1
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Karwig L, Moore PF, Alber G, Eschke M. Distinct characteristics of unique immunoregulatory canine non-conventional TCRαβ pos CD4 negCD8α neg double-negative T cell subpopulations. Front Immunol 2024; 15:1439213. [PMID: 39185407 PMCID: PMC11341405 DOI: 10.3389/fimmu.2024.1439213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024] Open
Abstract
Conventional CD4pos regulatory T (Treg) cells characterized by expression of the key transcription factor forkhead box P3 (FoxP3) are crucial to control immune responses, thereby maintaining homeostasis and self-tolerance. Within the substantial population of non-conventional T cell receptor (TCR)αβpos CD4negCD8αneg double-negative (dn) T cells of dogs, a novel FoxP3pos Treg-like subset was described that, similar to conventional CD4pos Treg cells, is characterized by high expression of CD25. Noteworthy, human and murine TCRαβpos regulatory dn T cells lack FoxP3. Immunosuppressive capacity of canine dn T cells was hypothesized based on expression of inhibitory molecules (interleukin (IL)-10, cytotoxic T-lymphocyte associated protein 4, CTLA4). Here, to verify their regulatory function, the dnCD25pos (enriched for FoxP3pos Treg-like cells) and the dnCD25neg fraction, were isolated by fluorescence-activated cell sorting from peripheral blood mononuclear cells (PBMC) of Beagle dogs and analyzed in an in vitro suppression assay in comparison to conventional CD4posCD25pos Treg cells (positive control) and CD4posCD25neg T cells (negative control). Canine dnCD25pos T cells suppressed the Concanavalin A-driven proliferation of responder PBMC to a similar extent as conventional CD4posCD25pos Treg cells. Albeit to a lesser extent than FoxP3-enriched dn and CD4posCD25pos populations, even dnCD25neg T cells reduced the proliferation of responder cells. This is remarkable, as dnCD25neg T cells have a FoxP3neg phenotype comparable to non-suppressive CD4posCD25neg T cells. Both, CD25pos and CD25neg dn T cells, can mediate suppression independent of cell-cell contact and do not require additional signals from CD4posCD25neg T cells to secrete inhibitory factors in contrast to CD4posCD25pos T cells. Neutralization of IL-10 completely abrogated the suppression by dnCD25pos and CD4posCD25pos Treg cells in a Transwell™ system, while it only partially reduced suppression by dnCD25neg T cells. Taken together, unique canine non-conventional dnCD25pos FoxP3pos Treg-like cells are potent suppressor cells in vitro. Moreover, inhibition of proliferation of responder T cells by the dnCD25neg fraction indicates suppressive function of a subset of dn T cells even in the absence of FoxP3. The identification of unique immunoregulatory non-conventional dn T cell subpopulations of the dog in vitro is of high relevance, given the immunotherapeutic potential of manipulating regulatory T cell responses in vivo.
Collapse
Affiliation(s)
- Laura Karwig
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Peter F. Moore
- Department of Veterinary Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Gottfried Alber
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Maria Eschke
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
3
|
Karmi N, Uniken Venema WTC, van der Heide F, Festen EAM, Dijkstra G. Biologicals in the prevention and treatment of intestinal graft rejection: The state of the art Biologicals in Intestinal Transplantation. Hum Immunol 2024; 85:110810. [PMID: 38788483 DOI: 10.1016/j.humimm.2024.110810] [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: 12/22/2023] [Revised: 04/10/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024]
Abstract
Intestinal transplantation is the standard treatment for patients with intestinal failure with severe complications due to parenteral nutrition; however, rejection leads to graft failure in approximately half of both adult and pediatric recipients within 5 years of transplantation. Although intensive immunosuppressive therapy is used in an attempt to reduce this risk, commonly used treatment strategies are generally practice- and/or expert-based, as head-to-head comparisons are lacking. In this ever-developing field, biologicals designed to prevent or treat rejection are used increasingly, with both infliximab and vedolizumab showing potential in the treatment of acute cellular rejection in individual cases and in relatively small patient cohorts. To help advance progress in clinical care, we review the current use of biologicals in intestinal transplantation, and we provide future perspectives to guide this progress.
Collapse
Affiliation(s)
- Naomi Karmi
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Werna T C Uniken Venema
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Frans van der Heide
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Eleonora A M Festen
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Gerard Dijkstra
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands.
| |
Collapse
|
4
|
Iguchi K, Yamamoto Y, Uchiyama M, Masaoka H, Nakamura M, Shizuka H, Imazuru T, Shimokawa T. Graft protective effects and donor-specific antibody suppression by CD4 +CD25 +Foxp3 + regulatory T cell induced by HMG-CoA reductase inhibitor rosuvastatin in a murine heart transplant model. J Cardiothorac Surg 2024; 19:368. [PMID: 38918849 PMCID: PMC11197312 DOI: 10.1186/s13019-024-02888-4] [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: 10/27/2023] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND We previously demonstrated that the hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitor (statins) play an important role in the regulation of alloimmune responses. However, little is known regarding the effects of statin on allograft protection or donor-specific antibodies (DSA). In this study, we investigated the graft-protective and immunomodulatory effects of rosuvastatin in a model of fully major histocompatibility complex-mismatched murine cardiac allograft transplantation. METHODS CBA mice underwent transplantation of C57BL/6 (B6) hearts and received 50 and 500 μg/kg/day of rosuvastatin from the day of transplantation until seven days after the completion of transplantation. To confirm the requirement for regulatory T cells (Tregs), we administered an anti-interleukin-2 receptor alpha antibody (PC-61) to rosuvastatin-treated CBA recipients. Additionally, histological and fluorescent staining, cell proliferation analysis, flow cytometry, and DSA measurements were performed. RESULTS CBA recipients with no treatment rejected B6 cardiac graft acutely (median survival time [MST], 7 days). CBA mice treated with 500 μg/kg/day of rosuvastatin prolonged allograft survival (MSTs, 77 days). Fluorescent staining studies showed that rosuvastatin-treated recipients had strong aggregation of CD4+Foxp3+ cells in the myocardium and around the coronary arteries of cardiac allografts two weeks after grafting. Flow cytometry studies performed two weeks after transplantation showed an increased number of splenic CD4+CD25+Foxp3+ T cells in rosuvastatin-treated recipients. The addition of rosuvastatin to mixed leukocyte cultures suppressed cell proliferation by increasing the number of CD4+CD25+Foxp3+ Tregs. Additionally, Tregs suppressed DSA production in rosuvastatin-treated recipients. CONCLUSION Rosuvastatin treatment may be a complementary graft-protective strategy for suppressing DSA production in the acute phase, driven by the promotion of splenic and graft-infiltrating CD4+CD25+Foxp3+ Tregs.
Collapse
Affiliation(s)
- Kazuhito Iguchi
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Yasuto Yamamoto
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Masateru Uchiyama
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan.
| | - Hisanori Masaoka
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Masahiro Nakamura
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Hiroyuki Shizuka
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Tomohiro Imazuru
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| |
Collapse
|
5
|
Juang JH, Chen CL, Kao CW, Wu ST, Shen CR. In Vivo Imaging of Immune Rejection of MIN6 Cells Transplanted in C3H Mice. Cells 2024; 13:1044. [PMID: 38920672 PMCID: PMC11201743 DOI: 10.3390/cells13121044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Recently, we successfully utilized noninvasive magnetic resonance and bioluminescence imaging to track MIN6 cells subcutaneously transplanted in immunocompromised nude mice for up to 64 days. In this study, we further used bioluminescence imaging to investigate the immune rejection of MIN6 cells in immunocompetent C3H mice. A total of 5 × 106 luciferase-transfected MIN6 cells were implanted into the subcutaneous space of each nude or C3H mouse. After transplantation, hypoglycemia and persistent bioluminescence signals were observed in eight of eight (100%) nude mice and five of nine (56%) C3H mice (p < 0.05). We then presensitized a group of C3H mice with C57BL/6 spleen cells just prior to transplantation (n = 14). Interestingly, none of them had hypoglycemia or persistent bioluminescence signals (p < 0.01 vs. C3H mice without presensitization). Histological examination of the grafts revealed a lack or minimal presence of insulin-positive cells in recipients without hypoglycemia and persistent bioluminescence signals. In contrast, recipients with hypoglycemia and persistent bioluminescence signals showed a significant presence of insulin-positive cells in their grafts. Our results indicate that rejection of MIN6 cells occurred in C3H mice and could be enhanced by presensitization with C57BL/6 spleen cells and that bioluminescence imaging is a useful noninvasive tool for detecting rejection of subcutaneously transplanted MIN6 cells.
Collapse
Affiliation(s)
- Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; (C.-L.C.); (C.-W.K.)
| | - Chen-Ling Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; (C.-L.C.); (C.-W.K.)
| | - Chen-Wei Kao
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; (C.-L.C.); (C.-W.K.)
| | - Shu-Ting Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Chia-Rui Shen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- R&D Center of Biochemical Engineering Technology, Department of Chemical Engineering, Ming Chi University of Technology, New Taipei 24301, Taiwan
| |
Collapse
|
6
|
Fouza A, Fylaktou A, Tagkouta A, Daoudaki M, Vagiotas L, Kasimatis E, Stangou M, Xochelli A, Nikolaidou V, Katsanos G, Tsoulfas G, Skoura L, Papagianni A, Antoniadis N. Evaluation of Regulatory B Cell Subpopulations CD24++CD38++, CD24++CD27+, Plasmablasts and Their Correlation with T Regs CD3+CD4+CD25+FOXP3+ in Dialysis Patients and Early Post-Transplant Rejection-Free Kidney Recipients. J Clin Med 2024; 13:3080. [PMID: 38892795 PMCID: PMC11173263 DOI: 10.3390/jcm13113080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Background: B and T regulatory cells, also known as Bregs and Tregs, are involved in kidney transplantation. The purpose of this study is to monitor changes in the frequency and absolute numbers of Tregs (CD3+CD4+CD25+FoxP3+), transitional Bregs (tBregs) (CD24++CD38++), memory Bregs (mBregs) (CD24++CD27+), and plasmablasts before (T0) and six months (T6) after transplantation. Additionally, we aim to investigate any correlation between Tregs and tBregs, mBregs, or plasmablasts and their relationship with graft function. Methods: Flow cytometry was used to immunophenotype cells from 50 kidney recipients who did not experience rejection. Renal function was assessed using the estimated glomerular filtration rate (eGFR). Results: At T6, there was a significant decrease in the frequency of Tregs, plasmablasts, and tBregs, as well as in the absolute number of tBregs. The frequency of mBregs, however, remained unchanged. Graft function was found to have a positive correlation with the frequency of tBregs and plasmablasts. A significant correlation was observed between the frequency and absolute number of tBregs only when the eGFR was greater than 60 but not at lower values. At an eGFR greater than 60, there was a positive correlation between the absolute numbers of Tregs and mBregs but not between Tregs and tBregs. No correlation was observed for any cell population in dialysis patients. Conclusions: The data show a correlation between the frequency and absolute number of tBregs and the absolute number of Tregs and mBregs with good renal function in the early post-transplant period.
Collapse
Affiliation(s)
- Ariadni Fouza
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Asimina Fylaktou
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Anneta Tagkouta
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Daoudaki
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Lampros Vagiotas
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Efstratios Kasimatis
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Maria Stangou
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Aliki Xochelli
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Vasiliki Nikolaidou
- National Peripheral Histocompatibility Center, Department of Immunology, Ippokration General Hospital, 54642 Thessaloniki, Greece; (A.F.); (A.X.); (V.N.)
| | - Georgios Katsanos
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Georgios Tsoulfas
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| | - Lemonia Skoura
- Microbiology Laboratory, Department of Immunology, AHEPA University Hospital, 54636 Thessaloniki, Greece;
| | - Aikaterini Papagianni
- 1st Department of Nephrology, School of Medicine, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece; (E.K.); (M.S.); (A.P.)
| | - Nikolaos Antoniadis
- Department of Transplant Surgery, Center for Research and Innovation in Solid Organ Transplantation School of Medicine, Aristotle University of Thessaloniki, Ippokratio General Hospital, 54642 Thessaloniki, Greece; (L.V.); (G.K.); (G.T.); (N.A.)
| |
Collapse
|
7
|
Belyayev L, Kang J, Sadat M, Loh K, Patil D, Muralidaran V, Khan K, Kaufman S, Subramanian S, Gusev Y, Bhuvaneshwar K, Ressom H, Varghese R, Ekong U, Matsumoto CS, Robson SC, Fishbein TM, Kroemer A. Suppressor T helper type 17 cell responses in intestinal transplant recipients with allograft rejection. Hum Immunol 2024; 85:110773. [PMID: 38494386 DOI: 10.1016/j.humimm.2024.110773] [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: 12/12/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Intestinal transplant (ITx) rejection is associated with memory T helper type 17 cell (Th17) infiltration of grafted tissues. Modulation of Th17 effector cell response is facilitated by T regulatory (Treg) cells, but a phenotypic characterization of this process is lacking in the context of allograft rejection. METHODS Flow cytometry was performed to examine the expression of surface receptors, cytokines, and transcription factors in Th17 and Treg cells in ITx control (n = 34) and rejection patients (n = 23). To elucidate key pathways guiding the rejection biology, we utilized RNA sequencing (RNAseq) and assessed epigenetic stability through pyrosequencing of the Treg-specific demethylated region (TSDR). RESULTS We found that intestinal allograft rejection is characterized by Treg cellular infiltrates, which are polarized toward Th17-type chemokine receptor, ROR-γt transcription factor expression, and cytokine production. These Treg cell subsets have maintained epigenetic stability, as defined by FoxP3-TSDR methylation status, but displayed upregulation of functional Treg and purinergic signaling genes by RNAseq analysis such as CD39, in keeping with suppressor Th17 properties. CONCLUSION We show that ITx rejection is associated with increased polarized cells that express a Th17-like phenotype concurrent with regulatory purinergic markers.
Collapse
Affiliation(s)
- Leonid Belyayev
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA; Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20814, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20007, USA
| | - Mohammed Sadat
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Katrina Loh
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Stuart Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Sukanya Subramanian
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington, DC 20075, USA
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington, DC 20075, USA
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20008, USA
| | - Rency Varghese
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20008, USA
| | - Udeme Ekong
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Cal S Matsumoto
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Thomas M Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA.
| |
Collapse
|
8
|
Masaoka H, Yamamoto Y, Uchiyama M, Iguchi K, Nakamura M, Yagita H, Imazuru T, Shimokawa T. Graft Protective and Intercellular Immunomodulatory Effects by Adoptive Transfer of an Agonistic Anti-BTLA mAb (3C10) Induced CD4 +CD25 + Regulatory T Cells in Murine Cardiac Allograft Transplant Model. Transplant Proc 2024; 56:692-700. [PMID: 38360464 DOI: 10.1016/j.transproceed.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND We demonstrated that an agonistic anti-B and T lymphocyte attenuator antibody (3C10) prolonged cardiac survival by inducing regulatory T cells (Treg). However, the mechanisms of immune tolerance in the recipients remained unclear. In this study, we investigated the graft-protective and intercellular immunomodulatory effects of adoptive transfer (AT) of 3C10-induced Tregs in a murine cardiac allograft transplant model. METHODS Thirty days after transplantation of a C57BL/6 heart into the primary 3C10-treated CBA recipients, splenic CD4+CD25+ cells from these recipients (3C10/AT group) or naïve CBA mice (no-treatment group) were adoptively transferred into secondary CBA recipients with a C57BL/6 heart. To confirm the requirement for 3C10-induced Tregs, we administered an anti-interleukin-2 receptor alpha antibody (PC-61) to secondary CBA recipients. Additionally, histologic and fluorescent staining, cell proliferation analysis, flow cytometry, and donor-specific antibody (DSA) measurements were performed. RESULTS 3C10/AT-treated CBA recipients resulted in significantly prolonged allograft survival (median survival time [MST], >50 days). Allografts displayed prolonged function with preservation of vessel structure by maintaining high numbers of splenic CD4+CD25+Foxp3+ Treg and intramyocardial CD4+Foxp3+ cells. DSA levels were suppressed in 3C10/AT-treated CBA recipients. Moreover, PC-61 administration resulted in a shorter MSTs of cardiac allograft survivals, a detrimental increase in DSA production, and enhanced expression of programmed cell death (PD)-1. CONCLUSION AT of 3C10-induced Tregs may be a promising graft-protective strategy to prolong allograft survival and suppress DSA production, driven by the promotion of splenic and graft-infiltrating Tregs and collaboration with PD-1+ T cells and Treg.
Collapse
Affiliation(s)
- Hisanori Masaoka
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Yasuto Yamamoto
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Masateru Uchiyama
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan.
| | - Kazuhito Iguchi
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Masahiro Nakamura
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University, Tokyo, Japan
| | - Tomohiro Imazuru
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| |
Collapse
|
9
|
Vafadar A, Vosough P, Jahromi HK, Tajbakhsh A, Savardshtaki A, Butler AE, Sahebkar A. The role of efferocytosis and transplant rejection: Strategies in promoting transplantation tolerance using apoptotic cell therapy and/or synthetic particles. Cell Biochem Funct 2023; 41:959-977. [PMID: 37787641 DOI: 10.1002/cbf.3852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/26/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
Recently, efforts have been made to recognize the precise reason(s) for transplant failure and the process of rejection utilizing the molecular signature. Most transplant recipients do not appreciate the unknown length of survival of allogeneic grafts with the existing standard of care. Two noteworthy immunological pathways occur during allogeneic transplant rejection. A nonspecific innate immune response predominates in the early stages of the immune reaction, and allogeneic antigens initiate a donor-specific adaptive reaction. Though the adaptive response is the major cause of allograft rejection, earlier pro-inflammatory responses that are part of the innate immune response are also regarded as significant in graft loss. The onset of the innate and adaptive immune response causes chronic and acute transplant rejection. Currently employed immunosuppressive medications have shown little or no influence on chronic rejection and, as a result, on overall long-term transplant survival. Furthermore, long-term pharmaceutical immunosuppression is associated with side effects, toxicity, and an increased risk of developing diseases, both infectious and metabolic. As a result, there is a need for the development of innovative donor-specific immunosuppressive medications to regulate the allorecognition pathways that induce graft loss and to reduce the side effects of immunosuppression. Efferocytosis is an immunomodulatory mechanism with fast and efficient clearance of apoptotic cells (ACs). As such, AC therapy strategies have been suggested to limit transplant-related sequelae. Efferocytosis-based medicines/treatments can also decrease the use of immunosuppressive drugs and have no detrimental side effects. Thus, this review aims to investigate the impact of efferocytosis on transplant rejection/tolerance and identify approaches using AC clearance to increase transplant viability.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Kargar Jahromi
- Research Center for Non-Communicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardshtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland - Bahrain, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Ge J, Pan W, Feeney NJ, Ott L, Anderson E, Alessandrini A, Zanoni I, Markmann JF, Cuenca AG. Adjuvant conditioning induces an immunosuppressive milieu that delays alloislet rejection through the expansion of myeloid-derived suppressor cells. Am J Transplant 2023; 23:935-945. [PMID: 37080464 PMCID: PMC10330215 DOI: 10.1016/j.ajt.2023.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
Advances in immunosuppression have been relatively stagnant over the past 2 decades, and transplant recipients continue to experience long-term morbidity associated with immunosuppression regimens. Strategies to reduce or eliminate the dosage of immunosuppression medications are needed. We discovered a novel administration strategy using the classic adjuvant alum to condition murine islet transplant recipients, known as adjuvant conditioning (AC), to expand both polymorphonuclear and monocytic myeloid-derived suppressive cells (MDSCs) in vivo. These AC MDSCs potently suppress T cell proliferation when cultured together in vitro. AC MDSCs also facilitate naïve CD4+ T cells to differentiate into regulatory T cells. In addition, we were able to demonstrate a significant delay in alloislet rejection compared with that by saline-treated control following adjuvant treatment in a MDSC-dependent manner. Furthermore, AC MDSCs produce significantly more interleukin (IL)-10 than saline-treated controls, which we demonstrated to be critical for the increased T cell suppressor function of AC MDSCs as well as the observed protective effect of AC against alloislet rejection. Our data suggest that adjuvant-related therapeutics designed to expand MDSCs could be a useful strategy to prevent transplant rejection and curb the use of toxic immunosuppressive regimens currently used in transplant patients.
Collapse
Affiliation(s)
- Jifu Ge
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weikang Pan
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Noel J Feeney
- Division of Transplant Surgery, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leah Ott
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Emily Anderson
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alessandro Alessandrini
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Massachusetts, USA; Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan Zanoni
- Division of Gastroenterology/Immunology, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - James F Markmann
- Division of Transplant Surgery, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Massachusetts, USA
| | - Alex G Cuenca
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Massachusetts, USA.
| |
Collapse
|
11
|
Saireito (TJ-114) Suppressed Donor-Specific Antibody Through Immunomodulatory Effects and Regulatory T Cell Induction in a Murine Heart Transplant Model. Transplant Proc 2022; 54:482-486. [DOI: 10.1016/j.transproceed.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
|
12
|
Chruscinski A, Rojas-Luengas V, Moshkelgosha S, Issachar A, Luo J, Yowanto H, Lilly L, Smith R, Renner E, Zhang J, Epstein M, Grant D, McEvoy CM, Konvalinka A, Humar A, Adeyi O, Fischer S, Volmer FH, Taubert R, Jaeckel E, Juvet S, Selzner N, Levy GA. Evaluation of a gene expression biomarker to identify operationally tolerant liver transplant recipients: the LITMUS trial. Clin Exp Immunol 2022; 207:123-139. [PMID: 35020854 PMCID: PMC8802178 DOI: 10.1093/cei/uxab011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
LITMUS was a single-centre, Phase 2a study designed to investigate whether the gene biomarker FGL2/IFNG previously reported for the identification of tolerance in murine models could identify operationally tolerant liver transplant recipients. Multiplex RT-PCR was used to amplify eight immunoregulatory genes in peripheral blood mononuclear cells (PBMC) from 69 adult liver transplant recipients. Patients with PBMC FGL2/IFNG ≥ 1 and a normal liver biopsy underwent immunosuppression (IS) withdrawal. The primary end point was the development of operational tolerance. Secondary end points included correlation of tolerance with allograft gene expression and immune cell markers. Twenty-eight of 69 patients (38%) were positive for the PBMC tolerance biomarker and 23 proceeded to IS withdrawal. Nine of the 23 patients had abnormal baseline liver biopsies and were excluded. Of the 14 patients with normal biopsies, eight (57%) have achieved operational tolerance and are off IS (range 12–57 months). Additional studies revealed that all of the tolerant patients and only one non-tolerant patient had a liver gene ratio of FOXP3/IFNG ≥ 1 prior to IS withdrawal. Increased CD4+ T regulatory T cells were detected both in PBMC and livers of tolerant patients following IS withdrawal. Higher expression of SELE (gene for E-selectin) and lower expression of genes associated with inflammatory responses (GZMB, CIITA, UBD, LSP1, and CXCL9) were observed in the pre-withdrawal liver biopsies of tolerant patients by RNA sequencing. These results suggest that measurement of PBMC FGL2/IFNG may enrich for the identification of operationally tolerant liver transplant patients, especially when combined with intragraft measurement of FOXP3/IFNG. Clinical Trial Registration: ClinicalTrials.gov (LITMUS: NCT02541916).
Collapse
Affiliation(s)
- Andrzej Chruscinski
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Vanessa Rojas-Luengas
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Sajad Moshkelgosha
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Assaf Issachar
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | | | | | - Leslie Lilly
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Robert Smith
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Eberhard Renner
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Jianhua Zhang
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Maor Epstein
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - David Grant
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Caitriona M McEvoy
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Ana Konvalinka
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Atul Humar
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Oyedele Adeyi
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Sandra Fischer
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Felix H Volmer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Stephen Juvet
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Nazia Selzner
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| | - Gary A Levy
- Multi-Organ Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, Canada
| |
Collapse
|
13
|
Yamamoto Y, Ikeda T, Uchiyama M, Iguchi K, Imazuru T, Shimokawa T. Effects of Each Domain in Recombinant Human Soluble Thrombomodulin on Prolongation of Murine Cardiac Allograft Survival. Transplant Proc 2022; 54:487-491. [DOI: 10.1016/j.transproceed.2021.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 10/19/2022]
|
14
|
Yamamoto Y, Uchiyama M, Iguchi K, Kawai K, Imazuru T, Kawamura M, Shimokawa T. Effects of Glycyrrhizic Acid in Licorice on Prolongation of Murine Cardiac Allograft Survival. Transplant Proc 2022; 54:476-481. [DOI: 10.1016/j.transproceed.2021.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/02/2021] [Accepted: 12/27/2021] [Indexed: 11/29/2022]
|
15
|
Sampani E, Vagiotas L, Daikidou DV, Nikolaidou V, Xochelli A, Kasimatis E, Lioulios G, Dimitriadis C, Fylaktou A, Papagianni A, Stangou M. End stage renal disease has an early and continuous detrimental effect on regulatory T cells. Nephrology (Carlton) 2021; 27:281-287. [PMID: 34781412 DOI: 10.1111/nep.13996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022]
Abstract
End stage renal disease (ESRD) is followed by disturbed adaptive immunity, together with alterations in T cell subsets, including CD4+CD25+FoxP3+ cells (Tregs). In the present study, we assessed the effect of haemodialysis (HD) on the Treg population. CD3+CD4+, CD3+CD8+ and CD4+CD25+FoxP3+ cells were estimated by flow cytometry in 142 ESRD patients (45 ESRD-preHD, 97 on HD) and 30 healthy controls (HC). Patients on HD were classified into three groups according to time on dialysis (HD vintage - HDV): A < 2 years, B: 2-5 years and C: >5 years on HD. The mean age of patients on HD (M/F 53/44) was 54.8 ± 14 years and the median HDV 58 (78) months. We observed a significant progressive reduction in the percentage and count of lymphocytes (p < .001, p < .001, respectively), CD3+CD4+ (p = .003 and, p < .001, respectively) and Tregs (p = .001 and, p < .001, respectively), between HC, ESRD-preHD and HD patients. HDV had a significant inverse correlation with total lymphocyte, CD3+CD4+ and Treg cell counts (p = .001, p < .001, p < .001, respectively) and, the percentage of lymphocytes and CD3+CD4+ cells (p = .005, p = .01, respectively). Furthermore, we stratified patients on HD into three groups according to HDV: A < 2 years, B: 2-5 years and C: >5 years on HD. Total lymphocytes and Tregs were significantly different among the three vintage groups (Kruskal-Wallis H test, p < .001, p < .001 respectively). CD3+CD4+ and CD3+CD8+ cells were also significantly affected (p < .001 and p = .001, respectively), after at least 2 years of HD. Tregs show prompt and significant reduction at the pre-dialysis stage, and continue to decrease gradually even after long-term HD, in a context of total lymphocyte reduction.
Collapse
Affiliation(s)
- Erasmia Sampani
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lampis Vagiotas
- Department of Transplant Surgery, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitra-Vasilia Daikidou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vasiliki Nikolaidou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration Hospital, Thessaloniki, Greece
| | - Aliki Xochelli
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration Hospital, Thessaloniki, Greece
| | - Efstratios Kasimatis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Lioulios
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Chrysostomos Dimitriadis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration Hospital, Thessaloniki, Greece
| | - Aikaterini Papagianni
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Stangou
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
16
|
Li Y, Cao L, Qian Z, Guo Q, Niu X, Huang L. Mifepristone regulates Tregs function mediated by dendritic cells through suppressing the expression of TGF-β. Immunopharmacol Immunotoxicol 2021; 43:85-93. [PMID: 33406939 DOI: 10.1080/08923973.2020.1867998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Previous studies have demonstrated that mifepristone in the daily low-dose affects the function of endometrium. These researches also implied an alteration of endometrium immune balance, which might be involved in regulating endometrial function. However, the detailed mechanisms remain to be further explored. METHODS In this study, the expressions of CD80, CD86, and ICAM-1 in dendritic cells (DCs), which were stimulated with different concentrations of mifepristone (20, 65, and 200 nM), were detected by FACS. After that, we further evaluated the expression of Forkhead box P3 (FOXP3) and IL-10 in Tregs, which co-cultured with mifepristone treated DCs. In mechanism, we compared the indoleamine 2,3-dioxygenase (IDO) and TGF-β expression with enzyme-linked immunosorbent assay (ELISA). RESULTS The results indicated that mifepristone promoted the expressions of CD80, CD86, and ICAM-1 in a dosage dependent manner. Reversely, FOXP3 and IL-10 expression levels in Tregs co-cultured with mifepristone-treated DCs were significantly decreased compared with those co-cultured with nontreated DC. Furthermore, a significant reduce in IDO and TGF-β expression was observed in DCs treated with mifepristone. By using the IDO inhibitor (1-methyl tryptophan, 1-MT) or TGF-b supplement, we confirmed that TGF-β, but not IDO could rescue the downregulation of FOXP3 and IL-10 in Tregs co-cultured with mifepristone treated DCs. All of these results suggest that mifepristone may regulate DC function by decreasing TGF-β expression, which further results in the downregulations of FOXP3 and IL-10 in Tregs. CONCLUSION Therefore, our research provides a theoretical basis for a potentially clinical application of mifepristone as a novel contraceptive.
Collapse
Affiliation(s)
- Yinghua Li
- Hangzhou Women's Hospital, Hangzhou, China.,Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Cao
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhida Qian
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingyun Guo
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaocen Niu
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Huang
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
17
|
Hwang W, Lee J. Pathophysiologic Implications of Cytokines Secretion during Liver Transplantation Surgery. Int J Med Sci 2018; 15:1737-1745. [PMID: 30588198 PMCID: PMC6299421 DOI: 10.7150/ijms.28382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/18/2018] [Indexed: 12/29/2022] Open
Abstract
We introduce the following issues of the cytokines secretion during liver transplantation surgery in this review article; 1) the aspect of cytokines secretion during liver transplantation surgery, 2) the evidences of association of cytokines concentration with post-transplantation graft survival, 3) a variety of factors that may influence the secretion of cytokines during liver transplantation, 4) pro-inflammatory and anti-inflammatory cytokine balance during the surgery, and 5) the issues of T helper 1 and T helper 2, and T helper 17 and regulatory T cell signature cytokines secretion and their ratio during liver transplantation surgery. Primary failure of the liver is associated with the secondary dysfunction of virtually all other organ systems, including the cardiovascular, pulmonary, renal, coagulation and central nervous systems. In addition, liver transplantation surgery itself is a major surgical procedure with accompanying life-threatening hemorrhage, massive transfusion, clamping and unclamping of great vessels and resulting ischemia-reperfusion injury and cardiovascular instability. Both the underlying liver failure and the surgical events act as stressors and promote the secretion of various cytokines. So it is clinically important to understand above issues regarding the cytokines secretion during liver transplantation surgery. As cytokines secretion has clear relationship with post-transplantation clinical outcomes, future study directions for artificially manipulating cytokines secretion is also suggested for enhancing outcomes of the patients.
Collapse
Affiliation(s)
- Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Jaemin Lee
- Department of Anesthesiology and Pain Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| |
Collapse
|
18
|
Liu Y, Sun L, Chen W, Chuai J, Shang Y, Zhang D, Fu B, Tian H, Jiang S. Combined treatment with simvastatin and rapamycin attenuates cardiac allograft rejection through the regulation of T helper 17 and regulatory T cells. Exp Ther Med 2017; 15:1941-1949. [PMID: 29434788 PMCID: PMC5776515 DOI: 10.3892/etm.2017.5635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 07/07/2017] [Indexed: 12/11/2022] Open
Abstract
Allograft rejection is an important issue post cardiac transplantation. In order to investigate the effect of combined treatment with simvastatin and rapamycin on allograft rejection, a cardiac transplantation rat model was employed in the present study. The survival time of rats following cardiac transplantation was recorded, while histopathological alterations were assessed by hematoxylin and eosin staining. The levels of transcription factors were measured by reverse transcription-quantitative polymerase chain reaction. In addition, the levels of CD4+ interleukin (IL)-17+ cells and CD4+ forkhead box P3 (FOXP3)+ cells in the allografts and CD4+ T cells and CD8+ T cells in the spleens were detected by flow cytometry. The results of the current study demonstrated that, following treatment with simvastatin and rapamycin, the survival time of model rats was prolonged, and the histopathological damage was attenuated. Treatment with simvastatin and rapamycin also led to decreased retinoic acid receptor-related orphan receptor γt (RORγt) level, increased FOXP3 level, reduced levels of CD4+IL-17+, CD4+ T and CD8+ T cells, and increased level of CD4+FOXP3+ cells. In conclusion, the current study observed that simvastatin and rapamycin performed a synergistic effect to reduce cardiac transplantation rejection. Thus, combined therapy of simvastatin and rapamycin may be a promising adjuvant therapy to reduce rejection post cardiac transplantation.
Collapse
Affiliation(s)
- Yingjie Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Lu Sun
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wei Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Junbo Chuai
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Shang
- Department of Respiratory Medicine, Harbin First Hospital, Harbin, Heilongjiang 150010, P.R. China
| | - Dongyang Zhang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bicheng Fu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hai Tian
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shulin Jiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
19
|
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
|
20
|
Baroja-Mazo A, Revilla-Nuin B, Parrilla P, Martínez-Alarcón L, Ramírez P, Pons JA. Tolerance in liver transplantation: Biomarkers and clinical relevance. World J Gastroenterol 2016; 22:7676-91. [PMID: 27678350 PMCID: PMC5016367 DOI: 10.3748/wjg.v22.i34.7676] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/04/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Transplantation is the optimal treatment for end-stage organ failure, and modern immunosuppression has allowed important progress in short-term outcomes. However, immunosuppression poorly influences chronic rejection and elicits chronic toxicity in current clinical practice. Thus, a major goal in transplantation is to understand and induce tolerance. It is well established that human regulatory T cells expressing the transcription factor FoxP3 play important roles in the maintenance of immunological self-tolerance and immune homeostasis. The major regulatory T cell subsets and mechanisms of expansion that are critical for induction and long-term maintenance of graft tolerance and survival are being actively investigated. Likewise, other immune cells, such as dendritic cells, monocyte/macrophages or natural killer cells, have been described as part of the process known as "operational tolerance". However, translation of these results towards clinical practice needs solid tools to identify accurately and reliably patients who are going to be tolerant. In this way, a plethora of genetic and cellular biomarkers is raising and being validated worldwide in large multi-center clinical trials. Few of the studies performed so far have provided a detailed analysis of the impact of immunosuppression withdrawal on pre-existing complications derived from the long-term administration of immunosuppressive drugs and the side effects associated with them. The future of liver transplantation is aimed to develop new therapies which increase the actual low tolerant vs non-tolerant recipients ratio.
Collapse
|
21
|
A Critical Role for TGF-β/Fc and Nonlytic IL-2/Fc Fusion Proteins in Promoting Chimerism and Donor-Specific Tolerance. Transplantation 2016; 101:294-301. [PMID: 27306533 DOI: 10.1097/tp.0000000000001283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Immunoglobulin-cytokine fusion molecules have been shown to be the new generation of immunomodulating agents in transplantation tolerance induction. In the present study, we tested whether immunoregulatory cytokine fusion proteins of IL-10/Fc, TGF-β/Fc, or IL-2/Fc would enhance allogeneic bone marrow cell (BMC) engraftment and promote tolerance induction. METHODS B6 (H2) mice were conditioned with anti-CD154 (MR1) and rapamycin (Rapa) plus 100 cGy total body irradiation (MR1/Rapa/100 cGy) and transplanted with allogeneic B10.D2 (H2) BMC. Recipients were treated with lytic IL-2/Fc, nonlytic IL-2/Fc, TGF-β/Fc, or IL-10/Fc fusion proteins to promote chimerism to induce tolerance. RESULTS Donor chimerism was achieved in 20% of recipients conditioned with MR1/Rapa/100 cGy. The addition of TGF-β/Fc (5- or 10-day treatment) or nonlytic IL-2/Fc (10-day treatment) fusion proteins to the conditioning resulted in engraftment in nearly 100% of recipients. In contrast, lytic IL-2/Fc or IL-10/Fc had no effect. The combination of nonlytic IL-2/Fc and TGF-β/Fc had a synergistic effect to promote engraftment and resulted in significantly higher donor chimerism compared with recipients conditioned with TGF-β/MR1/Rapa/100 cGy. Engraftment was durable in the majority of chimeras and increased over time. The chimeras accepted donor skin grafts and promptly rejected third-party skin grafts. Moreover, specific T cell receptor-Vβ5.½ and TCR-Vβ11 clonal deletion was detected in host T cells in chimeras, suggesting central tolerance to donor alloantigens. CONCLUSIONS Allogeneic BMC engraftment is enhanced with TGF-β/Fc fusion protein treatment. TGF-β/Fc and nonlytic IL-2/Fc exert a synergistic effect in promotion of alloengraftment and donor-specific transplant tolerance, significantly decreasing the minimum total body irradiation dose required.
Collapse
|
22
|
Latorre I, Esteve-Sole A, Redondo D, Giest S, Argilaguet J, Alvarez S, Peligero C, Forstmann I, Crespo M, Pascual J, Meyerhans A. Calcineurin and mTOR inhibitors have opposing effects on regulatory T cells while reducing regulatory B cell populations in kidney transplant recipients. Transpl Immunol 2016; 35:1-6. [DOI: 10.1016/j.trim.2016.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/24/2016] [Accepted: 01/30/2016] [Indexed: 01/19/2023]
|
23
|
Induction of tolerance and prolongation of islet allograft survival by syngeneic hematopoietic stem cell transplantation in mice. Transpl Immunol 2015; 33:130-9. [DOI: 10.1016/j.trim.2015.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 08/13/2015] [Accepted: 08/18/2015] [Indexed: 12/29/2022]
|
24
|
Chen T, Yuan J, Duncanson S, Hibert ML, Kodish BC, Mylavaganam G, Maker M, Li H, Sremac M, Santosuosso M, Forbes B, Kashiwagi S, Cao J, Lei J, Thomas M, Hartono C, Sachs D, Markmann J, Sambanis A, Poznansky MC. Alginate encapsulant incorporating CXCL12 supports long-term allo- and xenoislet transplantation without systemic immune suppression. Am J Transplant 2015; 15:618-27. [PMID: 25693473 DOI: 10.1111/ajt.13049] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 10/03/2014] [Accepted: 10/04/2014] [Indexed: 01/25/2023]
Abstract
Islet transplantation represents a potentially curative approach for individuals with Type I Diabetes. The requirement for systemic immune suppression to control immune-mediated rejection of transplanted islets and the limited human islet supply represent significant roadblocks to progress for this approach. Islet microencapsulation in alginate offers limited protection in the absence of systemic immunosuppression, but does not support long-term islet survival. The chemokine, CXCL12, can repel effector T cells while recruiting immune-suppressive regulatory T cells (Tregs) to an anatomic site while providing a prosurvival signal for beta-cells. We proposed that coating or encapsulating donor islets with CXCL12 would induce local immune-isolation and protect and support the function of an allo- or xenograft without systemic immune suppression. This study investigated the effect of alginate microcapsules incorporating CXCL12 on islet function. Islet transplantation was performed in murine models of insulin-dependent diabetes. Coating of islets with CXCL12 or microencapsulation of islets with alginate incorporating the chemokine, resulted in long-term allo- and xenoislet survival and function, as well as a selective increase in intragraft Tregs. These data support the use of CXCL12 as a coating or a component of an alginate encapsulant to induce sustained local immune-isolation for allo- or xenoislet transplantation without systemic immunosuppression.
Collapse
Affiliation(s)
- T Chen
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Charlestown, MA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
The ratio of circulating regulatory T cells (Tregs)/Th17 cells is associated with acute allograft rejection in liver transplantation. PLoS One 2014; 9:e112135. [PMID: 25372875 PMCID: PMC4221545 DOI: 10.1371/journal.pone.0112135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/13/2014] [Indexed: 12/31/2022] Open
Abstract
CD4+CD25+FoxP3+ regulatory T cells (Tregs) and Th17 cells are known to be involved in the alloreactive responses in organ transplantation, but little is known about the relationship between Tregs and Th17 cells in the context of liver alloresponse. Here, we investigated whether the circulating Tregs/Th17 ratio is associated with acute allograft rejection in liver transplantation. In present study, thirty-eight patients who received liver transplant were enrolled. The patients were divided into two groups: acute allograft rejection group (Gr-AR) (n = 16) and stable allograft liver function group (Gr-SF) (n = 22). The frequencies of circulating Tregs and circulating Th17 cells, as well as Tregs/Th17 ratio were determined using flow cytometry. The association between Tregs/Th17 ratio and acute allograft rejection was then analyzed. Our results showed that the frequency of circulating Tregs was significantly decreased, whereas the frequency of circulating Th17 cells was significantly increased in liver allograft recipients who developed acute rejection. Tregs/Th17 ratio had a negative correlation with liver damage indices and the score of rejection activity index (RAI) after liver transplantation. In addition, the percentages of CTLA-4+, HLA-DR+, Ki67+, and IL-10+ Tregs were higher in Gr-SF group than in Gr-AR group. Our results suggested that the ratio of circulating Tregs/Th17 cells is associated with acute allograft rejection, thus the ratio may serve as an alternative marker for the diagnosis of acute rejection.
Collapse
|
26
|
Yan F, Cai L, Hui Y, chen S, Meng H, Huang Z. Tolerogenic dendritic cells suppress murine corneal allograft rejection by modulating CD28/CTLA-4 expression on regulatory T cells. Cell Biol Int 2014; 38:835-48. [PMID: 24604878 DOI: 10.1002/cbin.10268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/21/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Feng Yan
- Department of Ophthalmology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing 210002 China
- Department of Ophthalmology; Xijing Hospital; The Fourth Military Medical University; Xi'an China
| | - Li Cai
- Department of Ophthalmology; Xijing Hospital; The Fourth Military Medical University; Xi'an China
| | - Yannian Hui
- Department of Ophthalmology; Xijing Hospital; The Fourth Military Medical University; Xi'an China
| | - Suihua chen
- Department of Ophthalmology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing 210002 China
| | - Hao Meng
- Department of Ophthalmology; Xijing Hospital; The Fourth Military Medical University; Xi'an China
- Department of Ophthalmology; NO.313 Hospital; Hulu Dao China
| | - Zhenping Huang
- Department of Ophthalmology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing 210002 China
| |
Collapse
|
27
|
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
|
28
|
Zhao Z, Yang C, Wang L, Li L, Zhao T, Hu L, Rong R, Xu M, Zhu T. The regulatory T cell effector soluble fibrinogen-like protein 2 induces tubular epithelial cell apoptosis in renal transplantation. Exp Biol Med (Maywood) 2014; 239:193-201. [PMID: 24414480 DOI: 10.1177/1535370213514921] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute rejection (AR) hinders renal allograft survival. Tubular epithelial cell (TEC) apoptosis contributes to premature graft loss in AR, while the mechanism remains unclear. Soluble fibrinogen-like protein 2 (sFGL2), a novel effector of regulatory T cells (Treg), induces apoptosis to mediate tissue injury. We previously found that serum sFGL2 significantly increased in renal allograft rejection patients. In this study, the role of sFGL2 in AR was further investigated both in vivo and in vitro. The serum level of sFGL2 and the percentage of CD4(+)CD25(+)Foxp3(+) Treg in the peripheral blood were measured in renal allograft recipients with AR or stable renal function (n = 30 per group). The human TEC was stimulated with sFGL2, tumor necrosis factor (TNF)-α, or phosphate buffered saline and investigated for apoptosis in vitro. Apoptosis-associated genes expression in TEC was further assessed. Approval for this study was obtained from the Ethics Committee of Fudan University. Our results showed that the serum level of sFGL2, correlated with Treg in the peripheral blood, was significantly increased in the AR patients. In vitro, sFGL2 remarkably induced TEC apoptosis, with a significant up-regulation of proapoptotic genes, including CASP-3, CASP-8, CASP-9, CASP-10, TRADD, TNFSF10, FADD, FAS, FASLG, BAK1, BAD, BAX, and NF-KB1. However, no significant changes were observed in the expression of antiapoptotic genes, including CARD-18, NAIP, BCL2, IKBKB, and TBK1. Therefore, sFGL2, an effector of Treg, induces TEC apoptosis. Our study suggests that sFGL2 is a potential mediator in the pathogenesis of allograft rejection and provides novel insights into the role of Treg in AR.
Collapse
Affiliation(s)
- Zitong Zhao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhao Z, Yang C, Tang Q, Zhao T, Jia Y, Ma Z, Rong R, Xu M, Zhu T. Serum level of soluble fibrinogen-like protein 2 in renal allograft recipients with acute rejection: a preliminary study. Transplant Proc 2013. [PMID: 23195010 DOI: 10.1016/j.transproceed.2012.05.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Soluble fibrinogen-like protein 2 (sfgl2), which is mainly secreted by T cells, is a novel effector of regulatory T cells with immunosuppressive functions. The aim of this study was to investigate serum levels of sfgl2 among renal allograft recipients. METHODS From November 2010 to August 2011 we retrospectively divided 47 renal allograft recipients into an acute rejection (n = 19) versus a stable group (n = 28) according to allograft biopsy results, using the Banff 2007 classification. The acute rejection group was subdivided into grade I (n = 8) versus grade II T-cell-mediated (n = 6) or antibody-mediated rejection episodes (n = 5). Peripheral blood samples were collected at the time of biopsy. Fourteen healthy volunteers were included as normal group controls. Serum levels of sfgl2 were analyzed by enzyme-linked immunosorbent assay. RESULTS Serum levels of sfgl2 were increased among renal allograft recipients suffering from biopsy-proven acute rejection episodes (61.91 ± 45.68 ng/mL), versus those with stable allografts (38.59 ± 19.92 ng/mL, P < .05) or healthy volunteers (29.10 ± 18.08 ng/mL, P < .05). The sfgl2 level was significantly higher among patients with antibody-mediated (118.48 ± 55.54 ng/mL) than T-cell-mediated acute rejection episodes (41.71 ± 16.44 ng/mL, P < .01). Serum sfgl2 levels were remarkably elevated in patients with grade II (51.87 ± 19.13 ng/mL) versus grade I T-cell-mediated rejection (34.10 ± 9.26 ng/mL, P < .05). CONCLUSIONS Serum sfgl2 levels were increased among renal allograft recipients with acute rejection episodes to an extent dependent upon the pathological type and severity of the response.
Collapse
Affiliation(s)
- Z Zhao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yuan Q, Hong S, Shi B, Kers J, Li Z, Pei X, Xu L, Wei X, Cai M. CD4(+)CD25(-)Nrp1(+) T cells synergize with rapamycin to prevent murine cardiac allorejection in immunocompetent recipients. PLoS One 2013; 8:e61151. [PMID: 23577203 PMCID: PMC3618334 DOI: 10.1371/journal.pone.0061151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/06/2013] [Indexed: 12/18/2022] Open
Abstract
Besides CD4+CD25+Foxp3+ regulatory T cells (Tregs), other immunosuppressive T cells also participated in the regulation of immune tolerance. Reportedly, neuropilin-1 (Nrp1) might be one of the molecules by which regulatory cells exert their suppressive effects. Indeed, CD4+CD25−Nrp1+ T cells exhibit potent suppressive function in autoimmune inflammatory responses. Here we investigated the specific role of CD4+CD25−Nrp1+ T cells in the setting of the transplant immune response. Through MLR assays, we found that CD4+CD25−Nrp1+ T cells suppressed the proliferation of naive CD4+CD25− T cells activated by allogeneic antigen-stimulation. Adoptive transfer of CD4+CD25−Nrp1+ T cells synergized with rapamycin to induce long-term graft survival in fully MHC-mismatched murine heart transplantation, which was associated with decreased IFN-γ, IL-17 and increased IL-10, TGF-β, Foxp3 and Nrp1 expression in the grafts. Importantly, our data indicated that CD4+CD25−Nrp1+ T cell transfer augments the accumulation of Tregs in the recipient, and creates conditions that favored induction of hyporesponsiveness of the T effector cells. In conclusion, this translational study indicates the possible therapeutic potential of CD4+CD25−Nrp1+ T cells in preventing allorejection. CD4+Nrp1+ T cells might therefore be used in bulk as a population of immunosuppressive cells with more beneficial properties concerning ex vivo isolation as compared to Foxp3+ Tregs.
Collapse
Affiliation(s)
- Qing Yuan
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Shanjuan Hong
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | - Bingyi Shi
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Jesper Kers
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Zhouli Li
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Xiangke Pei
- Organ Transplant Center, The 281th Hospital of PLA, Qinhuangdao, Hebei, China
| | - Liang Xu
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Xing Wei
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Ming Cai
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
- * E-mail:
| |
Collapse
|
31
|
Chandrasekharan D, Issa F, Wood KJ. Achieving operational tolerance in transplantation: how can lessons from the clinic inform research directions? Transpl Int 2013; 26:576-89. [PMID: 23517251 DOI: 10.1111/tri.12081] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 11/23/2012] [Accepted: 02/04/2013] [Indexed: 01/03/2023]
Abstract
Since the first solid organ transplant between the Herrick twins in 1954, transplantation immunology has sought to move away from harmful immunosuppressive regimens towards tolerogenic strategies that promote long-term graft survival. This has required a concerted multinational effort with scientists and clinicians working towards a common goal. Reports of immunosuppression-free kidney and liver allograft recipients have provided the proof-of-principle, but intentional generation of tolerance in clinical transplantation is still only achieved infrequently. Recently, there have been an increasing number of encouraging developments in the field in both experimental and clinical studies. In this article, we review the latest advances in tolerance research and consider possible future barriers and solutions in achieving reliable graft acceptance in the long term.
Collapse
Affiliation(s)
- Deepak Chandrasekharan
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
32
|
Dummer CD, Carpio VN, da Silva Loreto M, Joelsons G, Carraro DM, Olivieri ER, Manfro RC, Gonçalves LFS, Veronese FV. Analysis of FOXP3 gene and protein expressions in renal allograft biopsies and their association with graft outcomes. Ren Fail 2013; 35:521-30. [PMID: 23438049 DOI: 10.3109/0886022x.2013.766568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The transcription factor FOXP3 is increased in acute renal rejection, but its influence on graft outcomes is unclear. This study correlated FOXP3 with dendritic cells and graft outcomes. METHODS We assessed 96 kidney transplants undergoing allograft biopsy for cause. FOXP3 mRNA was analyzed by real-time polymerase chain reaction (PCR) and FOXP3 protein and DCsCD83(+) by immunohistochemistry. Graft function and survival were assessed at 5 years post-transplantation, as well as by independent predictors of graft loss. RESULTS Intragraft FOXP3 gene and protein expression were significantly correlated (r = 0.541, p < 0.001). Both FOXP3 mRNA and protein were increased in patients with acute rejection (AR). High expression of FOXP3 mRNA or protein in biopsies did not correlate with clinical variables, but there was a trend to higher positive variation in the glomerular filtration rate (GFR) from biopsy to last follow-up. Patients with FOXP3-mRNA(high) had more DCsCD83(+) in biopsy, but these cells did not associate with AR. Five-year graft survival was not influenced by either FOXP3 mRNA or protein expressions. CONCLUSIONS FOXP3 mRNA and protein had a good correlation in archival renal graft tissue. Increased FOXP3 expression was found in AR and FOXP3 associated with high numbers of DCs. However, both FOXP3 mRNA and protein was not associated with better allograft outcomes.
Collapse
Affiliation(s)
- Claus Dieter Dummer
- Post-Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cantaluppi V, Biancone L, Quercia A, Deregibus MC, Segoloni G, Camussi G. Rationale of mesenchymal stem cell therapy in kidney injury. Am J Kidney Dis 2012; 61:300-9. [PMID: 22938846 DOI: 10.1053/j.ajkd.2012.05.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 05/23/2012] [Indexed: 01/06/2023]
Abstract
Numerous preclinical and clinical studies suggest that mesenchymal stem cells, also known as multipotent mesenchymal stromal cells (MSCs), may improve pathologic conditions involving different organs. These beneficial effects initially were ascribed to the differentiation of MSCs into organ parenchymal cells. However, at least in the kidney, this is a very rare event and the kidney-protective effects of MSCs have been attributed mainly to paracrine mechanisms. MSCs release a number of trophic, anti-inflammatory, and immune-modulatory factors that may limit kidney injury and favor recovery. In this article, we provide an overview of the biologic activities of MSCs that may be relevant for the treatment of kidney injury in the context of a case vignette concerning a patient at high immunologic risk who underwent a second kidney transplantation followed by the development of ischemia-reperfusion injury and acute allograft rejection. We discuss the possible beneficial effect of MSC treatment in the light of preclinical and clinical data supporting the regenerative and immunomodulatory potential of MSCs.
Collapse
Affiliation(s)
- Vincenzo Cantaluppi
- Nephrology, Dialysis and Renal Transplantation Unit, Centre for Experimental Medical Research (CeRMS) and Department of Internal Medicine, University of Torino, Torino, Italy
| | | | | | | | | | | |
Collapse
|
34
|
Zhang X, Han S, Kang Y, Guo M, Hong S, Liu F, Fu S, Wang L, Wang QX. SAHA, an HDAC inhibitor, synergizes with tacrolimus to prevent murine cardiac allograft rejection. Cell Mol Immunol 2012; 9:390-8. [PMID: 22922441 DOI: 10.1038/cmi.2012.28] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA), as a histone deacetylase (HDAC) inhibitor (HDACi), was recently found to exhibit an immunosuppressive effect. However, whether SAHA can synergize with calcineurin inhibitors (CNIs) to inhibit allograft rejection and its underlying mechanism remain elusive. In this study, we demonstrated the synergistic effects of SAHA and non-therapeutic dose of tacrolimus (FK506) in prolonging the allograft survival in a murine cardiac transplant model. Concomitant intragraft examination revealed that allografts from SAHA-treated recipients showed significantly lower levels of IL-17 expression, and no discernable difference for IL-17 expressions was detected between SAHA- and SAHA/FK506-treated allograft as compared with allografts from FK506-treated animals. In contrast, administration of FK506 significantly suppressed interferon (IFN)-γ but increased IL-10 expression as compared with that of SAHA-treated animals, and this effect was independent of SAHA. Interestingly, SAHA synergizes with FK506 to promote Foxp3 and CTLA4 expression. In vitro, SAHA reduced the proportion of Th17 cells in isolated CD4⁺ T-cell population and decreased expressions of IL-17A, IL-17F, STAT3 and RORγt in these cells. Moreover, SAHA enhances suppressive function of regulatory T (Treg) cells by upregulating the expression of CTLA-4 without affecting T effector cell proliferation, and increased the proportion of Treg by selectively promoting apoptosis of T effector cells. Therefore, SAHA, a HDACi, may be a promising immunosuppressive agent with potential benefit in conjunction with CNI drugs.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Organ Transplantation, Changzheng Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
FOXP3+ regulatory T cells: from suppression of rejection to induction of renal allograft tolerance. Transpl Immunol 2011; 26:1-10. [PMID: 21939765 DOI: 10.1016/j.trim.2011.08.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 08/31/2011] [Accepted: 08/31/2011] [Indexed: 12/21/2022]
Abstract
Naturally occurring and induced regulatory T cells (Tregs) can become hyporesponsive and anergic to antigen stimulation in autoimmune diseases and allograft rejection. The mechanisms of suppression of effector T cells by Tregs remain unclear, but there are in vitro and in vivo evidences showing that these cells are able to suppress antigen-specific responses via direct cell-to-cell contact, secrete anti-inflammatory cytokines such as TGF-β and IL-10, and inhibit the generation of memory T cells, among others. The transcription factor FOXP3 is a specific marker of Tregs and its deficiency is associated with autoimmune diseases and inflammation. During acute rejection of kidney allografts, an augmented FOXP3 gene expression as well as increased CD4(+)CD25(+)FOXP3(+) and other cell populations are observed in graft biopsies. However, it is not clear whether Tregs migrate into the graft and are retained there to suppress the inflammatory process, or whether they are directly associated with more complex mechanisms to induce immune tolerance. FOXP3(+) Tregs may direct the immune response toward a graft acceptance program, potentially affecting the long-term survival of transplanted organs and tissues. Immunosuppressive drugs modulate the number and function of circulating Tregs and FOXP3 expression. Experimental and clinical studies have shown that mTOR inhibitors have positive and calcineurin inhibitors negative effects on Tregs, but it is difficult to set apart the effect of multiple other factors known to be associated with short- and long-term renal graft outcomes. This review aimed to describe the functions of Tregs and its transcription factor FOXP3 in suppression of immune response during rejection and in induction of kidney graft tolerance, as well as to review the individual effects of immunosuppressive drugs on Tregs.
Collapse
|