1
|
Heron SD, Shaw J, Dapprich J. Anti-HLA antibodies may be a subset of polyreactive immunoglobulins generated after viral superinfection. Transpl Immunol 2025; 90:102197. [PMID: 39954820 DOI: 10.1016/j.trim.2025.102197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Chronic rejection remains an obstacle to long-term allograft survival. Donor-specific anti-HLA antibodies (DSA) play a significant role in causing chronic antibody-mediated allograft rejection. Exposure to mismatched HLA antigens via transfusion, pregnancy, or transplanted tissue has been described in the literature as an immunogenic stimulus of anti-HLA antibodies. Yet anti-HLA antibodies also develop in the absence of traditional sensitization events and molecular mimicry has been postulated as a stimulus for these naturally occurring alloantibodies. While heterologous reactivity has been documented between virus components and allogeneic T cells, there is insufficient evidence to support the development of anti-HLA antibodies from viral components. We hypothesized that anti-HLA antibodies may develop following viral coinfection or superinfection. The objectives of this investigation included: 1) developing an in-silico algorithm to identify viral peptide components that exhibit HLA-specific homology, and 2) identifying cellular changes that take place during ischemia/reperfusion injury which could facilitate the generation of novel anti-HLA antibodies from viral sources. We developed the neoepitope transplant rejection and autoimmune disease (NETRAD) algorithm to identify amino acid sequence homology between viral envelope proteins and HLA. The algorithm integrates post-translational protein modifications that are consistent with ischemia/reperfusion injury. Seventy-two HLA-specific epitopes were demarcated as examples using this approach. In conclusion, we present in-silico evidence which supports the identification of anti-HLA antibodies as a subset of polyreactive antibodies generated from stress-modified viral envelope proteins. Remarkably, each targeted HLA epitope associated with a distinct anti-HLA antibody could be consistently attributed to a major envelope glycoprotein component of Epstein Barr virus. Transplant Immunology manuscript # TRIM-D-24-00351. Dryad data repository:https://doi.org/10.5061/dryad.qjq2bvqpq.
Collapse
Affiliation(s)
| | - Jim Shaw
- Cellanalytics, Chesterbrook, PA USA
| | | |
Collapse
|
2
|
Khosravi-Maharlooei M, Vecchione A, Danzl N, Li HW, Nauman G, Madley R, Waffarn E, Winchester R, Ruiz A, Ding X, Fousteri G, Sykes M. Follicular helper- and peripheral helper-like T cells drive autoimmune disease in human immune system mice. eLife 2025; 13:RP99389. [PMID: 40293219 PMCID: PMC12037178 DOI: 10.7554/elife.99389] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Human immune system (HIS) mice constructed in various ways are widely used for investigations of human immune responses to pathogens, transplants, and immunotherapies. In HIS mice that generate T cells de novo from hematopoietic progenitors, T cell-dependent multisystem autoimmune disease occurs, most rapidly when the human T cells develop in the native NOD.Cg- Prkdcscid Il2rgtm1Wjl (NSG) mouse thymus, where negative selection is abnormal. Disease develops very late when human T cells develop in human fetal thymus grafts, where robust negative selection is observed. We demonstrate here that PD-1+CD4+ peripheral (Tph) helper-like and follicular (Tfh) helper-like T cells developing in HIS mice can induce autoimmune disease. Tfh-like cells were more prominent in HIS mice with a mouse thymus, in which the highest levels of IgG were detected in plasma, compared to those with a human thymus. While circulating IgG and IgM antibodies were autoreactive to multiple mouse antigens, in vivo depletion of B cells and antibodies did not delay the development of autoimmune disease. Conversely, adoptive transfer of enriched Tfh- or Tph-like cells induced disease and autoimmunity-associated B cell phenotypes in recipient mice containing autologous human APCs without T cells. Tfh/Tph cells from mice with a human thymus expanded and induced disease more rapidly than those originating in a murine thymus, implicating HLA-restricted T cell-APC interactions in this process. Since Tfh, Tph, autoantibodies, and lymphopenia-induced proliferation (LIP) have all been implicated in various forms of human autoimmune disease, the observations here provide a platform for the further dissection of human autoimmune disease mechanisms and therapies.
Collapse
Affiliation(s)
- Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- Department of Immunology, Department of Biochemistry and Molecular Biology, Mayo ClinicPhoenixUnited States
| | - Andrea Vecchione
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- San Raffaele HospitalMilanItaly
| | - Nichole Danzl
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Hao Wei Li
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Grace Nauman
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Elizabeth Waffarn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Robert Winchester
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Amanda Ruiz
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | - Xiaolan Ding
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| | | | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- Department of Microbiology and Immunology, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
- Department of Surgery, Columbia University Medical Center, Columbia UniversityNew YorkUnited States
| |
Collapse
|
3
|
Khosravi-Maharlooei M, Vecchione A, Danzl N, Li HW, Nauman G, Madley R, Waffarn E, Winchester R, Ruiz A, Ding X, Fousteri G, Sykes M. Follicular helper- and peripheral helper-like T cells drive autoimmune disease in human immune system mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.02.591692. [PMID: 38746102 PMCID: PMC11092663 DOI: 10.1101/2024.05.02.591692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Human immune system (HIS) mice constructed in various ways are widely used for investigations of human immune responses to pathogens, transplants and immunotherapies. In HIS mice that generate T cells de novo from hematopoietic progenitors, T cell-dependent multisystem autoimmune disease occurs, most rapidly when the human T cells develop in the native NOD.Cg- Prkdc scid Il2rg tm1Wjl (NSG) mouse thymus, where negative selection is abnormal. Disease develops very late when human T cells develop in human fetal thymus grafts, where robust negative selection is observed. We demonstrate here that PD-1 + CD4 + peripheral (Tph) helper-like and follicular (Tfh) helper-like T cells developing in HIS mice can induce autoimmune disease. Tfh- like cells were more prominent in HIS mice with a mouse thymus, in which the highest levels of IgG were detected in plasma, compared to those with a human thymus. While circulating IgG and IgM antibodies were autoreactive to multiple mouse antigens, in vivo depletion of B cells and antibodies did not delay the development of autoimmune disease. Conversely, adoptive transfer of enriched Tfh- or Tph-like cells induced disease and autoimmunity-associated B cell phenotypes in recipient mice containing autologous human APCs without T cells. Tfh/Tph cells from mice with a human thymus expanded and induced disease more rapidly than those originating in a murine thymus, implicating HLA-restricted T cell-APC interactions in this process. Since Tfh, Tph, autoantibodies and lymphopenia-induced proliferation (LIP) have all been implicated in various forms of human autoimmune disease, the observations here provide a platform for the further dissection of human autoimmune disease mechanisms and therapies.
Collapse
|
4
|
Hertel A, Aguiar T, Mashiko S, Núñez S, Moore C, Gao B, Ausmeier M, Roy P, Zorn E. Clones reactive to apoptotic cells and specific chemical adducts are prevalent among human thymic B cells. Front Immunol 2024; 15:1462126. [PMID: 39497815 PMCID: PMC11532181 DOI: 10.3389/fimmu.2024.1462126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction Thymus resident B cells were described more than 40 years ago. In early human life, these cells are found predominantly in the medulla and overwhelmingly display an unswitched IgM+ phenotype. The reactivity of thymic IgM B cells, however, is still unclear. Methods Here, we generated 120 IgM-producing B cell clones from 3 separate thymus specimens obtained from infant, adolescent, and adult donors. Using flow cytometry and a unique high-dimensional ELISA platform, we investigated the clones' reactivity to apoptotic cells as well as to common chemical adducts exposed on modified amino acids and other macromolecules. Results Regardless of the age, approximately 30-40% of thymic IgM B cells reacted to apoptotic cells. Further, 30-40% displayed reactivity to at least one adduct, including malondialdehyde, Homocysteine, and NEDD 8. Four distinct reactivity patterns were identified through this profiling. Notably, a significant association was observed between reactivity to apoptotic cells, and to one or more adducts, suggesting that the same determinants were recognized in both assays. Additionally, thymic IgM B cells reactive to adducts were more likely to recognize intra-nuclear or intra-cytoplasmic structures in Hep-2 cells as revealed by immunofluorescence staining. Conclusion/Discussion Collectively, our findings suggest that thymic IgM B cells actively uptake apoptotic bodies and cellular debris in the medulla by binding specific chemical adducts. This mechanism could underpin their antigen-presenting function and further support their role in T-cell negative selection.
Collapse
Affiliation(s)
- Andrea Hertel
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Medical Department IV - Großhadern, LMU University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Talita Aguiar
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Shunya Mashiko
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Sarah Núñez
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Ciencia y Vida, Santiago, Chile
| | - Carolina Moore
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Baoshan Gao
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Mattea Ausmeier
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Poloumi Roy
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Emmanuel Zorn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
5
|
Killian JT, Glenn King R, Lucander ACK, Kizziah JL, Fucile CF, Diaz-Avalos R, Qiu S, Silva-Sanchez A, Mousseau BJ, Macon KJ, Callahan AR, Yang G, Emon Hossain M, Akther J, Good DB, Kelso S, Houp JA, Rosenblum F, Porrett PM, Ong SC, Kumar V, Saphire EO, Kearney JF, Randall TD, Rosenberg AF, Green TJ, Lund FE. HLA topography enforces shared and convergent immunodominant B cell and antibody alloresponses in transplant recipients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.31.534734. [PMID: 37034637 PMCID: PMC10081326 DOI: 10.1101/2023.03.31.534734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Donor-specific antibody (DSA) responses against human leukocyte antigen (HLA) proteins mismatched between kidney transplant donors and recipients cause allograft loss. The rules governing the immunogenicity of non-self donor HLA are poorly understood. Using single-cell, molecular, structural, and proteomic techniques, we profiled the HLA-specific B cell response in the kidney and blood of a transplant recipient with antibody-mediated rejection (AMR). We observed an immunodominant B cell antibody response focused on topographically exposed, solvent-accessible mismatched HLA residues along the peptide-binding groove - a subregion comprising only 20% of the HLA molecule. We further demonstrated that, even within a diverse cohort of transplant recipients, the B cell alloresponse consistently converges on this same immunodominant subregion on the crown of the HLA molecule. Based on these findings, we propose that B cell immunodominance in transplant rejection relies on antigenic topography, and we suggest that this link could be exploited for organ matching and therapeutics.
Collapse
|
6
|
Han J, Rushakoff J, Moayedi Y, Henricksen E, Lee R, Luikart H, Shalakhti O, Gragert L, Benck L, Malinoski D, Kobashigawa J, Teuteberg J, Khush KK, Patel J, Kransdorf E. HLA sensitization is associated with an increased risk of primary graft dysfunction after heart transplantation. J Heart Lung Transplant 2024; 43:387-393. [PMID: 37802261 DOI: 10.1016/j.healun.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 08/30/2023] [Accepted: 09/24/2023] [Indexed: 10/08/2023] Open
Abstract
Primary graft dysfunction (PGD) is a leading cause of early morbidity and mortality following heart transplantation (HT). We sought to determine the association between pretransplant human leukocyte antigen (HLA) sensitization, as measured using the calculated panel reactive antibody (cPRA) value, and the risk of PGD. METHODS Consecutive adult HT recipients (n = 596) from 1/2015 to 12/2019 at 2 US centers were included. Severity of PGD was based on the 2014 International Society for Heart and Lung Transplantation consensus statement. For each recipient, unacceptable HLA antigens were obtained and locus-specific cPRA (cPRA-LS) and pre-HT donor-specific antibodies (DSA) were assessed. RESULTS Univariable logistic modeling showed that peak cPRA-LS for all loci and HLA-A was associated with increased severity of PGD as an ordinal variable (all loci: OR 1.78, 95% CI: 1.01-1.14, p = 0.025, HLA-A: OR 1.14, 95% CI: 1.03-1.26, p = 0.011). Multivariable analysis showed peak cPRA-LS for HLA-A, recipient beta-blocker use, total ischemic time, donor age, prior cardiac surgery, and United Network for Organ Sharing status 1 or 2 were associated with increased severity of PGD. The presence of DSA to HLA-B was associated with trend toward increased risk of mild-to-moderate PGD (OR 2.56, 95% CI: 0.99-6.63, p = 0.053), but DSA to other HLA loci was not associated with PGD. CONCLUSIONS Sensitization for all HLA loci, and specifically HLA-A, is associated with an increased severity of PGD. These factors should be included in pre-HT risk stratification to minimize the risk of PGD.
Collapse
Affiliation(s)
- Jiho Han
- Section of Cardiology, Department of Medicine, University of Chicago Medical Center, Chicago, Illinois
| | - Josh Rushakoff
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yasbanoo Moayedi
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Erik Henricksen
- Department of Transplant, Stanford Health Care, Stanford, California
| | - Roy Lee
- Department of Transplant, Stanford Health Care, Stanford, California
| | - Helen Luikart
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Omar Shalakhti
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Loren Gragert
- Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Lillian Benck
- Division of Cardiology, NorthShore University Health System, Chicago, Illinois
| | - Darren Malinoski
- Critical Care and Acute Care Surgery, Oregon Health Sciences University, Portland, Oregon
| | - Jon Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jeffrey Teuteberg
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Kiran K Khush
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, California
| | - Jignesh Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Evan Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
7
|
Mashiko S, Shihab RR, See SB, Schahadat LGZ, Aguiar TFM, Roy P, Porcheray F, Zorn E. Broad responses to chemical adducts shape the natural antibody repertoire in early infancy. SCIENCE ADVANCES 2023; 9:eade8872. [PMID: 37172087 PMCID: PMC10181178 DOI: 10.1126/sciadv.ade8872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Natural antibodies are an integral part of innate humoral immunity yet their development and polyreactive nature are still enigmatic. Here, we show that characteristic monoclonal natural antibodies recognize common chemical moieties or adducts, supporting the view that polyreactive antibodies may often correspond to anti-adduct antibodies. We next examined the development of immunoglobulin M (IgM) and IgG to 81 ubiquitous adducts from birth to old age. Newborn IgM only reacted to a limited number of consensus determinants. This highly restricted neonatal repertoire abruptly diversified around 6 months of age through the development of antibodies to environmental antigens and age-driven epigenetic modifications. In contrast, the IgG repertoire was diverse across the entire life span. Our studies reveal an unrecognized component of humoral immunity directed to common adducts. These findings set the ground for further investigations into the role of anti-adduct B cell responses in homeostatic functions and pathological conditions.
Collapse
Affiliation(s)
- Shunya Mashiko
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronzon R Shihab
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah B See
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Luca G Z Schahadat
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Talita F M Aguiar
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Poulomi Roy
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Fabrice Porcheray
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emmanuel Zorn
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Sayin I, Chong AS. Beyond Adaptive Alloreactivity: Contribution of Innate B Cells to Allograft Inflammation and Rejection. Transplantation 2023; 107:98-104. [PMID: 36404414 PMCID: PMC9772142 DOI: 10.1097/tp.0000000000004377] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Innate B cells are a heterogeneous group of cells that function in maintaining homeostatic levels of circulating natural antibodies and being the first line of defense against infections. Innate B-1 cells and marginal zone B cells may relocate to lymphoid follicles and differentiate into cytokine and antibody-secreting cells in T-independent and T-dependent manners. Although marginal zone B cells are widely described in humans, the presence of B-1 cells is more controversial. Here, we review the basic features of the innate B-cell subsets identified in mice and their equivalent in humans, as well as their potential roles in transplantation. We summarize the findings of Cascalho and colleagues on the unexpected protective role of tumor necrosis factor receptor superfamily member 13B in regulating circulating levels of protective natural immunoglobulin M, and the studies by Zorn and colleagues on the potential pathogenic role for polyreactive innate B cells infiltrating allograft explants. Finally, we discuss our studies that took a transcriptomic approach to identify innate B cells infiltrating kidney allografts with antibody-mediated rejection and to demonstrate that local antigens within the allograft together with inflammation may induce a loss of B-cell tolerance.
Collapse
Affiliation(s)
- Ismail Sayin
- Department of Surgery, The University of Chicago, Chicago, Illinois, United States
| | - Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
9
|
Innate-like B Cells: Local Drivers of Non-HLA Immunity in Rejecting Kidney Allografts? Transplantation 2022; 106:234-235. [PMID: 35100225 DOI: 10.1097/tp.0000000000003989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Innate-like self-reactive B cells infiltrate human renal allografts during transplant rejection. Nat Commun 2021; 12:4372. [PMID: 34272370 PMCID: PMC8285506 DOI: 10.1038/s41467-021-24615-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Intrarenal B cells in human renal allografts indicate transplant recipients with a poor prognosis, but how these cells contribute to rejection is unclear. Here we show using single-cell RNA sequencing that intrarenal class-switched B cells have an innate cell transcriptional state resembling mouse peritoneal B1 or B-innate (Bin) cells. Antibodies generated by Bin cells do not bind donor-specific antigens nor are they enriched for reactivity to ubiquitously expressed self-antigens. Rather, Bin cells frequently express antibodies reactive with either renal-specific or inflammation-associated antigens. Furthermore, local antigens can drive Bin cell proliferation and differentiation into plasma cells expressing self-reactive antibodies. These data show a mechanism of human inflammation in which a breach in organ-restricted tolerance by infiltrating innate-like B cells drives local tissue destruction. Intrarenal B cells are indicative of poor prognosis in human renal allografts. Here the authors use single cell RNA sequencing to examine how intrarenal B cells contribute to renal rejection and find a population of innate B cells reactive to renal-specific or inflammation-associated antigens.
Collapse
|
11
|
Siu JH, Motallebzadeh R, Pettigrew GJ. Humoral autoimmunity after solid organ transplantation: Germinal ideas may not be natural. Cell Immunol 2020; 354:104131. [DOI: 10.1016/j.cellimm.2020.104131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022]
|
12
|
Abstract
Antibody-mediated rejection continues to hinder long-term survival of solid organ allografts. Natural antibodies (Nabs) with polyreactive and autoreactive properties have recently emerged as potential contributors to antibody-mediated graft rejection. This review discusses Nabs, their functions in health and disease, their significance in rejection following kidney, heart, and lung transplantation, and their implication in serum reactivity to key antigens associated with rejection. Finally, potential effector mechanisms of Nabs in the context of transplantation are explored.
Collapse
|
13
|
Chong AS, Rothstein D, Safa K, Riella LV. Outstanding questions in transplantation: B cells, alloantibodies, and humoral rejection. Am J Transplant 2019; 19:2155-2163. [PMID: 30803121 PMCID: PMC6691724 DOI: 10.1111/ajt.15323] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/06/2019] [Accepted: 02/16/2019] [Indexed: 01/25/2023]
Abstract
Over the past three decades, improved immunosuppression has significantly reduced T cell-mediated acute rejection rates, but long-term graft survival rates have seen only marginal improvement. The cause of late graft loss has been under intense investigation, and chronic antibody-mediated rejection (AMR) has been identified as one of the leading causes, thus providing a strong rationale for basic science investigation into donor-specific B cells and antibodies in transplantation and ways to mitigate their pathogenicity. In 2018, the American Society of Transplantation launched a community-wide online discussion of Outstanding Questions in Transplantation, and the topic of B cell biology and donor-specific antibody prevention emerged as a major area of interest to the community, leading to a highly engaged dialogue, with comments from basic and translational scientists as well as physicians (http://community.myast.org/communities/community-home/digestviewer). We have summarized this discussion from a bedside to bench perspective and have organized this review into outstanding questions within the paradigm that AMR is a leading cause of graft loss in the clinic, and points of view that challenge aspects of this paradigm. We also highlight opportunities for basic and translational scientists to contribute to the resolution of these questions, mapping important future directions for the transplant research field.
Collapse
Affiliation(s)
- Anita S. Chong
- Department of Surgery, The University of Chicago,Corresponding author: Anita S. Chong, PhD, 5841 S. Maryland Ave, Chicago, IL 60637, Ofc: 773-702-5521; Fax: 773-702-5517;
| | - David Rothstein
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh
| | - Kassem Safa
- Transplant Center and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School
| | - Leonardo V. Riella
- Schuster Transplantation Research Center, Renal Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Desensitization in the Era of Precision Medicine: Moving From the Bench to Bedside. Transplantation 2019; 103:1574-1581. [DOI: 10.1097/tp.0000000000002737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
15
|
Dieudé M, Cardinal H, Hébert MJ. Injury derived autoimmunity: Anti-perlecan/LG3 antibodies in transplantation. Hum Immunol 2019; 80:608-613. [PMID: 31029511 DOI: 10.1016/j.humimm.2019.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/08/2019] [Accepted: 04/13/2019] [Indexed: 01/02/2023]
Abstract
Ischemic, immunologic or pharmacological stressors can induce vascular injury and endothelial apoptosis in organ donors, in transplant candidates due to the impact of end stage organ failure on the vasculature, and in association with peri-transplantation events. Vascular injury may shape innate and adaptive immune responses, leading to dysregulation in the balance between tolerance and immunoreactivity to vascular-derived antigens. Mounting evidence shows that the early stages of apoptosis, characterized by the absence of membrane permeabilization, are prone to trigger various modes of intercellular communication allowing neoantigen production, exposure, or both. In this review, we present the evidence for the release of LG3, an immunogenic fragment of perlecan, as a consequence of caspase-3 dependent vascular apoptosis leading to the genesis of anti-LG3 autoantibodies and the consequences of these autoantibodies in native and transplanted kidneys.
Collapse
Affiliation(s)
- Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Canadian Donation and Transplantation Research Program, Canada; Université de Montréal, Canada.
| | - Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Canadian Donation and Transplantation Research Program, Canada; Université de Montréal, Canada.
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Canadian Donation and Transplantation Research Program, Canada; Université de Montréal, Canada.
| |
Collapse
|
16
|
Padet L, Dieudé M, Karakeussian‐Rimbaud A, Yang B, Turgeon J, Cailhier J, Cardinal H, Hébert M. New insights into immune mechanisms of antiperlecan/LG3 antibody production: Importance of T cells and innate B1 cells. Am J Transplant 2019; 19:699-712. [PMID: 30129231 PMCID: PMC6519043 DOI: 10.1111/ajt.15082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/25/2018] [Accepted: 07/29/2018] [Indexed: 01/25/2023]
Abstract
Autoantibodies against perlecan/LG3 (anti-LG3) have been associated with increased risks of delayed graft function, acute rejection, and reduced long-term survival. High titers of anti-LG3 antibodies have been found in de novo renal transplants recipients in the absence of allosensitizing or autoimmune conditions. Here, we seek to understand the pathways controlling anti-LG3 production prior to transplantation. Mice immunized with recombinant LG3 produce concomitantly IgM and IgG anti-LG3 antibodies suggesting a memory response. ELISpot confirmed the presence of LG3-specific memory B cells in nonimmunized mice. Purification of B1 and B2 subtypes identified peritoneal B1 cells as the major source of memory B cells reactive to LG3. Although nonimmunized CD4-deficient mice were found to express LG3-specific memory B cells, depletion of CD4+ T cells in wild type mice during immunization significantly decreased anti-LG3 production. These results demonstrate that B cell memory to LG3 is T cell independent but that production of anti-LG3 antibodies requires T cell help. Further supporting an important role for T cells in controlling anti-LG3 levels, we found that human renal transplant recipients show a significant decrease in anti-LG3 titers upon the initiation of CNI-based immunosuppression. Collectively, these results identify T cell targeting interventions as a means of reducing anti-LG3 levels in renal transplant patients.
Collapse
Affiliation(s)
- Lauriane Padet
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| | - Mélanie Dieudé
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Annie Karakeussian‐Rimbaud
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Bing Yang
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| | - Julie Turgeon
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Jean‐François Cailhier
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Héloïse Cardinal
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada
| | - Marie‐Josée Hébert
- Research CentreCentre Hospitalier de l'Université de Montréal (CRCHUM)MontrealQuebecCanada,Canadian National Transplant Research ProgramEdmontonAlbertaCanada,Université de MontréalMontrealQuebecCanada
| |
Collapse
|
17
|
New Answers to Old Conundrums: What Antibodies, Exosomes and Inflammasomes Bring to the Conversation. Canadian National Transplant Research Program International Summit Report. Transplantation 2018; 102:209-214. [PMID: 28731910 PMCID: PMC5802265 DOI: 10.1097/tp.0000000000001872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antibody-mediated injury is a major cause of allograft dysfunction and loss. Antibodies to ABH(O) blood group antigens are classic mediators of ABO-incompatible graft rejection, whereas donor-specific anti-HLA antibodies and, more recently, autoantibodies are appreciated as important contributors to allograft inflammation and dysfunction. In August 2016, the International Summit of the Canadian National Transplant Research Program focused on recent advances in the field of antibody-mediated rejection. Here, we describe work presented and discussed at the meeting, with a focus on 3 major themes: the importance of (1) natural antibodies and autoantibodies, (2) tissue injury-derived exosomes and autoimmunity, (3) inflammasome activation and innate immune responses in regulating allograft inflammation and dysfunction. Finally, we explore novel areas of therapeutic intervention that have recently emerged from these 3 major and overlapping fields of transplantation research.
Collapse
|
18
|
Kinloch AJ, Kaiser Y, Wolfgeher D, Ai J, Eklund A, Clark MR, Grunewald J. In Situ Humoral Immunity to Vimentin in HLA-DRB1*03 + Patients With Pulmonary Sarcoidosis. Front Immunol 2018; 9:1516. [PMID: 30038611 PMCID: PMC6046378 DOI: 10.3389/fimmu.2018.01516] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022] Open
Abstract
Vimentin has been implicated in pulmonary sarcoidosis as a T-cell autoantigen, particularly in the context of HLA-DRB1*03, the Vα2.3/Vβ22 T-cell receptor (TCR), and Löfgren’s syndrome. As vimentin is a known antigenic target in B-cell-mediated autoimmunity, we investigated in situ humoral anti-vimentin responses in pulmonary sarcoidosis and their relationship with HLA-DRB1*03. Sarcoid and healthy control (HC) lung biopsies were analyzed by multi-color confocal microscopy for B-cells, T-cells, proliferation, and vimentin, and compared to tonsillectomy tissue. Bronchoalveolar lavage fluid (BALF) and serum from 48 sarcoidosis patients and 15 healthy volunteers were typed for HLA-DRB1*03 and titrated for antibodies to full-length vimentin, vimentin truncations, and total IgG and IgA by ELISA. Presence of extracellular vimentin in BALF was determined by mass spectrometry and T-cell populations measured by flow cytometry. Sarcoid lung samples, especially from HLA-DRB1*03+ patients, contained vimentin-rich tertiary lymphoid structures and corresponding BALF was highly enriched for both IgG and IgA anti-vimentin antibody (AVA) titers. Furthermore, sarcoidosis patient BALF AVA concentrations (expressed as arbitrary units per milligram of total immunoglobulin isotype) correlated with the percentage of CD4+ T-cells expressing the Vα2.3/Vβ22 TCR. BALF antibody reactivity to the vimentin N-terminus was most prominent in HCs, whereas reactivity to the C-terminus (VimC-term) was enriched in the sarcoid lung. Specifically, HLA-DRB1*03+ patient BALF contained higher concentrations of anti-VimC-term antibodies than BALF from both HCs and HLA-DRB1*03− patients. Consistent with the lung as a site of AVA production, the concentration of AVAs in BALF was dramatically higher than in matched serum samples. Overall, there was a poor correlation between BALF and serum AVA concentrations. Together, these studies reveal the presence of linked in situ recognition of vimentin by both T- and B-cells in HLA-DRB1*03+ sarcoidosis patients, associated with a selective humoral immune response to the vimentin C-terminus.
Collapse
Affiliation(s)
- Andrew J Kinloch
- Department of Medicine, Section of Rheumatology, Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, United States
| | - Ylva Kaiser
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Don Wolfgeher
- Proteomics Core Laboratory, Cummings Life Science Center, University of Chicago, Chicago, IL, United States
| | - Junting Ai
- Department of Medicine, Section of Rheumatology, Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, United States
| | - Anders Eklund
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology, Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, United States
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
19
|
Chong AS, Ansari MJ. Heterogeneity of memory B cells. Am J Transplant 2018; 18:779-784. [PMID: 29359404 PMCID: PMC5962275 DOI: 10.1111/ajt.14669] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 12/29/2017] [Accepted: 12/30/2017] [Indexed: 01/25/2023]
Abstract
Potential solid organ transplant recipients broadly sensitized to HLA have long wait times, low transplant rates and poor outcomes. The new kidney allocation system has improved access to the most highly sensitized recipients; however, their long-term outcomes are unknown. Emerging data suggest that memory B cell repertoire is broader than the plasma cell repertoire, therefore, despite refinements in anti-HLA antibody detection technology, donor-specific HLA- specific memory B cells may in fact be present in some, if not most, highly sensitized recipients with no detectable donor-specific antibodies. In addition, new findings have underscored the heterogeneity in memory B cell generation, and in the signals that determine memory versus plasma cell fate during primary antigen encounter, as well as memory B cell differentiation upon antigen reencounter into plasma cells or reentry into germinal centers to subsequently emerge as higher affinity and class-switched plasma cells. Thus, heterogeneity memory B cells generation may affect the efficacy of specific immunomodulation during the recall response. We propose that the ability to quantify donor-specific B cell in transplant recipients is urgently required to provide insights into the mechanisms of sensitization and recall, and for the early detection of acute and chronic AMR.
Collapse
Affiliation(s)
- Anita S Chong
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, IL, USA
- Division of Nephrology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - M Javeed Ansari
- Department of Surgery, Section of Transplantation, University of Chicago, Chicago, IL, USA
- Division of Nephrology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection (ABMR), especially in its chronic manifestation, is increasingly recognized as a leading cause of late graft loss following solid organ transplantation. In recent years, autoantibodies have emerged as a significant component of the humoral response to allografts alongside anti-human leukocyte antigen antibodies. These include polyreactive antibodies also known as natural antibodies (Nabs) secreted by innate B cells. A hallmark of Nabs is their capacity to bind altered self such as oxidized lipids on apoptotic cells. This review provides an overview of these overlooked antibodies and their implication in the pathophysiology of ABMR. RECENT FINDINGS New evidence reported in the past few years support a contribution of immunoglobulin (Ig) G Nabs to ABMR. Serum IgG Nabs levels are significantly higher in patients with ABMR compared with control kidney transplant recipients with stable graft function. Pretransplant IgG Nabs are also associated with ABMR and late graft loss. IgG Nabs are almost exclusively of the IgG1 and IgG3 subclasses and have the capacity to activate complement. SUMMARY In conclusion, Nabs are important elements in host immune responses to solid organ grafts. The recent description of their implication in ABMR and late kidney graft loss warrants further investigation into their pathogenic potential.
Collapse
|
21
|
Alloimmune-induced intragraft lymphoid neogenesis promotes B-cell tolerance breakdown that accelerates chronic rejection. Curr Opin Organ Transplant 2017; 21:368-74. [PMID: 27258579 DOI: 10.1097/mot.0000000000000329] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Antibody-mediated rejection (AMR) has emerged as a leading cause of allograft loss in solid organ transplantation. A better understanding of AMR immunopathology is a prerequisite to improve its management. RECENT FINDINGS The prevalent dogma considers that AMR is the consequence of a thymo-dependent B-cell response against donor-specific polymorphic antigens (mainly mismatched human leukocyte antigen molecules).Nevertheless, antibodies directed against nonpolymorphic antigens expressed by the graft are also generated during chronic rejection and can contribute to allograft destruction. This implies that a breakdown of self-tolerance occurs during chronic rejection. Accumulating evidence suggests that this event occurs inside the ectopic 'tertiary' lymphoid tissue that develops within rejected allografts.Thus, AMR should be viewed as a complex interplay between allo- and autoimmune humoral responses. SUMMARY The interplay between allo- and autoimmune humoral responses in chronic rejection highlights several unmet medical issues like better diagnosis tools are needed to screen recipients for nonhuman leukocyte antigen alloantibodies and autoantibodies, therapeutic strategies shall aim at blocking the response against alloantigens but also the breakdown of self-tolerance that occurs within tertiary lymphoid tissue.
Collapse
|
22
|
Prevalence of polyreactive innate clones among graft--infiltrating B cells in human cardiac allograft vasculopathy. J Heart Lung Transplant 2017; 37:385-393. [PMID: 29055600 DOI: 10.1016/j.healun.2017.09.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/09/2017] [Accepted: 09/21/2017] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) has been associated with graft-infiltrating B cells, although their characteristics are still unclear. In this study we examined the frequency, localization and reactivity profile of graft-infiltrating B cells to determine their contribution to the pathophysiology of CAV. METHODS B cells, plasma cells and macrophages were examined by immunohistochemistry in 56 allografts with CAV, 49 native failed hearts and 25 autopsy specimens. A total of 102 B-cell clones were immortalized directly from the infiltrates of 3 fresh cardiac samples with CAV. Their secreted antibodies were assessed using enzyme-linked immunoassay and flow cytometry. RESULTS B-cell infiltration was observed around coronary arteries in 93% of allograft explants with CAV. Comparatively, intragraft B cells were less frequent and less dense in the intraventricular myocardium from where routine biopsies are obtained. Plasma cells and macrophages were also detected in 85% and 95% of explants, respectively. Remarkably, B-cell infiltrates were not associated with circulating donor-specific antibodies (DSA) or prior episodes of antibody-mediated rejection (AMR). Among all B-cell clones generated from 3 explants with CAV, a majority secreted natural antibodies reactive to multiple autoantigens and apoptotic cells, a characteristic of innate B cells. CONCLUSIONS Our study reveals a high frequency of infiltrating B cells around the coronary arteries of allografts with CAV, independent of DSA or AMR. These cells are enriched for innate B cells with a polyreactive profile. The findings shift the focus from conventional DSA-producing B cells to the potentially pathogenic polyreactive B cells in the development of clinical CAV.
Collapse
|
23
|
Cernoch M, Viklicky O. Complement in Kidney Transplantation. Front Med (Lausanne) 2017; 4:66. [PMID: 28611987 PMCID: PMC5447724 DOI: 10.3389/fmed.2017.00066] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/09/2017] [Indexed: 12/12/2022] Open
Abstract
The complement system is considered to be an important part of innate immune system with a significant role in inflammation processes. The activation can occur through classical, alternative, or lectin pathway, resulting in the creation of anaphylatoxins C3a and C5a, possessing a vast spectrum of immune functions, and the assembly of terminal complement cascade, capable of direct cell lysis. The activation processes are tightly regulated; inappropriate activation of the complement cascade plays a significant role in many renal diseases including organ transplantation. Moreover, complement cascade is activated during ischemia/reperfusion injury processes and influences delayed graft function of kidney allografts. Interestingly, complement system has been found to play a role in both acute cellular and antibody-mediated rejections and thrombotic microangiopathy. Therefore, complement system may represent an interesting therapeutical target in kidney transplant pathologies.
Collapse
Affiliation(s)
- Marek Cernoch
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Ondrej Viklicky
- Transplant Laboratory, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia.,Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| |
Collapse
|
24
|
Abstract
BACKGROUND Previous studies identified B cell gene signatures and predominance of specific B cell subsets as a marker of operational tolerance after kidney transplantation. These findings suggested a role for B cells in the establishment or maintenance of tolerance. Here we analyzed B cell recovery in 4 subjects, 3 of whom achieved tolerance after combined kidney/bone marrow transplantation. METHODS Peripheral B cell subsets were examined longitudinally by flow cytometry. Immunoglobulin heavy chain repertoire analysis was performed using next-generation sequencing. Lastly, the patients' serum reactivity to HLA was assessed by Luminex. RESULTS B cell counts recovered approximately 1 year posttransplant except for 1 subject who experienced delayed reconstitution. This subject resumed immunosuppression for acute rejection at 10 months posttransplant and underwent preemptive retransplantation at 3 years for chronic rejection. B cell recovery was accompanied by a high frequency of CD20 + CD24CD38 transitional B cells and a diversified clonal repertoire. However, all 4 subjects showed prevalence of CD20 + CD27+ memory B cells around 6 months posttransplant when B cell counts were still low and the clonal B cell repertoire very limited. The predominance of memory B cells was also associated with high levels of somatically mutated immunoglobulin heavy chain variable sequences and transient serum reactivity to HLA. CONCLUSIONS Our observations reveal the presence of memory B cells early posttransplant that likely escaped the preparative regimen at a time consistent with the establishment of tolerance. Further studies are warranted to characterize the functional properties of these persisting memory cells and evaluate their potential contribution to tolerance induction.
Collapse
|
25
|
Sullivan JA, Jankowska-Gan E, Hegde S, Pestrak MA, Agashe VV, Park AC, Brown ME, Kernien JF, Wilkes DS, Kaufman DB, Greenspan DS, Burlingham WJ. Th17 Responses to Collagen Type V, kα1-Tubulin, and Vimentin Are Present Early in Human Development and Persist Throughout Life. Am J Transplant 2017; 17:944-956. [PMID: 27801552 PMCID: PMC5626015 DOI: 10.1111/ajt.14097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/29/2016] [Accepted: 10/14/2016] [Indexed: 01/25/2023]
Abstract
T helper 17 (Th17)-dependent autoimmune responses can develop after heart or lung transplantation and are associated with fibro-obliterative forms of chronic rejection; however, the specific self-antigens involved are typically different from those associated with autoimmune disease. To investigate the basis of these responses, we investigated whether removal of regulatory T cells or blockade of function reveals a similar autoantigen bias. We found that Th17 cells specific for collagen type V (Col V), kα1-tubulin, and vimentin were present in healthy adult peripheral blood mononuclear cells, cord blood, and fetal thymus. Using synthetic peptides and recombinant fragments of the Col V triple helical region (α1[V]), we compared Th17 cells from healthy donors with Th17 cells from Col V-reactive heart and lung patients. Although the latter responded well to α1(V) fragments and peptides in an HLA-DR-restricted fashion, Th17 cells from healthy persons responded in an HLA-DR-restricted fashion to fragments but not to peptides. Col V, kα1-tubulin, and vimentin are preferred targets of a highly conserved, hitherto unknown, preexisting Th17 response that is MHC class II restricted. These data suggest that autoimmunity after heart and lung transplantation may result from dysregulation of an intrinsic mechanism controlling airway and vascular homeostasis.
Collapse
Affiliation(s)
- Jeremy A Sullivan
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792,To whom correspondence should be addressed: 600 Highland Avenue, Room G4/702, Madison, WI 53792. Tel: (608) 263-0119 Fax: (608)262-6280,
| | - Ewa Jankowska-Gan
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - Subramanya Hegde
- Current Address: Abbvie Bio-Research Center, 100 Research Dr., Worcester, MA 01605
| | - Matthew A Pestrak
- Current Address: Department of Surgery, Ohio State University, 410 W 10th Ave, Columbus, OH 43210
| | - Vrushali V Agashe
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - Arick C Park
- Department of Cell & Regenerative Biology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - Matthew E Brown
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - John F Kernien
- Department of Cell & Regenerative Biology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - David S Wilkes
- Department of Medicine, University of Indiana, 340 W 10th St Suite 6200 Indianapolis, IN 46202
| | - Dixon B Kaufman
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - Daniel S Greenspan
- Department of Cell & Regenerative Biology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792
| | - William J Burlingham
- Department of Surgery, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792,To whom correspondence should be addressed: 600 Highland Avenue, Room G4/702, Madison, WI 53792. Tel: (608) 263-0119 Fax: (608)262-6280,
| |
Collapse
|
26
|
Clinical value of non-HLA antibodies in kidney transplantation: Still an enigma? Transplant Rev (Orlando) 2016; 30:195-202. [DOI: 10.1016/j.trre.2016.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/22/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022]
|
27
|
Cellular immune profile of kidney transplant patients developing anti-HLA antibodies during childhood. Pediatr Nephrol 2016; 31:1001-10. [PMID: 26692023 DOI: 10.1007/s00467-015-3274-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND In the field of kidney transplantation, identifying early signatures of humoral rejection is a key challenge. METHODS We investigated the presence of anti-HLA antibodies and the distribution of lymphocyte subpopulations in 77 kidney-transplanted children and young adults compared to 23 healthy controls. Moreover, we tested whether the presence of anti-HLA antibodies could be related to modification in lymphocyte phenotype. Finally, we correlated the presence of anti-HLA antibodies and specific alteration of lymphocyte subsets with clinical outcomes. RESULTS In kidney-transplanted children who developed anti-HLA antibodies, we observed an expansion of double-negative B cells (CD19 + CD27-IgD-), indicating premature aging of this compartment. Moreover, we reported signs of impaired B cell regulation, indicated by a higher IL-21R+ B cell frequency associated with an abnormal increase of follicular helper T cells. Finally, a considerable reduction in CD8+ effector T and invariant Natural killer T (NKT) cells was observed. The stability of graft function over time is significantly correlated with the frequency of peripheral effector CD4+ and CD8+ T cells and invariant NKT cells. CONCLUSIONS This study supports the usefulness of lymphocyte subset as one of a spectrum of early diagnostic tools required to identify patients at risk of developing donor alloimmune response.
Collapse
|
28
|
Evidence to Support a Contribution of Polyreactive Antibodies to HLA Serum Reactivity. Transplantation 2016; 100:217-26. [PMID: 26285015 DOI: 10.1097/tp.0000000000000840] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Assessing the serum reactivity to HLA is essential for the evaluation of transplant candidates and the follow-up of allograft recipients. In this study, we look for evidence at the clonal level that polyreactive antibodies cross-reactive to apoptotic cells and multiple autoantigens can also react to HLA and contribute to the overall serum reactivity. METHODS We immortalized B cell clones from the blood of 2 kidney transplant recipients and characterized their reactivity to self-antigens, apoptotic cells as well as native, denatured, and cryptic HLA determinants using enzyme-linked immunosorbent assay (ELISA), immunofluorescence, flow cytometry and Luminex assays. We also assessed the reactivity of 300 pretransplant serum specimens to HLA and apoptotic cells. RESULTS We report here 4 distinct B cell clones cross-reactive to self and HLA class I. All 4 clones reacted to numerous HLA class I alleles but did not appear to target canonical "shared" epitopes. In parallel experiments, we observed a strong correlation between IgG reactivity to HLA and apoptotic cells in pretransplant serum samples collected from 300 kidney transplant recipients. Further analysis revealed that samples with higher reactivity to apoptotic cells displayed significantly higher class I percent panel-reactive antibodies compared to samples with low reactivity to apoptotic cells. CONCLUSIONS We provide here (1) proof of principle at the clonal level that human polyreactive antibodies can cross-react to HLA, multiple self-antigens and apoptotic cells and (2) supportive evidence that polyreactive antibodies contribute to overall HLA reactivity in the serum of patients awaiting kidney transplant.
Collapse
|
29
|
Abstract
BACKGROUND B-cell infiltrates are common in rejected kidney allografts, yet their composition is still unclear. The aim of our study was to characterize the clonal composition of B-cell infiltrates of rejected human kidney grafts. METHODS We used a molecular approach to characterize the partial B-cell repertoires of 5 failed human kidney grafts with detectable B-cell infiltrates. A comparison between the intragraft and blood repertoire was also conducted for 1 case. RESULTS Redundant sequences were observed in both blood and graft, although the level of clonal amplification was significantly higher for the graft. Somatic hypermutations (SHMs) were also more frequent in sequences found in the graft compared to the blood. The rate of nonsilent mutations was significantly higher in complementarity determining regions (CDRs) compared to framework regions in blood sequences as well as in graft sequences found at low frequency. In contrast, this preferential distribution was lost in sequences found at high frequency in the graft, suggesting a lack of affinity maturation in situ. Lastly, follicular dendritic cells were undetectable in CD20 infiltrates in all samples examined. CONCLUSIONS We provide here evidence that B-cell clones expand and undergo SHMs in situ. However, the even distribution of nonsilent SHM in high-frequency graft sequences together with the absence of follicular dendritic cells do not support the view that infiltrating B cells are part of functional germinal centers.
Collapse
|
30
|
Chong AS, Alegre ML. Transplantation tolerance and its outcome during infections and inflammation. Immunol Rev 2015; 258:80-101. [PMID: 24517427 DOI: 10.1111/imr.12147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Much progress has been made toward understanding the mechanistic basis of transplantation tolerance in experimental models, which implicates clonal deletion of alloreactive T and B cells, induction of cell-intrinsic hyporesponsiveness, and dominant regulatory cells mediating infectious tolerance and linked suppression. Despite encouraging success in the laboratory, achieving tolerance in the clinic remains challenging, although the basis for these challenges is beginning to be understood. Heterologous memory alloreactive T cells generated by infections prior to transplantation have been shown to be a critical barrier to tolerance induction. Furthermore, infections at the time of transplantation and tolerance induction provide a pro-inflammatory milieu that alters the stability and function of regulatory T cells as well as the activation requirements and differentiation of effector T cells. Thus, infections can result in enhanced alloreactivity, resistance to tolerance induction, and destabilization of the established tolerance state. We speculate that these experimental findings have relevance to the clinic, where infections have been associated with allograft rejection and may be a causal event precipitating the loss of grafts after long periods of stable operational tolerance. Understanding the mechanisms by which infections prevent and destabilize tolerance can lead to therapies that promote stable life-long tolerance in transplant recipients.
Collapse
Affiliation(s)
- Anita S Chong
- Section of Transplantation, Department of Surgery, The University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
31
|
Gao B, Moore C, Porcheray F, Rong C, Abidoglu C, DeVito J, Paine R, Girouard TC, Saidman SL, Schoenfeld D, Levin B, Wong W, Elias N, Schuetz C, Rosales IA, Fu Y, Zorn E. Pretransplant IgG reactivity to apoptotic cells correlates with late kidney allograft loss. Am J Transplant 2014; 14:1581-91. [PMID: 24935695 PMCID: PMC4120834 DOI: 10.1111/ajt.12763] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 02/03/2014] [Accepted: 02/20/2014] [Indexed: 01/25/2023]
Abstract
Preexisting serum antibodies have long been associated with graft loss in transplant recipients. While most studies have focused on HLA-specific antibodies, the contribution of non-HLA-reactive antibodies has been largely overlooked. We have recently characterized mAbs secreted by B cell clones derived from kidney allograft recipients with rejection that bind to apoptotic cells. Here, we assessed the presence of such antibodies in pretransplant serum from 300 kidney transplant recipients and examined their contribution to the graft outcomes. Kaplan-Meier survival analysis revealed that patients with high pretransplant IgG reactivity to apoptotic cells had a significantly increased rate of late graft loss. The effect was only apparent after approximately 1 year posttransplant. Moreover, the association between pretransplant IgG reactivity to apoptotic cells and graft loss was still significant after excluding patients with high reactivity to HLA. This reactivity was almost exclusively mediated by IgG1 and IgG3 with complement fixing and activating properties. Overall, our findings support the view that IgG reactive to apoptotic cells contribute to presensitization. Taking these antibodies into consideration alongside anti-HLA antibodies during candidate evaluation would likely improve the transplant risk assessment.
Collapse
Affiliation(s)
- Baoshan Gao
- Transplant Center, The First Hospital of Jilin University, Changchun, China,Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolina Moore
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrice Porcheray
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunshu Rong
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Cem Abidoglu
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Julie DeVito
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rosemary Paine
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy C. Girouard
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susan L. Saidman
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Schoenfeld
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
| | - Bruce Levin
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Waichi Wong
- Division of Nephrology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Nahel Elias
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Schuetz
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivy A. Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yaowen Fu
- Transplant Center, The First Hospital of Jilin University, Changchun, China
| | - Emmanuel Zorn
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Corresponding author: Emmanuel Zorn, Massachusetts General Hospital, Transplant Center, Thier 807, 55 Fruit Street, Boston, MA 02114, , Tel: (617) 643-3675, Fax: (617) 724-3471
| |
Collapse
|
32
|
Kawai T, Sachs DH, Sprangers B, Spitzer TR, Saidman SL, Zorn E, Tolkoff-Rubin N, Preffer F, Crisalli K, Gao B, Wong W, Morris H, LoCascio SA, Sayre P, Shonts B, Williams WW, Smith RN, Colvin RB, Sykes M, Cosimi AB. Long-term results in recipients of combined HLA-mismatched kidney and bone marrow transplantation without maintenance immunosuppression. Am J Transplant 2014; 14:1599-611. [PMID: 24903438 PMCID: PMC4228952 DOI: 10.1111/ajt.12731] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/05/2014] [Accepted: 03/05/2014] [Indexed: 01/25/2023]
Abstract
We report here the long-term results of HLA-mismatched kidney transplantation without maintenance immunosuppression (IS) in 10 subjects following combined kidney and bone marrow transplantation. All subjects were treated with nonmyeloablative conditioning and an 8- to 14-month course of calcineurin inhibitor with or without rituximab. All 10 subjects developed transient chimerism, and in seven of these, IS was successfully discontinued for 4 or more years. Currently, four subjects remain IS free for periods of 4.5-11.4 years, while three required reinstitution of IS after 5-8 years due to recurrence of original disease or chronic antibody-mediated rejection. Of the 10 renal allografts, three failed due to thrombotic microangiopathy or rejection. When compared with 21 immunologically similar living donor kidney recipients treated with conventional IS, the long-term IS-free survivors developed significantly fewer posttransplant complications. Although most recipients treated with none or two doses of rituximab developed donor-specific antibody (DSA), no DSA was detected in recipients treated with four doses of rituximab. Although further revisions of the current conditioning regimen are planned in order to improve consistency of the results, this study shows that long-term stable kidney allograft survival without maintenance IS can be achieved following transient mixed chimerism induction.
Collapse
Affiliation(s)
- T. Kawai
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA,Corresponding author: Tatsuo Kawai,
| | - D. H. Sachs
- Transplantation Biology Research Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - B. Sprangers
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - T. R. Spitzer
- Bone Marrow Transplant Unit, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - S. L. Saidman
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - E. Zorn
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - N. Tolkoff-Rubin
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - F. Preffer
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - K. Crisalli
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - B. Gao
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - W. Wong
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - H. Morris
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - S. A. LoCascio
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - P. Sayre
- Immune Tolerance Network, San Francisco, CA
| | - B. Shonts
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - W. W. Williams
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - R.-N. Smith
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - R. B. Colvin
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - M. Sykes
- Columbia Center for Translational Immunology, Columbia University, New York, NY
| | - A. B. Cosimi
- Transplant Center, Harvard Medical School, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
33
|
Iyer HS, Jackson AM, Montgomery RA. Sensitized Patients, Transplant, and Management. CURRENT TRANSPLANTATION REPORTS 2014. [DOI: 10.1007/s40472-014-0010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Increased negative impact of donor HLA-specific together with non-HLA-specific antibodies on graft outcome. Transplantation 2014; 97:595-601. [PMID: 24162250 DOI: 10.1097/01.tp.0000436927.08026.a8] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND De novo donor HLA-specific (dnDSA) and non-HLA antibodies including antiangiotensin type 1 receptor antibodies (AT1R-abs) have been associated with antibody-mediated rejection (AMR) and decreased graft survival as well as cellular-mediated rejection (CMR) and early onset of microvasculopathy in heart transplantation. The aim of our study was to determine the impact of anti-AT1R-ab and anti-donor HLA-specific antibody (DSA) on clinical outcomes. METHODS Pretransplant and posttransplant sera from 200 recipients transplanted between May 2007 and August 2011 were tested for DSA (Luminex-based single antigen bead assay) and AT1R-ab (enzyme-linked immunosorbent assay). Two cutoff levels (≥ 17 and ≥ 12 units) were used to define high and intermediate binding of AT1R-ab. Clinical parameters examined were 5-year AMR/CMR (≥ grade 2), coronary artery vasculopathy, and survival. RESULTS At 2 years after transplant, freedom from AMR and/or CMR was 95.4% for those with no DSA (n=175), 66.9% for those with dnDSA (n=19), and 25% for those with DSA at transplant (n=6) (P<0.0001). Neither ≥ 17 nor ≥ 12 units of pretransplant levels indicated a significant difference in freedom from AMR and/or CMR. When both dnDSA and AT1R-ab ≥ 17 or ≥ 12 units were considered, freedom from AMR and/or CMR decreased to 50% and 45% (P<0.0001), respectively. Coronary artery vasculopathy and survival were not significantly impacted. CONCLUSIONS These results show the increased negative impact of dnDSA and AT1R-ab on freedom from AMR and/or CMR and an increased hazard ratio when both parameters are considered. Both HLA- and non-HLA-specific antibodies seem to impact graft outcome in heart transplantation.
Collapse
|
35
|
Mengel M, Chong A, Rothstein DM, Zorn E, Maltzman JS. AST Cutting Edge of Transplantation 2013 Meeting Report: a comprehensive look at B cells and antibodies in transplantation. Am J Transplant 2014; 14:524-30. [PMID: 24674597 PMCID: PMC4046165 DOI: 10.1111/ajt.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 01/25/2023]
Abstract
Antibody-mediated rejection (ABMR) represents a significant clinical challenge for solid organ transplantation. Mechanistic understanding of ABMR is incomplete and diagnostic accuracy for ABMR is limited, and as a result, targeted treatment remains elusive and new treatment modalities are difficult to validate. Three hundred twenty-six participants from 15 countries met for the first Cutting Edge of Transplantation (CEOT) symposium organized by the American Society of Transplantation (AST) in Chandler, Arizona, February 14-16, 2013. During the 3-day interactive symposium, presentations, moderated poster sessions and round table discussions addressed cutting edge knowledge of B and plasma cell biology, mechanisms of antibody-mediated tissue injury, advances and limitations in ABMR diagnostics, as well as current and potential new treatment options for ABMR. The outcome of the meeting identified the following unmet needs for: (a) improved understanding of the regulation of B cell maturation and antibody response to enable targeted therapies; (b) more precise diagnostics of ABMR, including molecular pathology, risk stratification by sensitive antibody testing and monitoring of treatment effects; and (c) innovative multicenter trial designs that enhance observational power, in particular, in assessing synergistic multimodality therapies with reduced toxicities.
Collapse
Affiliation(s)
- Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Anita Chong
- Section of Transplant Surgery, University of Chicago, Chicago, IL
| | - David. M. Rothstein
- Thomas E Starzl Transplant Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Emanuel Zorn
- Harvard Med School Transplant Center, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Jonathan S. Maltzman
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
36
|
Porcheray F, Fraser JW, Gao B, McColl A, DeVito J, Dargon I, Helou Y, Wong W, Girouard TC, Saidman SL, Colvin RB, Palmisano A, Maggiore U, Vaglio A, Smith RN, Zorn E. Polyreactive antibodies developing amidst humoral rejection of human kidney grafts bind apoptotic cells and activate complement. Am J Transplant 2013; 13:2590-600. [PMID: 23919437 PMCID: PMC3864117 DOI: 10.1111/ajt.12394] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 06/05/2013] [Accepted: 06/20/2013] [Indexed: 01/25/2023]
Abstract
Antibody mediated rejection (AMR) is associated with a variety of graft-reactive antibodies following kidney transplant. To characterize these antibodies, we immortalized 107 B cell clones from a patient with AMR. In a previous study, we showed that six clones were reacting to multiple self-antigens as well as to HLA and MICA for two of them, thus displaying a pattern of polyreactivity. We show here that all six polyreactive clones also reacted to apoptotic but not viable cells. More generally we observed a nearly perfect overlap between polyreactivity and reactivity to apoptotic cells. Functionally, polyreactive antibodies can activate complement, resulting in the deposition of C3d and C4d at the surface of target cells. Testing the serum of 88 kidney transplant recipients revealed a significantly higher IgG reactivity to apoptotic cells in AMR patients than in patients with stable graft function. Moreover, total IgG purified from AMR patients had increased complement activating properties compared to IgG from non-AMR patients. Overall, our studies show the development of polyreactive antibodies cross-reactive to apoptotic cells during AMR. Further studies are now warranted to determine their contribution to the detection of C4d in graft biopsies as well as their role in the pathophysiology of AMR.
Collapse
Affiliation(s)
- Fabrice Porcheray
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - James W. Fraser
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Baoshan Gao
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA,Transplant Center, First Hospital of Jilin University, Changchun, China
| | - Aisleen McColl
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Julie DeVito
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Ian Dargon
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Ynes Helou
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Waichi Wong
- Renal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Timothy C. Girouard
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Susan L. Saidman
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Robert B. Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | | | | | - Augusto Vaglio
- Unit of Nephrology, University Hospital of Parma, Parma, Italy
| | - Rex Neal Smith
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston MA
| | - Emmanuel Zorn
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston MA
| |
Collapse
|
37
|
Non-HLA antibodies in solid organ transplantation: recent concepts and clinical relevance. Curr Opin Organ Transplant 2013; 18:430-5. [PMID: 23838648 DOI: 10.1097/mot.0b013e3283636e55] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Humoral responses beyond major histocompatibility antigens continue to receive the attention of the transplantation community. We report on clinical studies testing clinical relevance of non-human leukocyte antigen (HLA) antigens in solid organ transplantation and provide an update on novel experimental findings. A conceptual framework on the role of graft microenvironment during initiation of non-HLA-related humoral immunity is addressed as well. RECENT FINDINGS Clinical relevance of antibodies targeting angiotensin type 1 receptor (AT1R-Abs) is broadly confirmed in renal and cardiac transplantation, where in addition antibodies against endothelin type A receptor (ETAR-Abs) were found. Obliterative lesions in lung allografts occur more commonly in the presence of antibodies directed against K-α 1 tubulin and collagen-V. Anti-perlecan antibodies are newly identified as accelerators of obliterative vascular lesions. Changes in the intragraft microenvironment, ischemia and alloimmunity seem to represent important permissive factors for non-HLA antibody responses. SUMMARY Confirmed clinical relevance of non-HLA humoral responses in solid organ transplantation emphasizes the need for revision of classical diagnostic approaches based solely on detection of HLA-donor-specific antibodies (DSA). A better understanding of intersections of HLA- and non-HLA-related mechanisms and identification of common effector mechanisms would represent an important step towards targeted therapies.
Collapse
|
38
|
Yamanaga S, Watarai Y, Yamamoto T, Tsujita M, Hiramitsu T, Nanmoku K, Goto N, Takeda A, Morozumi K, Katayama A, Saji H, Uchida K, Kobayashi T. Frequent development of subclinical chronic antibody-mediated rejection within 1year after renal transplantation with pre-transplant positive donor-specific antibodies and negative CDC crossmatches. Hum Immunol 2013; 74:1111-8. [DOI: 10.1016/j.humimm.2013.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/26/2013] [Accepted: 06/07/2013] [Indexed: 11/25/2022]
|