1
|
Saha I, Chawla AS, Oliveira APBN, Elfers EE, Warrick K, Meibers HE, Jain VG, Hagan T, Katz JD, Pasare C. Alloreactive memory CD4 T cells promote transplant rejection by engaging DCs to induce innate inflammation and CD8 T cell priming. Proc Natl Acad Sci U S A 2024; 121:e2401658121. [PMID: 39136987 PMCID: PMC11348247 DOI: 10.1073/pnas.2401658121] [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: 02/02/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Alloreactive memory T cells have been implicated as central drivers of transplant rejection. Perplexingly, innate cytokines, such as IL-6, IL-1β, and IL-12, are also associated with rejection of organ transplants. However, the pathways of innate immune activation in allogeneic transplantation are unclear. While the role of microbial and cell death products has been previously described, we identified alloreactive memory CD4 T cells as the primary triggers of innate inflammation. Memory CD4 T cells engaged MHC II-mismatched dendritic cells (DCs), leading to the production of innate inflammatory cytokines. This innate inflammation was independent of several pattern recognition receptors and was primarily driven by TNF superfamily ligands expressed by alloreactive memory CD4 T cells. Blocking of CD40L and TNFα resulted in dampened inflammation, and mice genetically deficient in these molecules exhibited prolonged survival of cardiac allografts. Furthermore, myeloid cell and CD8 T cell infiltration into cardiac transplants was compromised in both CD40L- and TNFα-deficient recipients. Strikingly, we found that priming of naive alloreactive CD8 T cells was dependent on licensing of DCs by memory CD4 T cells. This study unravels the key mechanisms by which alloreactive memory CD4 T cells contribute to destructive pathology and transplant rejection.
Collapse
Affiliation(s)
- Irene Saha
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Amanpreet Singh Chawla
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Ana Paula B. N. Oliveira
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Eileen E. Elfers
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Kathrynne Warrick
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH45220
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Hannah E. Meibers
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH45220
| | - Viral G. Jain
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Thomas Hagan
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| | - Jonathan D. Katz
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH45220
| |
Collapse
|
2
|
Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, Zhang W, Dowdell AK, Kim JI, Andrijevic D, Khalil K, Jaffe IS, Loza BL, Gragert L, Camellato BR, Oliveira MF, O'Brien DP, Chen HM, Weldon E, Gao H, Gandla D, Chang A, Bhatt R, Gao S, Lin X, Reddy KP, Kagermazova L, Habara AH, Widawsky S, Liang FX, Sall J, Loupy A, Heguy A, Taylor SEB, Zhu Y, Michael B, Jiang L, Jian R, Chong AS, Fairchild RL, Linna-Kuosmanen S, Kaikkonen MU, Tatapudi V, Lorber M, Ayares D, Mangiola M, Narula N, Moazami N, Pass H, Herati RS, Griesemer A, Kellis M, Snyder MP, Montgomery RA, Boeke JD, Keating BJ. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med 2024; 30:1448-1460. [PMID: 38760586 DOI: 10.1038/s41591-024-02972-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 05/19/2024]
Abstract
In a previous study, heart xenografts from 10-gene-edited pigs transplanted into two human decedents did not show evidence of acute-onset cellular- or antibody-mediated rejection. Here, to better understand the detailed molecular landscape following xenotransplantation, we carried out bulk and single-cell transcriptomics, lipidomics, proteomics and metabolomics on blood samples obtained from the transplanted decedents every 6 h, as well as histological and transcriptomic tissue profiling. We observed substantial early immune responses in peripheral blood mononuclear cells and xenograft tissue obtained from decedent 1 (male), associated with downstream T cell and natural killer cell activity. Longitudinal analyses indicated the presence of ischemia reperfusion injury, exacerbated by inadequate immunosuppression of T cells, consistent with previous findings of perioperative cardiac xenograft dysfunction in pig-to-nonhuman primate studies. Moreover, at 42 h after transplantation, substantial alterations in cellular metabolism and liver-damage pathways occurred, correlating with profound organ-wide physiological dysfunction. By contrast, relatively minor changes in RNA, protein, lipid and metabolism profiles were observed in decedent 2 (female) as compared to decedent 1. Overall, these multi-omics analyses delineate distinct responses to cardiac xenotransplantation in the two human decedents and reveal new insights into early molecular and immune responses after xenotransplantation. These findings may aid in the development of targeted therapeutic approaches to limit ischemia reperfusion injury-related phenotypes and improve outcomes.
Collapse
Affiliation(s)
- Eloi Schmauch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Brian Piening
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Maedeh Mohebnasab
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jeffrey Stern
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Weimin Zhang
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
| | - Alexa K Dowdell
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | - Jacqueline I Kim
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - David Andrijevic
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Karen Khalil
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Ian S Jaffe
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bao-Li Loza
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Loren Gragert
- Division of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | - Han M Chen
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Elaina Weldon
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Hui Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Divya Gandla
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Chang
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Riyana Bhatt
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Gao
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiangping Lin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Kriyana P Reddy
- Penn Transplant Institute, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alawi H Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sophie Widawsky
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Joseph Sall
- DART Microscopy Laboratory, NYU Langone Health, New York, NY, USA
| | - Alexandre Loupy
- Université Paris Cité, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | - Adriana Heguy
- Genome Technology Center, NYU Langone Health, New York, NY, USA
| | | | - Yinan Zhu
- Division of Molecular Genetics Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Basil Michael
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suvi Linna-Kuosmanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vasishta Tatapudi
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Massimo Mangiola
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Navneet Narula
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Nader Moazami
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Harvey Pass
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | - Ramin S Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | | | - Robert A Montgomery
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Jef D Boeke
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
| | - Brendan J Keating
- Institute for Systems Genetics, NYU Langone Health, New York, NY, USA.
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA.
- Department of Surgery, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Ningoo M, Cruz-Encarnación P, Khilnani C, Heeger PS, Fribourg M. T-cell receptor sequencing reveals selected donor-reactive CD8 + T cell clones resist antithymocyte globulin depletion after kidney transplantation. Am J Transplant 2024; 24:755-764. [PMID: 38141722 PMCID: PMC11070313 DOI: 10.1016/j.ajt.2023.12.016] [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: 05/31/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023]
Abstract
High frequencies of donor-reactive memory T cells in the periphery of transplant candidates prior to transplantation are linked to the development of posttransplant acute rejection episodes and reduced allograft function. Rabbit antithymocyte globulin (rATG) effectively depletes naïve CD4+ and CD8+ T cells for >6 months posttransplant, but rATG's effects on human donor-reactive T cells have not been carefully determined. To address this, we performed T cell receptor β-chain sequencing on peripheral blood mononuclear cells aliquots collected pretransplant and serially posttransplant in 7 kidney transplant recipients who received rATG as induction therapy. We tracked the evolution of the donor-reactive CD4+ and CD8+ T cell repertoires and identified stimulated pretransplant, CTV-(surface dye)-labeled, peripheral blood mononuclear cells from each patient with donor cells or third-party cells. Our analyses showed that while rATG depleted CD4+ T cells in all tested subjects, a subset of donor-reactive CD8+ T cells that were present at high frequencies pretransplant, consistent with expanded memory cells, resisted rATG depletion, underwent posttransplant expansion and were functional. Together, our data support the conclusion that a subset of human memory CD8+ T cells specifically reactive to donor antigens expand in vivo despite induction therapy with rATG and thus have the potential to mediate allograft damage.
Collapse
Affiliation(s)
- Mehek Ningoo
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pamela Cruz-Encarnación
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Calla Khilnani
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter S Heeger
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Miguel Fribourg
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
4
|
Prickler L, Baranyi U, Mengrelis K, Weijler AM, Kainz V, Kratzer B, Steiner R, Mucha J, Rudoph E, Pilat N, Bohle B, Strobl H, Pickl WF, Valenta R, Linhart B, Wekerle T. Adoptive transfer of allergen-expressing B cells prevents IgE-mediated allergy. Front Immunol 2023; 14:1286638. [PMID: 38077381 PMCID: PMC10703460 DOI: 10.3389/fimmu.2023.1286638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Prophylactic strategies to prevent the development of allergies by establishing tolerance remain an unmet medical need. We previously reported that the transfer of autologous hematopoietic stem cells (HSC) expressing the major timothy grass pollen allergen, Phl p 5, on their cell surface induced allergen-specific tolerance in mice. In this study, we investigated the ability of allergen-expressing immune cells (dendritic cells, CD4+ T cells, CD8+ T cells, and CD19+ B cells) to induce allergen-specific tolerance in naive mice and identified CD19+ B cells as promising candidates for allergen-specific cell therapy. Methods For this purpose, CD19+ B cells were isolated from Phl p 5-transgenic BALB/c mice and transferred to naive BALB/c mice, pre-treated with a short course of rapamycin and an anti-CD40L antibody. Subsequently, the mice were subcutaneously sensitized three times at 4-week intervals to Phl p 5 and Bet v 1 as an unrelated control allergen. Allergen-expressing cells were followed in the blood to monitor molecular chimerism, and sera were analyzed for Phl p 5- and Bet v 1-specific IgE and IgG1 levels by RBL assay and ELISA, respectively. In vivo allergen-induced lung inflammation was measured by whole-body plethysmography, and mast cell degranulation was determined by skin testing. Results The transfer of purified Phl p 5-expressing CD19+ B cells to naive BALB/c mice induced B cell chimerism for up to three months and prevented the development of Phl p 5-specific IgE and IgG1 antibody responses for a follow-up period of 26 weeks. Since Bet v 1 but not Phl p 5-specific antibodies were detected, the induction of tolerance was specific for Phl p 5. Whole-body plethysmography revealed preserved lung function in CD19+ B cell-treated mice in contrast to sensitized mice, and there was no Phl p 5-induced mast cell degranulation in treated mice. Discussion Thus, we demonstrated that the transfer of Phl p 5-expressing CD19+ B cells induces allergen-specific tolerance in a mouse model of grass pollen allergy. This approach could be further translated into a prophylactic regimen for the prevention of IgE-mediated allergy in humans.
Collapse
Affiliation(s)
- Lisa Prickler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulrike Baranyi
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Konstantinos Mengrelis
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Anna Marianne Weijler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Verena Kainz
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Romy Steiner
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Jasmin Mucha
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Elisa Rudoph
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Nina Pilat
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Division of Immunology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Winfried Franz Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, National Research Center (NRC), Moscow, Russia
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Birgit Linhart
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|