1
|
Baron KJ, Turnquist HR. Clinical Manufacturing of Regulatory T Cell Products For Adoptive Cell Therapy and Strategies to Improve Therapeutic Efficacy. Organogenesis 2023; 19:2164159. [PMID: 36681905 PMCID: PMC9870008 DOI: 10.1080/15476278.2022.2164159] [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] [Indexed: 01/23/2023] Open
Abstract
Based on successes in preclinical animal transplant models, adoptive cell therapy (ACT) with regulatory T cells (Tregs) is a promising modality to induce allograft tolerance or reduce the use of immunosuppressive drugs to prevent rejection. Extensive work has been done in optimizing the best approach to manufacture Treg cell products for testing in transplant recipients. Collectively, clinical evaluations have demonstrated that large numbers of Tregs can be expanded ex vivo and infused safely. However, these trials have failed to induce robust drug-free tolerance and/or significantly reduce the level of immunosuppression needed to prevent solid organ transplant (SOTx) rejection. Improving Treg therapy effectiveness may require increasing Treg persistence or orchestrating Treg migration to secondary lymphatic tissues or places of inflammation. In this review, we describe current clinical Treg manufacturing methods used for clinical trials. We also highlight current strategies being implemented to improve delivered Treg ACT persistence and migration in preclinical studies.
Collapse
Affiliation(s)
- Kassandra J. Baron
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Infectious Disease and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,CONTACT Hēth R. Turnquist Departments of Surgery, University of Pittsburgh School of Medicine, Thomas E. Starzl Transplantation Institute 200 Lothrop Street, BST W1542, PittsburghPA 15213, USA
| |
Collapse
|
2
|
Rosado-Sánchez I, Haque M, Salim K, Speck M, Fung VC, Boardman DA, Mojibian M, Raimondi G, Levings MK. Tregs integrate native and CAR-mediated costimulatory signals for control of allograft rejection. JCI Insight 2023; 8:e167215. [PMID: 37669115 PMCID: PMC10619441 DOI: 10.1172/jci.insight.167215] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
Tregs expressing chimeric antigen receptors (CAR-Tregs) are a promising tool to promote transplant tolerance. The relationship between CAR structure and Treg function was studied in xenogeneic, immunodeficient mice, revealing advantages of CD28-encoding CARs. However, these models could underrepresent interactions between CAR-Tregs, antigen-presenting cells (APCs), and donor-specific Abs. We generated Tregs expressing HLA-A2-specific CARs with different costimulatory domains and compared their function in vitro and in vivo using an immunocompetent model of transplantation. In vitro, the CD28-encoding CAR had superior antigen-specific suppression, proliferation, and cytokine production. In contrast, in vivo, Tregs expressing CARs encoding CD28, ICOS, programmed cell death 1, and GITR, but not 4-1BB or OX40, all extended skin allograft survival. To reconcile in vitro and in vivo data, we analyzed effects of a CAR encoding CD3ζ but no costimulatory domain. These data revealed that exogenous costimulation from APCs can compensate for the lack of a CAR-encoded CD28 domain. Thus, Tregs expressing a CAR with or without CD28 are functionally equivalent in vivo, mediating similar extension of skin allograft survival and controlling the generation of anti-HLA-A2 alloantibodies. This study reveals a dimension of CAR-Treg biology and has important implications for the design of CARs for clinical use in Tregs.
Collapse
Affiliation(s)
- Isaac Rosado-Sánchez
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Biomedical Engineering and
| | - Manjurul Haque
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin Salim
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Madeleine Speck
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vivian C.W. Fung
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dominic A. Boardman
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Megan K. Levings
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Biomedical Engineering and
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
3
|
Abdeladhim M, Karnell JL, Rieder SA. In or out of control: Modulating regulatory T cell homeostasis and function with immune checkpoint pathways. Front Immunol 2022; 13:1033705. [PMID: 36591244 PMCID: PMC9799097 DOI: 10.3389/fimmu.2022.1033705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/16/2022] [Indexed: 12/16/2022] Open
Abstract
Regulatory T cells (Tregs) are the master regulators of immunity and they have been implicated in different disease states such as infection, autoimmunity and cancer. Since their discovery, many studies have focused on understanding Treg development, differentiation, and function. While there are many players in the generation and function of truly suppressive Tregs, the role of checkpoint pathways in these processes have been studied extensively. In this paper, we systematically review the role of different checkpoint pathways in Treg homeostasis and function. We describe how co-stimulatory and co-inhibitory pathways modulate Treg homeostasis and function and highlight data from mouse and human studies. Multiple checkpoint pathways are being targeted in cancer and autoimmunity; therefore, we share insights from the clinic and discuss the effect of experimental and approved therapeutics on Treg biology.
Collapse
|
4
|
Halliday N, Williams C, Kennedy A, Waters E, Pesenacker AM, Soskic B, Hinze C, Hou TZ, Rowshanravan B, Janman D, Walker LSK, Sansom DM. CD86 Is a Selective CD28 Ligand Supporting FoxP3+ Regulatory T Cell Homeostasis in the Presence of High Levels of CTLA-4. Front Immunol 2020; 11:600000. [PMID: 33363541 PMCID: PMC7753196 DOI: 10.3389/fimmu.2020.600000] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
CD80 and CD86 are expressed on antigen presenting cells and are required to engage their shared receptor, CD28, for the costimulation of CD4 T cells. It is unclear why two stimulatory ligands with overlapping roles have evolved. CD80 and CD86 also bind the regulatory molecule CTLA-4. We explored the role of CD80 and CD86 in the homeostasis and proliferation of CD4+FoxP3+ regulatory T cells (Treg), which constitutively express high levels of CTLA-4 yet are critically dependent upon CD28 signals. We observed that CD86 was the dominant ligand for Treg proliferation, survival, and maintenance of a regulatory phenotype, with higher expression of CTLA-4, ICOS, and OX40. We also explored whether CD80-CD28 interactions were specifically compromised by CTLA-4 and found that antibody blockade, clinical deficiency of CTLA-4 and CRISPR-Cas9 deletion of CTLA-4 all improved Treg survival following CD80 stimulation. Taken together, our data suggest that CD86 is the dominant costimulatory ligand for Treg homeostasis, despite its lower affinity for CD28, because CD80-CD28 interactions are selectively impaired by the high levels of CTLA-4. These data suggest a cell intrinsic role for CTLA-4 in regulating CD28 costimulation by direct competition for CD80, and indicate that that CD80 and CD86 have discrete roles in CD28 costimulation of CD4 T cells in the presence of high levels of CTLA-4.
Collapse
Affiliation(s)
- Neil Halliday
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Institute of Liver and Digestive Health, University College London, London, United Kingdom
| | - Cayman Williams
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Alan Kennedy
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Erin Waters
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Blagoje Soskic
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Claudia Hinze
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Tie Zheng Hou
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Behzad Rowshanravan
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Daniel Janman
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lucy S K Walker
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
5
|
Cano-Martínez D, Monserrat J, Hernández-Breijo B, Sanmartín Salinas P, Álvarez-Mon M, Val Toledo-Lobo M, Guijarro LG. Extracellular allograft inflammatory factor-1 (AIF-1) potentiates Th1 cell differentiation and inhibits Treg response in human peripheral blood mononuclear cells from normal subjects. Hum Immunol 2020; 81:91-100. [PMID: 32057519 DOI: 10.1016/j.humimm.2020.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 11/17/2022]
Abstract
We identified the presence of AIF-1 (allograft inflammatory factor-1) in human peripheral blood mononuclear cells (PBMCs) from normal subjects by immunocytological methods. After isolation of different types of mononuclear cells by FACS (Fluorescence-activated cell sorting) with >95% purity, we studied the transcript levels of AIF-1 using qPCR. We observed the following order of AIF-1 mRNA expression in mononuclear cells: T-lymphocytes ˃ Monocytes ˃ B-lymphocytes ˃ NK. After T cell expansion of isolated PBMCs using anti-CD3-CD28 magnetic beads (Dynabeads®), AIF-1 increased intracellularly in the presence of brefeldin A; this finding, along with an increase in the medium in the absence of the drug, suggests that AIF-1 is processed in the Golgi apparatus and may be secreted extracellularly. In another set of experiments, interleukin-12 and anti-interleukin-4 were added to PBMCs during T cell expansion to promote Th1 polarization and to inhibit Th2 differentiation. In this case, the presence of 6 nM of rhAIF-1 (recombinant human AIF-1) increased the mRNA expression of interferon-ϒ and interleukin-2. In the same set of experiments, the incubation of PBMCs with rhAIF-1 (6 nM) promoted the decrease of mRNA expression of IL-10 and TGF-β, along with the decrease of CD25 and Foxp3 proteins. Furthermore, extracellular rhAIF-1 (6 nM) increased the survival of naive and effector T cells during Th1 polarization by inhibition of apoptosis, without causing changes in cell cycle rate and in retinoblastoma-cyclin-dependent kinase (Rb-CDK) activation. Taken together, rhAIF-1 treatment of PBMCs potentiates Th1 response and inhibits functionally suppressive CD25 + Foxp3 + Treg, which suggests an important immunomodulatory role in governing T cell response.
Collapse
Affiliation(s)
- David Cano-Martínez
- Department of Systems Biology, University of Alcalá, Alcalá de Henares. Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD), Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialties, University of Alcalá, Alcalá de Henares, Spain
| | - Borja Hernández-Breijo
- Department of Systems Biology, University of Alcalá, Alcalá de Henares. Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD), Spain
| | - Patricia Sanmartín Salinas
- Department of Systems Biology, University of Alcalá, Alcalá de Henares. Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD), Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, University of Alcalá, Alcalá de Henares, Spain
| | - M Val Toledo-Lobo
- Department of Biomedicine and Biotechnology, Unit of Cell Biology, University of Alcalá, Alcalá de Henares, Spain
| | - Luis G Guijarro
- Department of Systems Biology, University of Alcalá, Alcalá de Henares. Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas CIBEREHD), Spain.
| |
Collapse
|
6
|
Wing JB, Tay C, Sakaguchi S. Control of Regulatory T Cells by Co-signal Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:179-210. [DOI: 10.1007/978-981-32-9717-3_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
7
|
Signal Transduction Via Co-stimulatory and Co-inhibitory Receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:85-133. [PMID: 31758532 DOI: 10.1007/978-981-32-9717-3_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T-cell receptor (TCR)-mediated antigen-specific stimulation is essential for initiating T-cell activation. However, signaling through the TCR alone is not sufficient for inducing an effective response. In addition to TCR-mediated signaling, signaling through antigen-independent co-stimulatory or co-inhibitory receptors is critically important not only for the full activation and functional differentiation of T cells but also for the termination and suppression of T-cell responses. Many studies have investigated the signaling pathways underlying the function of each molecular component. Co-stimulatory and co-inhibitory receptors have no kinase activity, but their cytoplasmic region contains unique functional motifs and potential phosphorylation sites. Engagement of co-stimulatory receptors leads to recruitment of specific binding partners, such as adaptor molecules, kinases, and phosphatases, via recognition of a specific motif. Consequently, each co-stimulatory receptor transduces a unique pattern of signaling pathways. This review focuses on our current understanding of the intracellular signaling pathways provided by co-stimulatory and co-inhibitory molecules, including B7:CD28 family members, immunoglobulin, and members of the tumor necrosis factor receptor superfamily.
Collapse
|
8
|
He T, Liu S, Chen S, Ye J, Wu X, Bian Z, Chen X. The p38 MAPK Inhibitor SB203580 Abrogates Tumor Necrosis Factor-Induced Proliferative Expansion of Mouse CD4 +Foxp3 + Regulatory T Cells. Front Immunol 2018; 9:1556. [PMID: 30038619 PMCID: PMC6046375 DOI: 10.3389/fimmu.2018.01556] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/25/2018] [Indexed: 12/27/2022] Open
Abstract
There is now compelling evidence that tumor necrosis factor (TNF) preferentially activates and expands CD4+Foxp3+ regulatory T cells (Tregs) through TNF receptor type II (TNFR2). However, it remains unclear which signaling transduction pathway(s) of TNFR2 is required for the stimulation of Tregs. Previously, it was shown that the interaction of TNF–TNFR2 resulted in the activation of a number of signaling pathways, including p38 MAPK, NF-κB, in T cells. We thus examined the role of p38 MAPK and NF-κB in TNF-mediated activation of Tregs, by using specific small molecule inhibitors. The results show that treatment with specific p38 MAPK inhibitor SB203580, rather than NF-κB inhibitors (Sulfasalazine and Bay 11-7082), abrogated TNF-induced expansion of Tregs in vitro. Furthermore, upregulation of TNFR2 and Foxp3 expression in Tregs by TNF was also markedly inhibited by SB203580. The proliferative expansion and the upregulation of TNFR2 expression on Tregs in LPS-treated mice were mediated by TNF–TNFR2 interaction, as shown by our previous study. The expansion of Tregs in LPS-treated mice were also markedly inhibited by in vivo treatment with SB203580. Taken together, our data clearly indicate that the activation of p38 MAPK is attributable to TNF/TNFR2-mediated activation and proliferative expansion of Tregs. Our results also suggest that targeting of p38 MAPK by pharmacological agent may represent a novel strategy to up- or downregulation of Treg activity for therapeutic purposes.
Collapse
Affiliation(s)
- Tianzhen He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Shuoyang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Shaokui Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jingyi Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xueqiang Wu
- Department of Oncology, Beijing Aerospace General Hospital, Beijing, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
9
|
Wakamatsu E, Omori H, Ohtsuka S, Ogawa S, Green JM, Abe R. Regulatory T cell subsets are differentially dependent on CD28 for their proliferation. Mol Immunol 2018; 101:92-101. [PMID: 29909367 DOI: 10.1016/j.molimm.2018.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/21/2018] [Accepted: 05/24/2018] [Indexed: 01/08/2023]
Abstract
It is thought that CD28 plays a crucial role in the maintenance of regulatory T cell (Treg) pool size through promoting the development and proliferation of these cells. However, recently we found that the dependency on CD28 co-stimulation for their development is different between Treg subsets, thymus-derived Tregs (tTregs, CD28-dependent) and peripherally-derived Tregs (pTregs, CD28-independent), suggesting that CD28 may also have differential influences on the homeostasis of each Treg subset. Here, we demonstrated that both Treg subsets were reduced in secondary lymphoid organs of CD28 deficient mice, and that this reduction was due to impaired proliferation in both Treg subsets by the intrinsic CD28 defect. However, we found that the massive proliferation of both Treg subsets under lymphopenic condition was regulated by CD28, whereas the proliferative activity of tTregs but not pTregs in the steady state was dependent on CD28. Also, experiments using mutant CD28 knock-in mice revealed that proliferation of pTregs under lymphopenic condition required only the Lck-NFκB pathway of CD28, whereas tTregs required an additional unknown pathway. These findings indicate that the dependency on CD28 for proliferation in each Treg subset differs depending on the environment.
Collapse
Affiliation(s)
- Ei Wakamatsu
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba, 278-0022, Japan; Department of Immunology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Hiroki Omori
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba, 278-0022, Japan
| | - Shizuka Ohtsuka
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba, 278-0022, Japan
| | - Shuhei Ogawa
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba, 278-0022, Japan
| | - Jonathan M Green
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda City, Chiba, 278-0022, Japan.
| |
Collapse
|
10
|
Sundqvist KG. T Cell Co-Stimulation: Inhibition of Immunosuppression? Front Immunol 2018; 9:974. [PMID: 29774033 PMCID: PMC5943593 DOI: 10.3389/fimmu.2018.00974] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/19/2018] [Indexed: 11/18/2022] Open
Affiliation(s)
- Karl-Gösta Sundqvist
- Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, Clinical Immunology and Transfusion Medicine at Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
11
|
Hombach AA, Abken H. Most Do, but Some Do Not: CD4⁺CD25 - T Cells, but Not CD4⁺CD25⁺ Treg Cells, Are Cytolytic When Redirected by a Chimeric Antigen Receptor (CAR). Cancers (Basel) 2017; 9:cancers9090112. [PMID: 28850063 PMCID: PMC5615327 DOI: 10.3390/cancers9090112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/20/2017] [Accepted: 08/22/2017] [Indexed: 01/30/2023] Open
Abstract
Evidences are accumulating that CD4⁺ T cells can physiologically mediate antigen specific target cell lysis. By circumventing major histocompatibility complex (MHC)-restrictions through an engineered chimeric antigen receptor (CAR), CD4⁺ T cells lyse defined target cells as efficiently as do CD8⁺ T cells. However, the cytolytic capacity of redirected CD4⁺CD25- T cells, in comparison with CD4⁺CD25⁺ regulatory T (Treg) cells was so far not thoroughly defined. Treg cells require a strong CD28 signal together with CD3ζ for activation. We consequently used a CAR with combined CD28-CD3ζ signalling for redirecting CD4⁺CD25- T cells and CD4⁺CD25⁺ Treg cells from the same donor. CAR redirected activation of these T cell subsets and induced a distinct cytokine pattern with high IL-10 and a lack of IL-2 release by Treg cells. Despite strong antigen-specific activation, CAR Treg cells produced only weak target cell lysis, whereas CD4⁺CD25- CAR T cells were potent killers. Cytolysis did not correlate with the target cell sensitivity to Fas/FasL mediated killing; CD4⁺CD25- T cells upregulated perforin and granzyme B upon CAR activation, whereas Treg cells did less. The different cytolytic capacities of CAR redirected conventional CD4⁺ cells and Treg cells imply their use for different purposes in cell therapy.
Collapse
Affiliation(s)
- Andreas A Hombach
- Department I Internal Medicine, University Hospital Cologne, Cologne D-50931, Germany.
| | - Hinrich Abken
- Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch-Str. 21, Cologne D-50931, Germany.
| |
Collapse
|
12
|
He X, Koenen HJ, Slaats JH, Joosten I. Stabilizing human regulatory T cells for tolerance inducing immunotherapy. Immunotherapy 2017; 9:735-751. [PMID: 28771099 DOI: 10.2217/imt-2017-0017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Many autoimmune diseases develop as a consequence of an altered balance between autoreactive immune cells and suppressive FOXP3+ Treg. Restoring this balance through amplification of Treg represents a promising strategy to treat disease. However, FOXP3+ Treg might become unstable especially under certain inflammatory conditions, and might transform into proinflammatory cytokine-producing cells. The issue of heterogeneity and instability of Treg has caused considerable debate in the field and has important implications for Treg-based immunotherapy. In this review, we discuss how Treg stability is defined and what the molecular mechanisms underlying the maintenance of FOXP3 expression and the regulation of Treg stability are. Also, we elaborate on current strategies used to stabilize human Treg for clinical purposes. This review focuses on human Treg, but considering that cell-intrinsic mechanisms to regulate Treg stability in mice and in humans might be similar, data derived from mice studies are also discussed in this paper.
Collapse
Affiliation(s)
- Xuehui He
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,College of Computer Science, Qinghai Normal University, Xining, Qinghai, China
| | - Hans Jpm Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen Hr Slaats
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
13
|
Boardman DA, Philippeos C, Fruhwirth GO, Ibrahim MAA, Hannen RF, Cooper D, Marelli-Berg FM, Watt FM, Lechler RI, Maher J, Smyth LA, Lombardi G. Expression of a Chimeric Antigen Receptor Specific for Donor HLA Class I Enhances the Potency of Human Regulatory T Cells in Preventing Human Skin Transplant Rejection. Am J Transplant 2017; 17:931-943. [PMID: 28027623 DOI: 10.1111/ajt.14185] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/30/2016] [Accepted: 12/17/2016] [Indexed: 01/25/2023]
Abstract
Regulatory T cell (Treg) therapy using recipient-derived Tregs expanded ex vivo is currently being investigated clinically by us and others as a means of reducing allograft rejection following organ transplantation. Data from animal models has demonstrated that adoptive transfer of allospecific Tregs offers greater protection from graft rejection compared to polyclonal Tregs. Chimeric antigen receptors (CAR) are clinically translatable synthetic fusion proteins that can redirect the specificity of T cells toward designated antigens. We used CAR technology to redirect human polyclonal Tregs toward donor-MHC class I molecules, which are ubiquitously expressed in allografts. Two novel HLA-A2-specific CARs were engineered: one comprising a CD28-CD3ζ signaling domain (CAR) and one lacking an intracellular signaling domain (ΔCAR). CAR Tregs were specifically activated and significantly more suppressive than polyclonal or ΔCAR Tregs in the presence of HLA-A2, without eliciting cytotoxic activity. Furthermore, CAR and ΔCAR Tregs preferentially transmigrated across HLA-A2-expressing endothelial cell monolayers. In a human skin xenograft transplant model, adoptive transfer of CAR Tregs alleviated the alloimmune-mediated skin injury caused by transferring allogeneic peripheral blood mononuclear cells more effectively than polyclonal Tregs. Our results demonstrated that the use of CAR technology is a clinically applicable refinement of Treg therapy for organ transplantation.
Collapse
Affiliation(s)
- D A Boardman
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| | - C Philippeos
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - G O Fruhwirth
- Department of Imaging Chemistry and Biology, Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, London, UK
| | - M A A Ibrahim
- Department of Clinical Immunology and Allergy, King's College London, King's College Hospital, London, UK.,Division of Asthma, Allergy & Lung Biology, King's College London, Guy's Hospital, London, UK
| | - R F Hannen
- Centre for Cell Biology & Cutaneous Research, Bart's and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - D Cooper
- William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, London, UK
| | - F M Marelli-Berg
- William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, London, UK
| | - F M Watt
- NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK.,Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Hospital, London, UK
| | - R I Lechler
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| | - J Maher
- NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK.,Department of Clinical Immunology and Allergy, King's College London, King's College Hospital, London, UK.,CAR Mechanics Group, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | - L A Smyth
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,School of Health, Sport and Bioscience, Stratford Campus, University of East London, London, UK
| | - G Lombardi
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK.,NIHR Biomedical Research Centre, Guy's & St Thomas' NHS Foundation Trust & King's College London, Guy's Hospital, London, UK
| |
Collapse
|
14
|
Esensten JH, Helou YA, Chopra G, Weiss A, Bluestone JA. CD28 Costimulation: From Mechanism to Therapy. Immunity 2016; 44:973-88. [PMID: 27192564 PMCID: PMC4932896 DOI: 10.1016/j.immuni.2016.04.020] [Citation(s) in RCA: 552] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Indexed: 02/07/2023]
Abstract
Ligation of the CD28 receptor on T cells provides a critical second signal alongside T cell receptor (TCR) ligation for naive T cell activation. Here, we discuss the expression, structure, and biochemistry of CD28 and its ligands. CD28 signals play a key role in many T cell processes, including cytoskeletal remodeling, production of cytokines, survival, and differentiation. CD28 ligation leads to unique epigenetic, transcriptional, and post-translational changes in T cells that cannot be recapitulated by TCR ligation alone. We discuss the function of CD28 and its ligands in both effector and regulatory T cells. CD28 is critical for regulatory T cell survival and the maintenance of immune homeostasis. We outline the roles that CD28 and its family members play in human disease and we review the clinical efficacy of drugs that block CD28 ligands. Despite the centrality of CD28 and its family members and ligands to immune function, many aspects of CD28 biology remain unclear. Translation of a basic understanding of CD28 function into immunomodulatory therapeutics has been uneven, with both successes and failures. Such real-world results might stem from multiple factors, including complex receptor-ligand interactions among CD28 family members, differences between the mouse and human CD28 families, and cell-type specific roles of CD28 family members.
Collapse
Affiliation(s)
- Jonathan H Esensten
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA.
| | - Ynes A Helou
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA 94143, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue Center for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Arthur Weiss
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, University of California, San Francisco, CA 94143, USA
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
15
|
Koh S, Shimasaki N, Bertoletti A. Redirecting T Cell Specificity Using T Cell Receptor Messenger RNA Electroporation. Methods Mol Biol 2016; 1428:285-96. [PMID: 27236807 DOI: 10.1007/978-1-4939-3625-0_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Autologous T lymphocytes genetically modified to express T cell receptors or chimeric antigen receptors have shown great promise in the treatment of several cancers, including melanoma and leukemia. In addition to tumor-associated antigens and tumor-specific neoantigens, tumors expressing viral peptides can also be recognized by specific T cells and are attractive targets for cell therapy. Hepatocellular carcinoma cells often have hepatitis B virus DNA integration and can be targeted by hepatitis B virus-specific T cells. Here, we describe a method to engineer hepatitis B virus-specific T cell receptors in primary human T lymphocytes based on electroporation of hepatitis B virus T cell receptor messenger RNA. This method can be extended to a large scale therapeutic T cell production following current good manufacturing practice compliance and is applicable to the redirection of T lymphocytes with T cell receptors of other virus specificities such as Epstein-Barr virus, cytomegalovirus, and chimeric receptors specific for other antigens expressed on cancer cells.
Collapse
Affiliation(s)
- Sarene Koh
- Singapore Institute for Clinical Sciences, Brenner Center for Molecular Medicine, Agency for Science, Technology and Research, (A*STAR), 30 Medical Drive, Singapore, Singapore.
| | - Noriko Shimasaki
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonio Bertoletti
- Emerging Infectious Diseases Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
16
|
Abstract
Induction of specific immune tolerance to grafts remains the sought-after standard following transplantation. Defined by expression of the Foxp3 (forkhead box protein 3) transcription factor, the regulatory T-cell (Treg) lineage has been noted to exert potent immunoregulatory functions that contribute to specific graft tolerance. In this review, we discuss the known signals and pathways which govern Treg development, both in the thymus and in peripheral sites, as well as lineage maintenance and homeostasis. In particular, we highlight the roles of T-cell receptor signaling, CD28 costimulation, and signals through phosphatidyl inositol 3-kinase (PI3K) and related metabolic pathways in multiple aspects of Treg biology.
Collapse
Affiliation(s)
- Alexandria Huynh
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA; Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
17
|
Li P, Mao L, Liu X, Gan Y, Zheng J, Thomson AW, Gao Y, Chen J, Hu X. Essential role of program death 1-ligand 1 in regulatory T-cell-afforded protection against blood-brain barrier damage after stroke. Stroke 2014; 45:857-64. [PMID: 24496394 DOI: 10.1161/strokeaha.113.004100] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Our recent research revealed that adoptively transferred regulatory T cells (Tregs) reduced acute ischemic brain injury by inhibiting neutrophil-derived matrix metalloproteinase-9 (MMP-9) and protecting against blood-brain barrier damage. The mechanisms underlying Treg interactions with neutrophils remain elusive. This study evaluates the contribution of program death 1-ligand 1 (PD-L1) to Treg-mediated neutrophil inhibition and neuroprotection after cerebral ischemia. METHODS In vitro experiments were performed using a transwell system or a coculture system allowing cell-to-cell contact. Focal cerebral ischemia was induced in mice for 60 minutes. Tregs (2×10(6)) isolated from donor animals (wild-type or PD-L1-/-) were intravenously injected into ischemic recipients 2 hours after middle cerebral artery occlusion (MCAO). MMP-9 production, blood-brain barrier permeability, and brain infarct were assessed at 1 or 3 days after MCAO. RESULTS In vitro experiments reveal that Treg-mediated inhibition of neutrophil MMP-9 required direct cell-to-cell contact. The suppression of MMP-9 was abolished when Tregs were pretreated with PD-L1 neutralizing antibodies or when neutrophils were pretreated with PD-1 antibodies. In vivo studies confirmed that intravenous administration of Tregs pretreated with PD-L1 antibodies or Tregs isolated from PD-L1-deficient mice failed to inhibit MMP-9 production by blood neutrophils 1 day after 60 minutes MCAO. Furthermore, the blood-brain barrier damage after MCAO was greatly ameliorated in PD-L1-competent Treg-treated mice but not in PD-L1-compromised Treg-treated mice. Consequently, PD-L1 dysfunction abolished Treg-mediated brain protection and neurological improvements 3 days after MCAO. CONCLUSIONS PD-L1 plays an essential role in the neuroprotection afforded by Tregs against cerebral ischemia by mediating the suppressive effect of Tregs on neutrophil-derived MMP-9.
Collapse
Affiliation(s)
- Peiying Li
- From the State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China (P.L., L.M., X.L., Y. Gao, J.C., X.H.); Center of Cerebrovascular Disease Research (P.L., L.M., Y. Gan, J.Z., J.C., X.H.) and Departments of Surgery and Immunology, Starzl Transplantation Institute (A.W.T.), University of Pittsburgh School of Medicine, PA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (P.L., X.H.); and Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, People's Republic of China (X.L.)
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Li P, Gan Y, Sun BL, Zhang F, Lu B, Gao Y, Liang W, Thomson AW, Chen J, Hu X. Adoptive regulatory T-cell therapy protects against cerebral ischemia. Ann Neurol 2013; 74:458-71. [PMID: 23674483 DOI: 10.1002/ana.23815] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/26/2012] [Accepted: 11/16/2012] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Recent evidence suggests that functional deficiency in regulatory T cells (Tregs), an innate immunomodulator, exacerbates brain damage after cerebral ischemia. We therefore evaluated the effect of Treg transfer in rodent models of ischemic stroke and further investigated the mechanism underlying Treg-afforded neuroprotection. METHODS We examined the therapeutic potential of Tregs and the mechanisms of neuroprotection in vivo in 2 rodent models of ischemic stroke and in vitro in Treg-neutrophil cocultures using a combined approach including cell-specific depletion, gene knockout mice, and bone marrow chimeras. RESULTS Systemic administration of purified Tregs at 2, 6, or even 24 hours after middle cerebral artery occlusion resulted in a marked reduction of brain infarct and prolonged improvement of neurological functions lasting out to 4 weeks. Treg-afforded neuroprotection was accompanied by attenuated blood-brain barrier (BBB) disruption during early stages of ischemia, decreased cerebral inflammation, and reduced infiltration of peripheral inflammatory cells into the lesioned brain. Surprisingly, Tregs exerted early neuroprotection without penetrating into the brain parenchyma or inhibiting the activation of residential microglia. Rather, both in vivo and in vitro studies demonstrated that Tregs suppressed peripheral neutrophil-derived matrix metallopeptidase-9 production, thus preventing proteolytic damage of the BBB. In addition to its potent central neuroprotection, Treg treatment was shown to ameliorate poststroke lymphopenia, suggesting a beneficial effect on immune status. INTERPRETATION Our study suggests that Treg adoptive therapy is a novel and potent cell-based therapy targeting poststroke inflammatory dysregulation and neurovascular disruption.
Collapse
Affiliation(s)
- Peiying Li
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai, China; Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
The role of positive costimulatory molecules in transplantation and tolerance. Curr Opin Organ Transplant 2012; 13:366-72. [PMID: 18685331 DOI: 10.1097/mot.0b013e328306115b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW The manipulation of costimulatory pathways holds tremendous potential for treating immunologically mediated diseases. In this article, we review the role of molecules that deliver a positive second signal that, together with an antigen-specific signal from the T-cell receptor, is necessary to promote complete T-cell activation, differentiation and development of effector function. RECENT FINDINGS Numerous positive costimulatory molecules have been identified: CD28/B7, induced costimulatory/induced costimulatory ligand, CD40/CD154, OX40/OX40L, CD27/CD70, 4-1BB/4-1BBL, LIGHT/herpes virus entry mediator, glucosyltransferase R and T-cell immunoglobulin mucin molecules. Many of these have been only recently discovered and remain incompletely studied. Recent work has demonstrated that some costimulatory molecules bind ligands expressed by nonprofessional activated protein C, some modulate regulatory T cells and some sustain rather than initiate immune responses. Emerging data suggest that the costimulatory pathways are redundant and that the various costimulatory molecules affect different T-cell populations and act at different times during the course of the immune response. SUMMARY These observations suggest that the therapeutic exploitation of strategies targeting costimulatory molecules will require carefully timed interventions directed against multiple pathways.
Collapse
|
20
|
Chikamatsu K, Sakakura K, Toyoda M, Takahashi K, Yamamoto T, Masuyama K. Immunosuppressive activity of CD14+ HLA-DR- cells in squamous cell carcinoma of the head and neck. Cancer Sci 2012; 103:976-83. [PMID: 22360618 PMCID: PMC7685071 DOI: 10.1111/j.1349-7006.2012.02248.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/10/2012] [Accepted: 02/20/2012] [Indexed: 01/04/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSC) represent a heterogeneous population and have the potential to suppress immune responses via diverse mechanisms. In recent studies, a new subset of MDSC was identified by the markers CD14(+) and HLA-DR(-) in the peripheral blood from cancer patients. In this study, we investigated the proportions and characteristics of CD14(+) HLA-DR(-) cells in patients with squamous cell carcinoma of the head and neck (SCCHN). As expected, the percentage of CD14(+) HLA-DR(-) cells was significantly elevated in patients relative to healthy donors and the sorted CD14(+) HLA-DR(-) cells were able to suppress effectively both the proliferation and IFN-γ production of anti-CD3/anti-CD28 stimulated T cells, suggesting that CD14(+) HLA-DR(-) cells in patients with SCCHN contribute to the immune suppressive status. Furthermore, CD14(+) HLA-DR(-) cells revealed a higher level of CD86 and PD-L1 expression and transforming growth factor (TGF)-β production than CD14(+) HLA-DR(+) cells. Addition of anti-CD86 mAb, anti-PD-L1 mAb and anti-TGF-β mAb partially restored T-cell proliferation and IFN-γ production, respectively, indicating that the suppressive effects of CD14(+) HLA-DR(-) cells appear to be mediated by various molecules, including coinhibitory molecules and cytokines. Our data suggest that CD14(+) HLA-DR(-) cells act as potent immunosuppressive cells and particularly contribute to tumor escape from the host immune system in patients with SCCHN.
Collapse
Affiliation(s)
- Kazuaki Chikamatsu
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan.
| | | | | | | | | | | |
Collapse
|
21
|
Bonacci B, Edwards B, Jia S, Williams CB, Hessner MJ, Gauld SB, Verbsky JW. Requirements for growth and IL-10 expression of highly purified human T regulatory cells. J Clin Immunol 2012; 32:1118-28. [PMID: 22562448 DOI: 10.1007/s10875-012-9701-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/13/2012] [Indexed: 01/07/2023]
Abstract
Human regulatory T cells (T(R)) cells have potential for the treatment of a variety of immune mediated diseases but the anergic phenotype of these cells makes them difficult to expand in vitro. We have examined the requirements for growth and cytokine expression from highly purified human T(R) cells, and correlated these findings with the signal transduction events of these cells. We demonstrate that these cells do not proliferate or secrete IL-10 even in the presence of high doses of IL-2. Stimulation with a superagonistic anti-CD28 antibody (clone 9.3) and IL-2 partially reversed the proliferative defect, and this correlated with reversal of the defective calcium mobilization in these cells. Dendritic cells were effective at promoting T(R) cell proliferation, and under these conditions the proliferative capacity of T(R) cells was comparable to conventional CD4 lymphocytes. Blocking TGF-β activity abrogated IL-10 expression from these cells, while addition of TGF-β resulted in IL-10 production. These data demonstrate that highly purified populations of T(R) cells are anergic even in the presence of high doses of IL-2. Furthermore, antigen presenting cells provide proper co-stimulation to overcome the anergic phenotype of T(R) cells, and under these conditions they are highly sensitive to IL-2. In addition, these data demonstrate for the first time that TGF-β is critical to enable human T(R) cells to express IL-10.
Collapse
Affiliation(s)
- Benedetta Bonacci
- Department of Pediatrics, Medical College of Wisconsin and the Children's Research Institute, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Lichtenfels R, Rappl G, Hombach AA, Recktenwald CV, Dressler SP, Abken H, Seliger B. A proteomic view at T cell costimulation. PLoS One 2012; 7:e32994. [PMID: 22539942 PMCID: PMC3335147 DOI: 10.1371/journal.pone.0032994] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 02/07/2012] [Indexed: 12/31/2022] Open
Abstract
The "two-signal paradigm" in T cell activation predicts that the cooperation of "signal 1," provided by the T cell receptor (TCR) through engagement of major histocompatility complex (MHC)-presented peptide, with "signal 2″ provided by costimulatory molecules, the prototype of which is CD28, is required to induce T cell effector functions. While the individual signalling pathways are well understood, little is known about global changes in the proteome pattern during TCR/CD28-mediated activation. Therefore, comparative 2-DE-based proteome analyses of CD3(+) CD69(-) resting T cells versus cells incubated with (i) the agonistic anti-CD3 antibody OKT3 mimicking signal 1 in absence or presence of IL-2 and/or with (ii) the agonistic antibody 15E8 triggering CD28-mediated signaling were performed. Differentially regulated spots were defined leading to the identification of proteins involved in the regulation of the metabolism, shaping and maintenance of the cytoskeleton and signal transduction. Representative members of the differentially expressed protein families, such as calmodulin (CALM), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), L-lactate dehydrogenase (LDH), Rho GDP-dissociation inhibitor 2 (GDIR2), and platelet basic protein (CXCL7), were independently verified by flow cytometry. Data provide a detailed map of individual protein alterations at the global proteome level in response to TCR/CD28-mediated T cell activation.
Collapse
Affiliation(s)
- Rudolf Lichtenfels
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Andreas A. Hombach
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | | | - Sven P. Dressler
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC) and Tumor Genetics Section, Clinic I Internal Medicine, University Cologne, Cologne, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
23
|
Hess J, Ruf P, Lindhofer H. Cancer therapy with trifunctional antibodies: linking innate and adaptive immunity. Future Oncol 2012; 8:73-85. [PMID: 22149036 DOI: 10.2217/fon.11.138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Trifunctional antibodies (trAbs) are promising novel anticancer biologics with a particular mode of action capable of linking innate with adaptive immunity. Based on their unique structure, trifunctional IgG-like heterodimeric antibodies, consisting of nonhuman mouse and rat immunoglobulin halves are able to redirect T lymphocytes, as well as accessory cells, to the tumor site. This recruitment of immune cells is accompanied by cellular activation events elicited by anti-CD3, as well as Fcγ-receptor engagement of trAbs supported by a proinflammatory Th1-biased cytokine milieu. All necessary immunological factors required for long-term vaccination-like effects are stimulated along trAb-mediated therapeutic interventions. Thus, the concerted interplay of antibody-dependent cellular cytotoxicity plus the polyclonal T-cell cytotoxicity and Fcγ-receptor-driven induction of long-lasting immune responses after the initial tumor cell elimination represent the major hallmarks of trAb-mediated treatment of malignant diseases.
Collapse
Affiliation(s)
- Juergen Hess
- TRION Pharma GmbH, Frankfurter Ring 193a, 80807 Munich, Germany
| | | | | |
Collapse
|
24
|
Dangi A, Sumpter TL, Kimura S, Stolz DB, Murase N, Raimondi G, Vodovotz Y, Huang C, Thomson AW, Gandhi CR. Selective expansion of allogeneic regulatory T cells by hepatic stellate cells: role of endotoxin and implications for allograft tolerance. THE JOURNAL OF IMMUNOLOGY 2012; 188:3667-77. [PMID: 22427640 DOI: 10.4049/jimmunol.1102460] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatic stellate cells (HSCs) may play an important role in hepatic immune regulation by producing numerous cytokines/chemokines and expressing Ag-presenting and T cell coregulatory molecules. Due to disruption of the endothelial barrier during cold-ischemic storage and reperfusion of liver grafts, HSCs can interact directly with cells of the immune system. Endotoxin (LPS), levels of which increase in liver diseases and transplantation, stimulates the synthesis of many mediators by HSCs. We hypothesized that LPS-stimulated HSCs might promote hepatic tolerogenicity by influencing naturally occurring immunosuppressive CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Following their portal venous infusion, allogeneic CD4(+) T cells, including Tregs, were found closely associated with HSCs, and this association increased in LPS-treated livers. In vitro, both unstimulated and LPS-stimulated HSCs upregulated Fas (CD95) expression on conventional CD4(+) T cells and induced their apoptosis in a Fas/Fas ligand-dependent manner. By contrast, HSCs induced Treg proliferation, which required cell-cell contact and was MHC class II-dependent. This effect was augmented when HSCs were pretreated with LPS. LPS increased the expression of MHC class II, CD80, and CD86 and stimulated the production of IL-1α, IL-1β, IL-6, IL-10 and TNF-α by HSCs. Interestingly, production of IL-1α, IL-1β, IL-6, and TNF-α was strongly inhibited, but that of IL-10 enhanced in LPS-pretreated HSC/Treg cocultures. Adoptively transferred allogeneic HSCs migrated to the secondary lymphoid tissues and induced Treg expansion in lymph nodes. These data implicate endotoxin-stimulated HSCs as important immune regulators in liver transplantation by inducing selective expansion of tolerance-promoting Tregs and reducing inflammation and alloimmunity.
Collapse
Affiliation(s)
- Anil Dangi
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen Y, Shen S, Gorentla B, Gao J, Zhong XP. Murine regulatory T cells contain hyperproliferative and death-prone subsets with differential ICOS expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:1698-707. [PMID: 22231701 PMCID: PMC3273604 DOI: 10.4049/jimmunol.1102448] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Regulatory T cells (Treg) are crucial for self-tolerance. It has been an enigma that Treg exhibit an anergic phenotype reflected by hypoproliferation in vitro after TCR stimulation but undergo vigorous proliferation in vivo. We report in this study that murine Treg are prone to death but hyperproliferative in vitro and in vivo, which is different from conventional CD4(+)Foxp3(-) T cells (Tcon). During in vitro culture, most Treg die with or without TCR stimulation, correlated with constitutive activation of the intrinsic death pathway. However, a small portion of the Treg population is more sensitive to TCR stimulation, particularly weak stimulation, proliferates more vigorously than CD4(+) Tcon, and is resistant to activation-induced cell death. Treg proliferation is enhanced by IL-2 but is less dependent on CD28-mediated costimulation than that of Tcon. We demonstrate further that the surviving and proliferative Treg are ICOS(+) whereas the death-prone Treg are ICOS(-). Moreover, ICOS(+) Treg contain much stronger suppressive activity than that of ICOS(-) Treg. Our data indicate that massive death contributes to the anergic phenotype of Treg in vitro and suggest modulation of Treg survival as a therapeutic strategy for treatment of autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Yong Chen
- Department of Pediatrics-Allergy and Immunology Duke University Medical Center, Durham, NC 27710
- School of Laboratory Medicine, Wenzhou Medical College, Wenzhou, Zhejiang Province 325035, China
| | - Shudan Shen
- Department of Pediatrics-Allergy and Immunology Duke University Medical Center, Durham, NC 27710
| | - Balachandra Gorentla
- Department of Pediatrics-Allergy and Immunology Duke University Medical Center, Durham, NC 27710
| | - Jimin Gao
- School of Laboratory Medicine, Wenzhou Medical College, Wenzhou, Zhejiang Province 325035, China
| | - Xiao-Ping Zhong
- Department of Pediatrics-Allergy and Immunology Duke University Medical Center, Durham, NC 27710
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
26
|
Zhao Y, Jiang Z, Zhao T, Ye M, Hu C, Yin Z, Li H, Zhang Y, Diao Y, Li Y, Chen Y, Sun X, Fisk MB, Skidgel R, Holterman M, Prabhakar B, Mazzone T. Reversal of type 1 diabetes via islet β cell regeneration following immune modulation by cord blood-derived multipotent stem cells. BMC Med 2012; 10:3. [PMID: 22233865 PMCID: PMC3322343 DOI: 10.1186/1741-7015-10-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/10/2012] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Inability to control autoimmunity is the primary barrier to developing a cure for type 1 diabetes (T1D). Evidence that human cord blood-derived multipotent stem cells (CB-SCs) can control autoimmune responses by altering regulatory T cells (Tregs) and human islet β cell-specific T cell clones offers promise for a new approach to overcome the autoimmunity underlying T1D. METHODS We developed a procedure for Stem Cell Educator therapy in which a patient's blood is circulated through a closed-loop system that separates lymphocytes from the whole blood and briefly co-cultures them with adherent CB-SCs before returning them to the patient's circulation. In an open-label, phase1/phase 2 study, patients (n=15) with T1D received one treatment with the Stem Cell Educator. Median age was 29 years (range: 15 to 41), and median diabetic history was 8 years (range: 1 to 21). RESULTS Stem Cell Educator therapy was well tolerated in all participants with minimal pain from two venipunctures and no adverse events. Stem Cell Educator therapy can markedly improve C-peptide levels, reduce the median glycated hemoglobin A1C (HbA1C) values, and decrease the median daily dose of insulin in patients with some residual β cell function (n=6) and patients with no residual pancreatic islet β cell function (n=6). Treatment also produced an increase in basal and glucose-stimulated C-peptide levels through 40 weeks. However, participants in the Control Group (n=3) did not exhibit significant change at any follow-up. Individuals who received Stem Cell Educator therapy exhibited increased expression of co-stimulating molecules (specifically, CD28 and ICOS), increases in the number of CD4+CD25+Foxp3+ Tregs, and restoration of Th1/Th2/Th3 cytokine balance. CONCLUSIONS Stem Cell Educator therapy is safe, and in individuals with moderate or severe T1D, a single treatment produces lasting improvement in metabolic control. Initial results indicate Stem Cell Educator therapy reverses autoimmunity and promotes regeneration of islet β cells. Successful immune modulation by CB-SCs and the resulting clinical improvement in patient status may have important implications for other autoimmune and inflammation-related diseases without the safety and ethical concerns associated with conventional stem cell-based approaches. TRIAL REGISTRATION ClinicalTrials.gov number, NCT01350219.
Collapse
Affiliation(s)
- Yong Zhao
- Section of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Illinois at Chicago, 1819 West Polk Street, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family. Immunol Rev 2011; 241:180-205. [PMID: 21488898 PMCID: PMC3077803 DOI: 10.1111/j.1600-065x.2011.01011.x] [Citation(s) in RCA: 296] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Positive and negative costimulation by members of the CD28 family is critical for the development of productive immune responses against foreign pathogens and their proper termination to prevent inflammation-induced tissue damage. In addition, costimulatory signals are critical for the establishment and maintenance of peripheral tolerance. This paradigm has been established in many animal models and has led to the development of immunotherapies targeting costimulation pathways for the treatment of cancer, autoimmune disease, and allograft rejection. During the last decade, the complexity of the biology of costimulatory pathways has greatly increased due to the realization that costimulation does not affect only effector T cells but also influences regulatory T cells and antigen-presenting cells. Thus, costimulation controls T-cell tolerance through both intrinsic and extrinsic pathways. In this review, we discuss the influence of costimulation on intrinsic and extrinsic pathways of peripheral tolerance, with emphasis on members of the CD28 family, CD28, cytotoxic T-lymphocyte antigen-4 (CTLA-4), and programmed death-1 (PD-1), as well as the downstream cytokine interleukin-1 (IL-2).
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA 94143-0400, USA
| | | | | | | | | | | |
Collapse
|
28
|
Baltcheva I, Codarri L, Pantaleo G, Le Boudec JY. Lifelong dynamics of human CD4+CD25+ regulatory T cells: insights from in vivo data and mathematical modeling. J Theor Biol 2010; 266:307-22. [PMID: 20600134 DOI: 10.1016/j.jtbi.2010.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Revised: 06/14/2010] [Accepted: 06/15/2010] [Indexed: 11/25/2022]
Abstract
Despite their limited proliferation capacity, regulatory T cells (T(regs)) constitute a population maintained over the entire lifetime of a human organism. The means by which T(regs) sustain a stable pool in vivo are controversial. Using a mathematical model, we address this issue by evaluating several biological scenarios of the origins and the proliferation capacity of two subsets of T(regs): precursor CD4(+)CD25(+)CD45RO(-) and mature CD4(+)CD25(+)CD45RO(+) cells. The lifelong dynamics of T(regs) are described by a set of ordinary differential equations, driven by a stochastic process representing the major immune reactions involving these cells. The model dynamics are validated using data from human donors of different ages. Analysis of the data led to the identification of two properties of the dynamics: (1) the equilibrium in the CD4(+)CD25(+)FoxP3(+)T(regs) population is maintained over both precursor and mature T(regs) pools together, and (2) the ratio between precursor and mature T(regs) is inverted in the early years of adulthood. Then, using the model, we identified three biologically relevant scenarios that have the above properties: (1) the unique source of mature T(regs) is the antigen-driven differentiation of precursors that acquire the mature profile in the periphery and the proliferation of T(regs) is essential for the development and the maintenance of the pool; there exist other sources of mature T(regs), such as (2) a homeostatic density-dependent regulation or (3) thymus- or effector-derived T(regs), and in both cases, antigen-induced proliferation is not necessary for the development of a stable pool of T(regs). This is the first time that a mathematical model built to describe the in vivo dynamics of regulatory T cells is validated using human data. The application of this model provides an invaluable tool in estimating the amount of regulatory T cells as a function of time in the blood of patients that received a solid organ transplant or are suffering from an autoimmune disease.
Collapse
Affiliation(s)
- Irina Baltcheva
- Laboratory for Computer Communications and Applications, Ecole Polytechnique Fédérale de Lausanne, EPFL IC-LCA, Batiment BC 258, Station 14, CH-1015 Lausanne, Switzerland.
| | | | | | | |
Collapse
|
29
|
Mittag D, Scholzen A, Varese N, Baxter L, Paukovics G, Harrison LC, Rolland JM, O'Hehir RE. The effector T cell response to ryegrass pollen is counterregulated by simultaneous induction of regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:4708-16. [PMID: 20308632 DOI: 10.4049/jimmunol.0901036] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Allergy is associated with pathological Th2 responses to otherwise harmless environmental Ags. In contrast, nonallergic individuals mount nonpathological immune responses to allergens, partly attributed to regulatory T cell (Treg) activity. Although thymus-derived natural Tregs have been shown to maintain tolerance to self-Ags and prevent autoimmunity, the generation of Tregs specific to non-self-Ags is less well understood. We investigated the potential for induction of Tregs from PBMCs of ryegrass pollen-allergic or healthy subjects by stimulation in vitro with ryegrass pollen extract in the absence of additional exogenous stimuli. We found that two subsets of proliferating CD4(+) T cells were induced, one expressing intermediate levels of Foxp3 (and IFN-gamma, IL-4, IL-17, or IL-2) and the other expressing high levels of Foxp3 (and no effector cytokines). After enrichment based on CD39 expression, the Foxp3(hi) subset suppressed CD4(+) T cell proliferation and IFN-gamma production. The Foxp3(hi) Treg originated from both conversion of dividing non-Tregs (CD4(+)CD25(-)CD127(hi)) and expansion of natural Tregs (CD4(+)CD25(+)CD127(lo)). Stable functional Tregs expressing high levels of Foxp3 were induced simultaneously with effector T cells by allergen stimulation. Induction of Foxp3(hi) Tregs was reduced in allergic subjects. These results indicate that the cogeneration of Foxp3(hi) Tregs in response to allergen may be a mechanism for controlling allergic reactions in healthy individuals, which is impaired in those with allergies.
Collapse
Affiliation(s)
- Diana Mittag
- Department of Immunology, Monash University, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Schatton T, Schütte U, Frank NY, Zhan Q, Hoerning A, Robles SC, Zhou J, Hodi FS, Spagnoli GC, Murphy GF, Frank MH. Modulation of T-cell activation by malignant melanoma initiating cells. Cancer Res 2010; 70:697-708. [PMID: 20068175 DOI: 10.1158/0008-5472.can-09-1592] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Highly immunogenic cancers such as malignant melanoma are capable of inexorable tumor growth despite the presence of antitumor immunity. Thus, only a restricted minority of tumorigenic malignant cells may possess the phenotypic and functional characteristics needed to modulate tumor-directed immune activation. Here we provide evidence supporting this hypothesis. Tumorigenic ABCB5(+) malignant melanoma initiating cells (MMICs) possessed the capacity to preferentially inhibit IL-2-dependent T-cell activation and to support, in a B7.2-dependent manner, induction of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs). Compared with melanoma bulk cell populations, ABCB5(+) MMICs displayed lower levels of MHC class I, aberrant positivity for MHC class II, and lower expression levels of the melanoma-associated antigens MART-1, ML-IAP, NY-ESO-1, and MAGE-A. Additionally, these tumorigenic ABCB5(+) subpopulations preferentially expressed the costimulatory molecules B7.2 and PD-1, both in established melanoma xenografts and in clinical tumor specimens. In immune activation assays, MMICs inhibited mitogen-dependent human peripheral blood mononuclear cell (PBMC) proliferation and IL-2 production more efficiently than ABCB5(-) melanoma cell populations. Moreover, coculture with ABCB5(+) MMICs increased the abundance of Tregs, in a B7.2 signaling-dependent manner, along with IL-10 production by mitogen-activated PBMCs. Consistent with these findings, MMICs also preferentially inhibited IL-2 production and induced IL-10 secretion by cocultured patient-derived, syngeneic PBMCs. Our findings identify novel T-cell modulatory functions of ABCB5(+) melanoma subpopulations and suggest specific roles for these MMICs in the evasion of antitumor immunity and in cancer immunotherapeutic resistance.
Collapse
Affiliation(s)
- Tobias Schatton
- Transplantation Research Center, Children's Hospital Boston, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Self-renewing cancer stem cells (CSC) capable of spawning more differentiated tumor cell progeny are required for tumorigenesis and neoplastic progression of leukemias and several solid cancers. The mechanisms by which CSC cause tumor initiation and growth are currently unknown. Recent findings that suggest a negative correlation between degrees of host immunocompetence and rates of cancer development raise the possibility that only a restricted minority of malignant cells, namely CSC, may possess the phenotypic and functional characteristics to evade host antitumor immunity. In human malignant melanoma, a highly immunogenic cancer, we recently identified malignant melanoma initiating cells (MMIC), a novel type of CSC, based on selective expression of the chemoresistance mediator ABCB5. Here we present evidence of a relative immune privilege of ABCB5(+) MMIC, suggesting refractoriness to current immunotherapeutic treatment strategies. We discuss our findings in the context of established immunomodulatory functions of physiologic stem cells and in relation to mechanisms responsible for the downregulation of immune responses against tumors. We propose that the MMIC subset might be responsible for melanoma immune evasion and that immunomodulation might represent one mechanism by which CSC advance tumorigenic growth and resistance to immunotherapy. Accordingly, the possibility of an MMIC-driven tumor escape from immune-mediated rejection has important implications for current melanoma immunotherapy.
Collapse
Affiliation(s)
- Tobias Schatton
- Transplantation Research Center, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
32
|
Zouggari Y, Ait-Oufella H, Waeckel L, Vilar J, Loinard C, Cochain C, Récalde A, Duriez M, Levy BI, Lutgens E, Lutgens E, Mallat Z, Silvestre JS. Regulatory T cells modulate postischemic neovascularization. Circulation 2009; 120:1415-25. [PMID: 19770391 DOI: 10.1161/circulationaha.109.875583] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND CD4+ and CD8+ T lymphocytes are key regulators of postischemic neovascularization. T-cell activation is promoted by 2 major costimulatory signalings, the B7/CD28 and CD40-CD40 ligand pathways. Interestingly, CD28 interactions with the structurally related ligands B7-1 and B7-2 are also required for the generation and homeostasis of CD4+CD25+ regulatory T cells (Treg cells), which play a critical role in the suppression of immune responses and the control of T-cell homeostasis. We hypothesized that Treg cell activation may modulate the immunoinflammatory response to ischemic injury, leading to alteration of postischemic vessel growth. METHODS AND RESULTS Ischemia was induced by right femoral artery ligation in CD28-, B7-1/2-, or CD40-deficient mice (n=10 per group). CD40 deficiency led to a significant reduction in the postischemic inflammatory response and vessel growth. In contrast, at day 21 after ischemia, angiographic score, foot perfusion, and capillary density were increased by 2.0-, 1.2-, and 1.8-fold, respectively, in CD28-deficient mice, which showed a profound reduction in the number of Treg cells compared with controls. Similarly, disruption of B7-1/2 signaling or anti-CD25 treatment and subsequent Treg deletion significantly enhanced postischemic neovascularization. These effects were associated with enhanced accumulation of CD3-positive T cells and Mac-3-positive macrophages in the ischemic leg. Conversely, treatment of CD28(-/-) mice with the nonmitogenic anti-CD3 monoclonal antibody enhanced the number of endogenous Treg cells and led to a significant reduction of the postischemic inflammatory response and neovascularization. Finally, coadministration of Treg cells and CD28(-/-) splenocytes in Rag1(-/-) mice with hindlimb ischemia abrogated the CD28(-/-) splenocyte-induced activation of the inflammatory response and neovascularization. CONCLUSIONS Treg cell response modulates postischemic neovascularization.
Collapse
Affiliation(s)
- Yasmine Zouggari
- Paris-Cardiovascular Research Center, INSERM U970, Hôpital Européen Georges Pompidou, Université Paris 5, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hombach AA, Kofler D, Rappl G, Abken H. Redirecting human CD4+CD25+ regulatory T cells from the peripheral blood with pre-defined target specificity. Gene Ther 2009; 16:1088-96. [PMID: 19554034 DOI: 10.1038/gt.2009.75] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent insight into the balance of self-tolerance and auto-aggression has raised interest in using human regulatory T (Treg) cells for adoptive immunotherapy of unlimited autoimmune diseases including type-1 diabetes, rhematoid arthritis and multiple sclerosis. The therapeutic use of Treg cells, however, is so far hampered by the inefficiency of current protocols in making them accessible for genetic manipulations. We report here that TCR/CD3 stimulation that is accompanied by extensive CD28 costimulation makes human Treg cells susceptible to retroviral gene transfer ex vivo while preserving their properties in vitro and in vivo. To show the power of genetic manipulation of human Treg cells, we engineered 'designer Treg cells' by retroviral expression of a chimeric immunoreceptor with defined specificity, which activates Treg cells in a ligand-dependent manner to proliferate, to secrete high amounts of interleukin-10 and to repress an ongoing cytolytic T-cell response in vivo. The procedure in genetically modifying human Treg cells ex vivo will open a panel of applications for their use in the adoptive therapy of deregulated immune responses.
Collapse
Affiliation(s)
- A A Hombach
- Zentrum für Molekulare Medizin Köln (ZMMK) and Klinik I für Innere Medizin, Labor Tumorgenetik, Universität zu Köln, Köln, Germany.
| | | | | | | |
Collapse
|
34
|
Moon C, Kim S, Park K, Choi B, Lee H, Park J, Choi G, Kwan J, Joh J, Kim S. Use of Epigenetic Modification to Induce FOXP3 Expression in Naïve T Cells. Transplant Proc 2009; 41:1848-54. [DOI: 10.1016/j.transproceed.2009.02.101] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2008] [Accepted: 02/17/2009] [Indexed: 01/19/2023]
|
35
|
Elinav E, Adam N, Waks T, Eshhar Z. Amelioration of colitis by genetically engineered murine regulatory T cells redirected by antigen-specific chimeric receptor. Gastroenterology 2009; 136:1721-31. [PMID: 19208357 DOI: 10.1053/j.gastro.2009.01.049] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Revised: 01/01/2009] [Accepted: 01/22/2009] [Indexed: 01/24/2023]
Abstract
BACKGROUND & AIMS The therapeutic application of regulatory T cells (Tregs) for the treatment of inflammatory diseases is limited by the scarcity of antigen-specific Tregs. A preferred approach to endow effector T cells (Teff) with a desired specificity uses chimeric immune receptors with antibody-type specificity. Accordingly, employing such chimeric immune receptors to redirect Tregs to sites of inflammation may be a useful therapeutic approach to alleviate a broad scope of diseases in which an uncontrolled inflammatory response plays a major role. METHODS To enable application of the approach in clinical setting, which requires the genetic modification of the patient's own Tregs, we describe here a novel protocol that allows the efficient retroviral transduction and 2,4,6-trinitrophenol-specific expansion of murine naturally occurring regulatory T cells (nTregs), with a 2,4,6-trinitrophenol-specific tripartite chimeric receptor. RESULTS Transduced Tregs maintained their Foxp3 level, could undergo repeated expansion upon ex vivo encounter with their cognate antigen in a major histocompatibility complex-independent, costimulation-independent, and contact-dependent manner and specifically suppressed Teff cells. Adoptive transfer of small numbers of the transduced nTregs was associated with antigen-specific, dose-dependent amelioration of trinitrobenzenesulphonic acid colitis. CONCLUSIONS This study demonstrates that nTregs can be efficiently transduced to express functional, antigen-specific chimeric receptors that enable the specific suppression of effector T cells both in vitro and in vivo. This approach may enable future cell-based therapeutic application in inflammatory bowel disease, as well as other inflammatory disorders.
Collapse
Affiliation(s)
- Eran Elinav
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | |
Collapse
|
36
|
Bour-Jordan H, Bluestone JA. Regulating the regulators: costimulatory signals control the homeostasis and function of regulatory T cells. Immunol Rev 2009; 229:41-66. [PMID: 19426214 PMCID: PMC2714548 DOI: 10.1111/j.1600-065x.2009.00775.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SUMMARY Costimulation is a concept that goes back to the early 1980s when Lafferty and others hypothesized that cell surface and soluble molecules must exist that are essential for initiating immune responses subsequent to antigen exposure. The explosion in this field of research ensued as over a dozen molecules have been identified to function as second signals following T-cell receptor engagement. By 1994, it seemed clear that the most prominent costimulatory pathway CD28 and functionally related costimulatory molecules, such as CD154, were the major drivers of a positive immune response. Then the immunology world turned upside down. CD28 knockout mice, which were, in most cases, immunodeficient, led to increased autoimmunity when bred into the non-obese diabetic background. Another CD28 family member, cytotoxic T-lymphocyte-associated protein 4, which was presumed to be a costimulatory molecule on activated T cells, turned out to be critical in downregulating immunity. These results, coupled with the vast suppressor cell literature which had been largely rebuked, suggested that the immune system was not poised for response but controlled in such a way that regulation was dominant. Over the last decade, we have learned that these costimulatory molecules play a key role in the now classical CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) that provide critical control of unwanted autoimmune responses. In this review, we discuss the connections between costimulation and Tregs that have changed the costimulation paradigm.
Collapse
Affiliation(s)
- Hélène Bour-Jordan
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA USA
| | - Jeffrey A. Bluestone
- UCSF Diabetes Center, University of California at San Francisco, San Francisco, CA USA
| |
Collapse
|
37
|
Putnam AL, Brusko TM, Lee MR, Liu W, Szot GL, Ghosh T, Atkinson MA, Bluestone JA. Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes 2009; 58:652-62. [PMID: 19074986 PMCID: PMC2646064 DOI: 10.2337/db08-1168] [Citation(s) in RCA: 287] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Regulatory T-cells (Tregs) have catalyzed the field of immune regulation. However, translating Treg-based therapies from animal models of autoimmunity to human clinical trials requires robust methods for the isolation and expansion of these cells-a need forming the basis for these studies. RESEARCH DESIGN AND METHODS Tregs from recent-onset type 1 diabetic patients and healthy control subjects were isolated by fluorescence-activated cell sorting and compared for their capacity to expand in vitro in response to anti-CD3-anti-CD28-coated microbeads and IL-2. Expanded cells were examined for suppressive function, lineage markers and FOXP3, and cytokine production. RESULTS Both CD4+CD127(lo/-) and CD4+CD127(lo/-)CD25+ T-cells could be expanded and used as Tregs. However, expansion of CD4+CD127(lo/-) cells required the addition of rapamycin to maintain lineage purity. In contrast, expansion of CD4+CD127(lo/-)CD25+ T-cells, especially the CD45RA+ subset, resulted in high yield, functional Tregs that maintained higher FOXP3 expression in the absence of rapamycin. Tregs from type 1 diabetic patients and control subjects expanded similarly and were equally capable of suppressing T-cell proliferation. Regulatory cytokines were produced by Tregs after culture; however, a portion of FOXP3+ cells were capable of producing interferon (IFN)-gamma after reactivation. IFN-gamma production was observed from both CD45RO+ and CD45RA+ Treg populations. CONCLUSIONS The results support the feasibility of isolating Tregs for in vitro expansion. Based on expansion capacity, FOXP3 stability, and functional properties, the CD4+CD127(lo/-)CD25+ T-cells represent a viable cell population for cellular therapy in this autoimmune disease.
Collapse
Affiliation(s)
- Amy L Putnam
- Diabetes Center at the University of California, San Francisco (UCSF), San Francisco, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Milojevic D, Nguyen KD, Wara D, Mellins ED. Regulatory T cells and their role in rheumatic diseases: a potential target for novel therapeutic development. Pediatr Rheumatol Online J 2008; 6:20. [PMID: 19046457 PMCID: PMC2633304 DOI: 10.1186/1546-0096-6-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 12/01/2008] [Indexed: 01/01/2023] Open
Abstract
Regulatory T cells have an important role in limiting immune reactions and are essential regulators of self-tolerance. Among them, CD4+CD25high regulatory T cells are the best-described subset. In this article, we summarize current knowledge on the phenotype, function, and development of CD4+CD25high regulatory T cells. We also review the literature on the role of these T cells in rheumatic diseases and discuss the potential for their use in immunotherapy.
Collapse
Affiliation(s)
- Diana Milojevic
- Department of Pediatrics, UCSF, San Francisco, CA 94143, USA
| | - Khoa D Nguyen
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Diane Wara
- Department of Pediatrics, UCSF, San Francisco, CA 94143, USA
| | | |
Collapse
|
39
|
Rappl G, Schmidt A, Mauch C, Hombach AA, Abken H. Extensive Amplification of Human Regulatory T Cells Alters Their Functional Capacities and Targets Them to the Periphery. Rejuvenation Res 2008; 11:915-33. [DOI: 10.1089/rej.2008.0723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Gunter Rappl
- Tumorgenetics, Department of Internal Medicine I, University of Cologne, Cologne, Germany
| | - Annette Schmidt
- Institute for Circulation Research and Sports Medicine, German Sports University, Cologne, Germany
| | - Cornelia Mauch
- Tumorbiology, Department of Dermatology, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Andreas A. Hombach
- Tumorgenetics, Department of Internal Medicine I, University of Cologne, Cologne, Germany
| | - Hinrich Abken
- Tumorgenetics, Department of Internal Medicine I, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|