1
|
Weijler AM, Wekerle T. Combining Treg Therapy With Donor Bone Marrow Transplantation: Experimental Progress and Clinical Perspective. Transplantation 2024; 108:1100-1108. [PMID: 37789519 DOI: 10.1097/tp.0000000000004814] [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: 10/05/2023]
Abstract
Donor-specific tolerance remains a goal in transplantation because it could improve graft survival and reduce morbidity. Cotransplantation of donor hematopoietic cells to achieve chimerism is a promising approach for tolerance induction, which was successfully tested in clinical trials. However, current protocols are associated with side effects related to the myelosuppressive recipient conditioning, which makes it difficult to introduce them as standard therapy. More recently, adoptive cell therapy with polyclonal or donor-specific regulatory T cells (Treg) proved safe and feasible in several transplant trials, but it is unclear whether it can induce tolerance on its own. The combination of both approaches-Treg therapy and hematopoietic cell transplantation-leads to chimerism and tolerance without myelosuppressive treatment in murine models. Treg therapy promotes engraftment of allogeneic hematopoietic cells, reducing conditioning requirements and enhancing regulatory mechanisms maintaining tolerance. This review discusses possible modes of action of transferred Treg in experimental chimerism models and describes translational efforts investigating the potent synergy of Treg and chimerism.
Collapse
Affiliation(s)
- Anna Marianne Weijler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
2
|
Sasaki K, Kubo M, Wang YC, Lu L, Vujevich V, Wood-Trageser MA, Golnoski K, Lesniak A, Gunabushanam V, Ganoza A, Wijkstrom MJ, Humar A, Demetris AJ, Thomson AW, Ezzelarab MB. Multiple infusions of ex vivo-expanded regulatory T cells promote CD163 + myeloid cells and kidney allograft survival in non-lymphodepleted non-human primates. Kidney Int 2024; 105:84-98. [PMID: 37839695 DOI: 10.1016/j.kint.2023.09.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 08/18/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023]
Abstract
Clinical verification of adoptively transferred regulatory T cell (Treg) efficacy in transplantation remains challenging. Here, we examined the influence of autologous ex vivo-expanded polyclonal Tregs on kidney graft survival in a clinically relevant non-human primate model. Peripheral blood Tregs were isolated and expanded using artificial antigen presenting cells. Immunosuppression was comprised of tapered tacrolimus and CTLA4 immunoglobulin, in five animals each without or with Treg infusions. Escalating Treg doses were administered 6, 10, 13, 16, 20, 23, 27 and 30 days after transplant. Infused Tregs were monitored for Treg signature, anti-apoptotic (Bcl-2) and proliferation (Ki67) marker expression. Treg infusions prolonged median graft survival time significantly from 35 to 70 days. Treg marker (Ki67 and Bcl-2) expression by infused Tregs diminished after their infusion but remained comparable to that of circulating native Tregs. No major changes in circulating donor-reactive T cell responses or total Treg percentages, or in graft-infiltrating T cell subsets were observed with Treg infusion. However, Treg infusion was associated with significant increases in CD163 expression by circulating HLA-DR+ myeloid cells and elevated levels of circulating soluble CD163. Further, graft-infiltrating CD163+ cells were increased with Treg infusion. Thus, multiple Treg infusions were associated with M2-like myeloid cell enhancement that may mediate immunomodulatory, anti-inflammatory and graft reparative effects.
Collapse
Affiliation(s)
- Kazuki Sasaki
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Masahiko Kubo
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yu-Chao Wang
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lien Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Veronica Vujevich
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michelle A Wood-Trageser
- Department of Pathology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kayla Golnoski
- Department of Pathology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrew Lesniak
- Department of Pathology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vikraman Gunabushanam
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Armando Ganoza
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Martin J Wijkstrom
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abhinav Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anthony J Demetris
- Department of Pathology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Department of Immunology, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed B Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
3
|
Sharma P, Arora A. Basic Understanding of Liver Transplant Immunology. J Clin Exp Hepatol 2023; 13:1091-1102. [PMID: 37975047 PMCID: PMC10643508 DOI: 10.1016/j.jceh.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/14/2023] [Indexed: 11/19/2023] Open
Abstract
The liver is a specialized organ and plays an important role in our immune system. The liver constitutes parenchymal cells which are hepatocytes and cholangiocytes (60-80%) and non-parenchymal cells like liver sinusoidal endothelial cells (LSECs), hepatic satellite/Ito cells, Kupffer cells, neutrophils, mononuclear cells, T and B lymphocytes (conventional and non-conventional), natural killer cells, and natural killer T (NKT) cells. The liver mounts a rapid and strong immune response, under unfavorable conditions and acts as an immune tolerance to a variety of non-pathogenic antigens. This delicate and dynamic interaction between different kinds of immune cells in the liver maintains a balance between immune screening and immune tolerance. The liver allografts are privileged immunologically; however, allograft rejection is not uncommon and is classified as cell or antibody-mediated. Advancements in transplant immunology help in the prevention of allografts rejection by immune reactions of the host thus leading to better graft and host survival. Fewer patients may not require immunosuppression due to systemic donor-specific T-cell tolerance. The liver tolerance mechanism is poorly studied, and LSEC and unconventional lymphocytes play an important role that dampens T cell response either by inducing apoptosis of cells or inhibiting co-stimulatory pathways. Newer cell-based therapy based on Treg, dendritic cells, and mesenchymal stromal cells will probably change the future of immunosuppression. Various invasive and non-invasive biomarkers and artificial intelligence have also been investigated to predict graft survival, post-transplant complications, and immunotolerance in the future.
Collapse
Affiliation(s)
- Praveen Sharma
- Department of Gastroenterology, Sir Ganga Ram Hospital, New Delhi, India
| | - Anil Arora
- Department of Gastroenterology and Hepatology, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
4
|
P. Singh R, S. Bischoff D, S Singh S, H. Hahn B. Peptide-based immunotherapy in lupus: Where are we now? RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:139-149. [PMID: 37781681 PMCID: PMC10538607 DOI: 10.2478/rir-2023-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023]
Abstract
In autoimmune rheumatic diseases, immune hyperactivity and chronic inflammation associate with immune dysregulation and the breakdown of immune self-tolerance. A continued, unresolved imbalance between effector and regulatory immune responses further exacerbates inflammation that ultimately causes tissue and organ damage. Many treatment modalities have been developed to restore the immune tolerance and immmunoregulatory balance in autoimmune rheumatic diseases, including the use of peptide-based therapeutics or the use of nanoparticles-based nanotechnology. This review summarizes the state-of-the-art therapeutic use of peptide-based therapies in autoimmune rheumatic diseases, with a specific focus on lupus.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, 90073 CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| | | | - Bevra H. Hahn
- Department of Medicine, University of California, Los Angeles, Los Angeles, 90095 CA, USA
| |
Collapse
|
5
|
Muckenhuber M, Mucha J, Mengrelis K, How C, Reindl-Schwaighofer R, Heinzel A, Kainz V, Worel N, Berlakovich G, Edinger M, Oberbauer R, Wekerle T. Optimum timing of antithymocyte globulin in relation to adoptive regulatory T cell therapy. Am J Transplant 2023; 23:84-92. [PMID: 36695625 DOI: 10.1016/j.ajt.2022.09.002] [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: 06/19/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 01/13/2023]
Abstract
Reducing the recipient's T cell repertoire is considered to increase the efficacy of regulatory T cell (Treg) therapy. This necessitates timing the administration of antithymocyte globulin (ATG) early enough before adoptive cell therapy (ACT) so that residual serum ATG does not deplete the transferred Tregs. The optimum time point in this regard has not been defined. Herein, we report the effects of residual serum ATG on the viability of an in vitro expanded Treg cell product used in a clinical trial of ACT in kidney transplant recipients (NCT03867617). Patients received ATG monotherapy (either 6 or 3 mg/kg body weight) without concomitant immunosuppression 2 to 3 weeks before transplantation and Treg transfer. An anti-ATG immunoglobulin G (IgG) immune response was elicited in all patients within 14 days. In turn, the elimination of total and Treg-specific ATG was accelerated substantially over control patients receiving the same dose of ATG with concomitant immunosuppression. However, ATG serum concentrations of <1 μg/mL, which had previously been reported as subtherapeutic threshold, triggered apoptosis of Tregs in vitro. Therefore, ATG levels need to decline to lower levels than those previously thought for efficacious Treg transfer. In 5 of 6 patients, such low levels of serum ATG considered safe for Treg transfer were reached within 2 weeks after ATG administration.
Collapse
Affiliation(s)
- Moritz Muckenhuber
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Jasmin Mucha
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Konstantinos Mengrelis
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Christopher How
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Heinzel
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Verena Kainz
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Nina Worel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Gabriela Berlakovich
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Matthias Edinger
- Leibniz Institute of Immunotherapy, Regensburg, Germany; Department of Internal Medicine 3 (Hematology and Oncology), University Hospital Regensburg, Regensburg, Germany
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Singh RP, Bischoff DS, Hahn BH. CD8 + T regulatory cells in lupus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:147-156. [PMID: 35880241 PMCID: PMC9242525 DOI: 10.2478/rir-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 04/11/2023]
Abstract
T regulatory cells (Tregs) have a key role in the maintenance of immune homeostasis and the regulation of immune tolerance by preventing the inflammation and suppressing the autoimmune responses. Numerical and functional deficits of these cells have been reported in systemic lupus erythematosus (SLE) patients and mouse models of SLE, where their imbalance and dysregulated activities have been reported to significantly influence the disease pathogenesis, progression and outcomes. Most studies in SLE have focused on CD4+ Tregs and it has become clear that a critical role in the control of immune tolerance after the breakdown of self-tolerance is provided by CD8+ Tregs. Here we review the role, cellular and molecular phenotypes, and mechanisms of action of CD8+ Tregs in SLE, including ways to induce these cells for immunotherapeutic modulation in SLE.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bevra H. Hahn
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
7
|
Ezzelarab MB, Zhang H, Sasaki K, Lu L, Zahorchak AF, van der Windt DJ, Dai H, Perez-Gutierrez A, Bhama JK, Thomson AW. Ex Vivo Expanded Donor Alloreactive Regulatory T Cells Lose Immunoregulatory, Proliferation, and Antiapoptotic Markers After Infusion Into ATG-lymphodepleted, Nonhuman Primate Heart Allograft Recipients. Transplantation 2021; 105:1965-1979. [PMID: 33587433 PMCID: PMC8239063 DOI: 10.1097/tp.0000000000003617] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Regulatory T cell (Treg) therapy is a promising approach to amelioration of allograft rejection and promotion of organ transplant tolerance. However, the fate of infused Treg, and how this relates to their therapeutic efficacy using different immunosuppressive regimens is poorly understood. Our aim was to analyze the tissue distribution, persistence, replicative activity and phenotypic stability of autologous, donor antigen alloreactive Treg (darTreg) in anti-thymocyte globulin (ATG)-lymphodepleted, heart-allografted cynomolgus monkeys. METHODS darTreg were expanded ex vivo from flow-sorted, circulating Treg using activated donor B cells and infused posttransplant into recipients of major histocompatibility complex-mismatched heart allografts. Fluorochrome-labeled darTreg were identified and characterized in peripheral blood, lymphoid, and nonlymphoid tissues and the graft by flow cytometric analysis. RESULTS darTreg selectively suppressed autologous T cell responses to donor antigens in vitro. However, following their adoptive transfer after transplantation, graft survival was not prolonged. Early (within 2 wk posttransplant; under ATG, tacrolimus, and anti-IL-6R) or delayed (6-8 wk posttransplant; under rapamycin) darTreg infusion resulted in a rapid decline in transferred darTreg in peripheral blood. Following their early or delayed infusion, labeled cells were evident in lymphoid and nonlymphoid organs and the graft at low percentages (<4% CD4+ T cells). Notably, infused darTreg showed reduced expression of immunoregulatory molecules (Foxp3 and CTLA4), Helios, the proliferative marker Ki67 and antiapoptotic Bcl2, compared with preinfusion darTreg and endogenous CD4+CD25hi Treg. CONCLUSIONS Lack of therapeutic efficacy of infused darTreg in lymphodepleted heart graft recipients appears to reflect loss of a regulatory signature and proliferative and survival capacity shortly after infusion.
Collapse
Affiliation(s)
- Mohamed B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hong Zhang
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kazuki Sasaki
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lien Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alan F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dirk J. van der Windt
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Helong Dai
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Angelica Perez-Gutierrez
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jay K. Bhama
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Angus W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Macedo C, Tran LM, Zahorchak AF, Dai H, Gu X, Ravichandran R, Mohanakumar T, Elinoff B, Zeevi A, Styn MA, Humar A, Lakkis FG, Metes DM, Thomson AW. Donor-derived regulatory dendritic cell infusion results in host cell cross-dressing and T cell subset changes in prospective living donor liver transplant recipients. Am J Transplant 2021; 21:2372-2386. [PMID: 33171019 PMCID: PMC8215622 DOI: 10.1111/ajt.16393] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/13/2020] [Accepted: 11/01/2020] [Indexed: 01/25/2023]
Abstract
Regulatory dendritic cells (DCreg) promote transplant tolerance following their adoptive transfer in experimental animals. We investigated the feasibility, safety, fate, and impact on host T cells of donor monocyte-derived DCreg infused into prospective, living donor liver transplant patients, 7 days before transplantation. The DCreg expressed a tolerogenic gene transcriptional profile, high cell surface programed death ligand-1 (PD-L1):CD86 ratios, high IL-10/no IL-12 productivity and poor ability to stimulate allogeneic T cell proliferation. Target DCreg doses (range 2.5-10 × 106 cells/kg) were achieved in all but 1 of 15 recipients, with no infusion reactions. Following DCreg infusion, transiently elevated levels of donor HLA and immunoregulatory PD-L1, CD39, and CD73 were detected in circulating small extracellular vesicles. At the same time, flow and advanced image stream analysis revealed intact DCreg and "cross-dressing" of host DCs in blood and lymph nodes. PD-L1 co-localization with donor HLA was observed at higher levels than with recipient HLA. Between DCreg infusion and transplantation, T-bethi Eomeshi memory CD8+ T cells decreased, whereas regulatory (CD25hi CD127- Foxp3+ ): T-bethi Eomeshi CD8+ T cell ratios increased. Thus, donor-derived DCreg infusion may induce systemic changes in host antigen-presenting cells and T cells potentially conducive to modulated anti-donor immune reactivity at the time of transplant.
Collapse
Affiliation(s)
- Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lillian M. Tran
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alan F. Zahorchak
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Helong Dai
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xinyan Gu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | - Beth Elinoff
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adriana Zeevi
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Mindi A. Styn
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Abhinav Humar
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Fadi G. Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Diana M. Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Mansourabadi AH, Mohamed Khosroshahi L, Noorbakhsh F, Amirzargar A. Cell therapy in transplantation: A comprehensive review of the current applications of cell therapy in transplant patients with the focus on Tregs, CAR Tregs, and Mesenchymal stem cells. Int Immunopharmacol 2021; 97:107669. [PMID: 33965760 DOI: 10.1016/j.intimp.2021.107669] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Organ transplantation is a practical treatment for patients with end-stage organ failure. Despite the advances in short-term graft survival, long-term graft survival remains the main challenge considering the increased mortality and morbidity associated with chronic rejection and the toxicity of immunosuppressive drugs. Since a novel therapeutic strategy to induce allograft tolerance seems urgent, focusing on developing novel and safe approaches to prolong graft survival is one of the main goals of transplant investigators. Researchers in the field of organ transplantation are interested in suppressing or optimizing the immune responses by focusing on immune cells including mesenchymal stem cells (MSCs), polyclonal regulatory Tcells (Tregs), and antigen-specific Tregs engineered with chimeric antigen receptors (CAR Tregs). We review the mechanistic pathways, phenotypic and functional characteristics of these cells, and their promising application in organ transplantation.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran; Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran
| | - Leila Mohamed Khosroshahi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| |
Collapse
|
10
|
Verma D, Chan ED, Ordway DJ. The double-edged sword of Tregs in M tuberculosis, M avium, and M absessus infection. Immunol Rev 2021; 301:48-61. [PMID: 33713043 DOI: 10.1111/imr.12959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Immunity against different Mycobacteria species targeting the lung requires distinctly different pulmonary immune responses for bacterial clearance. Many parameters of acquired and regulatory immune responses differ quantitatively and qualitatively from immunity during infection with Mycobacteria species. Nontuberculosis Mycobacteria species (NTM) Mycobacterium avium- (M avium), Mycobacterium abscessus-(M abscessus), and the Mycobacteria species Mycobacterium tuberculosis-(Mtb). Herein, we discuss the potential implications of acquired and regulatory immune responses in the context of animal and human studies, as well as future directions for efforts to treat Mycobacteria diseases.
Collapse
Affiliation(s)
- Deepshikha Verma
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Edward D Chan
- Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Denver, CO, USA.,Departments of Medicine and Academic Affairs, National Jewish Health, Denver, CO, USA.,Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Diane J Ordway
- Mycobacteria Research Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
11
|
López-Abente J, Martínez-Bonet M, Bernaldo-de-Quirós E, Camino M, Gil N, Panadero E, Gil-Jaurena JM, Clemente M, Urschel S, West L, Pion M, Correa-Rocha R. Basiliximab impairs regulatory T cell (TREG) function and could affect the short-term graft acceptance in children with heart transplantation. Sci Rep 2021; 11:827. [PMID: 33436905 PMCID: PMC7803770 DOI: 10.1038/s41598-020-80567-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
CD25, the alpha chain of the IL-2 receptor, is expressed on activated effector T cells that mediate immune graft damage. Induction immunosuppression is commonly used in solid organ transplantation and can include antibodies blocking CD25. However, regulatory T cells (Tregs) also rely on CD25 for their proliferation, survival, and regulatory function. Therefore, CD25-blockade may compromise Treg protective role against rejection. We analysed in vitro the effect of basiliximab (BXM) on the viability, phenotype, proliferation and cytokine production of Treg cells. We also evaluated in vivo the effect of BXM on Treg in thymectomized heart transplant children receiving BXM in comparison to patients not receiving induction therapy. Our results show that BXM reduces Treg counts and function in vitro by affecting their proliferation, Foxp3 expression, and IL-10 secretion capacity. In pediatric heart-transplant patients, we observed decreased Treg counts and a diminished Treg/Teff ratio in BXM-treated patients up to 6-month after treatment, recovering baseline values at the end of the 12-month follow up period. These results reveal that the use of BXM could produce detrimental effects on Tregs, and support the evidence suggesting that BXM induction could impair the protective role of Tregs in the period of highest incidence of acute graft rejection.
Collapse
Affiliation(s)
- Jacobo López-Abente
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Manuela Camino
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Nuria Gil
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Esther Panadero
- Pediatric-Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Juan Miguel Gil-Jaurena
- Pediatric Cardiac Surgery Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Maribel Clemente
- Cell Culture Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Simon Urschel
- Pediatric Cardiac Transplantation, University of Alberta/Stollery Children's Hospital, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada
| | - Lori West
- Pediatric Cardiac Transplantation, University of Alberta/Stollery Children's Hospital, Edmonton, AB, Canada.,Alberta Transplant Institute, University of Alberta, Edmonton, AB, Canada.,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Pabellón de Medicina Experimental, Planta Baja. C/ Maiquez, 6., 28006, Madrid, Spain. .,Canadian National Transplant Research Program Investigator, CNTRP, Edmonton, AB, Canada.
| |
Collapse
|
12
|
Gezinir E, Podlech J, Gergely KM, Becker S, Reddehase MJ, Lemmermann NAW. Enhancement of Antigen Presentation by Deletion of Viral Immune Evasion Genes Prevents Lethal Cytomegalovirus Disease in Minor Histocompatibility Antigen-Mismatched Hematopoietic Cell Transplantation. Front Cell Infect Microbiol 2020; 10:279. [PMID: 32582572 PMCID: PMC7296086 DOI: 10.3389/fcimb.2020.00279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/12/2020] [Indexed: 12/17/2022] Open
Abstract
Hematoablative treatment followed by hematopoietic cell transplantation (HCT) for reconstituting the co-ablated immune system is a therapeutic option to cure aggressive forms of hematopoietic malignancies. In cases of family donors or unrelated donors, immunogenetic mismatches in major histocompatibility complex (MHC) and/or minor histocompatibility (minor-H) loci are unavoidable and bear a risk of graft-vs.-host reaction and disease (GvHR/D). Transient immunodeficiency inherent to the HCT protocol favors a productive reactivation of latent cytomegalovirus (CMV) that can result in multiple-organ CMV disease. In addition, there exists evidence from a mouse model of MHC class-I-mismatched GvH-HCT to propose that mismatches interfere with an efficient reconstitution of antiviral immunity. Here we used a mouse model of MHC-matched HCT with C57BL/6 donors and MHC-congenic BALB.B recipients that only differ in polymorphic autosomal background genes, including minor-H loci coding for minor-H antigens (minor-HAg). Minor-HAg mismatch is found to promote lethal CMV disease in absence of a detectable GvH response to an immunodominant minor-HAg, the H60 locus-encoded antigenic peptide LYL8. Lethality of infection correlates with inefficient reconstitution of viral epitope-specific CD8+ T cells. Notably, lethality is prevented and control of cytopathogenic infection is restored when viral antigen presentation is enhanced by deletion of immune evasion genes from the infecting virus. We hypothesize that any kind of mismatch in GvH-HCT can induce "non-cognate transplantation tolerance" that dampens not only a mismatch-specific GvH response, which is beneficial, but adversely affects also responses to mismatch-unrelated antigens, such as CMV antigens in the specific case, with the consequence of lethal CMV disease.
Collapse
Affiliation(s)
- Emin Gezinir
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Kerstin M Gergely
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Sara Becker
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Matthias J Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Niels A W Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI) at the University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| |
Collapse
|
13
|
Barabadi M, Shahbaz SK, Foroughi F, Hosseinzadeh M, Nafar M, Yekaninejad MS, Amirzargar A. High Expression of FOXP3 mRNA in Blood and Urine as a Predictive Marker in Kidney Transplantation. Prog Transplant 2019; 28:134-141. [PMID: 29798728 DOI: 10.1177/1526924818765812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diagnosis of allograft dysfunction by noninvasive biomarker tests is preferable to invasive allograft biopsies and has been extensively considered in recent years. This study aims to evaluate blood and urinary forkhead box P3 (FOXP3) messenger RNA (mRNA) expression in renal transplant recipients in an attempt to determine whether differential diagnosis of graft dysfunction is feasible using mRNA profiles. METHODS We analyzed FOXP3 mRNA expression in paired urinary and peripheral blood mononuclear cell (PBMC) samples. A total of 91 kidney transplant recipients enrolled in this study that were classified into 3 groups: biopsy-proven acute rejection (AR; n = 27), chronic allograft nephropathy (n = 19), and well-functioning graft (n = 45). The FOXP3 mRNA expression was quantified by TaqMan probe real-time polymerase chain reaction. RESULTS Acute rejection patients had a higher expression level of transcription factor FOXP3 compared to the chronic nephropathy and control groups. Analysis of receiver operating characteristic curves showed that rejection could be diagnosed with 100% sensitivity and 96% specificity in urine, and 92% sensitivity and 86% specificity in PBMC samples using the optimal FOXP3 mRNA cutoff value. We subdivided the AR group into progressive and nonprogressive patients, which showed a significant difference in FOXP3 mRNA expression. This result confirmed the role of FOXP3 as a diagnostic marker in predicting transplantation outcomes. CONCLUSION Our results suggested that elevated expression of FOXP3 in blood and urine samples from kidney transplant recipients could be a useful noninvasive biomarker to diagnose graft dysfunction.
Collapse
Affiliation(s)
- Mehri Barabadi
- 1 Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanaz Keshavarz Shahbaz
- 1 Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Foroughi
- 2 Department of Immunology, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Morteza Hosseinzadeh
- 3 Department of Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohsen Nafar
- 4 Department of Nephrology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Saeed Yekaninejad
- 5 Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Amirzargar
- 1 Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,6 Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Yang F, Zhang Y, Liu B, Cao M, Yang J, Tian F, Yang P, Qin K, Zhao D. Basic fibroblast growth factor and agarose gel promote the ability of immune privilege of allogeneic cartilage transplantation in rats. J Orthop Translat 2019; 22:73-80. [PMID: 32440502 PMCID: PMC7231919 DOI: 10.1016/j.jot.2019.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 01/16/2023] Open
Abstract
Objective Allogeneic cartilage transplantation is used to treat severe osteochondral defects or cartilaginous injury. However, acute immune rejection has been a key problem interfering with graft healing. Methods Full-thickness osteochondral defects were performed in Sprague Dawley rats. The allograft implants were set into the defect region. Blood and spleen samples from Postoperative Day 3 onward were collected for inflammatory cell analysis, including analysis of monocytes, natural killer cells, CD4+CD25+Foxp3+ regulatory T cells, CD4+ T cells, and CD8+ T cells. Gross observation and histologic staining (hematoxylin and eosin, toluidine blue) were carried out at the same time point to assess the repair effect of the cartilage graft and the degree of immune rejection. Results Treatment with basic fibroblast growth factor, agarose gel, and allogeneic cartilage was similar to that of the autologous group. The percentage of monocytes in allografts was at a higher level in the spleen and blood; the frequency of CD4+ T cells in the allogeneic group was higher than in the autologous group and the other agarose groups at 6 weeks after transplantation. The number of regulatory T cells in the autograft was increased from Postoperative Week 1; similar results were observed in groups containing basic fibroblast growth factor beginning at Postoperative Week 3. Conclusions Allogeneic cartilage transplantation induces acute immune rejection, which compromises the validity of the implant. The combination of basic fibroblast growth factor and agarose gel facilitates the goal of immune privilege and promotes the success of the allograft tissues. The translational potential of this article This study investigated the combination of basic fibroblast growth factor (bFGF) and agarose gel facilitates promotes the success of the allograft tissues transplantation. This work may help clinicians find a new way to repair articular cartilage damage. This will affect the treatment of articular cartilage movement injuries and arthritis.
Collapse
Affiliation(s)
- Fan Yang
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Linggong Road, Dalian, Liaoning, China.,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Yu Zhang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Baoyi Liu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China.,Postdoctoral Workstation, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Meng Cao
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Jiahui Yang
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Fengde Tian
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Linggong Road, Dalian, Liaoning, China.,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| | - Pei Yang
- The First Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, 710000, China
| | - Kairong Qin
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Linggong Road, Dalian, Liaoning, China
| | - Dewei Zhao
- Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Linggong Road, Dalian, Liaoning, China.,Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, No 6 Jiefang Street, Dalian, Liaoning, China
| |
Collapse
|
15
|
See Hoe LE, Bartnikowski N, Wells MA, Suen JY, Fraser JF. Hurdles to Cardioprotection in the Critically Ill. Int J Mol Sci 2019; 20:E3823. [PMID: 31387264 PMCID: PMC6695809 DOI: 10.3390/ijms20153823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/26/2019] [Accepted: 08/03/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the largest contributor to worldwide mortality, and the deleterious impact of heart failure (HF) is projected to grow exponentially in the future. As heart transplantation (HTx) is the only effective treatment for end-stage HF, development of mechanical circulatory support (MCS) technology has unveiled additional therapeutic options for refractory cardiac disease. Unfortunately, despite both MCS and HTx being quintessential treatments for significant cardiac impairment, associated morbidity and mortality remain high. MCS technology continues to evolve, but is associated with numerous disturbances to cardiac function (e.g., oxidative damage, arrhythmias). Following MCS intervention, HTx is frequently the destination option for survival of critically ill cardiac patients. While effective, donor hearts are scarce, thus limiting HTx to few qualifying patients, and HTx remains correlated with substantial post-HTx complications. While MCS and HTx are vital to survival of critically ill cardiac patients, cardioprotective strategies to improve outcomes from these treatments are highly desirable. Accordingly, this review summarizes the current status of MCS and HTx in the clinic, and the associated cardiac complications inherent to these treatments. Furthermore, we detail current research being undertaken to improve cardiac outcomes following MCS/HTx, and important considerations for reducing the significant morbidity and mortality associated with these necessary treatment strategies.
Collapse
Affiliation(s)
- Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia.
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia.
| | - Nicole Bartnikowski
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Science and Engineering Faculty, Queensland University of Technology, Chermside 4032, Australia
| | - Matthew A Wells
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- School of Medical Science, Griffith University, Southport 4222, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Chermside 4032, Australia
- Faculty of Medicine, University of Queensland, Chermside 4032, Australia
| |
Collapse
|
16
|
Mortazavi H, Soltani‐Zangbar MS, Eghbal‐Fard S, Mehdizadeh A, Kamrani A, Chakeri‐Khiavi F, Kafil HS, Jadidi‐Niaragh F, Rahimifar S, Khosroshahi HT, Yousefi M. Cytokine profile, Treg/Th17 cell frequency changes during different posttransplantational time points in patients undergoing renal transplantation. J Cell Physiol 2019; 234:20935-20943. [DOI: 10.1002/jcp.28698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Hamid Mortazavi
- Research Center for Chronic Kidney Disease Tabriz University of Medical Sciences Tabriz Iran
| | - Mohammad Sadegh Soltani‐Zangbar
- Student committee Research Center Tabriz University of Medical Sciences Tabriz Iran
- Stem Cells Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Shadi Eghbal‐Fard
- Department of Immunology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | - Amir Mehdizadeh
- Endocrine Research Center Tabriz University of Medical Sciences Tabriz Iran
| | - Amin Kamrani
- Stem Cells Research Center Tabriz University of Medical Sciences Tabriz Iran
| | | | | | - Farhad Jadidi‐Niaragh
- Department of Immunology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| | | | | | - Mehdi Yousefi
- Stem Cells Research Center Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology, Faculty of Medicine Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
17
|
Jofra T, Di Fonte R, Galvani G, Kuka M, Iannacone M, Battaglia M, Fousteri G. Tr1 cell immunotherapy promotes transplant tolerance via de novo Tr1 cell induction in mice and is safe and effective during acute viral infection. Eur J Immunol 2018; 48:1389-1399. [PMID: 29684247 DOI: 10.1002/eji.201747316] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/07/2018] [Accepted: 04/18/2018] [Indexed: 01/16/2023]
Abstract
Tr1 cell therapy is considered an emerging approach to improve transplant tolerance and enhance allogeneic graft survival. However, it remains unclear how Tr1 cells promote transplant tolerance and whether they will be safe and stable in the face of an acute viral infection. By employing a mouse model of pancreatic islet transplantation, we report that Tr1 cell therapy promoted transplant tolerance via de novo induction of Tr1 cells in the recipients. Acute viral infection with lymphocytic choriomeningitis virus (LCMV) had no impact on Tr1 cell number and function, neither on the Tr1 cells infused nor on the ones induced, and that was reflected in the robust maintenance of the graft. Moreover, Tr1 cell immunotherapy had no detrimental effect on CD8 and CD4 anti-LCMV effector T-cell responses and viral control. Together, these data suggest that Tr1 cells did not convert to effector cells during acute infection with LCMV, maintained transplant tolerance and did not inhibit antiviral immunity.
Collapse
Affiliation(s)
- Tatiana Jofra
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Di Fonte
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Galvani
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mirela Kuka
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University
| | - Matteo Iannacone
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Manuela Battaglia
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Georgia Fousteri
- Division of Immunology Transplantation and Infectious Diseases (DITID), Diabetes Research Institute (DRI) IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
18
|
Hua J, Inomata T, Chen Y, Foulsham W, Stevenson W, Shiang T, Bluestone JA, Dana R. Pathological conversion of regulatory T cells is associated with loss of allotolerance. Sci Rep 2018; 8:7059. [PMID: 29728574 PMCID: PMC5935752 DOI: 10.1038/s41598-018-25384-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/11/2018] [Indexed: 01/26/2023] Open
Abstract
CD4+CD25+Foxp3+ Regulatory T cells (Tregs) play a critical role in immune tolerance. The plasticity and functional adaptability of Tregs in an inflammatory microenvironment has been demonstrated in autoimmunity. Here, using a double transgenic mouse model that permits Foxp3 lineage tracing, we investigated the phenotypic plasticity of Foxp3+ Tregs in a well-characterized murine model of corneal transplantation. In order to subvert the normal immune privilege of the cornea and foster an inflammatory milieu, host mice were exposed to desiccating stress prior to transplantation. Treg frequencies and function were decreased following desiccating stress, and this corresponded to decreased graft survival. A fraction of Tregs converted to IL-17+ or IFNγ+ 'exFoxp3' T cells that were phenotypically indistinguishable from effector Th17 or Th1 cells, respectively. We investigated how Foxp3 expression is modulated in different Treg subsets, demonstrating that neuropilin-1- peripherally-derived Tregs are particularly susceptible to conversion to IL-17+/IFNγ+ exFoxp3 cells in response to cues from their microenvironment. Finally, we show that IL-6 and IL-23 are implicated in the conversion of Tregs to exFoxp3 cells. This report demonstrates that the pathological conversion of Tregs contributes to the loss of corneal immune privilege.
Collapse
Affiliation(s)
- Jing Hua
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Takenori Inomata
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - William Stevenson
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tina Shiang
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Bal SH, Heper Y, Kumaş LT, Guvenc F, Budak F, Göral G, Oral HB. Effect of storage period of red blood cell suspensions on helper T-cell subpopulations. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2018; 16:262-272. [PMID: 28488961 PMCID: PMC5919838 DOI: 10.2450/2017.0238-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND The aim of this study was to investigate the immunological alterations that occur during the storage of erythrocyte suspensions which may lead to transfusion-related immunomodulation following allogeneic blood transfusion. MATERIALS AND METHODS One part of the erythrocyte suspensions obtained from donors was leucoreduced while the other part was not. The leucoreduced (LR) and non-leucoreduced (NL) erythrocyte suspensions were then further divided into three equal amounts which were stored for 0, 21 or 42 days prior to measurements, by enzyme-linked immunosorbent assays, of cytokine levels in their supernatants. T-helper (Th) lymphocyte subgroups and gene expression were analysed in the NL erythrocyte suspensions by flow cytometry and real-time polymerase chain reaction, respectively. Results were compared to those of storage day 0. RESULTS By day 21, the number of Th2 cells had increased significantly and the numbers of Th1, Th22 and Treg cells had decreased significantly in the NL erythrocyte suspensions. On day 42 the numbers of Th2 and Treg cells in the NL suspensions were significantly increased while the number of Th1 cells was significantly decreased. The levels of transcription factors (TBX21, GATA3, and SPI.1) were significantly decreased on days 21 and 42, and AHR, FOXP3 and RORC2 levels were significantly increased on day 42 in NL erythrocyte suspensions. The decrease in interleukin-22 and increase in transforming growth factor-β levels found in NL erythrocyte suspensions on day 21 were statistically significant. Elevated levels of interleukin-17A were found in both LR and NL erythrocyte suspensions on day 42. DISCUSSION Our results suggest that allogeneic leucocytes and cytokines may play significant roles in the development of transfusion-related immunomodulation.
Collapse
Affiliation(s)
- Salih H Bal
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Microbiology/Immunology, Institute of Health Sciences, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Yasemin Heper
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Levent T Kumaş
- "Dr. Rasit Durusoy" Blood Bank, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
- Department of Microbiology/Immunology, Institute of Health Sciences, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Furkan Guvenc
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Ferah Budak
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Güher Göral
- Department of Medical Microbiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Haluk B Oral
- Department of Immunology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
20
|
The ratio of circulating regulatory cluster of differentiation 4 T cells to endothelial progenitor cells predicts clinically significant acute rejection after heart transplantation. J Heart Lung Transplant 2018; 37:496-502. [DOI: 10.1016/j.healun.2017.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/27/2017] [Accepted: 10/18/2017] [Indexed: 11/23/2022] Open
|
21
|
Liu XG, Liu Y, Chen F. Soluble fibrinogen like protein 2 (sFGL2), the novel effector molecule for immunoregulation. Oncotarget 2018; 8:3711-3723. [PMID: 27732962 PMCID: PMC5356913 DOI: 10.18632/oncotarget.12533] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023] Open
Abstract
Soluble fibrinogen-like protein 2 (sFGL2) is the soluble form of fibrinogen-like protein 2 belonging to the fibrinogen-related protein superfamily. It is now well characterized that sFGL2 is mainly secreted by regulatory T cell (Treg) populations, and exerts potently immunosuppressive activities. By repressing not only the differentiation and proliferation of T cells but also the maturation of dendritic cells (DCs), sFGL2 acts largely as an immunosuppressant. Moreover, sFGL2 also induces apoptosis of B cells, tubular epithelial cells (TECs), sinusoidal endothelial cells (SECs), and hepatocytes. This mini-review focuses primarily on the recent literature with respect to the signaling mechanism of sFGL2 in immunomodulation, and discusses the clinical implications of sFGL2 in transplantation, hepatitis, autoimmunity, and tumors.
Collapse
Affiliation(s)
- Xin-Guang Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P. R. China
| | - Yu Liu
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan, P. R. China
| | - Feng Chen
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P. R. China.,Capital Medical University Cancer Center, Beijing Shijitan Hospital, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| |
Collapse
|
22
|
Chandran S, Tang Q, Sarwal M, Laszik ZG, Putnam AL, Lee K, Leung J, Nguyen V, Sigdel T, Tavares EC, Yang JY, Hellerstein M, Fitch M, Bluestone JA, Vincenti F. Polyclonal Regulatory T Cell Therapy for Control of Inflammation in Kidney Transplants. Am J Transplant 2017; 17:2945-2954. [PMID: 28675676 PMCID: PMC5662482 DOI: 10.1111/ajt.14415] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 01/25/2023]
Abstract
Early subclinical inflammation in kidney transplants is associated with later graft fibrosis and dysfunction. Regulatory T cells (Tregs) can reverse established inflammation in animal models. We conducted a pilot safety and feasibility trial of autologous Treg cell therapy in three kidney transplant recipients with subclinical inflammation noted on 6-month surveillance biopsies. Tregs were purified from peripheral blood and polyclonally expanded ex vivo using medium containing deuterated glucose to label the cells. All patients received a single infusion of ~320 × 106 (319, 321, and 363.8 × 106 ) expanded Tregs. Persistence of the infused Tregs was tracked. Graft inflammation was monitored with follow-up biopsies and urinary biomarkers. Nearly 1 × 109 (0.932, 0.956, 1.565 × 109 ) Tregs were successfully manufactured for each patient. There were no infusion reactions or serious therapy-related adverse events. The infused cells demonstrated patterns of persistence and stability similar to those observed in non-immunosuppressed subjects receiving the same dose of Tregs. Isolation and expansion of Tregs is feasible in kidney transplant patients on immunosuppression. Infusion of these cells was safe and well tolerated. Future trials will test the efficacy of polyclonal and donor alloantigen-reactive Tregs for the treatment of inflammation in kidney transplants.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, CA 94143, USA,Diabetes Center, University of California, San Francisco, CA 94143, USA
| | - Minnie Sarwal
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Zoltan G. Laszik
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Amy L. Putnam
- Diabetes Center, University of California, San Francisco, CA 94143, USA
| | - Karim Lee
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Joey Leung
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Tara Sigdel
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Erica C. Tavares
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Joshua Y.C. Yang
- Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Marc Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Mark Fitch
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | | | - Flavio Vincenti
- Department of Medicine, University of California, San Francisco, CA 94143, USA,Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
23
|
Hall BM, Robinson CM, Plain KM, Verma ND, Tran GT, Nomura M, Carter N, Boyd R, Hodgkinson SJ. Changes in Reactivity In Vitro of CD4 +CD25 + and CD4 +CD25 - T Cell Subsets in Transplant Tolerance. Front Immunol 2017; 8:994. [PMID: 28878770 PMCID: PMC5572370 DOI: 10.3389/fimmu.2017.00994] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 08/03/2017] [Indexed: 01/03/2023] Open
Abstract
Transplant tolerance induced in adult animals is mediated by alloantigen-specific CD4+CD25+ T cells, yet in many models, proliferation of CD4+ T cells from hosts tolerant to specific-alloantigen in vitro is not impaired. To identify changes that may diagnose tolerance, changes in the patterns of proliferation of CD4+, CD4+CD25+, and CD4+CD25− T cells from DA rats tolerant to Piebald Virol Glaxo rat strain (PVG) cardiac allografts and from naïve DA rats were examined. Proliferation of CD4+ T cells from both naïve and tolerant hosts was similar to both PVG and Lewis stimulator cells. In mixed lymphocyte culture to PVG, proliferation of naïve CD4+CD25− T cells was greater than naïve CD4+ T cells. In contrast, proliferation of CD4+CD25− T cells from tolerant hosts to specific-donor PVG was not greater than CD4+ T cells, whereas their response to Lewis and self-DA was greater than CD4+ T cells. Paradoxically, CD4+CD25+ T cells from tolerant hosts did not proliferate to PVG, but did to Lewis, whereas naïve CD4+CD25+ T cells proliferate to both PVG and Lewis but not to self-DA. CD4+CD25+ T cells from tolerant, but not naïve hosts, expressed receptors for interferon (IFN)-γ and IL-5 and these cytokines promoted their proliferation to specific-alloantigen PVG but not to Lewis or self-DA. We identified several differences in the patterns of proliferation to specific-donor alloantigen between cells from tolerant and naïve hosts. Most relevant is that CD4+CD25+ T cells from tolerant hosts failed to proliferate or suppress to specific donor in the absence of either IFN-γ or IL-5. The proliferation to third-party and self of each cell population from tolerant and naïve hosts was similar and not affected by IFN-γ or IL-5. Our findings suggest CD4+CD25+ T cells that mediate transplant tolerance depend on IFN−γ or IL-5 from alloactivated Th1 and Th2 cells.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Catherine M Robinson
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Karren M Plain
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia.,Faculty of Veterinary Sciences, University of Sydney, Cobbity, NSW, Australia
| | - Nirupama D Verma
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Giang T Tran
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| | - Masaru Nomura
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia.,Department of Surgery, Nakashibetsu Hospital Shibetu-gun Nakashibetsu-cho, Hokkaido, Japan
| | - Nicole Carter
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia.,Faculty of Veterinary Sciences, University of Sydney, Cobbity, NSW, Australia
| | - Rochelle Boyd
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia.,Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Suzanne J Hodgkinson
- Immune Tolerance Laboratory, Department of Medicine, Ingham Institute, University of New South Wales, Liverpool Hospital, Liverpool, NSW, Australia
| |
Collapse
|
24
|
Kang N, Toyofuku WM, Yang X, Scott MD. Inhibition of allogeneic cytotoxic T cell (CD8 +) proliferation via polymer-induced Treg (CD4 +) cells. Acta Biomater 2017; 57:146-155. [PMID: 28442414 DOI: 10.1016/j.actbio.2017.04.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 01/16/2023]
Abstract
T cell-mediated immune rejection remains a barrier to successful transplantation. Polymer-based bioengineering of cells may provide an effective means of preventing allorecognition and the proliferation of cytotoxic (CD8+) T lymphocytes (CTL). Using MHC-disparate murine splenocytes modified with succinimidyl valerate activated methoxypoly(ethylene glycol) [SVA-mPEG] polymers, the effects of leukocyte immunocamouflage on CD8+ and CD4+ alloproliferation and T regulatory (Treg) cell induction were assessed in a mixed lymphocyte reaction (MLR) model. Polymer-grafting effectively camouflaged multiple leukocyte markers (MHC class I and II, TCR and CD3) essential for effective allorecognition. Consequent to the polymer-induced immunocamouflage of the cell membrane, both CD8+ and CD4+ T cell alloproliferation were significantly inhibited in a polymer dose-dependent manner. The loss of alloproliferation correlated with the induction of Treg cells (CD4+CD25+Foxp3+). The Tregs, surprisingly, arose primarily via differentiation of naive, non-proliferating, CD4+ cells. Of biologic importance, the polymer-induced Treg were functional and exhibited potent immunosuppressive activity on allogeneic CTL proliferation. These results suggest that immunocamouflage-mediated attenuation of alloantigen-TCR recognition can prevent the tissue destructive allogeneic CD8+ T cell response, both directly and indirectly, through the generation/differentiation of functional Tregs. Immunocamouflage induced tolerance could be clinically valuable in attenuating T cell-mediated transplant rejection and in the treatment of autoimmune diseases. STATEMENT OF SIGNIFICANCE While our previous studies have demonstrated that polymer-grafting to MHC disparate leukocytes inhibits CD4+ cell proliferation, the effects of PEGylation on the alloproliferation of CD8+ cytotoxic T cells (CTL) was not examined. As shown here, PEGylation of allogeneic leukocytes prevents the generation of the CTL response responsible for acute rejection. The loss of CTL proliferation is consequent to the polymer-based attenuation of allorecognition and the induction of T regulatory cells (Tregs). Interestingly, the Tregs are primarily generated via the differentiation of non-proliferating naive T cells. Importantly, the Tregs are functional and effectively induce a tolerogenic environment when transferred to an alloresponsive environment. The use of polymer-modified leukocytes provides a unique approach to effectively maximize the biologic production of functional Tregs both in vitro and in vivo. By using this approach it may be possible to attenuate unwanted alloresponses (e.g., graft rejection) or to treat autoimmune diseases.
Collapse
Affiliation(s)
- Ning Kang
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Wendy M Toyofuku
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Xining Yang
- University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Mark D Scott
- Canadian Blood Services, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; University of British Columbia Centre for Blood Research, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pathology and Laboratory Medicine, Life Sciences Centre, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
25
|
Abstract
Immunity against Mycobacterium tuberculosis requires a balance between adaptive immune responses to constrain bacterial replication and the prevention of potentially damaging immune activation. Regulatory T (Treg) cells express the transcription factor Foxp3+ and constitute an essential counterbalance of inflammatory Th1 responses and are required to maintain immune homeostasis. The first reports describing the presence of Foxp3-expressing CD4+ Treg cells in tuberculosis (TB) emerged in 2006. Different Treg cell subsets, most likely specialized for different tissues and microenvironments, have been shown to expand in both human TB and animal models of TB. Recently, additional functional roles for Treg cells have been demonstrated during different stages and spectrums of TB disease. Foxp3+ regulatory cells can quickly expand during early infection and impede the onset of cellular immunity and persist during chronic TB infection. Increased frequencies of Treg cells have been associated with a detrimental outcome of active TB, and may be dependent on the M. tuberculosis strain, animal model, local environment, and the stage of infection. Some investigations also suggest that Treg cells are required together with effector T cell responses to obtain reduced pathology and sterilizing immunity. In this review, we will first provide an overview of the regulatory cells and mechanisms that control immune homeostasis. Then, we will review what is known about the phenotype and function of Treg cells from studies in human TB and experimental animal models of TB. We will discuss the potential role of Treg cells in the progression of TB disease and the relevance of this knowledge for future efforts to prevent, modulate, and treat TB.
Collapse
|
26
|
Nelsen MK, Beard KS, Plenter RJ, Kedl RM, Clambey ET, Gill RG. Disruption of Transplant Tolerance by an "Incognito" Form of CD8 T Cell-Dependent Memory. Am J Transplant 2017; 17:1742-1753. [PMID: 28066981 PMCID: PMC5489385 DOI: 10.1111/ajt.14194] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/27/2016] [Accepted: 12/29/2016] [Indexed: 01/25/2023]
Abstract
Several approaches successfully achieve allograft tolerance in preclinical models but are challenging to translate into clinical practice. Many clinically relevant factors can attenuate allograft tolerance induction, including intrinsic genetic resistance, peritransplant infection, inflammation, and preexisting antidonor immunity. The prevailing view for immune memory as a tolerance barrier is that the host harbors memory cells that spontaneously cross-react to donor MHC antigens. Such preexisting "heterologous" memory cells have direct reactivity to donor cells and resist most tolerance regimens. In this study, we developed a model system to determine if an alternative form of immune memory could also block tolerance. We posited that host memory T cells could potentially respond to donor-derived non-MHC antigens, such as latent viral antigens or autoantigens, to which the host is immune. Results show that immunity to a model nonself antigen, ovalbumin (OVA), can dramatically disrupt tolerance despite undetectable initial reactivity to donor MHC antigens. Importantly, this blockade of tolerance was CD8+ T cell-dependent and required linked antigen presentation of alloantigens with the test OVA antigen. As such, this pathway represents an unapparent, or "incognito," form of immunity that is sufficient to prevent tolerance and that can be an unforeseen additional immune barrier to clinical transplant tolerance.
Collapse
Affiliation(s)
- M. K. Nelsen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - K. S. Beard
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - R. J. Plenter
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - R. M. Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - E. T. Clambey
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - R. G. Gill
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
27
|
Cytokines affecting CD4 +T regulatory cells in transplant tolerance. III. Interleukin-5 (IL-5) promotes survival of alloantigen-specific CD4 + T regulatory cells. Transpl Immunol 2017; 43-44:33-41. [PMID: 28652007 DOI: 10.1016/j.trim.2017.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022]
Abstract
CD4+T cells mediate antigen-specific allograft tolerance, but die in culture without activated lymphocyte derived cytokines. Supplementation of the media with cytokine rich supernatant, from ConA activated spleen cells, preserves the capacity of tolerant cells to transfer tolerance and suppress rejection. rIL-2 or rIL-4 alone are insufficient to maintain these cells, however. We observed that activation of naïve CD4+CD25+FOXP3+Treg with alloantigen and the Th2 cytokine rIL-4 induces them to express interleukin-5 specific receptor alpha (IL-5Rα) suggesting that IL-5, a Th2 cytokine that is produced later in the immune response may promote tolerance mediating Treg. This study examined if recombinant IL-5(rIL-5) promoted survival of tolerant CD4+, especially CD4+CD25+T cells. CD4+T cells, from DA rats tolerant to fully allogeneic PVG heart allografts surviving over 100days without on-going immunosuppression, were cultured with PVG alloantigen and rIL-5. The ability of these cells to adoptively transfer tolerance to specific-donor allograft and suppress normal CD4+T cell mediated rejection in adoptive DA hosts was examined. Tolerant CD4+CD25+T cells' response to rIL-5 and expression of IL-5Rα was also assessed. rIL-5 was sufficient to promote transplant tolerance mediating CD4+T cells' survival in culture with specific-donor alloantigen. Tolerant CD4+T cells cultured with rIL-5 retained the capacity to transfer alloantigen-specific tolerance and inhibited naïve CD4+T cells' capacity to effect specific-donor graft rejection. rIL-5 promoted tolerant CD4+CD25+T cells' proliferation in vitro when stimulated with specific-donor but not third-party stimulator cells. Tolerant CD4+CD25+T cells expressed IL-5Rα. This study demonstrated that IL-5 promoted the survival of alloantigen-specific CD4+CD25+T cells that mediate transplant tolerance.
Collapse
|
28
|
Effect of Ex Vivo-Expanded Recipient Regulatory T Cells on Hematopoietic Chimerism and Kidney Allograft Tolerance Across MHC Barriers in Cynomolgus Macaques. Transplantation 2017; 101:274-283. [PMID: 27846155 DOI: 10.1097/tp.0000000000001559] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Infusion of recipient regulatory T (Treg) cells promotes durable mixed hematopoietic chimerism and allograft tolerance in mice receiving allogeneic bone marrow transplant (BMT) with minimal conditioning. We applied this strategy in a Cynomolgus macaque model. METHODS CD4 CD25 Treg cells that were polyclonally expanded in culture were highly suppressive in vitro and maintained high expression of FoxP3. Eight monkeys underwent nonmyeloablative conditioning and major histocompatibility complex mismatched BMT with or without Treg cell infusion. Renal transplantation (from the same BMT donor) was performed 4 months post-BMT without immunosuppression to assess for robust donor-specific tolerance. RESULTS Transient mixed chimerism, without significant T cell chimerism, was achieved in the animals that received BMT without Treg cells (N = 3). In contrast, 2 of 5 recipients of Treg cell BMT that were evaluable displayed chimerism in all lineages, including T cells, for up to 335 days post-BMT. Importantly, in the animal that survived long-term, greater than 90% of donor T cells were CD45RA CD31, suggesting they were new thymic emigrants. In this animal, the delayed (to 4 months) donor kidney graft was accepted more than 294 days without immunosuppression, whereas non-Treg cell BMT recipients rejected delayed donor kidneys within 3 to 4 weeks. Early CMV reactivation and treatment was associated with early failure of chimerism, regardless of Treg cell administration. CONCLUSIONS Our studies provide proof-of-principle that, in the absence of early CMV reactivation (and BM-toxic antiviral therapy), cotransplantation of host Treg cell can promote prolonged and high levels of multilineage allogeneic chimerism and robust tolerance to the donor.
Collapse
|
29
|
Metzker M, Shipkova M, von Ahsen N, Andag R, Abe M, Canzler O, Klett C, Leicht S, Olbricht C, Wieland E. Analytical evaluation of a real-time PCR-based DNA demethylation assay to assess the frequency of naturally occurring regulatory T cells in peripheral blood. Clin Biochem 2016; 49:1173-1180. [DOI: 10.1016/j.clinbiochem.2016.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/02/2016] [Accepted: 05/19/2016] [Indexed: 02/07/2023]
|
30
|
Sá H, Leal R, Rosa MS. Renal transplant immunology in the last 20 years: A revolution towards graft and patient survival improvement. Int Rev Immunol 2016; 36:182-203. [PMID: 27682364 DOI: 10.1080/08830185.2016.1225300] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
To deride the hope of progress is the ultimate fatuity, the last word in poverty of spirit and meanness of mind. There is no need to be dismayed by the fact that we cannot yet envisage a definitive solution of our problems, a resting-place beyond which we need not try to go. -P.B. Medawar, 1969 * Thomas E. Starlz, also known as the Father of Clinical Transplantation, once said that organ transplantation was the supreme exception to the rule that most major advances in medicine spring from discoveries in basic science [Starzl T. The mystique of organ transplantation. J Am Coll Surg 2005 Aug;201(2):160-170]. In fact, the first successful identical-twin kidney transplantation performed by Murray's team in December 1954 (Murray J et al. Renal homotransplantations in identical twins. Surg Forum 1955;6:432-436) was the example of an upside down translation medicine: Human clinical transplantation began and researchers tried to understand the underlying immune response and how to control the powerful rejection pathways through experimental models. In the last 20 years, we have witnessed an amazing progress in the knowledge of immunological mechanisms regarding alloimmune response and an outstanding evolution on the identification and characterization of major and minor histocompatibility antigens. This review presents an historical and clinical perspective of those important advances in kidney transplantation immunology in the last 20 years, which contributed to the improvement in patients' quality of life and the survival of end-stage renal patients. In spite of these significant progresses, some areas still need substantial progress, such as the definition of non-invasive biomarkers for acute rejection; the continuous reduction of immunosuppression; the extension of graft survival, and finally the achievement of real graft tolerance extended to HLA mismatch donor: recipient pairs.
Collapse
Affiliation(s)
- Helena Sá
- a Department of Nephrology , Centro Hospitalar e Universitário de Coimbra , Coimbra , Portugal.,b Faculty of Medicine , University of Coimbra , Coimbra , Portugal.,c Immunology Center, Faculty of Medicine , University of Coimbra , Coimbra , Portugal
| | - Rita Leal
- a Department of Nephrology , Centro Hospitalar e Universitário de Coimbra , Coimbra , Portugal
| | | |
Collapse
|
31
|
IL-7 Mediated Homeostatic Expansion of Human CD4+CD25+FOXP3+ Regulatory T Cells After Depletion With Anti-CD25 Monoclonal Antibody. Transplantation 2016; 100:1853-61. [DOI: 10.1097/tp.0000000000001276] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Montane J, Obach M, Alvarez S, Bischoff L, Dai DL, Soukhatcheva G, Priatel JJ, Hardenberg G, Levings MK, Tan R, Orban PC, Verchere CB. CCL22 Prevents Rejection of Mouse Islet Allografts and Induces Donor-Specific Tolerance. Cell Transplant 2016; 24:2143-54. [PMID: 26423995 DOI: 10.3727/096368914x685249] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Manipulation of regulatory T cell (Treg) migration by islet expression of the chemokine CCL22 prevents diabetes in NOD mice and delays recurrent autoimmunity in syngeneic islet transplants. We sought to determine whether attracting Tregs with CCL22 also prevents islet allograft rejection. Isolated Bl/6 mouse islets were transduced overnight with adenovirus expressing CCL22 (Ad-CCL22) downstream of the CMV promoter. Islets were transplanted under the renal capsule of Balb/c recipients made diabetic by streptozotocin. To assess immunologic tolerance, graft-bearing kidneys from recipients of CCL22-expressing islet grafts were removed, and mice received a second transplant of naive islets from the same donor strain or third-party islets into the contralateral kidney. Adenoviral expression of CCL22 conferred prolonged protection of islet allografts in MHC-mismatched, diabetic recipients, maintaining normoglycemia in 75% of recipients for at least 80 days. Increased frequency of Treg cells was observed in islet grafts transduced with Ad-CCL22 compared with untreated grafts. Normoglycemic recipients of CCL22-expressing islet grafts showed complete absence of antidonor antibodies and no lymphocyte proliferation after exposure to donor splenocytes. After removal of the primary graft at day 80, mice that received a second transplant with untreated islets from the same donor strain did not reject the grafts, suggesting the development of tolerance. Expression of CCL22 recruits Treg cells to transplanted islets, prevents activation of alloreactive T-cells and islet allograft failure and induces alloantigen-specific tolerance. Manipulation of Treg cells by CCL22 in transplanted islets may be a novel therapeutic strategy for diabetes.
Collapse
Affiliation(s)
- Joel Montane
- Department of Pathology and Laboratory Medicine, University of British Columbia and Child and Family Research Institute, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Clinical potential of DNA methylation in organ transplantation. J Heart Lung Transplant 2016; 35:843-50. [DOI: 10.1016/j.healun.2016.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/18/2016] [Accepted: 02/26/2016] [Indexed: 01/17/2023] Open
|
34
|
The Critical Role of Induced CD4+ FoxP3+ Regulatory Cells in Suppression of Interleukin-17 Production and Attenuation of Mouse Orthotopic Lung Allograft Rejection. Transplantation 2016; 99:1356-64. [PMID: 25856405 DOI: 10.1097/tp.0000000000000526] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Lung transplantation is the only definitive therapy for many forms of end-stage lung disease. Studies have demonstrated the critical role of interleukin (IL)-17 in the development of lung rejection. Regulatory T cells (Tregs) are essential for the establishment and maintenance of immune tolerance. METHODS We established mouse orthotopic lung transplantation models to investigate the importance of IL-17 and IL-17-producing cell types in acute lung allograft rejection and the efficacy of the adoptive transfer of induced Tregs (iTregs) in attenuating pathologic lesions of lung allografts. RESULTS We found that the IL-17 produced by Th17 cells and γδ T cells might make the primary contributions to the progression of acute lung allograft rejection. Interleukin-17 deficiency decreased lung allograft lesions. Exogenous iTregs maintained their FoxP3 expression levels in lung allograft recipients. Induced Tregs therapy downregulated the expressions of Th17 and IL-17 γδ T cells and increased IL-10 production in the mouse orthotopic lung transplantation models. Moreover, the adoptive transfer of iTregs prolonged the survivals of the lung allografts and attenuated the progression of acute rejection. CONCLUSION These data suggested that the adoptive transfer of iTregs could suppress the Th17 cells and IL-17 γδ cells of the recipients, decrease the expression of IL-17, and attenuate the pathology of acute lung allograft rejection. Exogenous iTregs upregulated immunosuppressive factors, such as IL-10 and suppressed IL-17-producing cells, which was one of the pathways to play a role in protecting lung allografts.
Collapse
|
35
|
Hall BM, Tran GT, Robinson CM, Hodgkinson SJ. Induction of antigen specific CD4+CD25+Foxp3+T regulatory cells from naïve natural thymic derived T regulatory cells. Int Immunopharmacol 2015; 28:875-86. [DOI: 10.1016/j.intimp.2015.03.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
|
36
|
Thymus-Derived Regulatory T Cells Infiltrate the Cardiac Allograft Before Rejection. Transplantation 2015; 99:1839-46. [DOI: 10.1097/tp.0000000000000730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Indoleamine 2, 3-Dioxgenase Transfected Mesenchymal Stem Cells Induce Kidney Allograft Tolerance by Increasing the Production and Function of Regulatory T Cells. Transplantation 2015; 99:1829-38. [DOI: 10.1097/tp.0000000000000856] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
38
|
Karimi MH, Marzban S, Hajiyan MR, Geramizadeh B, Pourfathollah AA, Rajabiyan MH, Ebrahimnezhad S. Effect of CD40 silenced dendritic cells by RNA interference on mice skin allograft rejection. Immunotherapy 2015; 7:111-8. [DOI: 10.2217/imt.14.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: Tolerogenic dendritic cells (DCs) play a critical role in inducing and maintaining tolerance. CD40 is a member of tumor necrosis factor receptor super family and is a potent T-cell costimulatory molecule. Therefore, in this study we evaluated the effect of CD40 silenced DCs by RNA interference on mice skin allograft rejection. Materials & methods: Skin transplantation was performed from C57BL/6 to BALB/c mouse. Skin allograft recipients were assigned to four groups (n = 5). CD40 downregulated DCs were injected to the BALB/c mice intravenously 7 days before transplantation. Then, graft survival time, Treg generation, CD4+ and CD8+ T cells infiltration and cytokine levels in serum of this group were compared with those of untreated and cyclosporine groups. Results: In comparison with untreated group, BALB/c mice injected with CD40 siRNA transfected DCs showed an increased graft survival time, Treg cells, IL-4 and IL-10 cytokine levels as well as decreased number of intragraft CD4+ and CD8+ T cells. IFN-γ and IL-12 secretion were diminished, too. Conclusion: Taken together, these data demonstrate that downregulation of CD40 in DCs can expand Treg cells and increase skin allograft survival.
Collapse
Affiliation(s)
| | - Saeed Marzban
- Plastic & Reconstructive Surgery Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Hajiyan
- Plastic & Reconstructive Surgery Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Akbar Pourfathollah
- Immunology Department, School of Basic Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | |
Collapse
|
39
|
Jiang X, Liu C, Hao J, Guo D, Guo J, Yao J, Jiang K, Cui Z, Zhu L, Sun W, Lin L, Liang J. CD4(+)CD25 (+) regulatory T cells are not required for mesenchymal stem cell function in fully MHC-mismatched mouse cardiac transplantation. Cell Tissue Res 2014; 358:503-14. [PMID: 25103227 DOI: 10.1007/s00441-014-1956-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 07/01/2014] [Indexed: 01/12/2023]
Abstract
Although the immunomodulative properties of mesenchymal stem cells (MSCs) open up attractive possibilities in solid-organ transplantation, information concerning the optimal dose, route, timing of administration, major histocompatibility complex (MHC)-restriction and relevant mechanisms is currently lacking. Therefore, better characterization of MSC immunoregulatory activity and elucidation of its mechanisms are crucial. In this study, we confirmed that MSCs did not elicit proliferation by allogeneic CD4(+) T cells, suggesting that MSCs were not immunogenic. By using C57BL/6 mouse MSCs as donor-derived or recipient-derived or as third-party MSCs, we discovered that MSCs suppressed CD4(+) T cell proliferation and prolonged mouse cardiac allograft survival in a dose-dependent and non-MHC-restricted manner. We also found that intraperitoneal administration favored survival prolongation, although this prolongation was weaker than that via the intravenous route. Only infusion at earlier time points favored survival prolongation. Depletion of CD4(+)CD25(+) T cells did not affect the immunosuppression of MSCs on CD4(+) T cells. Moreover, MSCs did not induce regulatory T cells. The in vivo data revealed that MSCs did not increase the percentage of CD4(+)CD25(+) T cells and FoxP3 expression. More importantly, we demonstrated for the first time that depletion of CD4(+)CD25(+) T cells did not hinder MSC-induced survival prolongation, indicating that CD4(+)CD25(+) regulatory T cells were not essential for the prolongation of MSC-mediated allograft survival.
Collapse
Affiliation(s)
- Xiaofeng Jiang
- Department of Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, Liaoning Province, People's Republic of China,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Short-term TNF-alpha inhibition reduces short-term and long-term inflammatory changes post-ischemia/reperfusion in rat intestinal transplantation. Transplantation 2014; 97:732-9. [PMID: 24598936 DOI: 10.1097/tp.0000000000000032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Tumor necrosis factor (TNF)-α inhibition was shown to reduce ischemia/reperfusion injury (IRI) after intestinal transplantation (ITX). We studied the effects of different TNFα inhibitors on acute IRI and long-term inflammatory responses in experimental ITX. METHODS Orthotopic ITX was performed in an isogenic ischemia/reperfusion model in Lewis rats. The TNFα inhibition groups received infliximab post-reperfusion; etanercept pre-reperfusion and at postoperative days (POD) 1, 3, 5, and 7; or pentoxifylline pre-reperfusion and at POD 1 to 5. Tissue samples were taken from proximal and distal graft sections and mesenteric lymph nodes at 20 min, 12 hr, 7 day, and 6 months post-reperfusion for histopathology, immunohistology, terminal deoxyribosyl transferase-mediated dUTP nick-end labeling (TUNEL) assay, and real-time RT-PCR. Lung sections were stained for the myeloperoxidase assay. RESULTS TNFα inhibitors decreased inflammatory changes after IRI in all treatment groups. Infliximab significantly improved 7-day survival and reduced the histological and immunohistochemical signs of IRI, the numbers of graft-infiltrating T cells and ED1 monocytes and macrophages, and pulmonary neutrophil infiltration, and also enhanced the accumulation of cytoprotective markers. Graft injury was more prominent in the distal graft than in the proximal graft in all groups, regardless of TNFα inhibition. CONCLUSION Infliximab significantly reduced both acute IRI and, as with other TNFα inhibitors, long-term inflammatory responses after rat ITX. TNFα inhibition may help diminish chronic inflammatory long-term effects and avoid chronic allograft enteropathy.
Collapse
|
41
|
EXP CLIN TRANSPLANTExp Clin Transplant 2014; 12. [DOI: 10.6002/ect.2013.0148] [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]
|
42
|
Su Y, Jevnikar AM, Huang X, Lian D, Zhang ZX. Spi6 protects alloreactive CD4(+) but not CD8 (+) memory T cell from granzyme B attack by double-negative T regulatory cell. Am J Transplant 2014; 14:580-93. [PMID: 24730048 DOI: 10.1111/ajt.12614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Memory T (Tm) cells pose a major barrier to long-term transplant survival. Whether regulatory T cells (Tregs)can control them remains poorly defined. Previously,we established that double-negative (DN) Tregs suppress effector T (Teff) cells. Here, we demonstrate that DNTregs effectively suppress CD4+/CD8+Teff and CD8+Tm but not CD4+Tm cells, whereas the suppression on CD8+Tm is abrogated by perforin (PFN) deficiency in DNTregs. Consistently, in a BALB/c to B6-Rag1-/-skin transplantation, transfer of DN Tregs suppressed the rejection mediated by CD4þ/CD8+Teff and CD8+Tmcells (76.0±4.9, 87.5±5.0 and 63.0±4.7 days, respectively)but not CD4þTmcells (25.3±1.4 days). Both CD8þ effector memory T and central memory T compartments significantly reduced after DN Treg transfer. CD4+Tm highly expresses granzyme B (GzmB) inhibitor serine protease inhibitor-6 (Spi6). Spi6 deficiency renders CD4þTm susceptible to DN Treg suppression. In addition,transfer of WT DN Tregs, but not PFN-/-DN Tregs,inhibited the skin allograft rejection mediated by Spi6-/-CD4þTm(75.5±7.9 days). In conclusion, CD4+ and CD8+Tm cells differentially respond toDNTregs’ suppression.The GzmB resistance conferred by Spi6 in CD4þTm cells might hint at the physiological significance of Tmpersistence
Collapse
|
43
|
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
|
44
|
Lee K, Nguyen V, Lee KM, Kang SM, Tang Q. Attenuation of donor-reactive T cells allows effective control of allograft rejection using regulatory T cell therapy. Am J Transplant 2014; 14:27-38. [PMID: 24354870 PMCID: PMC5262439 DOI: 10.1111/ajt.12509] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/01/2013] [Accepted: 08/01/2013] [Indexed: 01/25/2023]
Abstract
Regulatory T cells (Tregs) are essential for the establishment and maintenance of immune tolerance, suggesting a potential therapeutic role for Tregs in transplantation. However, Treg administration alone is insufficient in inducing long-term allograft survival in normal hosts, likely due to the high frequency of alloreactive T cells. We hypothesized that a targeted reduction of alloreactive T effector cells would allow a therapeutic window for Treg efficacy. Here we show that preconditioning recipient mice with donor-specific transfusion followed by cyclophosphamide treatment deleted 70-80% donor-reactive T cells, but failed to prolong islet allograft survival. However, infusion of either 5 × 10(6) Tregs with direct donor reactivity or 25 × 10(6) polyclonal Tregs led to indefinite survival of BALB/c islets in more than 70% of preconditioned C57BL/6 recipients. Notably, protection of C3H islets in autoimmune nonobese diabetic mice required islet autoantigen-specific Tregs together with polyclonal Tregs. Treg therapy led to significant reduction of CD8(+) T cells and concomitant increase in endogenous Tregs among graft-infiltrating cells early after transplantation. Together, these results demonstrate that reduction of the donor-reactive T cells will be an important component of Treg-based therapies in transplantation.
Collapse
Affiliation(s)
- K. Lee
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Department of Biochemistry and Molecular Biology, Korea University, Seoul, Republic of Korea
| | - V. Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - K.-M. Lee
- Department of Biochemistry and Molecular Biology, Korea University, Seoul, Republic of Korea
| | - S.-M. Kang
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Corresponding authors: Qizhi Tang, and Sang-Mo Kang,
| | - Q. Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA,Corresponding authors: Qizhi Tang, and Sang-Mo Kang,
| |
Collapse
|
45
|
Bansal D, Yadav AK, Kumar V, Minz M, Sakhuja V, Jha V. Deferred pre-emptive switch from calcineurin inhibitor to sirolimus leads to improvement in GFR and expansion of T regulatory cell population: a randomized, controlled trial. PLoS One 2013; 8:e75591. [PMID: 24146762 PMCID: PMC3795731 DOI: 10.1371/journal.pone.0075591] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/09/2013] [Indexed: 01/05/2023] Open
Abstract
Background Measures to prevent chronic calcineurin inhibitor (CNI) toxicity have included limiting exposure by switching to sirolimus (SIR). SIR may favorably influence T regulator cell (Treg) population. This randomized controlled trial compares the effect of switching from CNI to SIR on glomerular filtration rate (GFR) and Treg frequency. Methods In this prospective open label randomized trial, primary living donor kidney transplant recipients on CNI-based immunosuppression were randomized to continue CNI or switched to sirolimus 2 months after surgery; 29 were randomized to receive CNI and 31 to SIR. All patients received mycophenolate mofetil and steroids. The main outcome parameter was estimated GFR (eGFR) at 180 days. Treg population was estimated by flowcytometry. Results Baseline characteristics in the two groups were similar. Forty-eight patients completed the trial. At six months, patients in the SIR group had significantly higher eGFR as compared to those in the CNI group (88.94±11.78 vs 80.59±16.51 mL/min, p = 0.038). Patients on SIR had a 12 mL/min gain of eGFR of at the end of six months. Patients in the SIR group showed significant increase in Treg population at 30 days, which persisted till day 180. There was no difference in the adverse events in terms of number of acute rejection episodes, death, infections, proteinuria, lipid profile, blood pressure control and hematological parameters between the two groups. Four patients taking SIR developed enthesitis. No patient left the study or switched treatment because of adverse event. Conclusions A deferred pre-emptive switch over from CNI to SIR safely improves renal function and Treg population at 6 months in living donor kidney transplant recipients. Registered in Clinical Trials Registry of India (CTRI/2011/091/000034)
Collapse
Affiliation(s)
- Dinesh Bansal
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashok K. Yadav
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vinod Kumar
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mukut Minz
- Department of Renal Transplant Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vinay Sakhuja
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vivekanand Jha
- Department of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
- Department of Regenerative and Translational Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
- George Institute of International Health, Hyderabad, India
- * E-mail:
| |
Collapse
|
46
|
Larocca RA, Moraes-Vieira PM, Bassi ÊJ, Semedo P, de Almeida DC, da Silva MB, Thornley T, Pacheco-Silva A, Câmara NOS. Adipose tissue-derived mesenchymal stem cells increase skin allograft survival and inhibit Th-17 immune response. PLoS One 2013; 8:e76396. [PMID: 24124557 PMCID: PMC3790669 DOI: 10.1371/journal.pone.0076396] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 08/29/2013] [Indexed: 12/29/2022] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ADSC) exhibit immunosuppressive capabilities both in vitro and in vivo. Their use for therapy in the transplant field is attractive as they could render the use of immunosuppressive drugs unnecessary. The aim of this study was to investigate the effect of ADSC therapy on prolonging skin allograft survival. Animals that were treated with a single injection of donor allogeneic ADSC one day after transplantation showed an increase in donor skin graft survival by approximately one week. This improvement was associated with preserved histological morphology, an expansion of CD4(+) regulatory T cells (Treg) in draining lymph nodes, as well as heightened IL-10 expression and down-regulated IL-17 expression. In vitro, ADSC inhibit naïve CD4(+) T cell proliferation and constrain Th-1 and Th-17 polarization. In summary, infusion of ADSC one day post-transplantation dramatically increases skin allograft survival by inhibiting the Th-17 pathogenic immune response and enhancing the protective Treg immune response. Finally, these data suggest that ADSC therapy will open new opportunities for promoting drug-free allograft survival in clinical transplantation.
Collapse
Affiliation(s)
- Rafael Assumpção Larocca
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Harvard Medical School, Department of Medicine, The Transplant Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| | - Pedro Manoel Moraes-Vieira
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Harvard Medical School, Department of Medicine, Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Ênio José Bassi
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia Semedo
- Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Danilo Candido de Almeida
- Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Marina Burgos da Silva
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thomas Thornley
- Harvard Medical School, Department of Medicine, The Transplant Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Alvaro Pacheco-Silva
- Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
- Instituto Israelita de Ensino e Pesquisa Albert Einstein Hospital, Renal Transplantation Division, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Laboratory of Clinical and Experimental Immunology, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
FoxP3, Helios, and SATB1: Roles and relationships in regulatory T cells. Int Immunopharmacol 2013; 16:343-7. [DOI: 10.1016/j.intimp.2013.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 02/01/2013] [Indexed: 11/19/2022]
|
48
|
Lim DG, Park YH, Kim SE, Jeong SH, Kim SC. Diagnostic value of tolerance-related gene expression measured in the recipient alloantigen-reactive T cell fraction. Clin Immunol 2013; 148:219-26. [PMID: 23778261 DOI: 10.1016/j.clim.2013.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/17/2013] [Accepted: 05/19/2013] [Indexed: 10/26/2022]
Abstract
The efficient development of tolerance-inducing therapies and safe reduction of immunosuppression should be supported by early diagnosis and prediction of tolerance in transplantation. Using mouse models of donor-specific tolerance to allogeneic skin and islet grafts we tested whether measurement of tolerance-related gene expression in their alloantigen-reactive peripheral T cell fraction efficiently reflected the tolerance status of recipients. We found that Foxp3, Nrn1, and Klrg1 were preferentially expressed in conditions of tolerance compared with rejection or unmanipulated controls if their expression is measured in CD69(+) T cells prepared from coculture of recipient peripheral T cells and donor antigen-presenting cells. The same pattern of gene expression was observed in recipients grafted with either skin or islets, recipients of different genetic origins, and even those taking immunosuppressive drugs. These findings suggest that the expression of tolerance-related genes in the alloantigen-reactive T cell fraction could be used to detect tolerance in the clinic.
Collapse
Affiliation(s)
- Dong-Gyun Lim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
49
|
Leech JM, Sharif-Paghaleh E, Maher J, Livieratos L, Lechler RI, Mullen GE, Lombardi G, Smyth LA. Whole-body imaging of adoptively transferred T cells using magnetic resonance imaging, single photon emission computed tomography and positron emission tomography techniques, with a focus on regulatory T cells. Clin Exp Immunol 2013; 172:169-77. [PMID: 23574314 DOI: 10.1111/cei.12087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 01/03/2023] Open
Abstract
Cell-based therapies using natural or genetically modified regulatory T cells (T(regs)) have shown significant promise as immune-based therapies. One of the main difficulties facing the further advancement of these therapies is that the fate and localization of adoptively transferred T(regs) is largely unknown. The ability to dissect the migratory pathway of these cells in a non-invasive manner is of vital importance for the further development of in-vivo cell-based immunotherapies, as this technology allows the fate of the therapeutically administered cell to be imaged in real time. In this review we will provide an overview of the current clinical imaging techniques used to track T cells and T(regs) in vivo, including magnetic resonance imaging (MRI) and positron emission tomography (PET)/single photon emission computed tomography (SPECT). In addition, we will discuss how the finding of these studies can be used, in the context of transplantation, to define the most appropriate T(reg) subset required for cellular therapy.
Collapse
Affiliation(s)
- J M Leech
- Medical Research Council, Centre for Transplantation, King's College London, King's Health Partners, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Saso S, Ghaem-Maghami S, Louis LS, Ungar L, Del Priore G, Smith JR. Uterine transplantation: What else needs to be done before it can become a reality? J OBSTET GYNAECOL 2013; 33:232-8. [DOI: 10.3109/01443615.2012.734870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|