1
|
Meier RPH, Ben Nasr M, Fife BT, Finger EB, Fiorina P, Luo X, Bromberg JS. Best practices in islet transplantation in mice. Am J Transplant 2025:S1600-6135(25)00137-6. [PMID: 40089068 DOI: 10.1016/j.ajt.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/30/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Islet transplantation in mice serves as a crucial preclinical model for understanding alloimmune and autoimmune mechanisms, optimizing immunosuppressive strategies, and developing novel therapies for diabetes. This review provides a comprehensive overview of best practices in murine islet transplantation, including diabetes induction models, technical aspects of islet transplantation, and criteria for transplant graft and rejection. We discuss the immunological challenges posed by major histocompatibility complex disparities, the impact of various transplantation sites, and the limitations of murine models in translating findings to clinical settings. Special emphasis is placed on emerging strategies such as stem cell-derived insulin-producing cells, immune tolerance induction, and alternative transplantation sites. Although mouse models have significantly advanced our understanding of diabetes and β-cell replacement, their inherent differences from human physiology necessitate careful interpretation of findings. The review also highlights novel imaging modalities, immunosuppressive protocols, and biomarkers for graft monitoring, underscoring the need for further refinement of these models to bridge the gap between experimental research and clinical application. By standardizing methodologies and addressing translational limitations, murine islet transplantation studies remain a key model in transplantation and can continue to shape the future of β-cell replacement therapies for insulin-dependent diabetes.
Collapse
Affiliation(s)
- Raphael P H Meier
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| | - Moufida Ben Nasr
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian T Fife
- Department of Medicine, Division of Rheumatic and Autoimmune Diseases, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Erik B Finger
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland, School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
2
|
Tunbridge MJ, Luo X, Thomson AW. Negative Vaccination Strategies for Promotion of Transplant Tolerance. Transplantation 2024; 108:1715-1729. [PMID: 38361234 PMCID: PMC11265982 DOI: 10.1097/tp.0000000000004911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Organ transplantation requires the use of immunosuppressive medications that lack antigen specificity, have many adverse side effects, and fail to induce immunological tolerance to the graft. The safe induction of tolerance to allogeneic tissue without compromising host responses to infection or enhancing the risk of malignant disease is a major goal in transplantation. One promising approach to achieve this goal is based on the concept of "negative vaccination." Vaccination (or actively acquired immunity) involves the presentation of both a foreign antigen and immunostimulatory adjuvant to the immune system to induce antigen-specific immunity. By contrast, negative vaccination, in the context of transplantation, involves the delivery of donor antigen before or after transplantation, together with a "negative adjuvant" to selectively inhibit the alloimmune response. This review will explore established and emerging negative vaccination strategies for promotion of organ or pancreatic islet transplant tolerance. These include donor regulatory myeloid cell infusion, which has progressed to early-phase clinical trials, apoptotic donor cell infusion that has advanced to nonhuman primate models, and novel nanoparticle antigen-delivery systems.
Collapse
Affiliation(s)
- Matthew J. Tunbridge
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Department of Medicine (Nephrology), Duke University Medical Center, Durham, North Carolina, USA
| | - Xunrong Luo
- Department of Medicine (Nephrology), Duke University Medical Center, Durham, North Carolina, USA
| | - Angus W. Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Vafadar A, Vosough P, Jahromi HK, Tajbakhsh A, Savardshtaki A, Butler AE, Sahebkar A. The role of efferocytosis and transplant rejection: Strategies in promoting transplantation tolerance using apoptotic cell therapy and/or synthetic particles. Cell Biochem Funct 2023; 41:959-977. [PMID: 37787641 DOI: 10.1002/cbf.3852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
Recently, efforts have been made to recognize the precise reason(s) for transplant failure and the process of rejection utilizing the molecular signature. Most transplant recipients do not appreciate the unknown length of survival of allogeneic grafts with the existing standard of care. Two noteworthy immunological pathways occur during allogeneic transplant rejection. A nonspecific innate immune response predominates in the early stages of the immune reaction, and allogeneic antigens initiate a donor-specific adaptive reaction. Though the adaptive response is the major cause of allograft rejection, earlier pro-inflammatory responses that are part of the innate immune response are also regarded as significant in graft loss. The onset of the innate and adaptive immune response causes chronic and acute transplant rejection. Currently employed immunosuppressive medications have shown little or no influence on chronic rejection and, as a result, on overall long-term transplant survival. Furthermore, long-term pharmaceutical immunosuppression is associated with side effects, toxicity, and an increased risk of developing diseases, both infectious and metabolic. As a result, there is a need for the development of innovative donor-specific immunosuppressive medications to regulate the allorecognition pathways that induce graft loss and to reduce the side effects of immunosuppression. Efferocytosis is an immunomodulatory mechanism with fast and efficient clearance of apoptotic cells (ACs). As such, AC therapy strategies have been suggested to limit transplant-related sequelae. Efferocytosis-based medicines/treatments can also decrease the use of immunosuppressive drugs and have no detrimental side effects. Thus, this review aims to investigate the impact of efferocytosis on transplant rejection/tolerance and identify approaches using AC clearance to increase transplant viability.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland - Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Li F, Li F, Urie R, Bealer E, Ruiz RO, Saito E, Turan A, Yolcu E, Shirwan H, Shea LD. Membrane-coated nanoparticles for direct recognition by T cells. Biotechnol Bioeng 2023; 120:767-777. [PMID: 36515455 DOI: 10.1002/bit.28304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
The direct modulation of T cell responses is an emerging therapeutic strategy with the potential to modulate undesired immune responses including, autoimmune disease, and allogeneic cells transplantation. We have previously demonstrated that poly(lactide-co-glycolide) particles were able to modulate T cell responses indirectly through antigen-presenting cells (APCs). In this report, we investigated the design of nanoparticles that can directly interact and modulate T cells by coating the membranes from APCs onto nanoparticles to form membrane-coated nanoparticles (MCNPs). Proteins within the membranes of the APCs, such as Major Histocompatibility Complex class II and co-stimulatory factors, were effectively transferred to the MCNP. Using alloreactive T cell models, MCNP derived from allogeneic dendritic cells were able to stimulate proliferation, which was not observed with membranes from syngeneic dendritic cells and influenced cytokine secretion. Furthermore, we investigated the engineering of the membranes either on the dendritic cells or postfabrication of MCNP. Engineered membranes could be to promote antigen-specific responses, to differentially activate T cells, or to directly induce apoptosis. Collectively, MCNPs represent a tunable platform that can directly interact with and modulate T cell responses.
Collapse
Affiliation(s)
- Feiran Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Fanghua Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Russell Urie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth Bealer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ramon Ocadiz Ruiz
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ali Turan
- Department of Child Health and Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Esma Yolcu
- Department of Child Health and Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Haval Shirwan
- Department of Child Health and Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
5
|
Shaw BI, Ord JR, Nobuhara C, Luo X. Cellular Therapies in Solid Organ Allotransplantation: Promise and Pitfalls. Front Immunol 2021; 12:714723. [PMID: 34526991 PMCID: PMC8435835 DOI: 10.3389/fimmu.2021.714723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/30/2022] Open
Abstract
Donor specific transfusions have been the basis of tolerance inducing protocols since Peter Medawar showed that it was experimentally feasible in the 1950s. Though trials of cellular therapies have become increasingly common in solid organ transplantation, they have not become standard practice. Additionally, whereas some protocols have focused on cellular therapies as a method for donor antigen delivery-thought to promote tolerance in and of itself in the correct immunologic context-other approaches have alternatively focused on the intrinsic immunosuppressive properties of the certain cell types with less emphasis on their origin, including mesenchymal stem cells, regulatory T cells, and regulatory dendritic cells. Regardless of intent, all cellular therapies must contend with the potential that introducing donor antigen in a new context will lead to sensitization. In this review, we focus on the variety of cellular therapies that have been applied in human trials and non-human primate models, describe their efficacy, highlight data regarding their potential for sensitization, and discuss opportunities for cellular therapies within our current understanding of the immune landscape.
Collapse
Affiliation(s)
- Brian I. Shaw
- Department of Surgery, Duke University, Durham, NC, United States
| | - Jeffrey R. Ord
- School of Medicine, Duke University, Durham, NC, United States
| | - Chloe Nobuhara
- School of Medicine, Duke University, Durham, NC, United States
| | - Xunrong Luo
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Husain I, Luo X. Apoptotic Donor Cells in Transplantation. Front Immunol 2021; 12:626840. [PMID: 33717145 PMCID: PMC7947657 DOI: 10.3389/fimmu.2021.626840] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/18/2021] [Indexed: 12/31/2022] Open
Abstract
Despite significant advances in prevention and treatment of transplant rejection with immunosuppressive medications, we continue to face challenges of long-term graft survival, detrimental medication side effects to both the recipient and transplanted organ together with risks for opportunistic infections. Transplantation tolerance has so far only been achieved through hematopoietic chimerism, which carries with it a serious and life-threatening risk of graft versus host disease, along with variability in persistence of chimerism and uncertainty of sustained tolerance. More recently, numerous in vitro and in vivo studies have explored the therapeutic potential of silent clearance of apoptotic cells which have been well known to aid in maintaining peripheral tolerance to self. Apoptotic cells from a donor not only have the ability of down regulating the immune response, but also are a way of providing donor antigens to recipient antigen-presenting-cells that can then promote donor-specific peripheral tolerance. Herein, we review both laboratory and clinical evidence that support the utility of apoptotic cell-based therapies in prevention and treatment of graft versus host disease and transplant rejection along with induction of donor-specific tolerance in solid organ transplantation. We have highlighted the potential limitations and challenges of this apoptotic donor cell-based therapy together with ongoing advancements and attempts made to overcome them.
Collapse
Affiliation(s)
- Irma Husain
- Department of Medicine, Duke University, Durham, NC, United States
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
7
|
Dangi A, Yu S, Lee FT, Burnette M, Knechtle S, Kwun J, Luo X. Donor apoptotic cell-based therapy for effective inhibition of donor-specific memory T and B cells to promote long-term allograft survival in allosensitized recipients. Am J Transplant 2020; 20:2728-2739. [PMID: 32275799 PMCID: PMC7896418 DOI: 10.1111/ajt.15878] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 01/25/2023]
Abstract
Allosensitization constitutes a major barrier in transplantation. Preexisting donor-reactive memory T and B cells and preformed donor-specific antibodies (DSAs) have all been implicated in accelerated allograft rejection in sensitized recipients. Here, we employ a sensitized murine model of islet transplantation to test strategies that promote long-term immunosuppression-free allograft survival. We demonstrate that donor-specific memory T and B cells can be effectively inhibited by peritransplant infusions of donor apoptotic cells in combination with anti-CD40L and rapamycin, and this treatment leads to significant prolongation of islet allograft survival in allosensitized recipients. We further demonstrate that late graft rejection in recipients treated with this regimen is associated with a breakthrough of B cells and their aggressive graft infiltration. Consequently, additional posttransplant B cell depletion effectively prevents late rejection and promotes permanent acceptance of islet allografts. In contrast, persistent low levels of DSAs do not seem to impair graft outcome in these recipients. We propose that B cells contribute to late rejection as antigen-presenting cells for intragraft memory T cell expansion but not to alloantibody production and that a therapeutic strategy combining donor apoptotic cells, anti-CD40L, and rapamycin effectively inhibits proinflammatory B cells and promotes long-term islet allograft survival in such recipients.
Collapse
Affiliation(s)
- Anil Dangi
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Shuangjin Yu
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Frances T. Lee
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Melanie Burnette
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Stuart Knechtle
- Department of Surgery, Duke University Medical Center, Durham, North Carolina,Duke Transplant Center, Duke University Medical Center, Durham, North Carolina
| | - Jean Kwun
- Department of Surgery, Duke University Medical Center, Durham, North Carolina,Duke Transplant Center, Duke University Medical Center, Durham, North Carolina
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham, North Carolina,Duke Transplant Center, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
8
|
Shah S, DeBerge M, Iovane A, Yan S, Qiu L, Wang JJ, Kanwar YS, Hummel M, Zhang ZJ, Abecassis MM, Luo X, Thorp EB. MCMV Dissemination from Latently-Infected Allografts Following Transplantation into Pre-Tolerized Recipients. Pathogens 2020; 9:pathogens9080607. [PMID: 32722544 PMCID: PMC7460028 DOI: 10.3390/pathogens9080607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Transplantation tolerance is achieved when recipients are unresponsive to donor alloantigen yet mobilize against third-party antigens, including virus. After transplantation, cytomegalovirus (CMV) reactivation in latently-infected transplants reduces allograft viability. To determine if pre-tolerized recipients are resistant to viral dissemination in this setting, we transfused chemically-fixed donor splenocytes (1-ethyl-3- (3′-dimethyl-aminopropyl)-carbo-diimide (ECDI)-treated splenocytes (ECDIsp)) to induce donor antigen tolerance without immunosuppression. In parallel, we implanted donor islet cells to validate operational tolerance. These pre-tolerized recipients were implanted with murine CMV (MCMV) latently-infected donor kidneys (a validated model of CMV latency) to monitor graft inflammation and viral dissemination. Our results indicate that tolerance to donor islets was sustained in recipients after implantation of donor kidneys. In addition, kidney allografts implanted after ECDIsp and islet implantation exhibited low levels of fibrosis and tubulitis. In contrast, kidney cellular and innate immune infiltrates trended higher in the CMV group and exhibited increased markers of CD8+ T cell activation. Tolerance induction was unable to prevent increases in MCMV-specific CD8+ T cells or dissemination of viral IE-1 DNA. Our data suggest that latently-infected allografts are inherently more susceptible to inflammation that is associated with viral dissemination in pre-tolerized recipients. Thus, CMV latently-infected allografts require enhanced strategies to protect allograft integrity and viral spread.
Collapse
Affiliation(s)
- Sahil Shah
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA;
| | - Matthew DeBerge
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (M.D.); (Y.S.K.)
| | - Andre Iovane
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
| | - Shixian Yan
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
| | - Longhui Qiu
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
| | - Yashpal S. Kanwar
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (M.D.); (Y.S.K.)
| | - Mary Hummel
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Zheng J. Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27705, USA;
| | - Edward B. Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (M.D.); (Y.S.K.)
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (A.I.); (S.Y.); (L.Q.); (J.-J.W.); (M.H.); (Z.J.Z.)
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-312-503-4309
| |
Collapse
|
9
|
Zhou B, Zhang Y, Zhang D, Zhang Y, Xie J, Zhang X, Ding J, Su Y, Guo S, Zhuang R. ECDI-fixed donor splenocytes prolong skin allograft survival by promoting M2 macrophage polarization and inducing regulatory T cells. FASEB Bioadv 2019; 1:706-718. [PMID: 32123816 PMCID: PMC6996306 DOI: 10.1096/fba.2019-00029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 03/29/2019] [Accepted: 09/30/2019] [Indexed: 11/27/2022] Open
Abstract
Rejection is a common complication of allogeneic tissue transplantation. Fixation of splenocytes (SP) with 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide (ECDI) induces immune tolerance in recipients post-transplantation; however, the mechanism underlying this effect remains unclear. Here, we determined the mechanisms of ECDI-fixed donor SP (ECDI-SP) in inducing tolerance in skin allograft transplantation. C57BL/6-recipient mice that received Balb/c full-thickness skin transplants with two infusions of donor-derived ECDI-SP, along with rapamycin showed superior skin allograft survival and lower inflammatory cell infiltration than mice that received rapamycin-only treatment. In ECDI-SP-treated mice, the levels of anti-inflammatory cytokines such as interleukin (IL)-10 in sera were markedly increased, whereas the expression of inflammatory cytokines was significantly suppressed. Splenic macrophages were significantly polarized to the alternative activated macrophage (M2) phenotype, with expansion of CD4+Foxp3+ regulatory T cells (Tregs) in the spleen and draining lymph nodes. Allostimulatory activity of ECDI-SP in vitro and donor-specific ex vivo hyporesponsiveness were observed. C57BL/6 macrophages engulfed allogeneic Balb/c-derived ECDI-SP, polarized to the M2 phenotype, with pronounced cAMP response element-binding (CREB) protein phosphorylation. By facilitating increased IL-10 expression, ECDI-SP induced M2 polarization and Treg production, inhibiting effector T-cell proliferation. Thus, ECDI-SP modulates macrophage M2 polarization by increasing CREB phosphorylation and promoting Treg production to suppress allogeneic skin graft rejection.
Collapse
Affiliation(s)
- Bo Zhou
- Department of ImmunologyFourth Military Medical UniversityXi'anChina
- Department of Plastic and Reconstructive SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Yuan Zhang
- Transplant Immunology LaboratoryFourth Military Medical UniversityXi'anChina
| | - Dongliang Zhang
- Department of Plastic and Reconstructive SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Yun Zhang
- Department of ImmunologyFourth Military Medical UniversityXi'anChina
| | - Jiangang Xie
- Transplant Immunology LaboratoryFourth Military Medical UniversityXi'anChina
| | - Xuexin Zhang
- Transplant Immunology LaboratoryFourth Military Medical UniversityXi'anChina
| | - Jianke Ding
- Department of Plastic and Reconstructive SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Yingjun Su
- Department of Plastic and Reconstructive SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Shuzhong Guo
- Department of Plastic and Reconstructive SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Ran Zhuang
- Department of ImmunologyFourth Military Medical UniversityXi'anChina
- Transplant Immunology LaboratoryFourth Military Medical UniversityXi'anChina
| |
Collapse
|
10
|
Dangi A, Yu S, Luo X. Apoptotic cell-based therapies for promoting transplantation tolerance. Curr Opin Organ Transplant 2019; 23:552-558. [PMID: 30024416 DOI: 10.1097/mot.0000000000000562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW This article is aimed to provide readers with an updated review on the applicability, efficacy, and challenges of employing donor apoptotic cell-based therapies to promote transplantation tolerance in various experimental and clinical settings. RECENT FINDINGS Recently, donor apoptotic cell-based therapies have been employed in various models of cell (including pancreatic islets and bone marrow hematopoietic stem cells) and solid organ (heart and kidney) transplantation to promote donor-specific tolerance. Published data, thus far, have revealed a high potential of this approach in inducing robust transplantation tolerance. Recent clinical trials have also underscored the safety and potential efficacy of this approach in alleviating graft-versus-host disease (GVHD) in bone marrow transplantation (BMT). Host factors including prior allo-sensitization and opportunistic infections pose major obstacles in establishing transplantation tolerance employing this strategy. However, emerging data provide strategies for overcoming such obstacles in these clinically relevant settings. SUMMARY Donor apoptotic cell therapy is an emerging strategy in promoting transplantation tolerance, with recent data emphasizing its efficacy and applicability for transplantation tolerance in the clinic.
Collapse
Affiliation(s)
- Anil Dangi
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute.,Division of Nephrology and Hypertension, Department of Medicine
| | - Shuangjin Yu
- Division of Nephrology and Hypertension, Department of Medicine
| | - Xunrong Luo
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute.,Division of Nephrology and Hypertension, Department of Medicine.,Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Ilinois, USA
| |
Collapse
|
11
|
Optimizing PLG nanoparticle-peptide delivery platforms for transplantation tolerance using an allogeneic skin transplant model. Biomaterials 2019; 210:70-82. [PMID: 31077862 DOI: 10.1016/j.biomaterials.2019.04.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022]
Abstract
A robust regimen for inducing allogeneic transplantation tolerance involves pre-emptive recipient treatment with donor splenocytes (SP) rendered apoptotic by 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide(ECDI) treatment. However, such a regimen is limited by availability of donor cells, cost of cell procurement, and regulatory hurdles associated with cell-based therapies. Nanoparticles (NP) delivering donor antigens are a promising alternative for promoting transplantation tolerance. Here, we used a B6.C-H-2bm12(bm12) to C57BL/6(B6) skin transplant model involving a defined major histocompatibility antigen mismatch to investigate design parameters of poly(lactide-co-glycolide) (PLG) NPs delivering peptides containing the donor antigen for optimizing skin allograft survival. We showed that an epitope-containing short peptide (P1) was more effective than a longer peptide (P2) at providing graft protection. Importantly, the NP and P1 complex (NP-ECDI-P1) resulted in a significant expansion of graft-infiltrating Tregs. Interestingly, in comparison to donor ECDI-SP that provided indefinite graft protection, NP-ECDI-P1 targeted different splenic phagocytes and skin allografts in these recipients harbored significantly more graft-infiltrating CD8+IFN-γ+ cells. Collectively, the current study provides initial engineering parameters for a cell-free and biocompatible NP-peptide platform for transplant immunoregulation. Moreover, it also provides guidance to future NP engineering endeavors to recapitulate the effects of donor ECDI-SP as a goal for maximizing tolerance efficacy of NP formulations.
Collapse
|
12
|
Zhang L, DeBerge M, Wang J, Dangi A, Zhang X, Schroth S, Zhang Z, Thorp E, Luo X. Receptor tyrosine kinase MerTK suppresses an allogenic type I IFN response to promote transplant tolerance. Am J Transplant 2019; 19:674-685. [PMID: 30133807 PMCID: PMC6393931 DOI: 10.1111/ajt.15087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/27/2018] [Accepted: 08/15/2018] [Indexed: 01/25/2023]
Abstract
Recipient infusion of donor apoptotic cells is an emerging strategy for inducing robust transplantation tolerance. Daily clearance of billions of self-apoptotic cells relies on homeostatic engagement of phagocytic receptors, in particular, receptors of the tyrosine kinase family TAM (Tyro3, Axl, and MerTK), to maintain self-tolerance. However, an outstanding question is if allogeneic apoptotic cells trigger the same receptor system for inducing allogeneic tolerance. Here, we employed allogeneic apoptotic splenocytes and discovered that the efferocytic receptor MerTK on recipient phagocytes is a critical mediator for transplantation tolerance induced by this strategy. Our findings indicate that the tolerogenic properties of allogeneic apoptotic splenocytes require MerTK transmission of intracellular signaling to suppress the production of inflammatory cytokine interferon α (IFN-α). We further demonstrate that MerTK is crucial for subsequent expansion of myeloid-derived suppressor cells and for promoting their immunomodulatory function, including maintaining graft-infiltrating CD4+ CD25+ Foxp3+ regulatory T cells. Consequently, recipient MerTK deficiency resulted in failure of tolerance by donor apoptotic cells, and this failure could be effectively rescued by IFN-α receptor blockade. These findings underscore the importance of the efferocytic receptor MerTK in mediating transplantation tolerance by donor apoptotic cells and implicate MerTK agonism as a promising target for promoting transplantation tolerance.
Collapse
Affiliation(s)
- Lei Zhang
- Center for Kidney Research and Therapeutics, Feinberg
Cardiovascular Research Institute, Northwestern University Feinberg School of
Medicine, Chicago, IL, United States,Northwestern University Feinberg School of Medicine,
Division of Nephrology and Hypertension, Chicago, IL, United States
| | - Mathew DeBerge
- Northwestern University Feinberg School of Medicine,
Department of Pathology & Feinberg Cardiovascular and Renal Research Institute,
Chicago, IL, United States
| | - Jiaojin Wang
- Comprehensive Transplant Center, Northwestern University
Feinberg School of Medicine, Chicago, IL, United States
| | - Anil Dangi
- Center for Kidney Research and Therapeutics, Feinberg
Cardiovascular Research Institute, Northwestern University Feinberg School of
Medicine, Chicago, IL, United States,Northwestern University Feinberg School of Medicine,
Division of Nephrology and Hypertension, Chicago, IL, United States
| | - Xiaomin Zhang
- Comprehensive Transplant Center, Northwestern University
Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha Schroth
- Northwestern University Feinberg School of Medicine,
Department of Pathology & Feinberg Cardiovascular and Renal Research Institute,
Chicago, IL, United States
| | - Zheng Zhang
- Comprehensive Transplant Center, Northwestern University
Feinberg School of Medicine, Chicago, IL, United States
| | - Edward Thorp
- Northwestern University Feinberg School of Medicine,
Department of Pathology & Feinberg Cardiovascular and Renal Research Institute,
Chicago, IL, United States
| | - Xunrong Luo
- Center for Kidney Research and Therapeutics, Feinberg
Cardiovascular Research Institute, Northwestern University Feinberg School of
Medicine, Chicago, IL, United States,Northwestern University Feinberg School of Medicine,
Division of Nephrology and Hypertension, Chicago, IL, United States,Comprehensive Transplant Center, Northwestern University
Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
Gammon JM, Jewell CM. Engineering Immune Tolerance with Biomaterials. Adv Healthc Mater 2019; 8:e1801419. [PMID: 30605264 PMCID: PMC6384133 DOI: 10.1002/adhm.201801419] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/05/2018] [Indexed: 12/28/2022]
Abstract
Autoimmune diseases, rejection of transplanted organs and grafts, chronic inflammatory diseases, and immune-mediated rejection of biologic drugs impact a large number of people across the globe. New understanding of immune function is revealing exciting opportunities to help tackle these challenges by harnessing-or correcting-the specificity of immune function. However, realizing this potential requires precision control over the interaction between regulatory immune cues, antigens attacked during inflammation, and the tissues where these processes occur. Engineered materials-such as polymeric and lipid particles, scaffolds, and inorganic materials-offer powerful features that can help to selectively regulate immune function during disease without compromising healthy immune functions. This review highlights some of the exciting developments to leverage biomaterials as carriers, depots, scaffolds-and even as agents with intrinsic immunomodulatory features-to promote immunological tolerance.
Collapse
Affiliation(s)
- Joshua M. Gammon
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive RM 5110, College Park, MD 20742, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive RM 5110, College Park, MD 20742, USA ; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD 20742, USA; United States Department of Veterans Affairs, Baltimore VA Medical center, 10. N Green Street, Baltimore, Maryland 21201, USA; Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
14
|
Murine CMV induces type 1 IFN that impairs differentiation of MDSCs critical for transplantation tolerance. Blood Adv 2019; 2:669-680. [PMID: 29563123 DOI: 10.1182/bloodadvances.2017012187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/01/2018] [Indexed: 01/03/2023] Open
Abstract
Clinical tolerance without immunosuppression has now been achieved for organ transplantation, and its scope will likely continue to expand. In this context, a previously understudied and now increasingly relevant area is how microbial infections might affect the efficacy of tolerance. A highly prevalent and clinically relevant posttransplant pathogen is cytomegalovirus (CMV). Its impact on transplantation tolerance and graft outcomes is not well defined. Employing a mouse model of CMV (MCMV) infection and allogeneic pancreatic islet transplantation in which donor-specific tolerance was induced by infusing donor splenocytes rendered apoptotic by treatment with ethylenecarbodiimide, we investigated the effect of CMV infection on transplantation tolerance induction. We found that acute MCMV infection abrogated tolerance induction and that this abrogation correlated with an alteration in the differentiation and function of myeloid-derived suppressor cells (MDSCs). These effects on MDSCs were mediated in part through MCMV induced type 1 interferon (IFN) production. During MCMV infection, the highly immunosuppressive Gr1HI-granulocytic MDSCs were markedly reduced in numbers, and the accumulating Ly6CHI-monocytic cells lost their MDSC-like function but instead acquired an immunostimulatory phenotype to cross-present alloantigens and prime alloreactive CD8 T cells. Consequently, the islet allograft exhibited an altered effector to regulatory T-cell ratio that correlated with the ultimate graft demise. Blocking type 1 IFN signaling during MCMV infection rescued MDSC populations and partially restored transplantation tolerance. Our mechanistic studies now provide a solid foundation for seeking effective therapies for promoting transplantation tolerance in settings of CMV infection.
Collapse
|
15
|
Zou J, Gao X, Liu T, Liang R, Liu Y, Wang G, Wang L, Liu N, Sun P, Wang Z, Wang S, Shen Z. Ethylenecarbodiimide-fixed splenocytes carrying whole islet antigens decrease the incidence of diabetes in NOD mice via down-regulation of effector memory T cells and autoantibodies. Endocr J 2018; 65:943-952. [PMID: 29998909 DOI: 10.1507/endocrj.ej18-0158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a syndrome of loss of glucose homeostasis caused by the loss of β cell chronic autoimmunity against islet cells. Islet-specific epitopes coupled antigen presenting cells by Ethylenecarbodiimide (ECDI) is a promising strategy to induce antigen-specific tolerance. However, single epitope induced tolerance is insufficient to prevent the onset of T1DM. The aim of this study is to evaluate the efficacy of whole islet antigens in preventing the onset and progression of T1DM and identify the underlying immune mechanism in NOD mice. In this study, the whole islet antigens, derived from islet lysate isolated from BALB/c mice, were coupled to splenocytes of BALB/c mice by ECDI fixation (SP-Islet lysate), and then intravenously administrated to NOD mice. The results showed that, compared with control group, SP-Islet lysate group significantly decreased T1DM incidence and improved the survival of NOD mice. SP-Islet lysate treated mice had reduced insulitis score and autoantibody levels, and improved glucose tolerance and insulin/glucagon production. Furthermore, the effector memory T cells (TEMs) were downregulated and regulatory T cells (Tregs) were upregulated by the SP-Islet lysate treatment, with reduced populations of Th1&Th17 cells. In conclusion, ECDI-fixed splenocytes carrying whole islet antigens effectively prevented the onset of T1DM in NOD mice, via suppressing the production of autoantibodies and inducing anergy of autoreactive T cells.
Collapse
Affiliation(s)
- Jiaqi Zou
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Tianjin, China
| | - Xinpu Gao
- Tianjin Medical University, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China
| | - Tengli Liu
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Rui Liang
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Yaojuan Liu
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Guanqiao Wang
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Le Wang
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Na Liu
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Peng Sun
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
| | - Zhiping Wang
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Clinical Research Center for Organ Transplantation, Tianjin First Center Hospital, Tianjin, China
| | - Shusen Wang
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Clinical Research Center for Organ Transplantation, Tianjin First Center Hospital, Tianjin, China
| | - Zhongyang Shen
- Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin First Center Hospital, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Tianjin, China
- Tianjin Clinical Research Center for Organ Transplantation, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
16
|
Ding J, Liu S, Zhang D, Song Y, Ma X, Yi C, Song B, Xiao B, Su Y, Guo S. Transfusion of ethylene carbodiimide–fixed donor splenocytes prolongs survival of vascularized skin allografts. J Surg Res 2018; 221:343-352. [DOI: 10.1016/j.jss.2017.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/24/2017] [Accepted: 06/06/2017] [Indexed: 10/18/2022]
|
17
|
Dangi A, Luo X. Harnessing Apoptotic Cells for Transplantation Tolerance: Current Status and Future Perspectives. CURRENT TRANSPLANTATION REPORTS 2017; 4:270-279. [PMID: 29177124 PMCID: PMC5697727 DOI: 10.1007/s40472-017-0167-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW The use of donor apoptotic cells is an emerging therapy for inducing transplantation tolerance. In this review, we will discuss current understanding of mechanisms of this approach, as well as crucial aspects necessary for successful translation of this approach to clinical transplantation. RECENT FINDINGS Transplantation tolerance by donor apoptotic cells is mediated by their homeostatic interaction with recipient phagocytes, and subsequent expansion of suppressor cell populations as well as inhibition of effector T cells via deletion and anergy. To ensure their tolerogenicity, it is critical to procure non-stressed donor cells, and to induce and arrest their apoptosis at the appropriate stage prior to their administration. Equally important is the monitoring of dynamics of recipient immunological status, and its influences on tolerance efficacy and longevity. Emerging concepts and technologies may significantly streamline tolerogen manufacture and delivery of this approach, and smooth its transition to clinical application. SUMMARY Hijacking homeostatic clearance of donor apoptotic cells is a promising strategy for transplantation tolerance. Timing is now mature for concerted efforts for transitioning this strategy to clinical transplantation.
Collapse
Affiliation(s)
- Anil Dangi
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xunrong Luo
- Center for Kidney Research and Therapeutics, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
18
|
Differential Role of B Cells and IL-17 Versus IFN-γ During Early and Late Rejection of Pig Islet Xenografts in Mice. Transplantation 2017; 101:1801-1810. [PMID: 27893617 DOI: 10.1097/tp.0000000000001489] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Xenogeneic islet transplantation is an emerging therapeutic option for diabetic patients. However, immunological tolerance to xenogeneic islets remains a challenge. METHODS The current study used a pig-to-mouse discordant xenogeneic islet transplant model to examine antidonor xenogeneic immune responses during early and late rejection and to determine experimental therapeutic interventions that promote durable pig islet xenograft survival. RESULTS We found that during early acute rejection of pig islet xenografts, the rejecting hosts exhibited a heavy graft infiltration with B220 B cells and a robust antipig antibody production. In addition, early donor-stimulated IL-17 production, but not IFN-γ production, dominated during early acute rejection. Recipient treatment with donor apoptotic 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide-treated splenocytes significantly inhibited antidonor IL-17 response, and when combined with B cell depletion and a short course of rapamycin led to survival of pig islet xenografts beyond 100 days in approximately 65% recipients. Interestingly, treated recipients in this model experienced late rejection between 100 and 200 days posttransplant, which coincided with B cell reconstitution and an ensuing emergence of a robust antidonor IFN-γ, but not IL-17, response. CONCLUSIONS These findings reveal that early and late rejection of pig islet xenografts may be dominated by different immune responses and that maintenance of long-term xenogeneic tolerance will require strategies that target the temporal sequence of antixenogeneic immune responses.
Collapse
|
19
|
McCarthy DP, Yap JWT, Harp CT, Song WK, Chen J, Pearson RM, Miller SD, Shea LD. An antigen-encapsulating nanoparticle platform for T H1/17 immune tolerance therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2017; 13:191-200. [PMID: 27720992 PMCID: PMC5237397 DOI: 10.1016/j.nano.2016.09.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/12/2016] [Accepted: 09/20/2016] [Indexed: 01/19/2023]
Abstract
Tolerogenic nanoparticles (NPs) are rapidly being developed as specific immunotherapies to treat autoimmune disease. However, many NP-based therapies conjugate antigen (Ag) directly to the NP posing safety concerns due to antibody binding or require the co-delivery of immunosuppressants to induce tolerance. Here, we developed Ag encapsulated NPs comprised of poly(lactide-co-glycolide) [PLG(Ag)] and investigated the mechanism of action for Ag-specific tolerance induction in an autoimmune model of T helper type 1/17 dysfunction - relapse-remitting experimental autoimmune encephalomyelitis (R-EAE). PLG(Ag) completely abrogated disease induction in an organ specific manner, where the spleen was dispensable for tolerance induction. PLG(Ag) delivered intravenously distributed to the liver, associated with macrophages, and recruited Ag-specific T cells. Furthermore, programmed death ligand 1 (PD-L1) was increased on Ag presenting cells and PD-1 blockade lessened tolerance induction. The robust promotion of tolerance by PLG(Ag) without co-delivery of immunosuppressive drugs, suggests that these NPs effectively deliver antigen to endogenous tolerogenic pathways.
Collapse
Affiliation(s)
- Derrick P McCarthy
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Christopher T Harp
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - W Kelsey Song
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Jeane Chen
- Department of Chemical and Biological Engineering, Evanston, IL, USA
| | - Ryan M Pearson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Lonnie D Shea
- Department of Biomedical Engineering, Evanston, IL, USA; Department of Chemical and Biological Engineering, Evanston, IL, USA; Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA; The Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Burrack AL, Martinov T, Fife BT. T Cell-Mediated Beta Cell Destruction: Autoimmunity and Alloimmunity in the Context of Type 1 Diabetes. Front Endocrinol (Lausanne) 2017; 8:343. [PMID: 29259578 PMCID: PMC5723426 DOI: 10.3389/fendo.2017.00343] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022] Open
Abstract
Type 1 diabetes (T1D) results from destruction of pancreatic beta cells by T cells of the immune system. Despite improvements in insulin analogs and continuous blood glucose level monitoring, there is no cure for T1D, and some individuals develop life-threatening complications. Pancreas and islet transplantation have been attractive therapeutic approaches; however, transplants containing insulin-producing cells are vulnerable to both recurrent autoimmunity and conventional allograft rejection. Current immune suppression treatments subdue the immune system, but not without complications. Ideally a successful approach would target only the destructive immune cells and leave the remaining immune system intact to fight foreign pathogens. This review discusses the autoimmune diabetes disease process, diabetic complications that warrant a transplant, and alloimmunity. First, we describe the current understanding of autoimmune destruction of beta cells including the roles of CD4 and CD8 T cells and several possibilities for antigen-specific tolerance induction. Second, we outline diabetic complications necessitating beta cell replacement. Third, we discuss transplant recognition, potential sources for beta cell replacement, and tolerance-promoting therapies under development. We hypothesize that a better understanding of autoreactive T cell targets during disease pathogenesis and alloimmunity following transplant destruction could enhance attempts to re-establish tolerance to beta cells.
Collapse
Affiliation(s)
- Adam L. Burrack
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Brian T. Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- *Correspondence: Brian T. Fife,
| |
Collapse
|
21
|
Abstract
The undesired destruction of healthy cells, either endogenous or transplanted, by the immune system results in the loss of tissue function or limits strategies to restore tissue function. Current therapies typically involve nonspecific immunosuppression that may prevent the appropriate response to an antigen, thereby decreasing humoral immunity and increasing the risks of patient susceptibility to opportunistic infections, viral reactivation, and neoplasia. The induction of antigen-specific immunological tolerance to block undesired immune responses to self- or allogeneic antigens, while maintaining the integrity of the remaining immune system, has the potential to transform the current treatment of autoimmune disease and serve as a key enabling technology for therapies based on cell transplantation.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Division of Nephrology and Hypertension.,Comprehensive Cancer Center, and
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611; ,
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109;
| |
Collapse
|
22
|
Dumont CM, Park J, Shea LD. Controlled release strategies for modulating immune responses to promote tissue regeneration. J Control Release 2015; 219:155-166. [PMID: 26264833 DOI: 10.1016/j.jconrel.2015.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 01/06/2023]
Abstract
Advances in the field of tissue engineering have enhanced the potential of regenerative medicine, yet the efficacy of these strategies remains incomplete, and is limited by the innate and adaptive immune responses. The immune response associated with injury or disease combined with that mounted to biomaterials, transplanted cells, proteins, and gene therapies vectors can contribute to the inability to fully restore tissue function. Blocking immune responses such as with anti-inflammatory or immunosuppressive agents are either ineffective, as the immune response contributes significantly to regeneration, or have significant side effects. This review describes targeted strategies to modulate the immune response in order to limit tissue damage following injury, promote an anti-inflammatory environment that leads to regeneration, and induce antigen (Ag)-specific tolerance that can target degenerative diseases that destroy tissues and promote engraftment of transplanted cells. Focusing on targeted immuno-modulation, we describe local delivery techniques to sites of inflammation as well as systemic approaches that preferentially target subsets of immune populations.
Collapse
Affiliation(s)
- Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
23
|
McCarthy DP, Bryant J, Galvin JP, Miller SD, Luo X. Tempering allorecognition to induce transplant tolerance with chemically modified apoptotic donor cells. Am J Transplant 2015; 15:1475-83. [PMID: 25807873 PMCID: PMC4439351 DOI: 10.1111/ajt.13237] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/12/2015] [Accepted: 01/23/2015] [Indexed: 01/25/2023]
Abstract
The development of organ transplantation as a therapy for end-stage organ failure is among the most significant achievements of 20th century medicine, but chronic rejection remains a barrier to achieving long-term success. Current therapeutic regimens consist of immunosuppressive drugs that are efficient at delaying rejection but are associated with significant risks such as opportunistic infections, toxicity, and malignancy. Thus, the induction of specific immune tolerance to transplant antigens is the coveted aim of researchers. The use of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (ECDI)-treated, autoantigen-coupled syngeneic leukocytes has been developed as a specific immunotherapy in preclinical models of autoimmunity and is currently in a phase II clinical trial for the treatment of multiple sclerosis. In this review, we discuss the use of allogeneic ECDI-treated apoptotic donor leukocytes (allo-ECDI-SP) as a strategy for inducing antigen-specific tolerance in allogeneic transplantation. Allo-ECDI-SP therapy induces long-term systemic immune tolerance to transplant antigens by subverting alloimmune recognition and exploiting apoptotic cell uptake pathways to recapitulate innate mechanisms of peripheral tolerance. Lastly, we discuss potential indications and challenges for transitioning allo-ECDI-SP therapy into clinical practice.
Collapse
Affiliation(s)
- D. P. McCarthy
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - J. Bryant
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
| | - J. P. Galvin
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
| | - S. D. Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - X. Luo
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Evanston, IL
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Evanston, IL
| |
Collapse
|