1
|
Hill AE, Son ET, Paul-Heng M, Wang C, Ratnaseelan S, Denkova M, Faridi P, Braun A, Purcell AW, Mifsud NA, Sharland AF. Discovery of conserved peptide-MHC epitopes for directly alloreactive CD8 + T cells. FRONTIERS IN TRANSPLANTATION 2025; 4:1525003. [PMID: 39949593 PMCID: PMC11814428 DOI: 10.3389/frtra.2025.1525003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025]
Abstract
Mass Spectrometry allied with in-vivo generation of activated alloreactive T cell populations and tetramer screening facilitates the identification of endogenous peptides that are directly recognised in complex with allogeneic Major Histocompatibility class I (MHC I) molecules by alloreactive CD8+ T cells. We had previously used this approach for the discovery of immunogenic self-peptides presented by the allomorph H-2Kb (Kb). In this study, we identified 22 highly immunogenic self-peptides presented by H-2Kd (Kd). Peptide abundance across skin, spleen and liver samples (estimated as the product of the spectral intensity obtained for these samples) was the principal factor influencing recognition of peptide-Kd epitopes. Predicted binding affinity (BA score) and overall peptide hydrophobicity were also independently correlated with immunogenicity, while there was no significant correlation between the IEDB immunogenicity score and the proportion of T cells recognising a given epitope. Eight peptide-Kd epitopes were selected for inclusion in a tetramer panel to detect directly alloreactive CD8+ T cells. This panel bound over 30% of activated alloreactive CD8+ T cells after a prime-boost against Kd. Moreover, the panel identified alloreactive CD8+ T cells within the graft infiltrate, spleen and draining lymph node during rejection of a Kd-bearing heart graft. In conclusion, small animal studies have demonstrated the feasibility of high-throughput approaches for the discovery of pMHC epitopes recognised by directly alloreactive T cells. Translating this approach to the human setting is achievable and will yield both critical insights into the fundamental basis of alloreactivity and powerful tools for immune monitoring in transplantation.
Collapse
Affiliation(s)
- Alexandra E. Hill
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Eric T. Son
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Moumita Paul-Heng
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Shivanjali Ratnaseelan
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Martina Denkova
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Pouya Faridi
- Department of Biochemistry and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Asolina Braun
- Department of Biochemistry and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Anthony W. Purcell
- Department of Biochemistry and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Nicole A. Mifsud
- Department of Biochemistry and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Alexandra F. Sharland
- Transplantation Immunobiology Group, Sydney Medical School, University of Sydney Faculty of Medicine and Health, Sydney, NSW, Australia
| |
Collapse
|
2
|
Ishida M, Masuda T, Sakai N, Nakai-Futatsugi Y, Kamao H, Shiina T, Takahashi M, Sugita S. Graft survival of major histocompatibility complex deficient stem cell-derived retinal cells. COMMUNICATIONS MEDICINE 2024; 4:187. [PMID: 39349587 PMCID: PMC11442691 DOI: 10.1038/s43856-024-00617-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Gene editing of immunomodulating molecules is a potential transplantation strategy to control immune rejection. As we noticed the successful transplantation of retinal pigment epithelium (RPE) derived from embryonic stem cells of a cynomolgus monkey that accidentally lacked MHC class II (MHC-II) molecules, we hypothesized immune rejection could be evaded by suppressing MHC-II. METHODS Gene editing by the Crispr/Cas9 system was performed in induced pluripotent stem cells derived from a cynomolgus monkey (miPSCs) for targeted deletion of the gene coding class II MHC trans-activator (CIITA). Then the CIITA-knocked out miPSCs were differentiated into RPE cells to generate miPSC-derived MHC-II knockout RPE. The MHC-II knockout or wild-type RPEs were transplanted into the eyes of healthy cynomolgus monkeys. All monkeys used in this study were male. RESULTS Here we show when MHC-II knockout RPE are transplanted into monkey eyes, they show suppressed immunogenicity with no infiltration of inflammatory cells, leading to successful engraftment. CONCLUSIONS Our results reasonably evidence the efficacy of MHC-II knockout iPSC-RPE transplants for clinical application.
Collapse
Affiliation(s)
- Masaaki Ishida
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan
- Department of Ophthalmology, Toyama University, Toyama, Japan
| | - Tomohiro Masuda
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan
- VC Cell Therapy Inc, Kobe, Japan
- Ritsumeikan University, Research Organization of Science and Technology, Kusatsu, Japan
| | - Noriko Sakai
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan
- VC Cell Therapy Inc, Kobe, Japan
| | - Yoko Nakai-Futatsugi
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan.
- VC Cell Therapy Inc, Kobe, Japan.
- Ritsumeikan University, Research Organization of Science and Technology, Kusatsu, Japan.
| | - Hiroyuki Kamao
- Department of Ophthalmology, Kawasaki Medical School, Okayama, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Tokai University, School of Medicine, Kanagawa, Isehara, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan
- Ritsumeikan University, Research Organization of Science and Technology, Kusatsu, Japan
- Kobe City Eye Hospital, Department of Ophthalmology, Kobe, Japan
- Vision Care Inc, Kobe, Japan
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Kobe, Japan.
- Kobe City Eye Hospital, Department of Ophthalmology, Kobe, Japan.
- Vision Care Inc, Kobe, Japan.
| |
Collapse
|
3
|
Chen G, Hu X, Huang Y, Xiang X, Pan S, Chen R, Xu X. Role of the immune system in liver transplantation and its implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e444. [PMID: 38098611 PMCID: PMC10719430 DOI: 10.1002/mco2.444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Liver transplantation (LT) stands as the gold standard for treating end-stage liver disease and hepatocellular carcinoma, yet postoperative complications continue to impact survival rates. The liver's unique immune system, governed by a microenvironment of diverse immune cells, is disrupted during processes like ischemia-reperfusion injury posttransplantation, leading to immune imbalance, inflammation, and subsequent complications. In the posttransplantation period, immune cells within the liver collaboratively foster a tolerant environment, crucial for immune tolerance and liver regeneration. While clinical trials exploring cell therapy for LT complications exist, a comprehensive summary is lacking. This review provides an insight into the intricacies of the liver's immune microenvironment, with a specific focus on macrophages and T cells as primary immune players. Delving into the immunological dynamics at different stages of LT, we explore the disruptions after LT and subsequent immune responses. Focusing on immune cell targeting for treating liver transplant complications, we provide a comprehensive summary of ongoing clinical trials in this domain, especially cell therapies. Furthermore, we offer innovative treatment strategies that leverage the opportunities and prospects identified in the therapeutic landscape. This review seeks to advance our understanding of LT immunology and steer the development of precise therapies for postoperative complications.
Collapse
Affiliation(s)
- Guanrong Chen
- The Fourth School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Xin Hu
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Yingchen Huang
- The Fourth School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaonan Xiang
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Sheng Pan
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
| | - Ronggao Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiao Xu
- Zhejiang University School of MedicineHangzhouChina
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceHangzhouChina
- Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
4
|
Yang L, Meng Y, Shi Y, Fang H, Zhang L. Maternal hepatic immunology during pregnancy. Front Immunol 2023; 14:1220323. [PMID: 37457700 PMCID: PMC10348424 DOI: 10.3389/fimmu.2023.1220323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The liver plays pivotal roles in immunologic responses, and correct hepatic adaptations in maternal immunology are required during pregnancy. In this review, we focus on anatomical and immunological maternal hepatic adaptations during pregnancy, including our recent reports in this area. Moreover, we summarize maternal pregnancy-associated liver diseases, including hyperemesis gravidarum; intrahepatic cholestasis of pregnancy; preeclampsia, specifically hemolysis, elevated liver enzymes, and low platelet count syndrome; and acute fatty liver of pregnancy. In addition, the latest information about the factors that regulate hepatic immunology during pregnancy are reviewed for the first time, including human chorionic gonadotropin, estrogen, progesterone, growth hormone, insulin like growth factor 1, oxytocin, adrenocorticotropic hormone, adrenal hormone, prolactin, melatonin and prostaglandins. In summary, the latest progress on maternal hepatic anatomy and immunological adaptations, maternal pregnancy-associated diseases and the factors that regulate hepatic immunology during pregnancy are discussed, which may be used to prevent embryo loss and abortion, as well as pregnancy-associated liver diseases.
Collapse
|
5
|
Son ET, Paul-Heng M, Leong M, Wang C, Hill AE, Denkova M, Purcell AW, Mifsud NA, Sharland AF. Screening self-peptides for recognition by mouse alloreactive CD8 + T cells using direct ex vivo multimer staining. STAR Protoc 2023; 4:101943. [PMID: 36525346 PMCID: PMC9792548 DOI: 10.1016/j.xpro.2022.101943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/31/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
Here, we present a protocol to identify immunogenic self-peptide/allogeneic major histocompatibility complex (MHC) epitopes. We describe the generation of enriched alloreactive CD8+ T cells by priming mice with a skin graft expressing the allogeneic MHC class I molecule of interest, followed by boosting with a liver-specific AAV vector encoding the heavy chain of that donor MHC allomorph. We then use a peptide-exchange approach to assemble a range of peptide-MHC (pMHC) multimers for measuring recognition of the various epitopes by these alloreactive T cells. For complete details on the use and execution of this protocol, please refer to Son et al. (2021).1.
Collapse
Affiliation(s)
- Eric T Son
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Moumita Paul-Heng
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Mario Leong
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Chuanmin Wang
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Alexandra E Hill
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Martina Denkova
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia
| | - Anthony W Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Nicole A Mifsud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, NSW 2006, Australia.
| |
Collapse
|
6
|
Son ET, Faridi P, Paul-Heng M, Leong ML, English K, Ramarathinam SH, Braun A, Dudek NL, Alexander IE, Lisowski L, Bertolino P, Bowen DG, Purcell AW, Mifsud NA, Sharland AF. The self-peptide repertoire plays a critical role in transplant tolerance induction. J Clin Invest 2021; 131:e146771. [PMID: 34428180 PMCID: PMC8553557 DOI: 10.1172/jci146771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
While direct allorecognition underpins both solid organ allograft rejection and tolerance induction, the specific molecular targets of most directly alloreactive CD8+ T cells have not been defined. In this study, we used a combination of genetically engineered major histocompatibility complex class I (MHC I) constructs, mice with a hepatocyte-specific mutation in the class I antigen-presentation pathway, and immunopeptidomic analysis to provide definitive evidence for the contribution of the peptide cargo of allogeneic MHC I molecules to transplant tolerance induction. We established a systematic approach for the discovery of directly recognized pMHC epitopes and identified 17 strongly immunogenic H-2Kb-associated peptides recognized by CD8+ T cells from B10.BR (H-2k) mice, 13 of which were also recognized by BALB/c (H-2d) mice. As few as 5 different tetramers used together were able to identify a high proportion of alloreactive T cells within a polyclonal population, suggesting that there are immunodominant allogeneic MHC-peptide complexes that can account for a large component of the alloresponse.
Collapse
Affiliation(s)
- Eric T. Son
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Pouya Faridi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Moumita Paul-Heng
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Mario L. Leong
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Kieran English
- Liver Immunology Group and AW Morrow Gastroenterology and Liver Centre, The University of Sydney and Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Sri H. Ramarathinam
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Asolina Braun
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Nadine L. Dudek
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ian E. Alexander
- Gene Therapy Research Unit, Children’s Medical Research Institute, The University of Sydney, Faculty of Medicine and Health and Sydney Children’s Hospitals Network, Westmead, New South Wales, Australia
- The University of Sydney, Sydney Medical School, Discipline of Child and Adolescent Health, Westmead, New South Wales, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
- Vector and Genome Engineering Facility, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
- Military Institute of Medicine, Laboratory of Molecular Oncology and Innovative Therapies, Warsaw, Poland
| | - Patrick Bertolino
- Liver Immunology Group and AW Morrow Gastroenterology and Liver Centre, The University of Sydney and Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - David G. Bowen
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Liver Immunology Group and AW Morrow Gastroenterology and Liver Centre, The University of Sydney and Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Anthony W. Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Nicole A. Mifsud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Alexandra F. Sharland
- Transplantation Immunobiology Group, University of Sydney Central Clinical School, Charles Perkins Centre, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Damo M, Wilson DS, Watkins EA, Hubbell JA. Soluble N-Acetylgalactosamine-Modified Antigens Enhance Hepatocyte-Dependent Antigen Cross-Presentation and Result in Antigen-Specific CD8 + T Cell Tolerance Development. Front Immunol 2021; 12:555095. [PMID: 33746941 PMCID: PMC7965950 DOI: 10.3389/fimmu.2021.555095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 02/10/2021] [Indexed: 01/27/2023] Open
Abstract
Hepatocytes compose up to 80% of the total liver and have been indicated as important players in the induction of immunologic tolerance in this organ. We show that hepatocytes possess the molecular machinery required for the cross-presentation of extracellular antigens. Using a derivative of the model antigen ovalbumin (OVA) covalently modified with a polymer containing multiple N-acetylgalactosamine residues (pGal-OVA) that enhance extracellular antigen uptake by mimicking the glycome of apoptotic debris, we show efficient hepatocyte-dependent induction of cross-tolerance of both adoptively transferred OT-I cells and endogenous OVA-specific CD8+ T lymphocytes, for example inducing tolerance to OVA-expressing skin transplants. Our study confirms that hepatocytes are capable of inducing peripheral tolerogenesis and provides proof of concept that they may be a valuable candidate for in vivo targeted tolerogenic treatments.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 2/immunology
- ATP Binding Cassette Transporter, Subfamily B, Member 2/metabolism
- Acetylgalactosamine/immunology
- Adoptive Transfer/methods
- Animals
- Antigen Presentation/immunology
- Antigens/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Cross-Priming/immunology
- Hepatocytes/cytology
- Hepatocytes/immunology
- Immune Tolerance/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Ovalbumin/immunology
- Skin Transplantation/methods
- Solubility
- Vesicular Transport Proteins/immunology
- Vesicular Transport Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Martina Damo
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
- Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - D. Scott Wilson
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
- Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Elyse A. Watkins
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Jeffrey A. Hubbell
- Institute for Molecular Engineering, University of Chicago, Chicago, IL, United States
- Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
8
|
Thomson AW, Vionnet J, Sanchez-Fueyo A. Understanding, predicting and achieving liver transplant tolerance: from bench to bedside. Nat Rev Gastroenterol Hepatol 2020; 17:719-739. [PMID: 32759983 DOI: 10.1038/s41575-020-0334-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
In the past 40 years, liver transplantation has evolved from a high-risk procedure to one that offers high success rates for reversal of liver dysfunction and excellent patient and graft survival. The liver is the most tolerogenic of transplanted organs; indeed, immunosuppressive therapy can be completely withdrawn without rejection of the graft in carefully selected, stable long-term liver recipients. However, in other recipients, chronic allograft injury, late graft failure and the adverse effects of anti-rejection therapy remain important obstacles to improved success. The liver has a unique composition of parenchymal and immune cells that regulate innate and adaptive immunity and that can promote antigen-specific tolerance. Although the mechanisms underlying liver transplant tolerance are not well understood, important insights have been gained into how the local microenvironment, hepatic immune cells and specific molecular pathways can promote donor-specific tolerance. These insights provide a basis for the identification of potential clinical biomarkers that might correlate with tolerance or rejection and for the development of novel therapeutic targets. Innovative approaches aimed at promoting immunosuppressive drug minimization or withdrawal include the adoptive transfer of donor-derived or recipient-derived regulatory immune cells to promote liver transplant tolerance. In this Review, we summarize and discuss these developments and their implications for liver transplantation.
Collapse
Affiliation(s)
- Angus W Thomson
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Julien Vionnet
- Institute of Liver Studies, Medical Research Council (MRC) Centre for Transplantation, School of Immunology and Infectious Diseases, King's College London University, King's College Hospital, London, UK.,Transplantation Center, University Hospital of Lausanne, Lausanne, Switzerland.,Service of Gastroenterology and Hepatology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, Medical Research Council (MRC) Centre for Transplantation, School of Immunology and Infectious Diseases, King's College London University, King's College Hospital, London, UK
| |
Collapse
|
9
|
Lei H, Reinke P, Volk HD, Lv Y, Wu R. Mechanisms of Immune Tolerance in Liver Transplantation-Crosstalk Between Alloreactive T Cells and Liver Cells With Therapeutic Prospects. Front Immunol 2019; 10:2667. [PMID: 31803188 PMCID: PMC6877506 DOI: 10.3389/fimmu.2019.02667] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Liver transplantation (LTx) is currently the most powerful treatment for end-stage liver disease. Although liver allograft is more tolerogenic compared to other solid organs, the majority of LTx recipients still require long-term immune suppression (IS) to control the undesired alloimmune responses, which can lead to severe side effects. Thus, understanding the mechanism of liver transplant tolerance and crosstalk between immune cells, especially alloreactive T cells and liver cells, can shed light on more specific tolerance induction strategies for future clinical translation. In this review, we focus on alloreactive T cell mediated immune responses and their crosstalk with liver sinusoidal endothelial cells (LSECs), hepatocytes, hepatic stellate cells (HSCs), and cholangiocytes in transplant setting. Liver cells mainly serve as antigen presenting cells (APCs) to T cells, but with low expression of co-stimulatory molecules. Crosstalk between them largely depends on the different expression of adhesion molecules and chemokine receptors. Inflammatory cytokines secreted by immune cells further elaborate this crosstalk and regulate the fate of naïve T cells differentiation within the liver graft. On the other hand, regulatory T cells (Tregs) play an essential role in inducing and keeping immune tolerance in LTx. Tregs based adoptive cell therapy provides an excellent therapeutic option for clinical transplant tolerance induction. However, many questions regarding cell therapy still need to be solved. Here we also address the current clinical trials of adoptive Tregs therapy and other tolerance induction strategies in LTx, together with future challenges for clinical translation from bench to bedside.
Collapse
Affiliation(s)
- Hong Lei
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Berlin Center of Advanced Therapies, Berlin, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
10
|
Poupiot J, Costa Verdera H, Hardet R, Colella P, Collaud F, Bartolo L, Davoust J, Sanatine P, Mingozzi F, Richard I, Ronzitti G. Role of Regulatory T Cell and Effector T Cell Exhaustion in Liver-Mediated Transgene Tolerance in Muscle. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 15:83-100. [PMID: 31649958 PMCID: PMC6804827 DOI: 10.1016/j.omtm.2019.08.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022]
Abstract
The pro-tolerogenic environment of the liver makes this tissue an ideal target for gene replacement strategies. In other peripheral tissues such as the skeletal muscle, anti-transgene immune response can result in partial or complete clearance of the transduced fibers. Here, we characterized liver-induced transgene tolerance after simultaneous transduction of liver and muscle. A clinically relevant transgene, α-sarcoglycan, mutated in limb-girdle muscular dystrophy type 2D, was fused with the SIINFEKL epitope (hSGCA-SIIN) and expressed with adeno-associated virus vectors (AAV-hSGCA-SIIN). Intramuscular delivery of AAV-hSGCA-SIIN resulted in a strong inflammatory response, which could be prevented and reversed by concomitant liver expression of the same antigen. Regulatory T cells and upregulation of checkpoint inhibitor receptors were required to establish and maintain liver-mediated peripheral tolerance. This study identifies the fundamental role of the synergy between Tregs and upregulation of checkpoint inhibitor receptors in the liver-mediated control of anti-transgene immunity triggered by muscle-directed gene transfer.
Collapse
Affiliation(s)
- Jérôme Poupiot
- INTEGRARE, Genethon, INSERM, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | | | | | - Pasqualina Colella
- INTEGRARE, Genethon, INSERM, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Laurent Bartolo
- UMR 1151, Necker-Institut Enfants Malades-Molecular Medicine Center, Paris, France
| | - Jean Davoust
- UMR 1151, Necker-Institut Enfants Malades-Molecular Medicine Center, Paris, France
| | | | | | - Isabelle Richard
- INTEGRARE, Genethon, INSERM, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Univ Evry, Université Paris-Saclay, 91002 Evry, France
| |
Collapse
|
11
|
Bartolo L, Li Chung Tong S, Chappert P, Urbain D, Collaud F, Colella P, Richard I, Ronzitti G, Demengeot J, Gross DA, Mingozzi F, Davoust J. Dual muscle-liver transduction imposes immune tolerance for muscle transgene engraftment despite preexisting immunity. JCI Insight 2019; 4:127008. [PMID: 31167976 DOI: 10.1172/jci.insight.127008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
Immune responses to therapeutic transgenes are a potential hurdle to treat monogenic muscle disorders. These responses result from the neutralizing activity of transgene-specific B cells and cytotoxic T cells recruited upon gene transfer. We explored here how dual muscle-liver expression of a foreign transgene allows muscle transgene engraftment after adenoassociated viral vector delivery. We found in particular that induction of transgene-specific tolerance is imposed by concurrent muscle and liver targeting, resulting in the absence of CD8+ T cell responses to the transgene. This tolerance can be temporally decoupled, because transgene engraftment can be achieved in muscle weeks after liver transduction. Importantly, transgene-specific CD8+ T cell tolerance can be established despite preexisting immunity to the transgene. Whenever preexisting, transgene-specific CD4+ and CD8+ memory T cell responses are present, dual muscle-liver transduction turns polyclonal, transgene-specific CD8+ T cells into typically exhausted T cells with high programmed cell death 1 (PD-1) expression and lack of IFN-γ production. Our results demonstrate that successful transduction of muscle tissue can be achieved through liver-mediated control of humoral and cytotoxic T cell responses, even in the presence of preexisting immunity to the muscle-associated transgene.
Collapse
Affiliation(s)
- Laurent Bartolo
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| | - Stéphanie Li Chung Tong
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| | - Pascal Chappert
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| | - Dominique Urbain
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| | - Fanny Collaud
- Integrare Research Unit UMR S951, Genethon, INSERM, Université Evry, Université Paris Saclay, École Pratique des Hautes Études, Evry, France
| | - Pasqualina Colella
- Integrare Research Unit UMR S951, Genethon, INSERM, Université Evry, Université Paris Saclay, École Pratique des Hautes Études, Evry, France
| | - Isabelle Richard
- Integrare Research Unit UMR S951, Genethon, INSERM, Université Evry, Université Paris Saclay, École Pratique des Hautes Études, Evry, France
| | - Giuseppe Ronzitti
- Integrare Research Unit UMR S951, Genethon, INSERM, Université Evry, Université Paris Saclay, École Pratique des Hautes Études, Evry, France
| | | | - David A Gross
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| | - Federico Mingozzi
- Integrare Research Unit UMR S951, Genethon, INSERM, Université Evry, Université Paris Saclay, École Pratique des Hautes Études, Evry, France
| | - Jean Davoust
- Institut Necker Enfants-Malades, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France; INSERM, U1151, Paris, France; CNRS UMR 8253, Paris, France
| |
Collapse
|