51
|
Stead SO, McInnes SJP, Kireta S, Rose PD, Jesudason S, Rojas-Canales D, Warther D, Cunin F, Durand JO, Drogemuller CJ, Carroll RP, Coates PT, Voelcker NH. Manipulating human dendritic cell phenotype and function with targeted porous silicon nanoparticles. Biomaterials 2017; 155:92-102. [PMID: 29175084 DOI: 10.1016/j.biomaterials.2017.11.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/13/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DC) are the most potent antigen-presenting cells and are fundamental for the establishment of transplant tolerance. The Dendritic Cell-Specific Intracellular adhesion molecule-3-Grabbing Non-integrin (DC-SIGN; CD209) receptor provides a target for dendritic cell therapy. Biodegradable and high-surface area porous silicon (pSi) nanoparticles displaying anti-DC-SIGN antibodies and loaded with the immunosuppressant rapamycin (Sirolimus) serve as a fit-for-purpose platform to target and modify DC. Here, we describe the fabrication of rapamycin-loaded DC-SIGN displaying pSi nanoparticles, the uptake efficiency into DC and the extent of nanoparticle-induced modulation of phenotype and function. DC-SIGN antibody displaying pSi nanoparticles favourably targeted and were phagocytosed by monocyte-derived and myeloid DC in whole human blood in a time- and dose-dependent manner. DC preconditioning with rapamycin-loaded nanoparticles, resulted in a maturation resistant phenotype and significantly suppressed allogeneic T-cell proliferation.
Collapse
Affiliation(s)
| | - Steven J P McInnes
- Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Svjetlana Kireta
- Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Peter D Rose
- University of Adelaide, Department of Medicine, Adelaide, Australia
| | - Shilpanjali Jesudason
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Darling Rojas-Canales
- Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - David Warther
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Frédérique Cunin
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Jean-Olivier Durand
- Institut Charles Gerhardt Montpellier, UMR 5253 CNRS -ENSCM-UM2-UM1, Ecole Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier, France
| | - Christopher J Drogemuller
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - Robert P Carroll
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia
| | - P Toby Coates
- University of Adelaide, Department of Medicine, Adelaide, Australia; Central Northern Adelaide Renal and Transplantation Service (CNARTS), The Royal Adelaide Hospital, Australia.
| | - Nicolas H Voelcker
- Future Industries Institute, University of South Australia, Adelaide, Australia; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, VIC, Australia; Monash Institute of Medical Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
52
|
Radu CA, Fischer S, Diehm Y, Hetzel O, Neubrech F, Dittmar L, Kleist C, Gebhard MM, Terness P, Kneser U, Kiefer J. The combination of mitomycin-induced blood cells with a temporary treatment of ciclosporin A prolongs allograft survival in vascularized composite allotransplantation. Langenbecks Arch Surg 2017; 403:83-92. [PMID: 28823033 DOI: 10.1007/s00423-017-1616-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 08/10/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Vascularized composite allotransplantation (VCA) is a rapidly expanding field of transplantation and provides a potential treatment for complex tissue defects. Peripheral blood mononuclear cells (PBMCs) shortly incubated with the antibiotic and chemotherapeutic agent mitomycin C (MMC) can suppress allogeneic T cell response and control allograft rejection in various organ transplantation models. MMC-incubated PBMCs (MICs) are currently being tested in a phase I clinical trial in kidney transplant patients. Previous studies with MICs in a complex VCA model showed the immunomodulatory potential of these cells. The aim of this study is to optimize and evaluate the use of MICs in combination with a standard immunosuppressive drug in VCA. METHODS Fully mismatched rats were used as hind limb donors [Lewis (RT11)] and recipients [Brown-Norway (RT1n)]. Sixty allogeneic hind limb transplantations were performed in six groups. Group A received donor-derived MICs combined with a temporary ciclosporin A (CsA) treatment. Group B received MICs in combination with a temporarily administered reduced dose of CsA. Group C served as a control and received a standard CsA dose temporarily without an additional administration of MICs, whereas Group D was solely medicated with a reduced CsA dose. Group E received no immunosuppressive therapy, neither CsA nor MICs. Group F was given a continuous standard immunosuppressive regimen consisting of CsA and prednisolone. The endpoint of the study was the onset of allograft rejection which was assessed clinically and histologically. RESULTS In group A and B, the rejection-free interval of the allograft was significantly prolonged to an average of 23.1 ± 1.7 and 24.7 ± 1.8 days compared to the corresponding control groups (p < 0.01). Rejection in groups C, D, and E was noted after 14.3 ± 1.1, 7.8 ± 0.7, and 6.9 ± 0.6 days. No rejection occurred in control group F during the follow-up period of 100 days. No adverse events have been noted. CONCLUSION The findings of this study show that the combination of MICs with a temporary CsA treatment significantly prolongs the rejection-free interval in a complex VCA model. The combination of MICs with CsA showed no adverse events such as graft-versus-host disease. MICs, which are generated by a simple and reliable in vitro technique, represent a potential therapeutic tool for prolonging allograft survival through immunomodulation.
Collapse
Affiliation(s)
- Christian Andreas Radu
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Sebastian Fischer
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Yannick Diehm
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Otto Hetzel
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Florian Neubrech
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Laura Dittmar
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany.,Department of Nuclear Medicine, University of Heidelberg, Heidelberg, Germany
| | - Martha Maria Gebhard
- Department of Experimental Surgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Terness
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Jurij Kiefer
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany.
| |
Collapse
|
53
|
Cho JJ, Stewart JM, Drashansky TT, Brusko MA, Zuniga AN, Lorentsen KJ, Keselowsky BG, Avram D. An antigen-specific semi-therapeutic treatment with local delivery of tolerogenic factors through a dual-sized microparticle system blocks experimental autoimmune encephalomyelitis. Biomaterials 2017; 143:79-92. [PMID: 28772190 DOI: 10.1016/j.biomaterials.2017.07.029] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/11/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Antigen-specific treatments are highly desirable for autoimmune diseases in contrast to treatments which induce systemic immunosuppression. A novel antigen-specific therapy has been developed which, when administered semi-therapeutically, is highly efficacious in the treatment of the mouse model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). The treatment uses dual-sized, polymeric microparticles (dMPs) loaded with specific antigen and tolerizing factors for intra- and extra-cellular delivery, designed to recruit and modulate dendritic cells toward a tolerogenic phenotype without systemic release. This approach demonstrated robust efficacy and provided complete protection against disease. Therapeutic efficacy required encapsulation of the factors in controlled-release microparticles and was antigen-specific. Disease blocking was associated with a reduction of infiltrating CD4+ T cells, inflammatory cytokine-producing pathogenic CD4+ T cells, and activated macrophages and microglia in the central nervous system. Furthermore, CD4+ T cells isolated from dMP-treated mice were anergic in response to disease-specific, antigen-loaded splenocytes. Additionally, the frequency of CD86hiMHCIIhi dendritic cells in draining lymph nodes of EAE mice treated with Ag-specific dMPs was reduced. Our findings highlight the efficacy of microparticle-based drug delivery platform to mediate antigen-specific tolerance, and suggest that such a multi-factor combinatorial approach can act to block autoimmunity.
Collapse
Affiliation(s)
- Jonathan J Cho
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Theodore T Drashansky
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Ashley N Zuniga
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Kyle J Lorentsen
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Dorina Avram
- Division of Pulmonary Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
54
|
Ezzelarab M, Raich-Regue D, Lu L, Zahorchak A, Perez-Gutierrez A, Humar A, Wijkstrom M, Minervini M, Wiseman R, Cooper D, Morelli A, Thomson A. Renal Allograft Survival in Nonhuman Primates Infused With Donor Antigen-Pulsed Autologous Regulatory Dendritic Cells. Am J Transplant 2017; 17:1476-1489. [PMID: 28009481 PMCID: PMC5444942 DOI: 10.1111/ajt.14182] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/30/2016] [Accepted: 12/15/2016] [Indexed: 01/25/2023]
Abstract
Systemic administration of autologous regulatory dendritic cells (DCreg; unpulsed or pulsed with donor antigen [Ag]), prolongs allograft survival and promotes transplant tolerance in rodents. Here, we demonstrate that nonhuman primate (NHP) monocyte-derived DCreg preloaded with cell membrane vesicles from allogeneic peripheral blood mononuclear cells induce T cell hyporesponsiveness to donor alloantigen (alloAg) in vitro. These donor alloAg-pulsed autologous DCreg (1.4-3.6 × 106 /kg) were administered intravenously, 1 day before MHC-mismatched renal transplantation to rhesus monkeys treated with costimulation blockade (cytotoxic T lymphocyte Ag 4 immunoglobulin [CTLA4] Ig) and tapered rapamycin. Prolongation of graft median survival time from 39.5 days (no DCreg infusion; n = 6 historical controls) and 29 days with control unpulsed DCreg (n = 2), to 56 days with donor Ag-pulsed DCreg (n = 5) was associated with evidence of modulated host CD4+ and CD8+ T cell responses to donor Ag and attenuation of systemic IL-17 production. Circulating anti-donor antibody (Ab) was not detected until CTLA4 Ig withdrawal. One monkey treated with donor Ag-pulsed DCreg rejected its graft in association with progressively elevated anti-donor Ab, 525 days posttransplant (160 days after withdrawal of immunosuppression). These findings indicate a modest but not statistically significant beneficial effect of donor Ag-pulsed autologous DCreg infusion on NHP graft survival when administered with a minimal immunosuppressive drug regimen.
Collapse
Affiliation(s)
- M.B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D. Raich-Regue
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. Perez-Gutierrez
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Wijkstrom
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Minervini
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - R.W. Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI
| | - D.K.C. Cooper
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.E. Morelli
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Angus W. Thomson, PhD, DSc, University of Pittsburgh School of Medicine, 200 Lothrop Street, W1540 BST, Pittsburgh, PA 15261, Phone: (412) 624-6392,
| |
Collapse
|
55
|
Combinatorial drug delivery approaches for immunomodulation. Adv Drug Deliv Rev 2017; 114:161-174. [PMID: 28532690 DOI: 10.1016/j.addr.2017.05.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022]
Abstract
Immunotherapy has been widely explored for applications to both augment and suppress intrinsic host immunity. Clinical achievements have seen a number of immunotherapeutic drugs displace established strategies like chemotherapy in treating immune-associated diseases. However, single drug approaches modulating an individual arm of the immune system are often incompletely effective. Imperfect mechanistic understanding and heterogeneity within disease pathology have seen monotherapies inadequately equipped to mediate complete disease remission. Recent success in applications of combinatorial immunotherapy has suggested that targeting multiple biological pathways simultaneously may be critical in treating complex immune pathologies. Drug delivery approaches through engineered biomaterials offer the potential to augment desired immune responses while mitigating toxic side-effects by localizing immunotherapy. This review discusses recent advances in immunotherapy and highlights newly explored combinatorial drug delivery approaches. Furthermore, prospective future directions for immunomodulatory drug delivery to exploit are provided.
Collapse
|
56
|
Cai S, Hou J, Fujino M, Zhang Q, Ichimaru N, Takahara S, Araki R, Lu L, Chen JM, Zhuang J, Zhu P, Li XK. iPSC-Derived Regulatory Dendritic Cells Inhibit Allograft Rejection by Generating Alloantigen-Specific Regulatory T Cells. Stem Cell Reports 2017; 8:1174-1189. [PMID: 28434942 PMCID: PMC5425686 DOI: 10.1016/j.stemcr.2017.03.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023] Open
Abstract
Regulatory dendritic cell (DCregs)-based immunotherapy is a potential therapeutic tool for transplant rejection. We generated DCregs from murine induced pluripotent stem cells (iPSCs), which could remain in a “stable immature stage” even under strong stimulation. Harnessing this characteristic, we hypothesized that iPS-DCregs worked as a negative vaccine to generate regulatory T cells (Tregs), and induced donor-specific allograft acceptance. We immunized naive CBA (H-2Kk) mice with B6 (H-2Kb) iPS-DCregs and found that Tregs (CD4+CD25+FOXP3+) significantly increased in CBA splenocytes. Moreover, immunized CBA recipients permanently accepted B6 cardiac grafts in a donor-specific pattern. We demonstrated mechanistically that donor-type iPS-DCregs triggered transforming growth factor β1 secretion, under which the donor-antigen peptides directed naive CD4+ T cells to differentiate into donor-specific FOXP3+ Tregs instead of into effector T cells in vivo. These findings highlight the potential of iPS-DCregs as a key cell therapy resource in clinical transplantation. iPS-DCregs keep in stable immature stage that makes them a powerful cellular vaccine Donor-type iPS-DCregs lead to permanent acceptance of allogeneic cardiac grafts iPS-DCregs reduce CTL and downregulate proinflammatory cytokine iPS-DCregs enhance Tregs transmigration capability in a TGF-β1-dependent manner
Collapse
Affiliation(s)
- Songjie Cai
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Jiangang Hou
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Qi Zhang
- Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Naotsugu Ichimaru
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shiro Takahara
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Ryoko Araki
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Lina Lu
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ji-Mei Chen
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Jian Zhuang
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ping Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan.
| |
Collapse
|
57
|
Abstract
Dendritic cells (DCs) are antigen-presenting cells that normally play a critical role in stimulating T-cell-dependent immune responses. However, tolerogenic DCs (CD11cMHC-IICD80CD86) induce immune tolerance by stimulating regulatory T cells (Tregs: CD4CD25Foxp3). Although tolerogenic DCs are used to treat autoimmune diseases and to prevent transplantation rejection, the mechanisms by which they regulate alloimmunity are poorly understood. Here, we review our previous studies aiming to elucidate the mechanisms involved in immune rejection of corneal allografts using a corneal transplant model. We found that donor-derived tolerogenic DCs significantly prolonged corneal allograft survival by suppressing indirect allosensitization. We also reported the precise distribution of intraepithelial corneal DCs, termed Langerhans cells (LCs: CD11cLangerinMHC-II) in the cornea, which we maintain play a critical role in regulating corneal immunity. By confocal microscopy, we constructed 3-dimensional images of corneal LCs, which demonstrated that their cell bodies are present in the basal cell layer of the corneal epithelium. Furthermore, LC dendrites extend toward the ocular surface, but do not connect to epithelial tight junctions, indicating that they cannot directly interact with ocular surface antigens. We confirm the potential of DC therapy for corneal graft rejection and report the function of intraepithelial DCs (LCs) in the normal cornea.
Collapse
|
58
|
Horton C, Shanmugarajah K, Fairchild PJ. Harnessing the properties of dendritic cells in the pursuit of immunological tolerance. Biomed J 2017; 40:80-93. [PMID: 28521905 PMCID: PMC6138597 DOI: 10.1016/j.bj.2017.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
The acquisition of self-perpetuating, immunological tolerance specific for graft alloantigens has long been described as the "holy grail" of clinical transplantation. By removing the need for life-long immunosuppression following engraftment, the adverse consequences of immunosuppressive regimens, including chronic infections and malignancy, may be avoided. Furthermore, autoimmune diseases and allergy are, by definition, driven by aberrant immunological responses to ordinarily innocuous antigens. The re-establishment of permanent tolerance towards instigating antigens may, therefore, provide a cure to these common diseases. Whilst various cell types exhibiting a tolerogenic phenotype have been proposed for such a task, tolerogenic dendritic cells (tol-DCs) are exquisitely adapted for antigen presentation and interact with many facets of the immune system: as such, they are attractive candidates for use in strategies for immune intervention. We review here our current understanding of tol-DC mediated induction and maintenance of immunological tolerance. Additionally, we discuss recent in vitro findings from animal models and clinical trials of tol-DC immunotherapy in the setting of transplantation, autoimmunity and allergy which highlight their promising therapeutic potential, and speculate how tol-DC therapy may be developed in the future.
Collapse
|
59
|
Moreau A, Alliot-Licht B, Cuturi MC, Blancho G. Tolerogenic dendritic cell therapy in organ transplantation. Transpl Int 2016; 30:754-764. [DOI: 10.1111/tri.12889] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 09/13/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Aurélie Moreau
- INSERM UMR1064; Center for Research in Transplantation and Immunology; Nantes France
- CHU de Nantes; Institut de Transplantation Urologie Nephrologie (ITUN); Nantes France
- Université de Nantes; Nantes France
| | - Brigitte Alliot-Licht
- INSERM UMR1064; Center for Research in Transplantation and Immunology; Nantes France
- CHU de Nantes; Institut de Transplantation Urologie Nephrologie (ITUN); Nantes France
- Université de Nantes; Nantes France
| | - Maria-Cristina Cuturi
- INSERM UMR1064; Center for Research in Transplantation and Immunology; Nantes France
- CHU de Nantes; Institut de Transplantation Urologie Nephrologie (ITUN); Nantes France
- Université de Nantes; Nantes France
| | - Gilles Blancho
- INSERM UMR1064; Center for Research in Transplantation and Immunology; Nantes France
- CHU de Nantes; Institut de Transplantation Urologie Nephrologie (ITUN); Nantes France
- Université de Nantes; Nantes France
| |
Collapse
|
60
|
Madelon N, Puga Yung GL, Seebach JD. Human anti-pig NK cell and CD8 + T-cell responses in the presence of regulatory dendritic cells. Xenotransplantation 2016; 23:479-489. [PMID: 27862343 DOI: 10.1111/xen.12279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/28/2016] [Accepted: 10/09/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Dendritic cells (DC) play a major role in natural killer (NK) cell and cytotoxic T lymphocyte (CTL) activation leading to cell-mediated xenogeneic responses. In contrast, the use of in vitro differentiated regulatory DC may represent an attractive approach to protect porcine endothelial cells (pEC) from human cell-mediated immune responses. In this study, we evaluated the potential of human regulatory DC to reduce xenogeneic NK cell and CTL responses to pEC. METHODS Human monocytes were differentiated into DC with GM-CSF and IL-4 in the absence or presence of rapamycin or IL-10. The effect of regulatory DC on xenogeneic NK cell and CTL responses was evaluated by analyzing phenotype, IFNγ production, degranulation, and cytotoxicity by flow cytometry and cytotoxicity assays. RESULTS Upon maturation with LPS, Rapa-DC and IL-10-DC displayed different phenotypes and cytokine production profiles. In contrast to untreated DC, both Rapa-DC and IL-10-DC induced significantly less IFNγ production and NK cell degranulation in response to pEC, but did not affect NK cell-mediated pEC lysis. Low production of IL-18 by Rapa-DC, and of IL-12 by IL-10-DC were linked to the deficient IFNγ production by NK cells as shown by partial reversion of IFNγ production upon cytokine reconstitution. In contrast to untreated DC efficiently generating xenoantigen-specific CTL, priming of CTL in the presence of IL-10-DC was impaired as shown by lower IFNγ production and cytotoxicity of CTL in response to pEC. CONCLUSION Both Rapa-DC and IL-10-DC controlled human anti-porcine NK cell responses, in particular IFNγ production, whereas IL-10-DC presented stronger regulatory properties of anti-porcine CTL responses. These in vitro findings indicate that regulatory DC could be a useful tool to promote xenograft tolerance in vivo.
Collapse
Affiliation(s)
- Natacha Madelon
- Laboratory of Transplantation Immunology, Division of Immunology and Allergology, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Gisella L Puga Yung
- Laboratory of Transplantation Immunology, Division of Immunology and Allergology, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Jörg D Seebach
- Laboratory of Transplantation Immunology, Division of Immunology and Allergology, Department of Medical Specialties, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
61
|
Combining Exosomes Derived from Immature DCs with Donor Antigen-Specific Treg Cells Induces Tolerance in a Rat Liver Allograft Model. Sci Rep 2016; 6:32971. [PMID: 27640806 PMCID: PMC5027549 DOI: 10.1038/srep32971] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022] Open
Abstract
Allograft tolerance is the ultimate goal in the field of transplantation immunology. Immature dendritic cells (imDCs) play an important role in establishing tolerance but have limitations, including potential for maturation, short lifespan in vivo and short storage times in vitro. However, exosomes (generally 30–100 nm) from imDCs (imDex) retain many source cell properties and may overcome these limitations. In previous reports, imDex prolonged the survival time of heart or intestine allografts. However, tolerance or long-term survival was not achieved unless immune suppressants were used. Regulatory T cells (Tregs) can protect allografts from immune rejection, and our previous study showed that the effects of imDex were significantly associated with Tregs. Therefore, we incorporated Tregs into the treatment protocol to further reduce or avoid suppressant use. We defined the optimal exosome dose as approximately 20 μg (per treatment before, during and after transplantation) in rat liver transplantation and the antigen-specific role of Tregs in protecting liver allografts. In the co-treatment group, recipients achieved long-term survival, and tolerance was induced. Moreover, imDex amplified Tregs, which required recipient DCs and were enhanced by IL-2. Fortunately, the expanded Tregs retained their regulatory ability and donor-specificity. Thus, imDex and donor-specific Tregs can collaboratively induce graft tolerance.
Collapse
|
62
|
Baroja-Mazo A, Revilla-Nuin B, Parrilla P, Martínez-Alarcón L, Ramírez P, Pons JA. Tolerance in liver transplantation: Biomarkers and clinical relevance. World J Gastroenterol 2016; 22:7676-91. [PMID: 27678350 PMCID: PMC5016367 DOI: 10.3748/wjg.v22.i34.7676] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/04/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Transplantation is the optimal treatment for end-stage organ failure, and modern immunosuppression has allowed important progress in short-term outcomes. However, immunosuppression poorly influences chronic rejection and elicits chronic toxicity in current clinical practice. Thus, a major goal in transplantation is to understand and induce tolerance. It is well established that human regulatory T cells expressing the transcription factor FoxP3 play important roles in the maintenance of immunological self-tolerance and immune homeostasis. The major regulatory T cell subsets and mechanisms of expansion that are critical for induction and long-term maintenance of graft tolerance and survival are being actively investigated. Likewise, other immune cells, such as dendritic cells, monocyte/macrophages or natural killer cells, have been described as part of the process known as "operational tolerance". However, translation of these results towards clinical practice needs solid tools to identify accurately and reliably patients who are going to be tolerant. In this way, a plethora of genetic and cellular biomarkers is raising and being validated worldwide in large multi-center clinical trials. Few of the studies performed so far have provided a detailed analysis of the impact of immunosuppression withdrawal on pre-existing complications derived from the long-term administration of immunosuppressive drugs and the side effects associated with them. The future of liver transplantation is aimed to develop new therapies which increase the actual low tolerant vs non-tolerant recipients ratio.
Collapse
|
63
|
Sukhbaatar N, Hengstschläger M, Weichhart T. mTOR-Mediated Regulation of Dendritic Cell Differentiation and Function. Trends Immunol 2016; 37:778-789. [PMID: 27614799 DOI: 10.1016/j.it.2016.08.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/10/2016] [Accepted: 08/15/2016] [Indexed: 01/09/2023]
Abstract
Dendritic cells (DCs) are essential antigen-presenting cells that sample the extra- and intracellular milieu to process antigens for the instruction of T cell responses. The mammalian target of rapamycin (mTOR) network senses environmental cues and is important for numerous cellular processes. This review discusses how DCs use mTOR complexes (mTORC1 and 2) to adapt their cellular metabolism, transcriptional responses, and translation machinery to control DC development, antigen processing, cytokine production, and T cell stimulation. We present a spatiotemporal model suggesting that the mTOR network integrates pattern recognition and growth factor receptor activation with nutritional information from the cell and surrounding tissue to support T cell stimulation and tolerance. mTOR develops into a central player that regulates DC differentiation and immune functions.
Collapse
Affiliation(s)
- Nyamdelger Sukhbaatar
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Markus Hengstschläger
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Thomas Weichhart
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria.
| |
Collapse
|
64
|
Marín E, Cuturi MC, Moreau A. Potential of Tolerogenic Dendritic Cells in Transplantation. CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0109-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
65
|
Microfilariae of Brugia malayi Inhibit the mTOR Pathway and Induce Autophagy in Human Dendritic Cells. Infect Immun 2016; 84:2463-72. [PMID: 27297394 DOI: 10.1128/iai.00174-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/08/2016] [Indexed: 01/04/2023] Open
Abstract
Immune modulation is a hallmark of patent filarial infection, including suppression of antigen-presenting cell function and downmodulation of filarial antigen-specific T cell responses. The mammalian target of rapamycin (mTOR) signaling pathway has been implicated in immune regulation, not only by suppressing T cell responses but also by regulating autophagy (through mTOR sensing amino acid availability). Global proteomic analysis (liquid chromatography-tandem mass spectrometry) of microfilaria (mf)-exposed monocyte-derived dendritic cells (DC) indicated that multiple components of the mTOR signaling pathway, including mTOR, eIF4A, and eIF4E, are downregulated by mf, suggesting that mf target this pathway for immune modulation in DC. Utilizing Western blot analysis, we demonstrate that similar to rapamycin (a known mTOR inhibitor), mf downregulate the phosphorylation of mTOR and its regulatory proteins, p70S6K1 and 4E-BP1, a process essential for DC protein synthesis. As active mTOR signaling regulates autophagy, we examined whether mf exposure alters autophagy-associated processes. mf-induced autophagy was reflected in marked upregulation of phosphorylated Beclin 1, known to play an important role in both autophagosome formation and autolysosome fusion, in induction of LC3II, a marker of autophagosome formation, and in induced degradation of p62, a ubiquitin-binding protein that aggregates protein in autophagosomes and is degraded upon autophagy that was reduced significantly by mf exposure and by rapamycin. Together, these results suggest that Brugia malayi mf employ mechanisms of metabolic modulation in DC to influence the regulation of the host immune response by downregulating mTOR signaling, resulting in increased autophagy. Whether this is a result of the parasite-secreted rapamycin homolog is currently under study.
Collapse
|
66
|
Effects of Adoptive Transfer of Tolerogenic Dendritic Cells on Allograft Survival in Organ Transplantation Models: An Overview of Systematic Reviews. J Immunol Res 2016; 2016:5730674. [PMID: 27547767 PMCID: PMC4980535 DOI: 10.1155/2016/5730674] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/06/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023] Open
Abstract
Objective. To dissect the efficacy of Tol-DC therapy with or without IS in multiple animal models of transplantation. Methods and Results. PubMed, Medline, Embase, and the Cochrane Library were searched for reviews published up to April 2015. Six systematic reviews and a total of 61 articles were finally included. Data were grouped by organ transplantation models and applied to meta-analysis. Our meta-analysis shows that Tol-DC therapy successfully prolonged allograft survival to varying extents in all except the islet transplantation models and with IS drugs further prolonged the survival of heart, skin, and islet allografts in mice, but not of heart allografts in rats. Compared with IS drugs alone, Tol-DC therapy with IS extended islet allograft survival in rats but failed to influence the survival of skin, small intestine, and heart allografts in rats or of heart and skin allografts in mice. Conclusion. Tol-DC therapy significantly prolonged multiple allograft survival and further prolonged survival with IS. However, standardized protocols for modification of Tol-DC should be established before its application in clinic.
Collapse
|
67
|
Stallone G, Infante B, Di Lorenzo A, Rascio F, Zaza G, Grandaliano G. mTOR inhibitors effects on regulatory T cells and on dendritic cells. J Transl Med 2016; 14:152. [PMID: 27245075 PMCID: PMC4886438 DOI: 10.1186/s12967-016-0916-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/18/2016] [Indexed: 01/03/2023] Open
Abstract
The mammalian target of rapamycin (mTOR), a cytoplasmic serine/threonine kinase, represents a key biologic "switch" modulating cell metabolisms in response to environmental signals and is now recognized as a central regulator of the immune system. There is an increasing body of evidence supporting the hypothesis that mTOR inhibitors exhibit several biological properties in addition to immunosuppression, including anti-neoplastic effects, cardio-protective activities, and an array of immunomodulatory actions facilitating the development of an operational graft tolerance. The biological mechanisms explaining how mTOR inhibition can enable a tolerogenic state are still largely unclear. The induction of transplant tolerance might at the same time decrease rejection rate and minimize immunosuppression-related side effects, leading to an improvement in long-term graft outcome. In this scenario, T cell immunoregulation has been defined as the hallmark of peripheral tolerance. Two main immunologic cell populations have been reported to play a central role in this setting: regulatory T cells (Tregs) and dendritic cells (DCs). In this review we focus on mTOR inhibitors effects on Treg and DCs differentiation, activation, and function in the transplantation setting.
Collapse
Affiliation(s)
- Giovanni Stallone
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Barbara Infante
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Adelaide Di Lorenzo
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Federica Rascio
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy.
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Tranplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Luigi Pinto, 1, 71100, Foggia, Italy.
| |
Collapse
|
68
|
Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases. Pharmacol Res 2016; 107:27-41. [DOI: 10.1016/j.phrs.2016.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
|
69
|
Metabolism Is Central to Tolerogenic Dendritic Cell Function. Mediators Inflamm 2016; 2016:2636701. [PMID: 26980944 PMCID: PMC4766347 DOI: 10.1155/2016/2636701] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 12/31/2015] [Indexed: 12/13/2022] Open
Abstract
Immunological tolerance is a fundamental tenant of immune homeostasis and overall health. Self-tolerance is a critical component of the immune system that allows for the recognition of self, resulting in hyporeactivity instead of immunogenicity. Dendritic cells are central to the establishment of dominant immune tolerance through the secretion of immunosuppressive cytokines and regulatory polarization of T cells. Cellular metabolism holds the key to determining DC immunogenic or tolerogenic cell fate. Recent studies have demonstrated that dendritic cell maturation leads to a shift toward a glycolytic metabolic state and preferred use of glucose as a carbon source. In contrast, tolerogenic dendritic cells favor oxidative phosphorylation and fatty acid oxidation. This dichotomous metabolic reprogramming of dendritic cells drives differential cellular function and plays a role in pathologies, such as autoimmune disease. Pharmacological alterations in metabolism have promising therapeutic potential.
Collapse
|
70
|
Thomson AW, Zahorchak AF, Ezzelarab MB, Butterfield LH, Lakkis FG, Metes DM. Prospective Clinical Testing of Regulatory Dendritic Cells in Organ Transplantation. Front Immunol 2016; 7:15. [PMID: 26858719 PMCID: PMC4729892 DOI: 10.3389/fimmu.2016.00015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/12/2016] [Indexed: 01/03/2023] Open
Abstract
Dendritic cells (DC) are rare, professional antigen-presenting cells with ability to induce or regulate alloimmune responses. Regulatory DC (DCreg) with potential to down-modulate acute and chronic inflammatory conditions that occur in organ transplantation can be generated in vitro under a variety of conditions. Here, we provide a rationale for evaluation of DCreg therapy in clinical organ transplantation with the goal of promoting sustained, donor-specific hyporesponsiveness, while lowering the incidence and severity of rejection and reducing patients’ dependence on anti-rejection drugs. Generation of donor- or recipient-derived DCreg that suppress T cell responses and prolong transplant survival in rodents or non-human primates has been well-described. Recently, good manufacturing practice (GMP)-grade DCreg have been produced at our Institution for prospective use in human organ transplantation. We briefly review experience of regulatory immune therapy in organ transplantation and describe our experience generating and characterizing human monocyte-derived DCreg. We propose a phase I/II safety study in which the influence of donor-derived DCreg combined with conventional immunosuppression on subclinical and clinical rejection and host alloimmune responses will be examined in detail.
Collapse
Affiliation(s)
- Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alan F Zahorchak
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Mohamed B Ezzelarab
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine , Pittsburgh, PA , USA
| | - Lisa H Butterfield
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fadi G Lakkis
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diana M Metes
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
71
|
Zhang AH, Rossi RJ, Yoon J, Wang H, Scott DW. Tolerogenic nanoparticles to induce immunologic tolerance: Prevention and reversal of FVIII inhibitor formation. Cell Immunol 2015; 301:74-81. [PMID: 26687613 DOI: 10.1016/j.cellimm.2015.11.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/27/2015] [Accepted: 11/20/2015] [Indexed: 01/06/2023]
Abstract
The immune response of hemophilia A patients to administered FVIII is a major complication that obviates this very therapy. We have recently described the use of synthetic, biodegradable nanoparticles carrying rapamycin and FVIII peptide antigens, to induce antigen-specific tolerance. Herein we test the tolerogenicity of nanoparticles that contains full length FVIII protein in hemophilia A mice, focusing on anti-FVIII humoral immune response. As expected, recipients of tolerogenic nanoparticles remained unresponsive to FVIII despite multiple challenges for up to 6 months. Furthermore, therapeutic treatments in FVIII-immunized mice with pre-existing anti-FVIII antibodies resulted in diminished antibody titers, albeit efficacy required longer therapy with the tolerogenic nanoparticles. Interestingly, durable FVIII-specific tolerance was also achieved in animals co-administered with FVIII admixed with nanoparticles encapsulating rapamycin alone. These results suggest that nanoparticles carrying rapamycin and FVIII can be employed to induce specific tolerance to prevent and even reverse inhibitor formation.
Collapse
Affiliation(s)
- Ai-Hong Zhang
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert J Rossi
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jeongheon Yoon
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Hong Wang
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - David W Scott
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
72
|
Abstract
From the early days of transplantation onwards, increased cancer development in transplant recipients, who require immunosuppression to avoid graft rejection, has been recognized. Registry data indicate that approximately 10-30% of deaths are attributed to post-transplant malignancy, with an upward trend in this incidence as more patients have been exposed to chronic lifelong immunosuppression. In this Review, the overall incidence and most frequent types of cancer encountered are summarized, along with information about which transplant recipients are at the greatest risk of malignancy. Reasons for why differences exist in susceptibility to cancer in this patient population are examined, and approaches that might improve our understanding of the options available for reducing the incidence of this adverse effect of immunosuppression are described. Whether anti-rejection drugs have been successful in diminishing overall immunosuppressive burden, and consequently show any promise for decreasing post-transplant malignancies is also discussed. The topic shifts to one class of conventional anti-rejection drugs, the mammalian target of rapamycin (mTOR) inhibitors, which paradoxically have both immunosuppressive and anti-neoplastic properties. The complex activities of mTOR are reviewed in order to provide context for how these seemingly opposing effects are possible, and the latest clinical data on use of mTOR inhibitors in the clinic are discussed. The current and future perspectives on how best to normalize these unacceptably high rates of post-transplantation malignancies are highlighted.
Collapse
|
73
|
Osorio F, Fuentes C, López MN, Salazar-Onfray F, González FE. Role of Dendritic Cells in the Induction of Lymphocyte Tolerance. Front Immunol 2015; 6:535. [PMID: 26539197 PMCID: PMC4611163 DOI: 10.3389/fimmu.2015.00535] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/02/2015] [Indexed: 01/07/2023] Open
Abstract
The ability of dendritic cells (DCs) to trigger tolerance or immunity is dictated by the context in which an antigen is encountered. A large body of evidence indicates that antigen presentation by steady-state DCs induces peripheral tolerance through mechanisms such as the secretion of soluble factors, the clonal deletion of autoreactive T cells, and feedback control of regulatory T cells. Moreover, recent understandings on the function of DC lineages and the advent of murine models of DC depletion have highlighted the contribution of DCs to lymphocyte tolerance. Importantly, these findings are now being applied to human research in the contexts of autoimmune diseases, allergies, and transplant rejection. Indeed, DC-based immunotherapy research has made important progress in the area of human health, particularly in regards to cancer. A better understanding of several DC-related aspects including the features of DC lineages, milieu composition, specific expression of surface molecules, the control of signaling responses, and the identification of competent stimuli able to trigger and sustain a tolerogenic outcome will contribute to the success of DC-based immunotherapy in the area of lymphocyte tolerance. This review will discuss the latest advances in the biology of DC subtypes related to the induction of regulatory T cells, in addition to presenting current ex vivo protocols for tolerogenic DC production. Particular attention will be given to the molecules and signals relevant for achieving an adequate tolerogenic response for the treatment of human pathologies.
Collapse
Affiliation(s)
- Fabiola Osorio
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Camila Fuentes
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Mercedes N López
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile ; Cell Therapy Laboratory, Blood Bank Service, University of Chile Clinical Hospital , Santiago , Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Fermín E González
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Laboratory of Experimental Immunology and Cancer, Faculty of Dentistry, University of Chile , Santiago , Chile
| |
Collapse
|
74
|
Roehrich ME, Wyss JC, Kumar R, Pascual M, Golshayan D, Vassalli G. Additive effects of rapamycin and aspirin on dendritic cell allostimulatory capacity. Immunopharmacol Immunotoxicol 2015; 37:434-41. [DOI: 10.3109/08923973.2015.1081606] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
75
|
|
76
|
Abstract
The innate immune system is central for the maintenance of tissue homeostasis and quickly responds to local or systemic perturbations by pathogenic or sterile insults. This rapid response must be metabolically supported to allow cell migration and proliferation and to enable efficient production of cytokines and lipid mediators. This Review focuses on the role of mammalian target of rapamycin (mTOR) in controlling and shaping the effector responses of innate immune cells. mTOR reconfigures cellular metabolism and regulates translation, cytokine responses, antigen presentation, macrophage polarization and cell migration. The mTOR network emerges as an integrative rheostat that couples cellular activation to the environmental and intracellular nutritional status to dictate and optimize the inflammatory response. A detailed understanding of how mTOR metabolically coordinates effector responses by myeloid cells will provide important insights into immunity in health and disease.
Collapse
Affiliation(s)
- Thomas Weichhart
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Markus Hengstschläger
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Monika Linke
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| |
Collapse
|
77
|
Nakamura T, Nakao T, Yoshimura N, Ashihara E. Rapamycin Prolongs Cardiac Allograft Survival in a Mouse Model by Inducing Myeloid-Derived Suppressor Cells. Am J Transplant 2015; 15:2364-77. [PMID: 25943210 DOI: 10.1111/ajt.13276] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/25/2023]
Abstract
Mammalian target of rapamycin (mTOR) inhibitors are the main immunosuppressive drugs for organ transplant recipients. Nevertheless, the mechanisms by which mTOR inhibitors induce immunosuppression is not fully understood. Myeloid-derived suppressor cells (MDSCs) maintain host immunity; however, the relationship between mTOR inhibitors and MDSCs is unclear. Here, the results from a murine cardiac transplantation model revealed that rapamycin treatment (3 mg/kg, intraperitoneally on postoperative days 0, 2, 4, and 6) led to the recruitment of MDSCs and increased their expression of inducible nitric oxide synthase (iNOS). Immunohistochemical analysis revealed that rapamycin induced the migration of iNOS-expressing MDSCs into the subintimal space within the allograft vessels, resulting in a significant prolongation of graft survival compared with that in the untreated group (67 days vs. 7 days, respectively). These effects were counterbalanced by the administration of an anti-Gr-1, which reduced allograft survival to 21 days. Moreover, adoptive transcoronary arterial transfer of MDSCs from rapamycin-treated recipients prolonged allograft survival; this increase was reversed by the anti-Gr-1 antibody. Finally, co-administration of rapamycin and a mitogen-activated protein kinase kinase (MEK) inhibitor trametinib reversed rapamycin-mediated MDSC recruitment. Thus, the mTOR and Raf/MEK/extracellular signal regulated kinase (ERK) signaling pathways appear to play an important role in MDSC expansion.
Collapse
Affiliation(s)
- T Nakamura
- Department of Transplantation and Regenerative Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto-Prefecture, Japan.,Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - T Nakao
- Department of Transplantation and Regenerative Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto-Prefecture, Japan.,Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - N Yoshimura
- Department of Transplantation and Regenerative Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto-Prefecture, Japan
| | - E Ashihara
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| |
Collapse
|
78
|
Podestà MA, Cucchiari D, Ponticelli C. The diverging roles of dendritic cells in kidney allotransplantation. Transplant Rev (Orlando) 2015; 29:114-20. [DOI: 10.1016/j.trre.2015.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/09/2015] [Accepted: 04/08/2015] [Indexed: 12/23/2022]
|
79
|
Pidala J, Kim J, Alsina M, Ayala E, Betts BC, Fernandez HF, Field T, Jim H, Kharfan-Dabaja MA, Locke FL, Mishra A, Nishihori T, Ochoa-Bayona L, Perez L, Riches M, Anasetti C. Prolonged sirolimus administration after allogeneic hematopoietic cell transplantation is associated with decreased risk for moderate-severe chronic graft-versus-host disease. Haematologica 2015; 100:970-7. [PMID: 25840599 DOI: 10.3324/haematol.2015.123588] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/20/2015] [Indexed: 11/09/2022] Open
Abstract
Effective pharmacological strategies employed in allogeneic hematopoietic cell transplantation should prevent serious chronic graft-versus-host disease and facilitate donor-recipient immune tolerance. Based on demonstrated pro-tolerogenic activity, sirolimus (rapamycin) is an agent with promise to achieve these goals. In a long-term follow-up analysis of a randomized phase II trial comparing sirolimus/tacrolimus versus methotrexate/tacrolimus for graft-versus-host disease prevention in matched sibling or unrelated donor transplant, we examined the impact of prolonged sirolimus administration (≥ 1 year post-transplant). Median follow-up time for surviving patients at time of this analysis was 41 months (range 27-60) for sirolimus/tacrolimus and 49 months (range 29-63) for methotrexate/tacrolimus. Sirolimus/tacrolimus patients had significantly lower National Institutes of Health Consensus moderate-severe chronic graft-versus-host disease (34% vs. 65%; P=0.004) and late acute graft-versus-host disease (20% vs. 43%; P=0.04). While sirolimus/tacrolimus patients had lower prednisone exposure and earlier discontinuation of tacrolimus (median time to tacrolimus discontinuation 368 days vs. 821 days; P=0.002), there was no significant difference in complete immune suppression discontinuation (60-month estimate: 43% vs. 31%; P=0.78). Prolonged sirolimus administration represents a viable approach to mitigate risk for moderate-severe chronic and late acute graft-versus-host disease. Further study of determinants of successful immune suppression discontinuation is needed.
Collapse
Affiliation(s)
- Joseph Pidala
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Jongphil Kim
- Oncologic Sciences, College of Medicine at University of South Florida, USA Biostatistics, Moffitt Cancer Center; USA
| | - Melissa Alsina
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Ernesto Ayala
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Brian C Betts
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Hugo F Fernandez
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Teresa Field
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Heather Jim
- Oncologic Sciences, College of Medicine at University of South Florida, USA Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Fl, USA
| | - Mohamed A Kharfan-Dabaja
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Frederick L Locke
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Asmita Mishra
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Taiga Nishihori
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Leonel Ochoa-Bayona
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Lia Perez
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Marcie Riches
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| | - Claudio Anasetti
- Blood and Marrow Transplantation, Moffitt Cancer Center, USA Oncologic Sciences, College of Medicine at University of South Florida, USA
| |
Collapse
|
80
|
Jeong SH, Ji YH, Yoon ES. Immunosuppressive activity of adipose tissue-derived mesenchymal stem cells in a rat model of hind limb allotransplantation. Transplant Proc 2015; 46:1606-14. [PMID: 24935335 DOI: 10.1016/j.transproceed.2013.12.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/15/2013] [Accepted: 12/16/2013] [Indexed: 02/07/2023]
Abstract
Many reports have shown that bone marrow-derived mesenchymal stem cells exhibit immunosuppressive effects in allogeneic transplantation. However, few reports have evaluated the immunosuppressive properties of adipose tissue-derived mesenchymal stem cells (ASCs) in vitro and in vivo. In this study, we investigated the immunosuppressive characteristics of ASCs, and investigated whether ASCs originating from donor rats prolong allotransplant survival in a rat hind limb allotransplantation model. T-cell proliferation stimulated by allogeneic stimuli or mitogen with or without ASCs originating from the donor was assessed in vitro. The effects of cellular contact or soluble factors on the inhibition of T-cell proliferation were also evaluated. In the in vivo study, cultured ASCs (1 × 10(5)) that originated from the donor were injected into recipient animals intravenously immediately after operation, followed by 1 dose per day for 3 consecutive days post-transplantation. When immune rejection occurred, the survival time of allotransplants was determined and rejected tissue was histologically and immunochemically assessed for determining regulatory T-cell infiltration. ASCs inhibited the T-cell proliferation stimulated by alloantigen or mitogen in a dose-dependent manner, and recipient T cells proliferated less in animals treated with ASCs than in controls. Although ASCs were separated from T cells, ASCs persisted to elicit a suppressive effect. ASC culture supernatants did not inhibit T-cell proliferation; however, supernatants obtained from the mixed lymphocyte reaction in the presence of ASCs suppressed T-cell proliferation. ASCs prolonged allotransplant survival time, reduced inflammatory cell infiltration, and induced regulatory T cells. In conclusion, ASCs can exhibit in vitro immunosuppressive properties and prolong allotransplant survival time in a rat hind limb composite tissue allotransplantation model, possibly through the induction of regulatory T cells.
Collapse
Affiliation(s)
- S-H Jeong
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Y-H Ji
- Department of Plastic Surgery, Korea University Guro Hospital, Seoul, Korea
| | - E-S Yoon
- Department of Plastic Surgery, Korea University Anam Hospital, Seoul, Korea.
| |
Collapse
|
81
|
Serra P, Santamaria P. Nanoparticle-based autoimmune disease therapy. Clin Immunol 2015; 160:3-13. [PMID: 25704658 DOI: 10.1016/j.clim.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 01/10/2023]
Abstract
The goal of immunotherapy against autoimmunity is to block pathogenic inflammation without impairing immunity against infections and tumours. Regulatory T-cells (Tregs) play a central role in maintaining immune homeostasis, and autoimmune inflammation is frequently associated with decreased numbers and/or function of these T-cells. Therapies harnessing Tregs to treat autoimmune inflammation remain under-developed with caveats ranging from the lack of antigenic and disease specificity to the potential phenotypic and functional instability of in vitro-expanded Treg cells in vivo. Here, we review nanotechnology-based approaches designed to promote immune tolerance through various mechanisms, ranging from systemic or local suppression of antigen-presenting cells and deletion of antigen-specific T-cells, to the systemic expansion of antigen- and disease-specific Treg cells in vivo.
Collapse
Affiliation(s)
- Pau Serra
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain.
| | - Pere Santamaria
- Institut D'Investigacions Biomediques August Pi i Sunyer, Barcelona 08036, Spain; Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cummings School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
82
|
Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci U S A 2014; 112:E156-65. [PMID: 25548186 DOI: 10.1073/pnas.1408686111] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Current treatments to control pathological or unwanted immune responses often use broadly immunosuppressive drugs. New approaches to induce antigen-specific immunological tolerance that control both cellular and humoral immune responses are desirable. Here we describe the use of synthetic, biodegradable nanoparticles carrying either protein or peptide antigens and a tolerogenic immunomodulator, rapamycin, to induce durable and antigen-specific immune tolerance, even in the presence of potent Toll-like receptor agonists. Treatment with tolerogenic nanoparticles results in the inhibition of CD4+ and CD8+ T-cell activation, an increase in regulatory cells, durable B-cell tolerance resistant to multiple immunogenic challenges, and the inhibition of antigen-specific hypersensitivity reactions, relapsing experimental autoimmune encephalomyelitis, and antibody responses against coagulation factor VIII in hemophilia A mice, even in animals previously sensitized to antigen. Only encapsulated rapamycin, not the free form, could induce immunological tolerance. Tolerogenic nanoparticle therapy represents a potential novel approach for the treatment of allergies, autoimmune diseases, and prevention of antidrug antibodies against biologic therapies.
Collapse
|
83
|
Immunosuppressive therapy in allograft transplantation: from novel insights and strategies to tolerance and challenges. Cent Eur J Immunol 2014; 39:400-9. [PMID: 26155155 PMCID: PMC4440012 DOI: 10.5114/ceji.2014.45955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 07/03/2014] [Indexed: 01/07/2023] Open
Abstract
Immunosuppression therapy is the key to successful post-transplantation outcomes. The need for ideal immunosuppression became durable maintenance of long-term graft survival. In spite of current immunosuppressive therapy regimens advances, surgical procedures, and preservation methods, organ transplantation is associated with a long-term poor survival and significant mortality. This has led to an increased interest to optimize outcomes while minimizing associated toxicity by using alternative methods for maintenance immunosuppression, organ rejection treatment, and monitoring of immunosuppression. T regulatory (Treg) cells, which have immunosuppressive functions and cytokine profiles, have been studied during the last decades. Treg cells are able to inhibit the development of allergen-specific cell responses and consequently play a key role in a healthy immune response to allergens. Mature dendritic cells (DCs) play a crucial role in the differentiation of Tregs, which are known to regulate allergic inflammatory responses. Advance in long-standing allograft outcomes may depend on new drugs with novel mechanisms of action with minimal toxicity. Newer treatment techniques have been developed, including using novel stem cell-based therapies such as mesenchymal stem cells, phagosomes and exosomes. Immunoisolation techniques and salvage therapies, including photopheresis and total lymphoid irradiation have emerged as alternative therapeutic choices. The present review evaluates the recent clinical advances in immunosuppressive therapies for organ transplantation.
Collapse
|
84
|
Jeon HJ, Yang J. Cell Therapy in Kidney Transplantation. KOREAN JOURNAL OF TRANSPLANTATION 2014. [DOI: 10.4285/jkstn.2014.28.3.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hee Jung Jeon
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
85
|
Baas MC, Kuhn C, Valette F, Mangez C, Duarte MS, Hill M, Besançon A, Chatenoud L, Cuturi MC, You S. Combining Autologous Dendritic Cell Therapy with CD3 Antibodies Promotes Regulatory T Cells and Permanent Islet Allograft Acceptance. THE JOURNAL OF IMMUNOLOGY 2014; 193:4696-703. [DOI: 10.4049/jimmunol.1401423] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
86
|
Monguió-Tortajada M, Lauzurica-Valdemoros R, Borràs FE. Tolerance in organ transplantation: from conventional immunosuppression to extracellular vesicles. Front Immunol 2014; 5:416. [PMID: 25278936 PMCID: PMC4166341 DOI: 10.3389/fimmu.2014.00416] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/18/2014] [Indexed: 12/26/2022] Open
Abstract
Organ transplantation is often the unique solution for organ failure. However, rejection is still an unsolved problem. Although acute rejection is well controlled, the chronic use of immunosuppressive drugs for allograft acceptance causes numerous side effects in the recipient and do not prevent chronic allograft dysfunction. Different alternative therapies have been proposed to replace the classical treatment for allograft rejection. The alternative therapies are mainly based in pre-infusions of different types of regulatory cells, including DCs, MSCs, and Tregs. Nevertheless, these approaches lack full efficiency and have many problems related to availability and applicability. In this context, the use of extracellular vesicles, and in particular exosomes, may represent a cell-free alternative approach in inducing transplant tolerance and survival. Preliminary approaches in vitro and in vivo have demonstrated the efficient alloantigen presentation and immunomodulation abilities of exosomes, leading to alloantigen-specific tolerance and allograft acceptance in rodent models. Donor exosomes have been used alone, processed by recipient antigen-presenting cells, or administered together with suboptimal doses of immunosuppressive drugs, achieving specific allograft tolerance and infinite transplant survival. In this review, we gathered the latest exosome-based strategies for graft acceptance and discuss the tolerance mechanisms involved in organ tolerance mediated by the administration of exosomes. We will also deal with the feasibility and difficulties that arise from the application of this strategy into the clinic.
Collapse
Affiliation(s)
- Marta Monguió-Tortajada
- Innovation in Vesicles and Cells for Application Therapy Group (IVECAT), Institut d’Investigació Germans Trias i Pujol, Badalona, Spain
| | | | - Francesc E. Borràs
- Innovation in Vesicles and Cells for Application Therapy Group (IVECAT), Institut d’Investigació Germans Trias i Pujol, Badalona, Spain
- Nephrology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
87
|
Mackern-Oberti JP, Vega F, Llanos C, Bueno SM, Kalergis AM. Targeting dendritic cell function during systemic autoimmunity to restore tolerance. Int J Mol Sci 2014; 15:16381-417. [PMID: 25229821 PMCID: PMC4200801 DOI: 10.3390/ijms150916381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/29/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022] Open
Abstract
Systemic autoimmune diseases can damage nearly every tissue or cell type of the body. Although a great deal of progress has been made in understanding the pathogenesis of autoimmune diseases, current therapies have not been improved, remain unspecific and are associated with significant side effects. Because dendritic cells (DCs) play a major role in promoting immune tolerance against self-antigens (self-Ags), current efforts are focusing at generating new therapies based on the transfer of tolerogenic DCs (tolDCs) during autoimmunity. However, the feasibility of this approach during systemic autoimmunity has yet to be evaluated. TolDCs may ameliorate autoimmunity mainly by restoring T cell tolerance and, thus, indirectly modulating autoantibody development. In vitro induction of tolDCs loaded with immunodominant self-Ags and subsequent cell transfer to patients would be a specific new therapy that will avoid systemic immunosuppression. Herein, we review recent approaches evaluating the potential of tolDCs for the treatment of systemic autoimmune disorders.
Collapse
Affiliation(s)
- Juan P Mackern-Oberti
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Fabián Vega
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Carolina Llanos
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Marcoleta 350, Santiago 8330033, Chile.
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Portugal 49, Santiago 8330025, Chile.
| |
Collapse
|
88
|
Zhu L, Yang T, Li L, Sun L, Hou Y, Hu X, Zhang L, Tian H, Zhao Q, Peng J, Zhang H, Wang R, Yang Z, Zhang L, Zhao Y. TSC1 controls macrophage polarization to prevent inflammatory disease. Nat Commun 2014; 5:4696. [PMID: 25175012 DOI: 10.1038/ncomms5696] [Citation(s) in RCA: 234] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/15/2014] [Indexed: 12/29/2022] Open
Abstract
Macrophages acquire distinct phenotypes during tissue stress and inflammatory responses, but the mechanisms that regulate the macrophage polarization are poorly defined. Here we show that tuberous sclerosis complex 1 (TSC1) is a critical regulator of M1 and M2 phenotypes of macrophages. Mice with myeloid-specific deletion of TSC1 exhibit enhanced M1 response and spontaneously develop M1-related inflammatory disorders. However, TSC1-deficient mice are highly resistant to M2-polarized allergic asthma. Inhibition of the mammalian target of rapamycin (mTOR) fails to reverse the hypersensitive M1 response of TSC1-deficient macrophages, but efficiently rescues the defective M2 polarization. Deletion of mTOR also fails to reverse the enhanced inflammatory response of TSC1-deficient macrophages. Molecular studies indicate that TSC1 inhibits M1 polarization by suppressing the Ras GTPase-Raf1-MEK-ERK pathway in mTOR-independent manner, whereas TSC1 promotes M2 properties by mTOR-dependent CCAAT/enhancer-binding protein-β pathways. Overall, these findings define a key role for TSC1 in orchestrating macrophage polarization via mTOR-dependent and independent pathways.
Collapse
Affiliation(s)
- Linnan Zhu
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China [2]
| | - Tao Yang
- 1] State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China [2]
| | - Longjie Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lina Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuzhu Hou
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuelian Hu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lianjun Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongling Tian
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Qingjie Zhao
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxia Peng
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongbing Zhang
- 1] National Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China [2] Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116085, China
| | - Ruoyu Wang
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Zhongzhou Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, CAMS &PUMC, Beijing 100021, China
| | - Yong Zhao
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
89
|
Park CS, Im SA, Song S, Kim K, Lee CK. Identification of HLA-A2-restricted immunogenic peptides derived from a xenogenic porcine major histocompatibility complex. Xenotransplantation 2014; 21:465-72. [DOI: 10.1111/xen.12119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/17/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Chan-Su Park
- College of Pharmacy; Chungbuk National University; Cheongju South Korea
| | - Sun-A Im
- College of Pharmacy; Chungbuk National University; Cheongju South Korea
| | - Sukgil Song
- College of Pharmacy; Chungbuk National University; Cheongju South Korea
| | - Kyungjae Kim
- College of Pharmacy; SahmYook University; Seoul South Korea
| | - Chong-Kil Lee
- College of Pharmacy; Chungbuk National University; Cheongju South Korea
| |
Collapse
|
90
|
Segovia-Gamboa N, Rodríguez-Arellano ME, Rangel-Cruz R, Sánchez-Díaz M, Ramírez-Reyes JC, Faradji R, González-Domínguez É, Sánchez-Torres C. Tolerogenic dendritic cells induce antigen-specific hyporesponsiveness in insulin- and glutamic acid decarboxylase 65-autoreactive T lymphocytes from type 1 diabetic patients. Clin Immunol 2014; 154:72-83. [PMID: 24993292 DOI: 10.1016/j.clim.2014.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/18/2014] [Accepted: 06/20/2014] [Indexed: 12/19/2022]
Abstract
Tolerogenic dendritic cells (tDC) constitute a promising therapy for autoimmune diseases, since they can anergize T lymphocytes recognizing self-antigens. Patients with type 1 diabetes mellitus (T1D) have autoreactive T cells against pancreatic islet antigens (insulin, glutamic acid decarboxylase 65 -GAD65-). We aimed to determine the ability of tDC derived from T1D patients to inactivate their insulin- and GAD65-reactive T cells. CD14+ monocytes and CD4+CD45RA- effector/memory lymphocytes were isolated from 25 patients. Monocyte-derived DC were generated in the absence (control, cDC) or presence of IL-10 and TGF-β1 (tDC), and loaded with insulin or GAD65. DC were cultured with T lymphocytes (primary culture), and cell proliferation and cytokine secretion were determined. These lymphocytes were rechallenged with insulin-, GAD65- or candidin-pulsed cDC (secondary culture) to assess whether tDC rendered T cells hyporesponsive to further stimulation. In the primary cultures, tDC induced significant lower lymphocyte proliferation and IL-2 and IFN-γ secretion than cDC; in contrast, tDC induced higher IL-10 production. Lymphocytes from 60% of patients proliferated specifically against insulin or GAD65 (group 1), whereas 40% did not (group 2). Most patients from group 1 had controlled glycemia. The secondary cultures showed tolerance induction to insulin or GAD65 in 14 and 10 patients, respectively. A high percentage of these patients (70-80%) belonged to group 1. Importantly, tDC induced antigen-specific T-cell hyporesponsiveness, since the responses against unrelated antigens were unaffected. These results suggest that tDC therapy against multiple antigens might be useful in a subset of T1D patients.
Collapse
Affiliation(s)
- Norma Segovia-Gamboa
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N. (CINVESTAV-IPN). Av. I.P.N. 2508, C.P. 07360, Mexico City, Mexico
| | | | - Rafael Rangel-Cruz
- Department of Endocrinology, Hospital Regional "Lic. Adolfo López Mateos", ISSSTE, Av. Universidad 1321, Mexico City, Mexico
| | - Moisés Sánchez-Díaz
- Department of Pediatrics, Hospital Regional "Lic. Adolfo López Mateos", ISSSTE, Av. Universidad 1321, Mexico City, Mexico
| | - Julio César Ramírez-Reyes
- Department of Pediatrics, Hospital Regional "Lic. Adolfo López Mateos", ISSSTE, Av. Universidad 1321, Mexico City, Mexico
| | - Raquel Faradji
- Medicina Interna, Asociación Médica, Centro Médico ABC, Sur 136 #116, Mexico City, Mexico
| | - Érika González-Domínguez
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N. (CINVESTAV-IPN). Av. I.P.N. 2508, C.P. 07360, Mexico City, Mexico
| | - Carmen Sánchez-Torres
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del I.P.N. (CINVESTAV-IPN). Av. I.P.N. 2508, C.P. 07360, Mexico City, Mexico.
| |
Collapse
|
91
|
He Y, Jie Y, Wang B, Zeng H, Zhang Y, Pan Z. Adoptive transfer of donor corneal antigen-specific regulatory T cells can prolong mice corneal grafts survival. Cornea 2014; 29 Suppl 1:S25-31. [PMID: 20935538 DOI: 10.1097/ico.0b013e3181ea4999] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To explore the effects of adoptive transferring T regulatory cells (T reg cells) stimulated by donor corneal antigen (Ag) to prevent corneal transplantation immune rejection in mice. METHODS C57BL/6 mice were used as donors and BALB/c mice as recipients. Corneal Ag was harvested by homogenization and centrifugation. Bone marrow dendritic cells (DCs) from BALB/c mice were cultured with stimulation of granulocyte-macrophage colony-stimulating factor and interleukin-4 for 5 days. Then, donor corneal Ag was added to obtain donor corneal Ag-loaded DCs. The DCs were used to stimulate CD4+CD25+ and CD4+CD25+ T cells from the recipient to yield Ag-stimulated T reg cells. Penetrating keratoplasty was performed in the mice. The recipients were randomly divided into 3 groups receiving 0.1 mL of phosphate-buffered saline, 1 × 10(6) naive T reg cells, and Ag-stimulated T reg cells, respectively, given by retroorbital injection at the end of surgery. The allografts were observed, and histopathological examination was performed 15 days after surgery. RESULTS : The corneal Ag mainly comprised 2 proteins with molecular weight 54 and 42 kD, respectively. Corneal Ag-loaded DCs expressed higher levels of CD11c, CD80, and CD86 than bone marrow precursor cells. Both CD4CD25 and CD4+CD25+ T cells showed vigorous proliferative responses to corneal Ag-loaded DCs. Mean survival time of the mice corneal allografts in phosphate-buffered saline, naive T reg cells, and Ag-stimulated groups was 8.1 ± 1.1, 14.3 ± 2.0, and 23.3 ± 2.6 days, respectively (P < 0.01 among groups). Histopathological examination revealed less inflammatory cells infiltration in Ag-stimulated than in naive mice. CONCLUSIONS : Adoptive transfer of donor corneal Ag-specific T reg cells prolonged survival time of corneal allografts in our mouse model, which might suggest a useful approach to cellular immunotherapy for corneal transplantation immune rejection.
Collapse
Affiliation(s)
- Yan He
- From the *Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China; and †Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
92
|
Abstract
Regulatory myeloid cells (RMC) are emerging as novel targets for immunosuppressive (IS) agents and hold considerable promise as cellular therapeutic agents. Herein, we discuss the ability of regulatory macrophages, regulatory dendritic cells, and myeloid-derived suppressor cells to regulate alloimmunity, their potential as cellular therapeutic agents, and the IS agents that target their function. We consider protocols for the generation of RMC and the selection of donor- or recipient-derived cells for adoptive cell therapy. Additionally, the issues of cell trafficking and antigen (Ag) specificity after RMC transfer are discussed. Improved understanding of the immunobiology of these cells has increased the possibility of moving RMC into the clinic to reduce the burden of current IS agents and to promote Ag-specific tolerance. In the second half of this review, we discuss the influence of established and experimental IS agents on myeloid cell populations. IS agents believed historically to act primarily on T cell activation and proliferation are emerging as important regulators of RMC function. Better insights into the influence of IS agents on RMC will enhance our ability to develop cell therapy protocols to promote the function of these cells. Moreover, novel IS agents may be designed to target RMC in situ to promote Ag-specific immune regulation in transplantation and to usher in a new era of immune modulation exploiting cells of myeloid origin.
Collapse
Affiliation(s)
- Brian R. Rosborough
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Dàlia Raïch-Regué
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Heth R. Turnquist
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Angus W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
93
|
Takasato F, Morita R, Schichita T, Sekiya T, Morikawa Y, Kuroda T, Niimi M, Yoshimura A. Prevention of allogeneic cardiac graft rejection by transfer of ex vivo expanded antigen-specific regulatory T-cells. PLoS One 2014; 9:e87722. [PMID: 24498362 PMCID: PMC3912059 DOI: 10.1371/journal.pone.0087722] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 12/30/2013] [Indexed: 01/09/2023] Open
Abstract
The rate of graft survival has dramatically increased using calcineurin inhibitors, however chronic graft rejection and risk of infection are difficult to manage. Induction of allograft-specific regulatory T-cells (Tregs) is considered an ideal way to achieve long-term tolerance for allografts. However, efficient in vitro methods for developing allograft-specific Tregs which is applicable to MHC full-mismatched cardiac transplant models have not been established. We compared antigen-nonspecific polyclonal-induced Tregs (iTregs) as well as antigen-specific iTregs and thymus-derived Tregs (nTregs) that were expanded via direct and indirect pathways. We found that iTregs induced via the indirect pathway had the greatest ability to prolong graft survival and suppress angiitis. Antigen-specific iTregs generated ex vivo via both direct and indirect pathways using dendritic cells from F1 mice also induced long-term engraftment without using MHC peptides. In antigen-specific Treg transferred models, activation of dendritic cells and allograft-specific CTL generation were suppressed. The present study demonstrated the potential of ex vivo antigen-specific Treg expansion for clinical cell-based therapeutic approaches to induce lifelong immunological tolerance for allogeneic cardiac transplants.
Collapse
Affiliation(s)
- Fumika Takasato
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, CREST, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, CREST, Tokyo, Japan
| | - Takashi Schichita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, CREST, Tokyo, Japan
| | - Takashi Sekiya
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, CREST, Tokyo, Japan
| | - Yasuhide Morikawa
- Department of Pediatric Surgery, International University Medical Welfare Hospital, Nasushiobara, Tochigi, Japan
| | - Tatsuo Kuroda
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masanori Niimi
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Japan Science and Technology Agency, CREST, Tokyo, Japan
- * E-mail:
| |
Collapse
|
94
|
Stenger EO, Rosborough BR, Mathews LR, Ma H, Mapara MY, Thomson AW, Turnquist HR. IL-12hi rapamycin-conditioned dendritic cells mediate IFN-γ-dependent apoptosis of alloreactive CD4+ T cells in vitro and reduce lethal graft-versus-host disease. Biol Blood Marrow Transplant 2014; 20:192-201. [PMID: 24239650 PMCID: PMC3964782 DOI: 10.1016/j.bbmt.2013.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 11/06/2013] [Indexed: 12/30/2022]
Abstract
Rapamycin (RAPA) inhibits the mechanistic target of rapamycin (mTOR), a crucial immune system regulator. Dendritic cells (DC) generated in RAPA (RAPA-DC) enrich for CD4(+) forkhead box p3 (FoxP3(+)) regulatory T cells and induce T cell apoptosis by an unknown mechanism. RAPA-DC also promote experimental allograft survival, yet paradoxically secrete increased IL-12, crucial for the generation of IFN-γ(+) CD4(+) T cells. However, IFN-γ is pro-apoptotic and IL-12-driven IFN-γ inhibits experimental graft-versus-host disease (GVHD). We hypothesized that IL-12(hi) RAPA-DC would facilitate IFN-γ-mediated apoptosis of alloreactive T cells and, unlike control (CTR)-DC, would reduce lethal GVHD. Following LPS stimulation, RAPA-DC exhibited decreased MHCII and co-stimulatory molecules and contained a significant population of CD86(lo) IL-12(hi) cells. Consistent with our hypothesis, both unstimulated and LPS-stimulated RAPA-DC enhanced alloreactive CD4(+) T cell apoptosis in culture. Augmented T cell apoptosis was ablated by IFN-γ neutralization or using T cells lacking the IFN-γ receptor, and it was associated with increased expression of Fas and cleaved caspase 8. DC production or responses to IFN-γ were not important to increased apoptotic functions of RAPA-DC. LPS-stimulated IL-12p40(-/-) RAPA-DC induced lower levels of T cell apoptosis in culture, which was further decreased with addition of anti-IFN-γ. Finally, whereas CTR-DC accelerated mortality from GVHD, LPS-treated RAPA-DC significantly prolonged host survival. In conclusion, increased apoptosis of allogeneic CD4(+) T cells induced by LPS-stimulated IL-12(hi) RAPA-DC is mediated in vitro through IFN-γ and in part by increased IL-12 expression. Enhanced production of IL-12, the predominant inducer of IFN-γ by immune cells, is a probable mechanism underlying the capacity of LPS-treated RAPA-DC to reduce GVHD.
Collapse
Affiliation(s)
- Elizabeth O Stenger
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Division of Pediatric Hematology/Oncology, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Brian R Rosborough
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lisa R Mathews
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Huihui Ma
- Division of Hematology/Oncology, Department of Medicine, Hematologic Malignancies Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Markus Y Mapara
- Division of Hematology/Oncology, Department of Medicine, Hematologic Malignancies Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hēth R Turnquist
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
95
|
Gordon JR, Ma Y, Churchman L, Gordon SA, Dawicki W. Regulatory dendritic cells for immunotherapy in immunologic diseases. Front Immunol 2014; 5:7. [PMID: 24550907 PMCID: PMC3907717 DOI: 10.3389/fimmu.2014.00007] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/07/2014] [Indexed: 12/12/2022] Open
Abstract
We recognize well the abilities of dendritic cells to activate effector T cell (Teff cell) responses to an array of antigens and think of these cells in this context as pre-eminent antigen-presenting cells, but dendritic cells are also critical to the induction of immunologic tolerance. Herein, we review our knowledge on the different kinds of tolerogenic or regulatory dendritic cells that are present or can be induced in experimental settings and humans, how they operate, and the diseases in which they are effective, from allergic to autoimmune diseases and transplant tolerance. The primary conclusions that arise from these cumulative studies clearly indicate that the agent(s) used to induce the tolerogenic phenotype and the status of the dendritic cell at the time of induction influence not only the phenotype of the dendritic cell, but also that of the regulatory T cell responses that they in turn mobilize. For example, while many, if not most, types of induced regulatory dendritic cells lead CD4+ naïve or Teff cells to adopt a CD25+Foxp3+ Treg phenotype, exposure of Langerhans cells or dermal dendritic cells to vitamin D leads in one case to the downstream induction of CD25+Foxp3+ regulatory T cell responses, while in the other to Foxp3− type 1 regulatory T cells (Tr1) responses. Similarly, exposure of human immature versus semi-mature dendritic cells to IL-10 leads to distinct regulatory T cell outcomes. Thus, it should be possible to shape our dendritic cell immunotherapy approaches for selective induction of different types of T cell tolerance or to simultaneously induce multiple types of regulatory T cell responses. This may prove to be an important option as we target diseases in different anatomic compartments or with divergent pathologies in the clinic. Finally, we provide an overview of the use and potential use of these cells clinically, highlighting their potential as tools in an array of settings.
Collapse
Affiliation(s)
- John R Gordon
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Yanna Ma
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Laura Churchman
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Sara A Gordon
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| | - Wojciech Dawicki
- Department of Medicine, University of Saskatchewan , Saskatoon, SK , Canada
| |
Collapse
|
96
|
Regulatory dendritic cell therapy: from rodents to clinical application. Immunol Lett 2013; 161:216-21. [PMID: 24316407 DOI: 10.1016/j.imlet.2013.11.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/24/2013] [Indexed: 12/21/2022]
Abstract
Dendritic cells (DC) are highly-specialized, bone marrow-derived antigen-presenting cells that induce or regulate innate and adaptive immunity. Regulatory or "tolerogenic" DC play a crucial role in maintaining self tolerance in the healthy steady-state. These regulatory innate immune cells subvert naïve or memory T cell responses by various mechanisms. Regulatory DC (DCreg) also exhibit the ability to induce or restore T cell tolerance in many animal models of autoimmune disease or transplant rejection. There is also evidence that adoptive transfer of DCreg can regulate T cell responses in non-human primates and humans. Important insights gained from in vitro studies and animal models have led recently to the development of clinical grade human DCreg, with potential to treat autoimmune disease or enhance transplant survival while reducing patient dependency on immunosuppressive drugs. Phase I trials have been conducted in type-1 diabetes and rheumatoid arthritis, with results that emphasize the feasibility and safety of DCreg therapy. This mini-review will outline how observations made using animal models have been translated into human use, and discuss the challenges faced in further developing this form of regulatory immune cell therapy in the fields of autoimmunity and transplantation.
Collapse
|
97
|
Seliger B, Massa C. The dark side of dendritic cells: development and exploitation of tolerogenic activity that favor tumor outgrowth and immune escape. Front Immunol 2013; 4:419. [PMID: 24348482 PMCID: PMC3845009 DOI: 10.3389/fimmu.2013.00419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/17/2013] [Indexed: 01/27/2023] Open
Abstract
Dendritic cells (DC) play a central role in the regulation of the immune responses by providing the information needed to decide between tolerance, ignorance, or active responses. For this reason different therapies aim at manipulating DC to obtain the desired response, such as enhanced cell-mediated toxicity against tumor and infected cells or the induction of tolerance in autoimmunity and transplantation. In the last decade studies performed in these settings have started to identify (some) molecules/factors involved in the acquisition of a tolerogenic DC phenotype as well as the underlying mechanisms of their regulatory function on different immune cell populations.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg , Halle (Saale) , Germany
| | - Chiara Massa
- Institute for Medical Immunology, Martin Luther University Halle-Wittenberg , Halle (Saale) , Germany
| |
Collapse
|
98
|
Geissler EK. The influence of mTOR inhibitors on immunity and the relationship to post-transplant malignancy. Transplant Res 2013; 2:S2. [PMID: 24565200 PMCID: PMC3834556 DOI: 10.1186/2047-1440-2-s1-s2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The known role of mammalian target of rapamycin (mTOR) in the immune response has been rapidly evolving, from what was once thought to be a simple immunosuppressive antiproliferative effect on T cells to a very complex central role that serves to integrate multiple signals given to T cells, B cells and antigen-presenting cells. The complexity of this topic is demonstrated by recent data suggesting that mTOR inhibition can either inhibit or promote certain aspects of immune responses, depending on the nature of the antigenic stimulus, and the environmental conditions cueing the cellular immunological players. There is even evidence that, under mTOR inhibition, an immune response to one foreign entity (for example, an organ transplant) may be simultaneously completely different to that of another (for example, tumour or microorganism). To understand how this might be possible, it is necessary to investigate the central role that mTOR seems to have in shaping the immune response. This review is aimed at examining how mTOR controls the development and function of key immune cells, and puts this information primarily in the context of organ transplant rejection and post-transplant malignancy.
Collapse
|
99
|
Kazma I, Lemoine R, Herr F, Chadet S, Meley D, Velge-Roussel F, Lebranchu Y, Baron C. Mycophenolic acid-treated dendritic cells generate regulatory CD4+ T cells that suppress CD8+ T cells’ allocytotoxicity. Int Immunol 2013; 26:173-81. [DOI: 10.1093/intimm/dxt054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
100
|
Mercalli A, Calavita I, Dugnani E, Citro A, Cantarelli E, Nano R, Melzi R, Maffi P, Secchi A, Sordi V, Piemonti L. Rapamycin unbalances the polarization of human macrophages to M1. Immunology 2013; 140:179-90. [PMID: 23710834 DOI: 10.1111/imm.12126] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 12/12/2022] Open
Abstract
Plasticity is a hallmark of macrophages, and in response to environmental signals these cells undergo different forms of polarized activation, the extremes of which are called classic (M1) and alternative (M2). Rapamycin (RAPA) is crucial for survival and functions of myeloid phagocytes, but its effects on macrophage polarization are not yet studied. To address this issue, human macrophages obtained from six normal blood donors were polarized to M1 or M2 in vitro by lipopolysaccharide plus interferon-γ or interleukin-4 (IL-4), respectively. The presence of RAPA (10 ng/ml) induced macrophage apoptosis in M2 but not in M1. Beyond the impact on survival in M2, RAPA reduced CXCR4, CD206 and CD209 expression and stem cell growth factor-β, CCL18 and CCL13 release. In contrast, in M1 RAPA increased CD86 and CCR7 expression and IL-6, tumour necrosis factor-α and IL-1β release but reduced CD206 and CD209 expression and IL-10, vascular endothelial growth factor and CCL18 release. In view of the in vitro data, we examined the in vivo effect of RAPA monotherapy (0·1 mg/kg/day) in 12 patients who were treated for at least 1 month before islet transplant. Cytokine release by Toll-like receptor 4-stimulated peripheral blood mononuclear cells showed a clear shift to an M1-like profile. Moreover, macrophage polarization 21 days after treatment showed a significant quantitative shift to M1. These results suggest a role of mammalian target of rapamycin (mTOR) into the molecular mechanisms of macrophage polarization and propose new therapeutic strategies for human M2-related diseases through mTOR inhibitor treatment.
Collapse
Affiliation(s)
- Alessia Mercalli
- Diabetes Research Institute (HSR-DRI), San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|