities in Tregs Bone marrow chimera and adoptive transfer experiments demonstrated that IL-2RβY3 Tregs resulted in impaired homeostasis of lymphoid-residing central Tregs and inefficient development of highly activated effector Tregs and that they were less suppressive. Pancreatic IL-2RβY3 Tregs showed impaired development into IL-10-secreting effector Tregs The pancreatic lymph nodes and pancreases of NOD-Y3 mice had increased numbers of antigen-experienced CD4+ effector T cells, which was largely due to impaired Tregs, because adoptively transferred pancreatic autoantigen-specific CD4+ Foxp3- T cells from NOD-Y3 mice did not accelerate diabetes in NOD.SCID recipients. Our study indicates that the primary defect associated with chronic, mildly reduced IL-2R signaling is due to impaired Tregs that cannot effectively produce and maintain highly functional tissue-seeking effector Treg subsets.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Allison L Bayer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Carmen Fotino
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Cecilia Cabello-Kindelan
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Natasha C Ward
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Kevin H Toomer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA. .,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
27
|
Yu A, Dee MJ, Adeegbe D, Dwyer CJ, Altman NH, Malek TR. The Lower Limit of Regulatory CD4 + Foxp3 + TCRβ Repertoire Diversity Required To Control Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2017; 198:3127-3135. [PMID: 28264971 DOI: 10.4049/jimmunol.1601966] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/13/2017] [Indexed: 01/18/2023]
Abstract
The TCR repertoire of regulatory T cells (Tregs) is highly diverse. The relevance of this diversity to maintain self-tolerance remains unknown. We established a model where the TCR repertoire of normal polyclonal Tregs was limited by serial transfers into IL-2Rβ-/- mice, which lack functional Tregs. After a primary transfer, the donor Treg TCR repertoire was substantially narrowed, yet the recipients remained autoimmune-free. Importantly, upon purification and transfer of donor-derived Tregs from an individual primary recipient into neonatal IL-2Rβ-/- mice, the secondary recipients developed autoimmunity. In this study, the Treg TCRβ repertoire was reshaped and further narrowed. In contrast, secondary IL-2Rβ recipients showed fewer symptoms of autoimmunity when they received donor Tregs that were premixed from several primary recipients to increase their TCRβ repertoire diversity. About 8-11% of the Treg TCRβ repertoire was estimated to be the minimum required to establish and maintain tolerance in primary IL-2Rβ-/- recipients. Collectively, these data quantify where limitations imposed on the Treg TCRβ repertoire results in a population of Tregs that cannot fully suppress polyclonal autoreactive T cells. Our data favor a model where the high diversity of the Treg TCR provides a mechanism for Tregs to actively adapt and effectively suppress autoreactive T cells, which are not fixed, but are evolving as they encounter self-antigens.
Collapse
Affiliation(s)
- Aixin Yu
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Michael J Dee
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Dennis Adeegbe
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Norman H Altman
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136; and
| | - Thomas R Malek
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136; .,Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136
| |
Collapse
|
28
|
Wolf D, Barreras H, Bader CS, Copsel S, Lightbourn CO, Pfeiffer BJ, Altman NH, Podack ER, Komanduri KV, Levy RB. Marked in Vivo Donor Regulatory T Cell Expansion via Interleukin-2 and TL1A-Ig Stimulation Ameliorates Graft-versus-Host Disease but Preserves Graft-versus-Leukemia in Recipients after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2017; 23:757-766. [PMID: 28219835 DOI: 10.1016/j.bbmt.2017.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/16/2017] [Indexed: 01/23/2023]
Abstract
Regulatory T cells (Tregs) are critical for self-tolerance. Although adoptive transfer of expanded Tregs limits graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation (HSCT), ex vivo generation of large numbers of functional Tregs remains difficult. Here, we demonstrate that in vivo targeting of the TNF superfamily receptor TNFRSF25 using the TL1A-Ig fusion protein, along with IL-2, resulted in transient but massive Treg expansion in donor mice, which peaked within days and was nontoxic. Tregs increased in multiple compartments, including blood, lymph nodes, spleen, and colon (GVHD target tissue). Tregs did not expand in bone marrow, a critical site for graft-versus-malignancy responses. Adoptive transfer of in vivo-expanded Tregs in the setting of MHC-mismatched or MHC-matched allogeneic HSCT significantly ameliorated GVHD. Critically, transplantation of Treg-expanded donor cells facilitated transplant tolerance without GVHD, with complete sparing of graft-versus-malignancy. This approach may prove valuable as a therapeutic strategy promoting transplantation tolerance.
Collapse
Affiliation(s)
- Dietlinde Wolf
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Henry Barreras
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Cameron S Bader
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Sabrina Copsel
- Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Casey O Lightbourn
- Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Brent J Pfeiffer
- Department of Pediatrics, University of Miami, Miller School of Medicine, Miami, Florida
| | - Norman H Altman
- Department of Pathology and Laboratory Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Eckhard R Podack
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Robert B Levy
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami, Miller School of Medicine, Miami, Florida; Department of Ophthalmology, University of Miami, Miller School of Medicine, Miami, Florida.
| |
Collapse
|