1
|
Pipic D, Rasmussen M, Saleh QW, Tepel M. Induction Therapies Determine the Distribution of Perforin and Granzyme B Transcripts in Kidney Transplant Recipients. Biomedicines 2024; 12:1258. [PMID: 38927465 PMCID: PMC11200803 DOI: 10.3390/biomedicines12061258] [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: 05/06/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Peripheral blood mononuclear cells contain secretory granules with Perforin and Granzyme B for defense against pathogens. The objective of the present study was to compare the effects of immunosuppressive induction therapies on Perforin and Granzyme B transcripts in kidney transplant recipients. Transcripts were determined in 408 incident kidney transplant recipients eight days posttransplant using quantitative real-time PCR. Compared to 90 healthy subjects, the median Perforin transcripts were lower in kidney transplant recipients with blood-group ABO-incompatible donors (N = 52), compatible living donors (N = 130), and deceased donors (N = 226) (25.7%; IQR, 6.5% to 46.0%; 31.5%; IQR, 10.9% to 57.7%; and 35.6%; IQR, 20.6% to 60.2%; respectively; p = 0.015 by the Kruskal-Wallis test). Kidney transplant recipients who were treated with thymoglobulin (N = 64) had significantly lower Perforin as well as Granzyme B compared to all other induction therapies (N = 344) (each p < 0.001). Receiver operator characteristics analysis showed that both Perforin (area under curve, 0.919) and Granzyme B (area under curve, 0.915) indicated thyroglobulin-containing induction therapies. Regression analysis showed that both reduction in plasma creatinine and human leukocyte antigen mismatches were positively associated with elevated Perforin/Granzyme B transcript ratio posttransplant. We conclude clinical parameters and therapies affect Perforin and Granzyme B transcripts posttransplant.
Collapse
Affiliation(s)
- Dino Pipic
- Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, 5000 Odense, Denmark
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
| | - Marianne Rasmussen
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
| | - Qais W. Saleh
- Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, 5000 Odense, Denmark
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
| | - Martin Tepel
- Institute of Molecular Medicine, Clinical Institute, University of Southern Denmark, 5000 Odense, Denmark
- Department of Nephrology, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
2
|
Hendrawan K, Khoo MLM, Visweswaran M, Massey JC, Withers B, Sutton I, Ma DDF, Moore JJ. Long-Term Suppression of Circulating Proinflammatory Cytokines in Multiple Sclerosis Patients Following Autologous Haematopoietic Stem Cell Transplantation. Front Immunol 2022; 12:782935. [PMID: 35126353 PMCID: PMC8807525 DOI: 10.3389/fimmu.2021.782935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Autologous haematopoietic stem cell transplantation (AHSCT) is a therapeutic option for haematological malignancies, such as non-Hodgkin’s lymphoma (NHL), and more recently, for autoimmune diseases, such as treatment-refractory multiple sclerosis (MS). The immunological mechanisms underlying remission in MS patients following AHSCT likely involve an anti-inflammatory shift in the milieu of circulating cytokines. We hypothesised that immunological tolerance in MS patients post-AHSCT is reflected by an increase in anti-inflammatory cytokines and a suppression of proinflammatory cytokines in the patient blood. We investigated this hypothesis using a multiplex-ELISA assay to compare the concentrations of secreted cytokine in the peripheral blood of MS patients and NHL patients undergoing AHSCT. In MS patients, we detected significant reductions in proinflammatory T helper (Th)17 cytokines interleukin (IL)-17, IL-23, IL-1β, and IL-21, and Th1 cytokines interferon (IFN)γ and IL-12p70 in MS patients from day 8 to 24 months post-AHSCT. These changes were not observed in the NHL patients despite similar pre-conditioning treatment for AHSCT. Some proinflammatory cytokines show similar trends in both cohorts, such as IL-8 and tumour necrosis factor (TNF)-α, indicating a probable treatment-related AHSCT response. Anti-inflammatory cytokines (IL-10, IL-4, and IL-2) were only transiently reduced post-AHSCT, with only IL-10 exhibiting a significant surge at day 14 post-AHSCT. MS patients that relapsed post-AHSCT exhibited significantly elevated levels of IL-17 at 12 months post-AHSCT, unlike non-relapse patients which displayed sustained suppression of Th17 cytokines at all post-AHSCT timepoints up to 24 months. These findings suggest that suppression of Th17 cytokines is essential for the induction of long-term remission in MS patients following AHSCT.
Collapse
Affiliation(s)
- Kevin Hendrawan
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Melissa L. M. Khoo
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Malini Visweswaran
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Jennifer C. Massey
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Neurology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
| | - Barbara Withers
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
| | - Ian Sutton
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Neurology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
| | - David D. F. Ma
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
| | - John J. Moore
- Blood, Stem Cells and Cancer Research Programme, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Haematology, St Vincent’s Hospital, Darlinghurst, NSW, Australia
- *Correspondence: John J. Moore,
| |
Collapse
|
3
|
Beider K, Naor D, Voevoda V, Ostrovsky O, Bitner H, Rosenberg E, Varda-Bloom N, Marcu-Malina V, Canaani J, Danilesko I, Shimoni A, Nagler A. Dissecting the mechanisms involved in anti-human T-lymphocyte immunoglobulin (ATG)-induced tolerance in the setting of allogeneic stem cell transplantation - potential implications for graft versus host disease. Oncotarget 2017; 8:90748-90765. [PMID: 29207601 PMCID: PMC5710882 DOI: 10.18632/oncotarget.21797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023] Open
Abstract
Polyclonal anti-human thymocyte globulins (ATG) have been recently shown to significantly reduce the incidence of graft versus host disease (GVHD) post allogeneic stem cell transplantation (HSCT) from both sibling and unrelated donors. Induction of regulatory T cells has been suggested as one of the possible mechanisms. The aim of current study was to further characterize the T cell populations induced by ATG treatment and to delineate the mechanisms involved in ATG-induced tolerance. Phenotypic characterization revealed a significant increase in the expression of FoxP3, GITR, CD95, PD-1 and ICOS as well as the complement inhibitory molecules CD55, CD58 and CD59 on CD4+CD25+ T cells upon ATG treatment. Addition of ATG-treated cells to autologous and allogeneic peripheral blood mononuclear cells (PBMCs) stimulated with anti-CD3/anti-CD28 antibodies resulted in significant inhibition of proliferation. Moreover, T-cell activation and IFNγ secretion were reduced in the presence of ATG-induced Treg cells. The CD4+CD25+CD127-low Treg fraction sorted from ATG-treated culture demonstrated greater suppressive potency than negative fraction. Conditioned medium produced by ATG-treated but not IgG-treated cells contained TGFβ and suppressed T cell proliferation and activation in a TGFβ receptor-dependent manner. TGFβ receptor kinase inhibitor SB431542 interfered with the suppressive activity of ATG-primed cells, enabling partial rescue of proliferation and IFNγ secretion. Moreover, SB431542 prevented Treg phenotype induction upon ATG treatment. Altogether, our data reveal the role of TGFβ signaling in ATG-mediated immunosuppression and further support the use of ATG, a potent inducer of regulatory T cells, for prevention of GVHD post HSCT and potentially other therapeutic applications.
Collapse
Affiliation(s)
- Katia Beider
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - David Naor
- 2 Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Valeria Voevoda
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Olga Ostrovsky
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Hanna Bitner
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Evgenia Rosenberg
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Nira Varda-Bloom
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Victoria Marcu-Malina
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Jonathan Canaani
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Ivetta Danilesko
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Avichai Shimoni
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center and Tel Aviv University, Tel-Hashomer, Ramat Gan, Israel
| |
Collapse
|
4
|
Choi I, Yoon SR, Park SY, Kim H, Jung SJ, Kang YL, Lee JH, Lee JH, Kim DY, Lee JL, Park HS, Choi EJ, Lee YS, Kang YA, Jeon M, Seol M, Baek S, Yun SC, Kim HJ, Lee KH. Donor-Derived Natural Killer Cell Infusion after Human Leukocyte Antigen-Haploidentical Hematopoietic Cell Transplantation in Patients with Refractory Acute Leukemia. Biol Blood Marrow Transplant 2016; 22:2065-2076. [PMID: 27530969 DOI: 10.1016/j.bbmt.2016.08.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/08/2016] [Indexed: 11/29/2022]
Abstract
The optimum method of donor natural killer cell infusion (DNKI) after allogeneic hematopoietic cell transplantation (HCT) remains unclear. Fifty-one patients (age range, 19 years to 67 years) with refractory acute leukemia underwent HLA-haploidentical HCT and underwent DNKI on days 6, 9, 13, and 20 of HCT. Median DNKI doses were .5, .5, 1.0, and 2.0 × 108/kg cells, respectively. During DNKI, 33 of the 45 evaluated patients (73%) developed fever (>38.3°C) along with weight gain (median, 13%; range, 2% to 31%) and/or hyperbilirubinemia (median, 6.2 mg/dL; range, 1.0 mg/dL to 35.1 mg/dL); the toxicity was reversible in 90% of patients. After transplantation, we observed cumulative incidences of neutrophil engraftment (≥500/µL), grade 2 to 4 acute graft-versus-host disease (GVHD), chronic GVHD, and nonrelapse mortality of 84%, 28%, 30%, and 16%, respectively. The leukemia complete remission rate was 57% at 1 month after HCT and 3-year cumulative incidence of leukemia progression was 75%. When analyzed together with our historical cohort of 40 patients with refractory acute leukemia who underwent haploidentical HCT and DNKI on days 14 and 21 only, higher expression of NKp30 (>90%) on donor NK cells was an independent predictor of higher complete remission (hazard ratio, 5.59) and less leukemia progression (hazard ratio, .57). Additional DNKI on days 6 and 9 was not associated with less leukemia progression (75% versus 55%).
Collapse
Affiliation(s)
- Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
| | - Suk Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Soo-Yeon Park
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hanna Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sol-Ji Jung
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - You-Lee Kang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Je-Hwan Lee
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Jung-Hee Lee
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Dae-Young Kim
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Jae-Lyun Lee
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Han-Seung Park
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Eun-Ji Choi
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Young-Shin Lee
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Young-A Kang
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Mijin Jeon
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Miee Seol
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Seunghyun Baek
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Sung-Cheol Yun
- Department of Clinical Epidemiology and Biostatistics, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatistics, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Kyoo-Hyung Lee
- Hematology and Oncology Sections, Department of Internal Medicine, College of Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Alemtuzumab as Antirejection Therapy: T Cell Repopulation and Cytokine Responsiveness. Transplant Direct 2016; 2:e83. [PMID: 27500273 PMCID: PMC4946521 DOI: 10.1097/txd.0000000000000595] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/10/2016] [Indexed: 01/05/2023] Open
Abstract
Supplemental digital content is available in the text. Background Alemtuzumab induction therapy in kidney transplant patients results in T cell depletion followed by slow immune reconstitution of memory T cells with reduced immune functions. The kinetics and functional characteristics of T cell reconstitution when alemtuzumab is given during immune activation, ie, as antirejection therapy, are unknown. Methods Patients (n = 12) with glucocorticoid-resistant or severe vascular kidney transplant rejection were treated with alemtuzumab. Flow cytometric analysis was performed on whole blood to measure cell division by the marker Ki-67, and cytokine responsiveness by IL-2–mediated and IL-7–mediated phosphorylation of signal transducer and activator of transcription 5 of T cells before and during the first year after rejection therapy. Results At 1 year after alemtuzumab antirejection therapy, the total T cell population recovered to baseline level. Repopulation of CD4+ and CD8+ T cells was associated with increased percentages of Ki-67+ proliferating T cells (P < 0.05). In addition, both populations showed a phenotypic shift toward relatively more memory T cells (P < 0.01). At the functional level, IL-7 reactivity of CD4+ memory T cells was diminished, reflected by a decreased capacity to phosphorylate signal transducer and activator of transcription 5 during the first 6 months after alemtuzumab treatment (P < 0.05), whereas reactivity to IL-2 was preserved. CD8+ T cells were affected in terms of both IL-2 and IL-7 responses (both P < 0.05). After reconstitution, relatively more regulatory T cells were present, and a relatively high proportion of Ki-67+ T cells was observed. Conclusions Preliminary data from this small series suggest that alemtuzumab antirejection therapy induces homeostatic proliferation of memory and regulatory T cells with diminished responsiveness to the homeostatic cytokine IL-7. IL-2 responsiveness was affected in repopulated CD8+ T cells.
Collapse
|
6
|
Effect of induction therapy on the expression of molecular markers associated with rejection and tolerance. BMC Nephrol 2015; 16:146. [PMID: 26286066 PMCID: PMC4545708 DOI: 10.1186/s12882-015-0141-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/06/2015] [Indexed: 12/02/2022] Open
Abstract
Background Induction therapy can improve kidney transplantation (KTx) outcomes, but little is known about the mechanisms underlying its effects. Methods The mRNA levels of T cell-related genes associated with tolerance or rejection (CD247, GZMB, PRF1, FOXP3, MAN1A1, TCAIM, and TLR5) and lymphocyte subpopulations were monitored prospectively in the peripheral blood of 60 kidney transplant recipients before and 7, 14, 21, 28, 60, 90 days, 6 months, and 12 months after KTx. Patients were treated with calcineurin inhibitor-based triple immunosuppression and induction with rabbit anti-thymocyte globulin (rATG, n = 24), basiliximab (n = 17), or without induction (no-induction, n = 19). A generalized linear mixed model with gamma distribution for repeated measures, adjusted for rejection, recipient/donor age and delayed graft function, was used for statistical analysis. Results rATG treatment caused an intense reduction in all T cell type population and natural killer (NK) cells within 7 days, then a slow increase and repopulation was observed. This was also noticed in the expression levels of CD247, FOXP3, GZMB, and PRF1. The basiliximab group exhibited higher CD247, GZMB, FOXP3 and TCAIM mRNA levels and regulatory T cell (Treg) counts than the no-induction group. The levels of MAN1A1 and TLR5 mRNA expressions were increased, whereas TCAIM decreased in the rATG group as compared with those in the no-induction group. Conclusion The rATG induction therapy was associated with decreased T and NK cell-related transcript levels and with upregulation of two rejection-associated transcripts (MAN1A1 and TLR5) shortly after KTx. Basiliximab treatment was associated with increased absolute number of Treg cells, and increased level of FOXP3 and TCAIM expression.
Collapse
|
7
|
Roedder S, Vitalone M, Khatri P, Sarwal MM. Biomarkers in solid organ transplantation: establishing personalized transplantation medicine. Genome Med 2011; 3:37. [PMID: 21658299 PMCID: PMC3218811 DOI: 10.1186/gm253] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Technological advances in molecular and in silico research have enabled significant progress towards personalized transplantation medicine. It is now possible to conduct comprehensive biomarker development studies of transplant organ pathologies, correlating genomic, transcriptomic and proteomic information from donor and recipient with clinical and histological phenotypes. Translation of these advances to the clinical setting will allow assessment of an individual patient's risk of allograft damage or accommodation. Transplantation biomarkers are needed for active monitoring of immunosuppression, to reduce patient morbidity, and to improve long-term allograft function and life expectancy. Here, we highlight recent pre- and post-transplantation biomarkers of acute and chronic allograft damage or adaptation, focusing on peripheral blood-based methodologies for non-invasive application. We then critically discuss current findings with respect to their future application in routine clinical transplantation medicine. Complement-system-associated SNPs present potential biomarkers that may be used to indicate the baseline risk for allograft damage prior to transplantation. The detection of antibodies against novel, non-HLA, MICA antigens, and the expression of cytokine genes and proteins and cytotoxicity-related genes have been correlated with allograft damage and are potential post-transplantation biomarkers indicating allograft damage at the molecular level, although these do not have clinical relevance yet. Several multi-gene expression-based biomarker panels have been identified that accurately predicted graft accommodation in liver transplant recipients and may be developed into a predictive biomarker assay.
Collapse
Affiliation(s)
- Silke Roedder
- Department of Pediatrics and Immunology, Stanford University, G306 300 Pasteur Drive, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
8
|
Kim YH, Wee YM, Choi MY, Lim DG, Kim SC, Han DJ. Interleukin (IL)-10 induced by CD11b(+) cells and IL-10-activated regulatory T cells play a role in immune modulation of mesenchymal stem cells in rat islet allografts. Mol Med 2011; 17:697-708. [PMID: 21365122 DOI: 10.2119/molmed.2010.00098] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 02/24/2011] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are suggested to be immune modulators because of their therapeutic potential in transplantation. In the present study, we evaluated the therapeutic potential of autologous MSCs for preventing graft rejection after allogeneic rat islet transplantation. We assessed the ability of MSCs to elicit an antiproliferative response in alloreactive lymphocytes and tested the immunosuppressive effect of MSCs in allogeneic islet transplantation. In islet allotransplantation, injection of autologous MSCs or a subtherapeutic dose of cyclosporine A (CsA; 5 mg/kg) alone did not prolong allograft survival. However, graft survival was attained for >100 d in 33% of autologous MSC-plus-CsA-treated recipients, indicating that graft acceptance was achieved in a subgroup of allograft recipients. Splenocytes from autologous MSC-plus-CsA-treated rats exhibited a reduced mixed lymphocyte reaction (MLR)-proliferative response to donor stimulators and increased interleukin (IL)-10 release. Interestingly, after excluding host CD11b(+) cells, splenic T cells from autologous MSC-plus-CsA-treated rats did not produce IL-10 or did not inhibit proliferative responses under the same conditions. The use of autologous MSC-plus-CsA downregulated immune responses, inducing donor-specific T-cell hyporesponsiveness by reducing the production of proinflammatory cytokines and inducing antiinflammatory cytokine production, especially that of IL-10, during the early posttransplantation period. T-regulatory cells made a contribution at a later phase. In conclusion, the combined use of autologous MSCs and low-dose CsA exerted a synergistic immunosuppressive effect in an islet allograft model, suggesting a role for autologous MSCs as an immune modulator.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Department of Surgery, University of Ulsan College of Medicine, and Asan Medical Center, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
9
|
Fishman JA, Issa NC. Infection in Organ Transplantation: Risk Factors and Evolving Patterns of Infection. Infect Dis Clin North Am 2010; 24:273-83. [DOI: 10.1016/j.idc.2010.01.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Sewgobind VDKD, van der Laan LJW, Kho MML, Kraaijeveld R, Korevaar SS, Mol W, Weimar W, Baan CC. The calcineurin inhibitor tacrolimus allows the induction of functional CD4CD25 regulatory T cells by rabbit anti-thymocyte globulins. Clin Exp Immunol 2010; 161:364-77. [PMID: 20528886 DOI: 10.1111/j.1365-2249.2010.04183.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rabbit anti-thymocyte globulins (rATG) induce CD4(+)CD25(+)forkhead box P3 (FoxP3(+)) regulatory T cells that control alloreactivity. In the present study, we investigated whether rATG convert T cells into functional CD4(+)CD25(+)FoxP3(+)CD127(-/low) regulatory T cells in the presence of drugs that may hamper their induction and function, i.e. calcineurin inhibitors. CD25(neg) T cells were stimulated with rATG or control rabbit immunoglobulin G (rIgG) in the absence and presence of tacrolimus for 24 h. Flow cytometry was performed for CD4, CD25, FoxP3 and CD127 and the function of CD25(+) T cells was examined in suppression assays. MRNA expression profiles were composed to study the underlying mechanisms. After stimulation, the percentage CD4(+)CD25(+)FoxP3(+)CD127(-/low) increased (from 2% to 30%, mean, P < 0.01) and was higher in the rATG samples than in control rIgG samples (2%, P < 0.01). Interestingly, FoxP3(+)T cells were also induced when tacrolimus was present in the rATG cultures. Blockade of the interleukin (IL)-2 pathway did not affect the frequency of rATG-induced FoxP3(+) T cells. The rATG tacrolimus-induced CD25(+) T cells inhibited proliferative responses of alloantigen-stimulated effector T cells as vigorously as rATG-induced and natural CD4(+)CD25(+)FoxP3(+)CD127(-/low) T cells (67% +/- 18% versus 69% +/- 16% versus 45% +/- 20%, mean +/- standard error of the mean, respectively). At the mRNA-expression level, rATG-induced CD25(+) T cells abundantly expressed IL-10, IL-27, interferon (IFN)-gamma, perforin and granzyme B in contrast to natural CD25(+) T cells (all P = 0.03), while FoxP3 was expressed at a lower level (P = 0.03). These mRNA data were confirmed in regulatory T cells from kidney transplant patients. Our findings demonstrate that tacrolimus does not negatively affect the induction, phenotype and function of CD4(+)CD25(+) T cells, suggesting that rATG may induce regulatory T cells in patients who receive tacrolimus maintenance therapy.
Collapse
Affiliation(s)
- V D K D Sewgobind
- Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
11
|
One center's experience with antithymocyte globulin treatment for acute rejection in renal transplantation. Transplant Proc 2008; 40:123-5. [PMID: 18261564 DOI: 10.1016/j.transproceed.2007.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Antithymocyte globulin (ATG) is a polyclonal antibody used in renal transplantation for prevention and treatment of acute rejection. In this study we have presented the outcomes of 23 cases treated with ATG due to steroid-resistant acute rejection episodes in 17 male and 6 female recipients. Sixteen transplantations were performed from cadaver donors and the other 7 from living-related donors. The mean recipient age was 31.9 +/- 9 years and the mean donor age was 56.3 +/- 10.8 years. ATG treatment was administered in doses of 3-5 mg/kg/d for 10 or 14 days. All patients received the same premedication before the ATG treatment; we did not encounter any ATG-related side effects. ATG doses were adjusted according to the T-lymphocyte levels. All recipients were followed up for infectious complications, for graft function, and for immunologic parameters of CD3 levels, CD4 levels, CD3/CD4 ratios, lymphocyte, and polymorphonuclear leukocyte numbers. According to the Banff criteria introduced in 1997, 4 patients displayed humoral rejection: 3 had type 3; 5 had type 2; and 11 had type 1 acute rejection episodes. Nine patients developed infectious complication during the follow-up. Three had pulmonary aspergillosis; 2 had cytomegalovirus infection; and 4 had bacterial infections. One patient who experienced aspergillosis died with a functioning graft, and the remaining 8 patients were treated successfully. Graft function improved in 19 (83%) cases. The other 4 patients returned to hemodialysis. Mean creatinine levels decreased from 4 +/- 1.7 to 2.1 +/- 0.2 mg/dL. We did not observe any relationship between the immunologic parameters and infectious complications. In conclusion, although ATG is a powerful drug to treat steroid-resistant acute rejection episodes, there was no precise way to monitor the intensity of immunosuppression to prevent infectious complications.
Collapse
|
12
|
Derfuss T, Segerer S, Herberger S, Sinicina I, Hüfner K, Ebelt K, Knaus HG, Steiner I, Meinl E, Dornmair K, Arbusow V, Strupp M, Brandt T, Theil D. Presence of HSV-1 immediate early genes and clonally expanded T-cells with a memory effector phenotype in human trigeminal ganglia. Brain Pathol 2007; 17:389-98. [PMID: 17784877 PMCID: PMC8095593 DOI: 10.1111/j.1750-3639.2007.00088.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The latent persistence of herpes simplex virus type 1 (HSV-1) in human trigeminal ganglia (TG) is accompanied by a chronic CD8 T-cell infiltrate. The focus of the current work was to look for HSV-1 transcription activity as a potential trigger of the immune response and to characterize the immune cell infiltrates by this feature. We combined in situ hybridization, laser cutting microscopy, and single cell RT-PCR to demonstrate the expression of the HSV-1 immediate early (IE) genes ICP0 and ICP4 in human trigeminal neurons. Using CDR3 spectratyping, we showed that the infiltrating T-cells are clonally expanded, indicating an antigen-driven immune response. Moreover, the persisting CD8+ T-cells had features of the memory effector phenotype. The voltage-gated potassium channel Kv1.3, a marker of chronic activated memory effector cells, and the chemokines CCL5 and CXCL10 were expressed by a subpopulation of infiltrating cells. The corresponding chemokine receptors CCR5 and CXCR3 were co-expressed on virtually all CD8 T-cells. In addition, T-cells expressed granzymes and perforin. In contrast to animal models of HSV-1 latency, hardly any FoxP3-positive regulatory T-cells were detected in human TG. Thus, HSV-1 IE genes are expressed in human TG and the infiltrating T-cells bear several characteristics that suggest viral antigenic stimulation.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/virology
- Chemokines/immunology
- Chemokines/metabolism
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/immunology
- Clone Cells/immunology
- Clone Cells/virology
- Female
- Gene Expression Regulation, Viral/genetics
- Genes, Immediate-Early/genetics
- Genes, Viral/genetics
- Herpes Simplex/genetics
- Herpes Simplex/physiopathology
- Herpes Simplex/virology
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/immunology
- Humans
- Immunologic Memory/genetics
- Immunologic Memory/immunology
- Kv1.3 Potassium Channel/metabolism
- Male
- Middle Aged
- Neurons, Afferent/immunology
- Neurons, Afferent/virology
- Phenotype
- Receptors, Chemokine/immunology
- Receptors, Chemokine/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/virology
- Trigeminal Ganglion/cytology
- Trigeminal Ganglion/immunology
- Trigeminal Ganglion/virology
- Virus Latency/genetics
- Virus Latency/immunology
Collapse
Affiliation(s)
- Tobias Derfuss
- Institute of Clinical Neuroimmunology, University Clinic, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Lytton SD, Denton CP, Nutzenberger AM. Treatment of Autoimmune Disease with Rabbit Anti-T Lymphocyte Globulin: Clinical Efficacy and Potential Mechanisms of Action. Ann N Y Acad Sci 2007; 1110:285-96. [PMID: 17911443 DOI: 10.1196/annals.1423.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The rabbit anti-T lymphocyte globulins (rATGs) are immune-suppressive anti-T cell agents with beneficial effects in solid organ and hematological transplantation. The present review evaluates the potential mechanisms of rATGs and their impact on pilot and exploratory studies of diffuse cutaneous systemic sclerosis (scleroderma-SSc), inclusion body myositis (IBM), vasculitis, and type 1 diabetes mellitus (T1DM). The rATGs are associated with improvements in well-defined parameters of clinical autoimmunity: insulin usage, tissue inflammation, and systemic organ functions. Meta-analysis of a retrospective database of SSc, N = 196 and two prospective randomized pilot studies; IBM, N = 11 and T1DM, N = 17 shows a two- to ninefold increase in the relative response to treatments with intravenous infusions of rATG. The rATGs deplete T cells and are associated with increases in the percentage of CD25+ T cell subsets. This may underlie the apparent long-lasting immunomodulation associated with these agents. The future optimization of rATG adjunct therapy requires statistically powered-controlled prospective trials of rATG dose-finding and timing of administration. The potential mechanisms of rATGs:depletion of autoreactive T cells, generation CDCD25+Foxp3+ regulatory T cells (Tregs), and the acquisition of regulatory immune cell functions, need to be examined in patients prior to rATG infusion and at time intervals following rATG treatment to identify those mechanisms relevant to the improvement of their clinical outcome.
Collapse
Affiliation(s)
- Simon D Lytton
- SeraDiaLogistics, Hertlingstr 1, 81545 München, Germany.
| | | | | |
Collapse
|
14
|
Yannaraki M, Rebibou JM, Ducloux D, Saas P, Duperrier A, Felix S, Rifle G, Chalopin JM, Hervé P, Tiberghien P, Ferrand C. Urinary cytotoxic molecular markers for a noninvasive diagnosis in acute renal transplant rejection. Transpl Int 2006; 19:759-68. [PMID: 16918537 DOI: 10.1111/j.1432-2277.2006.00351.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Perforin (P), Granzyme B (GB) and Fas-Ligand (FAS-L) are cytotoxic molecules involved in acute rejection (AR) after renal transplantation. A noninvasive diagnostic test to monitor AR and other complications could improve clinical management. We investigated the predictive and diagnostic interest of target mRNA measurements, with a quantitative PCR assay, in AR, as well as in other clinical complications recurrent in kidney transplantation. One hundred and sixty-two urine specimens from 37 allograft recipients were investigated. Clinical settings were AR, urinary tract infection (UTI), cytomegalovirus infection (CMVi) or disease (CMVd), chronic allograft nephropathy (CAN), delayed graft function (DGF) and stable graft course (controls). In the case of AR, mRNA levels of all three molecules were significantly higher than in recipients not showing any clinically evident signs of complication. Indeed, it was observed that expression levels of P, GB and Fas-L mRNA also increase in other clinical situations such as UTI, CMV and DGF. Finally, kinetic studies in three patients with AR revealed that increased P, GB and Fas-L mRNA levels could precede or were concomitant with increased serum creatinin levels. P, GB and Fas-L gene expression in urine specimens were upregulated in AR episodes but also in UTI, CMV infection and DGF. Therefore, this technique would appear to be of limited clinical value as a noninvasive method of diagnosing AR.
Collapse
|
15
|
Simon T, Opelz G, Wiesel M, Pelzl S, Ott RC, Süsal C. Serial peripheral blood interleukin-18 and perforin gene expression measurements for prediction of acute kidney graft rejection. Transplantation 2004; 77:1589-95. [PMID: 15239627 DOI: 10.1097/01.tp.0000121764.92730.fb] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We and others have shown that expression of the cytotoxic T-lymphocyte effector gene perforin in the peripheral blood is a strong predictor of acute rejection in the early posttransplant period. In the present study we investigated whether interleukin (IL)-18, an immunostimulatory gene that up-regulates perforin-dependent cytotoxicity and promotes tissue damage through other noncytotoxic T-lymphocyte mechanisms alone or in combination with perforin gene expression, may serve as a better predictor of renal allograft rejection in the first weeks after transplantation. METHODS Peripheral blood was collected twice weekly, and gene expression was measured using real-time polymerase chain reaction. RESULTS Recipients with acute rejection (n = 17) showed higher levels of perforin and IL-18 transcript on days 5 to 7, 8 to 10, and 11 to 13, compared with patients without rejection (n = 37, P < 0.01 in all cases). Rejection diagnosis using gene expression criteria was possible 1 to 32 days before traditional diagnosis (median 11 days). High specificity was associated with IL-18 expression (72%-93%), and high sensitivity was associated with perforin expression (63%-90%). Positive predictive value was optimized (78%-100%) by using combined up-regulation in both genes as a diagnostic criterion (double-positive). Using high expression in "either or both" genes as a diagnostic criterion yielded high sensitivity (82%-91%) and negative predictive value (91%-96%). CONCLUSIONS Our data indicate that combined perforin and IL-18 gene expression measurements are useful tools for the recognition of graft rejection in its earliest stages. Serial measurements could be implemented as a monitoring system to identify patients at higher risk of rejection, making them candidates for biopsy or prophylactic increases in immunosuppression.
Collapse
Affiliation(s)
- Tania Simon
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Süsal C, Pelzl S, Simon T, Opelz G. Advances in pre- and posttransplant immunologic testing in kidney transplantation. Transplant Proc 2004; 36:29-34. [PMID: 15013292 DOI: 10.1016/j.transproceed.2003.11.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pre- and posttransplant risk estimation in kidney transplantation is important for the selection of appropriate treatment strategies. Recently, using new immunologic tests, we made observations within the framework of the Collaborative Transplant Study that may influence clinical practice. Complement-dependent lymphocytotoxic panel reactivity as a measure of anti-HLA sensitization, although criticized for its low sensitivity, is a useful indicator of an increased risk of rejection. Using the more sensitive complement-independent ELISA methodology, which utilizes solubilized HLA molecules instead of lymphocytes, we found that recipients with preformed complement-dependent anti-HLA antibodies showed a decreased graft survival only if their antibodies were directed against both HLA class I and class II, whereas isolated reactivity only against HLA class I or class II was of no clinical consequence. Pretransplant serum-soluble CD30 (sCD30) was found to be an independent and highly predictive factor of immunologic risk. The effects of sCD30 and anti-HLA antibodies were additive. Importantly, even patients without anti-HLA antibodies showed a strong HLA matching effect if their pretransplant serum contained high levels of sCD30. Although the role of anti-HLA antibody formation after transplantation remains uncertain, ELISA-detected sCD30 was shown to indicate impending graft rejection as early as on posttransplant days 3 to 5. Another very sensitive indicator of impending rejection is provided by posttransplant monitoring of the cytotoxic T-lymphocyte effector genes, perforin and granzyme B, in peripheral blood using real-time PCR.
Collapse
Affiliation(s)
- C Süsal
- Department of Transplantation Immunology, Institute of Immunology University of Heidelberg, Heidelberg, Germany.
| | | | | | | |
Collapse
|
17
|
Ensom MHH, Partovi N, Decarie D, Ignaszewski AP, Fradet GJ, Levy RD. Mycophenolate Pharmacokinetics in Early Period Following Lung or Heart Transplantation. Ann Pharmacother 2003; 37:1761-7. [PMID: 14632536 DOI: 10.1345/aph.1d099] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The available pharmacokinetic and pharmacodynamic data on mycophenolic acid (MPA), the pharmacologically active metabolite of mycophenolate mofetil (MMF), are derived largely from renal transplant patients, not thoracic transplant recipients. OBJECTIVE To evaluate, in a pilot study, the pharmacokinetics of MPA at 3 different times in the early period (up to the first 9 mo) following lung or heart transplantation. METHODS Nine patients were entered into this open-label study. Upon administration of a steady-state morning MMF dose, blood samples were collected at 0, 20, 40, 60, and 90 minutes and at 2, 4, 6, 8, 10, and 12 hours after the dose at 3 times (denoted as sampling periods 1, 2, and 3) in the early posttransplant period. Total MPA concentrations were measured by a validated HPLC method with ultraviolet detection and followed by ultrafiltration of pooled samples for unbound MPA concentrations. Pharmacokinetic parameters (maximal concentration [C(max)], dose-normalized C(max), time to C(max), minimum concentration, predose concentration, AUC, dose-normalized AUC, free fraction, free AUC) were calculated by traditional noncompartmental methods. RESULTS Patient characteristics included 7 men and 2 women, 5 lung and 4 heart transplant recipients, mean +/- SD age 53 +/- 11 years, and weight 77 +/- 14 kg. All patients were receiving prednisone and cyclosporine (with the exception of 2 pts. on tacrolimus during sampling periods 2 and 3). Sampling periods 1, 2, and 3 occurred on posttransplant days 15 +/- 13, 56 +/- 33, and 125 +/- 73, respectively. No significant differences were found between sampling periods in any pharmacokinetic parameter. Drug exposure as evaluated by AUC was 39.95 +/- 44.86, 25.24 +/- 25.68, and 43.96 +/- 38.67 micro g*h/mL during sampling periods 1, 2, and 3, respectively, (p > 0.05). CONCLUSIONS As of September 26, 2003, this is the first study to systematically evaluate MPA pharmacokinetics in thoracic transplant recipients at 3 different time points during the early posttransplant period. Wide interpatient variability in MPA pharmacokinetics was observed, thus emphasizing the need to individualize dosing of MMF and to further evaluate important pharmacokinetic/pharmacodynamic parameters and endpoints that impact on clinical outcomes. Further studies involving more patients and pharmacodynamic outcomes are underway to help identify optimal MMF strategies.
Collapse
Affiliation(s)
- Mary H H Ensom
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada and Clinical Pharmacy Specialist, Pharmacy Department, Children's & Women's Health Centre of British Columbia.
| | | | | | | | | | | |
Collapse
|
18
|
Simon T, Opelz G, Wiesel M, Ott RC, Süsal C. Serial peripheral blood perforin and granzyme B gene expression measurements for prediction of acute rejection in kidney graft recipients. Am J Transplant 2003; 3:1121-7. [PMID: 12919092 DOI: 10.1034/j.1600-6143.2003.00187.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In the present study we investigated whether peripheral blood gene expression measurements may serve as an early and non-invasive tool to predict renal allograft rejection. Peripheral blood was collected twice weekly after transplantation and gene expression was measured using real-time polymerase chain reaction (PCR). Recipients with acute rejection (n = 17) had higher levels of perforin and granzyme B transcript on days 5-7, 8-10, 11-13, 17-19, 20-22, and 26-29, as compared to patients without rejection (n = 50, p < 0.05 in all cases). Rejection diagnosis using gene expression criteria, determined with receiver operating characteristic (ROC) curves, was possible 2-30 days before traditional diagnosis (median 11 days). The best diagnostic result was obtained from samples taken on days 8-10, with a specificity of 90% and a sensitivity of 82% for perforin, and a specificity of 87% and sensitivity of 72% for granzyme B. Decreases in perforin (p < 0.01) and granzyme B expression (p < 0.05) were observed after initiation of anti-rejection therapy. Our data indicate that gene expression measurement is a useful tool for the recognition of graft rejection in its earliest stages. Serial measurements could be implemented as a monitoring system to highlight patients at higher risk of rejection, making them candidates for biopsy or pre-emptive anti-rejection therapy.
Collapse
Affiliation(s)
- Tania Simon
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|