1
|
Rim JG, Hellkamp AS, Neely ML, Reynolds JM, Belperio JA, Budev M, Eason L, Frankel CW, Keshavjee S, Kirchner J, Singer LG, Shah PD, Snyder LD, Samuel Weigt S, Palmer SM, Todd JL. Basiliximab induction immunosuppression and lung transplant outcomes: Propensity analysis in a multicenter cohort. J Heart Lung Transplant 2024:S1053-2498(24)02003-5. [PMID: 39642952 DOI: 10.1016/j.healun.2024.11.033] [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: 05/03/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Basiliximab induction immunosuppression is increasingly employed in lung transplant recipients despite limited prospective evidence to support its use in this population. We sought to determine the relationship between basiliximab induction and development of acute rejection, chronic lung allograft dysfunction, and other clinically relevant outcomes in a multicenter lung transplant cohort with variable induction practice patterns. METHODS We applied propensity-based statistical methods to rigorous, prospectively collected longitudinal data from 768 newly transplanted adult lung recipients at 5 North American centers (368 who received basiliximab induction immunosuppression and 400 who received no induction immunosuppression). Treatment effects were estimated using outcome-specific propensity score regression models, weighted by the outcome-specific overlap weights, and stratified by center strata. RESULTS Basiliximab induction immunosuppression was associated with a significant reduction in any grade acute rejection (HR 0.65, 95% CI 0.46-0.92; p=0.015), organizing pneumonia histology (HR 0.38, 95% CI 0.16-0.90; p=0.028), acute lung injury histology (HR 0.28, 95% CI 0.13-0.61; p=0.001), and development of class II donor specific antibodies (HR 0.51, 95% CI 0.27-0.95; p=0.034) within the first posttransplant year. However, there was no significant association between basiliximab and development of chronic lung allograft dysfunction, mortality, or graft loss. For select infections during the first posttransplant year, there was no evidence of a difference in risk between patients who did versus did not receive basiliximab. CONCLUSIONS Basiliximab induction immunosuppression is associated with a significant reduction in early posttransplant cellular and humoral immune events and lung injury histologies but not chronic lung allograft dysfunction or mortality.
Collapse
Affiliation(s)
- Jeeyon G Rim
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC
| | - Anne S Hellkamp
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Megan L Neely
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - John M Reynolds
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC
| | | | | | - Lerin Eason
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC
| | - Courtney W Frankel
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC
| | - Shaf Keshavjee
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jerry Kirchner
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | | | - Pali D Shah
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Laurie D Snyder
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | | | - Scott M Palmer
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Jamie L Todd
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Duke University Medical Center, Durham, NC.
| |
Collapse
|
2
|
Furian L, Bestard O, Budde K, Cozzi E, Diekmann F, Mamode N, Naesens M, Pengel LHM, Schwartz Sorensen S, Vistoli F, Thaunat O. European Consensus on the Management of Sensitized Kidney Transplant Recipients: A Delphi Study. Transpl Int 2024; 37:12475. [PMID: 38665475 PMCID: PMC11043529 DOI: 10.3389/ti.2024.12475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 04/28/2024]
Abstract
An increasing number of sensitized patients awaiting transplantation face limited options, leading to fatalities during dialysis and higher costs. The absence of established evidence highlights the need for collaborative consensus. Donor-specific antibodies (DSA)-triggered antibody-mediated rejection (AMR) significantly contributes to kidney graft failure, especially in sensitized patients. The European Society for Organ Transplantation (ESOT) launched the ENGAGE initiative, categorizing sensitized candidates by AMR risk to improve patient care. A systematic review assessed induction and maintenance regimens as well as antibody removal strategies, with statements subjected to the Delphi methodology. A Likert-scale survey was distributed to 53 European experts (Nephrologists, Transplant surgeons and Immunologists) with experience in kidney transplant recipient care. A rate ≥75% with the same answer was considered consensus. Consensus was achieved in 95.3% of statements. While most recommendations aligned, two statements related to complement inhibitors for AMR prophylaxis lacked consensus. The ENGAGE consensus presents contemporary recommendations for desensitization and immunomodulation strategies, grounded in predefined risk categories. The adoption of tailored, patient-specific measures is anticipated to streamline the care of sensitized recipients undergoing renal allografts. While this approach holds the promise of enhancing transplant accessibility and fostering long-term success in transplantation outcomes, its efficacy will need to be assessed through dedicated studies.
Collapse
Affiliation(s)
- Lucrezia Furian
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, School of Medicine and Surgery, University of Padua, Padua, Italy
| | - Oriol Bestard
- Kidney Transplant Unit, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| | - Emanuele Cozzi
- Transplant Immunology Unit, Department of Cardiac, Thoracic and Vascular Sciences, School of Medicine and Surgery, University of Padua, Padua, Italy
| | - Fritz Diekmann
- Experimental Nephrology and Transplant Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Faculty of Medicine, KU Leuven, Leuven, Belgium
- Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Liset H. M. Pengel
- Erasmus MC Transplant Institute, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Soren Schwartz Sorensen
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Fabio Vistoli
- University of Pisa, Pisa, Italy
- Department of Biothecnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
3
|
Negi S, Rutman AK, Saw CL, Paraskevas S, Tchervenkov J. Pretransplant, Th17 dominant alloreactivity in highly sensitized kidney transplant candidates. FRONTIERS IN TRANSPLANTATION 2024; 3:1336563. [PMID: 38993777 PMCID: PMC11235243 DOI: 10.3389/frtra.2024.1336563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/21/2024] [Indexed: 07/13/2024]
Abstract
Introduction Sensitization to donor human leukocyte antigen (HLA) molecules prior to transplantation is a significant risk factor for delayed access to transplantation and to long-term outcomes. Memory T cells and their cytokines play a pivotal role in shaping immune responses, thereby increasing the risk of allograft rejection among highly sensitized patients. This study aims to elucidate the precise contribution of different CD4+ memory T cell subsets to alloreactivity in highly sensitized (HS) kidney transplant recipients. Methods and results Stimulation of peripheral blood mononuclear cells (PBMC) with various polyclonal stimulating agents to assess non-specific immune responses revealed that HS patients exhibit elevated immune reactivity even before kidney transplantation, compared to non-sensitized (NS) patients. HS patients' PBMC displayed higher frequencies of CD4+ T cells expressing IFNγ, IL4, IL6, IL17A, and TNFα and secreted relatively higher levels of IL17A and IL21 upon stimulation with PMA/ionomycin. Additionally, PBMC from HS patients stimulated with T cell stimulating agent phytohemagglutinin (PHA) exhibited elevated expression levels of IFNγ, IL4 and, IL21. On the other hand, stimulation with a combination of resiquimod (R848) and IL2 for the activation of memory B cells demonstrated higher expression of IL17A, TNFα and IL21, as determined by quantitative real-time PCR. A mixed leukocyte reaction (MLR) assay, employing third-party donor antigen presenting cells (APCs), was implemented to evaluate the direct alloreactive response. HS patients demonstrated notably higher frequencies of CD4+ T cells expressing IL4, IL6 and IL17A. Interestingly, APCs expressing recall HLA antigens triggered a stronger Th17 response compared to APCs lacking recall HLA antigens in sensitized patients. Furthermore, donor APCs induced higher activation of effector memory T cells in HS patients as compared to NS patients. Conclusion These results provide an assessment of pretransplant alloreactive T cell subsets in highly sensitized patients and emphasize the significance of Th17 cells in alloimmune responses. These findings hold promise for the development of treatment strategies tailored to sensitized kidney transplant recipients, with potential clinical implications.
Collapse
Affiliation(s)
- Sarita Negi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Human Islet Transplantation Laboratory, McGill University Health Centre, Montréal, QC, Canada
| | | | - Chee Loong Saw
- HLA Laboratory, Division of Hematology, McGill University Health Centre, Montréal, QC, Canada
| | - Steven Paraskevas
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Human Islet Transplantation Laboratory, McGill University Health Centre, Montréal, QC, Canada
- Department of Surgery, McGill University, Montréal, QC, Canada
- Division of General Surgery and Multi-Organ Transplant Program, Department of Surgery, McGill University Health Centre, Montréal, QC, Canada
| | - Jean Tchervenkov
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
- Department of Surgery, McGill University, Montréal, QC, Canada
- Division of General Surgery and Multi-Organ Transplant Program, Department of Surgery, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
4
|
DiToro D, Murakami N, Pillai S. T-B Collaboration in Autoimmunity, Infection, and Transplantation. Transplantation 2024; 108:386-398. [PMID: 37314442 PMCID: PMC11345790 DOI: 10.1097/tp.0000000000004671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel DiToro
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Naoka Murakami
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
5
|
Kanai D, Wakui H, Hanaoka M, Haze T, Azushima K, Shinoda S, Tsukamoto S, Taguchi S, Kinguchi S, Kanaoka T, Toya Y, Hirawa N, Kato H, Watanabe F, Hanaoka K, Mitsuhashi H, Yamaguchi S, Ohnishi T, Tamura K. Blunted humoral immune response to the fourth dose of BNT162b2 COVID-19 vaccine in patients undergoing hemodialysis. Clin Exp Nephrol 2023:10.1007/s10157-023-02342-0. [PMID: 36977892 PMCID: PMC10047466 DOI: 10.1007/s10157-023-02342-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND We aimed to investigate the impact of a fourth dose of BNT162b2 vaccine (Comirnaty®, Pfizer-BioNTech) on anti-SARS-CoV-2 (anti-S IgG) antibody titers in patients receiving hemodialysis (HD) and healthcare workers (HCWs). METHODS A multi-institutional retrospective study at five dialysis clinics in Japan was conducted using 238 HD patients and 58 HCW controls who received four doses of the BNT162b2 mRNA vaccine. Anti-S IgG titers were measured at 1, 3, and 6 months after the second dose, at 1 and 5/6 months after the third dose, and at 1 month after the fourth dose of vaccine. RESULTS The log anti-S IgG titers of the HD patients after the second vaccination were significantly lower than those of the control group, but equalized 1 month after the third vaccination: 9.94 (95% CI 9.82-10.10) vs. 9.81 (95% CI 9.66-9.96), (P = 0.32). In both groups, the fold-increase in anti-S IgG titers was significantly lower after the fourth dose than after the third dose of vaccine. In addition, there was a strong negative correlation between antibody titers 1 month after the fourth vaccination and antibody titers immediately before the vaccination. In both groups, the waning rate of anti-S IgG titers from the post-vaccination peak level after the third vaccine dose was significantly slower than that after the second dose. CONCLUSIONS These findings suggest that the humoral immune response was blunted after the fourth dose of the conventional BNT162b2 vaccine. However, multiple vaccinations could extend the window of humoral immune protection.
Collapse
Affiliation(s)
- Daisuke Kanai
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Masaaki Hanaoka
- Kohsaikai Kamioooka Jinsei Clinic, Yokohama, Kanagawa, Japan
| | - Tatsuya Haze
- YCU Center for Novel and Exploratory Clinical Trials (Y-NEXT), Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Satoru Shinoda
- Department of Biostatistics, School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Shinya Taguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Yoshiyuki Toya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Nobuhito Hirawa
- Department of Nephrology and Hypertension, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | | | - Kanako Hanaoka
- Kohsaikai Kamioooka Jinsei Clinic, Yokohama, Kanagawa, Japan
| | | | | | | | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
6
|
Pascual J, Crespo M, Portoles J, Jimenez C, Ortega-Carrion A, Diez T, Portero I. The IMBG Test for Evaluating the Pharmacodynamic Response to Immunosuppressive Therapy in Kidney Transplant Patients: Current Evidence and Future Applications. Int J Mol Sci 2023; 24:ijms24065201. [PMID: 36982276 PMCID: PMC10049381 DOI: 10.3390/ijms24065201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Immunosuppressive drugs are widely used to prevent rejection after kidney transplantation. However, the pharmacological response to a given immunosuppressant can vary markedly between individuals, with some showing poor treatment responses and/or experiencing serious side effects. There is an unmet need for diagnostic tools that allow clinicians to individually tailor immunosuppressive therapy to a patient’s immunological profile. The Immunobiogram (IMBG) is a novel blood-based in vitro diagnostic test that provides a pharmacodynamic readout of the immune response of individual patients to a range of immunosuppressants commonly used in kidney transplant recipients. Here, we discuss the current approaches used to measure the pharmacodynamic responses of individual patients to specific immunosuppressive drugs in vitro, which can then be correlated with patient’s clinical outcomes. We also describe the procedure of the IMBG assay, and summarize the results obtained using the IMBG in different kidney transplant populations. Finally, we outline future directions and other novel applications of the IMBG, both in kidney transplant patients and other autoimmune diseases.
Collapse
Affiliation(s)
- Julio Pascual
- Nephrology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- Nephrology Department, Hospital del Mar, Institut Mar for Medical Research, 08003 Barcelona, Spain
- Correspondence:
| | - Marta Crespo
- Nephrology Department, Hospital del Mar, Institut Mar for Medical Research, 08003 Barcelona, Spain
| | - Jose Portoles
- Nephrology Department, Hospital Puerta de Hierro Mahadahonda, Institute IDHIPHIM for Medical Research, 28222 Madrid, Spain
| | - Carlos Jimenez
- Nephrology Department, Hospital La Paz, 28046 Madrid, Spain
| | | | - Teresa Diez
- Biohope Scientific Solutions for Human Health, 28760 Madrid, Spain
| | - Isabel Portero
- Biohope Scientific Solutions for Human Health, 28760 Madrid, Spain
| |
Collapse
|
7
|
Bouchet A, Muller B, Olagne J, Barba T, Joly M, Obrecht A, Rabeyrin M, Dijoud F, Picard C, Mezaache S, Sicard A, Koenig A, Parissiadis A, Dubois V, Morelon E, Caillard S, Thaunat O. Evolution of humoral lesions on follow-up biopsy stratifies the risk for renal graft loss after antibody-mediated rejection treatment. Nephrol Dial Transplant 2022; 37:2555-2568. [PMID: 35675302 DOI: 10.1093/ndt/gfac192] [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: 07/14/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The standard-of-care protocol, based on plasma exchanges, high-dose intravenous immunoglobulin and optimization of maintenance immunosuppression, can slow down the evolution of antibody-mediated rejection (AMR), but with high interindividual variability. Identification of a reliable predictive tool of the response to AMR treatment is a mandatory step for personalization of the follow-up strategy and to guide second-line therapies. METHODS Interrogation of the electronic databases of 2 French university hospitals (Lyon and Strasbourg) retrospectively identified 81 renal transplant recipients diagnosed with AMR without chronic lesions (cg score ≤1) at diagnosis and for whom a follow-up biopsy had been performed 3-6 months after initiation of therapy. RESULTS The evolution of humoral lesions on follow-up biopsy (disappearance versus persistence versus progression) correlated with the risk for allograft loss (logrank test, P = .001). Patients with disappearance of humoral lesions had ∼80% graft survival at 10 years. The hazard ratio for graft loss in multivariate analysis was 3.91 (P = .04) and 5.15 (P = .02) for patients with persistence and progression of lesions, respectively. The non-invasive parameters classically used to follow the intensity of humoral alloimmune response (evolution of immunodominant DSA mean fluorescence intensity) and the decline of renal graft function (estimated glomerular filtration rate decrease and persistent proteinuria) showed little clinical value to predict the histological response to AMR therapy. CONCLUSION We conclude that invasive monitoring of the evolution of humoral lesions by the mean of follow-up biopsy performed 3-6 months after the initiation of therapy is an interesting tool to predict long-term outcome after AMR treatment.
Collapse
Affiliation(s)
- Antonin Bouchet
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France
| | - Brieuc Muller
- Service de Néphrologie et Transplantation, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jerome Olagne
- Service de Néphrologie et Transplantation, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Thomas Barba
- Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| | - Mélanie Joly
- Service de Néphrologie et Transplantation, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Augustin Obrecht
- Service de Néphrologie et Transplantation, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Maud Rabeyrin
- Institut de Pathologie Multisite, Groupement Hospitalier Est, Bron, France
| | - Frédérique Dijoud
- Institut de Pathologie Multisite, Groupement Hospitalier Est, Bron, France
| | - Cécile Picard
- Institut de Pathologie Multisite, Groupement Hospitalier Est, Bron, France
| | - Sarah Mezaache
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France
| | - Antoine Sicard
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France
| | - Alice Koenig
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| | - Anne Parissiadis
- Laboratoire d'Histocompatibilité, Etablissement Français du Sang, Strasbourg, France
| | - Valérie Dubois
- Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France.,Laboratoire d'Histocompatibilité, Etablissement Français du Sang, Lyon, France
| | - Emmanuel Morelon
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| | - Sophie Caillard
- Service de Néphrologie et Transplantation, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Unité de Formation et de Recherche de Médecine Lyon Est, Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| |
Collapse
|
8
|
Charmetant X, Chen CC, Hamada S, Goncalves D, Saison C, Rabeyrin M, Rabant M, Duong van Huyen JP, Koenig A, Mathias V, Barba T, Lacaille F, le Pavec J, Brugière O, Taupin JL, Chalabreysse L, Mornex JF, Couzi L, Graff-Dubois S, Jeger-Madiot R, Tran-Dinh A, Mordant P, Paidassi H, Defrance T, Morelon E, Badet L, Nicoletti A, Dubois V, Thaunat O. Inverted direct allorecognition triggers early donor-specific antibody responses after transplantation. Sci Transl Med 2022; 14:eabg1046. [PMID: 36130013 DOI: 10.1126/scitranslmed.abg1046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The generation of antibodies against donor-specific major histocompatibility complex (MHC) antigens, a type of donor-specific antibodies (DSAs), after transplantation requires that recipient's allospecific B cells receive help from T cells. The current dogma holds that this help is exclusively provided by the recipient's CD4+ T cells that recognize complexes of recipient's MHC II molecules and peptides derived from donor-specific MHC alloantigens, a process called indirect allorecognition. Here, we demonstrated that, after allogeneic heart transplantation, CD3ε knockout recipient mice lacking T cells generate a rapid, transient wave of switched alloantibodies, predominantly directed against MHC I molecules. This is due to the presence of donor CD4+ T cells within the graft that recognize intact recipient's MHC II molecules expressed by B cell receptor-activated allospecific B cells. Indirect evidence suggests that this inverted direct pathway is also operant in patients after transplantation. Resident memory donor CD4+ T cells were observed in perfusion liquids of human renal and lung grafts and acquired B cell helper functions upon in vitro stimulation. Furthermore, T follicular helper cells, specialized in helping B cells, were abundant in mucosa-associated lymphoid tissue of lung and intestinal grafts. In the latter, more graft-derived passenger T cells correlated with the detection of donor T cells in recipient's circulation; this, in turn, was associated with an early transient anti-MHC I DSA response and worse transplantation outcomes. We conclude that this inverted direct allorecognition is a possible explanation for the early transient anti-MHC DSA responses frequently observed after lung or intestinal transplantations.
Collapse
Affiliation(s)
- Xavier Charmetant
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Sarah Hamada
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - David Goncalves
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Carole Saison
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, 69500 Bron, France
| | - Marion Rabant
- Pathology Department, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, 75015 Paris, France
| | | | - Alice Koenig
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | - Virginie Mathias
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Thomas Barba
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Florence Lacaille
- Pediatric Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants malades, 75015 Paris, France
| | - Jérôme le Pavec
- Department of Pulmonology and Lung Transplantation, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - Olivier Brugière
- Pulmonology Department, Adult Cystic Fibrosis Centre and Lung Transplantation Department, Foch Hospital, 92150 Suresnes, France
| | - Jean-Luc Taupin
- Laboratory of Immunology and Histocompatibility, Hôpital Saint-Louis APHP, 75010 Paris, France
- INSERM U976 Institut de Recherche Saint-Louis, Université Paris Diderot, 75010 Paris, France
| | - Lara Chalabreysse
- Department of Pathology, Hospices Civils de Lyon, Groupement Hospitalier Est, 69500 Bron, France
| | - Jean-François Mornex
- Université de Lyon, Université Lyon 1, INRAE, IVPC, UMR754, 69000 Lyon, France
- Department of Pneumology, GHE, Hospices Civils de Lyon, 69000 Lyon, France
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis, Apheresis, Pellegrin Hospital, 33000 Bordeaux, France
| | - Stéphanie Graff-Dubois
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Raphaël Jeger-Madiot
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), 75013 Paris, France
| | - Alexy Tran-Dinh
- Université de Paris, LVTS, INSERM U1148, 75018 Paris, France
| | - Pierre Mordant
- Department of Vascular and Thoracic Surgery, Assistance Publique-Hôpitaux de Paris, Bichat-Claude Bernard Hospital, 75018 Paris, France
| | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Thierry Defrance
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Emmanuel Morelon
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | - Lionel Badet
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Urology and Transplantation Surgery, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| | | | - Valérie Dubois
- French National Blood Service (EFS), HLA Laboratory, 69150 Décines, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 69008 Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, 69003 Lyon, France
| |
Collapse
|
9
|
Shen J, Liu C, Yan P, Wang M, Guo L, Liu S, Chen J, Rosenholm JM, Huang H, Wang R, Zhang H. Helper T Cell (CD4 +) Targeted Tacrolimus Delivery Mediates Precise Suppression of Allogeneic Humoral Immunity. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9794235. [PMID: 35958106 PMCID: PMC9343082 DOI: 10.34133/2022/9794235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/24/2022] [Indexed: 01/15/2023]
Abstract
Antibody-mediated rejection (ABMR) is a major cause of dysfunction and loss of transplanted kidney. The current treatments for ABMR involve nonspecific inhibition and clearance of T/B cells or plasma cells. However, the prognosis of patients following current treatment is poor. T follicular helper cells (Tfh) play an important role in allograft-specific antibodies secreting plasma cell (PC) development. Tfh cells are therefore considered to be important therapeutic targets for the treatment of antibody hypersecretion disorders, such as transplant rejection and autoimmune diseases. Tacrolimus (Tac), the primary immunosuppressant, prevents rejection by reducing T cell activation. However, its administration should be closely monitored to avoid serious side effects. In this study, we investigated whether Tac delivery to helper T (CD4+) cells using functionalized mesoporous nanoparticles can block Tfh cell differentiation after alloantigen exposure. Results showed that Tac delivery ameliorated humoral rejection injury in rodent kidney graft by suppressing Tfh cell development, PC, and donor-specific antibody (DSA) generation without causing severe side effects compared with delivery through the drug administration pathway. This study provides a promising therapeutic strategy for preventing humoral rejection in solid organ transplantation. The specific and controllable drug delivery avoids multiple disorder risks and side effects observed in currently used clinical approaches.
Collapse
Affiliation(s)
- Jia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Chang Liu
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Pengpeng Yan
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Meifang Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Luying Guo
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Shuaihui Liu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
| | - Hongfeng Huang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
| | - Rending Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, China
- Organ Donation and Coordination Office, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku 20520, Finland
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
10
|
Charmetant X, Espi M, Barba T, Ovize A, Morelon E, Mathieu C, Thaunat O. Predictive factors of a viral neutralizing humoral response after a third dose of COVID-19 mRNA vaccine. Am J Transplant 2022; 22:1442-1450. [PMID: 35114060 PMCID: PMC10149236 DOI: 10.1111/ajt.16990] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 01/25/2023]
Abstract
Kidney transplant recipients (KTRs) have reduced ability to mount adequate antibody response after two doses of the COVID-19 mRNA vaccine. French health authorities have allowed a third booster dose (D3) for KTRs, but their response is heterogeneous and tools able to discriminate the responders are lacking. Anti-RBD IgG titers (chemiluminescence immunoassay), spike-specific cellular responses (IFN-γ-releasing assay, IGRA), and in vitro serum neutralization of the virus (the best available correlate of protection), were evaluated 7-14 days after the second dose (D2) of BNT162b2 vaccine in 93 KTRs. Among the 73 KTRs, whose serum did not neutralize SARS-CoV-2 in vitro after D2, 14 (19%) acquired this capacity after D3, and were considered as "responders." Exploratory univariate analysis identified short time from transplantation and high maintenance immunosuppression as detrimental factors for the response to D3. In addition, any of the presence of anti-RBD IgGs and/or positive IGRA after D2 was predictive of response to D3. By contrast, none of the KTRs with both a negative serology and IGRA responded to D3. In summary, routinely available bioassays performed after D2 allow identifying KTRs that will respond to a booster D3. These results pave the way for the personalization of vaccination strategy in KTRs.
Collapse
Affiliation(s)
- Xavier Charmetant
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
| | - Maxime Espi
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
| | - Thomas Barba
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
| | - Anne Ovize
- Eurofins Biomnis Laboratory, Lyon, France
| | - Emmanuel Morelon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
- Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Cyrille Mathieu
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
- Claude Bernard University (Lyon 1), Villeurbanne, France
| |
Collapse
|
11
|
Charmetant X, Espi M, Benotmane I, Barateau V, Heibel F, Buron F, Gautier-Vargas G, Delafosse M, Perrin P, Koenig A, Cognard N, Levi C, Gallais F, Manière L, Rossolillo P, Soulier E, Pierre F, Ovize A, Morelon E, Defrance T, Fafi-Kremer S, Caillard S, Thaunat O. Infection or a third dose of mRNA vaccine elicits neutralizing antibody responses against SARS-CoV-2 in kidney transplant recipients. Sci Transl Med 2022; 14:eabl6141. [PMID: 35103481 PMCID: PMC8939774 DOI: 10.1126/scitranslmed.abl6141] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/25/2022] [Indexed: 12/12/2022]
Abstract
Transplant recipients, who receive therapeutic immunosuppression to prevent graft rejection, are characterized by high coronavirus disease 2019 (COVID-19)-related mortality and defective response to vaccines. We observed that previous infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but not the standard two-dose regimen of vaccination, provided protection against symptomatic COVID-19 in kidney transplant recipients. We therefore compared the cellular and humoral immune responses of these two groups of patients. Neutralizing anti-receptor-binding domain (RBD) immunoglobulin G (IgG) antibodies were identified as the primary correlate of protection for transplant recipients. Analysis of virus-specific B and T cell responses suggested that the generation of neutralizing anti-RBD IgG may have depended on cognate T-B cell interactions that took place in germinal center, potentially acting as a limiting checkpoint. High-dose mycophenolate mofetil, an immunosuppressive drug, was associated with fewer antigen-specific B and T follicular helper (TFH) cells after vaccination; this was not observed in patients recently infected with SARS-CoV-2. Last, we observed that, in two independent prospective cohorts, administration of a third dose of SARS-CoV-2 mRNA vaccine restored neutralizing titers of anti-RBD IgG in about 40% of individuals who had not previously responded to two doses of vaccine. Together, these findings suggest that a third dose of SARS-CoV-2 mRNA vaccine improves the RBD-specific responses of transplant patients treated with immunosuppressive drugs.
Collapse
Affiliation(s)
- Xavier Charmetant
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
| | - Maxime Espi
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
| | - Ilies Benotmane
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
- Department of Virology, Strasbourg University Hospital, 67000 Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Véronique Barateau
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
| | - Francoise Heibel
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Fanny Buron
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
| | - Gabriela Gautier-Vargas
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Marion Delafosse
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
| | - Peggy Perrin
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
- Claude Bernard University (Lyon 1), 43 boulevard du 11 Novembre 1918, 69622 Villeurbanne France
| | - Noëlle Cognard
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
| | - Charlène Levi
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
| | - Floriane Gallais
- Department of Virology, Strasbourg University Hospital, 67000 Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Louis Manière
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
| | - Paola Rossolillo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique (CNRS), UMR 7104, Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Université de Strasbourg, 67400 Illkirch, France
| | - Eric Soulier
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Florian Pierre
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Anne Ovize
- Eurofins Biomnis Laboratory, 69007 Lyon, France
| | - Emmanuel Morelon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
- Claude Bernard University (Lyon 1), 43 boulevard du 11 Novembre 1918, 69622 Villeurbanne France
| | - Thierry Defrance
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
| | - Samira Fafi-Kremer
- Department of Virology, Strasbourg University Hospital, 67000 Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Sophie Caillard
- Department of Nephrology and Transplantation, Strasbourg University Hospital, 67000 Strasbourg, France
- Department of Virology, Strasbourg University Hospital, 67000 Strasbourg, France
- Inserm UMR S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21 avenue Tony Garnier, 69007 Lyon, France
- Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d’Arsonval, 69003 Lyon, France
- Claude Bernard University (Lyon 1), 43 boulevard du 11 Novembre 1918, 69622 Villeurbanne France
| |
Collapse
|
12
|
Yeo WS, Ng QX. Biomarkers of immune tolerance in kidney transplantation: an overview. Pediatr Nephrol 2022; 37:489-498. [PMID: 33712863 DOI: 10.1007/s00467-021-05023-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 11/30/2022]
Abstract
Kidney failure, one of the most prevalent diseases in the world and with increasing incidence, is associated with substantial morbidity and mortality. Currently available modes of kidney replacement therapy include dialysis and kidney transplantation. Though kidney transplantation is the preferred and ideal mode of kidney replacement therapy, this modality, however, is not without its risks. Kidney transplant recipients are constantly at risk of complications associated with immunosuppression, namely, opportunistic infections (e.g., Epstein-Barr virus and cytomegalovirus infections), post-transplant lymphoproliferative disorder, and complications associated with immunosuppressants (e.g., calcineurin inhibitor- and corticosteroid-associated new onset diabetes after transplantation and calcineurin inhibitor-associated nephrotoxicity). Transplantation tolerance, an acquired state in which immunocompetent recipients have developed donor-specific unresponsiveness, may be the Holy Grail in enabling optimal allograft survival and obviating the risks associated with immunosuppression in kidney transplant recipients. This review aims to discuss the biomarkers available to predict, identify, and define the transplant immune tolerant state and various tolerance induction strategies. Regrettably, pediatric patients have not been included in any tolerance studies and this should be the focus of future studies.
Collapse
Affiliation(s)
- Wee-Song Yeo
- Mount Elizabeth Hospital, 3 Mount Elizabeth, Singapore, 228510, Singapore.
| | - Qin Xiang Ng
- MOH Holdings Pte Ltd, 1 Maritime Square, Singapore, 099253, Singapore
| |
Collapse
|
13
|
Espi M, Charmetant X, Barba T, Mathieu C, Pelletier C, Koppe L, Chalencon E, Kalbacher E, Mathias V, Ovize A, Cart-Tanneur E, Bouz C, Pellegrina L, Morelon E, Juillard L, Fouque D, Couchoud C, Thaunat O. A prospective observational study for justification, safety, and efficacy of a third dose of mRNA vaccine in patients receiving maintenance hemodialysis. Kidney Int 2022; 101:390-402. [PMID: 34856313 PMCID: PMC8628628 DOI: 10.1016/j.kint.2021.10.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 11/29/2022]
Abstract
The level of protection achieved by the standard two doses of COVID-19 mRNA vaccines in patients receiving maintenance hemodialysis (MHD) remains unclear. To study this we used the French Renal Epidemiology and Information Network (REIN) Registry to compare the incidence and severity of 1474 cases of COVID-19 diagnosed in patients receiving MHD after none, one or two doses of vaccine. Vaccination significantly reduce COVID-19 incidence and severity, but 11% of patients infected after two doses still died. Lack of vaccinal protection in patients naïve for SARS-CoV-2 could be due to defective Tfh response [38% of patients with negative spike-specific CD4+ T-cell interferon gamma release assay] and failure to generate viral neutralizing titers of anti-spike receptor binding domain (RBD) IgGs (63% of patients with titer at or under 997 BAU/ml, defining low/no responders) after two doses of vaccine. To improve protection, a third dose of vaccine was administered to 75 patients [57 low/no responders, 18 high responders after two doses] from the ROMANOV cohort that prospectively enrolled patients receiving MHD vaccinated with BNT162b2 (Pfizer). Tolerance to the third dose was excellent. High responders to two doses did not generate more anti-RBD IgGs after three doses but had more side effects. Importantly, 31 (54%) of low/no responders to two doses reached neutralizing titers of anti-RBD IgGs after three doses. A positive interferon gamma release assay and/or suboptimal titer of anti-RBD IgGs after two doses were the only predictive variables for response to three doses in multivariate analysis. Thus, the standard scheme of vaccination insufficiently protects patients receiving MHD. Anti-RBD IgG and specific CD4+ T-cell response after two doses can guide personalized administration of the third dose, which improves the humoral response of SARS-CoV-2-naïve patients receiving MHD.
Collapse
Affiliation(s)
- Maxime Espi
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France; Department of Nephrology, Nutrition, and Hemodialysis, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Xavier Charmetant
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Thomas Barba
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France; Department of Internal Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France; Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Cyrille Mathieu
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
| | - Caroline Pelletier
- Department of Nephrology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Laetitia Koppe
- Department of Nephrology, Nutrition, and Hemodialysis, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Elodie Chalencon
- Department of Nephrology, Nutrition, and Hemodialysis, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Emilie Kalbacher
- Department of Nephrology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Virginie Mathias
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France; Human Leukocyte Antigen Laboratory, French National Blood Service, Décines-Charpieu, France
| | - Anne Ovize
- Eurofins Biomnis Laboratory, Lyon, France
| | | | | | | | - Emmanuel Morelon
- Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France; Department of Transplantation, Nephrology, and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Laurent Juillard
- Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France; Department of Nephrology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Denis Fouque
- Department of Nephrology, Nutrition, and Hemodialysis, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France
| | - Cécile Couchoud
- REIN Registry, Agence de la Biomédecine, Saint-Denis La Plaine, France; Biostatistique Santé Department, Laboratoire de Biométrie et Biologie Evolutive, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5558, Université Lyon I, Villeurbanne, France
| | - Olivier Thaunat
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale U1111, Université Claude Bernard Lyon I, Centre National de la Recherche Scientifique Unité Mixte de Recherch 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France; Medical School, Claude Bernard University (Lyon 1), Villeurbanne, France; Department of Transplantation, Nephrology, and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
14
|
Steines L, Scharf M, Hoffmann P, Schuster A, Banas B, Bergler T. Monitoring B cell alloresponses in rats. J Immunol Methods 2022; 501:113212. [PMID: 34971633 DOI: 10.1016/j.jim.2021.113212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/01/2021] [Accepted: 12/23/2021] [Indexed: 11/24/2022]
Abstract
Antibody-mediated rejection is a major cause of graft failure in organ transplantation. For this reason, B cell responses are of particular interest to transplantation research. Rats are important model organisms for transplant studies, but B cell alloimmune assays and B cell subset markers are poorly established in rats. We alloimmunized rats by donor blood injection using the high responder rat strain combination Brown Norway (donor) and Lewis (recipient) rats. Using splenocytes from alloimmunized and control rats, we established assays to assess allospecific B cell proliferation and the capacity to generate allospecific B memory cells and alloantibody-secreting cells after antigenic rechallenge in vitro using a mixed lymphocyte reaction. Furthermore, we defined a simple gating and sorting strategy for pre- and post-germinal center follicular B cells, as well as non-switched and switched plasmablasts. Our protocols for assessing B cell alloresponses and B cell subsets in rats may help to accelerate research into the role of B cells and manipulation of humoral alloresponses in transplant research.
Collapse
Affiliation(s)
- Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Germany.
| | - Mona Scharf
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, University Hospital Regensburg, Germany; Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| | - Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Germany
| |
Collapse
|
15
|
Liu J, Tang T, Qu Z, Wang L, Si R, Wang H, Jiang Y. Elevated number of IL-21+ TFH and CD86+CD38+ B cells in blood of renal transplant recipients with AMR under conventional immuno-suppression. Int J Immunopathol Pharmacol 2022; 36:20587384211048027. [PMID: 35012395 PMCID: PMC8755922 DOI: 10.1177/20587384211048027] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/03/2021] [Indexed: 11/12/2022] Open
Abstract
The objective of this study is to detect the number of different subsets of TFH and B cells in renal transplant recipients (RTR) with antibody-mediated acute rejection (AMR), acute rejection (AR), chronic rejection (CR), or transplant stable (TS). The present study was a prospective study. The numbers of ICOS +, PD-1+ and IL-21+ TFH, CD86+, CD38+, CD27+, and IgD- B cells in 21 patients with end-stage renal disease (ESRD) and post-transplant times were measured by flow cytometry. The level of serum IL-21 was detected by ELISA. The numbers of circulating CD4+CXCR5+, CD4+CXCR5+ICOS+, CD4+CXCR5+PD-1+, CD4+CXCR5+IL-21+ TFH, CD19+CD86+, and CD19 +CD86+CD38+ B cells as well as the level of serum IL-21 in the AMR, AR, and CR groups at post-transplantation were significantly higher than those at pre-transplantation. In contrast, the number of circulating CD19+CD27+IgD B cells was significantly increased in the TS groups in respect to the other groups. Moreover, the numbers of circulating CD4+CXCR5+IL-21+ TFH cells, CD19+CD86+CD38+ B cells as well as the level of serum IL-21 were positive related to the level of serum Cr while showing negative correlated with the values of eGFR in the AMR groups at post-transplantation for 4 and 12 weeks. Circulating TFH cells may be a biomarker in RTR with AMR, which can promote the differentiation of B cells into plasma cells by activating B cells, thereby promoting disease progression.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Tongyu Tang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Qu
- Department of Nephrology, the First Hospital of Jilin University, Changchun, China
| | - Li Wang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
- Xu Zhou Central Hospital, Xuzhou, China
| | - Rui Si
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Haifeng Wang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Zoonoses Research, Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Abstract
Unconventional T cells are a diverse and underappreciated group of relatively rare lymphocytes that are distinct from conventional CD4+ and CD8+ T cells, and that mainly recognize antigens in the absence of classical restriction through the major histocompatibility complex (MHC). These non-MHC-restricted T cells include mucosal-associated invariant T (MAIT) cells, natural killer T (NKT) cells, γδ T cells and other, often poorly defined, subsets. Depending on the physiological context, unconventional T cells may assume either protective or pathogenic roles in a range of inflammatory and autoimmune responses in the kidney. Accordingly, experimental models and clinical studies have revealed that certain unconventional T cells are potential therapeutic targets, as well as prognostic and diagnostic biomarkers. The responsiveness of human Vγ9Vδ2 T cells and MAIT cells to many microbial pathogens, for example, has implications for early diagnosis, risk stratification and targeted treatment of peritoneal dialysis-related peritonitis. The expansion of non-Vγ9Vδ2 γδ T cells during cytomegalovirus infection and their contribution to viral clearance suggest that these cells can be harnessed for immune monitoring and adoptive immunotherapy in kidney transplant recipients. In addition, populations of NKT, MAIT or γδ T cells are involved in the immunopathology of IgA nephropathy and in models of glomerulonephritis, ischaemia-reperfusion injury and kidney transplantation.
Collapse
|
17
|
Louis K, Macedo C, Metes D. Targeting T Follicular Helper Cells to Control Humoral Allogeneic Immunity. Transplantation 2021; 105:e168-e180. [PMID: 33909968 PMCID: PMC8484368 DOI: 10.1097/tp.0000000000003776] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Humoral allogeneic immunity driven by anti-HLA donor-specific antibodies and antibody-mediated rejection (AMR) significantly impede prolonged survival of organ allografts after transplantation. Although the importance of T follicular helper (TFH) cells in controlling antibody responses has been long established, their role in directing donor-specific antibody generation leading to AMR was only recently appreciated in the clinical setting of organ transplantation. In this review, we provide a comprehensive summary of the current knowledge on the biology of human TFH cells as well as their circulating counterparts and describe their pivotal role in driving humoral alloimmunity. In addition, we discuss the intrinsic effects of current induction therapies and maintenance immunosuppressive drugs as well as of biotherapies on TFH cells and provide future directions and novel opportunities of biotherapeutic targeting of TFH cells that have the potential of bringing the prophylactic and curative treatments of AMR toward personalized and precision medicine.
Collapse
Affiliation(s)
- Kevin Louis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Human Immunology and Immunopathology, Inserm UMR 976, Université de Paris, Paris, France
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diana Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
The role of circulating T follicular helper cells in kidney transplantation. Transpl Immunol 2021; 69:101459. [PMID: 34461243 DOI: 10.1016/j.trim.2021.101459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/21/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Humoral rejection plays a crucial role in the chronic deterioration of kidney allografts, but there is no effective therapeutic strategy to prevent or treat it. T follicular helper (Tfh) cells provide help to B cells, subsequently contributing to humoral rejection. Investigation of Tfh cells may be a useful strategy for assessing the risk and level of humoral rejection. However, it is difficult to investigate Tfh cells from patient-derived lymphoid tissue. Recent studies have shown that circulating Tfh (cTfh) cells, working in parallel to Tfh cells, have the capacity to promote antibody-secreting B cell differentiation and antibody secretion. Here, we review recent studies of cTfh cells in kidney transplantation and discuss the characteristics and functions of cTfh cells in kidney transplant recipients.
Collapse
|
19
|
Olaso D, Manook M, Moris D, Knechtle S, Kwun J. Optimal Immunosuppression Strategy in the Sensitized Kidney Transplant Recipient. J Clin Med 2021; 10:3656. [PMID: 34441950 PMCID: PMC8396983 DOI: 10.3390/jcm10163656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/09/2021] [Accepted: 08/15/2021] [Indexed: 01/10/2023] Open
Abstract
Patients with previous sensitization events against anti-human leukocyte antigens (HLA) often have circulating anti-HLA antibodies. Following organ transplantation, sensitized patients have higher rates of antibody-mediated rejection (AMR) compared to those who are non-sensitized. More stringent donor matching is required for these patients, which results in a reduced donor pool and increased time on the waitlist. Current approaches for sensitized patients focus on reducing preformed antibodies that preclude transplantation; however, this type of desensitization does not modulate the primed immune response in sensitized patients. Thus, an optimized maintenance immunosuppressive regimen is necessary for highly sensitized patients, which may be distinct from non-sensitized patients. In this review, we will discuss the currently available therapeutic options for induction, maintenance, and adjuvant immunosuppression for sensitized patients.
Collapse
Affiliation(s)
| | | | | | - Stuart Knechtle
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; (D.O.); (M.M.); (D.M.)
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; (D.O.); (M.M.); (D.M.)
| |
Collapse
|
20
|
The ROMANOV study found impaired humoral and cellular immune responses to SARS-CoV-2 mRNA vaccine in virus-unexposed patients receiving maintenance hemodialysis. Kidney Int 2021; 100:928-936. [PMID: 34284044 PMCID: PMC8286235 DOI: 10.1016/j.kint.2021.07.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
Patients on maintenance hemodialysis (MHD), which are at high risk of infection by SARS-CoV-2 virus and death due to COVID-19, have been prioritized for vaccination. However, because they were excluded from pivotal studies and have weakened immune responses, it is not known whether these patients are protected after the “standard” two doses of mRNA vaccines. To answer this, anti-spike receptor binding domain (RBD) IgG and interferon gamma-producing CD4+ and CD8+ specific-T cells were measured in the circulation 10-14 days after the second injection of BNT162b2 vaccine in 106 patients receiving MHD (14 with history of COVID-19) and compared to 30 healthy volunteers (four with history of COVID-19). After vaccination, most (72/80, 90%) patients receiving MHD naïve for the virus generated at least one type of immune effector, but their response was weaker and less complete than that of healthy volunteers. In multivariate analysis, hemodialysis and immunosuppressive therapy were significantly associated with absence of both anti-RBD IgGs and anti-spike CD8+ T cells. In contrast, previous history of COVID-19 in patients receiving MHD correlated with the generation of both types of immune effectors anti-RBD IgG and anti-spike CD8+ T cells at levels similar to healthy volunteers. Patients receiving MHD naïve for SARS-Cov-2 generate mitigated immune responses after two doses of mRNA vaccine. Thus, the good response to vaccine of patients receiving MHD with a history of COVID-19 suggest that these patients may benefit from a third vaccine injection.
Collapse
|
21
|
Hamada S, Dubois V, Koenig A, Thaunat O. Allograft recognition by recipient's natural killer cells: Molecular mechanisms and role in transplant rejection. HLA 2021; 98:191-199. [PMID: 34050618 DOI: 10.1111/tan.14332] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
The current transplant immunology dogma defends that allograft rejection is initiated by recipient's adaptive immune system. In this prevalent model, innate immune cells in general, and natural killer (NK) cells in particular, are merely considered as downstream effectors which participate in the destruction of the graft only upon recruitment by adaptive effectors: alloreactive T cells or donor-specific antibodies (DSA). Challenging this vision, recent data demonstrated that recipients' NK cells are capable of a form of allorecognition because they can sense the absence of self HLA class I molecules on the surface of graft endothelial cells. Missing-self triggers mTORC1-dependent activation of NK cells, which in turn promote the development of graft microvascular inflammation and detrimentally impact graft survival. The fact that some patients develop chronic vascular rejection in absence of DSA or genetically-predicted missing self suggests that other molecular mechanisms could underly NK cell allorecognition. This review provides an overview of these proven and putative molecular mechanisms and discusses future research directions in this emerging field in organ transplant immunology.
Collapse
Affiliation(s)
- Sarah Hamada
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Valérie Dubois
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,HLA Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
22
|
Steines L, Poth H, Schuster A, Amann K, Banas B, Bergler T. Disruption of Tfh:B Cell Interactions Prevents Antibody-Mediated Rejection in a Kidney Transplant Model in Rats: Impact of Calcineurin Inhibitor Dose. Front Immunol 2021; 12:657894. [PMID: 34135891 PMCID: PMC8201497 DOI: 10.3389/fimmu.2021.657894] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022] Open
Abstract
We aimed to investigate the mechanisms of humoral immune activation in ABMR using a MHC-mismatched rat kidney transplant model. We applied low dose cyclosporine A (loCNI) to allow donor-specific antibody (DSA) formation and rejection and high dose cyclosporine A (hiCNI) for non-rejection. DSA and leukocyte subsets were measured by flow cytometry. Germinal centers (GC), T follicular helper cells (Tfh), plasma cells and interleukin-21 (IL-21) expression were analyzed by immunofluorescence microscopy. Expression of important costimulatory molecules and cytokines was measured by qRT-PCR. Allograft rejection was evaluated by a nephropathologist. We found that DSA formation correlated with GC frequency and expansion, and that GC size was linked to the number of activated Tfh. In hiCNI, GC and activated Tfh were virtually absent, resulting in fewer plasma cells and no DSA or ABMR. Expression of B cell activating T cell cytokine IL-21 was substantially inhibited in hiCNI, but not in loCNI. In addition, hiCNI showed lower expression of ICOS ligand and IL-6, which stimulate Tfh differentiation and maintenance. Overall, Tfh:B cell crosstalk was controlled only by hiCNI treatment, preventing the development of DSA and ABMR. Additional strategies targeting Tfh:B cell interactions are needed for preventing alloantibody formation and ABMR.
Collapse
Affiliation(s)
- Louisa Steines
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Helen Poth
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Antonia Schuster
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Bernhard Banas
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Bergler
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
23
|
Buron F, Reffet S, Badet L, Morelon E, Thaunat O. Immunological Monitoring in Beta Cell Replacement: Towards a Pathophysiology-Guided Implementation of Biomarkers. Curr Diab Rep 2021; 21:19. [PMID: 33895937 DOI: 10.1007/s11892-021-01386-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 01/23/2023]
Abstract
PURPOSE OF REVIEW Grafted beta cells are lost because of recurrence of T1D and/or allograft rejection, two conditions diagnosed with pancreas graft biopsy, which is invasive and impossible in case of islet transplantation. This review synthetizes the current pathophysiological knowledge and discusses the interest of available immune biomarkers. RECENT FINDINGS Despite the central role of auto-(recurrence of T1D) and allo-(T-cell mediated rejection) immune cellular responses, the latter are not directly monitored in routine. In striking contrast, there have been undisputable progresses in monitoring of auto and alloantibodies. Except for pancreas recipients in whom anti-donor HLA antibodies can be directly responsible for antibody-mediated rejection, autoantibodies (and alloantibodies in islet recipients) have no direct pathogenic effect. However, their fluctuation offers a surrogate marker for the activation status of T cells (because antibody generation depends on T cells). This illustrates the necessity to understand the pathophysiology when interpreting a biomarker and selecting the appropriate treatment.
Collapse
Affiliation(s)
- Fanny Buron
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France
| | - Sophie Reffet
- Department of Endocrinology and Diabetes, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310, Pierre-Bénite, France
| | - Lionel Badet
- Department of Urology and Transplantation surgery, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France.
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France.
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, 5 Place d'Arsonval, 69003, Lyon, France.
| |
Collapse
|
24
|
Bestard O, Couzi L, Crespo M, Kessaris N, Thaunat O. Stratifying the humoral risk of candidates to a solid organ transplantation: a proposal of the ENGAGE working group. Transpl Int 2021; 34:1005-1018. [PMID: 33786891 DOI: 10.1111/tri.13874] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Detection of circulating antibodies directed against human leukocyte antigen (HLA) molecules, which corresponds to the current definition of 'sensitized patient', has been shown to have a severe impact on both access to transplantation and, if the anti-HLA antibodies are specific to the selected donor, survival of the graft. However, not all donor-specific antibodies (DSA) are equally harmful to the graft and progress in the understanding of humoral memory has led to the conclusion that absence of DSA at transplantation does not rule out the possibility that the patient has a preformed cellular humoral memory against the graft (thereby defining a category of DSA-negative sensitized recipients). Technological progress has led to the generation of new assays that offer unprecedented precision in exploring the different layers (serological and cellular) of alloimmune humoral memory. Based on this recent knowledge, the EuropeaN Guidelines for the mAnagement of Graft rEcipients (ENGAGE) working group to propose an updated definition of sensitization in candidates for solid organ transplantation - one that moves away from the current binary division towards a definition based on homogenous strata with similar humoral risk.
Collapse
Affiliation(s)
- Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain.,Bellvitge Research Institute (IDIBELL), Barcelona, Spain
| | - Lionel Couzi
- Nephrology-Transplantation-Dialysis, CHU Bordeaux, Bordeaux, France.,CNRS-UMR 5164 Immuno ConcEpT, Bordeaux University, Bordeaux, France
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain.,Nephropathies Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Nicos Kessaris
- Department of Nephrology and Transplantation, Guy's Hospital, London, UK.,King's College London, London, UK
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Lyon, France.,Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| |
Collapse
|
25
|
Abstract
BACKGROUND Norovirus (NoV) infection frequently progresses to chronic disease after kidney transplant (KTx). This study aims to assess potential risk factors helping to determine patients at risk of chronic NoV infection and to analyse the effect of NoV on allograft outcome. Additionally, we assessed the effectiveness of intravenous immunoglobulin (IVIg) therapy for chronic NoV infection. METHODS The study enrolled 60 KTx patients requiring hospitalization because of NoV infection. Clinical parameters, severity of NoV infection and potential risk factors were evaluated. Outcome parameters were clinical symptoms, rehospitalizations, persistent shedding of virus and effects on allograft function. RESULTS Patients were divided into 2 groups: 29 had acute NoV infection only, 31 progressed to chronic NoV infection. Chronic NoV infection was defined as a recurrence of clinical symptoms plus redetection of NoV in stool. Lymphocyte-depleting induction therapy and diabetes mellitus were independent risk factors for chronic infection. For patients with chronic NoV infection, length of stay in hospital was significantly prolonged (p= 0.024). Allograft function remained impaired in the chronic NoV group 6 and 12 months after initial admission.IVIg was administered to 18 patients with chronic NoV infection. No further clinical symptoms of NoV infection occurred in 13 (72%) of these patients. However, NoV was still detectable in stool specimens from 10 (77%) of these patients. CONCLUSIONS Chronic NoV infection is associated with reduced allograft function. Administration of IVIg to patients with chronic NoV infection seems beneficial in achieving freedom from clinical symptoms, despite limited effects on shedding of virus.
Collapse
|
26
|
Abstract
Initially overlooked in favor of T cell-mediated rejection, the importance of the humoral alloimmune response has progressively emerged. As a result, antibody-mediated rejection is now widely recognized as the main cause of late allograft loss in most (if not all) types of solid-organ transplantation. Over the last 2 decades, vascularized composite allotransplantation (VCA) has appeared for replacing tissue defects in patients for whom no other satisfactory reconstructive options were available. Although it is now clear that VCA recipients can develop donor-specific antibodies, conclusions made in solid organ transplantation regarding antibody-mediated rejection may not systematically apply to VCA. Here, we propose to use the experience gained in organ transplantation to shed light on the path that shall be followed to evaluate and manage humoral alloreactivity in VCA recipients.
Collapse
|
27
|
Koenig A, Mezaache S, Callemeyn J, Barba T, Mathias V, Sicard A, Charreau B, Rabeyrin M, Dijoud F, Picard C, Meas-Yedid V, Olivo-Marin JC, Morelon E, Naesens M, Dubois V, Thaunat O. Missing Self-Induced Activation of NK Cells Combines with Non-Complement-Fixing Donor-Specific Antibodies to Accelerate Kidney Transplant Loss in Chronic Antibody-Mediated Rejection. J Am Soc Nephrol 2021; 32:479-494. [PMID: 33239394 PMCID: PMC8054908 DOI: 10.1681/asn.2020040433] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/06/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules ("missing self") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR. METHODS AND RESULTS Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage. CONCLUSIONS The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.
Collapse
Affiliation(s)
- Alice Koenig
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Sarah Mezaache
- Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Jasper Callemeyn
- Department of Microbiology, Immunology and Transplantation, Catholic University (KU) Leuven, University of Leuven, Leuven, Belgium,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Thomas Barba
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Virginie Mathias
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Human Leukocyte Antigen (HLA) Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Antoine Sicard
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Béatrice Charreau
- Centre de Recherche en Transplantation et Immunologie (CRTI), University Hospital Center (CHU) Nantes, Université de Nantes, National Institute for Health and Medical Research (INSERM), Mixed University Unit (UMR) 1064, Transplantation Urology Nephrology Institute (ITUN), Nantes, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | | | - Cécile Picard
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | | | | | - Emmanuel Morelon
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Catholic University (KU) Leuven, University of Leuven, Leuven, Belgium,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Valérie Dubois
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Human Leukocyte Antigen (HLA) Laboratory, French National Blood Service (EFS), Décines-Charpieu, France
| | - Olivier Thaunat
- International Center of Infectiology research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
28
|
Thaunat O, Legeai C, Anglicheau D, Couzi L, Blancho G, Hazzan M, Pastural M, Savoye E, Bayer F, Morelon E, Le Meur Y, Bastien O, Caillard S. IMPact of the COVID-19 epidemic on the moRTAlity of kidney transplant recipients and candidates in a French Nationwide registry sTudy (IMPORTANT). Kidney Int 2020; 98:1568-1577. [PMID: 33137341 PMCID: PMC7604114 DOI: 10.1016/j.kint.2020.10.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/10/2020] [Accepted: 10/11/2020] [Indexed: 12/31/2022]
Abstract
End stage kidney disease increase the risk of COVID-19 related death but how the kidney replacement strategy should be adapted during the pandemic is unknown. Chronic hemodialysis makes social distancing difficult to achieve. Alternatively, kidney transplantation could increase the severity of COVID-19 due to therapeutic immunosuppression and contribute to saturation of intensive care units. For these reasons, kidney transplantation was suspended in France during the first epidemic wave. Here, we retrospectively evaluated this strategy by comparing the overall and COVID-19 related mortality in kidney transplant recipients and candidates over the last three years. Cross-interrogation of two national registries for the period 1 March and 1 June 2020, identified 275 deaths among the 42812 kidney transplant recipients and 144 deaths among the 16210 candidates. This represents an excess of deaths for both populations, as compared with the same period the two previous years (mean of two previous years: 253 in recipients and 112 in candidates). This difference was integrally explained by COVID-19, which accounted for 44% (122) and 42% (60) of the deaths in recipients and candidates, respectively. Taking into account the size of the two populations and the geographical heterogeneity of virus circulation, we found that the excess of risk of death due to COVID-19 was similar for recipients and candidates in high viral risk area but four-fold higher for candidates in the low viral risk area. Thus, in case of a second epidemic wave, kidney transplantation should be suspended in high viral risk areas but maintained outside those areas, both to reduce the excess of deaths of candidates and avoid wasting precious resources.
Collapse
Affiliation(s)
- Olivier Thaunat
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France; Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France; CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, 21, Lyon, France.
| | | | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Lionel Couzi
- Department of Nephrology and Transplantation, University of Bordeaux, Bordeaux, France
| | - Gilles Blancho
- Institut de Transplantation-Urologie-Néphrologie (ITUN), CHU Nantes, France; Centre de Recherche en Transplantation et Immunologie, UMR 1064 Inserm, Université de Nantes, Nantes, France
| | - Marc Hazzan
- University Lille, Inserm, CHU Lille, U1286-Infinite-Institute for Translational Research in Inflammation, Lille, France
| | | | - Emilie Savoye
- Agence de la Biomédecine, Saint Denis la Plaine, France
| | - Florian Bayer
- Agence de la Biomédecine, Saint Denis la Plaine, France
| | - Emmanuel Morelon
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France; Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, Lyon, France; CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, University Lyon, 21, Lyon, France
| | - Yann Le Meur
- Department of Nephrology and Transplantation, University of Brest, Brest, France
| | | | - Sophie Caillard
- Department of Nephrology and Transplantation, Strasbourg University Hospital, Strasbourg, France
| |
Collapse
|
29
|
Chen CC, Lin WC, Lee CY, Yang CY, Tsai MK. Two-year protocol biopsy after kidney transplantation in clinically stable recipients - a retrospective study. Transpl Int 2020; 34:185-193. [PMID: 33152140 DOI: 10.1111/tri.13785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/14/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
The idea of protocol biopsy is to detect subclinical pathologies, including rejection, recurrent disease, or infection for early intervention and adjustment of immunosuppressants. Nevertheless, it is not adopted by most clinicians because of its low yield rate and uncertain long-term benefits. This retrospective study evaluated the impact of protocol biopsy on renal function and allograft survival. A two-year protocol biopsy was proposed for 190 stable patients; 68 of them accepted [protocol biopsy (PB) group], while 122 did not [nonprotocol biopsy (NPB) group]. The rejection diagnosis was made in 13 patients by protocol biopsy, and 11 of them had borderline rejection. In the following 5 years, graft survival was better in the PB group than in the NPB group (P = 0.0143). A total of 4 and 17 patients in the PB and NPB groups, respectively, had rejection events proven by indication biopsy. Renal function was better preserved in the PB group than in the NPB group (P = 0.0107) for patients with rejection events. Nevertheless, the survival benefit disappeared by a longer follow-up period (12-year, P = 0.2886). In conclusion, 2-year protocol biopsy detects subclinical pathological changes in rejection and preserves renal function by early intervention so as to prolong graft survival within 5 years.
Collapse
Affiliation(s)
- Chien-Chia Chen
- Division of General Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Chou Lin
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yuan Lee
- Division of General Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Yao Yang
- Division of General Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Meng-Kun Tsai
- Division of General Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| |
Collapse
|
30
|
Xiu MX, Liu YM, Wang WJ. Investigation of hub genes and immune status in heart transplant rejection using endomyocardial biopsies. J Cell Mol Med 2020; 25:763-773. [PMID: 33230903 PMCID: PMC7812257 DOI: 10.1111/jcmm.16127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
T cell‒mediated rejection (TCMR) and antibody‐mediated rejection (ABMR) are severe post‐transplantation complications for heart transplantation (HTx), whose molecular and immunological pathogenesis remains unclear. In the present study, the mRNA microarray data set GSE124897 containing 645 stable, 52 TCMR and 144 ABMR endomyocardial biopsies was obtained to screen for differentially expressed genes (DEGs) between rejected and stable HTx samples and to investigate immune cell infiltration. Functional enrichment analyses indicated roles of the DEGs primarily in immune‐related mechanisms. Protein‐protein interaction networks were then constructed, and ICAM1, CD44, HLA‐A and HLA‐B were identified as hub genes using the maximal clique centrality method. Immune cell infiltration analysis revealed differences in adaptive and innate immune cell populations between TCMR, ABMR and stable HTx samples. Additionally, hub gene expression levels significantly correlated with the degree and composition of immune cell infiltration in HTx rejection samples. Furthermore, drug‐gene interactions were constructed, and 12 FDA‐approved drugs were predicted to target hub genes. Finally, an external GSE2596 data set was used to validate the expression of the hub genes, and ROC curves indicated all four hub genes had promising diagnostic value for HTx rejection. This study provides a comprehensive perspective of molecular and immunological regulatory mechanisms underlying HTx rejection.
Collapse
Affiliation(s)
- Meng-Xi Xiu
- Medical School of Nanchang University, Nanchang, China
| | - Yuan-Meng Liu
- Medical School of Nanchang University, Nanchang, China
| | - Wen-Jun Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Gierczak V, Jouve T, Malvezzi P, Terrec F, Naciri-Bennani H, Janbon B, Rostaing L, Noble J. Impact of Immunosuppressive Strategies on Post-Kidney Transplantation Thrombocytopenia. Transplant Proc 2020; 53:941-949. [PMID: 33121819 DOI: 10.1016/j.transproceed.2020.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/11/2020] [Accepted: 09/18/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Thrombocytopenia after kidney transplantation is a common complication, partly induced by immunosuppressive therapies. Peritransplant thrombocytopenia may cause serious hemorrhages. We assessed the incidence of early posttransplantation thrombocytopenia (defined as a platelet count of <150,000 mm3 or <150 G/L) in de novo kidney transplant recipients (KTRs) across 4 immunosuppressive regimens. METHODS This was a single-center observational study that included all consecutive KTRs who received either Thymoglobulin (THY) or Grafalon (GRA) and maintenance therapy of either mycophenolate-mofetil (MMF) or everolimus (EVR), associated with tacrolimus/corticosteroids. RESULTS Between July 27, 2016, and September 7, 2018, 237 KTRs were included; 64.6% experienced thrombocytopenia within the first week. Thrombocytopenia was significantly more frequent (P = .004) among GRA-treated patients (73.4%) compared to THY-treated patients (61.3%). These patients also had lower nadir platelet count (120 ± 52 vs 142 ± 48 G/L; P = .002) and lower platelet count at discharge (227 ± 94 vs 243 ± 92 G/L; P = .25). More of the GRA-EVR group had thrombocytopenia (81.0% vs 61.4% in THY-MMF, 60.9% in THY-EVR, and 69.8% in GRA-MMF; P = .081) and a worse nadir platelet count (109 ± 41 in GRA-EVR vs 141 ± 47G/L in THY-MMF, 145 ± 52 G/L in THY-EVR, and 125 ± 56 G/L in GRA-MMF; P = .011) but GRA was the only risk factor for thrombocytopenia in multivariate analyses (P = .002). Rates of hemorrhage, red blood cell transfusions, reoperations needed within the first week, delayed graft function, acute rejection, graft loss, and death did not differ between the groups after a mean follow-up of 25 ± 8 months. CONCLUSIONS GRA associated with EVR led to more frequent and severe thrombocytopenia, although we found no significant clinical consequences.
Collapse
Affiliation(s)
- Valentine Gierczak
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France; Grenoble Alpes University, Grenoble, France
| | - Thomas Jouve
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France; Grenoble Alpes University, Grenoble, France
| | - Paolo Malvezzi
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France
| | - Florian Terrec
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France
| | - Hamza Naciri-Bennani
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France
| | - Benedicte Janbon
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France
| | - Lionel Rostaing
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France; Grenoble Alpes University, Grenoble, France.
| | - Johan Noble
- Nephrology Department, Hémodialyse, Apheresis and Renal Transplantation, CHU Grenoble-Alpes, Grenoble, France
| |
Collapse
|
32
|
Donor-specific B Cell Memory in Alloimmunized Kidney Transplant Recipients: First Clinical Application of a Novel Method. Transplantation 2020; 104:1026-1032. [DOI: 10.1097/tp.0000000000002909] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
33
|
Koenig A, Chen CC, Marçais A, Barba T, Mathias V, Sicard A, Rabeyrin M, Racapé M, Duong-Van-Huyen JP, Bruneval P, Loupy A, Dussurgey S, Ducreux S, Meas-Yedid V, Olivo-Marin JC, Paidassi H, Guillemain R, Taupin JL, Callemeyn J, Morelon E, Nicoletti A, Charreau B, Dubois V, Naesens M, Walzer T, Defrance T, Thaunat O. Missing self triggers NK cell-mediated chronic vascular rejection of solid organ transplants. Nat Commun 2019; 10:5350. [PMID: 31767837 PMCID: PMC6877588 DOI: 10.1038/s41467-019-13113-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Current doctrine is that microvascular inflammation (MVI) triggered by a transplant -recipient antibody response against alloantigens (antibody-mediated rejection) is the main cause of graft failure. Here, we show that histological lesions are not mediated by antibodies in approximately half the participants in a cohort of 129 renal recipients with MVI on graft biopsy. Genetic analysis of these patients shows a higher prevalence of mismatches between donor HLA I and recipient inhibitory killer cell immunoglobulin-like receptors (KIRs). Human in vitro models and transplantation of β2-microglobulin-deficient hearts into wild-type mice demonstrates that the inability of graft endothelial cells to provide HLA I-mediated inhibitory signals to recipient circulating NK cells triggers their activation, which in turn promotes endothelial damage. Missing self-induced NK cell activation is mTORC1-dependent and the mTOR inhibitor rapamycin can prevent the development of this type of chronic vascular rejection. ‘Missing self’ is a mode of natural killer (NK) cell activation aimed to detect the lack of HLA-I molecules on infected or neoplastic cells. Here, the authors show that mismatch between donor HLA-I and cognate receptors on recipient NK cells mediates microvascular inflammation-associated graft rejection, a pathology that is preventable by mTOR inhibition.
Collapse
Affiliation(s)
- Alice Koenig
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d'Arsonval, 69003, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 8, avenue Rockfeller, 69373, Lyon, France
| | - Chien-Chia Chen
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France
| | - Antoine Marçais
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France
| | - Thomas Barba
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d'Arsonval, 69003, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 8, avenue Rockfeller, 69373, Lyon, France
| | - Virginie Mathias
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,French National Blood Service (EFS), HLA Laboratory, 111, rue Elisée-Reclus, 69153, Décines-Charpieu, France
| | - Antoine Sicard
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d'Arsonval, 69003, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 8, avenue Rockfeller, 69373, Lyon, France
| | - Maud Rabeyrin
- Hospices Civils de Lyon, Department of Pathology, 59, boulevard Pinel, 69500, Bron, France
| | - Maud Racapé
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, 12, rue de l'Ecole de Médecine, 75006, Paris, France
| | - Jean-Paul Duong-Van-Huyen
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, 12, rue de l'Ecole de Médecine, 75006, Paris, France
| | - Patrick Bruneval
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, 12, rue de l'Ecole de Médecine, 75006, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Centre for Organ Transplantation, Paris Descartes University, 12, rue de l'Ecole de Médecine, 75006, Paris, France
| | - Sébastien Dussurgey
- SFR Biosciences (UMS3444/CNRS, US8/Inserm, ENS de Lyon, UCBL), 50, avenue Tony-Garnier, 69007, Lyon, France
| | - Stéphanie Ducreux
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,French National Blood Service (EFS), HLA Laboratory, 111, rue Elisée-Reclus, 69153, Décines-Charpieu, France
| | - Vannary Meas-Yedid
- Unité d'Analyse d'Images Biologiques, Pasteur Institut, 25-28, rue du Docteur-Roux, 75015, Paris, France
| | | | - Héléna Paidassi
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France
| | - Romain Guillemain
- Assistance Publique - Hôpitaux de Paris, Georges Pompidou Hospital, Cardiology and Heart Transplant Department, 20, rue Leblanc, 75015, Paris, France
| | - Jean-Luc Taupin
- Assistance Publique - Hôpitaux de Paris, Immunology and HLA Laboratory, Saint-Louis Hospital, 1, avenue Claude-Vellefaux, 75010, Paris, France.,French National Institute of Health and Medical Research (Inserm) Unit 1160, 1, avenue Claude-Vellefaux, 75010, Paris, France.,Paris Diderot University, 5, rue Thomas-Mann, 75013, Paris, France
| | - Jasper Callemeyn
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Herestraat 49, Box 7003, 3000, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Emmanuel Morelon
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d'Arsonval, 69003, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 8, avenue Rockfeller, 69373, Lyon, France
| | - Antonino Nicoletti
- Paris Diderot University, 5, rue Thomas-Mann, 75013, Paris, France.,French National Institute of Health and Medical Research (Inserm) Unit 1148, Laboratory of Vascular Translational Science, 46, rue Henri-Huchard, 75018, Paris, France
| | - Béatrice Charreau
- French National Institute of Health and Medical Research (Inserm) UMR1064, 30, boulevard Jean-Monnet, 44093, Nantes Cedex 01, France
| | - Valérie Dubois
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France.,French National Blood Service (EFS), HLA Laboratory, 111, rue Elisée-Reclus, 69153, Décines-Charpieu, France
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, University of Leuven, Herestraat 49, Box 7003, 3000, Leuven, Belgium.,Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Thierry Walzer
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France
| | - Thierry Defrance
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France
| | - Olivier Thaunat
- CIRI, INSERM U1111, Université Claude Bernard Lyon I, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Univ. Lyon, 21, avenue Tony Garnier, 69007, Lyon, France. .,Hospices Civils de Lyon, Edouard Herriot Hospital, Department of Transplantation, Nephrology and Clinical Immunology, 5, place d'Arsonval, 69003, Lyon, France. .,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), 8, avenue Rockfeller, 69373, Lyon, France.
| |
Collapse
|
34
|
Niu Q, Kraaijeveld R, Li Y, Mendoza Rojas A, Shi Y, Wang L, Van Besouw NM, Baan CC. An overview of T follicular cells in transplantation: spotlight on their clinical significance. Expert Rev Clin Immunol 2019; 15:1249-1262. [PMID: 31721600 DOI: 10.1080/1744666x.2020.1693262] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: For late stage organ failure patients, transplantation is the best option to increase life expectancy with a superior quality of life. Unfortunately, after transplantation many patients are at risk of cellular and antibody-mediated rejection (ABMR). The latter is initiated by donor specific antibodies (DSA) which depend on the actions of B cells, T follicular helper (Tfh) cells and T follicular regulatory (Tfr) cells that are present in the germinal center of lymphoid organs.Areas covered: In this overview paper, we discuss the biology and function of Tfh and Tfr cells in lymphoid tissues, transplanted organs and their circulating counterparts. We report on their relevance to alloimmunity and on the effects of immunosuppressive drugs on these immunocompetent cell populations.Expert opinion: Growing knowledge about the actions of Tfh and Tfr allows for a better understanding of the immunological mechanisms of ABMR after organ transplantation. This understanding feeds the hypothesis that immunosuppressive drugs targeting the actions of Tfh cells have huge therapeutic potential. This new concept in the treatment of the humoral rejection response will improve graft and patient survival after organ transplantation.
Collapse
Affiliation(s)
- Qian Niu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.,Department Internal Medicine - Sector Nephrology & Transplantation, The Rotterdam Transplant Group, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Rens Kraaijeveld
- Department Internal Medicine - Sector Nephrology & Transplantation, The Rotterdam Transplant Group, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Yi Li
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Aleixandra Mendoza Rojas
- Department Internal Medicine - Sector Nephrology & Transplantation, The Rotterdam Transplant Group, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Yunying Shi
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lanlan Wang
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Nicole M Van Besouw
- Department Internal Medicine - Sector Nephrology & Transplantation, The Rotterdam Transplant Group, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Carla C Baan
- Department Internal Medicine - Sector Nephrology & Transplantation, The Rotterdam Transplant Group, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Lattimore S, Skill NJ, Maluccio MA, Elliott H, Dobben E, Shafuddin A, Goggins WC. Antithymocyte Globulin Antibody Titer Congruent With Kidney Transplantation: Analysis of Incidence, Outcomes, Cost, and Alternative Targets. Transplant Direct 2019; 5:e493. [PMID: 31723588 PMCID: PMC6791597 DOI: 10.1097/txd.0000000000000933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/19/2019] [Indexed: 11/25/2022] Open
Abstract
Rabbit antithymocyte globulin (rATG) use for immunosuppression induction is widespread but is contraindicated by the presence of anti-rATG antibodies. This study reports the incidence of positive anti-rATG antibody titers in patients before and after renal transplant and evaluates associated outcomes and costs. In addition, it will correlate CD40L and interleukin (IL)-21 with anti-rATG antibody titers. METHODS Clinical and billing records from the Indiana University Transplant Laboratory were reviewed for positive versus negative anti-rATG antibody titers, graft survival, and 7-day readmission costs between 2004 and 2018. Serum from patients with positive and negative rATG antibody titers were quantitated for CD40L and IL-21 by enzyme-linked immunosorbent assay. RESULTS On average, between 2004 and May 2018, 163 kidney transplants per year were performed. Anti-rATG antibody titers were ordered for 17 patients/year, of which 18.2% were positive at 1:100 titer either pre- or post-transplant. Time to graft loss correlated with a positive rATG titer at time of readmission. Moreover, second kidney transplant increased the anti-rATG positive rate. A weak correlation was observed between anti-rATG titer and recipient age. Seven-day readmission treatment costs were significantly lower in patients with positive anti-rATG titer. IL-21 and CD40L were significantly greater in patients with positive anti-rATG titers after transplant when compared with negative anti rATG patients. CONCLUSIONS Positive anti-rATG antibody titer is associated with a significant negative impact on outcomes. Monitoring of anti-rATG antibody titer is recommended to optimize treatment options in patients, especially in the setting of second transplants. Elucidation of the mechanisms associated with positive anti-rATG antibody is required. IL-21 and CD40L are potential targets for future study.
Collapse
Affiliation(s)
- Sherene Lattimore
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Nicholas J. Skill
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Mary A. Maluccio
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Holly Elliott
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Elizabeth Dobben
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Asif Shafuddin
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - William C. Goggins
- Division of Transplant, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
36
|
Danger R, Chesneau M, Delbos F, Le Bot S, Kerleau C, Chenouard A, Ville S, Degauque N, Conchon S, Cesbron A, Giral M, Brouard S. CXCR5 +PD1 +ICOS + Circulating T Follicular Helpers Are Associated With de novo Donor-Specific Antibodies After Renal Transplantation. Front Immunol 2019; 10:2071. [PMID: 31552030 PMCID: PMC6746839 DOI: 10.3389/fimmu.2019.02071] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/16/2019] [Indexed: 12/21/2022] Open
Abstract
Donor-specific anti-HLA antibodies (DSAs) are a major risk factor associated with renal allograft outcomes. As a trigger of B cell antibody production, T follicular helper cells (Tfhs) promote DSA appearance. Herein, we evaluated whether circulating Tfhs (cTfhs) are associated with the genesis of antibody-mediated rejection. We measured cTfh levels on the day of transplantation and 1 year after transplantation in blood from a prospective cohort of 237 renal transplantation patients without DSA during the first year post-transplantation. Total cTfhs were characterized as CD4+CD45RA−CXCR5+, and the three following subsets of activated cTfh were analyzed: CXCR5+PD1+, CXCR5+PD1+ICOS+, an CXCR5+PD1+CXCR3−. Immunizing events (previous blood transfusion and/or pregnancy) and the presence of class II anti-HLA antibodies were associated with increased frequencies of activated CXCR5+PD1+, CXCR5+PD1+ICOS+, and CXCR5+PD1+CXCR3− cTfh subsets. In addition, ATG-depleting induction and calcineurin inhibitor treatments were associated with a relative increase of activated cTfh subsets frequencies at 1 year post-transplantation. In multivariate survival analysis, we reported that a decrease in activated CXCR5+PD1+ICOS+ at 1 year after transplantation in the blood of DSA-free patients was significantly associated with the risk of developing de novo DSA after the first year (p = 0.018, HR = 0.39), independently of HLA mismatches (p = 0.003, HR = 3.79). These results highlight the importance of monitoring activated Tfhs in patients early after transplantation and show that current treatments cannot provide early, efficient prevention of Tfh activation and migration. These findings indicate the need to develop innovative treatments to specifically target Tfhs to prevent DSA appearance in renal transplantation.
Collapse
Affiliation(s)
- Richard Danger
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Mélanie Chesneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Florent Delbos
- Laboratoire Histocompatibilité et Immunogénétique - Etablissement Français du sang, Nantes, France
| | - Sabine Le Bot
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Clarisse Kerleau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Alexis Chenouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Simon Ville
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Nicolas Degauque
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Sophie Conchon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Anne Cesbron
- Laboratoire Histocompatibilité et Immunogénétique - Etablissement Français du sang, Nantes, France
| | - Magali Giral
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Centre d'Investigation Clinique en Biothérapie, Centre de Ressources Biologiques (CRB), Labex IGO, Nantes, France
| |
Collapse
|
37
|
Singh A, Ramachandran S, Graham ML, Daneshmandi S, Heller D, Suarez-Pinzon WL, Balamurugan AN, Ansite JD, Wilhelm JJ, Yang A, Zhang Y, Palani NP, Abrahante JE, Burlak C, Miller SD, Luo X, Hering BJ. Long-term tolerance of islet allografts in nonhuman primates induced by apoptotic donor leukocytes. Nat Commun 2019; 10:3495. [PMID: 31375697 PMCID: PMC6677762 DOI: 10.1038/s41467-019-11338-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Immune tolerance to allografts has been pursued for decades as an important goal in transplantation. Administration of apoptotic donor splenocytes effectively induces antigen-specific tolerance to allografts in murine studies. Here we show that two peritransplant infusions of apoptotic donor leukocytes under short-term immunotherapy with antagonistic anti-CD40 antibody 2C10R4, rapamycin, soluble tumor necrosis factor receptor and anti-interleukin 6 receptor antibody induce long-term (≥1 year) tolerance to islet allografts in 5 of 5 nonsensitized, MHC class I-disparate, and one MHC class II DRB allele-matched rhesus macaques. Tolerance in our preclinical model is associated with a regulatory network, involving antigen-specific Tr1 cells exhibiting a distinct transcriptome and indirect specificity for matched MHC class II and mismatched class I peptides. Apoptotic donor leukocyte infusions warrant continued investigation as a cellular, nonchimeric and translatable method for inducing antigen-specific tolerance in transplantation. Injection of donor apoptotic cells induces graft tolerance in mice. Here the authors combine this approach with short immunosuppressive therapy to achieve long-term tolerance to allogeneic islets and restoration of normoglycemia in diabetic nonhuman primates, and delineate cellular and molecular correlates of tolerance induction.
Collapse
Affiliation(s)
- Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sabarinathan Ramachandran
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Melanie L Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Saeed Daneshmandi
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - David Heller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wilma Lucia Suarez-Pinzon
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Appakalai N Balamurugan
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA.,Center for Cellular Transplantation, Cardiovascular Innovation Institute, Department of Surgery, University of Louisville, Louisville, KY, 40202, USA
| | - Jeffrey D Ansite
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joshua J Wilhelm
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Amy Yang
- Biostatistics Collaboration Center, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nagendra P Palani
- University of Minnesota Genomics Center, Minneapolis, MN, 55455, USA
| | - Juan E Abrahante
- University of Minnesota Informatics Institute, Minneapolis, MN, 55455, USA
| | - Christopher Burlak
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunology Center, Northwestern University, Chicago, IL, 60611, USA.
| | - Xunrong Luo
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Biostatistics Collaboration Center, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Duke Transplant Center, Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
38
|
Laguna-Goya R, Suàrez-Fernández P, Paz-Artal E. Follicular helper T cells and humoral response in organ transplantation. Transplant Rev (Orlando) 2019; 33:183-190. [PMID: 31327572 DOI: 10.1016/j.trre.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/17/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
Antibody mediated rejection has been recognized as an important contributor to long-term graft loss in most solid organ transplants. Current immunosuppressive regimes are not capable of preventing anti-HLA antibody formation and eventual damage to the graft, and there is a need to develop drugs directed against novel targets to avoid graft allorecognition. In this review we introduce follicular helper T cells (Tfh), a subtype of lymphocyte specialized in helping B cells to differentiate into plasmablasts and produce class-switched antibodies. We focus on the role of Tfh in solid organ transplantation, what is known about Tfh and the production of alloantibodies, how current immunosuppressive therapies affect Tfh and what new molecules could be used to target Tfh in transplantation, with the goal of improving graft survival.
Collapse
Affiliation(s)
- R Laguna-Goya
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de investigación Hospital Universitario 12 de Octubre (Imas12), Madrid, Spain; School of Medicine, Universidad Complutense de Madrid, Spain.
| | - P Suàrez-Fernández
- Instituto de investigación Hospital Universitario 12 de Octubre (Imas12), Madrid, Spain
| | - E Paz-Artal
- Immunology Department, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de investigación Hospital Universitario 12 de Octubre (Imas12), Madrid, Spain; School of Medicine, Universidad Complutense de Madrid, Spain
| |
Collapse
|
39
|
Recent Topics on The Mechanisms of Immunosuppressive Therapy-Related Neurotoxicities. Int J Mol Sci 2019; 20:ijms20133210. [PMID: 31261959 PMCID: PMC6651704 DOI: 10.3390/ijms20133210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023] Open
Abstract
Although transplantation procedures have been developed for patients with end-stage hepatic insufficiency or other diseases, allograft rejection still threatens patient health and lifespan. Over the last few decades, the emergence of immunosuppressive agents such as calcineurin inhibitors (CNIs) and mammalian target of rapamycin (mTOR) inhibitors have strikingly increased graft survival. Unfortunately, immunosuppressive agent-related neurotoxicity commonly occurs in clinical practice, with the majority of neurotoxicity cases caused by CNIs. The possible mechanisms through which CNIs cause neurotoxicity include increasing the permeability or injury of the blood–brain barrier, alterations of mitochondrial function, and alterations in the electrophysiological state. Other immunosuppressants can also induce neuropsychiatric complications. For example, mTOR inhibitors induce seizures, mycophenolate mofetil induces depression and headaches, methotrexate affects the central nervous system, the mouse monoclonal immunoglobulin G2 antibody (used against the cluster of differentiation 3) also induces headaches, and patients using corticosteroids usually experience cognitive alteration. Therapeutic drug monitoring, individual therapy based on pharmacogenetics, and early recognition of symptoms help reduce neurotoxic events considerably. Once neurotoxicity occurs, a reduction in the drug dosage, switching to other immunosuppressants, combination therapy with drugs used to treat the neuropsychiatric manifestation, or blood purification therapy have proven to be effective against neurotoxicity. In this review, we summarize recent topics on the mechanisms of immunosuppressive drug-related neurotoxicity. In addition, information about the neuroprotective effects of several immunosuppressants is also discussed.
Collapse
|
40
|
Ho CM, Chen HL, Hu RH, Lee PH. Harnessing immunotherapy for liver recipients with hepatocellular carcinoma: a review from a transplant oncology perspective. Ther Adv Med Oncol 2019; 11:1758835919843463. [PMID: 31065295 PMCID: PMC6487770 DOI: 10.1177/1758835919843463] [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: 11/28/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
Without stringent criteria, liver transplantation for hepatocellular carcinoma (HCC) can lead to high cancer recurrence and poor prognosis in the current treatment context. Checkpoint inhibitors can lead to long survival by targeting coinhibitory pathways and promoting T-cell activity; thus, they have great potential for cancer immunotherapy. Therapeutic modulation of cosignaling pathways may shift paradigms from surgical prevention of recurrence to oncological intervention. Herein, we review the available evidence from a therapeutic perspective and focus on immune microenvironment perturbation by immunosuppressants and checkpoint inhibitors. Partial and reversible interleukin-2 signaling blockade is the mainstream strategy of immunosuppression for graft protection. Programmed cell death protein 1 (PD-1) is abundantly expressed on human liver allograft-infiltrating T-cells, which proliferate considerably after programmed death-ligand 1 (PD-L1) blockade. Clinically, checkpoint inhibitors are used in heart, liver, and kidney recipients with various cancers. Rejection can occur after checkpoint inhibitor administration through acute T-cell-mediated, antibody-mediated, or chronic allograft rejection mechanisms. Nevertheless, liver recipients may demonstrate favorable responses to treatment for HCC recurrence without rejection. Pharmacodynamically, substantial degrees of receptor occupancy can be achieved with lower doses, with favorable clinical outcomes. Manipulation of the immune microenvironment is a therapeutic niche that balances seemingly conflicting anticancer and graft protection needs. Additional translational and clinical studies emphasizing the comparative effectiveness of signaling networks within the immune microenvironment and conducting overall assessment of the immune microenvironment may aid in creating a therapeutic window and benefiting future liver recipients with HCC recurrence.
Collapse
Affiliation(s)
- Cheng-Maw Ho
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | - Hui-Ling Chen
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Rey-Heng Hu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Huang Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
41
|
Barba T, Harb J, Ducreux S, Koenig A, Mathias V, Rabeyrin M, Pouliquen E, Sicard A, Chartoire D, Dugast E, Defrance T, Morelon E, Brouard S, Dubois V, Thaunat O. Highly Variable Sialylation Status of Donor-Specific Antibodies Does Not Impact Humoral Rejection Outcomes. Front Immunol 2019; 10:513. [PMID: 30949173 PMCID: PMC6435580 DOI: 10.3389/fimmu.2019.00513] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Clinical outcome in antibody-mediated rejection (AMR) shows high inter-individual heterogeneity. Sialylation status of the Fc fragment of IgGs is variable, which could modulate their ability to bind to C1q and/or Fc receptors. In this translational study, we evaluated whether DSA sialylation influence AMR outcomes. Among 938 kidney transplant recipients for whom a graft biopsy was performed between 2004 and 2012 at Lyon University Hospitals, 69 fulfilled the diagnosis criteria for AMR and were enrolled. Sera banked at the time of the biopsy were screened for the presence of DSA by Luminex. The sialylation status of total IgG and DSA was quantified using Sambucus nigra agglutinin-based chromatography. All patients had similar levels of sialylation of serum IgGs (~2%). In contrast, the proportion of sialylated DSA were highly variable (median = 9%; range = 0–100%), allowing to distribute the patients in two groups: high DSA sialylation (n = 44; 64%) and low DSA sialylation (n = 25; 36%). The two groups differed neither on the intensity of rejection lesions (C4d, ptc, and g; p > 0.05) nor on graft survival rates (Log rank test, p = 0.99). in vitro models confirmed the lack of impact of Fc sialylation on the ability of a monoclonal antibody to trigger classical complement cascade and activate NK cells. We conclude that DSA sialylation status is highly variable but has not impact on DSA pathogenicity and AMR outcome.
Collapse
Affiliation(s)
- Thomas Barba
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Jean Harb
- French National Institute of Health and Medical Research (INSERM) UMR1064, Nantes, France.,Laboratory of Biochemistry, Nantes University Hospital, Nantes, France
| | | | - Alice Koenig
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Virginie Mathias
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | - Eric Pouliquen
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Antoine Sicard
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Dimitri Chartoire
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Emilie Dugast
- French National Institute of Health and Medical Research (INSERM) UMR1064, Nantes, France
| | - Thierry Defrance
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France
| | - Emmanuel Morelon
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Sophie Brouard
- French National Institute of Health and Medical Research (INSERM) UMR1064, Nantes, France
| | - Valérie Dubois
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,French National Blood Service (EFS), HLA Laboratory, Lyon, France
| | - Olivier Thaunat
- French National Institute of Health and Medical Research (INSERM) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
42
|
Ruszkowski J, Lisowska KA, Pindel M, Heleniak Z, Dębska-Ślizień A, Witkowski JM. T cells in IgA nephropathy: role in pathogenesis, clinical significance and potential therapeutic target. Clin Exp Nephrol 2019; 23:291-303. [PMID: 30406499 PMCID: PMC6394565 DOI: 10.1007/s10157-018-1665-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN), the most frequent cause of primary glomerulonephritis worldwide, is an autoimmune disease with complex pathogenesis. In this review, we focus on T cells and summarize knowledge about their involvement in pathophysiology and treatment of IgAN METHODS: We reviewed the literature for (1) alterations of T cell subpopulations in IgAN, (2) experimental and clinical proofs for T cells' participation in IgAN pathogenesis, (3) clinical correlations with T cell-associated alterations, and (4) influence of drugs used in IgAN therapy on T cell subpopulations. RESULTS We found that IgAN is characterized by higher proportions of circulatory Th2, Tfh, Th17, Th22 and γδ T cells, but lower Th1 and Treg cells. We discuss genetic and epigenetic makeup that may contribute to this immunological phenotype. We found that Th2, Th17 and Tfh-type interleukins contribute to elevated synthesis of galactose-deficient IgA1 (Gd-IgA1) and that the production of anti-Gd-IgA1 autoantibodies may be stimulated by Tfh cells. We described the roles of Th2, Th17, Th22 and Treg cells in the renal injury and summarized correlations between T cell-associated alterations and clinical features of IgAN (proteinuria, reduced GFR, hematuria). We detailed the impact of immunosuppressive drugs on T cell subpopulations and found that the majority of drugs have nonoptimal influence on T cells in IgAN patients. CONCLUSIONS T cells play an important role in IgAN pathogenesis and are correlated with its clinical severity. Clinical trials with the drugs targeting the reported alterations of the T-cell compartment are highly desirable.
Collapse
Affiliation(s)
- Jakub Ruszkowski
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland.
| | - Katarzyna A Lisowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland
| | - Małgorzata Pindel
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland
| | - Zbigniew Heleniak
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Jacek M Witkowski
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland
| |
Collapse
|
43
|
Dahdal S, Saison C, Valette M, Bachy E, Pallet N, Lina B, Koenig A, Monneret G, Defrance T, Morelon E, Thaunat O. Residual Activatability of Circulating Tfh17 Predicts Humoral Response to Thymodependent Antigens in Patients on Therapeutic Immunosuppression. Front Immunol 2019; 9:3178. [PMID: 30804950 PMCID: PMC6370621 DOI: 10.3389/fimmu.2018.03178] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/27/2018] [Indexed: 12/31/2022] Open
Abstract
The generation of antibodies against protein antigens (such as donor-specific HLA molecules) requires that T follicular helper cells (Tfh) provide help to B cells. Immunosuppressive (IS) armamentarium prevents T cell activation, yet a significant proportion of renal transplant patients develop donor-specific antibodies (DSA), which suggests that IS drugs do not efficiently block T follicular helper cells. To test this hypothesis, the number of circulating Tfh, their polarization profile, and ability to up-regulate (i) the co-stimulatory molecules CD40L and ICOS, and (ii) the activation marker CD25, following in vitro stimulation in presence of IS drugs, were compared between 36 renal transplant patients (6–72 months post transplantation) and nine healthy controls. IS drugs reduced the number of Tfh1 and 2 but had little impact on Tfh17, which was the dominant subset in transplant patients. Although, IS drugs decreased activation-induced expression of co-stimulatory molecules by Tfh, the impact was highly variable between individuals. Furthermore, 20% of transplant patients displayed normal expression of CD25 on Tfh following in vitro stimulation (i.e., “residual activatability”). To test whether residual activatability of Tfh correlates with antibody response against thymo-dependent antigens we took advantage of the 2015 influenza vaccination campaign, which provided a normalized setting for antigenic stimulation. In line with our hypothesis, responders to influenza vaccine exhibited significantly higher percentage of CD25-expressing Tfh17 after in vitro stimulation. A results that was confirmed retrospectively in nine transplanted patients at the time of first DSA detection. We concluded that “residual activatability” of Tfh17 might be used as a non-invasive biomarker to identify transplant patients at higher risk to develop DSA under immunosuppression. If validated in larger studies, this assay might help optimizing the prevention of DSA through personalized adaptation of immunosuppressive regimen.
Collapse
Affiliation(s)
- Suzan Dahdal
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France
| | - Carole Saison
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France
| | - Martine Valette
- Hospices Civils de Lyon, Croix-Rousse University Hospital, Infectious Agents Institute (IAI) Laboratory of Virology-National Reference Center for Respiratory Viruses (Including Influenza), Lyon, France
| | - Emmanuel Bachy
- Department of Hematology, Hospices Civils de Lyon, Lyon Sud University Hospital, Pierre Bénite, France.,Claude Bernard University (Lyon 1), Lyon, France
| | - Nicolas Pallet
- Laboratory of Biochemistry, Assistance Publique-Hôpitaux de Paris, Georges Pompidou Hospital, Paris, France.,Paris Descartes University, Paris, France
| | - Bruno Lina
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Hospices Civils de Lyon, Croix-Rousse University Hospital, Infectious Agents Institute (IAI) Laboratory of Virology-National Reference Center for Respiratory Viruses (Including Influenza), Lyon, France.,Claude Bernard University (Lyon 1), Lyon, France
| | - Alice Koenig
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France
| | - Guillaume Monneret
- Laboratory of Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France
| | - Thierry Defrance
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France
| | - Emmanuel Morelon
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France.,Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France.,Claude Bernard University (Lyon 1), Lyon, France
| |
Collapse
|
44
|
Macedo C, Hadi K, Walters J, Elinoff B, Marrari M, Zeevi A, Ramaswami B, Chalasani G, Landsittel D, Shields A, Alloway R, Lakkis FG, Woodle ES, Metes D. Impact of Induction Therapy on Circulating T Follicular Helper Cells and Subsequent Donor-Specific Antibody Formation After Kidney Transplant. Kidney Int Rep 2018; 4:455-469. [PMID: 30899873 PMCID: PMC6409398 DOI: 10.1016/j.ekir.2018.11.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
Introduction The cellular events that contribute to generation of donor-specific anti-HLA antibodies (DSA) post-kidney transplantation (KTx) are not well understood. Characterization of such mechanisms could allow tailoring of immunosuppression to benefit sensitized patients. Methods We prospectively monitored circulating T follicular helper (cTFH) cells in KTx recipients who received T-cell depleting (thymoglobulin, n = 54) or T-cell nondepleting (basiliximab, n = 20) induction therapy from pre-KTx to 1 year post-KTx and assessed their phenotypic changes due to induction and DSA occurrence, in addition to healthy controls (n = 13), for a total of 307 blood samples. Results Before KTx, patients displayed comparable levels of resting, central memory cTFH cells with similar polarization to those of healthy controls. Unlike basiliximab induction, thymoglobulin induction significantly depleted cTFH cells, triggered lymphopenia-induced proliferation that skewed cTFH cells toward increased Th1 polarization, effector memory, and elevated programmed cell death protein 1 (PD-1)int/hi expression, resembling activated phenotypes. Regardless of induction, patients who developed DSA post-KTx, harbored pre-KTx donor-reactive memory interleukin (IL)-21+ cTFH cells and showed higher % cTFH and lower % of T regulatory (TREG) cells post-KTx resulting in elevated cTFH:TREG ratio at DSA occurrence. Conclusion Induction therapy distinctly shapes cTFH cell phenotype post-KTx. Monitoring cTFH cells before and after KTx may help detect those patients prone to DSA generation post-KTx.
Collapse
Affiliation(s)
- Camila Macedo
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Hadi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Walters
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Beth Elinoff
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marilyn Marrari
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adriana Zeevi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bala Ramaswami
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Geetha Chalasani
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas Landsittel
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adele Shields
- Division of Transplantation, University of Cincinnati, Cincinnati, Ohio, USA
| | - Rita Alloway
- Division of Nephrology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - E Steve Woodle
- Division of Transplantation, University of Cincinnati, Cincinnati, Ohio, USA
| | - Diana Metes
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|