1
|
Yemaneberhan KH, Kang M, Jang JH, Kim JH, Kim KS, Park HB, Choi D. Beyond the icebox: modern strategies in organ preservation for transplantation. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:377-403. [PMID: 39743232 PMCID: PMC11732768 DOI: 10.4285/ctr.24.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 01/04/2025]
Abstract
Organ transplantation, a critical treatment for end-stage organ failure, has witnessed significant advancements due to the integration of improved surgical techniques, immunosuppressive therapies, and donor-recipient matching. This review explores the progress of organ preservation, focusing on the shift from static cold storage (SCS) to advanced machine perfusion techniques such as hypothermic (HMP) and normothermic machine perfusion (NMP). Although SCS has been the standard approach, its limitations in preserving marginal organs and preventing ischemia-reperfusion injury (IRI) have led to the adoption of HMP and NMP. HMP, which is now the gold standard for high-risk donor kidneys, reduces metabolic activity and improves posttransplant outcomes. NMP allows real-time organ viability assessment and reconditioning, especially for liver transplants. Controlled oxygenated rewarming further minimizes IRI by addressing mitochondrial dysfunction. The review also highlights the potential of cryopreservation for long-term organ storage, despite challenges with ice formation. These advances are crucial for expanding the donor pool, improving transplant success rates, and addressing organ shortages. Continued innovation is necessary to meet the growing demands of transplantation and save more lives.
Collapse
Affiliation(s)
- Kidus Haile Yemaneberhan
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| | - Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Jun Hwan Jang
- Department of Energy Engineering, Hanyang University, Seoul, Korea
| | - Jin Hee Kim
- Department of Energy Engineering, Hanyang University, Seoul, Korea
| | - Kyeong Sik Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Ho Bum Park
- Department of Energy Engineering, Hanyang University, Seoul, Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
2
|
Pottebaum AA, January SE, Liu C, Lavine S, Schilling JD, Lavine KJ. Feasibility of Interleukin-6 Receptor Blockade in Cardiac Antibody-mediated Rejection. Transplantation 2024; 108:539-544. [PMID: 37638881 PMCID: PMC10798586 DOI: 10.1097/tp.0000000000004784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/15/2023] [Accepted: 06/28/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Antibody-mediated rejection (AMR) remains a significant cause of heart transplant mortality with few effective therapies. METHODS This study aimed to describe initial experience of using interleukin-6 receptor blockade with tocilizumab in the treatment of acute cardiac AMR at Barnes-Jewish Hospital/Washington University Transplant Center from July 2017 to May 2021 (n = 7). Clinical, echocardiographic, and serum alloantibody data were analyzed before and after treatment. RESULTS All participants demonstrated marked improvement in functional status. Echocardiographic data following 4-6 mo of tocilizumab revealed significant improvements in biventricular systolic function for all participants. Consistent reductions in donor-specific HLA or angiotensin type I receptor antibodies were not observed, suggesting that tocilizumab may act downstream of antibody production. No patient experienced drug-related complications that necessitated discontinuation of therapy. CONCLUSIONS These findings provide initial insights into the safety and efficacy of interleukin-6 receptor blockade in the treatment of cardiac AMR and support the design of larger prospective studies.
Collapse
Affiliation(s)
| | | | - Chang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Steven Lavine
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Joel D Schilling
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
3
|
Praska CE, Tamburrini R, Danobeitia JS. Innate immune modulation in transplantation: mechanisms, challenges, and opportunities. FRONTIERS IN TRANSPLANTATION 2023; 2:1277669. [PMID: 38993914 PMCID: PMC11235239 DOI: 10.3389/frtra.2023.1277669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/23/2023] [Indexed: 07/13/2024]
Abstract
Organ transplantation is characterized by a sequence of steps that involve operative trauma, organ preservation, and ischemia-reperfusion injury in the transplant recipient. During this process, the release of damage-associated molecular patterns (DAMPs) promotes the activation of innate immune cells via engagement of the toll-like receptor (TLR) system, the complement system, and coagulation cascade. Different classes of effector responses are then carried out by specialized populations of macrophages, dendritic cells, and T and B lymphocytes; these play a central role in the orchestration and regulation of the inflammatory response and modulation of the ensuing adaptive immune response to transplant allografts. Organ function and rejection of human allografts have traditionally been studied through the lens of adaptive immunity; however, an increasing body of work has provided a more comprehensive picture of the pivotal role of innate regulation of adaptive immune responses in transplant and the potential therapeutic implications. Herein we review literature that examines the repercussions of inflammatory injury to transplantable organs. We highlight novel concepts in the pathophysiology and mechanisms involved in innate control of adaptive immunity and rejection. Furthermore, we discuss existing evidence on novel therapies aimed at innate immunomodulation and how this could be harnessed in the transplant setting.
Collapse
Affiliation(s)
- Corinne E. Praska
- Division of Transplantation, Department of Surgery, University of Wisconsin, Madison, WI, United States
| | - Riccardo Tamburrini
- Division of Transplantation, Department of Surgery, University of Wisconsin, Madison, WI, United States
| | - Juan Sebastian Danobeitia
- Division of Transplantation, Department of Surgery, University of Wisconsin, Madison, WI, United States
- Baylor Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, United States
| |
Collapse
|
4
|
Cassano A, Chong AS, Alegre ML. Tregs in transplantation tolerance: role and therapeutic potential. FRONTIERS IN TRANSPLANTATION 2023; 2:1217065. [PMID: 38993904 PMCID: PMC11235334 DOI: 10.3389/frtra.2023.1217065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/14/2023] [Indexed: 07/13/2024]
Abstract
CD4+ Foxp3+ regulatory T cells (Tregs) are indispensable for preventing autoimmunity, and they play a role in cancer and transplantation settings by restraining immune responses. In this review, we describe evidence for the importance of Tregs in the induction versus maintenance of transplantation tolerance, discussing insights into mechanisms of Treg control of the alloimmune response. Further, we address the therapeutic potential of Tregs as a clinical intervention after transplantation, highlighting engineered CAR-Tregs as well as expansion of donor and host Tregs.
Collapse
Affiliation(s)
- Alexandra Cassano
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Anita S. Chong
- Department of Surgery, University of Chicago, Chicago, IL, United States
| | - Maria-Luisa Alegre
- Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
See Hoe LE, Li Bassi G, Wildi K, Passmore MR, Bouquet M, Sato K, Heinsar S, Ainola C, Bartnikowski N, Wilson ES, Hyslop K, Skeggs K, Obonyo NG, Shuker T, Bradbury L, Palmieri C, Engkilde-Pedersen S, McDonald C, Colombo SM, Wells MA, Reid JD, O'Neill H, Livingstone S, Abbate G, Haymet A, Jung JS, Sato N, James L, He T, White N, Redd MA, Millar JE, Malfertheiner MV, Molenaar P, Platts D, Chan J, Suen JY, McGiffin DC, Fraser JF. Donor heart ischemic time can be extended beyond 9 hours using hypothermic machine perfusion in sheep. J Heart Lung Transplant 2023; 42:1015-1029. [PMID: 37031869 DOI: 10.1016/j.healun.2023.03.020] [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: 07/12/2022] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND The global shortage of donor hearts available for transplantation is a major problem for the treatment of end-stage heart failure. The ischemic time for donor hearts using traditional preservation by standard static cold storage (SCS) is limited to approximately 4 hours, beyond which the risk for primary graft dysfunction (PGD) significantly increases. Hypothermic machine perfusion (HMP) of donor hearts has been proposed to safely extend ischemic time without increasing the risk of PGD. METHODS Using our sheep model of 24 hours brain death (BD) followed by orthotopic heart transplantation (HTx), we examined post-transplant outcomes in recipients following donor heart preservation by HMP for 8 hours, compared to donor heart preservation for 2 hours by either SCS or HMP. RESULTS Following HTx, all HMP recipients (both 2 hours and 8 hours groups) survived to the end of the study (6 hours after transplantation and successful weaning from cardiopulmonary bypass), required less vasoactive support for hemodynamic stability, and exhibited superior metabolic, fluid status and inflammatory profiles compared to SCS recipients. Contractile function and cardiac damage (troponin I release and histological assessment) was comparable between groups. CONCLUSIONS Overall, compared to current clinical SCS, recipient outcomes following transplantation are not adversely impacted by extending HMP to 8 hours. These results have important implications for clinical transplantation where longer ischemic times may be required (e.g., complex surgical cases, transport across long distances). Additionally, HMP may allow safe preservation of "marginal" donor hearts that are more susceptible to myocardial injury and facilitate increased utilization of these hearts for transplantation.
Collapse
Affiliation(s)
- Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia.
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Uniting Care Hospitals, Intensive Care Units St Andrew's War Memorial Hospital and The Wesley Hospital, Brisbane, Queensland, Australia; Wesley Medical Research, Brisbane, Queensland, Australia; Queensland University of Technology, Brisbane, Queensland, Australia
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Cardiovascular Research Institute Basel, Basel, Switzerland
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Mahe Bouquet
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Department of Intensive Care, North Estonia Medical Centre, Tallinn, Estonia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Nicole Bartnikowski
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Queensland, Australia
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Kris Skeggs
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Nchafatso G Obonyo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Wellcome Trust Centre for Global Health Research, Imperial College London, London, United Kingdom; Initiative to Develop African Research Leaders (IDeAL), Kilifi, Kenya
| | - Tristan Shuker
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Lucy Bradbury
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Chiara Palmieri
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, Queensland, Australia
| | | | - Charles McDonald
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Department of Anesthesia and Perfusion, The Prince Charles Hospital, Queensland, Australia
| | - Sebastiano M Colombo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Matthew A Wells
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | - Janice D Reid
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Hollier O'Neill
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Samantha Livingstone
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Andrew Haymet
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jae-Seung Jung
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Lynnette James
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ting He
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Nicole White
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Meredith A Redd
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Institute for Molecular Bioscience, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jonathan E Millar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Maximillian V Malfertheiner
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Department of Internal Medicine II, Cardiology and Pneumology, University Medical Center Regensburg, Regensburg, Germany
| | - Peter Molenaar
- Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - David Platts
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jonathan Chan
- School of Medicine, Griffith University, Southport, Queensland, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David C McGiffin
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, Victoria, Australia; Monash University, Melbourne, Victoria, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia; Prince Charles Hospital Northside Clinical Unit, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Habibabady Z, McGrath G, Kinoshita K, Maenaka A, Ikechukwu I, Elias GF, Zaletel T, Rosales I, Hara H, Pierson RN, Cooper DKC. Antibody-mediated rejection in xenotransplantation: Can it be prevented or reversed? Xenotransplantation 2023; 30:e12816. [PMID: 37548030 PMCID: PMC11101061 DOI: 10.1111/xen.12816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Antibody-mediated rejection (AMR) is the commonest cause of failure of a pig graft after transplantation into an immunosuppressed nonhuman primate (NHP). The incidence of AMR compared to acute cellular rejection is much higher in xenotransplantation (46% vs. 7%) than in allotransplantation (3% vs. 63%) in NHPs. Although AMR in an allograft can often be reversed, to our knowledge there is no report of its successful reversal in a pig xenograft. As there is less experience in preventing or reversing AMR in models of xenotransplantation, the results of studies in patients with allografts provide more information. These include (i) depletion or neutralization of serum anti-donor antibodies, (ii) inhibition of complement activation, (iii) therapies targeting B or plasma cells, and (iv) anti-inflammatory therapy. Depletion or neutralization of anti-pig antibody, for example, by plasmapheresis, is effective in depleting antibodies, but they recover within days. IgG-degrading enzymes do not deplete IgM. Despite the expression of human complement-regulatory proteins on the pig graft, inhibition of systemic complement activation may be necessary, particularly if AMR is to be reversed. Potential therapies include (i) inhibition of complement activation (e.g., by IVIg, C1 INH, or an anti-C5 antibody), but some complement inhibitors are not effective in NHPs, for example, eculizumab. Possible B cell-targeted therapies include (i) B cell depletion, (ii) plasma cell depletion, (iii) modulation of B cell activation, and (iv) enhancing the generation of regulatory B and/or T cells. Among anti-inflammatory agents, anti-IL6R mAb and TNF blockers are increasingly being tested in xenotransplantation models, but with no definitive evidence that they reverse AMR. Increasing attention should be directed toward testing combinations of the above therapies. We suggest that treatment with a systemic complement inhibitor is likely to be most effective, possibly combined with anti-inflammatory agents (if these are not already being administered). Ultimately, it may require further genetic engineering of the organ-source pig to resolve the problem entirely, for example, knockout or knockdown of SLA, and/or expression of PD-L1, HLA E, and/or HLA-G.
Collapse
Affiliation(s)
- Zahra Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ileka Ikechukwu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela F. Elias
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Tjasa Zaletel
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Hara
- Yunnan Xenotransplantation Engineering Research Center, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Miller CL, Madsen JC. Targeting IL-6 to prevent cardiac allograft rejection. Am J Transplant 2022; 22 Suppl 4:12-17. [PMID: 36453706 PMCID: PMC10191185 DOI: 10.1111/ajt.17206] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 12/05/2022]
Abstract
Outcomes following heart transplantation remain suboptimal with acute and chronic rejection being major contributors to poor long-term survival. IL-6 is increasingly recognized as a critical pro-inflammatory cytokine involved in allograft injury and has been shown to play a key role in regulating the inflammatory and alloimmune responses following heart transplantation. Therapies that inhibit IL-6 signaling have emerged as promising strategies to prevent allograft rejection. Here, we review experimental and pre-clinical evidence that supports the potential use of IL-6 signaling blockade to improve outcomes in heart transplant recipients.
Collapse
Affiliation(s)
- Cynthia L. Miller
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joren C. Madsen
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
8
|
van Vugt LK, Schagen MR, de Weerd A, Reinders ME, de Winter BC, Hesselink DA. Investigational drugs for the treatment of kidney transplant rejection. Expert Opin Investig Drugs 2022; 31:1087-1100. [PMID: 36175360 DOI: 10.1080/13543784.2022.2130751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Kidney transplant rejection remains an important clinical problem despite the development of effective immunosuppressive drug combination therapy. Two major types of rejection are recognized, namely T-cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR), which have a different pathophysiology and are treated differently. Unfortunately, long-term outcomes of both TCMR and ABMR remain unsatisfactory despite current therapy. Hence, alternative therapeutic drugs are urgently needed. AREAS COVERED This review covers novel and investigational drugs for the pharmacological treatment of kidney transplant rejection. Potential therapeutic strategies and future directions are discussed. EXPERT OPINION The development of alternative pharmacologic treatment of rejection has focused mostly on ABMR, since this is the leading cause of kidney allograft loss and currently lacks an effective, evidence-based therapy. At present, there is insufficient high-quality evidence for any of the covered investigational drugs to support their use in ABMR. However, with the emergence of targeted therapies, this potential arises for individualized treatment strategies. In order to generate more high-quality evidence for such strategies and overcome the obstacles of classic, randomized, controlled trials, we advocate the implementation of adaptive trial designs and surrogate clinical endpoints. We believe such adaptive trial designs could help to understand the risks and benefits of promising drugs such as tocilizumab, clazakizumab, belimumab, and imlifidase.
Collapse
Affiliation(s)
- Lukas K van Vugt
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maaike R Schagen
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annelies de Weerd
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marlies Ej Reinders
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda Cm de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
9
|
Kim SJ, Kim JY, Kim EJ, Lee H, Cho Y, Kim MS, Kim YS, Kim BS, Huh KH. The Immunomodulating Effects of Thalidomide and Dexamethasone in a Murine Cardiac Allograft Transplantation Model. Yonsei Med J 2022; 63:158-165. [PMID: 35083901 PMCID: PMC8819408 DOI: 10.3349/ymj.2022.63.2.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 12/03/2022] Open
Abstract
PURPOSE The immunomodulatory effects of thalidomide (TM) and dexamethasone (DX) on immune cells and their co-stimulatory, co-inhibitory molecules in vitro and in vivo have been previously reported. The current study investigated the effects of TM and the combinatorial treatment with DX on immune cells using a murine cardiac allograft transplantation model. MATERIALS AND METHODS Intraabdominal transplant of cardiac allografts from BALB/c (H-2d) donors to C57BL/6 (H-2b) recipients was performed. After transplantation, mice were injected daily with TM or DX or a combination of both TM and DX (TM/DX) by intraperitoneal route until the time of graft loss. CD4+ T cell subsets and CD11c+ cells in the peripheral blood mononuclear cells and spleen were examined and quantified with flow cytometry. Serum IL-6 levels were measured by enzyme-linked immunosorbent assay on day 7. RESULTS The mean graft survivals were 6.86 days in the untreated group, and 10.0 days in the TM/DX group (p<0.001). The TM/DX treatment affected the CD4+ T cell subsets without suppressing the total CD4+ T cell population. The CD4+FOXP3+/CD4+CD44hi T cell ratio increased. Increase in cell counts and median fluorescence intensity on CD11c+CD85k+ with TM/DX were observed. The inhibition of pro-inflammatory cytokine interleukin-6 was also observed. CONCLUSION These outcomes suggest the immunomodulating effect of the TM/DX combinatorial treatment. In conclusion, TM/DX combination may be a promising immunomodulatory approach for preventing allograft rejection and improving graft survival by inducing tolerance in transplantation.
Collapse
Affiliation(s)
- Soo Jin Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Joon Ye Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jee Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojung Lee
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Yuri Cho
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Myoung Soo Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Seun Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
| | - Beom Seok Kim
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Kyu Ha Huh
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
- The Research Institute for Transplantation, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
10
|
Ma M, Sun Q, Li X, Deng G, Zhang Y, Yang Z, Han F, Huang Z, Fang Y, Liao T, Sun Q. Blockade of IL-6/IL-6R Signaling Attenuates Acute Antibody-Mediated Rejection in a Mouse Cardiac Transplantation Model. Front Immunol 2021; 12:778359. [PMID: 34777394 PMCID: PMC8581398 DOI: 10.3389/fimmu.2021.778359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Acute antibody-mediated rejection (AAMR) is an important cause of cardiac allograft dysfunction, and more effective strategies need to be explored to improve allograft prognosis. Interleukin (IL)-6/IL-6R signaling plays a key role in the activation of immune cells including B cells, T cells and macrophages, which participate in the progression of AAMR. In this study, we investigated the effect of IL-6/IL-6R signaling blockade on the prevention of AAMR in a mouse model. We established a mouse model of AAMR for cardiac transplantation via presensitization of skin grafts and addition of cyclosporin A, and sequentially analyzed its features. Tocilizumab, anti-IL-6R antibody, and recipient IL-6 knockout were used to block IL-6/IL-6R signaling. We demonstrated that blockade of IL-6/IL-6R signaling significantly attenuated allograft injury and improved survival. Further mechanistic research revealed that signaling blockade decreased B cells in circulation, spleens, and allografts, thus inhibiting donor-specific antibody production and complement activation. Moreover, macrophage, T cell, and pro-inflammatory cytokine infiltration in allografts was also reduced. Collectively, we provided a highly practical mouse model of AAMR and demonstrated that blockade of IL-6/IL-6R signaling markedly alleviated AAMR, which is expected to provide a superior option for the treatment of AAMR in clinic.
Collapse
Affiliation(s)
- Maolin Ma
- Organ Transplantation Research Institute, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qipeng Sun
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiujie Li
- Department of Obstetrics and Gynecology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gengguo Deng
- Organ Transplantation Research Institute, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yannan Zhang
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Zhe Yang
- Organ Transplantation Research Institute, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Fei Han
- Organ Transplantation Research Institute, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengyu Huang
- Organ Transplantation Research Institute, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Youqiang Fang
- Department of Urology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Liao
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Qiquan Sun
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Chandran S, Leung J, Hu C, Laszik ZG, Tang Q, Vincenti FG. Interleukin-6 blockade with tocilizumab increases Tregs and reduces T effector cytokines in renal graft inflammation: A randomized controlled trial. Am J Transplant 2021; 21:2543-2554. [PMID: 33331082 DOI: 10.1111/ajt.16459] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/11/2020] [Accepted: 12/11/2020] [Indexed: 01/25/2023]
Abstract
Interleukin-6 (IL-6) is a proinflammatory cytokine and key regulator of Treg: T effector cell (Teff) balance. We hypothesized that IL-6 blockade with tocilizumab, a monoclonal antibody to IL-6R, would increase Tregs, dampen Teff function, and control graft inflammation. We conducted a randomized controlled clinical trial (2014-2018) of clinically stable kidney transplant recipients on calcineurin inhibitor, mycophenolate mofetil, and prednisone, with subclinical graft inflammation noted on surveillance biopsies during the first year posttransplant. Subjects received tocilizumab (8 mg/kg IV every 4 weeks; 6 doses; n = 16) or no treatment (controls; n = 14) on top of usual maintenance immunosuppression. Kidney biopsies pre- and post-treatment were analyzed using Banff criteria. Blood was analyzed for serum cytokines, Treg frequencies, and T cell effector molecule expression (IFN-γ, IL-17, granzyme B) post-stimulation ex vivo. Tocilizumab-treated subjects were more likely to show improved Banff ti-score (62.5% vs. 21.4%, p = .03), increased Treg frequency (7.1% ± 5.55% vs. 3.6% ± 1.7%, p = .0168), and a blunted Teff cytokine response compared to controls. Changes in Banff i- and t-scores were not significantly different. The treatment was relatively well tolerated with no patient deaths or graft loss. Blockade of IL-6 is a novel and promising treatment option to regulate the T cell alloimmune response in kidney transplant recipients. NCT02108600.
Collapse
Affiliation(s)
- Sindhu Chandran
- Department of Medicine, University of California, San Francisco, California, USA
| | - Joey Leung
- Department of Surgery, University of California, San Francisco, California, USA
| | - Crystal Hu
- Department of Surgery, University of California, San Francisco, California, USA
| | - Zoltan G Laszik
- Department of Pathology, University of California, San Francisco, California, USA
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, California, USA
| | - Flavio G Vincenti
- Department of Medicine, University of California, San Francisco, California, USA.,Department of Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
12
|
Abstract
Purpose of Review IL-6 is a pleiotropic, pro-inflammatory cytokine that plays an integral role in the development of acute and chronic rejection after solid organ transplantation. This article reviews the experimental evidence and current clinical application of IL-6/IL-6 receptor (IL-6R) signaling inhibition for the prevention and treatment of allograft injury. Recent Findings There exists a robust body of evidence linking IL-6 to allograft injury mediated by acute inflammation, adaptive cellular/humoral responses, innate immunity, and fibrosis. IL-6 promotes the acute phase reaction, induces B cell maturation/antibody formation, directs cytotoxic T-cell differentiation, and inhibits regulatory T-cell development. Importantly, blockade of the IL-6/IL-6R signaling pathway has been shown to mitigate its harmful effects in experimental studies, particularly in models of kidney and heart transplant rejection. Currently, available agents for IL-6 signaling inhibition include monoclonal antibodies against IL-6 or IL-6R and janus kinase inhibitors. Recent clinical trials have investigated the use of tocilizumab, an anti-IL-6R mAb, for desensitization and treatment of antibody-mediated rejection (AMR) in kidney transplant recipients, with promising initial results. Further studies are underway investigating the use of alternative agents including clazakizumab, an anti-IL-6 mAb, and application of IL-6 signaling blockade to clinical cardiac transplantation. Summary IL-6/IL-6R signaling inhibition provides a novel therapeutic option for the prevention and treatment of allograft injury. To date, evidence from clinical trials supports the use of IL-6 blockade for desensitization and treatment of AMR in kidney transplant recipients. Ongoing and future clinical trials will further elucidate the role of IL-6 signaling inhibition in other types of solid organ transplantation.
Collapse
|
13
|
Hasgur S, Fan R, Zwick DB, Fairchild RL, Valujskikh A. B cell-derived IL-1β and IL-6 drive T cell reconstitution following lymphoablation. Am J Transplant 2020; 20:2740-2754. [PMID: 32342598 PMCID: PMC7956246 DOI: 10.1111/ajt.15960] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 01/25/2023]
Abstract
Understanding the mechanisms of T cell homeostatic expansion is crucial for clinical applications of lymphoablative therapies. We previously established that T cell recovery in mouse heart allograft recipients treated with anti-thymocyte globulin (mATG) critically depends on B cells and is mediated by B cell-derived soluble factors. B cell production of interleukin (IL)-1β and IL-6 is markedly upregulated after heart allotransplantation and lymphoablation. Neutralizing IL-1β or IL-6 with mAb or the use of recipients lacking mature IL-1β, IL-6, IL-1R, MyD88, or IL-6R impair CD4+ and CD8+ T cell recovery and significantly enhance the graft-prolonging efficacy of lymphoablation. Adoptive co-transfer experiments demonstrate a direct effect of IL-6 but not IL-1β on T lymphocytes. Furthermore, B cells incapable of IL-1β or IL-6 production have diminished capacity to mediate T cell reconstitution and initiate heart allograft rejection upon adoptive transfer into mATG treated B cell deficient recipients. These findings reveal the essential role of B cell-derived IL-1β and IL-6 during homeostatic T cell expansion in a clinically relevant model of lymphoablation.
Collapse
Affiliation(s)
- Suheyla Hasgur
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Ran Fan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Daniel B. Zwick
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
14
|
GGTA1/iGb3S Double Knockout Mice: Immunological Properties and Immunogenicity Response to Xenogeneic Bone Matrix. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9680474. [PMID: 32596401 PMCID: PMC7292995 DOI: 10.1155/2020/9680474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/05/2022]
Abstract
Background Animal tissues and tissue-derived biomaterials are widely used in the field of xenotransplantation and regenerative medicine. A potential immunogenic risk that affects the safety and effectiveness of xenografts is the presence of remnant α-Gal antigen (synthesized by GGTA1 or/and iGb3S). GGTA1 knockout mice have been developed as a suitable model for the analysis of anti-Gal antibody-mediated immunogenicity. However, we are yet to establish whether GGTA1/iGb3S double knockout (G/i DKO) mice are sensitive to Gal antigen-positive xenoimplants. Methods α-Gal antigen expression in the main organs of G/i DKO mice or bovine bone substitutes was detected via a standardized ELISA inhibition assay. Serum anti-α-Gal antibody titers of G/i DKO mice after immunization with rabbit red blood cells (RRBC) and implantation of raw lyophilized bone substitutes (Gal antigen content was 8.14 ± 3.17 × 1012/mg) or Guanhao Biotech bone substitutes (50% decrease in Gal antigen relative to the raw material) were assessed. The evaluation of total serum antibody, inflammatory cytokine, and splenic lymphocyte subtype populations and the histological analysis of implants and thymus were performed to systematically assess the immune response caused by bovine bone substitutes and bone substitute grafts in G/i DKO mice. Results α-Gal epitope expression was reduced by 100% in the main organs of G/i DKO mice, compared with their wild-type counterparts. Following immunization with RRBC, serum anti-Gal antibody titers of G/i DKO mice increased from 80- to 180-fold. After subcutaneous implantation of raw lyophilized bone substitutes and Guanhao Biotech bone substitutes into G/i DKO mice, specific anti-α-Gal IgG, anti-α-Gal IgM, and related inflammatory factors (IFN-γ and IL-6) were significantly increased in the raw lyophilized bone substitute group but showed limited changes in the Guanhao Biotech bone substitute group, compared with the control. Conclusion G/i DKO mice are sensitive to Gal antigen-positive xenogeneic grafts and can be effectively utilized for evaluating the α-Gal-mediated immunogenic risk of xenogeneic grafts.
Collapse
|
15
|
Yi L, Chen Y, Jin Q, Deng C, Wu Y, Li H, Liu T, Li Y, Yang Y, Wang J, Lv Q, Zhang L, Xie M. Antagomir-155 Attenuates Acute Cardiac Rejection Using Ultrasound Targeted Microbubbles Destruction. Adv Healthc Mater 2020; 9:e2000189. [PMID: 32548962 DOI: 10.1002/adhm.202000189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/27/2020] [Indexed: 12/22/2022]
Abstract
Antagomir-155 is an artificial inhibitor of miRNA-155, which is expected to be a promising therapeutic target to attenuate acute cardiac rejection (ACR). However, its vulnerability of being degraded by endogenous nuclease and potential off-target effect make the authors seek for a more suitable way to delivery it. In attribution of efficiency and safety, ultrasound targeted microbubbles destruction (UTMD) turns out to be an appropriate method to deliver gene to target tissues. Here, cationic microbubbles to deliver antagomir-155 downregulating miRNA-155 in murine allograft hearts triggered by UTMD are synthesized. The viability of this therapy is verified by fluorescent microscopy. The biodistribution of antagomir-155 is analyzed by optical imaging system. The results show antagomir-155 delivered by UTMD which significantly decreases the levels of miR-155. Also, this therapy downregulates the expression of cytokines and inflammation infiltration. And allograft survival time is significantly prolonged. Therefore, antagomir-loaded microbubbles trigged by UTMD may provide a novel platform for ACR target treatment.
Collapse
Affiliation(s)
- Luyang Yi
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yihan Chen
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Qiaofeng Jin
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Cheng Deng
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Ya Wu
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Huiling Li
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Tianshu Liu
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yuman Li
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Yali Yang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Jing Wang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Qing Lv
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Li Zhang
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| | - Mingxing Xie
- Department of UltrasoundUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology 1277 Jiefang Avenue Wuhan 430022 China
- Hubei Province Key Laboratory of Molecular Imaging 13 Hangkong Road Wuhan 430030 China
| |
Collapse
|
16
|
Zhang G, Iwase H, Wang L, Yamamoto T, Jagdale A, Ayares D, Li Y, Cooper DKC, Hara H. Is interleukin-6 receptor blockade (tocilizumab) beneficial or detrimental to pig-to-baboon organ xenotransplantation? Am J Transplant 2020; 20:999-1013. [PMID: 31733178 DOI: 10.1111/ajt.15712] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/08/2019] [Accepted: 10/29/2019] [Indexed: 01/25/2023]
Abstract
The interleukin (IL)-6/IL-6 receptor-α (IL-6Rα)/signal transduction and activation of the transcription 3 (STAT3) pathway plays an important role in inflammation. Anti-human IL-6Rα blockade by tocilizumab (TCZ) has been used in pig-to-baboon organ xenotransplant models, but whether it is beneficial remains uncertain. After xenotransplant, there were significant increases in both baboon and pig IL-6 in the baboon serum, especially in baboons that received TCZ before xenotransplant. In vitro observations demonstrated that human, baboon, and pig IL-6 can activate the IL-6/IL-6Rα/STAT3 pathway in human, baboon, and pig cells, respectively. Activation of the IL-6/IL-6Rα/STAT3 pathway was blocked by TCZ in human and baboon cells but not in pig cells (ie, pig IL-6R). Siltuximab (human IL-6 inhibitor) bound to both human and baboon, but not pig, IL-6 and suppressed activation of the IL-6/IL-6Rα/STAT3 pathway. These results indicate that TCZ and siltuximab do not cross-react with pig IL-6R and pig IL-6, respectively. Rapamycin partially inhibited human, baboon, and pig IL-6/IL-6Rα/STAT3 pathways and suppressed inflammatory gene expression. TCZ treatment increased serum IL-6 because it could no longer bind to baboon IL-6Rα. We suggest that increased serum IL-6 may be detrimental to the pig xenograft because it is likely to bind to pig IL-6R, resulting in activation of pig cells.
Collapse
Affiliation(s)
- Guoqiang Zhang
- Department of General Surgery, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Liaoran Wang
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Takayuki Yamamoto
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abhijit Jagdale
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Yong Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
17
|
Cao P, Sun Z, Feng C, Zhang J, Zhang F, Wang W, Zhao Y. Myeloid-derived suppressor cells in transplantation tolerance induction. Int Immunopharmacol 2020; 83:106421. [PMID: 32217462 DOI: 10.1016/j.intimp.2020.106421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous cells derived from bone marrow. These cells are developed from immature myeloid cells and have strong negative immunomodulatory effects. In the context of pathology (such as tumor, autoimmune disease, trauma, and burns), MDSCs accumulate around tumor and inflammatory tissues, where their main role is to inhibit the function of effector T cells and promote the recruitment of regulatory T cells. MDSCs can be used in organ transplantation to regulate the immune responses that participate in rejection of the transplanted organ. This effect is achieved by increasing the production of MDSCs in vivo or transfusion of MDSCs induced in vitro to establish immune tolerance and prolong the survival of the graft. In this review, we discuss the efficacy of MDSCs in a variety of transplantation studies as well as the induction of immune tolerance to prevent transplant rejection through the use of common clinical immunosuppressants combined with MDSCs.
Collapse
Affiliation(s)
- Peng Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chang Feng
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
18
|
Uehara M, Bahmani B, Jiang L, Jung S, Banouni N, Kasinath V, Solhjou Z, Jing Z, Ordikhani F, Bae M, Clardy J, Annabi N, McGrath MM, Abdi R. Nanodelivery of Mycophenolate Mofetil to the Organ Improves Transplant Vasculopathy. ACS NANO 2019; 13:12393-12407. [PMID: 31518498 PMCID: PMC7247279 DOI: 10.1021/acsnano.9b05115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Inflammation occurring within the transplanted organ from the time of harvest is an important stimulus of early alloimmune reactivity and promotes chronic allograft rejection. Chronic immune-mediated injury remains the primary obstacle to the long-term success of organ transplantation. However, organ transplantation represents a rare clinical setting in which the organ is accessible ex vivo, providing an opportunity to use nanotechnology to deliver therapeutics directly to the graft. This approach facilitates the directed delivery of immunosuppressive agents (ISA) to target local pathogenic immune responses prior to the transplantation. Here, we have developed a system of direct delivery and sustained release of mycophenolate mofetil (MMF) to treat the donor organ prior to transplantation. Perfusion of a donor mouse heart with MMF-loaded PEG-PLGA nanoparticles (MMF-NPs) prior to transplantation abrogated cardiac transplant vasculopathy by suppressing intragraft pro-inflammatory cytokines and chemokines. Our findings demonstrate that ex vivo delivery of an ISA to donor organs using a nanocarrier can serve as a clinically feasible approach to reduce transplant immunity.
Collapse
Affiliation(s)
- Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Baharak Bahmani
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sungwook Jung
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhao Jing
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Farideh Ordikhani
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Munhyung Bae
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Martina M. McGrath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Address correspondence to: Reza Abdi, MD, Transplantation Research Center, Brigham and Women’s Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254, ; Martina M. McGrath, Transplantation Research Center, Brigham and Women’s Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254,
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Address correspondence to: Reza Abdi, MD, Transplantation Research Center, Brigham and Women’s Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254, ; Martina M. McGrath, Transplantation Research Center, Brigham and Women’s Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254,
| |
Collapse
|
19
|
AAV-Mediated Expression of AP-1-Neutralizing RNA Decoy Oligonucleotides Attenuates Transplant Vasculopathy in Mouse Aortic Allografts. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:246-256. [PMID: 31720303 PMCID: PMC6838891 DOI: 10.1016/j.omtm.2019.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/24/2019] [Indexed: 11/21/2022]
Abstract
Transplant vasculopathy (TV), characterized by obstructive lesions in affected vessels, represents one of the long-term complications of cardiac transplantation. Activation of the transcription factor activator protein-1 (AP-1) is implicated in smooth muscle cell (SMC) phenotypic switch from contractile to synthetic function, increasing the migration and proliferation rate of these cells. We hypothesize that adeno-associated virus (AAV)-mediated delivery of an RNA hairpin AP-1 decoy oligonucleotide (dON) might effectively ameliorate TV severity in a mouse aortic allograft model. Aortic allografts from DBA/2 mice ex vivo transduced with modified AAV9-SLR carrying a targeting peptide within the capsid surface were transplanted into the infrarenal aorta of C57BL/6 mice. Cyclosporine A (10 mg/kg BW) was administered daily. AP-1 dONs were intracellularly expressed in the graft tissue as small hairpin RNA proved by fluorescent in situ hybridization. Explantation after 30 days and histomorphometric evaluation revealed that AP-1 dON treatment significantly reduced intima-to-media ratio by 41.5% (p < 0.05) in the grafts. In addition, expression of adhesion molecules, cytokines, as well as numbers of proliferative SMCs, matrix metalloproteinase-9-positive cells, and inflammatory cell infiltration were significantly decreased in treated aortic grafts. Our findings demonstrate the feasibility, efficacy, and specificity of the anti-AP-1 RNA dON approach for the treatment of allograft vasculopathy in an animal model. Moreover, the AAV-based approach in general provides the possibility to achieve a prolonged delivery of nucleic-acids-based therapeutics in to the blood vessel wall.
Collapse
|
20
|
Anti-IL-6 eluting immunomodulatory biomaterials prolong skin allograft survival. Sci Rep 2019; 9:6535. [PMID: 31024011 PMCID: PMC6484015 DOI: 10.1038/s41598-019-42349-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
A primary goal in the management of burn wounds is early wound closure. The use of skin allografts represents a lifesaving strategy for severe burn patients, but their ultimate rejection limits their potential efficacy and utility. IL-6 is a major pleiotropic cytokine which critically links innate and adaptive immune responses. Here, we devised anti-IL-6 receptor eluting gelatin methacryloyl (GelMA) biomaterials (GelMA/anti-IL-6), which were implanted at the interface between the wound beds and skin allografts. Our visible light crosslinked GelMA/anti-IL-6 immunomodulatory biomaterial (IMB) demonstrated a stable kinetic release profile of anti-IL-6. In addition, the incorporation of anti-IL-6 within the GelMA hydrogel had no effect on the mechanical properties of the hydrogels. Using a highly stringent skin transplant model, the GelMA/anti-IL-6 IMB almost doubled the survival of skin allografts. The use of GelMA/anti-IL-6 IMB was far superior to systemic anti-IL-6 receptor treatment in prolonging skin allograft survival. As compared to the untreated control group, skin from the GelMA/anti-IL-6 IMB group contained significantly fewer alloreactive T cells and macrophages. Interestingly, the environmental milieu of the draining lymph nodes (DLNs) of the mice implanted with the GelMA/anti-IL-6 IMB was also considerably less pro-inflammatory. The percentage of CD4+ IFNγ+ cells was much lower in the DLNs of the GelMA/anti-IL-6 IMB group in comparison to the GelMA group. These data highlight the importance of localized immune delivery in prolonging skin allograft survival and its potential utility in treating patients with severe burns.
Collapse
|
21
|
Chen J, Liu C, Liu B, Kong D, Wen L, Gong W. Donor IL-6 deficiency evidently reduces memory T cell responses in sensitized transplant recipients. Transpl Immunol 2018; 51:66-72. [PMID: 30287356 DOI: 10.1016/j.trim.2018.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/30/2018] [Accepted: 09/30/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Resistance of tolerance induction in sensitized transplantation is mainly caused by generation of memory T cells. It is unknown whether alteration of graft niche such as level of pro-inflammatory cytokines can affect generation of memory T cells. METHODS IL-6 deficient or wild-type (WT) C57BL/6 heart grafts were transplanted into pre-sensitized wild-type BALB/c recipients. Frequencies of memory T cells in the peripheral blood, grafts, and spleen were evaluated. RESULTS We revealed that deficiency of donor IL-6 could significant prolong sensitized allograft survival. Compared with counterpart of WT group, frequency of effector memory CD4 + T cells (CD4 + CD44 + CD62L-) in the peripheral blood was significantly lower in the IL-6 KO group (p = .026) at day 3 post-transplantation. Frequency of effector memory CD8 + T cells (CD8 + CD44 + CD62L-) in the peripheral blood was significantly lower in the IL-6 KO group (p < .0001) at day 3 post-transplant in comparison to that of WT group. No significant difference of central memory T cells was found between these groups. Histology demonstrated that deficiency of donor pro-inflammatory cytokine IL-6 (IL-6 KO group) preserved cardiac architecture with a mild infiltration of lymphocytes, whereas wild-type donor (control group) caused an evident lymphocytic infiltration within myocardial fibers of grafts and destruction of cardiac structure. CONCLUSION Deficiency of pro-inflammatory IL-6 of donor graft could effectively prolong sensitized allograft survival, which was caused by a remarkable decrease of peripheral memory T cells rather than central memory T cells. This unveiled mechanism of targeting IL-6 signaling pathway might provide a novel insight into preventing allograft rejection for sensitized transplant recipients.
Collapse
Affiliation(s)
- Juntao Chen
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Chen Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Baoqing Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Deqiang Kong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Liang Wen
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Weihua Gong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China.
| |
Collapse
|
22
|
Li S, Wang S, Murugan R, Al-Khafaji A, Lebovitz DJ, Souter M, Stuart SRN, Kellum JA. Donor biomarkers as predictors of organ use and recipient survival after neurologically deceased donor organ transplantation. J Crit Care 2018; 48:42-47. [PMID: 30172032 DOI: 10.1016/j.jcrc.2018.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/05/2018] [Accepted: 08/14/2018] [Indexed: 02/01/2023]
Abstract
PURPOSE We sought to build prediction models for organ transplantation and recipient survival using both biomarkers and clinical information. MATERIALS AND METHODS We abstracted clinical variables from a previous randomized trial (n = 556) of donor management. In a subset of donors (n = 97), we measured two candidate biomarkers in plasma at enrollment and just prior to explantation. RESULTS Secretory leukocyte protease inhibitor (SLPI) was significant for predicting liver transplantation (C-statistic 0.65 (0.53, 0.78)). SLPI also significantly improved the predictive performance of a clinical model for liver transplantation (integrated discrimination improvement (IDI): 0.090 (0.009, 0.210)). For other organs, clinical variables alone had strong predictive ability (C-statistic >0.80). Recipient 3-years survival was 80.0% (71.9%, 87.0%). Donor IL-6 was significantly associated with recipient 3-years survival (adjusted Hazard Ratio (95%CI): 1.26(1.08, 1.48), P = .004). Neither clinical variables nor biomarkers showed strong predictive ability for 3-year recipient survival. CONCLUSIONS Plasma biomarkers in neurologically deceased donors were associated with organ use. SLPI enhanced prediction within a liver transplantation model, whereas IL-6 before transplantation was significantly associated with recipient 3-year survival. Clinicaltrials.gov: NCT00987714.
Collapse
Affiliation(s)
- Shengnan Li
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Shu Wang
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Raghavan Murugan
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ali Al-Khafaji
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel J Lebovitz
- Department of Critical Care, Akron Children's Hospital, Akron, OH, United States
| | - Michael Souter
- Department of Anesthesiology & Pain Medicine, University of Washington, Harborview Medical Center, Seattle, WA, United States
| | - Susan R N Stuart
- Center for Organ Recovery and Education, Pittsburgh, PA, United States
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | | |
Collapse
|
23
|
Uehara M, Solhjou Z, Banouni N, Kasinath V, Xiaqun Y, Dai L, Yilmam O, Yilmaz M, Ichimura T, Fiorina P, Martins PN, Ohori S, Guleria I, Maarouf OH, Tullius SG, McGrath MM, Abdi R. Ischemia augments alloimmune injury through IL-6-driven CD4 + alloreactivity. Sci Rep 2018; 8:2461. [PMID: 29410442 PMCID: PMC5802749 DOI: 10.1038/s41598-018-20858-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023] Open
Abstract
Ischemia reperfusion injuries (IRI) are unavoidable in solid organ transplantation. IRI augments alloimmunity but the mechanisms involved are poorly understood. Herein, we examined the effect of IRI on antigen specific alloimmunity. We demonstrate that ischemia promotes alloimmune activation, leading to more severe histological features of rejection, and increased CD4+ and CD8+ T cell graft infiltration, with a predominantly CD8+ IFNγ+ infiltrate. This process is dependent on the presence of alloreactive CD4+ T cells, where depletion prevented infiltration of ischemic grafts by CD8+ IFNγ+ T cells. IL-6 is a known driver of ischemia-induced rejection. Herein, depletion of donor antigen-presenting cells reduced ischemia-induced CD8+ IFNγ+ allograft infiltration, and improved allograft outcomes. Following prolonged ischemia, accelerated rejection was observed despite treatment with CTLA4Ig, indicating that T cell costimulatory blockade failed to overcome the immune activating effect of IRI. However, despite severe ischemic injury, treatment with anti-IL-6 and CTLA4Ig blocked IRI-induced alloimmune injury and markedly improved allograft survival. We describe a novel pathway where IRI activates innate immunity, leading to upregulation of antigen specific alloimmunity, resulting in chronic allograft injury. Based on these findings, we describe a clinically relevant treatment strategy to overcome the deleterious effect of IRI, and provide superior long-term allograft outcomes.
Collapse
Affiliation(s)
- Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ye Xiaqun
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Dai
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Osman Yilmam
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mine Yilmaz
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Fiorina
- Division of Nephrology, Boston Children Hospital, Harvard Medical School, Boston, MA, USA
| | - Paulo N Martins
- Division of Surgery, University of Massachusetts Medical School, Boston, MA, USA
| | - Shunsuke Ohori
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Indira Guleria
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar H Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martina M McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Wei J, Chen S, Xue S, Zhu Q, Liu S, Cui L, Hua X, Wang Y. Blockade of Inflammation and Apoptosis Pathways by siRNA Prolongs Cold Preservation Time and Protects Donor Hearts in a Porcine Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:428-439. [PMID: 29246321 PMCID: PMC5701800 DOI: 10.1016/j.omtn.2017.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/28/2017] [Accepted: 10/28/2017] [Indexed: 02/08/2023]
Abstract
In donor hearts from mini pigs, overtime cold preservation and ischemia-reperfusion injury cause poor graft quality and impaired heart function. Blockage of complement, apoptosis, and inflammation is considered a strategy for attenuating ischemia-reperfusion injury and protecting cardiac function. Minipig donor hearts were perfused and preserved in Celsior solution or transfection reagent containing Celsior solution with scramble siRNA or siRNAs targeting complement 3, caspase-8, caspase-3, and nuclear factor κB-p65 genes at 4°C and subsequently hemo-reperfused ex vivo (38°C) or transplanted into recipients. The protective effect of the siRNA solution was evaluated by measuring cell apoptosis, structural alteration, protein markers for tissue damage and oxidative stress, and cardiac function. We found a reduction in cell apoptosis, myocardial damage, and tissue inflammation by reduced biochemistry and markers and protein expression of proinflammatory cytokines and improvement in cardiac function, as shown by the improved hemodynamic indices in 12-hr-preserved siRNA-treated hearts of both ex vivo and orthotopic transplantation models. These findings demonstrate that blockade of inflammation and apoptosis pathways using siRNA can prolong cold preservation time and better protect donor heart function in cardiac transplantation of large animals, which may be beneficial for human heart preservation.
Collapse
Affiliation(s)
- Jia Wei
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiaotong University, Shanghai 200240, China
| | - Shiyou Chen
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Song Xue
- Department of Cardiac Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Qiangru Zhu
- CCI Facility, Covidien (Shanghai) Management Consulting Co. Ltd., Shanghai 200233, China
| | - Sha Liu
- CCI Facility, Covidien (Shanghai) Management Consulting Co. Ltd., Shanghai 200233, China
| | - Li Cui
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiaotong University, Shanghai 200240, China
| | - Xiuguo Hua
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiaotong University, Shanghai 200240, China.
| | - Yongyi Wang
- Department of Cardiac Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| |
Collapse
|
25
|
Plasma Exosomes From HLA-Sensitized Kidney Transplant Recipients Contain mRNA Transcripts Which Predict Development of Antibody-Mediated Rejection. Transplantation 2017; 101:2419-2428. [PMID: 28557957 DOI: 10.1097/tp.0000000000001834] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Sensitization to HLA remains a significant immunologic barrier to successful transplantation. Identifying immune mechanisms responsible for antibody-mediated rejection (AMR) is an important goal. Here, we explored the possibility of predicting the risk for AMR by measuring mRNA transcripts of AMR-associated genes in plasma exosomes from kidney transplant patients. METHODS Total RNA was extracted from exosomes purified from 152 ethylenediaminetetraacetic acid-plasma samples of 64 patients (18 AMR, 8 cell-mediated rejection [CMR], 38 no rejection in desensitized [DES] and non-DES control groups) for reverse transcription into cDNA, preamplification and then real time quantitative polymerase chain reaction (qPCR) for 21 candidate genes. The mRNA transcript levels of each gene were calculated. Comparisons were made among 4 patient groups for each gene and also for a gene combination score based on selected genes. RESULTS Among 21 candidate genes, we identified multiple genes (gp130, CCL4, TNFα, SH2D1B, CAV1, atypical chemokine receptor 1 [duffy blood group]) whose mRNA transcript levels in plasma exosomes significantly increased among AMR compared with CMR and/or control patients. A gene combination score calculated from 4 genes of gp130, SH2D1B, TNFα, and CCL4 was significantly higher in the AMR than the CMR (P < 0.0001) and no rejection control groups (P < 0.01 vs DES control, P < 0.05 vs non-DES control). CONCLUSIONS Our results suggest that plasma exosomes may contain information indicating clinical conditions of kidney transplant patients. mRNA transcript profiles based on gp130, SH2D1B, TNFα, and CCL4 in plasma exosomes may be used to predict on-going and/or imminent AMR.
Collapse
|
26
|
Solhjou Z, Uehara M, Bahmani B, Maarouf OH, Ichimura T, Brooks CR, Xu W, Yilmaz M, Elkhal A, Tullius SG, Guleria I, McGrath M, Abdi R. Novel Application of Localized Nanodelivery of Anti-Interleukin-6 Protects Organ Transplant From Ischemia-Reperfusion Injuries. Am J Transplant 2017; 17:2326-2337. [PMID: 28296000 PMCID: PMC5573642 DOI: 10.1111/ajt.14266] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/03/2017] [Accepted: 02/25/2017] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury (IRI) evokes intragraft inflammatory responses, which markedly augment alloimmune responses against the graft. Understanding the mechanisms underlying these responses is fundamental to develop therapeutic regimens to prevent/ameliorate organ IRI. Here, we demonstrate that IRI results in a marked increase in mitochondrial damage and autophagy in dendritic cells (DCs). While autophagy is a survival mechanism for ischemic DCs, it also augments their production of interleukin (IL)-6. Allograft-derived dendritic cells (ADDCs) lacking autophagy-related gene 5 (Atg5) showed higher death rates posttransplantation. Transplanted ischemic hearts from CD11cCre/Atg5 conditional knockout mice showed marked reduction in intragraft expression of IL-6 compared with controls. To antagonize the effect of IL-6 locally in the heart, we synthesized novel anti-IL-6 nanoparticles with capacity for controlled release of anti-IL-6 over time. Compared with systemic delivery of anti-IL-6, localized delivery of anti-IL-6 significantly reduced chronic rejection with a markedly lower amount administered. Despite improved allograft histology, there were no changes to splenic T cell populations, illustrating the importance of local IL-6 in driving chronic rejection after IRI. These data carry potential clinical significance by identifying an innovative, targeted strategy to manipulate organs before transplantation to diminish inflammation, leading to improved long-term outcomes.
Collapse
Affiliation(s)
- Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Baharak Bahmani
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar H. Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig R. Brooks
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wanlong Xu
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mine Yilmaz
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abdala Elkhal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Indira Guleria
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martina McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA,Address correspondence to: Reza Abdi, MD, Transplant Research Center, Brigham and Women's Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254,
| |
Collapse
|
27
|
A20 Haploinsufficiency Aggravates Transplant Arteriosclerosis in Mouse Vascular Allografts: Implications for Clinical Transplantation. Transplantation 2017; 100:e106-e116. [PMID: 27495763 DOI: 10.1097/tp.0000000000001407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Inflammation is central to the pathogenesis of transplant arteriosclerosis (TA). We questioned whether physiologic levels of anti-inflammatory A20 influence TA severity. METHODS We performed major histocompatibility complex mismatched aorta to carotid artery interposition grafts, using wild type (WT) or A20 heterozygote (HET) C57BL/6 (H-2) donors and BALB/c (H-2) recipients, and conversely BALB/c donors and WT/HET recipients. We analyzed aortic allografts by histology, immunohistochemistry, immunofluorescence, and gene profiling (quantitative real-time reverse-transcriptase polymerase chain reaction). We validated select in vivo A20 targets in human and mouse smooth muscle cell (SMC) cultures. RESULTS We noted significantly greater intimal hyperplasia in HET versus WT allografts, indicating aggravated TA. Inadequate upregulation of A20 in HET allografts after transplantation was associated with excessive NF-кB activation, gauged by higher levels of IkBα, p65, VCAM-1, ICAM-1, CXCL10, CCL2, TNF, and IL-6 (mostly localized to SMC). Correspondingly, cytokine-induced upregulation of TNF and IL-6 in human and mouse SMC cultures inversely correlated with A20 expression. Aggravated TA in HET versus WT allografts correlated with increased intimal SMC proliferation, and a higher number of infiltrating IFNγ and Granzyme B CD4 T cells and natural killer cells, and lower number of FoxP3 regulatory T cells. A20 haploinsufficiency in allograft recipients did not influence TA. CONCLUSIONS A20 haploinsufficiency in vascular allografts aggravates lesions of TA by exacerbating inflammation, SMC proliferation, and infiltration of pathogenic T cells. A20 single nucleotide polymorphisms associating with lower A20 expression or function in donors of vascularized allografts may inform risk and severity of TA, highlighting the clinical implications of our findings.
Collapse
|
28
|
Kennelly KP, Holmes TM, Wallace DM, O'Farrelly C, Keegan DJ. Early Subretinal Allograft Rejection Is Characterized by Innate Immune Activity. Cell Transplant 2017; 26:983-1000. [PMID: 28105976 DOI: 10.3727/096368917x694697] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Successful subretinal transplantation is limited by considerable early graft loss despite pharmacological suppression of adaptive immunity. We postulated that early innate immune activity is a dominant factor in determining graft survival and chose a nonimmunosuppressed mouse model of retinal pigment epithelial (RPE) cell transplantation to explore this. Expression of almost all measured cytokines by DH01 RPE cells increased significantly following graft preparation, and the neutrophil chemoattractant KC/GRO/CINC was most significantly increased. Subretinal allografts of DH01 cells (C57BL/10 origin) into healthy, nonimmunosuppressed C57BL/6 murine eyes were harvested and fixed at 1, 3, 7, and 28 days postoperatively and subsequently cryosectioned and stained. Graft cells were detected using SV40 large T antigen (SV40T) immunolabeling and apoptosis/necrosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Sections were also immunolabeled for macrophage (CD11b and F4/80), neutrophil (Gr1 Ly-6G), and T-lymphocyte (CD3-ɛ) infiltration. Images captured with an Olympus FV1000 confocal microscope were analyzed using the Imaris software. The proportion of the subretinal bolus comprising graft cells (SV40T+) was significantly (p < 0.001) reduced between postoperative day (POD) 3 (90 ± 4%) and POD 7 (20 ± 7%). CD11b+, F4/80+, and Gr1 Ly-6G+ cells increased significantly (p < 0.05) from POD 1 and predominated over SV40T+ cells by POD 7. Colabeling confocal microscopic analysis demonstrated graft engulfment by neutrophils and macrophages at POD 7, and reconstruction of z-stacked confocal images confirmed SV40T inside Gr1 Ly-6G+ cells. Expression of CD3-ɛ was low and did not differ significantly between time points. By POD 28, no graft cells were detectable and few inflammatory cells remained. These studies reveal, for the first time, a critical role for innate immune mechanisms early in subretinal graft rejection. The future success of subretinal transplantation will require more emphasis on techniques to limit innate immune-mediated graft loss, rather than focusing exclusively on suppression of the adaptive immune response.
Collapse
|
29
|
Arciero JC, Maturo A, Arun A, Oh BC, Brandacher G, Raimondi G. Combining Theoretical and Experimental Techniques to Study Murine Heart Transplant Rejection. Front Immunol 2016; 7:448. [PMID: 27872621 PMCID: PMC5097940 DOI: 10.3389/fimmu.2016.00448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/10/2016] [Indexed: 12/21/2022] Open
Abstract
The quality of life of organ transplant recipients is compromised by complications associated with life-long immunosuppression, such as hypertension, diabetes, opportunistic infections, and cancer. Moreover, the absence of established tolerance to the transplanted tissues causes limited long-term graft survival rates. Thus, there is a great medical need to understand the complex immune system interactions that lead to transplant rejection so that novel and effective strategies of intervention that redirect the system toward transplant acceptance (while preserving overall immune competence) can be identified. This study implements a systems biology approach in which an experimentally based mathematical model is used to predict how alterations in the immune response influence the rejection of mouse heart transplants. Five stages of conventional mouse heart transplantation are modeled using a system of 13 ordinary differential equations that tracks populations of both innate and adaptive immunity as well as proxies for pro- and anti-inflammatory factors within the graft and a representative draining lymph node. The model correctly reproduces known experimental outcomes, such as indefinite survival of the graft in the absence of CD4+ T cells and quick rejection in the absence of CD8+ T cells. The model predicts that decreasing the translocation rate of effector cells from the lymph node to the graft delays transplant rejection. Increasing the starting number of quiescent regulatory T cells in the model yields a significant but somewhat limited protective effect on graft survival. Surprisingly, the model shows that a delayed appearance of alloreactive T cells has an impact on graft survival that does not correlate linearly with the time delay. This computational model represents one of the first comprehensive approaches toward simulating the many interacting components of the immune system. Despite some limitations, the model provides important suggestions of experimental investigations that could improve the understanding of rejection. Overall, the systems biology approach used here is a first step in predicting treatments and interventions that can induce transplant tolerance while preserving the capacity of the immune system to protect against legitimate pathogens.
Collapse
Affiliation(s)
- Julia C Arciero
- Department of Mathematical Sciences, Indiana University-Purdue University Indianapolis , Indianapolis, IN , USA
| | - Andrew Maturo
- Department of Mathematical Sciences, Indiana University-Purdue University Indianapolis , Indianapolis, IN , USA
| | - Anirudh Arun
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Byoung Chol Oh
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Gerald Brandacher
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| |
Collapse
|
30
|
Graft-Derived IL-6 Amplifies Proliferation and Survival of Effector T Cells That Drive Alloimmune-Mediated Vascular Rejection. Transplantation 2016; 100:2332-2341. [DOI: 10.1097/tp.0000000000001227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
31
|
Abstract
Immunosuppression strategies that selectively inhibit effector T cells while preserving and even enhancing CD4FOXP3 regulatory T cells (Treg) permit immune self-regulation and may allow minimization of immunosuppression and associated toxicities. Many immunosuppressive drugs were developed before the identity and function of Treg were appreciated. A good understanding of the interactions between Treg and immunosuppressive agents will be valuable to the effective design of more tolerable immunosuppression regimens. This review will discuss preclinical and clinical evidence regarding the influence of current and emerging immunosuppressive drugs on Treg homeostasis, stability, and function as a guideline for the selection and development of Treg-friendly immunosuppressive regimens.
Collapse
Affiliation(s)
- Akiko Furukawa
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Steven A Wisel
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Zhang Z, Sun H, Zhang J, Ge C, Dong S, Li Z, Li R, Chen X, Li M, Chen Y, Zou Y, Qian Z, Yang L, Yang J, Zhu Z, Liu Z, Song X. Safety and Efficacy of Transplantation with Allogeneic Skin Tumors to Treat Chemically-Induced Skin Tumors in Mice. Med Sci Monit 2016; 22:3113-23. [PMID: 27587310 PMCID: PMC5019137 DOI: 10.12659/msm.900148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Transplantation with allogeneic cells has become a promising modality for cancer therapy, which can induce graft-versus-tumor (GVT) effect. This study was aimed at assessing the safety, efficacy, and tissue type GVT (tGVT) response of transplantation with allogeneic skin tumors to treat chemically-induced skin tumors in mice. Material/Methods FVB/N and ICR mice were exposed topically to chemicals to induce skin tumors. Healthy ICR mice were transplanted with allogeneic skin tumors from FVB/N mice to test the safety. The tumor-bearing ICR mice were transplanted with, or without, allogeneic skin tumors to test the efficacy. The body weights (BW), body condition scores (BCS), tumor volumes in situ, metastasis tumors, overall survival, and serum cytokines were measured longitudinally. Results Transplantation with no more than 0.03 g allogeneic skin tumors from FVB/N mice to healthy ICR mice was safe. After transplantation with allogeneic skin tumors to treat tumor-bearing mice, it inhibited the growth of tumors slightly at early stage, accompanied by fewer metastatic tumors at 24 days after transplantation (21.05% vs. 47.37%), while there were no statistically significant differences in the values of BW, BCS, tumor volumes in situ, metastasis tumors, and overall survival between the transplanted and non-transplanted groups. The levels of serum interleukin (IL)-2 were significantly reduced in the controls (P<0.05), but not in the recipients, which may be associated with the tGVT response. Conclusions Our results suggest that transplantation with allogeneic skin tumors is a safe treatment in mice, which can induce short-term tGVT response mediated by IL-2.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Hua Sun
- PET/CT Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Jianhua Zhang
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Chunlei Ge
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Suwei Dong
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Zhen Li
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Ruilei Li
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Xiaodan Chen
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Mei Li
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Yun Chen
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Yingying Zou
- Department of Pathology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Zhongyi Qian
- Laboratory of Morphology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Lei Yang
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Jinyan Yang
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Zhitao Zhu
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Zhimin Liu
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| | - Xin Song
- Department of Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, China (mainland)
| |
Collapse
|
33
|
Iida S, Tsuda H, Tanaka T, Kish DD, Abe T, Su CA, Abe R, Tanabe K, Valujskikh A, Baldwin WM, Fairchild RL. IL-1 Receptor Signaling on Graft Parenchymal Cells Regulates Memory and De Novo Donor-Reactive CD8 T Cell Responses to Cardiac Allografts. THE JOURNAL OF IMMUNOLOGY 2016; 196:2827-37. [PMID: 26856697 DOI: 10.4049/jimmunol.1500876] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 01/04/2016] [Indexed: 01/03/2023]
Abstract
Reperfusion of organ allografts induces a potent inflammatory response that directs rapid memory T cell, neutrophil, and macrophage graft infiltration and their activation to express functions mediating graft tissue injury. The role of cardiac allograft IL-1 receptor (IL-1R) signaling in this early inflammation and the downstream primary alloimmune response was investigated. When compared with complete MHC-mismatched wild-type cardiac allografts, IL-1R(-/-) allografts had marked decreases in endogenous memory CD8 T cell and neutrophil infiltration and expression of proinflammatory mediators at early times after transplant, whereas endogenous memory CD4 T cell and macrophage infiltration was not decreased. IL-1R(-/-) allograft recipients also had marked decreases in de novo donor-reactive CD8, but not CD4, T cell development to IFN-γ-producing cells. CD8 T cell-mediated rejection of IL-1R(-/-) cardiac allografts took 3 wk longer than wild-type allografts. Cardiac allografts from reciprocal bone marrow reconstituted IL-1R(-/-)/wild-type chimeric donors indicated that IL-1R signaling on graft nonhematopoietic-derived, but not bone marrow-derived, cells is required for the potent donor-reactive memory and primary CD8 T cell alloimmune responses observed in response to wild-type allografts. These studies implicate IL-1R-mediated signals by allograft parenchymal cells in generating the stimuli-provoking development and elicitation of optimal alloimmune responses to the grafts.
Collapse
Affiliation(s)
- Shoichi Iida
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Hidetoshi Tsuda
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Urology, Osaka University School of Medicine, Osaka 565-0871, Japan; and
| | - Toshiaki Tanaka
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Danielle D Kish
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Toyofumi Abe
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Urology, Osaka University School of Medicine, Osaka 565-0871, Japan; and
| | - Charles A Su
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Kazunari Tanabe
- Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Anna Valujskikh
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - William M Baldwin
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
34
|
Pabois A, Pagie S, Gérard N, Laboisse C, Pattier S, Hulin P, Nedellec S, Toquet C, Charreau B. Notch signaling mediates crosstalk between endothelial cells and macrophages via Dll4 and IL6 in cardiac microvascular inflammation. Biochem Pharmacol 2016; 104:95-107. [PMID: 26826491 DOI: 10.1016/j.bcp.2016.01.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
Although short-term outcomes have improved with modern era immunosuppression, little progress has been made in long-term graft survival in cardiac transplantation. Antibody-mediated rejection (AMR) is one of the leading causes of graft failure and contributes significantly to poor long-term outcomes. Endothelial cell (EC) injury, intravascular macrophage infiltrate and microvascular inflammation are the histological features of AMR. Nevertheless, mechanisms of AMR remain unclear and treatment is still limited. Here, we investigated the mechanisms underlying vascular and inflammatory cell network involved in AMR at endothelial and macrophage levels, using endomyocardial transplant biopsies and EC/monocyte cocultures. First, we found that AMR associates with changes in Notch signaling at endothelium/monocyte interface including loss of endothelial Notch4 and the acquisition of the Notch ligand Dll4 in both cell types. We showed that endothelial Dll4 induces macrophage polarization into a pro-inflammatory fate (CD40(high)CD64(high)CD200R(low) HLA-DR(low)CD11b(low)) eliciting the production of IL-6. Dll4 and IL-6 are both Notch-dependent and are required for macrophage polarization through selective down and upregulation of M2- and M1-type markers, respectively. Overall, these findings highlight the impact of the graft's endothelium on macrophage recruitment and differentiation upon AMR via Notch signaling. We identified Dll4 and IL-6 as coregulators of vascular inflammation in cardiac transplantation and as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Angélique Pabois
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Sylvain Pagie
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Nathalie Gérard
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France
| | | | - Sabine Pattier
- Service de transplantation cardiaque, CHU de Nantes, Nantes F44000, France
| | - Philippe Hulin
- LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France; Plateforme MicroPICell SFR Santé - IRT, Nantes, France
| | - Steven Nedellec
- LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France; Plateforme MicroPICell SFR Santé - IRT, Nantes, France
| | - Claire Toquet
- Service d'Anatomie Pathologique, CHU de Nantes, Nantes F44000, France
| | - Béatrice Charreau
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France.
| |
Collapse
|
35
|
Chadet S, Ivanes F, Benoist L, Salmon-Gandonnière C, Guibon R, Velge-Roussel F, Babuty D, Baron C, Roger S, Angoulvant D. Hypoxia/Reoxygenation Inhibits P2Y11 Receptor Expression and Its Immunosuppressive Activity in Human Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:651-60. [PMID: 26078273 DOI: 10.4049/jimmunol.1500197] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/17/2015] [Indexed: 12/20/2022]
Abstract
High concentrations of extracellular ATP (eATP) resulting from cell damage may be found during an ischemia/reperfusion (I/R) episode at the site of injury. eATP activates purinergic receptors in dendritic cells (DCs) and may inhibit inflammation. This immunosuppressive activity could be of interest in the field of I/R, which is an inflammatory condition involved in myocardial infarction, stroke, and solid organ transplantation. However, the specific purinergic receptor responsible for this effect remains to be identified. In this study, we report that eATP induced maturation of human monocyte-derived DCs. Additionally, eATP inhibited IL-12 production whereas IL-10 levels remained unchanged in activated DCs. These effects were prevented by the P2Y11R antagonist NF340. Interestingly, a 5-h hypoxia prevented the effects of eATP on cytokine production whereas a 1-h hypoxia did not affect the eATP-mediated decrease of IL-12 and IL-6. We showed a time-dependent downregulation of P2Y11R at both mRNA and protein levels that was prevented by knocking down hypoxia-inducible factor-1α. In this study, we showed an immunosuppressive role of P2Y11R in human DCs. Additionally, we demonstrated that the time-dependent downregulation of P2Y11R by hypoxia orientates DCs toward a proinflammatory phenotype that may be involved in post-I/R injuries as observed after organ transplantation.
Collapse
Affiliation(s)
- Stéphanie Chadet
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France
| | - Fabrice Ivanes
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France; Service de Cardiologie, Hôpital Trousseau, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Lauriane Benoist
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France
| | - Charlotte Salmon-Gandonnière
- Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37000 Tours, France
| | - Roseline Guibon
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France
| | - Florence Velge-Roussel
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France; Fédération Hospitalo-Universitaire SUPPORT Tours, Poitiers, Limoges, Université François-Rabelais de Tours, 37032 Tours, France; and
| | - Dominique Babuty
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France; Service de Cardiologie, Hôpital Trousseau, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France
| | - Christophe Baron
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France; Service de Néphrologie et d'Immunologie Clinique, Centre Hospitalier Régional Universitaire de Tours, 37000 Tours, France; Fédération Hospitalo-Universitaire SUPPORT Tours, Poitiers, Limoges, Université François-Rabelais de Tours, 37032 Tours, France; and
| | - Sébastien Roger
- UMR INSERM 1069, "Nutrition, Croissance et Cancer," Université François-Rabelais de Tours, 37032 Tours, France
| | - Denis Angoulvant
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032 Tours, France; Service de Cardiologie, Hôpital Trousseau, Centre Hospitalier Régional Universitaire de Tours, 37044 Tours, France; Fédération Hospitalo-Universitaire SUPPORT Tours, Poitiers, Limoges, Université François-Rabelais de Tours, 37032 Tours, France; and
| |
Collapse
|
36
|
Ni Q, Yuan B, Liu T, Lan F, Luo X, Lu X, Huang P, Dai L, Jin X, Yin H. Sphingosine-1-phosphate receptor 1 agonist SEW2871 prolongs heterotopic heart allograft survival in mice. Int Immunopharmacol 2015; 26:37-42. [PMID: 25776899 DOI: 10.1016/j.intimp.2015.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 11/26/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a biologically active metabolite of plasma-membrane sphingolipids that is essential for immune cell trafficking. Recent studies have revealed immunomodulatory functions of S1P and its receptors (S1PR1-S1PR5) in many inflammatory conditions, such as asthma and autoimmunity. Here, we explore the efficacy of SEW2871, a selective S1PR1 agonist, in the prevention of acute allograft rejection in a murine cardiac transplantation model. Treatment of recipient mice with SEW2871 significantly prolongs cardiac allograft survival as compared to those recipients treated with control vehicle. The enhanced graft survival is associated with reduced circulating lymphocytes and allograft inflammatory cell infiltration. The cytokine analysis showed decreased allograft expression of TNF-α, IFN-γ and IL-2 in the SEW2871-treated mice. Moreover, administration of SEW2871 increases the percentage of CD4(+) T regulatory cells and FoxP3 expression in spleen of allograft recipients. Therefore, SEW2871 plays a critical role in regulation of lymphocyte trafficking and development, which directly contributes to prolongation of the allograft survival.
Collapse
Affiliation(s)
- Qian Ni
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Baohong Yuan
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Fang Lan
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaochun Luo
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoyan Lu
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ping Huang
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Liangcheng Dai
- Intensive Care Unit, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
37
|
Shen H, Heuzey E, Mori DN, Wong CK, Colangelo CM, Chung LM, Bruce C, Slizovskiy IB, Booth CJ, Kreisel D, Goldstein DR. Haptoglobin enhances cardiac transplant rejection. Circ Res 2015; 116:1670-9. [PMID: 25801896 DOI: 10.1161/circresaha.116.305406] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/23/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Early graft inflammation enhances both acute and chronic rejection of heart transplants, but it is unclear how this inflammation is initiated. OBJECTIVE To identify specific inflammatory modulators and determine their underlying molecular mechanisms after cardiac transplantation. METHODS AND RESULTS We used a murine heterotopic cardiac transplant model to identify inflammatory modulators of early graft inflammation. Unbiased mass spectrometric analysis of cardiac tissue before and ≤72 hours after transplantation revealed that 22 proteins including haptoglobin, a known antioxidant, are significantly upregulated in our grafts. Through the use of haptoglobin-deficient mice, we show that 80% of haptoglobin-deficient recipients treated with perioperative administration of the costimulatory blocking agent CTLA4 immunoglobulin exhibited >100-day survival of full major histocompatibility complex mismatched allografts, whereas all similarly treated wild-type recipients rejected their transplants by 21 days after transplantation. We found that haptoglobin modifies the intra-allograft inflammatory milieu by enhancing levels of the inflammatory cytokine interleukin-6 and the chemokine MIP-2 (macrophage inflammatory protein 2) but impair levels of the immunosuppressive cytokine interleukin-10. Haptoglobin also enhances dendritic cell graft recruitment and augments antidonor T-cell responses. Moreover, we confirmed that the protein is present in human cardiac allograft specimens undergoing acute graft rejection. CONCLUSIONS Our findings provide new insights into the mechanisms of inflammation after cardiac transplantation and suggest that, in contrast to its prior reported antioxidant function in vascular inflammation, haptoglobin is an enhancer of inflammation after cardiac transplantation. Haptoglobin may also be a key component in other sterile inflammatory conditions.
Collapse
Affiliation(s)
- Hua Shen
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Elizabeth Heuzey
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel N Mori
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Christine K Wong
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Christopher M Colangelo
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Lisa M Chung
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Can Bruce
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Ilya B Slizovskiy
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Carmen J Booth
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel Kreisel
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel R Goldstein
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO.
| |
Collapse
|
38
|
Khalifian S, Raimondi G, Lee WA, Brandacher G. Taming inflammation by targeting cytokine signaling: new perspectives in the induction of transplantation tolerance. Immunotherapy 2015; 6:637-53. [PMID: 24896631 DOI: 10.2217/imt.14.25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transplantation tolerance remains an elusive goal, partly due to limitations in our understanding of the interplay between inflammatory mediators and their role in the activation and regulation of T lymphocytes. Although multiple mechanisms acting both centrally and peripherally are responsible for tolerance induction, the signaling pathways leading to activation or regulation of adaptive immunity are often complex, branched, redundant and modulated by the microenvironment's inflammatory milieu. Accumulating evidence clearly indicates that inflammatory cytokines limit the tolerogenic potential of immunomodulatory protocols by supporting priming of the immune system and counteracting regulatory mechanisms, ultimately promoting rejection. In this review, we summarize recent progress in the development of novel therapeutics to manipulate this inflammatory environment and achievements in targeted inhibition of inflammatory cytokine signaling. Ultimately, robust transplant tolerance induction will probably require a multifaceted, holistic approach that integrates the various mechanisms of tolerance induction, incorporates the dynamic alterations in costimulatory requirements of alloreactive T cells, while maintaining endogenous mechanisms of immune regulation.
Collapse
Affiliation(s)
- Saami Khalifian
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
39
|
Gong W, Shou D, Cheng F, Shi J, Ge F, Liu D. Tolerance induced by IL-6 deficient donor heart is significantly involved in myeloid-derived suppressor cells (MDSCs). Transpl Immunol 2015; 32:72-5. [PMID: 25680847 DOI: 10.1016/j.trim.2015.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 10/30/2014] [Accepted: 02/02/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Transplant tolerance induced by IL-6 deficient donor is supported by regulatory T cells (Tregs). However, it is unknown whether innate immunoregulatory cells such as myeloid-derived suppressor cells (MDSCs) are involved in the process. MATERIALS AND METHODS In this study, we demonstrate the role of MDSCs by transplanting IL-6 deficient heart grafts into wild-type recipients in a murine allogeneic transplant model. RESULTS Our data further revealed that utilization of IL-6 deficient heart grafts could cause a significant prolongation of allograft survival (Mantel-Cox Test, p = 0.001; Gehan-Breslow-Wilcoxon Test, p = 0.0016) and a remarkable increase of the frequency of CD11b + Gr1(-low) in the recipients' spleens (p = 0.0028). CONCLUSIONS MDSCs rather than Th17 cells are closely involved in induced tolerance by IL-6 deficient donor heart. This unveiled mechanism of targeting IL-6 or its signaling pathway may provide a novel insight into preventing allograft rejection for non-sensitized transplant recipients.
Collapse
Affiliation(s)
- Weihua Gong
- Department of Surgery and Medicine, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China; Department of Medicine, Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Dawei Shou
- Department of Surgery and Medicine, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Fei Cheng
- Center for Stem Cell and Translation Medicine, School of Life Sciences, Anhui University, Hefei City, People's Republic of China
| | - Jianguang Shi
- Department of Thoracic Surgery, Ningbo First Hospital, Ningbo City, People's Republic of China
| | - Fangmin Ge
- Department of Surgery and Medicine, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Dahai Liu
- Center for Stem Cell and Translation Medicine, School of Life Sciences, Anhui University, Hefei City, People's Republic of China.
| |
Collapse
|
40
|
Solhjou Z, Athar H, Xu Q, Abdi R. Emerging therapies targeting intra-organ inflammation in transplantation. Am J Transplant 2015; 15:305-11. [PMID: 25612486 DOI: 10.1111/ajt.13073] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 09/16/2014] [Accepted: 10/12/2014] [Indexed: 01/25/2023]
Abstract
Over the past several years, the field of transplantation has witnessed significant progress on several fronts; in particular, achievements have been made in devising novel immunosuppressive strategies. An under-explored area that may hold great potential to improve transplantation outcomes is the design of novel strategies to apply specifically to organs to reduce intra-graft inflammation. A growing body of evidence indicates a key role of intra-graft inflammatory cascade in potently instigating the alloimmune response. Indeed, controlling the activation of innate immunity/inflammatory responses has been shown to be a promising strategy to increase the graft acceptance and survival. In this minireview, we provide an overview of emerging targeted strategies, which can be directly applied to grafts to down-regulate intra-graft inflammation prior to transplantation.
Collapse
Affiliation(s)
- Z Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | | |
Collapse
|
41
|
Sinclair J, Reeves M. The intimate relationship between human cytomegalovirus and the dendritic cell lineage. Front Microbiol 2014; 5:389. [PMID: 25147545 PMCID: PMC4124589 DOI: 10.3389/fmicb.2014.00389] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/11/2014] [Indexed: 11/13/2022] Open
Abstract
Primary infection of healthy individuals with human cytomegalovirus (HCMV) is normally asymptomatic but results in the establishment of a lifelong infection of the host. One important cellular reservoir of HCMV latency is the CD34+ haematopoietic progenitor cells resident in the bone marrow. Viral gene expression is highly restricted in these cells with an absence of viral progeny production. However, cellular differentiation into mature myeloid cells is concomitant with the induction of a full lytic transcription program, DNA replication and, ultimately, the production of infectious viral progeny. Such reactivation of HCMV is a major cause of morbidity and mortality in a number of immune-suppressed patient populations. Our current understanding of HCMV carriage and reactivation is that cellular differentiation of the CD34+ progenitor cells through the myeloid lineage, resulting in terminal differentiation to either a macrophage or dendritic cell (DC) phenotype, is crucial for the reactivation event. In this mini-review, we focus on the interaction of HCMV with DCs, with a particular emphasis on their role in reactivation, and discuss how the critical regulation of viral major immediate-early gene expression appears to be delicately entwined with the activation of cellular pathways in differentiating DCs. Furthermore, we also explore the possible immune consequences associated with reactivation in a professional antigen presenting cell and potential countermeasures HCMV employs to abrogate these.
Collapse
Affiliation(s)
- John Sinclair
- Department of Medicine, University of Cambridge - Addenbrooke's Hospital Cambridge, UK
| | - Matthew Reeves
- Institute of Immunity and Transplantation, University College London - Royal Free Hospital Hampstead, London, UK
| |
Collapse
|
42
|
Batal I, Azzi J, Mounayar M, Abdoli R, Moore R, Lee JY, Rosetti F, Wang C, Fiorina P, Sackstein R, Ichimura T, Abdi R. The mechanisms of up-regulation of dendritic cell activity by oxidative stress. J Leukoc Biol 2014; 96:283-93. [PMID: 24676276 PMCID: PMC4101089 DOI: 10.1189/jlb.3a0113-033rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 12/20/2022] Open
Abstract
Whereas DC have increasingly been recognized for their role in activating the inflammatory cascades during IRIs, the mechanisms by which oxidative stress enhances DC activation remain to be explored. We examined the role of oxidative stress on two important features of DC: T cell activation and trafficking. Bone marrow-derived OS-DC were compared with untreated DC. DC exposed to oxidative stress augmented allogeneic T cell proliferation and showed increased migration in a chemotaxis chamber. These results were confirmed by using hypoxanthine and xanthine oxidase as another inducer of oxidative stress. We used OT-II and OT-I mice to assess the effect of oxidative stress on DC activation of OVA-specific CD4(+) and CD8(+) T cells, respectively. Oxidative stress increased DC capacity to promote OVA-specific CD4(+) T cell activity, demonstrated by an increase in their proliferation and production of IFN-γ, IL-6, and IL-2 proinflammatory cytokines. Whereas oxidative stress increased the DC ability to stimulate IFN-γ production by OVA-specific CD8(+) T cells, cellular proliferation and cytotoxicity were not affected. Compared with untreated DC, oxidative stress significantly reduced the capacity of DC to generate T(regs), which were restored by using anti-IL-6. With regard to DC trafficking, whereas oxidative stress increased DC expression of p-Akt and p-NF-κB, targeting PI3Kγ and NF-κB pathways abrogated the observed increase in DC migration. Our data propose novel insights on the activation of DC by oxidative stress and provide rationales for targeted therapies, which can potentially attenuate IRI.
Collapse
Affiliation(s)
- Ibrahim Batal
- Transplantation Research Center, Departments of Pathology and
| | | | | | | | | | | | | | - Chang Wang
- Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
43
|
Ge F, Yuan S, Su L, Shen Z, He A, Huang T, Gong W. Alteration of innate immunity by donor IL-6 deficiency in a presensitized heart transplant model. PLoS One 2013; 8:e77559. [PMID: 24147024 PMCID: PMC3797753 DOI: 10.1371/journal.pone.0077559] [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: 04/24/2013] [Accepted: 09/03/2013] [Indexed: 11/21/2022] Open
Abstract
Engraftment of IL-6 deficient donor into wild-type recipient could significantly improve allograft survival through T cell lineage particularly regulatory T cells (Tregs) in non-sensitized transplant host. However, its effect on innate immune responses remains uncertain. Our data revealed that donor IL-6 deficiency significantly increased infiltration of two subsets of MDSCs (CD11b+Gr1+myeloid-derived suppressor cells), CD11b+Gr1(-low) and CD11b+Gr1(-int) with strong immunosuppression activity in the transplanted graft. It resulted in a dramatic increase of CD11b+Gr1(-low) frequency and a significant decrease of the frequency of CD11b+Gr1(-high) and CD4-CD8-NK1.1+ cells in the recipient's spleen. Unexpectedly, donor IL-6 deficiency could not significantly reduce macrophage frequency irrespective of in the host's spleen or graft. Taken together, suppression of innate immune effector cells and enhanced activity of regulatory MDSCs contributed to tolerance induction by blockade of IL-6 signaling pathway. The unveiled novel mechanism of targeting IL-6 might shed light on clinical therapeutic application in preventing accelerated allograft rejection for those pre-sensitized transplant recipients.
Collapse
Affiliation(s)
- Fangmin Ge
- Department of Surgery and Medicine, Transplant International Research Centre (TIRC), Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Shunzong Yuan
- Department of Lymphoma, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lida Su
- Department of Surgery and Medicine, Transplant International Research Centre (TIRC), Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Zhonghua Shen
- Department of Surgery and Medicine, Transplant International Research Centre (TIRC), Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| | - Aibin He
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tao Huang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Weihua Gong
- Department of Surgery and Medicine, Transplant International Research Centre (TIRC), Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou City, People's Republic of China
| |
Collapse
|
44
|
Yao J, Feng XW, Yu XB, Xie HY, Zhu LX, Yang Z, Wei BJ, Zheng SS, Zhou L. Recipient IL-6-572C/G genotype is associated with reduced incidence of acute rejection following liver transplantation. J Int Med Res 2013; 41:356-64. [PMID: 23569034 DOI: 10.1177/0300060513477264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Acute rejection resulting from alloimmune responses is a major risk factor affecting patient survival following liver transplantation. Since interleukin (IL)-6 can mediate acute rejection, the association between IL-6 gene single nucleotide polymorphisms (SNPs) and incidence of acute rejection in liver transplant recipients was investigated. METHODS Patients who received liver transplant between January 2005 and December 2010 were typed for IL6-572C/G (rs1800796) polymorphisms using the snapshot technique. Association between genotype and acute rejection was analysed using the SNP Statistics website: http://bioinfo.iconcologia.net/snpstats/start.htm. Allelic and genotypic distributions for rs1800796 were compared among 335 patients with or without acute rejection within the first 6 months following liver transplant. RESULTS Incidence of acute rejection was 11.94%. A heterozygous CG genotype for IL6-572C/G was associated with a lower acute rejection rate compared with homozygous CC or GG genotypes. CONCLUSION IL6-572 CG genotype may be related to protection from acute rejection following liver transplant in Han Chinese patients.
Collapse
Affiliation(s)
- Jia Yao
- Division of Hepatobiliary and Pancreatic Surgery, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Lee J, Nakagiri T, Oto T, Harada M, Morii E, Shintani Y, Inoue M, Iwakura Y, Miyoshi S, Okumura M, Hirano T, Murakami M. IL-6 Amplifier, NF-κB–Triggered Positive Feedback for IL-6 Signaling, in Grafts Is Involved in Allogeneic Rejection Responses. THE JOURNAL OF IMMUNOLOGY 2012; 189:1928-36. [DOI: 10.4049/jimmunol.1103613] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
46
|
Wu ZW, Ling ZX, Lu HF, Zuo J, Sheng JF, Zheng SS, Li LJ. Changes of gut bacteria and immune parameters in liver transplant recipients. Hepatobiliary Pancreat Dis Int 2012; 11:40-50. [PMID: 22251469 DOI: 10.1016/s1499-3872(11)60124-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Liver transplantation is one of the most effective therapeutic options for patients with end-stage liver diseases, and gut microbiota is actively involved in potential infections in pretransplant and posttransplant patients. However, the diversity of gut microbiota and its relationship with the immune parameter of liver transplantation recipients are not well understood. METHODS We collected fresh feces and blood samples from 190 participants in China from November 2004 to May 2008, including 28 healthy volunteers, 51 cirrhotic patients and 111 liver-transplanted patients. Six interesting gut bacteria, plasma endotoxin, serum cytokines (i.e., tumor necrosis factor alpha and interleukin-6) and fecal secretory IgA (SIgA) were investigated by real-time quantitative PCR, chromogenic limulus amoebocyte assay, sandwich-type enzyme-linked immunosorbent assay and radioimmunoassay, respectively. RESULTS All Eubacteria, Bifidobacterium spp., Faecalibacterium prausnitzii and Lactobacillus spp. were significantly lower in the liver transplantation recipients while Enterobacteriaceae and Enterococcus spp. were significantly higher (P<0.05). Except for Enterococcus spp., other bacteria showed a tendency to restore to normal level along with the time after liver transplantation. Plasma endotoxin, interleukin-6 and fecal SIgA in cirrhotic patients increased significantly, but not in liver transplantation recipients. Plasma endotoxin and interleukin-6 were negatively correlated with all Eubacteria and the Bacteroides-Prevotella group, while tumor necrosis factor alpha was not significantly correlated with these six gut bacteria in cirrhotic patients. CONCLUSIONS Our study demonstrates that abundant gut bacteria were altered significantly in both cirrhotic and liver transplantation patients, while plasma endotoxin and interleukin-6 increased remarkably in cirrhotic patients, showing significant correlations with gut microbiota. Interestingly, our data show a tendency for these gut bacteria to restore to normal levels in liver transplantation recipients.
Collapse
Affiliation(s)
- Zhong-Wen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases; Department of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Wang H, Guan Q, Lan Z, Li S, Ge W, Chen H, Nguan CYC, Du C. Prolonged renal allograft survival by donor interleukin-6 deficiency: association with decreased alloantibodies and increased intragraft T regulatory cells. Am J Physiol Renal Physiol 2012; 302:F276-83. [DOI: 10.1152/ajprenal.00258.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Both humoral and cellular immune responses are involved in renal allograft rejection. Interleukin (IL)-6 is a regulatory cytokine for both B and Foxp3 (forkhead box P3)-expressing regulatory T (Treg) cells. This study was designed to investigate the impact of donor IL-6 production on renal allograft survival. Donor kidneys from IL-6 knockout (KO) vs. wild-type (WT) C57BL/6 mice (H-2b) were orthotopically transplanted to nephrotomized BALB/c mice (H-2d). Alloantibodies and Treg cells were examined by fluorescence-activated cell sorting analysis. Graft survival was determined by the time to graft failure. Here, we showed that a deficiency in IL-6 expression in donor kidneys significantly prolonged renal allograft survival compared with WT controls. IL-6 protein was upregulated in renal tubules and endothelium of renal allografts following rejection, which correlated with an increase in serum IL-6 compared with that in those receiving KO grafts or naive controls. The absence of graft-producing IL-6 or lower levels of serum IL-6 in the recipients receiving IL-6 KO allografts was associated with decreased circulating anti-graft alloantibodies and increased the percentage of intragraft CD4+CD25+Foxp3+ Treg cells compared with those with WT allografts. In conclusion, the lack of graft-producing IL-6 significantly prolongs renal allograft survival, which is associated with reduced alloantibody production and/or increased intragraft Treg cell population, implying that targeting donor IL-6 may effectively prevent both humoral and cellular rejection of kidney transplants.
Collapse
Affiliation(s)
- Hao Wang
- Department of Surgery, The University of Western Ontario, London, Ontario
| | - Qiunong Guan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| | - Zhu Lan
- Department of Surgery, The University of Western Ontario, London, Ontario
| | - Shuyuan Li
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| | - Wei Ge
- Department of Surgery, The University of Western Ontario, London, Ontario
| | - Huifang Chen
- Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Christopher Y. C. Nguan
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| | - Caigan Du
- Department of Urologic Sciences, The University of British Columbia, Vancouver, British Columbia
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| |
Collapse
|
48
|
Zhao X, Boenisch O, Yeung M, Mfarrej B, Yang S, Turka LA, Sayegh MH, Iacomini J, Yuan X. Critical role of proinflammatory cytokine IL-6 in allograft rejection and tolerance. Am J Transplant 2012; 12:90-101. [PMID: 21992708 DOI: 10.1111/j.1600-6143.2011.03770.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The proinflammatory cytokine IL-6 plays an important role in controlling T-cell differentiation, especially the development of Th17 and regulatory T cells. To determine the function of IL-6 in regulating allograft rejection and tolerance, BALB/c cardiac grafts were transplanted into wild-type or IL-6-deficient C57BL/6 mice. We observed that production of IL-6 and IFN-γ was upregulated during allograft rejection in untreated wild-type mice. In IL-6-deficient mice, IFN-γ production was greater than that observed in wild-type controls, suggesting that IL-6 production affects Th1/Th2 balance during allograft rejection. CD28-B7 blockade by CTLA4-Ig inhibited IFN-γ production in C57BL/6 recipients, but had no effect on the production of IL-6. Although wild-type C57BL/6 recipients treated with CTLA4-Ig rejected fully MHC-mismatched BALB/c heart transplants, treatment of IL-6-deficient mice with CTLA4-Ig resulted in graft acceptance. Allograft acceptance appeared to result from the combined effect of costimulatory molecule blockade and IL-6-deficiency, which limited the differentiation of effector cells and promoted the migration of regulatory T cells into the grafts. These data suggest that the blockade of IL-6, or its signaling pathway, when combined with strategies that inhibit Th1 responses, has a synergistic effect on the promotion of allograft acceptance. Thus, targeting the effects of IL-6 production may represent an important part of costimulation blockade-based strategies to promote allograft acceptance and tolerance.
Collapse
Affiliation(s)
- X Zhao
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Fogal B, Yi T, Wang C, Rao DA, Lebastchi A, Kulkarni S, Tellides G, Pober JS. Neutralizing IL-6 reduces human arterial allograft rejection by allowing emergence of CD161+ CD4+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:6268-80. [PMID: 22084439 DOI: 10.4049/jimmunol.1003774] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Perioperative injuries to an allograft exacerbate graft rejection, which in humans is primarily mediated by effector memory T cells. IL-6 transcripts in human coronary artery segments rapidly increase posttransplantation into immunodeficient mouse hosts compared with those of pretransplant specimens and fall dramatically by 30 d. Adoptive transfer of human PBMCs allogeneic to the artery 2 d postoperatively results in T cell infiltrates and intimal expansion 4 wk later. Ab neutralization of human IL-6 reduces the magnitude of intimal expansion and total T cell infiltration but increases the relative expression of CD161 while decreasing other Th17 markers. Coculture of MHC class II-expressing human endothelial cells (ECs) with allogeneic CD4(+) memory T cells results in T cell activation and EC secretion of IL-6. Neutralizing IL-6 in primary allogeneic T cell-EC cocultures results in enhanced T cell proliferation of CD161(+) CD4(+) T cells, reduces total T cell proliferation upon restimulation in secondary cultures (an effect dependent on CD161(+) T cells), increases expression of FOXP3 in CD161(+) T cells, and generates T cells that suppress proliferation of freshly isolated T cells. These data suggest that IL-6 released from injured allograft vessels enhances allogeneic T cell infiltration and intimal expansion in a model of human allograft rejection by inhibiting an increase in CD161(+) regulatory T cells.
Collapse
Affiliation(s)
- Birgit Fogal
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Booth AJ, Grabauskiene S, Wood SC, Lu G, Burrell BE, Bishop DK. IL-6 promotes cardiac graft rejection mediated by CD4+ cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:5764-71. [PMID: 22025555 DOI: 10.4049/jimmunol.1100766] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
IL-6 mediates numerous immunologic effects relevant to transplant rejection; however, its specific contributions to these processes are not fully understood. To this end, we neutralized IL-6 in settings of acute cardiac allograft rejection associated with either CD8(+) or CD4(+) cell-dominant responses. In a setting of CD8(+) cell-dominant graft rejection, IL-6 neutralization delayed the onset of acute rejection while decreasing graft infiltrate and inverting anti-graft Th1/Th2 priming dominance in recipients. IL-6 neutralization markedly prolonged graft survival in the setting of CD4(+) cell-mediated acute rejection and was associated with decreased graft infiltrate, altered Th1 responses, and reduced serum alloantibody. Furthermore, in CD4(+) cell-dominated rejection, IL-6 neutralization was effective when anti-IL-6 administration was delayed by as many as 6 d posttransplant. Finally, IL-6-deficient graft recipients were protected from CD4(+) cell-dominant responses, suggesting that IL-6 production by graft recipients, rather than grafts, is necessary for this type of rejection. Collectively, these observations define IL-6 as a critical promoter of graft infiltration and a shaper of T cell lineage development in cardiac graft rejection. In light of these findings, the utility of therapeutics targeting IL-6 should be considered for preventing cardiac allograft rejection.
Collapse
Affiliation(s)
- Adam Jared Booth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|