1
|
Liu Y, Feng H, Li K, Li R, Zhang XJ, Tian Y, Fang Y, Zhou Y, Liu L, Zhang X. Donor MHC-specific thymus vaccination allows for immunocompatible allotransplantation. Cell Res 2025; 35:132-144. [PMID: 39748049 PMCID: PMC11770082 DOI: 10.1038/s41422-024-01049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/17/2024] [Indexed: 01/04/2025] Open
Abstract
Organ transplantation is the last-resort option to treat organ failure. However, less than 10% of patients benefit from this only option due to lack of major histocompatibility complex (MHC)-matched donor organs and 25%-80% of donated organs could not find MHC-matched recipients. T cell allorecognition is the principal mechanism for allogeneic graft rejection. We herein present a "donor MHC-specific thymus vaccination" (DMTV) strategy to induce T cell tolerance to both autologous and allogeneic donor MHC. Allogeneic MHC molecules were expressed in the recipient thymus through adeno-associated virus-mediated delivery, which led to stable expression of allogeneic MHC together with the autologous MHC in the engineered thymus. During local T cell education, those T cells recognizing either autologous MHC or allogeneic MHC were equally depleted. We constructed C57BL/6-MHC and BALB/c-MHC dual immunocompatible mice via thymus vaccination of C57BL/6-MHC into the BALB/c thymus and observed long-term graft tolerance after transplantation of C57BL/6 skin and C57BL/6 mouse embryonic stem cells into the vaccinated BALB/c mice. We also validated our DMTV strategy in a bone marrow, liver, thymus (BLT)-humanized mouse model for immunocompatible allotransplantation of human embryonic stem cells. Our study suggests that the DMTV strategy is a potent avenue to introduce a donor compatible immune system in recipients, which overcomes the clinical dilemma of the extreme shortage of MHC-matched donor organs for treating patients with end-stage organ failure.
Collapse
Affiliation(s)
- Yang Liu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Hexi Feng
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Ke Li
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Ruiyi Li
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Xiao-Jie Zhang
- Department of Gynaecology, Jing'an District Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Ye Tian
- School of Foreign Studies, Tongji University, Shanghai, China
| | - Yujiang Fang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Yanjie Zhou
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Ling Liu
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China.
| | - Xiaoqing Zhang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.
- Stem Cell Research Center and Key Laboratory of Neuroregeneration of Shanghai Universities, School of Medicine, Tongji University, Shanghai, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai, China.
| |
Collapse
|
2
|
Nicosia M, Valujskikh A. Recognizing Complexity of CD8 T Cells in Transplantation. Transplantation 2024; 108:2186-2196. [PMID: 38637929 PMCID: PMC11489323 DOI: 10.1097/tp.0000000000005001] [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: 04/20/2024]
Abstract
The major role of CD8 + T cells in clinical and experimental transplantation is well documented and acknowledged. Nevertheless, the precise impact of CD8 + T cells on graft tissue injury is not completely understood, thus impeding the development of specific treatment strategies. The goal of this overview is to consider the biology and functions of CD8 + T cells in the context of experimental and clinical allotransplantation, with special emphasis on how this cell subset is affected by currently available and emerging therapies.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
3
|
Viklicky O, Zahradka I, Bold G, Bestard O, Hruba P, Otto NM, Stein M, Sefrin A, Modos I, Meneghini M, Crespo E, Grinyo J, Volk HD, Christakoudi S, Reinke P. Tacrolimus After rATG and Infliximab Induction Immunosuppression-RIMINI Trial. Transplantation 2024; 108:242-251. [PMID: 37525369 DOI: 10.1097/tp.0000000000004736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
BACKGROUND Infliximab selectively targets recently activated effector cells and, as an induction agent, might enable the safe elimination of mycophenolate from maintenance immunosuppression in kidney transplantation. METHODS This is a phase II international multicenter open-label single-arm confidence interval (CI)-based clinical trial of the BIO-DrIM EU consortium aimed at assessing the efficacy and safety of rabbit antithymocyte globulin and infliximab induction in kidney transplantation. Sixty-seven primary kidney transplant recipients at low risk (panel-reactive antibodies <20%, no donor-specific antibodies [DSA]) received rabbit antithymocyte globulin (2 × 1.5 mg/kg, postoperative days 0 and 1) and infliximab (5 mg/kg, postoperative day 2), followed by mycophenolate-free tacrolimus-based immunosuppression for 12 mo. The primary endpoint was efficacy failure, defined as a composite of acute rejection, graft loss, or poor graft function (estimated glomerular filtration rate <40 mL/min) at 12 mo and was based on the endpoint of the comparator study. Additionally, a historical propensity-matched control cohort was established. RESULTS Primary endpoint occurred in 22 of 67 patients (32.84%), with upper bound of an exact 1-sided 95% CI of 43.47%, which met the predefined criteria (efficacy failure of <40% and upper-bound 95% CI of <50%) and was similar in the historical matched cohort. By 12 mo, 79.1% of patients remained on the study protocol. Lower rates of BK replication (6% versus 22.4%; P = 0.013) but higher rates of de novo DSAs (11.9% versus 1.5%; P = 0.039) were observed in the study cohort. CONCLUSIONS A similar efficacy of the study immunosuppression regimen to the comparator study and the historical matched cohort was found. However, a higher de novo DSA emergence points to an increased risk of antibody-mediated rejection (NCT04114188).
Collapse
Affiliation(s)
- Ondrej Viklicky
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Zahradka
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Gantuja Bold
- Department of Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin Center for Advanced Therapies (BeCAT), Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| | - Oriol Bestard
- Department of Nephrology and Kidney Transplantation, Vall d'Hebron University Hospital, Barcelona Hospital Campus, Barcelona, Spain
- Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona, Spain
| | - Petra Hruba
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Natalie M Otto
- Department of Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin Center for Advanced Therapies (BeCAT), Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| | - Maik Stein
- Department of Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin Center for Advanced Therapies (BeCAT), Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| | - Anett Sefrin
- Department of Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin Center for Advanced Therapies (BeCAT), Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| | - Istvan Modos
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Maria Meneghini
- Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona, Spain
| | - Elena Crespo
- Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona, Spain
| | - Josep Grinyo
- Department of Medicine, Barcelona University, Barcelona, Spain
| | - Hans-Dieter Volk
- Berlin Center for Advanced Therapies (BeCAT) and Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| | - Sofia Christakoudi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St. Mary's Campus, Norfolk Place, London, United Kingdom
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Petra Reinke
- Department of Nephrology and Intensive Care, Charité Universitätsmedizin Berlin, Berlin Center for Advanced Therapies (BeCAT), Berlin Institut of Health Center of Regenerative Therapies (BCRT), Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
4
|
Cui X, Li CG, Gao H, Cheng M, Jiang F. Boosting regulatory T cell-dependent immune tolerance by activation of p53. Int Immunopharmacol 2023; 125:111167. [PMID: 37931392 DOI: 10.1016/j.intimp.2023.111167] [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: 08/20/2023] [Revised: 10/13/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Regulatory T cells (Tregs) have critical roles in maintaining immune hemostasis and have important anti-inflammatory functions in diseases. Recently, we identified that CX-5461 (a selective RNA polymerase I inhibitor and p53 activator) acted as a potent immunosuppressive agent, which prevented allogeneic acute rejection in animal models via a molecular mechanism distinct from all those of conventional immunosuppressive drugs. Unexpectedly, we discovered that CX-5461 could promote Treg differentiation. In this review, we have summarized the evidence for a potential role of p53 in mediating Treg differentiation and its possible mechanisms, including regulation of FoxP3 transcription, regulation of the expression of PTEN (phosphatase and tensin homolog), as well as protein-protein interaction with the transcription factor STAT5 (signal transducer and activator of transcription 5). Evidence also suggests that pharmacological p53 activators may potentially be used to boost Treg-mediated immune tolerance. Based on these data, we argue that novel p53 activators such as CX-5461 may represent a distinct class of immunosuppressants that repress conventional T cell-mediated alloimmunity with concomitant boosting of Treg-dependent immune tolerance.
Collapse
Affiliation(s)
- Xiaopei Cui
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Haiqing Gao
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Mei Cheng
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Fan Jiang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
5
|
Li S, Anwar IJ, Canning AJ, Vo-Dinh T, Kirk AD, Xu H. Xenorecognition and costimulation of porcine endothelium-derived extracellular vesicles in initiating human porcine-specific T cell immune responses. Am J Transplant 2023; 23:904-919. [PMID: 37054891 PMCID: PMC10330644 DOI: 10.1016/j.ajt.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
Porcine vascular endothelial cells (PECs) form a mechanistic centerpiece of xenograft rejection. Here, we determined that resting PECs release swine leukocyte antigen class I (SLA-I) but not swine leukocyte antigen class-II DR (SLA-DR) expressing extracellular vesicles (EVs) and investigated whether these EVs proficiently initiate xenoreactive T cell responses via direct xenorecognition and costimulation. Human T cells acquired SLA-I+ EVs with or without direct contact to PECs, and these EVs colocalized with T cell receptors. Although interferon gamma-activated PECs released SLA-DR+ EVs, the binding of SLA-DR+ EVs to T cells was sparse. Human T cells demonstrated low levels of proliferation without direct contact to PECs, but marked T cell proliferation was induced following exposure to EVs. EV-induced proliferation proceeded independent of monocytes/macrophages, suggesting that EVs delivered both a T cell receptor signal and costimulation. Costimulation blockade targeting B7, CD40L, or CD11a significantly reduced T cell proliferation to PEC-derived EVs. These findings indicate that endothelial-derived EVs can directly initiate T cell-mediated immune responses, and suggest that inhibiting the release of SLA-I EVs from organ xenografts has the potential to modify the xenograft rejection. We propose a secondary-direct pathway for T cell activation via xenoantigen recognition/costimulation by endothelial-derived EVs.
Collapse
Affiliation(s)
- Shu Li
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Imran J Anwar
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Aidan J Canning
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, North Carolina, USA
| | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA; Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - He Xu
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
6
|
Chen Y, Zhu Y, Kramer A, Fang Y, Wilson M, Li YR, Yang L. Genetic engineering strategies to enhance antitumor reactivity and reduce alloreactivity for allogeneic cell-based cancer therapy. Front Med (Lausanne) 2023; 10:1135468. [PMID: 37064017 PMCID: PMC10090359 DOI: 10.3389/fmed.2023.1135468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
The realm of cell-based immunotherapy holds untapped potential for the development of next-generation cancer treatment through genetic engineering of chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapies for targeted eradication of cancerous malignancies. Such allogeneic "off-the-shelf" cell products can be advantageously manufactured in large quantities, stored for extended periods, and easily distributed to treat an exponential number of cancer patients. At current, patient risk of graft-versus-host disease (GvHD) and host-versus-graft (HvG) allorejection severely restrict the development of allogeneic CAR-T cell products. To address these limitations, a variety of genetic engineering strategies have been implemented to enhance antitumor efficacy, reduce GvHD and HvG onset, and improve the overall safety profile of T-cell based immunotherapies. In this review, we summarize these genetic engineering strategies and discuss the challenges and prospects these approaches provide to expedite progression of translational and clinical studies for adoption of a universal cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Adam Kramer
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Matthew Wilson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
7
|
Pascual J, Jiménez C, Krajewska M, Seron D, Kotton CN, Portolés J, Witzke O, Sorensen SS, Andrés A, Crespo M, Paz-Artal E, Díez T, Ortega A, Portero I. The Immunobiogram, a novel in vitro diagnostic test to measure the pharmacodynamic response to immunosuppressive therapy in kidney transplant patients. Transpl Immunol 2022; 75:101711. [PMID: 36096417 DOI: 10.1016/j.trim.2022.101711] [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: 05/07/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diagnostic tools to measure the response to individual immunosuppressive drugs for transplant patients are currently lacking. We previously developed the blood-based Immunobiogram bioassay for in-vitro characterization of the pharmacodynamic response of patients' own immune cells to a range of immunosuppressants. We used Immunobiogram to examine the association between patients' sensitivity to their prescribed immunosuppressants and clinical outcome. METHODS We conducted an international, multicenter, observational study in a kidney transplant population undergoing maintenance immunosuppressive therapy. Patients were selected by clinical course poor [PCC] N = 53 (with renal dysfunction, and rejection signs in biopsy or/and an increase in DSA strength in last 12 months) versus good [GCC] N = 50 (with stable renal function and treatment, no rejection and no DSA titers). Immunobiogram dose-response curve parameters were compared between both subgroups in patients treated with mycophenolate, tacrolimus, corticosteroids, cyclosporine A or everolimus. Parameters for which significant inter-group differences were observed were further analyzed by univariate and subsequent multivariate logistic regression. RESULTS Clinical outcome was associated with following parameters: area over the curve (AOC) and 25% (ID25) and 50% (ID50) inhibitory response in mycophenolate, tacrolimus, and corticosteroid-treated subgroups, respectively. These statistically significant associations persisted in mycophenolate (OR 0.003, CI95% <0.001-0.258; p = 0.01) and tacrolimus (OR < 0.0001, CI95% <0.00001-0.202; p = 0.016) subgroups after adjusting for concomitant corticosteroid treatment, and in corticosteroid subgroup after adjusting for concomitant mycophenolate or tacrolimus treatment (OR 0.003; CI95% <0.0001-0.499; p = 0.026). CONCLUSIONS Our results highlight the potential of Immunobiogram as a tool to test the pharmacodynamic response to individual immunosuppressive drugs.
Collapse
Affiliation(s)
- Julio Pascual
- Nephrology Department, Hospital del Mar, Institute Mar for Medical Research, Barcelona, Spain; Nephrology Department, Hospital 12 de Octubre, Madrid, Spain.
| | | | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Daniel Seron
- Nephrology Department, Hospital Vall d'Hebron, Barcelona, Spain
| | - Camille N Kotton
- Transplant Infectious Diseases Division, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jose Portolés
- Nephrology Department, Hospital Puerta de Hierro, Madrid, Spain
| | - Oliver Witzke
- Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Soren S Sorensen
- Department of Nephrology, Rigshospitalet University Hospital Copenhagen, Denmark
| | - Amado Andrés
- Nephrology Department, Hospital 12 de Octubre, Madrid, Spain
| | - Marta Crespo
- Nephrology Department, Hospital del Mar, Institute Mar for Medical Research, Barcelona, Spain
| | | | - Teresa Díez
- Biohope Scientific Solutions for Human Health, Madrid, Spain
| | - Alvaro Ortega
- Biohope Scientific Solutions for Human Health, Madrid, Spain
| | - Isabel Portero
- Biohope Scientific Solutions for Human Health, Madrid, Spain
| |
Collapse
|
8
|
Crespo E, Vidal-Alabró A, Jouve T, Fontova P, Stein M, Mocka S, Meneghini M, Sefrin A, Hruba P, Gomà M, Torija A, Donadeu L, Favà A, Cruzado JM, Melilli E, Moreso F, Viklicky O, Bemelman F, Reinke P, Grinyó J, Lloberas N, Bestard O. Tacrolimus CYP3A Single-Nucleotide Polymorphisms and Preformed T- and B-Cell Alloimmune Memory Improve Current Pretransplant Rejection-Risk Stratification in Kidney Transplantation. Front Immunol 2022; 13:869554. [PMID: 35833145 PMCID: PMC9272702 DOI: 10.3389/fimmu.2022.869554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
Achieving fast immunosuppression blood exposure after kidney transplantation is key to abrogating both preformed and de novo anti-donor humoral and cellular alloresponses. However, while tacrolimus (TAC) is the cornerstone immunosuppressant inhibiting adaptive alloimmunity, its blood exposure is directly impacted by different single-nucleotide polymorphisms (SNPs) in CYP3A TAC-metabolizing enzymes. Here, we investigated how functional TAC-CYP3A genetic variants (CYP3A4*22/CYP3A5*3) influence the main baseline clinical and immunological risk factors of biopsy-proven acute rejection (BPAR) by means of preformed donor-specific antibodies (DSAs) and donor-specific alloreactive T cells (DSTs) in a large European cohort of 447 kidney transplants receiving TAC-based immunosuppression. A total of 70 (15.7%) patients developed BPAR. Preformed DSAs and DSTs were observed in 12 (2.7%) and 227 (50.8%) patients, respectively. According to the different CYP3A4*22 and CYP3A5*3 functional allele variants, we found 4 differential new clusters impacting fasting TAC exposure after transplantation; 7 (1.6%) were classified as high metabolizers 1 (HM1), 71 (15.9%) as HM2, 324 (72.5%) as intermediate (IM), and 45 (10.1%) as poor metabolizers (PM1). HM1/2 showed significantly lower TAC trough levels and higher dose requirements than IM and PM (p < 0.001) and more frequently showed TAC underexposure (<5 ng/ml). Multivariate Cox regression analyses revealed that CYP3A HM1 and IM pharmacogenetic phenotypes (hazard ratio (HR) 12.566, 95% CI 1.99–79.36, p = 0.007, and HR 4.532, 95% CI 1.10–18.60, p = 0.036, respectively), preformed DSTs (HR 3.482, 95% CI 1.99–6.08, p < 0.001), DSAs (HR 4.421, 95% CI 1.63–11.98, p = 0.003), and delayed graft function (DGF) (HR 2.023, 95% CI 1.22–3.36, p = 0.006) independently predicted BPAR. Notably, a significant interaction between T-cell depletion and TAC underexposure was observed, showing a reduction of the BPAR risk (HR 0.264, 95% CI 0.08–0.92, p = 0.037). Such variables except for DSAs displayed a higher predictive risk for the development of T cell-mediated rejection (TCMR). Refinement of pretransplant monitoring by incorporating TAC CYP3A SNPs with preformed DSAs as well as DSTs may improve current rejection-risk stratification and help induction treatment decision-making.
Collapse
Affiliation(s)
- Elena Crespo
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
- *Correspondence: Oriol Bestard, ; Elena Crespo,
| | - Anna Vidal-Alabró
- Experimental Nephrology and Transplantation Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Thomas Jouve
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
- Faculty of Health, Université Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, INSERM 1209, CNRS 5309, Grenoble, France
| | - Pere Fontova
- Experimental Nephrology and Transplantation Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Maik Stein
- Berlin Center for Advanced Therapies (BeCAT), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Sonila Mocka
- Experimental Nephrology and Transplantation Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Maria Meneghini
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
- Kidney Transplant Unit and Nephrology Department, Vall d’Hebron Hospital, Barcelona, Spain
| | - Anett Sefrin
- Berlin Center for Advanced Therapies (BeCAT), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Petra Hruba
- Department of Nephrology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czechia
| | - Montserrat Gomà
- Pathology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Alba Torija
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Laura Donadeu
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Alex Favà
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Josep M. Cruzado
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Edoardo Melilli
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain
| | - Francesc Moreso
- Kidney Transplant Unit and Nephrology Department, Vall d’Hebron Hospital, Barcelona, Spain
| | - Ondrej Viklicky
- Department of Nephrology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czechia
| | - Frederike Bemelman
- Renal Transplant Unit, Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center—University of Amsterdam, Amsterdam, Netherlands
| | - Petra Reinke
- Berlin Center for Advanced Therapies (BeCAT), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Josep Grinyó
- Department of Clinical Sciences, Barcelona University, Barcelona, Spain
| | - Nuria Lloberas
- Experimental Nephrology and Transplantation Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Oriol Bestard
- Nephrology and Transplant Laboratory, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
- Kidney Transplant Unit and Nephrology Department, Vall d’Hebron Hospital, Barcelona, Spain
- *Correspondence: Oriol Bestard, ; Elena Crespo,
| |
Collapse
|
9
|
Repression of T cell-mediated alloimmunity by CX-5461 via the p53-DUSP5 pathway. Pharmacol Res 2022; 177:106120. [DOI: 10.1016/j.phrs.2022.106120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 12/19/2022]
|
10
|
Mangare C, Tischer-Zimmermann S, Bonifacius A, Riese SB, Dragon AC, Blasczyk R, Maecker-Kolhoff B, Eiz-Vesper B. Variances in Antiviral Memory T-Cell Repertoire of CD45RA- and CD62L-Depleted Lymphocyte Products Reflect the Need of Individual T-Cell Selection Strategies to Reduce the Risk of GvHD while Preserving Antiviral Immunity in Adoptive T-Cell Therapy. Transfus Med Hemother 2022; 49:30-43. [PMID: 35221866 PMCID: PMC8832244 DOI: 10.1159/000516284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/01/2021] [Indexed: 04/03/2025] Open
Abstract
INTRODUCTION Viral infections and reactivations still remain a cause of morbidity and mortality after hematopoietic stem cell transplantation due to immunodeficiency and immunosuppression. Transfer of unmanipulated donor-derived lymphocytes (DLI) represents a promising strategy for improving cellular immunity but carries the risk of graft versus host disease (GvHD). Depleting alloreactive naïve T cells (TN) from DLIs was implemented to reduce the risk of GvHD induction while preserving antiviral memory T-cell activity. Here, we compared two TN depletion strategies via CD45RA and CD62L expression and investigated the presence of antiviral memory T cells against human adenovirus (AdV) and Epstein-Barr virus (EBV) in the depleted fractions in relation to their functional and immunophenotypic characteristics. METHODS T-cell responses against ppEBV_EBNA1, ppEBV_Consensus and ppAdV_Hexon within TN-depleted (CD45RA-/CD62L-) and TN-enriched (CD45RA+/CD62L+) fractions were quantified by interferon-gamma (IFN-γ) ELISpot assay after short- and long-term in vitro stimulation. T-cell frequencies and immunophenotypic composition were assessed in all fractions by flow cytometry. Moreover, alloimmune T-cell responses were evaluated by mixed lymphocyte reaction. RESULTS According to differences in the phenotype composition, antigen-specific T-cell responses in CD45RA- fraction were up to 2 times higher than those in the CD62L- fraction, with the highest increase (up to 4-fold) observed after 7 days for ppEBV_EBNA1-specific T cells. The CD4+ effector memory T cells (TEM) were mainly responsible for EBV_EBNA1- and AdV_Hexon-specific T-cell responses, whereas the main functionally active T cells against ppEBV_Consensus were CD8+ central memory T cells (TCM) and TEM. Moreover, comparison of both depletion strategies indicated that alloreactivity in CD45RA- was lower than that in CD62L- fraction. CONCLUSION Taken together, our results indicate that CD45RA depletion is a more suitable strategy for generating TN-depleted products consisting of memory T cells against ppEBV_EBNA1 and ppAdV_Hexon than CD62L in terms of depletion effectiveness, T-cell functionality and alloreactivity. To maximally exploit the beneficial effects mediated by antiviral memory T cells in TN-depleted products, depletion methods should be selected individually according to phenotype composition and CD4/CD8 antigen restriction. TN-depleted DLIs may improve the clinical outcome in terms of infections, GvHD, and disease relapse if selection of pathogen-specific donor T cells is not available.
Collapse
Affiliation(s)
- Caroline Mangare
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sabine Tischer-Zimmermann
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Agnes Bonifacius
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sebastian B. Riese
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna Christina Dragon
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Britta Maecker-Kolhoff
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Bestard O, Meneghini M, Crespo E, Bemelman F, Koch M, Volk HD, Viklicky O, Giral M, Banas B, Ruiz JC, Melilli E, Hu L, van Duivenvoorden R, Nashan B, Thaiss F, Otto NM, Bold G, Stein M, Sefrin A, Lachmann N, Hruba P, Stranavova L, Brouard S, Braudeau C, Blancho G, Banas M, Irure J, Christakoudi S, Sanchez-Fueyo A, Wood KJ, Reinke P, Grinyó JM. Preformed T cell alloimmunity and HLA eplet mismatch to guide immunosuppression minimization with tacrolimus monotherapy in kidney transplantation: Results of the CELLIMIN trial. Am J Transplant 2021; 21:2833-2845. [PMID: 33725408 DOI: 10.1111/ajt.16563] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/08/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
Personalizing immunosuppression is a major objective in transplantation. Transplant recipients are heterogeneous regarding their immunological memory and primary alloimmune susceptibility. This biomarker-guided trial investigated whether in low immunological-risk kidney transplants without pretransplant DSA and donor-specific T cells assessed by a standardized IFN-γ ELISPOT, low immunosuppression (LI) with tacrolimus monotherapy would be non-inferior regarding 6-month BPAR than tacrolimus-based standard of care (SOC). Due to low recruitment rates, the trial was terminated when 167 patients were enrolled. ELISPOT negatives (E-) were randomized to LI (n = 48) or SOC (n = 53), E+ received the same SOC. Six- and 12-month BPAR rates were higher among LI than SOC/E- (4/35 [13%] vs. 1/43 [2%], p = .15 and 12/48 [25%] vs. 6/53 [11.3%], p = .073, respectively). E+ patients showed similarly high BPAR rates than LI at 6 and 12 months (12/55 [22%] and 13/66 [20%], respectively). These differences were stronger in per-protocol analyses. Post-hoc analysis revealed that poor class-II eplet matching, especially DQ, discriminated E- patients, notably E-/LI, developing BPAR (4/28 [14%] low risk vs. 8/20 [40%] high risk, p = .043). Eplet mismatch also predicted anti-class-I (p = .05) and anti-DQ (p < .001) de novo DSA. Adverse events were similar, but E-/LI developed fewer viral infections, particularly polyoma-virus-associated nephropathy (p = .021). Preformed T cell alloreactivity and HLA eplet mismatch assessment may refine current baseline immune-risk stratification and guide immunosuppression decision-making in kidney transplantation.
Collapse
Affiliation(s)
- Oriol Bestard
- Kidney Transplant Unit, Nephrology department, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain.,Nephrology and Transplantation Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Maria Meneghini
- Kidney Transplant Unit, Nephrology department, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain.,Nephrology and Transplantation Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Elena Crespo
- Nephrology and Transplantation Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Frederike Bemelman
- Renal Transplant Unit, Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center - University of Amsterdam, Amsterdam, the Netherlands
| | - Martina Koch
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans D Volk
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Ondrej Viklicky
- Transplant Laboratory, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic.,Department of Nephrology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Magali Giral
- Nantes Université, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, ITUN, Nantes, France
| | - Bernhard Banas
- Department of Nephrology, University Medical Center Regensburg, Regensburg, Germany
| | - Juan C Ruiz
- Department of Nephrology, Hospital Universitario "Marqués de Valdecilla", Instituto de Investigación "Marqués de Valdecilla" (IDIVAL, Santander, Spain
| | - Edoardo Melilli
- Kidney Transplant Unit, Nephrology department, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain
| | - Liu Hu
- Renal Transplant Unit, Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center - University of Amsterdam, Amsterdam, the Netherlands
| | - Raphael van Duivenvoorden
- Renal Transplant Unit, Department of Internal Medicine, Amsterdam University Medical Centers, Academic Medical Center - University of Amsterdam, Amsterdam, the Netherlands
| | - Björn Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Thaiss
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie M Otto
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Gantuja Bold
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Maik Stein
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anett Sefrin
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Nils Lachmann
- HLA-Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Hruba
- Transplant Laboratory, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic.,Department of Nephrology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Lucia Stranavova
- Transplant Laboratory, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic.,Department of Nephrology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Sophie Brouard
- Nantes Université, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, ITUN, Nantes, France
| | - Cécile Braudeau
- Nantes Université, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, ITUN, Nantes, France.,CHU Nantes, Laboratoire d'immunologie, CIMNA, Nantes, France
| | - Gilles Blancho
- Nantes Université, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie UMR1064, ITUN, Nantes, France
| | - Miriam Banas
- Department of Nephrology, University Medical Center Regensburg, Regensburg, Germany
| | - Juan Irure
- Immunology Department, University Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Sophia Christakoudi
- Institute of Liver Studies, MRC Centre for Transplantation, Department of Inflammation Biology, Faculty of Sciences & Medicine, King's College London, London, UK
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, MRC Centre for Transplantation, Department of Inflammation Biology, Faculty of Sciences & Medicine, King's College London, London, UK
| | - Kathryn J Wood
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Petra Reinke
- BeCAT, BCRT, and Department of Nephrology & Intensive Care, Charité Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - Josep M Grinyó
- Kidney Transplant Unit, Nephrology department, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain.,Nephrology and Transplantation Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| |
Collapse
|
12
|
Romphruk AV, Simtong P, Suntornnipat J, Sudwilai Y, Cheunta S, Chan-On C, Leelayuwat C. Prevalence and impact of HLA and MICA allele mismatching on donor-specific antibodies induction in kidney transplant rejection. Nephrology (Carlton) 2021; 26:833-841. [PMID: 34197005 DOI: 10.1111/nep.13921] [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: 01/19/2021] [Revised: 06/07/2021] [Accepted: 06/25/2021] [Indexed: 11/26/2022]
Abstract
AIM Donor-recipient antigen mismatching for anti-human leucocyte antigen (HLA) and MICA is one of the risk factors for antibody induction leading to graft rejection. Our aim was to analyze the incidence and specificity of the different DSAs developing and to investigate the impact of HLA and MICA allele mismatches on antibody production in kidney transplant patients experiencing antibody-mediated rejection (AMR). METHODS We retrospectively reviewed 253 consecutive recipients of kidney transplant who were diagnosed as experiencing AMR. RESULTS Our results showed that around 27% of our patients were positive for DSAs over a median follow-up period of 24 months. Antibody to HLA-DQ7 was the most prevalent DSA detected. The allele mismatch number was significantly lower for DQ loci than -A and -B loci (DQ vs. A, p < .001; DQ vs. B, p = .002). Considering each HLA antigen, the incidence rate of DQ-DSA [41.9 (32.92-51.46; 95%CI)] was much higher than the rate observed for DSA directed to -A, -DR and -B loci. Half of the recipients in the DQ-DSA-only group, and the DQ-DSA together with non-DQ group, had MFI > 5000. Only one case developed de novo MICA-DSA (MICA002). CONCLUSION Our study indicates that mismatching for HLA and MICA alleles leads to the development of HLA and MICA antibodies in some kidney transplant recipients. We have also demonstrated that DSA to the DQ locus is the most prevalent in kidney transplant patients with AMR. Thus, matching the DQ locus in kidney allocation algorithms may reduce post-transplant development of DSA.
Collapse
Affiliation(s)
- Amornrat V Romphruk
- Blood Transfusion Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,The Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Piyapong Simtong
- The Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand.,Department of Clinical Immunology and Transfusion Sciences, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Jidpinan Suntornnipat
- Biomedical Sciences Program, Faculty of Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Yupaporn Sudwilai
- Blood Transfusion Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Siriluk Cheunta
- Blood Transfusion Center, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chitranon Chan-On
- Internal Medicine Department, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chanvit Leelayuwat
- The Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand.,Department of Clinical Immunology and Transfusion Sciences, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
13
|
Takahagi A, Shindo T, Chen-Yoshikawa TF, Yoshizawa A, Gochi F, Miyamoto E, Saito M, Tanaka S, Motoyama H, Aoyama A, Takaori-Kondo A, Date H. Trametinib Attenuates Delayed Rejection and Preserves Thymic Function in Rat Lung Transplantation. Am J Respir Cell Mol Biol 2020; 61:355-366. [PMID: 30849233 DOI: 10.1165/rcmb.2018-0188oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Delayed immunological rejection after human lung transplantation causes chronic lung allograft dysfunction, which is associated with high mortality. Delayed rejection may be attributable to indirect alloantigen presentation by host antigen-presenting cells; however, its pathophysiology is not fully understood. The mitogen-activated protein kinase pathway is activated in T cells upon stimulation, and we previously showed that the MEK inhibitor, trametinib, suppresses graft-versus-host disease after murine bone marrow transplantation. We investigated whether trametinib suppresses graft rejection after two types of rat lung transplantation and analyzed its immunological mode of action. Major histocompatibility complex-mismatched transplantation from brown Norway rats into Lewis rats and minor histocompatibility antigen-mismatched transplantation from Fischer 344 rats into Lewis rats were performed. Cyclosporine (CsA) and/or trametinib were administered alone or consecutively. Acute and delayed rejection, lymphocyte infiltration, and pulmonary function were evaluated. Administration of trametinib after CsA suppressed delayed rejection, reduced inflammatory cell infiltration and fibrosis within the graft, and preserved pulmonary functions at Day 28. Trametinib suppressed functional differentiation of T and B cells in the periphery but preserved thymic T cell differentiation. Donor B cells within the graft disappeared by Day 14, indicating that delayed graft rejection at Day 28 was mainly due to indirect presentation by host antigen-presenting cells. Finally, trametinib administration without CsA preconditioning suppressed rejection after minor histocompatibility antigen-mismatched transplantation. Trametinib attenuates delayed rejection upon major histocompatibility complex-mismatched transplantation by suppressing indirect presentation and is a promising candidate to treat chronic lung allograft dysfunction in humans.
Collapse
Affiliation(s)
| | - Takero Shindo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | | | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | | - Akifumi Takaori-Kondo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan; and
| | | |
Collapse
|
14
|
Pretransplant Donor-specific IFNγ ELISPOT as a Predictor of Graft Rejection: A Diagnostic Test Accuracy Meta-analysis. Transplant Direct 2019; 5:e451. [PMID: 31165086 PMCID: PMC6511445 DOI: 10.1097/txd.0000000000000886] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Supplemental Digital Content is available in the text. Background. Pretransplant interferon-γ enzyme-linked immunospot (IFN-γ ELISPOT) has been proposed as a tool to quantify alloreactive memory T cells and estimate the risk of acute rejection (AR) after kidney transplantation, but studies have been inconclusive so far. We performed a meta-analysis to evaluate the association between pretransplant IFN-γ ELISPOT and AR and assess its predictive accuracy at the individual level. Methods. We estimated the pooled summary of odds ratio for AR and the joined sensitivity and specificity for predicting AR using random-effects and hierarchical summary receiver-operating characteristic models. We used meta-regression models with the Monte Carlo permutation method to adjust for multiple tests to explain sensitivity and specificity heterogeneity across studies. The meta-analytic estimates of sensitivity and specificity were used to calculate positive and negative predictive values across studies. Results. The analysis included 12 studies and 1181 patients. IFN-γ ELISPOT was significantly associated with increased AR risk (odds ratio: 3.29; 95% confidence interval (CI), 2.34-4.60); hierarchical summary receiver operating characteristic jointly estimated sensitivity and specificity values were 64.9% (95% CI, 53.7%-74.6%) and 65.8% (95% CI, 57.4%-73.5%), respectively, with moderate heterogeneity across studies. After adjusting for multiple testing, meta-regression models showed that thymoglobulin induction, recipient black ethnicity, living versus deceased donors, and geographical location did not affect sensitivity or specificity. Because of the varying AR incidence of the studies, positive and negative predictive values ranged between 16%–60% and 70%–95%, respectively. Conclusions. Pretransplant IFN-γ ELISPOT is significantly associated with increased risk of AR but provides suboptimal predictive ability at an individual level. Prospective randomized clinical trials are warranted.
Collapse
|
15
|
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018; 183:128-138. [PMID: 30165256 DOI: 10.1016/j.biomaterials.2018.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Nancy C Kirkiles-Smith
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
16
|
Gandolfini I, Crespo E, Baweja M, Jarque M, Donadei C, Luque S, Montero N, Allesina A, Perin L, Maggiore U, Cravedi P, Bestard O. Impact of preformed T-cell alloreactivity by means of donor-specific and panel of reactive T cells (PRT) ELISPOT in kidney transplantation. PLoS One 2018; 13:e0200696. [PMID: 30059561 PMCID: PMC6066206 DOI: 10.1371/journal.pone.0200696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/02/2018] [Indexed: 01/09/2023] Open
Abstract
Donor-specific (d-sp) interferon gamma enzyme-linked immunosorbent spot (d-sp ELISPOT) and Panel of reactive T-cell (PRT) ELISPOT assays have been developed to detect alloreactive memory T (Tmem) cells in order to estimate the risk of acute rejection after kidney transplantation. Adding IL15 to the PRT assay (PRT+IL15) may uncover the presence of pathogenic alloreactive CD28-Tmem. Face-to-face comparisons of these assays have not been done yet. We performed pre-transplant d-sp ELISPOT and PRT assays (±IL15, against six B-cell lines) in 168 consecutive kidney transplant recipients and evaluated the multivariable-adjusted associations with biopsy-proven acute rejection (BPAR), de novo donor-specific antibodies (DSA), and eGFR decline over a 48-month follow-up period. D-sp ELISPOT was positive in 81 (48%) subjects, while 71 (42%) and 81 (48%) subjects displayed positive PRT and PRT+IL15, respectively. Their median [interquartile range] numerical test result was 23 [6–65], 18 [8–37], and 26 [10–45] spots/3x105 PBMCs, respectively. The number of PRT spots were weakly correlated with those of d-sp ELISPOT, but highly correlated with PRT+IL15 (rho = 0.96, P<0.001). d-sp ELISPOT, but not PRT (±IL15) was independently associated with BPAR (adjusted Odds Ratio of BPAR associated with d-sp ELISPOT positivity: 4.20 [95%CI: 1.06 to 21.73; P = 0.041]). Unlike d-sp ELISPOT, median PRT and PRT+IL15 were independently associated with higher Δ3-48month eGFR decline post-transplantation (for both assays, about -3mL/min/1.73m2 per one standard deviation unit increase in the spot number). Pre-transplant T-cell immune-monitoring using d-sp ELISPOT and PRT assays identifies kidney transplant candidates at high risk of BPAR and worse kidney allograft progression.
Collapse
Affiliation(s)
- Ilaria Gandolfini
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Kidney and Kidney-Pancreas Transplant Unit (Department of Nephrology), Parma University Hospital, Parma, Italy
| | - Elena Crespo
- Experimental Nephrology Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Mukta Baweja
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Marta Jarque
- Experimental Nephrology Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Chiara Donadei
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Sergio Luque
- Experimental Nephrology Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Núria Montero
- Kidney Transplant Unit, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain
| | - Anna Allesina
- Experimental Nephrology Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
| | - Laura Perin
- GOFARR Laboratory, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, United States of America
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Umberto Maggiore
- Kidney and Kidney-Pancreas Transplant Unit (Department of Nephrology), Parma University Hospital, Parma, Italy
| | - Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Oriol Bestard
- Experimental Nephrology Laboratory, IDIBELL, Barcelona University, Barcelona, Spain
- Kidney Transplant Unit, Bellvitge University Hospital, IDIBELL, Barcelona University, Barcelona, Spain
- * E-mail:
| |
Collapse
|
17
|
HLA mismatches that are identical for the antigen recognition domain are less immunogenic. Bone Marrow Transplant 2018; 53:729-740. [DOI: 10.1038/s41409-018-0108-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/18/2017] [Accepted: 01/04/2018] [Indexed: 11/08/2022]
|
18
|
Menon MC, Heeger PS. Donor SIRP-α polymorphisms: widening the innate-to-adaptive continuum in allograft rejection. Kidney Int 2017; 92:1305-1308. [DOI: 10.1016/j.kint.2017.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/10/2017] [Indexed: 11/27/2022]
|
19
|
Gates KV, Pereira NL, Griffiths LG. Cardiac Non-Human Leukocyte Antigen Identification: Techniques and Troubles. Front Immunol 2017; 8:1332. [PMID: 29093713 PMCID: PMC5651233 DOI: 10.3389/fimmu.2017.01332] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022] Open
Abstract
Historically efforts have focused on the human leukocyte antigen (HLA) as the major cause for acute and chronic rejection following cardiac transplantation. However, rising evidence indicates that non-HLA antibodies can be both primary initiators and modifiers of antibody-mediated rejection (AMR) and cardiac allograft vasculopathy (CAV). The purpose of this review is to assess currently available technologies for non-HLA identification and leveraging such responses toward antibody quantification. Several techniques have been used to identify antigenic determinants of recipient graft-specific non-HLA humoral immune responses, but each comes with its own set of benefits and caveats. Improving our ability to detect non-HLA humoral immune response will aid in our understanding of the underlying antigenic determinants of AMR and CAV, as well as improve patient outcomes.
Collapse
Affiliation(s)
- Katherine V Gates
- Department of Veterinary Medicine and Epidemiology, University of California, Davis, Davis, CA, United States.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Naveen L Pereira
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Leigh G Griffiths
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
20
|
Sukma Dewi I, Celik S, Karlsson A, Hollander Z, Lam K, McManus JW, Tebbutt S, Ng R, Keown P, McMaster R, McManus B, Öhman J, Gidlöf O. Exosomal miR-142-3p is increased during cardiac allograft rejection and augments vascular permeability through down-regulation of endothelial RAB11FIP2 expression. Cardiovasc Res 2017; 113:440-452. [PMID: 28073833 DOI: 10.1093/cvr/cvw244] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 12/01/2016] [Indexed: 01/24/2023] Open
Abstract
Aims Exosome-mediated microRNA transfer is a recently discovered mode of cell-to-cell communication, in which microRNAs act as paracrine molecules, exerting their regulatory effects in recipient cells. T cells and endothelial cells are two main players in the mechanism of acute cellular cardiac rejection. The aim of this study was to investigate the role of exosomal microRNAs in the crosstalk between T cells and endothelial cells and its implications for the molecular mechanisms that drive acute cellular rejection in heart transplantation. Methods and results Exosomes isolated from serum samples of heart transplant patients with and without acute cardiac allograft rejection were profiled and showed enrichment of miR-142-3p, miR-92a-3p, miR-339-3p and miR-21-5p. Treatment of endothelial cells with the respected serum exosomes resulted the increased of miR-142-3p level in endothelial cells. Using T cells isolated from healthy donors and activated with either anti-CD3/CD28 antibody or IL-2/PHA, we could show that miR-142-3p is released from activated cells, is contained in exosomes and can be transferred to human vascular endothelial cells in vitro. Transcriptome analysis of endothelial cells treated with activated T cell supernatant with or without exosomes was used to identify mRNA targets of transferred miR-142-3-p. Overexpression of miR-142-3p in endothelial cells resulted in a significant down-regulation of RAB11FIP2, and interaction of miR-142-3p with its predicted target site was confirmed with a reporter assay. Moreover, treatment of endothelial cells with serum exosomes from heart transplant patients with acute cellular rejection resulted in down-regulation of RAB11FIP2 expression and increase in vascular endothelial permeability. Conclusion We have identified a novel mechanism whereby miR-142-3p, a microRNA enriched in exosomes during acute cellular rejection, is transferred to endothelial cells and compromises endothelial barrier function via down-regulation of RAB11FIP2. This study sheds new light on the interaction between host immune system and cardiac allograft endothelium during acute cellular rejection.
Collapse
Affiliation(s)
- Ihdina Sukma Dewi
- Department of Cardiology, Clinical Sciences, Skåne University Hospital, Lund University, BMC D12, S?gatan 19, 221 84, Lund, Sweden
| | - Selvi Celik
- Department of Cardiology, Clinical Sciences, Skåne University Hospital, Lund University, BMC D12, S?gatan 19, 221 84, Lund, Sweden
| | - Anna Karlsson
- Department of Cardiology, Clinical Sciences, Skåne University Hospital, Lund University, BMC D12, S?gatan 19, 221 84, Lund, Sweden
| | - Zsuzsanna Hollander
- Prevention of Organ Failure (PROOF) Centre of Excellence, 1190 Hornby Street, Vancouver, British Columbia, V6Z 2K5. Canada.,UBC James Hogg Research Centre, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Karen Lam
- Prevention of Organ Failure (PROOF) Centre of Excellence, 1190 Hornby Street, Vancouver, British Columbia, V6Z 2K5. Canada
| | - Janet-Wilson McManus
- netCAD, Canadian Blood Services, 2150 Western Parkway, Vancouver, British Columbia V6T 1V6, Canada
| | - Scott Tebbutt
- Prevention of Organ Failure (PROOF) Centre of Excellence, 1190 Hornby Street, Vancouver, British Columbia, V6Z 2K5. Canada.,UBC James Hogg Research Centre, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.,Department of Medicine, University of British Columbia, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.,Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada
| | - Raymond Ng
- Prevention of Organ Failure (PROOF) Centre of Excellence, 1190 Hornby Street, Vancouver, British Columbia, V6Z 2K5. Canada.,Department of Computer Science, University of British Columbia, 2366 Main Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Paul Keown
- Vancouver General Hospital, 899 West 12th Avenue, Vancouver, British Columbia V5Z 1M9, Canada
| | - Robert McMaster
- Department of Transplantation and Immunology, Vancouver Coastal Health Research Institute, 910 West 10th Avenue, Vancouver, British Columbia V5Z 1M9, Canada
| | - Bruce McManus
- Prevention of Organ Failure (PROOF) Centre of Excellence, 1190 Hornby Street, Vancouver, British Columbia, V6Z 2K5. Canada.,UBC James Hogg Research Centre, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.,Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia V6T 2B5, Canada
| | - Jenny Öhman
- Department of Cardiology, Clinical Sciences, Skåne University Hospital, Lund University, BMC D12, S?gatan 19, 221 84, Lund, Sweden
| | - Olof Gidlöf
- Department of Cardiology, Clinical Sciences, Skåne University Hospital, Lund University, BMC D12, S?gatan 19, 221 84, Lund, Sweden
| |
Collapse
|
21
|
Jacquemont L, Soulillou JP, Degauque N. Blood biomarkers of kidney transplant rejection, an endless search? Expert Rev Mol Diagn 2017; 17:687-697. [PMID: 28571481 DOI: 10.1080/14737159.2017.1337512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The tailoring of immunosuppressive treatment is recognized as a promising strategy to improve long-term kidney graft outcome. To guide the standard care of transplant recipients, physicians need objective biomarkers that can identify an ongoing pathology with the graft or low intensity signals that will be later evolved to accelerated transplant rejection. The early identification of 'high-risk /low-risk' patients enables the adjustment of standard of caring, including managing the frequency of clinical visits and the immunosuppression dosing. Given their ease of availability and the compatibility with a large technical array, blood-based biomarkers have been widely scrutinized for use as potential predictive and diagnostic biomarkers. Areas covered: Here, the authors report on non-invasive biomarkers, such as modification of immune cell subsets and mRNA and miRNA profiles, identified in the blood of kidney transplant recipients collected before or after transplantation. Expert commentary: Combined with functional tests, the identification of biomarkers will improve our understanding of pathological processes and will contribute to a global improvement in clinical management.
Collapse
Affiliation(s)
- Lola Jacquemont
- a Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM , Université de Nantes , Nantes , France.,b Institut de Transplantation Urologie Néphrologie (ITUN) , CHU Nantes , Nantes , France
| | - Jean-Paul Soulillou
- a Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM , Université de Nantes , Nantes , France.,b Institut de Transplantation Urologie Néphrologie (ITUN) , CHU Nantes , Nantes , France
| | - Nicolas Degauque
- a Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM , Université de Nantes , Nantes , France.,b Institut de Transplantation Urologie Néphrologie (ITUN) , CHU Nantes , Nantes , France.,c LabEx IGO , "Immunotherapy, Graft, Oncology" , Nantes , France
| |
Collapse
|
22
|
Fischer M, Leyking S, Schäfer M, Elsäßer J, Janssen M, Mihm J, van Bentum K, Fliser D, Sester M, Sester U. Donor-specific alloreactive T cells can be quantified from whole blood, and may predict cellular rejection after renal transplantation. Eur J Immunol 2017; 47:1220-1231. [PMID: 28426152 DOI: 10.1002/eji.201646826] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/06/2017] [Accepted: 04/18/2017] [Indexed: 11/07/2022]
Abstract
Preformed cellular alloreactivity can exist prior to transplantation and may contribute to rejection. Here, we used a rapid flow-cytometric whole-blood assay to characterize the extent of alloreactive T cells among 1491 stimulatory reactions from 61 renal transplant candidates and 75 controls. The role of preformed donor-specific alloreactive T cells in cellular rejection was prospectively analyzed in 21 renal transplant recipients. Alloreactive CD8+ T cells were more frequent than respective CD4+ T cells, and these levels were stable over time. CD8+ T cells were effector-memory T cells largely negative for expression of CD27, CD62L, and CCR7, and were susceptible to steroid and calcineurin inhibitor inhibition. Alloreactivity was more frequent in samples with higher number of HLA mismatches. Moreover, the percentage of individuals with alloreactive T cells was higher in transplant candidates than in controls. Among transplant candidates, 5/61 exhibited alloreactive CD8+ T cells against most stimulators, 23/61 toward a limited number of stimulators, and 33/61 did not show any alloreactivity. Among 21 renal transplant recipients followed prospectively, one had donor-specific preformed T-cell alloreactivity. She was the only patient who developed cellular rejection posttransplantation. In conclusion, donor-specific alloreactive T cells may be rapidly quantified from whole blood, and may predict cellular rejection after transplantation.
Collapse
Affiliation(s)
- Michaela Fischer
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany
| | - Sarah Leyking
- Department of Internal Medicine IV, Saarland University, Homburg, Germany
| | - Marco Schäfer
- HLA-Laboratory, Stefan-Morsch-Stiftung, Birkenfeld, Germany
| | - Julia Elsäßer
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany
| | - Martin Janssen
- Department of Urology and Paediatric Urology, Saarland University, Homburg, Germany
| | - Janine Mihm
- Department of Internal Medicine IV, Saarland University, Homburg, Germany
| | - Kai van Bentum
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany.,Department of Internal Medicine IV, Saarland University, Homburg, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University, Homburg, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, Homburg, Germany
| | - Urban Sester
- Department of Internal Medicine IV, Saarland University, Homburg, Germany
| |
Collapse
|
23
|
The Immunogenicity of HLA Class II Mismatches: The Predicted Presentation of Nonself Allo-HLA-Derived Peptide by the HLA-DR Phenotype of the Recipient Is Associated with the Formation of DSA. J Immunol Res 2017; 2017:2748614. [PMID: 28331856 PMCID: PMC5346368 DOI: 10.1155/2017/2748614] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/17/2017] [Accepted: 02/02/2017] [Indexed: 12/02/2022] Open
Abstract
The identification of permissible HLA class II mismatches can prevent DSA in mismatched transplantation. The HLA-DR phenotype of recipients contributes to DSA formation by presenting allo-HLA-derived peptides to T-helper cells, which induces the differentiation of B cells into plasma cells. Comparing the binding affinity of self and nonself allo-HLA-derived peptides for recipients' HLA class II antigens may distinguish immunogenic HLA mismatches from nonimmunogenic ones. The binding affinities of allo-HLA-derived peptides to recipients' HLA-DR and HLA-DQ antigens were predicted using the NetMHCIIpan 3.1 server. HLA class II mismatches were classified based on whether they induced DSA and whether self or nonself peptide was predicted to bind with highest affinity to recipients' HLA-DR and HLA-DQ. Other mismatch characteristics (eplet, hydrophobic, electrostatic, and amino acid mismatch scores and PIRCHE-II) were evaluated. A significant association occurred between DSA formation and the predicted HLA-DR presentation of nonself peptides (P = 0.0169; accuracy = 80%; sensitivity = 88%; specificity = 63%). In contrast, mismatch characteristics did not differ significantly between mismatches that induced DSA and the ones that did not, except for PIRCHE-II (P = 0.0094). This methodology predicts DSA formation based on HLA mismatches and recipients' HLA-DR phenotype and may identify permissible HLA mismatches to help optimize HLA matching and guide donor selection.
Collapse
|
24
|
Esensten JH, Bluestone JA, Lim WA. Engineering Therapeutic T Cells: From Synthetic Biology to Clinical Trials. ANNUAL REVIEW OF PATHOLOGY 2017; 12:305-330. [PMID: 27959633 PMCID: PMC5557092 DOI: 10.1146/annurev-pathol-052016-100304] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Engineered T cells are currently in clinical trials to treat patients with cancer, solid organ transplants, and autoimmune diseases. However, the field is still in its infancy. The design, and manufacturing, of T cell therapies is not standardized and is performed mostly in academic settings by competing groups. Reliable methods to define dose and pharmacokinetics of T cell therapies need to be developed. As of mid-2016, there are no US Food and Drug Administration (FDA)-approved T cell therapeutics on the market, and FDA regulations are only slowly adapting to the new technologies. Further development of engineered T cell therapies requires advances in immunology, synthetic biology, manufacturing processes, and government regulation. In this review, we outline some of these challenges and discuss the contributions that pathologists can make to this emerging field.
Collapse
Affiliation(s)
- Jonathan H Esensten
- Department of Laboratory Medicine, University of California, San Francisco, California 94143;
| | - Jeffrey A Bluestone
- Diabetes Center and Department of Medicine, University of California, San Francisco, California 94143;
| | - Wendell A Lim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco 94158-2517;
| |
Collapse
|
25
|
Conversion to Once-Daily Tacrolimus Results in Increased p38MAPK Phosphorylation in T Lymphocytes of Kidney Transplant Recipients. Ther Drug Monit 2016; 38:280-4. [PMID: 26606072 DOI: 10.1097/ftd.0000000000000264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND The once-daily formulation of tacrolimus (TAC(OD)) has been developed to overcome adherence problems. Conversion from the twice-daily TAC (TAC(BID)) formulation to TAC(OD) on a 1:1 basis, however, often leads to a decrease of TAC predose concentrations, which averages ∼15%. Switching between the two TAC formulations may thus influence drug efficacy and necessitates therapeutic drug monitoring. As an additional tool in transplantation diagnostics, phospho-specific flow cytometry was used to study the biological effects of conversion on p38MAPK phosphorylation, a kinase involved in T-lymphocyte activation. METHODS Stable renal transplant recipients (n = 12), at least 1 year after their transplantation, were converted from TAC(BID) to TAC(OD) on 1:1 mg for mg base. Comedication consisted of mycophenolate mofetil (n = 10) and prednisolone (n = 3). TAC whole-blood predose concentrations were determined by immunoassay before and 3 months after conversion. P38MAPK phosphorylation was measured in T lymphocytes by whole-blood phospho-specific flow cytometry. RESULTS Three months after conversion, no significant decreases in TAC predose concentrations (C0) were found (P = 0.54), whereas p38MAPK phosphorylation increased with 11.4% (P < 0.05) in CD4 and with 15.6% (P < 0.05) in CD8 T lymphocytes. The TAC C0 during treatment with TAC(BID) correlated inversely with p38MAPK phosphorylation in T lymphocytes (rs = -0.638; P < 0.05). CONCLUSIONS These results suggest that the measurement of p38MAPK phosphorylation status in T lymphocytes is a sensitive method to determine the biological effects of TAC before and after conversion from TAC(BID) to TAC(OD). This method could be a more sensitive tool for therapeutic drug monitoring of TAC.
Collapse
|
26
|
Arciero JC, Maturo A, Arun A, Oh BC, Brandacher G, Raimondi G. Combining Theoretical and Experimental Techniques to Study Murine Heart Transplant Rejection. Front Immunol 2016; 7:448. [PMID: 27872621 PMCID: PMC5097940 DOI: 10.3389/fimmu.2016.00448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/10/2016] [Indexed: 12/21/2022] Open
Abstract
The quality of life of organ transplant recipients is compromised by complications associated with life-long immunosuppression, such as hypertension, diabetes, opportunistic infections, and cancer. Moreover, the absence of established tolerance to the transplanted tissues causes limited long-term graft survival rates. Thus, there is a great medical need to understand the complex immune system interactions that lead to transplant rejection so that novel and effective strategies of intervention that redirect the system toward transplant acceptance (while preserving overall immune competence) can be identified. This study implements a systems biology approach in which an experimentally based mathematical model is used to predict how alterations in the immune response influence the rejection of mouse heart transplants. Five stages of conventional mouse heart transplantation are modeled using a system of 13 ordinary differential equations that tracks populations of both innate and adaptive immunity as well as proxies for pro- and anti-inflammatory factors within the graft and a representative draining lymph node. The model correctly reproduces known experimental outcomes, such as indefinite survival of the graft in the absence of CD4+ T cells and quick rejection in the absence of CD8+ T cells. The model predicts that decreasing the translocation rate of effector cells from the lymph node to the graft delays transplant rejection. Increasing the starting number of quiescent regulatory T cells in the model yields a significant but somewhat limited protective effect on graft survival. Surprisingly, the model shows that a delayed appearance of alloreactive T cells has an impact on graft survival that does not correlate linearly with the time delay. This computational model represents one of the first comprehensive approaches toward simulating the many interacting components of the immune system. Despite some limitations, the model provides important suggestions of experimental investigations that could improve the understanding of rejection. Overall, the systems biology approach used here is a first step in predicting treatments and interventions that can induce transplant tolerance while preserving the capacity of the immune system to protect against legitimate pathogens.
Collapse
Affiliation(s)
- Julia C Arciero
- Department of Mathematical Sciences, Indiana University-Purdue University Indianapolis , Indianapolis, IN , USA
| | - Andrew Maturo
- Department of Mathematical Sciences, Indiana University-Purdue University Indianapolis , Indianapolis, IN , USA
| | - Anirudh Arun
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Byoung Chol Oh
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Gerald Brandacher
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| | - Giorgio Raimondi
- Vascularized and Composite Allotransplantation Laboratory, Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine , Baltimore, MD , USA
| |
Collapse
|
27
|
Abstract
The analyses of indirect T cell responses in patients with
antibody-mediated renal transplant injury by Shiu et al. emphasize the complex
contribution of B cells in alloimmunity. The data suggest at least three
distinct but potentially overlapping consequences of T/B cell interactions:
antigen presentation by B cells, alloantibody production, and immune regulation.
These multifaceted functions of B cells should be taken into consideration while
developing diagnostic tools and therapeutic strategies.
Collapse
|
28
|
Torikai H, Cooper LJ. Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors. Mol Ther 2016; 24:1178-86. [PMID: 27203439 DOI: 10.1038/mt.2016.106] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) endows specificity to T-cells independent of human leukocyte antigen (HLA). This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR(+)T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR(+)T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product from lymphopenic patients or patient's immune cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a biobank of allogeneic genetically modified immune cells from healthy third-party donors, which are cryopreserved and validated in advance of administration, will facilitate the centralizing manufacturing and widespread distribution of CAR(+)T-cells to multiple points-of-care in a timely manner. To achieve this, it is necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to toxicity and rejection. We review the strategies to establish "off-the-shelf" donor-derived biobanks for human application of CAR(+)T-cells as a drug.
Collapse
Affiliation(s)
- Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laurence Jn Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Ziopharm Oncology Inc., Boston, Massachusetts, USA
| |
Collapse
|
29
|
Merino D, San Segundo D, Medina JM, Rodrigo E, Asensio E, Irure J, Fernández-Fresnedo G, Arias MA, López-Hoyos M. Different in vitro proliferation and cytokine-production inhibition of memory T-cell subsets after calcineurin and mammalian target of rapamycin inhibitors treatment. Immunology 2016; 148:206-15. [PMID: 26931075 DOI: 10.1111/imm.12603] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/10/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022] Open
Abstract
Calcineurin inhibitors (CNI) and mammalian target of rapamycin inhibitors (mTORi) are the main immunosuppressants used for long-term maintenance therapy in transplant recipients to avoid acute rejection episodes. Both groups of immunosuppressants have wide effects and are focused against the T cells, although different impacts on specific T-cell subsets, such as regulatory T cells, have been demonstrated. A greater knowledge of the impact of immunosuppression on the cellular components involved in allograft rejection could facilitate decisions for individualized immunosuppression when an acute rejection event is suspected. Memory T cells have recently gained focus because they might induce a more potent response compared with naive cells. The impact of immunosuppressants on different memory T-cell subsets remains unclear. In the present study, we have studied the specific impact of CNI (tacrolimus) and mTORi (rapamycin and everolimus) over memory and naive CD4(+) T cells. To do so, we have analysed the proliferation, phenotypic changes and cytokine synthesis in vitro in the presence of these immunosuppressants. The present work shows a more potent effect of CNI on proliferation and cytokine production in naive and memory T cells. However, the mTORi permit the differentiation of naive T cells to the memory phenotype and allow the production of interleukin-2. Taken together, our data show evidence to support the combined use of CNI and mTORi in transplant immunosuppression.
Collapse
Affiliation(s)
- David Merino
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - David San Segundo
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan M Medina
- Autoimmunity and Transplant Group-IDIVAL, Santander, Spain
| | - Emilio Rodrigo
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Esther Asensio
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Juan Irure
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | | | - Manuel A Arias
- Nephrology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Immunology Service Hospital Universitario Marqués de Valdecilla-IDIVAL, Santander, Spain
| |
Collapse
|
30
|
Transforming growth factor-beta 1 delivery from microporous scaffolds decreases inflammation post-implant and enhances function of transplanted islets. Biomaterials 2015; 80:11-19. [PMID: 26701143 DOI: 10.1016/j.biomaterials.2015.11.065] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 01/15/2023]
Abstract
Biomaterial scaffolds are central to many regenerative strategies as they create a space for infiltration of host tissue and provide a platform to deliver growth factors and progenitor cells. However, biomaterial implantation results in an unavoidable inflammatory response, which can impair tissue regeneration and promote loss or dysfunction of transplanted cells. We investigated localized TGF-β1 delivery to modulate this immunological environment around scaffolds and transplanted cells. TGF-β1 was delivered from layered scaffolds, with protein entrapped within an inner layer and outer layers designed for cell seeding and host tissue integration. Scaffolds were implanted into the epididymal fat pad, a site frequently used for cell transplantation. Expression of cytokines TNF-α, IL-12, and MCP-1 were decreased by at least 40% for scaffolds releasing TGF-β1 relative to control scaffolds. This decrease in inflammatory cytokine production corresponded to a 60% decrease in leukocyte infiltration. Transplantation of islets into diabetic mice on TGF-β1 scaffolds significantly improved the ability of syngeneic islets to control blood glucose levels within the first week of transplant and delayed rejection of allogeneic islets. Together, these studies emphasize the ability of localized TGF-β1 delivery to modulate the immune response to biomaterial implants and enhance cell function in cell-based therapies.
Collapse
|
31
|
de Mare-Bredemeijer ELD, Shi XL, Mancham S, van Gent R, van der Heide-Mulder M, de Boer R, Heemskerk MHM, de Jonge J, van der Laan LJW, Metselaar HJ, Kwekkeboom J. Cytomegalovirus-Induced Expression of CD244 after Liver Transplantation Is Associated with CD8+ T Cell Hyporesponsiveness to Alloantigen. THE JOURNAL OF IMMUNOLOGY 2015; 195:1838-48. [DOI: 10.4049/jimmunol.1500440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/02/2015] [Indexed: 12/31/2022]
|
32
|
Robinson RH, Meissler JJ, Fan X, Yu D, Adler MW, Eisenstein TK. A CB2-Selective Cannabinoid Suppresses T-Cell Activities and Increases Tregs and IL-10. J Neuroimmune Pharmacol 2015; 10:318-32. [PMID: 25980325 PMCID: PMC4528965 DOI: 10.1007/s11481-015-9611-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/26/2015] [Indexed: 01/03/2023]
Abstract
We have previously shown that agonists selective for the cannabinoid receptor 2 (CB2), including O-1966, inhibit the Mixed Lymphocyte Reaction (MLR), an in vitro correlate of organ graft rejection, predominantly through effects on T-cells. Current studies explored the mechanism of this immunosuppression by O-1966 using mouse spleen cells. Treatment with O-1966 dose-relatedly decreased levels of the active nuclear forms of the transcription factors NF-κB and NFAT in wild-type T-cells, but not T-cells from CB2 knockout (CB2R k/o) mice. Additionally, a gene expression profile of purified T-cells from MLR cultures generated using a PCR T-cell activation array showed that O-1966 decreased mRNA expression of CD40 ligand and CyclinD3, and increased mRNA expression of Src-like-adaptor 2 (SLA2), Suppressor of Cytokine Signaling 5 (SOCS5), and IL-10. The increase in IL-10 was confirmed by measuring IL-10 protein levels in MLR culture supernatants. Further, an increase in the percentage of regulatory T-cells (Tregs) was observed in MLR cultures. Pretreatment with anti-IL-10 resulted in a partial reversal of the inhibition of proliferation and blocked the increase of Tregs. Additionally, O-1966 treatment caused a dose-related decrease in the expression of CD4 in MLR cultures from wild-type, but not CB2R k/o, mice. These data support the potential of CB2-selective agonists as useful therapeutic agents to prolong graft survival in transplant patients, and strengthens their potential as a new class of immunosuppressive agents with broader applicability.
Collapse
MESH Headings
- Animals
- Anisoles/pharmacology
- Cannabinoid Receptor Agonists/pharmacology
- Coculture Techniques
- Cyclohexanols
- Dose-Response Relationship, Drug
- Female
- Interleukin-10/biosynthesis
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/metabolism
- T-Lymphocytes/drug effects
- T-Lymphocytes/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Rebecca H. Robinson
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Joseph J. Meissler
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Xiaoxuan Fan
- Manager, Flow Cytometry Facility, Temple University School of Medicine, Philadelphia, PA 19140
| | - Daohai Yu
- Department of Clinical Sciences, Temple University School of Medicine, Philadelphia, PA 19140
| | - Martin W. Adler
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Toby K. Eisenstein
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140
| |
Collapse
|
33
|
Crespo E, Lucia M, Cruzado JM, Luque S, Melilli E, Manonelles A, Lloberas N, Torras J, Grinyó JM, Bestard O. Pre-transplant donor-specific T-cell alloreactivity is strongly associated with early acute cellular rejection in kidney transplant recipients not receiving T-cell depleting induction therapy. PLoS One 2015; 10:e0117618. [PMID: 25689405 PMCID: PMC4331510 DOI: 10.1371/journal.pone.0117618] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/29/2014] [Indexed: 01/08/2023] Open
Abstract
Preformed T-cell immune-sensitization should most likely impact allograft outcome during the initial period after kidney transplantation, since donor-specific memory T-cells may rapidly recognize alloantigens and activate the effector immune response, which leads to allograft rejection. However, the precise time-frame in which acute rejection is fundamentally triggered by preformed donor-specific memory T cells rather than by de novo activated naïve T cells is still to be established. Here, preformed donor-specific alloreactive T-cell responses were evaluated using the IFN-γ ELISPOT assay in a large consecutive cohort of kidney transplant patients (n = 90), to assess the main clinical variables associated with cellular sensitization and its predominant time-frame impact on allograft outcome, and was further validated in an independent new set of kidney transplant recipients (n = 67). We found that most highly T-cell sensitized patients were elderly patients with particularly poor HLA class-I matching, without any clinically recognizable sensitizing events. While one-year incidence of all types of biopsy-proven acute rejection did not differ between T-cell alloreactive and non-alloreactive patients, Receiver Operating Characteristic curve analysis indicated the first two months after transplantation as the highest risk time period for acute cellular rejection associated with baseline T-cell sensitization. This effect was particularly evident in young and highly alloreactive individuals that did not receive T-cell depletion immunosuppression. Multivariate analysis confirmed preformed T-cell sensitization as an independent predictor of early acute cellular rejection. In summary, monitoring anti-donor T-cell sensitization before transplantation may help to identify patients at increased risk of acute cellular rejection, particularly in the early phases after kidney transplantation, and thus guide decision-making regarding the use of induction therapy.
Collapse
Affiliation(s)
- Elena Crespo
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Marc Lucia
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Josep M. Cruzado
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
| | - Sergio Luque
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Edoardo Melilli
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
| | - Anna Manonelles
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
| | - Nuria Lloberas
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Joan Torras
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
| | - Josep M. Grinyó
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
| | - Oriol Bestard
- Experimental Nephrology Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Renal Transplant Unit, Nephrology department, Bellvitge University Hospital, Barcelona, Spain
- * E-mail:
| |
Collapse
|
34
|
Noninvasive allograft imaging of acute rejection: evaluation of (131)I-anti-CXCL10 mAb. Inflammation 2014; 38:456-64. [PMID: 25338944 DOI: 10.1007/s10753-014-0050-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The purpose of this study was to investigate the use of iodine-131-labeled anti-CXCL10 mAb as tracer targeted at CXCL10 to detect acute rejection (AR) with mice model. Expression of CXCL10 was proved by RT-PCR, ELISA, and immunochemistry staining. All groups were submitted to whole-body autoradioimaging and ex vivo biodistribution studies after tail vein injection of (131)I-anti-CXCL10 mAb. The highest concentration/expression of CXCL10 was detected in allograft tissue compared with allograft treated with tacrolimus and isograft control. Tacrolimus could obviously inhibit the rejection of allograft. Allograft could be obviously imaged at all checking points, much clearer than the other two groups. The biodistribution results showed the highest uptake of radiotracer in allograft. T/NT (target/nontarget) ratio was 4.15 ± 0.25 at 72 h, apparently different from allograft treated with tacrolimus (2.29 ± 0.10), P < 0.05. These data suggest that CXCL10 is a promising target for early stage AR imaging and (131)I-CXCL10 mAb can successfully image AR and monitor the effect of immunosuppressant.
Collapse
|
35
|
Ishii D, Rosenblum JM, Nozaki T, Schenk AD, Setoguchi K, Su CA, Gorbacheva V, Baldwin WM, Valujskikh A, Fairchild RL. Novel CD8 T cell alloreactivities in CCR5-deficient recipients of class II MHC disparate kidney grafts. THE JOURNAL OF IMMUNOLOGY 2014; 193:3816-24. [PMID: 25172484 DOI: 10.4049/jimmunol.1303256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recipient CD4 T regulatory cells inhibit the acute T cell-mediated rejection of renal allografts in wild-type mice. The survival of single class II MHC-disparate H-2(bm12) renal allografts was tested in B6.CCR5(-/-) recipients, which have defects in T regulatory cell activities that constrain alloimmune responses. In contrast to wild-type C57BL/6 recipients, B6.CCR5(-/-) recipients rejected the bm12 renal allografts. However, donor-reactive CD8 T cells rather than CD4 T cells were the primary effector T cells mediating rejection. The CD8 T cells induced to bm12 allografts in CCR5-deficient recipients were reactive to peptides spanning the 3 aa difference in the I-A(bm12) versus I-A(b) β-chains presented by K(b) and D(b) class I MHC molecules. Allograft-primed CD8 T cells from CCR5-deficient allograft recipients were activated during culture either with proinflammatory cytokine-stimulated wild-type endothelial cells pulsed with the I-A(bm12) peptides or with proinflammatory cytokine-simulated bm12 endothelial cells, indicating their presentation of the I-A(bm12) β-chain peptide/class I MHC complexes. In addition to induction by bm12 renal allografts, the I-A(bm12) β-chain-reactive CD8 T cells were induced in CCR5-deficient, but not wild-type C57BL/6, mice by immunization with the peptides. These results reveal novel alloreactive CD8 T cell specificities in CCR5-deficient recipients of single class II MHC renal allografts that mediate rejection of the allografts.
Collapse
Affiliation(s)
- Daisuke Ishii
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Urology, Kitasato University, Kanagawa 228-8555, Japan
| | - Joshua M Rosenblum
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - Taiji Nozaki
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Austin D Schenk
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | - Kiyoshi Setoguchi
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Charles A Su
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and
| | | | - William M Baldwin
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| | - Anna Valujskikh
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and Glickman Urological Institute and the Transplant Center, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
36
|
Lorenz K, Rommel K, Mani J, Jin N, Hilgendorf I, Ho AD, Freund M, Schmitt M, Schmitt A. Modulation of lymphocyte subpopulations by extracorporeal photopheresis in patients with acute graft-versus-host disease or graft rejection. Leuk Lymphoma 2014; 56:671-5. [PMID: 24913503 DOI: 10.3109/10428194.2014.931956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Extracorporeal photopheresis (ECP) constitutes a promising treatment for patients with steroid-refractory acute graft-versus-host disease (aGvHD) after allogeneic stem cell transplantation and for patients with graft rejection after solid organ transplantation (SOT). There is an increasing body of evidence that modulation of lymphocyte subsets might play a crucial role in the mechanism of action in ECP. We therefore analyzed immunological effects concomitantly with clinical findings in patients under ECP therapy using multicolor flow cytometry. In a patient with steroid-refractory aGvHD and a patient with progressive bronchiolitis obliterans syndrome (BOS) after double-lung transplantation, clinical responses to ECP therapy were paralleled by an increase of CD4 + CD25hiFoxP3 + regulatory T cells and a decrease of T(EMRA) (CD3 + CD8+ CD45RA+ CD62L+ effector memory T) cells as well as of natural killer (NK)T cells. In summary, immunomonitoring of T cell subsets can elucidate the mechanism of action in ECP.
Collapse
Affiliation(s)
- Katrin Lorenz
- Department of Internal Medicine V, University Clinic Heidelberg , Heidelberg , Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tagliamacco A, Cioni M, Comoli P, Ramondetta M, Brambilla C, Trivelli A, Magnasco A, Biticchi R, Fontana I, Dulbecco P, Palombo D, Klersy C, Ghiggeri GM, Ginevri F, Cardillo M, Nocera A. DQ molecules are the principal stimulators ofde novodonor-specific antibodies in nonsensitized pediatric recipients receiving a first kidney transplant. Transpl Int 2014; 27:667-73. [DOI: 10.1111/tri.12316] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/13/2014] [Accepted: 03/11/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Augusto Tagliamacco
- Transplant Immunology Research Laboratory at Clinical Nephrology and Gastroenterology Units; Department of Internal Medicine -DIMI; University of Genova; Genova Italy
| | - Michela Cioni
- Nephrology, Dialysis and Transplantation Unit; G. Gaslini Institute; Genova Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology; Fondazione IRCCS Policlinico S. Matteo; Pavia Italy
| | - Miriam Ramondetta
- Transplantation Immunology; Fondazione Ca' Granda, Ospedale Maggiore Policlinico; Milano Italy
| | - Caterina Brambilla
- Transplantation Immunology; Fondazione Ca' Granda, Ospedale Maggiore Policlinico; Milano Italy
| | - Antonella Trivelli
- Nephrology, Dialysis and Transplantation Unit; G. Gaslini Institute; Genova Italy
| | - Alberto Magnasco
- Nephrology, Dialysis and Transplantation Unit; G. Gaslini Institute; Genova Italy
| | - Roberta Biticchi
- Transplant Immunology Research Laboratory at Clinical Nephrology and Gastroenterology Units; Department of Internal Medicine -DIMI; University of Genova; Genova Italy
| | - Iris Fontana
- Vascular and Endovascular Unit and Kidney Transplant Surgery Unit; University of Genova; IRCCS San Martino University Hospital IST; Genova Italy
| | - Pietro Dulbecco
- Gastroenterology Unit, DIMI; University of Genova; Genova Italy
| | - Domenico Palombo
- Vascular and Endovascular Unit and Kidney Transplant Surgery Unit; University of Genova; IRCCS San Martino University Hospital IST; Genova Italy
| | - Catherine Klersy
- Biometry and Statistics Service; Fondazione IRCCS Policlinico S. Matteo; Pavia Italy
| | - Gian Marco Ghiggeri
- Nephrology, Dialysis and Transplantation Unit; G. Gaslini Institute; Genova Italy
| | - Fabrizio Ginevri
- Nephrology, Dialysis and Transplantation Unit; G. Gaslini Institute; Genova Italy
| | - Massimo Cardillo
- Transplantation Immunology; Fondazione Ca' Granda, Ospedale Maggiore Policlinico; Milano Italy
| | - Arcangelo Nocera
- Transplant Immunology Research Laboratory at Clinical Nephrology and Gastroenterology Units; Department of Internal Medicine -DIMI; University of Genova; Genova Italy
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Cell therapy with mesenchymal stromal cells (MSC) has emerged as a promising tolerance-inducing strategy, as MSC are potent modifiers of immune cells within adaptive as well as innate arm of the immune system. Here, we review recent evidence on both the beneficial and deleterious effect of MSC in experimental models of solid organ transplantation as well as first clinical experiences of MSC therapy in kidney transplant recipients. RECENT FINDINGS MSC are able to reprogram macrophages toward an anti-inflammatory phenotype capable to regulate antigraft immune response. This interaction is mediated mainly by TNF-α-induced-protein-6. Conversely, MSC also take on a proinflammatory phenotype and actually could worsen graft outcome. MSC in clinical transplantation is in its infancy and nobody so far has attempted to or provided evidence that this cell-based therapy is capable to promote operational tolerance. There are, however, supporting data of the ex-vivo immunoregulatory activity of MSC in treated patients. SUMMARY MSC have a great potential as a tolerance-promoting cell therapy. Extensive investigations are still needed to dissect the mechanism(s) of action of MSC, particularly in the setting of a proinflammatory environment, and to establish specific assays for monitoring MSC-treated patients to define the protolerogenic potential of MSC-based therapy in kidney transplantation.
Collapse
|
39
|
Rambal V, Müller K, Dang-Heine C, Sattler A, Dziubianau M, Weist B, Luu SH, Stoyanova A, Nickel P, Thiel A, Neumann A, Schweiger B, Reinke P, Babel N. Differential influenza H1N1-specific humoral and cellular response kinetics in kidney transplant patients. Med Microbiol Immunol 2014; 203:35-45. [PMID: 24057515 DOI: 10.1007/s00430-013-0312-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 09/03/2013] [Indexed: 12/17/2022]
Abstract
Renal transplant recipients (RTR) are considered at high risk for influenza-associated complications due to immunosuppression. The efficacy of standard influenza vaccination in RTRs is unclear. Hence, we evaluated activation of the adaptive immunity by the pandemic influenza A(H1N1) 2009 (A(H1N1)pdm09) vaccine in RTRs as compared to healthy controls. To determine cross-reactivity and/or bystander activation, seasonal trivalent influenza vaccine and tetanus/diphteria toxoid (TT/DT) vaccine-specific T cells along with allospecific T cells were quantified before and after A(H1N1)pdm09 vaccination. Vaccination-induced alloimmunity was additionally determined by quantifying serum creatinine and proinflammatory protein IP-10. Contrary to healthy controls, RTRs required a booster vaccination to achieve seroconversion (13.3 % day 21; 90 % day 90). In contrast to humoral immunity, sufficient A(H1N1)pdm09-specific T-cell responses were mounted in RTRs already after the first immunization with a magnitude comparable with healthy controls. Interestingly, vaccination simultaneously boosted T cells reacting to seasonal flu but not to TT/DT, suggesting cross-activation. No alloimmune effects were recorded. In conclusion, protective antibody responses required booster vaccination. However, sufficient cellular immunity is established already after the first vaccination, demonstrating differential kinetics of humoral and cellular immunity.
Collapse
Affiliation(s)
- Vinay Rambal
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
van Gent R, Jaadar H, Tjon ASW, Mancham S, Kwekkeboom J. T-cell inhibitory capacity of hyperimmunoglobulins is influenced by the production process. Int Immunopharmacol 2014; 19:142-4. [PMID: 24462495 DOI: 10.1016/j.intimp.2014.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/11/2013] [Accepted: 01/09/2014] [Indexed: 01/11/2023]
Abstract
Intravenous immunoglobulin (IVIg) preparations are widely used for anti-inflammatory therapy of autoimmune and systemic inflammatory diseases. Hyperimmunoglobulins enriched in neutralizing antibodies against viruses can, in addition to their virus-neutralizing activity, also exert immunomodulatory activity. Previously, we observed that Cytotect®, an anti-CMV hyperimmunoglobulin, was less effective in suppressing human T-cell responses in vitro compared to Hepatect® CP, an anti-HBV hyperimmunoglobulin. We hypothesized that the poor immunomodulatory activity of Cytotect® results from treatment with β-propiolactone during the manufacturing process. The manufacturer of these hyperimmunoglobulins has now introduced a new anti-CMV hyperimmunoglobulin, called Cytotect® CP, in which β-propiolactone treatment is omitted. Here we show that Cytotect® CP inhibits PHA-driven T-cell proliferation and cytokine production with similar efficacy as Hepatect® CP, whereas the former Cytotect® does not. In addition, Cytotect® CP inhibits allogeneic T-cell responses better than Cytotect®. Our results advocate the use of hyperimmunoglobulins that have not been exposed to β-propiolactone in order to benefit from their immunomodulatory properties.
Collapse
Affiliation(s)
- Rogier van Gent
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Haziz Jaadar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Angela S W Tjon
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Shanta Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
|
42
|
Robinson RH, Meissler JJ, Breslow-Deckman JM, Gaughan J, Adler MW, Eisenstein TK. Cannabinoids inhibit T-cells via cannabinoid receptor 2 in an in vitro assay for graft rejection, the mixed lymphocyte reaction. J Neuroimmune Pharmacol 2013; 8:1239-50. [PMID: 23824763 PMCID: PMC3864984 DOI: 10.1007/s11481-013-9485-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 06/05/2013] [Indexed: 10/26/2022]
Abstract
Cannabinoids are known to have anti-inflammatory and immunomodulatory properties. Cannabinoid receptor 2 (CB2) is expressed mainly on leukocytes and is the receptor implicated in mediating many of the effects of cannabinoids on immune processes. This study tested the capacity of Δ(9)-tetrahydrocannabinol (Δ(9)-THC) and of two CB2-selective agonists to inhibit the murine Mixed Lymphocyte Reaction (MLR), an in vitro correlate of graft rejection following skin and organ transplantation. Both CB2-selective agonists and Δ(9)-THC significantly suppressed the MLR in a dose dependent fashion. The inhibition was via CB2, as suppression could be blocked by pretreatment with a CB2-selective antagonist, but not by a CB1 antagonist, and none of the compounds suppressed the MLR when splenocytes from CB2 deficient mice were used. The CB2 agonists were shown to act directly on T-cells, as exposure of CD3(+) cells to these compounds completely inhibited their action in a reconstituted MLR. Further, the CB2-selective agonists completely inhibited proliferation of purified T-cells activated by anti-CD3 and anti-CD28 antibodies. T-cell function was decreased by the CB2 agonists, as an ELISA of MLR culture supernatants revealed IL-2 release was significantly decreased in the cannabinoid treated cells. Together, these data support the potential of this class of compounds as useful therapies to prolong graft survival in transplant patients.
Collapse
Affiliation(s)
- Rebecca Hartzell Robinson
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | | | | | | | | | | |
Collapse
|
43
|
Angaswamy N, Tiriveedhi V, Sarma NJ, Subramanian V, Klein C, Wellen J, Shenoy S, Chapman WC, Mohanakumar T. Interplay between immune responses to HLA and non-HLA self-antigens in allograft rejection. Hum Immunol 2013; 74:1478-85. [PMID: 23876679 DOI: 10.1016/j.humimm.2013.07.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 07/09/2013] [Accepted: 07/09/2013] [Indexed: 01/02/2023]
Abstract
Recent studies strongly suggest an increasing role for immune responses against self-antigens (Ags) which are not encoded by the major histocompatibility complex in the immunopathogenesis of allograft rejection. Although, improved surgical techniques coupled with improved methods to detect and avoid sensitization against donor human leukocyte antigen (HLA) have improved the immediate and short term function of transplanted organs. However, acute and chronic rejection still remains a vexing problem for the long term function of the transplanted organ. Immediately following organ transplantation, several factors both immune and non immune mechanisms lead to the development of local inflammatory milieu which sets the stage for allograft rejection. Traditionally, development of antibodies (Abs) against mismatched donor HLA have been implicated in the development of Ab mediated rejection. However, recent studies from our laboratory and others have demonstrated that development of humoral and cellular immune responses against non-HLA self-Ags may contribute in the pathogenesis of allograft rejection. There are reports demonstrating that immune responses to self-Ags especially Abs to the self-Ags as well as cellular immune responses especially through IL17 has significant pro-fibrotic properties leading to chronic allograft failure. This review summarizes recent studies demonstrating the role for immune responses to self-Ags in allograft immunity leading to rejection as well as present recent evidence suggesting there is interplay between allo- and autoimmunity leading to allograft dysfunction.
Collapse
Affiliation(s)
- Nataraju Angaswamy
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bestard O, Crespo E, Stein M, Lúcia M, Roelen DL, de Vaal YJ, Hernandez-Fuentes MP, Chatenoud L, Wood KJ, Claas FH, Cruzado JM, Grinyó JM, Volk HD, Reinke P. Cross-validation of IFN-γ Elispot assay for measuring alloreactive memory/effector T cell responses in renal transplant recipients. Am J Transplant 2013; 13:1880-90. [PMID: 23763435 DOI: 10.1111/ajt.12285] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/12/2013] [Accepted: 03/25/2013] [Indexed: 01/25/2023]
Abstract
Assessment of donor-specific alloreactive memory/effector T cell responses using an IFN-γ Elispot assay has been suggested to be a novel immune-monitoring tool for evaluating the cellular immune risk in renal transplantation. Here, we report the cross-validation data of the IFN-γ Elispot assay performed within different European laboratories taking part of the EU RISET consortium. For this purpose, development of a standard operating procedure (SOP), comparisons of lectures of IFN-γ plates assessing intra- and interlaboratory assay variability of allogeneic or peptide stimuli in both healthy and kidney transplant individuals have been the main objectives. We show that the use of a same SOP and count-settings of the Elispot bioreader allow low coefficient variation between laboratories. Frozen and shipped samples display slightly lower detectable IFN-γ frequencies than fresh samples. Importantly, a close correlation between different laboratories is obtained when measuring high frequencies of antigen-specific primed/memory T cell alloresponses. Interestingly, significant high donor-specific alloreactive T cell responses can be similarly detected among different laboratories in kidney transplant patients displaying histological patterns of acute T cell mediated rejection. In conclusion, assessment of circulating alloreactive memory/effector T cells using an INF-γ Elispot assay can be accurately achieved using the same SOP, Elispot bioreader and experienced technicians in kidney transplantation.
Collapse
Affiliation(s)
- O Bestard
- Nephrology Department, Renal Transplant Unit, Bellvitge University Hospital, Barcelona University, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bestard O, Cruzado JM, Lucia M, Crespo E, Casis L, Sawitzki B, Vogt K, Cantarell C, Torras J, Melilli E, Mast R, Martinez-Castelao A, Gomà M, Reinke P, Volk HD, Grinyó JM. Prospective assessment of antidonor cellular alloreactivity is a tool for guidance of immunosuppression in kidney transplantation. Kidney Int 2013; 84:1226-36. [PMID: 23783240 DOI: 10.1038/ki.2013.236] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 04/21/2013] [Accepted: 04/25/2013] [Indexed: 01/08/2023]
Abstract
Current characterization of the immune risk in renal transplant patients is only focused on the assessment of preformed circulating alloantibodies; however, alloreactive memory T cells are key players in mediating allograft rejection. Immune monitoring of antidonor alloreactive memory/effector T cells using an IFN-γ Elispot has been shown to distinguish patients at risk for immune-mediated graft dysfunction, suggesting a potential tool for immunosuppression individualization. In this nonrandomized study, we prospectively assessed donor and nondonor T-cell alloreactivity in 60 highly alloreactive patients receiving calcineurin inhibitor-based immunosuppression and in non-T-cell alloreactive transplant recipients treated with a calcineurin inhibitor-free regimen. The impact was evaluated using 1-year allograft outcome. We found a strong association between ongoing antidonor T-cell alloreactivity and histological lesions of acute T cell-mediated rejection in 6-month protocol biopsies, distinguishing those patients with better 1-year graft function, regardless of immunosuppression regimen. Interestingly, evidence for enhanced immune regulation, driven by circulating Foxp3-demethylated regulatory T cells, was only observed among patients achieving antidonor T-cell hyporesponsiveness. Thus, prospective evaluation of donor-specific T-cell sensitization may add crucial information on the alloimmune state of transplanted patients to be used in daily clinical practice.
Collapse
Affiliation(s)
- Oriol Bestard
- 1] Renal Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain [2] Experimental Nephrology Department, IDIBELL, Barcelona University, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cohen Freue GV, Meredith A, Smith D, Bergman A, Sasaki M, Lam KKY, Hollander Z, Opushneva N, Takhar M, Lin D, Wilson-McManus J, Balshaw R, Keown PA, Borchers CH, McManus B, Ng RT, McMaster WR. Computational biomarker pipeline from discovery to clinical implementation: plasma proteomic biomarkers for cardiac transplantation. PLoS Comput Biol 2013; 9:e1002963. [PMID: 23592955 PMCID: PMC3617196 DOI: 10.1371/journal.pcbi.1002963] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 01/16/2013] [Indexed: 11/19/2022] Open
Abstract
Recent technical advances in the field of quantitative proteomics have stimulated a large number of biomarker discovery studies of various diseases, providing avenues for new treatments and diagnostics. However, inherent challenges have limited the successful translation of candidate biomarkers into clinical use, thus highlighting the need for a robust analytical methodology to transition from biomarker discovery to clinical implementation. We have developed an end-to-end computational proteomic pipeline for biomarkers studies. At the discovery stage, the pipeline emphasizes different aspects of experimental design, appropriate statistical methodologies, and quality assessment of results. At the validation stage, the pipeline focuses on the migration of the results to a platform appropriate for external validation, and the development of a classifier score based on corroborated protein biomarkers. At the last stage towards clinical implementation, the main aims are to develop and validate an assay suitable for clinical deployment, and to calibrate the biomarker classifier using the developed assay. The proposed pipeline was applied to a biomarker study in cardiac transplantation aimed at developing a minimally invasive clinical test to monitor acute rejection. Starting with an untargeted screening of the human plasma proteome, five candidate biomarker proteins were identified. Rejection-regulated proteins reflect cellular and humoral immune responses, acute phase inflammatory pathways, and lipid metabolism biological processes. A multiplex multiple reaction monitoring mass-spectrometry (MRM-MS) assay was developed for the five candidate biomarkers and validated by enzyme-linked immune-sorbent (ELISA) and immunonephelometric assays (INA). A classifier score based on corroborated proteins demonstrated that the developed MRM-MS assay provides an appropriate methodology for an external validation, which is still in progress. Plasma proteomic biomarkers of acute cardiac rejection may offer a relevant post-transplant monitoring tool to effectively guide clinical care. The proposed computational pipeline is highly applicable to a wide range of biomarker proteomic studies.
Collapse
Affiliation(s)
- Gabriela V. Cohen Freue
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Meredith
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Derek Smith
- University of Victoria Genome BC Proteomics Centre, Victoria, British Columbia, Canada
| | - Axel Bergman
- Immunity and Infection Research Centre, Vancouver, British Columbia, Canada
| | - Mayu Sasaki
- Immunity and Infection Research Centre, Vancouver, British Columbia, Canada
| | - Karen K. Y. Lam
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zsuzsanna Hollander
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nina Opushneva
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
| | - Mandeep Takhar
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
| | - David Lin
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Janet Wilson-McManus
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert Balshaw
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul A. Keown
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Immunology Laboratory, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Christoph H. Borchers
- University of Victoria Genome BC Proteomics Centre, Victoria, British Columbia, Canada
| | - Bruce McManus
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- UBC James Hogg Research Centre, Vancouver, British Columbia, Canada
| | - Raymond T. Ng
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Department of Computer Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - W. Robert McMaster
- NCE CECR Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, British Columbia, Canada
- Immunity and Infection Research Centre, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
47
|
Inhibitory effect of tacrolimus on p38 mitogen-activated protein kinase signaling in kidney transplant recipients measured by whole-blood phosphospecific flow cytometry. Transplantation 2013; 93:1245-51. [PMID: 22643331 DOI: 10.1097/tp.0b013e318250fc62] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tacrolimus (TAC), the cornerstone of immunosuppressive therapy after solid organ transplantation, inhibits calcineurin activation. Despite pharmacokinetic monitoring, patients frequently experience toxicity or lack of efficacy, which could be prevented by pharmacodynamic monitoring. In Jurkat T-cell lines, it has been shown that TAC, in addition to calcineurin, inhibits the p38 mitogen-activated protein kinase (MAPK) pathway, which is important in T-cell activation and is therefore a potential drug-specific biomarker. We studied whether TAC inhibits p38 MAPK signaling in primary human T cells and ex vivo in healthy volunteers and kidney transplant recipients. METHODS Phorbol-12-myristate-13-acetate/ionomycin-induced MAPK signaling was measured by whole-blood phosphospecific flow cytometry. RESULTS In vitro, 10-ng/mL TAC inhibited p38 MAPK phosphorylation by a mean of 27% in CD3, 26% in CD4, and 34% in CD8 T cells (P<0.01 compared with baseline). In healthy adults (n=4), 2 hr after a single oral dose of 10-mg TAC, the p38 MAPK activation was inhibited by 35% in CD3, CD4, and CD8 T cells (P<0.05 compared with baseline). In kidney transplant recipients (n=24), TAC predose concentrations (range, 3.2-10.5 ng/mL) were inversely correlated with p38 MAPK activation in CD3, CD4, and CD8 T cells (r=0.51, 0.34, and 0.37, respectively; P<0.01). CONCLUSIONS TAC inhibits activation of the MAPK pathway in a dose-dependent manner in kidney transplant patients and may be a potential marker for immune monitoring.
Collapse
|
48
|
Lee J, Nakagiri T, Kamimura D, Harada M, Oto T, Susaki Y, Shintani Y, Inoue M, Miyoshi S, Morii E, Hirano T, Murakami M, Okumura M. IL-6 amplifier activation in epithelial regions of bronchi after allogeneic lung transplantation. Int Immunol 2013; 25:319-32. [DOI: 10.1093/intimm/dxs158] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Ramsey H, Pilat N, Hock K, Klaus C, Unger L, Schwarz C, Baranyi U, Gattringer M, Schwaiger E, Wrba F, Wekerle T. Anti-LFA-1 or rapamycin overcome costimulation blockade-resistant rejection in sensitized bone marrow recipients. Transpl Int 2012; 26:206-18. [PMID: 23240587 DOI: 10.1111/tri.12021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/15/2012] [Accepted: 10/25/2012] [Indexed: 02/04/2023]
Abstract
While costimulation blockade-based mixed chimerism protocols work well for inducing tolerance in rodents, translation to preclinical large animal/nonhuman primate models has been less successful. One recognized cause for these difficulties is the high frequency of alloreactive memory T cells (Tmem) found in the (pre)clinical setting as opposed to laboratory mice. In the present study, we therefore developed a murine bone marrow transplantation (BMT) model employing recipients harboring polyclonal donor-reactive Tmem without concomitant humoral sensitization. This model was then used to identify strategies to overcome this additional immune barrier. We found that B6 recipients that were enriched with 3 × 10(7) T cells isolated from B6 mice that had been previously grafted with Balb/c skin, rejected Balb/c BM despite costimulation blockade with anti-CD40L and CTLA4Ig (while recipients not enriched developed chimerism). Adjunctive short-term treatment of sensitized BMT recipients with rapamycin or anti-LFA-1 mAb was demonstrated to be effective in controlling Tmem in this model, leading to long-term mixed chimerism and donor-specific tolerance. Thus, rapamycin and anti-LFA-1 mAb are effective in overcoming the potent barrier that donor-reactive Tmem pose to the induction of mixed chimerism and tolerance despite costimulation blockade.
Collapse
Affiliation(s)
- Haley Ramsey
- Division of Transplantation, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim Y, Park KH, Chung BH, Choi BS, Yang CW, Kim JI, Moon IS, Park YJ, Han K, Oh EJ. Pretransplant IFN-γ ELISPOT assay as a potential screening test to select immunosuppression protocols for patients receiving basiliximab induction therapy. Transl Res 2012; 160:230-6. [PMID: 22683414 DOI: 10.1016/j.trsl.2012.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 02/10/2012] [Accepted: 02/14/2012] [Indexed: 11/29/2022]
Abstract
The use of basiliximab induction therapy has increased in standard immunological risk patients. The objective of this study was to identify whether pretransplant donor-reactive interferon-γ enzyme-linked immunosorbent spot (ELISPOT) assay results were associated with post-transplant clinical outcomes in patients receiving basiliximab induction therapy and whether this could be helpful for choosing an efficacious immunosuppressive regimen. In 154 living donor renal transplant recipients who received basiliximab induction therapy without desensitization, we determined pretransplant ELISPOT frequencies and correlated the results with clinical outcomes based on the use of calcineurin inhibitors (tacrolimus [TAC] or cyclosporine [CSA]). The ELISPOT (+) patients had higher rate of post-transplant biopsy-proven acute rejection (AR) than ELISPOT (-) patients (P = 0.001) regardless of immunosuppressive regimen. In the logistic and multivariate regression analysis, ELISPOT was the only significant correlate of AR (P = 0.002), and the patients with increased ELISPOT results and CSA therapy were associated with AR. Our results suggest that the pretransplant ELISPOT (+) may assess the risk of poor post-transplant outcomes in patients with basiliximab induction.
Collapse
Affiliation(s)
- Yonggoo Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|