1
|
Tharmaraj D, Mulley WR, Dendle C. Current and emerging tools for simultaneous assessment of infection and rejection risk in transplantation. Front Immunol 2024; 15:1490472. [PMID: 39660122 PMCID: PMC11628869 DOI: 10.3389/fimmu.2024.1490472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Infection and rejection are major complications that impact transplant longevity and recipient survival. Balancing their risks is a significant challenge for clinicians. Current strategies aimed at interrogating the degree of immune deficiency or activation and their attendant risks of infection and rejection are imprecise. These include immune (cell counts, function and subsets, immunoglobulin levels) and non-immune (drug levels, viral loads) markers. The shared risk factors between infection and rejection and the bidirectional and intricate relationship between both entities further complicate transplant recipient care and decision-making. Understanding the dynamic changes in the underlying net state of immunity and the overall risk of both complications in parallel is key to optimizing outcomes. The allograft biopsy is the current gold standard for the diagnosis of rejection but is associated with inherent risks that warrant careful consideration. Several biomarkers, in particular, donor derived cell-free-DNA and urinary chemokines (CXCL9 and CXCL10), show significant promise in improving subclinical and clinical rejection risk prediction, which may reduce the need for allograft biopsies in some situations. Integrating conventional and emerging risk assessment tools can help stratify the individual's short- and longer-term infection and rejection risks in parallel. Individuals identified as having a low risk of rejection may tolerate immunosuppression wean to reduce medication-related toxicity. Serial monitoring following immunosuppression reduction or escalation with minimally invasive tools can help mitigate infection and rejection risks and allow for timely diagnosis and treatment of these complications, ultimately improving allograft and patient outcomes.
Collapse
Affiliation(s)
- Dhakshayini Tharmaraj
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - William R. Mulley
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Claire Dendle
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, VIC, Australia
- Monash Infectious Diseases, Monash Health, Clayton, VIC, Australia
| |
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
|
Süsal C, Alvarez CM, Benning L, Daniel V, Zeier M, Schaier M, Morath C, Speer C. The balance between memory and regulatory cell populations in kidney transplant recipients with operational tolerance. Clin Exp Immunol 2024; 216:318-330. [PMID: 38393856 PMCID: PMC11097908 DOI: 10.1093/cei/uxae018] [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: 06/30/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 02/25/2024] Open
Abstract
Donor-reactive memory cells represent a barrier to long-term kidney graft survival. A better understanding of regulatory mechanisms that counterbalance alloreactive memory responses may help to identify patients with operational tolerance. This prospective study investigated the equilibrium between memory T-cell subsets and regulatory T or B cells (Tregs, Bregs) in peripheral blood of kidney transplant recipients with operational tolerance (N = 8), chronic rejection (N = 8), and different immunosuppressive treatment regimens (N = 81). Patients on hemodialysis and healthy individuals served as controls (N = 50). In addition, the expression of Treg- and Breg-associated molecule genes was analyzed. Patients with chronic rejection showed a disrupted memory T-cell composition with a significantly higher frequency of circulating CD8+ terminally differentiated effector memory (TEMRA) T cells than patients with operational tolerance, patients on hemodialysis, or healthy controls (P < 0.001). Low frequency of CD8+ TEMRA and high frequency of Tregs and transitional Bregs were found in operationally tolerant patients. Consequently, operationally tolerant patients showed, as compared to all other transplant recipients with different immunosuppressive regiments, the lowest ratios between CD8+ TEMRA T cells and Tregs or Bregs (for both P < 0.001). Moreover, a specific peripheral blood transcription pattern was found in operationally tolerant patients with an increased expression of Breg- and Treg-associated genes CD22 and FoxP3 and a decreased FcγRIIA/FcγRIIB transcript ratio (for all P < 0.001). In conclusion, monitoring the balance between circulating CD8+ TEMRA T cells and regulatory cell subsets and their transcripts may help to distinguish transplant recipients with operational tolerance from recipients at risk of graft loss.
Collapse
Affiliation(s)
- Caner Süsal
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
- Transplant Immunology Research Center of Excellence, Koç University Hospital, Istanbul, Turkey
| | - Cristiam M Alvarez
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Louise Benning
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Volker Daniel
- Institute of Immunology, University of Heidelberg, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Christian Morath
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Claudius Speer
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Hu Y, Hao F, An Q, Jiang W. Immune cell signatures and inflammatory mediators: unraveling their genetic impact on chronic kidney disease through Mendelian randomization. Clin Exp Med 2024; 24:94. [PMID: 38703294 PMCID: PMC11069478 DOI: 10.1007/s10238-024-01341-z] [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: 12/18/2023] [Accepted: 03/27/2024] [Indexed: 05/06/2024]
Abstract
Prior research has established associations between immune cells, inflammatory proteins, and chronic kidney disease (CKD). Our Mendelian randomization study aims to elucidate the genetic causal relationships among these factors and CKD. We applied Mendelian randomization using genetic variants associated with CKD from a large genome-wide association study (GWAS) and inflammatory markers from a comprehensive GWAS summary. The causal links between exposures (immune cell subtypes and inflammatory proteins) and CKD were primarily analyzed using the inverse variance-weighted, supplemented by sensitivity analyses, including MR-Egger, weighted median, weighted mode, and MR-PRESSO. Our analysis identified both absolute and relative counts of CD28 + CD45RA + CD8 + T cell (OR = 1.01; 95% CI = 1.01-1.02; p < 0.001, FDR = 0.018) (OR = 1.01; 95% CI = 1.00-1.01; p < 0.001, FDR = 0.002), CD28 on CD39 + CD8 + T cell(OR = 0.97; 95% CI = 0.96-0.99; p < 0.001, FDR = 0.006), CD16 on CD14-CD16 + monocyte (OR = 1.02; 95% CI = 1.01-1.03; p < 0.001, FDR = 0.004) and cytokines, such as IL-17A(OR = 1.11, 95% CI = 1.06-1.16, p < 0.001, FDR = 0.001), and LIF-R(OR = 1.06, 95% CI = 1.02-1.10, p = 0.005, FDR = 0.043) that are genetically predisposed to influence the risk of CKD. Moreover, the study discovered that CKD itself may causatively lead to alterations in certain proteins, including CST5(OR = 1.16, 95% CI = 1.09-1.24, p < 0.001, FDR = 0.001). No evidence of reverse causality was found for any single biomarker and CKD. This comprehensive MR investigation supports a genetic causal nexus between certain immune cell subtypes, inflammatory proteins, and CKD. These findings enhance the understanding of CKD's immunological underpinnings and open avenues for targeted treatments.
Collapse
Affiliation(s)
- Yongzheng Hu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengyun Hao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qian An
- Department of Nephrology, Qingdao Central Hospital, Qingdao, Shandong, China
| | - Wei Jiang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
5
|
Ningoo M, Cruz-Encarnación P, Khilnani C, Heeger PS, Fribourg M. T-cell receptor sequencing reveals selected donor-reactive CD8 + T cell clones resist antithymocyte globulin depletion after kidney transplantation. Am J Transplant 2024; 24:755-764. [PMID: 38141722 PMCID: PMC11070313 DOI: 10.1016/j.ajt.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/21/2023] [Accepted: 12/19/2023] [Indexed: 12/25/2023]
Abstract
High frequencies of donor-reactive memory T cells in the periphery of transplant candidates prior to transplantation are linked to the development of posttransplant acute rejection episodes and reduced allograft function. Rabbit antithymocyte globulin (rATG) effectively depletes naïve CD4+ and CD8+ T cells for >6 months posttransplant, but rATG's effects on human donor-reactive T cells have not been carefully determined. To address this, we performed T cell receptor β-chain sequencing on peripheral blood mononuclear cells aliquots collected pretransplant and serially posttransplant in 7 kidney transplant recipients who received rATG as induction therapy. We tracked the evolution of the donor-reactive CD4+ and CD8+ T cell repertoires and identified stimulated pretransplant, CTV-(surface dye)-labeled, peripheral blood mononuclear cells from each patient with donor cells or third-party cells. Our analyses showed that while rATG depleted CD4+ T cells in all tested subjects, a subset of donor-reactive CD8+ T cells that were present at high frequencies pretransplant, consistent with expanded memory cells, resisted rATG depletion, underwent posttransplant expansion and were functional. Together, our data support the conclusion that a subset of human memory CD8+ T cells specifically reactive to donor antigens expand in vivo despite induction therapy with rATG and thus have the potential to mediate allograft damage.
Collapse
Affiliation(s)
- Mehek Ningoo
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pamela Cruz-Encarnación
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Calla Khilnani
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter S Heeger
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Miguel Fribourg
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
6
|
Jallah BP, Kuypers DRJ. Impact of Immunosenescence in Older Kidney Transplant Recipients: Associated Clinical Outcomes and Possible Risk Stratification for Immunosuppression Reduction. Drugs Aging 2024; 41:219-238. [PMID: 38386164 DOI: 10.1007/s40266-024-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2024] [Indexed: 02/23/2024]
Abstract
The number of older individuals receiving a kidney transplant as replacement therapy has significantly increased in the past decades and this increase is expected to continue. Older patients have a lower rate of acute rejection but an increased incidence of death with a functioning graft. Several factors, including an increased incidence of infections, post-transplant malignancy and cardiovascular comorbidity and mortality, contribute to this increased risk. Notwithstanding, kidney transplantation is still the best form of kidney replacement therapy in all patients with chronic kidney disease, including in older individuals. The best form of immunosuppression and the optimal dose of these medications in older recipients remains a topic of discussion. Pharmacological studies have usually excluded older patients and when included, patients were highly selected and their numbers insignificant to draw a reasonable conclusion. The reduced incidence of acute rejection in older recipients has largely been attributed to immunosenescence. Immunosenescence refers to the aging of the innate and adaptive immunity, accumulating in phenotypic and functional changes. These changes influences the response of the immune system to new challenges. In older individuals, immunosenescence is associated with increased susceptibility to infectious pathogens, a decreased response after vaccinations, increased risk of malignancies and cardiovascular morbidity and mortality. Chronic kidney disease is associated with premature immunosenescent changes, and these are independent of aging. The immunosenescent state is associated with low-grade sterile inflammation termed inflammaging. This chronic low-grade inflammation triggers a compensatory immunosuppressive state to avoid further tissue damage, leaving older individuals with chronic kidney disease in an immune-impaired state before kidney transplantation. Immunosuppression after transplantation may further enhance progression of this immunosenescent state. This review covers the role of immunosenescence in older kidney transplant recipients and it details present knowledge of the changes in chronic kidney disease and after transplantation. The impact of immunosuppression on the progression and complications of an immunosenescent state are discussed, and the future direction of a possible clinical implementation of immunosenescence to individualize/reduce immunosuppression in older recipients is laid out.
Collapse
Affiliation(s)
- Borefore P Jallah
- Department of Nephrology and Renal Transplantation, University Hospital Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Dirk R J Kuypers
- Department of Nephrology and Renal Transplantation, University Hospital Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of Microbiology, Immunology and Transplantation, University of Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Betjes MGH, Kal-van Gestel J, Roodnat JI, de Weerd AE. The Incidence of Antibody-Mediated Rejection Is Age-Related, Plateaus Late After Kidney Transplantation, and Contributes Little to Graft Loss in the Older Recipients. Transpl Int 2023; 36:11751. [PMID: 38188697 PMCID: PMC10768842 DOI: 10.3389/ti.2023.11751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
It is not known whether antibody-mediated rejection (ABMR) is age-related, whether it plateaus late after transplantation, and to what extent it contributes to graft loss in older recipients. Patients transplanted between 2010 and 2015 (n = 1,054) in a single center had regular follow-up until January 2023. Recipients were divided into age groups at transplantation: 18-39 years ("young"), 40-55 years ("middle age"), and >55 years ("elderly"). Ten years after transplantation the cumulative % of recipients with ABMR was 17% in young, 15% in middle age, and 12% in elderly recipients (p < 0.001). The cumulative incidence of ABMR increased over time and plateaued 8-10 years after transplantation. In the elderly, with a median follow-up of 7.5 years, on average 30% of the recipients with ABMR died with a functional graft and ABMR contributed only 4% to overall graft loss in this group. These results were cross-validated in a cohort of recipients with >15 years follow-up. Multivariate cox-regression analysis showed that increasing recipient age was independently associated with decreasing risk for ABMR. In conclusion, the cumulative risk for ABMR is age-dependent, plateaus late after transplantation, and contributes little to overall graft loss in older recipients.
Collapse
Affiliation(s)
- Michiel G. H. Betjes
- Rotterdam Transplantation Institute, Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
8
|
Cremen S, Santiago RM, Robinson MW, Gallagher TK. Biomarkers of biological aging in recipients of solid organ transplantation and clinical outcomes: A scoping review. Transpl Immunol 2023; 79:101851. [PMID: 37182719 DOI: 10.1016/j.trim.2023.101851] [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: 11/21/2022] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Biological aging is the accumulation of cellular and molecular damage within an individual over time. The biological age of a donor organ is known to influence clinical outcomes of solid organ transplantation, including delayed graft function and frequency of rejection episodes. While much research has focused on the biological age of donor organs, the recipient's biological age may also influence transplantation outcomes. The aim of this scoping review was to identify and provide an overview of the existing evidence regarding biological aging in solid organ transplant recipients and the impact on patient outcomes post-transplant. METHODS Literature searches were carried out on PubMed, Web of Science, Google Scholar, Embase and TRIP using the phrases 'solid organ transplant', 'cell senescence', 'cell aging' and 'outcomes', using boolean 'and/or' phrases and MeSH terms. Duplicates were removed and abstracts were reviewed by two independent reviewers. Full papers were then screened for inclusion by two reviewers. Data extraction was carried out using a standardised proforma agreed on prior to starting. RESULTS 32 studies, including data on a total of 7760 patients, were identified for inclusion in this review; 23 relating to kidney transplant recipients, three to liver transplant, five to lung transplant and one to heart transplantation. A wide range of biomarkers of biological aging have been assessed in kidney transplant recipients, whereas studies of liver, lung and heart transplant have predominantly assessed recipient telomere length. The most robust associations with clinical outcomes are observed in kidney transplant recipients, possibly influenced by the larger number of studies and the use of a wider range of biomarkers of biological aging. In kidney transplant recipients reduced thymic function and accumulation of terminally differentiated T cell populations was associated with reduced risk of acute rejection but increased risk of infection and mortality. CONCLUSION Studies to date on biological aging in transplant recipients have been heavily biased to kidney transplant recipients. The results from these studies suggest recipient biological age can influence clinical outcomes and future research is needed to prioritise robust biomarkers of biological aging in transplant recipients.
Collapse
Affiliation(s)
- S Cremen
- Department of Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - R M Santiago
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Kildare, Ireland
| | - M W Robinson
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Kildare, Ireland.
| | - T K Gallagher
- Department of Hepatobiliary and Transplant Surgery, St Vincent's University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Betjes MGH, De Weerd A. Lowering maintenance immune suppression in elderly kidney transplant recipients; connecting the immunological and clinical dots. Front Med (Lausanne) 2023; 10:1215167. [PMID: 37502354 PMCID: PMC10368955 DOI: 10.3389/fmed.2023.1215167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/09/2023] [Indexed: 07/29/2023] Open
Abstract
The management of long-term immune suppressive medication in kidney transplant recipients is a poorly explored field in the area of transplant medicine. In particular, older recipients are at an increased risk for side effects and have an exponentially increased risk of infection-related death. In contrast, an aged immune system decreases the risk of acute T-cell-mediated rejection in older recipients. Recent advances in alloimmunity research have shown a rapid and substantial decline in polyfunctional, high-risk CD4+ T cells post-transplantation. This lowers the direct alloreactivity responsible for T-cell-mediated rejection, also known as donor-specific hyporesponsiveness. Chronic antibody-mediated rejection (c-aABMR) is the most frequent cause of kidney graft loss in the long term. However, in older adults, c-aABMR as a cause of graft loss is outnumbered by death with a functioning graft. In addition, DSA development and a diagnosis of c-aABMR plateau ~10 years after transplantation, resulting in a very low risk for rejection thereafter. The intensity of immune suppression regimes could likely be reduced accordingly, but trials in this area are scarce. Tacrolimus monotherapy for 1 year after transplantation seems feasible in older kidney transplant recipients with standard immunological risk, showing the expected benefits of fewer infections and better vaccination responses.
Collapse
|
10
|
Lacquaniti A, Campo S, Falliti G, Caruso D, Gargano R, Giunta E, Monardo P. Free Light Chains, High Mobility Group Box 1, and Mortality in Hemodialysis Patients. J Clin Med 2022; 11:jcm11236904. [PMID: 36498479 PMCID: PMC9739300 DOI: 10.3390/jcm11236904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/15/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Uremic toxins are associated with immune dysfunction and inflammation. The inadequate removal by hemodialysis (HD) of serum free light chains (FLCs) determines their accumulation. This study evaluated FLCs in HD patients, analyzing their relations with other biomarkers, such as serum high mobility group box 1 (HMGB1). Methods: FLC and HMGB1 were evaluated in a cohort of 119 HD patients. κFLC and λFLC were summated to give a combined (c) FLC concentration. Patients were followed prospectively until the end of the observation period of four years, or until the endpoint: the patient’s death. Results: cFLC values in HD patients were 244.4 (197.9−273.5) mg/L. We detected a significant reduction in CD8+ cells and a decreased CD4+/CD8+ ratio. HMGB1 levels were 94.5 (55−302) pg/mL. After multivariate analysis, cFLCs correlated with β2-microglobulin and the CD4+/CD8+ ratio. Subjects with cFLC values above 263 mg/L and with sHMGB1 values < 80 pg/mL experienced a significantly faster evolution to the endpoint (mean follow-up time to progression of 27.5 and 28.5 months, respectively; p < 0.001). After an adjusted multivariate Cox analysis, cFLCs were associated with 11% increased risk of death, whereas low sHMGB1 increased this risk by 5%. Conclusions: cFLCs and HMGB1 reflect the inflammation and immune dysfunction in HD patients representing two strong and independent risk markers of mortality.
Collapse
Affiliation(s)
| | - Susanna Campo
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy
| | | | - Daniele Caruso
- Clinical Pathology Unit, Papardo Hospital, 98158 Messina, Italy
| | - Romana Gargano
- Department of Economics, University of Messina, 98122 Messina, Italy
| | - Elena Giunta
- Microbiology and Virology Unit, Papardo Hospital, 98158 Messina, Italy
| | - Paolo Monardo
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy
- Correspondence: ; Tel.: +39-090-3996062; Fax: +39-090-3992337
| |
Collapse
|
11
|
Cai X, Li H, Wang M, Chu E, Wei N, Lin J, Hu Y, Dai J, Chen A, Zheng H, Zhang Q, Zhong Y, Chang R, Wu S, Xiao Y, Liu C. mTOR Participates in the Formation, Maintenance, and Function of Memory CD8 +T Cells Regulated by Glycometabolism. Biochem Pharmacol 2022; 204:115197. [PMID: 35926651 DOI: 10.1016/j.bcp.2022.115197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022]
Abstract
Memory CD8+T cells participate in the fight against infection and tumorigenesis as well as in autoimmune disease progression because of their efficient and rapid immune response, long-term survival, and continuous differentiation. At each stage of their formation, maintenance, and function, the cell metabolism must be adjusted to match the functional requirements of the specific stage. Notably, enhanced glycolytic metabolism can generate sufficient levels of adenosine triphosphate (ATP) to form memory CD8+T cells, countering the view that glycolysis prevents the formation of memory CD8+T cells. This review focuses on how glycometabolism regulates memory CD8+T cells and highlights the key mechanisms through which the mammalian target of rapamycin (mTOR) signaling pathway affects memory CD8+T cell formation, maintenance, and function by regulating glycometabolism. In addition, different subpopulations of memory CD8+T cells exhibit different metabolic flexibility during their formation, survival, and functional stages, during which the energy metabolism may be critical. These findings which may explain why enhanced glycolytic metabolism can give rise to memory CD8+T cells. Modulating the metabolism of memory CD8+T cells to influence specific cell fates may be useful for disease treatment.
Collapse
Affiliation(s)
- Xuepei Cai
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Haokun Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Manyi Wang
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Edward Chu
- Department of Oncology and Cancer Therapeutics Program, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ning Wei
- Department of Oncology and Cancer Therapeutics Program, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jiayu Lin
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yun Hu
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jingtao Dai
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Aijie Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hua Zheng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuxia Zhong
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoshui Chang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory of Functional Proteomics of Guangdong Province, Guangzhou, China; National Demonstration Center for Experimental Education of Basic Medical Sciences of China, Guangzhou, China.
| | - Yaomu Xiao
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Chufeng Liu
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Betjes MGH, Roelen DL, van Agteren M, Kal-van Gestel J. Causes of Kidney Graft Failure in a Cohort of Recipients With a Very Long-Time Follow-Up After Transplantation. Front Med (Lausanne) 2022; 9:842419. [PMID: 35733857 PMCID: PMC9207199 DOI: 10.3389/fmed.2022.842419] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/05/2022] [Indexed: 01/03/2023] Open
Abstract
Background Biopsy-proven causes of graft loss many years after kidney transplantation are scarcely documented. Methods Patients transplanted between 1995 and 2005 (n = 737) in a single center were followed on a regular basis until 2021. The recipients were divided according to age at transplantation into 3 groups; 18–39 years (young), 40–55 years (middle age), and older than 55 years (elderly). For cause biopsies of renal transplants were clustered into the categories, rejection, IFTA, return original disease, and diagnosis of de novo kidney disease. Results Rejection was the main cause of graft failure censored for death at every time period after transplantation. The incidence of T cell-mediated rejection (TCMR) became rare 6 years after transplantation while the cumulative incidence of antibody-mediated rejection (ABMR) increased over time (1.1% per year). ABMR was not diagnosed anymore beyond 15 years of follow-up in recipients without pre-transplant donor-specific antibodies (DSA). An episode of TCMR was associated with an increased incidence of ABMR diagnosis in the short-term but did not increase the overall incidence of AMBR not in the long-term. Death as a cause of graft failure was an important competitive risk factor long after transplantation and resulted in a significantly lower frequency of rejection-related graft loss in the elderly group (11 vs. 23% in the young group at 15 year follow-up). Conclusion Rejection is a major cause of graft loss but recipient’s age, time after transplantation, and the presence of DSA before transplantation determine the relative contribution to overall graft loss and the type of rejection involved.
Collapse
Affiliation(s)
- Michiel G. H. Betjes
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
- *Correspondence: Michiel G. H. Betjes, ; orcid.org/0000-0001-9435-6208
| | - Dave L. Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Madelon van Agteren
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
| | - Judith Kal-van Gestel
- Department of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam Transplantation Institute, Rotterdam, Netherlands
| |
Collapse
|
13
|
Coppo L, Mishra P, Siefert N, Holmgren A, Pääbo S, Zeberg H. A substitution in the glutathione reductase lowers electron leakage and inflammation in modern humans. SCIENCE ADVANCES 2022; 8:eabm1148. [PMID: 34985944 PMCID: PMC8730399 DOI: 10.1126/sciadv.abm1148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Glutathione reductase is a critical enzyme for preventing oxidative stress and maintaining a reduced intracellular environment. Almost all present-day humans carry an amino acid substitution (S232G) in this enzyme relative to apes and Neanderthals. We express the modern human and the ancestral enzymes and show that whereas the activity and stability are unaffected by the amino acid substitution, the ancestral enzyme produces more reactive oxygen species and increases cellular levels of transcripts encoding cytokines. We furthermore show that the ancestral enzyme has been reintroduced into the modern human gene pool by gene flow from Neanderthals and is associated with multiple traits in present-day people, including increased susceptibility for inflammatory-associated disorders and vascular disease.
Collapse
Affiliation(s)
- Lucia Coppo
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165 Stockholm, Sweden
- Corresponding author. (L.C.); (S.P.); (H.Z.)
| | - Pradeep Mishra
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Nora Siefert
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Arne Holmgren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany
- Okinawa Institute of Science and Technology, Onna-son, Okinawa 904-0495, Japan
- Corresponding author. (L.C.); (S.P.); (H.Z.)
| | - Hugo Zeberg
- Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany
- Department of Neuroscience, Karolinska Institutet, SE-17165 Stockholm, Sweden
- Corresponding author. (L.C.); (S.P.); (H.Z.)
| |
Collapse
|
14
|
Abstract
Involvement of T lymphocytes in kidney transplantation is a well-developed topic; however, most of the scientific interest focused on the different type of CD4+ lymphocyte subpopulations. Few authors, instead, investigated the role of CD8+ T cells in renal transplantation and how deleterious they can be to long-term allograft survival. Recently, there has been a renewed interest in the CD8+ T cells involvement in the transplantation field with the aim to investigate the immunological mechanisms underlying the infiltration of CD8+ T cells and their biological functions in human kidney allografts. The purpose of the present review is to highlight the role of allo-reactive cytotoxic T lymphocytes (CTLs) CD8+ subset in allograft kidney recipients and their related clinical complications.
Collapse
|
15
|
Abstract
Immunologic memory is the ability of adaptive immune system to quickly and specifically recognize previously encountered antigens and initiate an effector response. Alloreactive memory cells can mount rapid and robust responses to the transplanted organ resulting in allograft injury. Thus preexisting humoral or cellular memory alloresponses are typically associated with poor graft outcomes in experimental and clinical transplantation. While both B and T lymphocytes exhibit memory responses, this review discusses recent updates on the biology of memory T cells and their relevance to the field of transplantation. Three major areas of focus are the emergence and characterization of tissue resident memory T cells, manipulation of T cell metabolic pathways, and the latest promising approaches to targeting detrimental T cell memory in the settings of organ transplantation.
Collapse
|
16
|
Courivaud C, Bamoulid J, Crepin T, Gaiffe E, Laheurte C, Saas P, Ducloux D. Pre-transplant Thymic Function Predicts Is Associated With Patient Death After Kidney Transplantation. Front Immunol 2020; 11:1653. [PMID: 32903778 PMCID: PMC7438875 DOI: 10.3389/fimmu.2020.01653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Accelerated thymic involution is a main feature of end-stage renal disease (ESRD)-associated immune senescence. Recent evidences suggest that ESRD-associated immune senescence is associated with adverse outcomes in dialysis patients. However, no study focused on the association between pre-transplant thymic function and patient survival after transplantation. We conducted a prospective, multicenter study to assess whether pre-transplant thymic function measured by recent thymic emigrants (RTE) may predict death after first kidney transplantation. Results were tested in a validation cohort. Nine hundred and sixty-seven incident kidney transplant recipients were included in the prospective study. Mean follow up was 5.1 + 2.9 years. Eighty two patients (8.5%) died during follow up. Lower RTE levels were associated with a higher risk of death (2.53; 95%CI, 1.54–4.39 for each decrease of 1 log in RTE; p < 0.001). Cancer-related death was particularly increased in patients with low RTE levels (4.23; 95%CI, 1.43–12.13; p = 0.007). One hundred and thirty-six patients having received a first kidney transplantation were included in the validation cohort. Lower TREC levels were associated with higher risk of death (1.90; 95%CI, 1.11–3.51 for each decrease of 1 log in RTE; p = 0.025). RTE were not associated with death-censored graft loss. Pre-transplant thymic function is strongly associated with death after transplantation. Attempt to reverse ESRD-related thymic loss may prevent premature death.
Collapse
Affiliation(s)
- Cécile Courivaud
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Jamal Bamoulid
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Thomas Crepin
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Emilie Gaiffe
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| | - Caroline Laheurte
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Philippe Saas
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Didier Ducloux
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| |
Collapse
|
17
|
Ko EJ, Seo JW, Kim KW, Kim BM, Cho JH, Kim CD, Seok J, Yang CW, Lee SH, Chung BH. Phenotype and molecular signature of CD8+ T cell subsets in T cell- mediated rejections after kidney transplantation. PLoS One 2020; 15:e0234323. [PMID: 32530943 PMCID: PMC7292394 DOI: 10.1371/journal.pone.0234323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/24/2020] [Indexed: 01/08/2023] Open
Abstract
We investigated the phenotype and molecular signatures of CD8+ T cell subsets in kidney-transplant recipients (KTRs) with biopsy-proven T cell-mediated rejection (TCMR). We included 121 KTRs and divided them into three groups according to the pathologic or clinical diagnosis: Normal biopsy control (NC)(n = 32), TCMR (n = 50), and long-term graft survival (LTGS)(n = 39). We used flowcytometry and microarray to analyze the phenotype and molecular signatures of CD8+ T cell subsets using peripheral blood from those patients and analyzed significant gene expressions according to CD8+ T cell subsets. We investigated whether the analysis of CD8+ T cell subsets is useful for predicting the development of TCMR. CCR7+CD8+ T cells significantly decreased, but CD28nullCD57+CD8+ T cells and CCR7-CD45RA+CD8+ T cells showed an increase in the TCMR group compared to other groups (p<0.05 for each); hence CCR7+CD8+ T cells showed significant negative correlations to both effector CD8+ T cells. We identified genes significantly associated with the change of CCR7+CD8+ T, CCR7-CD45RA+CD8+ T, and CD28nullCD57+CD8+ T cells in an ex vivo study and found that most of them were included in the significant genes on in vitro CCR7+CD8+ T cells. Finally, the decrease of CCR7+CD8+ T cells relative to CD28nullCD57+ T or CCR7-CD45RA+CD8+ T cells can predict TCMR significantly in the whole clinical cohort. In conclusion, phenotype and molecular signature of CD8+ T subsets showed a significant relationship to the development of TCMR; hence monitoring of CD8+ T cell subsets may be a useful for predicting TCMR in KTRs.
Collapse
Affiliation(s)
- Eun Jeong Ko
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung-Woo Seo
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Kyoung Woon Kim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bo-Mi Kim
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Junhee Seok
- School of Electrical Engineering, Korea University, Seoul, South Korea
| | - Chul Woo Yang
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang-Ho Lee
- Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Byung Ha Chung
- Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
18
|
Uremia-Associated Ageing of the Thymus and Adaptive Immune Responses. Toxins (Basel) 2020; 12:toxins12040224. [PMID: 32260178 PMCID: PMC7232426 DOI: 10.3390/toxins12040224] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
Progressive loss of renal function is associated with a series of changes of the adaptive immune system which collectively constitute premature immunological ageing. This phenomenon contributes significantly to the mortality and morbidity of end-stage renal disease (ESRD) patients. In this review, the effect of ESRD on the T cell part of the adaptive immune system is highlighted. Naïve T cell lymphopenia, in combination with the expansion of highly differentiated memory T cells, are the hallmarks of immunological ageing. The decreased production of newly formed T cells by the thymus is critically involved. This affects both the CD4 and CD8 T cell compartment and may contribute to the expansion of memory T cells. The expanding populations of memory T cells have a pro-inflammatory phenotype, add to low-grade inflammation already present in ESRD patients and destabilize atherosclerotic plaques. The effect of loss of renal function on the thymus is not reversed after restoring renal function by kidney transplantation and constitutes a long-term mortality risk factor. Promising results from animal experiments have shown that rejuvenation of the thymus is a possibility, although not yet applicable in humans.
Collapse
|
19
|
High numbers of differentiated CD28null CD8+ T cells are associated with a lowered risk for late rejection and graft loss after kidney transplantation. PLoS One 2020; 15:e0228096. [PMID: 32023273 PMCID: PMC7001918 DOI: 10.1371/journal.pone.0228096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/07/2020] [Indexed: 12/29/2022] Open
Abstract
Background The hypothesis was tested that parameters of an aged T-cell compartment associate with the risk for late rejection after kidney transplantation. Methods Recipients of a kidney transplant in the period 2007–2013 were (N = 365) were included. T cells were characterized prior to transplantation by flow cytometry as naive (CD45RO-CCR7+), central-memory (CD45RO+CCR7+), effector-memory (CD45RO-CCR7-) or terminally differentiated CD8+ Temra (CD45RO-/CCR7-/CD28-) cells. T cell telomere length and thymic output were assessed prior to transplantation in 202 recipients. Follow-up was until December 2018. The date of the first time of biopsy-proven late rejection (>6 months after transplantation) was used to calculate the rejection-free survival time. Results Fifty cases of biopsy-proven rejection were recorded. Thymic output and T cell telomere length did not associate with late rejection-free survival. However, the percentage and absolute numbers of CD8+Temra and CD28null CD8+ T cells were significantly lower in patients with late rejection. Specifically, in the highest tertile of percentages of CD28null CD8+ T cells, the cumulative incidence of late rejection at 5 and 10 years was only 5% and 8% compared to 16% and 20% in the middle to lowest tertile (p = 0.002). Multivariate proportional hazard analysis showed that percentage and absolute number of CD28null CD8+ T cells remained significantly associated with late rejection and rejection-related graft loss. Conclusion High numbers of differentiated CD28null CD8+ T cells decrease the risk for late rejection and rejection-related graft loss after kidney transplantation.
Collapse
|
20
|
Hartigan CR, Sun H, Ford ML. Memory T‐cell exhaustion and tolerance in transplantation. Immunol Rev 2019; 292:225-242. [DOI: 10.1111/imr.12824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Affiliation(s)
| | - He Sun
- Emory Transplant Center and Department of Surgery Emory University Atlanta GA USA
- Department of Hepatobiliary Surgery and Transplantation The First Hospital of China Medical University Shenyang China
| | - Mandy L. Ford
- Emory Transplant Center and Department of Surgery Emory University Atlanta GA USA
| |
Collapse
|
21
|
Nakid-Cordero C, Arzouk N, Gauthier N, Tarantino N, Larsen M, Choquet S, Burrel S, Autran B, Vieillard V, Guihot A. Skewed T cell responses to Epstein-Barr virus in long-term asymptomatic kidney transplant recipients. PLoS One 2019; 14:e0224211. [PMID: 31639143 PMCID: PMC6804993 DOI: 10.1371/journal.pone.0224211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/08/2019] [Indexed: 01/06/2023] Open
Abstract
Kidney transplant recipients (KTRs) abnormally replicate the Epstein Barr Virus (EBV). To better understand how long-term immunosuppression impacts the immune control of this EBV re-emergence, we systematically compared 10 clinically stable KTRs to 30 healthy controls (HCs). The EBV-specific T cell responses were determined in both groups by multiparameter flow cytometry with intra cellular cytokine staining (KTRs n = 10; HCs n = 15) and ELISpot-IFNγ assays (KTRs n = 7; HCs n = 7). The T/B/NK cell counts (KTRs n = 10; HCs n = 30) and the NK/T cell differentiation and activation phenotypes (KTRs n = 10; HCs n = 15/30) were also measured. We show that in KTRs, the Th1 effector CD4+ T cell responses against latent EBV proteins are weak (2/7 responders). Conversely, the frequencies total EBV-specific CD8+T cells are conserved in KTRs (n = 10) and span a wider range of EBNA-3A peptides (5/7responders) than in HCs (5/7responders). Those modifications of the EBV-specific T cell response were associated with a profound CD4+ T cell lymphopenia in KTRs compared to HCs, involving the naïve CD4+ T cell subset, and a persistent activation of highly-differentiated senescent CD8+ T cells. The proportion of total NK / CD8+ T cells expressing PD-1 was also increased in KTRs. Noteworthy, PD-1 expression on CD8+ T cells normalized with time after transplantation. In conclusion, we show modifications of the EBV-specific cellular immunity in long term transplant recipients. This may be the result of both persistent EBV antigenic stimulation and profound immunosuppression induced by anti-rejection treatments. These findings provide new insights into the immunopathology of EBV infection after renal transplantation.
Collapse
Affiliation(s)
- Cecilia Nakid-Cordero
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Nadia Arzouk
- Service de Néphrologie, Urologie et Transplantation Rénale, Hôpital Pitié Salpêtrière, Paris, France
| | - Nicolas Gauthier
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Nadine Tarantino
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Martin Larsen
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sylvain Choquet
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Service d’Hématologie, Hôpital Pitié Salpêtrière, Paris, France
| | - Sonia Burrel
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Service de Virologie, Hôpital Pitié Salpêtrière, Paris, France
| | - Brigitte Autran
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Vincent Vieillard
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- CNRS ERL8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Amélie Guihot
- Sorbonne Université (Univ. Paris 06), INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Département d’Immunologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| |
Collapse
|
22
|
DeWolfe D, Aid M, McGann K, Ghofrani J, Geiger E, Helzer C, Malik S, Kleiboeker S, Jost S, Tan CS. NK Cells Contribute to the Immune Risk Profile in Kidney Transplant Candidates. Front Immunol 2019; 10:1890. [PMID: 31507586 PMCID: PMC6716214 DOI: 10.3389/fimmu.2019.01890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/26/2019] [Indexed: 01/03/2023] Open
Abstract
Background: A previously proposed immune risk profile (IRP), based on T cell phenotype and CMV serotype, is associated with mortality in the elderly and increased infections post-kidney transplant. To evaluate if NK cells contribute to the IRP and if the IRP can be predicted by a clinical T cell functional assays, we conducted a cross sectional study in renal transplant candidates to determine the incidence of IRP and its association with specific NK cell characteristics and ImmuKnow® value. Material and Methods: Sixty five subjects were enrolled in 5 cohorts designated by age and dialysis status. We determined T and NK cell phenotypes by flow cytometry and analyzed multiple factors contributing to IRP. Results: We identified 14 IRP+ [CMV seropositivity and CD4/CD8 ratio < 1 or being in the highest quintile of CD8+ senescent (28CD–/CD57+) T cells] individuals equally divided amongst the cohorts. Multivariable linear regression revealed a distinct IRP+ group. Age and dialysis status did not predict immune senescence in kidney transplant candidates. NK cell features alone could discriminate IRP– and IRP+ patients, suggesting that NK cells significantly contribute to the overall immune status in kidney transplant candidates and that a combined T and NK cell phenotyping can provide a more detailed IRP definition. ImmuKnow® value was negatively correlated to age and significantly lower in IRP+ patients and predicts IRP when used alone or in combination with NK cell features. Conclusion: NK cells contribute to overall immune senescence in kidney transplant candidates.
Collapse
Affiliation(s)
- David DeWolfe
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Malika Aid
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Kevin McGann
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Joshua Ghofrani
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Emma Geiger
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Catherine Helzer
- Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Shaily Malik
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | | | - Stephanie Jost
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Chen Sabrina Tan
- Center for Virology and Vaccines Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Sablik KA, Litjens NH, Klepper M, Betjes MG. Increased CD16 expression on NK cells is indicative of antibody-dependent cell-mediated cytotoxicity in chronic-active antibody-mediated rejection. Transpl Immunol 2019; 54:52-58. [DOI: 10.1016/j.trim.2019.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 01/01/2023]
|
24
|
Mirzakhani M, Shahbazi M, Oliaei F, Mohammadnia-Afrouzi M. Immunological biomarkers of tolerance in human kidney transplantation: An updated literature review. J Cell Physiol 2018; 234:5762-5774. [PMID: 30362556 DOI: 10.1002/jcp.27480] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
The half-life of transplanted kidneys is <10 years. Acute or chronic rejections have a negative impact on transplant outcome. Therefore, achieving to allograft tolerance for improving long-term transplant outcome is a desirable goal of transplantation field. In contrast, there are evidence that distinct immunological characteristics lead to tolerance in some transplant recipients. In contrast, the main reason for allograft loss is immunological responses. Various immune cells including T cells, B cells, dendritic cells, macrophages, natural killer, and myeloid-derived suppressor cells damage graft tissue and, thereby, graft loss happens. Therefore, being armed with the comprehensive knowledge about either preimmunological or postimmunological characteristics of renal transplant patients may help us to achieve an operational tolerance. In the present study, we are going to review and discuss immunological characteristics of renal transplant recipients with rejection and compare them with tolerant subjects.
Collapse
Affiliation(s)
- Mohammad Mirzakhani
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran.,Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Shahbazi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Farshid Oliaei
- Kidney Transplantation Center, Shahid Beheshti Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Mousa Mohammadnia-Afrouzi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
25
|
Díaz-Molina B, Diaz-Bulnes P, Carvajal Palao R, Bernardo MJ, Rodriguez RM, Corte-Iglesias V, Moris de la Tassa C, Lambert JL, Suarez-Alvarez B. Early Everolimus Initiation Fails to Counteract the Cytotoxic Response Mediated by CD8 + T and NK Cells in Heart Transplant Patients. Front Immunol 2018; 9:2181. [PMID: 30319636 PMCID: PMC6168668 DOI: 10.3389/fimmu.2018.02181] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/04/2018] [Indexed: 12/22/2022] Open
Abstract
The positive long-term effects of conversion to everolimus (EVL) after heart transplantation (HT) have been evaluated in several studies. However, the timing of EVL initiation, the best way to combine it with other immunosuppressive treatments, and the impact of these combinations on the immune response are poorly understood aspects. Here, we analyzed the immune phenotype and function of HT patients (n = 56) at short and long terms (prospective and retrospective cohorts), taking into account the time of EVL initiation: early (3 months post-transplant, EVL-E group) or late (>1 year post-transplant, EVL-L group) compared with mycophenolate mofetil treatment (MMF group). We show that early EVL conversion from MMF allows the increase of cytotoxic (CD56dim CD16+) NK and effector-memory (EM, CD45RA− CCR7−) CD8+ T cell subsets, which show a significantly higher level of expression of cytotoxic molecules, IFN-γ production and degranulation ability under activation. NK cell expansion is accompanied by an altered balance of receptor expression, increasing the activation state, and lytic activity of those cells. Those changes are detected after as little as 1 month after EVL conversion in association with the expansion of regulatory T cells and the decrease in B cell frequency. However, no changes in the immune cells subsets were observed after late EVL initiation (EVL-L) compared with the MMF group. Our results imply that only early EVL conversion induces key changes in the post-transplant immune response, preserving an efficient anti-viral response, but simultaneously showing a limited ability to counteract the cytotoxic response to the allograft.
Collapse
Affiliation(s)
- Beatriz Díaz-Molina
- Advanced Heart Failure and Transplant Service, Department of Cardiology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Paula Diaz-Bulnes
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Reyes Carvajal Palao
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Maria José Bernardo
- Advanced Heart Failure and Transplant Service, Department of Cardiology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ramón M Rodriguez
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Jose Luis Lambert
- Advanced Heart Failure and Transplant Service, Department of Cardiology, Hospital Universitario Central de Asturias, Oviedo, Spain.,Faculty of Health Sciences, Universidad Católica San Antonio de Murcia, Murcia, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
26
|
Singh MD, Ni M, Sullivan JM, Hamerman JA, Campbell DJ. B cell adaptor for PI3-kinase (BCAP) modulates CD8 + effector and memory T cell differentiation. J Exp Med 2018; 215:2429-2443. [PMID: 30093532 PMCID: PMC6122975 DOI: 10.1084/jem.20171820] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/13/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
Singh et al. show that expression of B cell adaptor for PI3-kinase (BCAP) is induced upon T cell activation and that this helps control effector and memory CD8+ T cell differentiation. CD8+ T cells respond to signals via the T cell receptor (TCR), costimulatory molecules, and immunoregulatory cytokines by developing into diverse populations of effector and memory cells. The relative strength of phosphoinositide 3-kinase (PI3K) signaling early in the T cell response can dramatically influence downstream effector and memory T cell differentiation. We show that initial PI3K signaling during T cell activation results in up-regulation of the signaling scaffold B cell adaptor for PI3K (BCAP), which further potentiates PI3K signaling and promotes the accumulation of CD8+ T cells with a terminally differentiated effector phenotype. Accordingly, BCAP-deficient CD8+ T cells have attenuated clonal expansion and altered effector and memory T cell development following infection with Listeria monocytogenes. Thus, induction of BCAP serves as a positive feedback circuit to enhance PI3K signaling in activated CD8+ T cells, thereby acting as a molecular checkpoint regulating effector and memory T cell development.
Collapse
Affiliation(s)
- Mark D Singh
- Immunology Program, Benaroya Research Institute, Seattle, WA
| | - Minjian Ni
- Immunology Program, Benaroya Research Institute, Seattle, WA
| | - Jenna M Sullivan
- Immunology Program, Benaroya Research Institute, Seattle, WA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jessica A Hamerman
- Immunology Program, Benaroya Research Institute, Seattle, WA.,Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Daniel J Campbell
- Immunology Program, Benaroya Research Institute, Seattle, WA .,Department of Immunology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
27
|
Ducloux D, Legendre M, Bamoulid J, Rebibou JM, Saas P, Courivaud C, Crepin T. ESRD-associated immune phenotype depends on dialysis modality and iron status: clinical implications. Immun Ageing 2018; 15:16. [PMID: 30026783 PMCID: PMC6050655 DOI: 10.1186/s12979-018-0121-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND End-stage renal disease (ESRD) causes premature ageing of the immune system. However, it is not known whether hemodialysis (HD) and peritoneal dialysis (PD) similarly affect the T cell system. METHODS The aim of our study was to analyse whether dialysis modality may mitigate ESRD-induced immune senescence. We explored a large population of patients (675 ESRD patients) and both confirmed and refined the results in a second cohort (84 patients). RESULTS HD patients exhibited higher inflammatory monocytes counts (44/mm3 (1-520) vs 36/mm3 (1-161); p = 0.005). Patients on HD also had higher frequency of CD8 T cells (24% (7-61) vs 22% (8-42); p = 0.003) and reduced CD4/CD8 ratio. Such results were confirmed in the second cohort. Moreover, both CD4 + CD57 + CD28- (3.25% (0-38.2) vs 1.05% (0-28.5); p = 0.068) and CD8 + CD57 + CD28- (38.5% (3.6-76.8) vs 26.1 (2.1-46.9); p = 0.039) T cells frequencies were increased in HD patients. Telomere length did not differ according to dialysis modality, but was inversely related to ferritin levels (r = - 0.33; p = 0.003). There was a trend towards higher telomerase activity in PD patients (11 ± 13 vs 6 ± 11; p = 0.053). Thymic function was not different in PD and HD patients. Patients on PD before transplantation had a higher risk of acute rejection after kidney transplantation (HR, 1.61; 95%CI, 1.02 to 2.56; p = 0.041). CONCLUSIONS More pronounced inflammation with hemodialysis may induce premature aging of the immune system. This observation correlates with a lower risk of acute kidney rejection in patients previously on HD. Clinical consequences in patients maintained on dialysis should be determined. TRIAL REGISTRATION Trial registration: NCT02843867, registered July 8, 2016.
Collapse
Affiliation(s)
- Didier Ducloux
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Mathieu Legendre
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Dijon, Department of Nephrology, Dialysis, and Renal Transplantation, Dijon, France
| | - Jamal Bamoulid
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Jean-Michel Rebibou
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Dijon, Department of Nephrology, Dialysis, and Renal Transplantation, Dijon, France
| | - Philippe Saas
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, INSERM CIC 1431/UMR1098, Besançon, France
| | - Cécile Courivaud
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Thomas Crepin
- INSERM, UMR1098, Federation Hospitalo-Universitaire, INCREASE, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France
- Univ. Bourgogne-Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Dijon, France
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| |
Collapse
|
28
|
Clinical significance of CCR7 +CD8 + T cells in kidney transplant recipients with allograft rejection. Sci Rep 2018; 8:8827. [PMID: 29891963 PMCID: PMC5995850 DOI: 10.1038/s41598-018-27141-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/30/2018] [Indexed: 01/05/2023] Open
Abstract
The regulatory function of CCR7+CD8+ T cells against effector T-cells involved in T-cell mediated rejection (TCMR) in kidney transplant recipients was investigated. In vitro experiments explored the ability of CCR7+CD8+ T cells to suppress T-cell proliferation under T-cell activation conditions or during coculture with human renal proximal tubular epithelial cells (HRPTEpiC). In an ex vivo experiment, the proportion of CCR7+/CD8+, FOXP3+/CCR7+CD8+ T and effector T-cell subsets were compared between the normal biopsy control (NC, n = 17) and TCMR group (n = 17). The CCR7+CD8+ T cells significantly suppressed the proliferation of CD4+ T cells and significantly decreased the proportion of IFN-γ+ and IL-17+/CD4+ T cells and inflammatory cytokine levels (all p < 0.05). After coculturing with HRPTEpiC, CCR7+CD8+ T cells also suppressed T-cell differentiation into IL-2+, IFN-γ+, and IL-17+/CD4+ T cells (all p < 0.05). The TCMR group had significantly fewer CCR7+/CD8+ and FOXP3+/CCR7+CD8+ T in comparison with the NC group, but the proportions of all three effector T-cell subsets were increased in the TCMR group (all p < 0.05). The proportion of CCR7+/CD8+ T was inversely correlated with those of effector T-cell subsets. The results indicate that CCR7+CD8+ T cells may regulate effector T-cells involved in TCMR in an in vitro and in an ex vivo transplant model.
Collapse
|
29
|
Litjens NHR, van der Wagen L, Kuball J, Kwekkeboom J. Potential Beneficial Effects of Cytomegalovirus Infection after Transplantation. Front Immunol 2018; 9:389. [PMID: 29545802 PMCID: PMC5838002 DOI: 10.3389/fimmu.2018.00389] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/12/2018] [Indexed: 01/03/2023] Open
Abstract
Cytomegalovirus (CMV) infection can cause significant complications after transplantation, but recent emerging data suggest that CMV may paradoxically also exert beneficial effects in two specific allogeneic transplant settings. These potential benefits have been underappreciated and are therefore highlighted in this review. First, after allogeneic hematopoietic stem cell transplantation (HSCT) for acute myeloid leukemia (AML) using T-cell and natural killer (NK) cell-replete grafts, CMV reactivation is associated with protection from leukemic relapse. This association was not observed for other hematologic malignancies. This anti-leukemic effect might be mediated by CMV-driven expansion of donor-derived memory-like NKG2C+ NK and Vδ2negγδ T-cells. Donor-derived NK cells probably recognize recipient leukemic blasts by engagement of NKG2C with HLA-E and/or by the lack of donor (self) HLA molecules. Vδ2negγδ T cells probably recognize as yet unidentified antigens on leukemic blasts via their TCR. Second, immunological imprints of CMV infection, such as expanded numbers of Vδ2negγδ T cells and terminally differentiated TCRαβ+ T cells, as well as enhanced NKG2C gene expression in peripheral blood of operationally tolerant liver transplant patients, suggest that CMV infection or reactivation may be associated with liver graft acceptance. Mechanistically, poor alloreactivity of CMV-induced terminally differentiated TCRαβ+ T cells and CMV-induced IFN-driven adaptive immune resistance mechanisms in liver grafts may be involved. In conclusion, direct associations indicate that CMV reactivation may protect against AML relapse after allogeneic HSCT, and indirect associations suggest that CMV infection may promote allograft acceptance after liver transplantation. The causative mechanisms need further investigations, but are probably related to the profound and sustained imprint of CMV infection on the immune system.
Collapse
Affiliation(s)
- Nicolle H R Litjens
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus MC, University Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Lotte van der Wagen
- Laboratory of Translational Immunology, Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jurgen Kuball
- Laboratory of Translational Immunology, Department of Hematology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC, University Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
30
|
Dedeoglu B, Litjens NHR, de Weerd AE, Dor FJ, Klepper M, Reijerkerk D, Baan CC, Betjes MGH. T-Cell Composition of the Lymph Node Is Associated with the Risk for Early Rejection after Renal Transplantation. Front Immunol 2017; 8:1416. [PMID: 29163492 PMCID: PMC5663687 DOI: 10.3389/fimmu.2017.01416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/12/2017] [Indexed: 01/03/2023] Open
Abstract
Background The T-cell composition within the lymph node (LN) of end-stage renal disease (ESRD) patients differs from the composition within the circulation. Activation of the alloreactive T-cell response within secondary lymphoid organs is important after organ transplantation. However, to date no data are present on LN T-cell subsets and the risk for acute rejection after kidney transplantation. Methods T cells from LNs of ESRD patients were analyzed for frequency of recent thymic emigrants, relative telomere length, expression of differentiation markers, and were related to the development of early acute rejection (EAR), occurring within 3 months after renal transplantation (RT). Furthermore, the alloreactive potential of mononuclear cells isolated from the LN and peripheral blood of 10 patients was analyzed. Measures of alloreactive potential included proliferation, cytokine production, frequencies of interferon-gamma-producing cells, and the presence of cytotoxic molecules. Results Patients with EAR were younger (p = 0.019), cytomegalovirus-seropositive (p = 0.037) and usually received dialysis prior to RT (p = 0.030). Next to this, patients with EAR showed a lower CD4:CD8 ratio (p = 0.027) within the LN. T cells from the LN were similar with regard to alloreactive capacity compared with those within the circulation. Univariate regression analysis showed that the CD4:CD8 ratio (OR: 0.67, p = 0.039), patient age (OR: 0.93, p = 0.024), and preemptive RT (OR: 0.11, p = 0.046) were associated with EAR. After a multivariate analysis, only the CD4:CD8 ratio (OR: 0.58, p = 0.019) and preemptive RT (OR:0.05, p = 0.012) were associated with EAR. Conclusion A lower CD4:CD8 ratio in the LN is associated with a higher risk for the development of rejection within 3 months after RT.
Collapse
Affiliation(s)
- Burç Dedeoglu
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nicolle H R Litjens
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Annelies E de Weerd
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Frank Jmf Dor
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mariska Klepper
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Derek Reijerkerk
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
31
|
van den Bosch TPP, Hilbrands LB, Kraaijeveld R, Litjens NHR, Rezaee F, Nieboer D, Steyerberg EW, van Gestel JA, Roelen DL, Clahsen-van Groningen MC, Baan CC, Rowshani AT. Pretransplant Numbers of CD16 + Monocytes as a Novel Biomarker to Predict Acute Rejection After Kidney Transplantation: A Pilot Study. Am J Transplant 2017; 17:2659-2667. [PMID: 28332287 DOI: 10.1111/ajt.14280] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/08/2017] [Accepted: 03/12/2017] [Indexed: 01/25/2023]
Abstract
Acute rejection is one of the major immunological determinants of kidney graft function and survival. Early biomarkers to predict rejection are lacking. Emerging evidence reveals a crucial role for the monocyte/macrophage lineage cells in the pathogenesis of rejection. We hypothesized that higher pretransplant numbers of proinflammatory CD16+ monocytes can predict rejection. The study cohort consisted of 104 kidney transplant recipients (58 with no rejection and 46 with biopsy-proven rejection) and 33 healthy persons. Posttransplant median follow-up time was 14.7 mo (interquartile range 0.3-34 mo). Pretransplantation blood samples were analyzed by flow cytometry for monocyte immunophenotypes. Groups were compared by Cox regression models for the occurrence of acute rejection. We documented a significantly increased absolute number of pretransplant CD16+ monocytes in patients who developed biopsy-proven rejection after transplantation compared with those with no rejection (hazard ratio [HR] 1.60, 95% CI 1.28-2.00, p < 0.001) and healthy persons (HR 1.47, 95% CI 1.18-1.82, p < 0.001). In parallel, significantly fewer absolute numbers of CD16- monocytes were observed at pretransplant time points in rejectors versus nonrejectors (HR 0.74, 95% CI 0.58-0.94, p < 0,014). A higher pretransplant number of CD16+ monocytes is significantly associated with a higher risk of acute rejection after kidney transplantation.
Collapse
Affiliation(s)
- T P P van den Bosch
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R Kraaijeveld
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - N H R Litjens
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - F Rezaee
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - D Nieboer
- Department of Cell Biology, University Medical Center Groningen, Groningen, The Netherlands.,Department of Public Health, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - E W Steyerberg
- Department of Cell Biology, University Medical Center Groningen, Groningen, The Netherlands.,Department of Public Health, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - J A van Gestel
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - D L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | - C C Baan
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A T Rowshani
- Department Internal Medicine, Section of Nephrology and Transplantation, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
32
|
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
|
33
|
DeWolfe D, Gandhi J, Mackenzie MR, Broge TA, Bord E, Babwah A, Mandelbrot DA, Pavlakis M, Cardarelli F, Viscidi R, Chandraker A, Tan CS. Pre-transplant immune factors may be associated with BK polyomavirus reactivation in kidney transplant recipients. PLoS One 2017; 12:e0177339. [PMID: 28562595 PMCID: PMC5451008 DOI: 10.1371/journal.pone.0177339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/26/2017] [Indexed: 12/14/2022] Open
Abstract
BK polyomavirus (BKPyV) reactivation in kidney transplant recipients can lead to allograft damage and loss. The elements of the adaptive immune system that are permissive of reactivation and responsible for viral control remain incompletely described. We performed a prospective study evaluating BKPyV-specific T-cell response, humoral response and overall T-cell phenotype beginning pre-transplant through one year post-transplant in 28 patients at two centers. We performed an exploratory analysis of risk factors for the development of viremia and viruria as well as compared the immune response to BKPyV in these groups and those who remained BK negative. 6 patients developed viruria and 3 developed viremia. BKPyV-specific CD8+ T-cells increased post-transplant in viremic and viruric but not BK negative patients. BKPyV-specific CD4+ T-cells increased in viremic, but not viruric or BK negative patients. Anti-BKPyV IgG antibodies increased in viruric and viremic patients but remained unchanged in BK negative patients. Viremic patients had a greater proportion of CD8+ effector cells pre-transplant and at 12 months post-transplant. Viremic patients had fewer CD4+ effector memory cells at 3 months post-transplant. Exploratory analysis demonstrated lower CD4 and higher total CD8 proportions, higher anti-BKPyV antibody titers and the cause of renal failure were associated BKPyV reactivation. In conclusion, low CD4, high CD8 and increased effector CD8 cells were found pre-transplant in patients who became viremic, a phenotype associated with immune senescence. This pre-transplant T-cell senescence phenotype could potentially be used to identify patients at increased risk of BKPyV reactivation.
Collapse
Affiliation(s)
- David DeWolfe
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - Jinal Gandhi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Matthew R. Mackenzie
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Thomas A. Broge
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Evelyn Bord
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amaara Babwah
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Didier A. Mandelbrot
- The Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Martha Pavlakis
- The Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Francesca Cardarelli
- The Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Raphael Viscidi
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Anil Chandraker
- Transplantation Research Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Chen S. Tan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Disease Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Altered differentiation and activation of T-cell subsets occur in patients with chronic kidney disease (CKD), but the impact on graft rejection and protective immunity during transplantation are not fully understood. RECENT FINDINGS Patients with CKD have decreased frequency of naïve T cells, accumulation of activated, terminally differentiated memory cells, and skewed regulatory versus T helper 17 ratio. Naïve and memory T-cell subsets do not appear to improve following kidney transplantation. Retained thymic output is associated with acute rejection, whereas naïve lymphopenia and accumulation of CD8 TEMRA cells correlate with long-term graft dysfunction. CD28 memory cells accumulate during CKD and appear to confer protection against acute rejection under standard immunosuppression and possibly costimulation blockade. T cells bearing CD57 are also increased in patients with CKD and may underlie rejection during costimulation blockade. SUMMARY The mechanisms by which CKD alters the differentiation and activation status of T-cell subsets is poorly understood. Further research is also needed to understand which cell populations mediate rejection under various immunosuppressive regimens. To date, there is little use of animal models of organ failure in transplant immunology research. CKD mouse models may help identify novel pathways and targets to better control alloimmunity in posttransplant.
Collapse
Affiliation(s)
- Pamela D Winterberg
- aPediatric Nephrology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta bEmory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | |
Collapse
|
35
|
Boer K, de Wit LEA, Peters FS, Hesselink DA, Hofland LJ, Betjes MGH, Looman CWN, Baan CC. Variations in DNA methylation of interferon gamma and programmed death 1 in allograft rejection after kidney transplantation. Clin Epigenetics 2016; 8:116. [PMID: 27891189 PMCID: PMC5112717 DOI: 10.1186/s13148-016-0288-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/08/2016] [Indexed: 02/07/2023] Open
Abstract
Background The role of DNA methylation in the regulation of the anti-donor-directed immune response after organ transplantation is unknown. Here, we studied the methylation of two mediators of the immune response: the pro-inflammatory cytokine interferon γ (IFNγ) and the inhibitory receptor programmed death 1 (PD1) in naïve and memory CD8+ T cell subsets in kidney transplant recipients receiving immunosuppressive medication. Both recipients experiencing an episode of acute allograft rejection (rejectors) as well as recipients without rejection (non-rejectors) were included. Results CpGs in the promoter regions of both IFNγ and PD1 were significantly (p < 0.001) higher methylated in the naïve CD8+ T cells compared to the memory T cell subsets. The methylation status of both IFNγ and PD1 inversely correlated with the percentage of IFNγ or PD1-producing cells. Before transplantation, the methylation status of both IFNγ and PD1 was not significantly different from healthy donors. At 3 months after transplantation, irrespective of rejection and subsequent anti-rejection therapy, the IFNy methylation was significantly higher in the differentiated effector memory CD45RA+ (EMRA) CD8+ T cells (p = 0.01) whereas the PD1 methylation was significantly higher in all memory CD8+ T cell subsets (CD27+ memory; p = 0.02: CD27− memory; p = 0.02: EMRA; p = 0.002). Comparing the increase in methylation in the first 3 months after transplantation between rejectors and non-rejectors demonstrated a significantly more prominent increase in the PD1 methylation in the CD27− memory CD8+ T cells in rejectors (increase in rejectors 14%, increase in non-rejectors 1.9%, p = 0.04). The increase in DNA methylation in the other memory CD8+ T cells was not significantly different between rejectors and non-rejectors. At 12 months after transplantation, the methylation of both IFNγ and PD1 returned to baseline levels. Conclusions The DNA methylation of both IFNγ and PD1 increases the first 3 months after transplantation in memory CD8+ T cells in kidney transplant recipients. This increase was irrespective of a rejection episode indicating that general factors of the kidney transplantation procedure, including the use of immunosuppressive medication, contribute to these variations in DNA methylation.
Collapse
Affiliation(s)
- Karin Boer
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - L Elly A de Wit
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Fleur S Peters
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Internal Medicine, Section Endocrinology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Caspar W N Looman
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
36
|
Dedeoglu B, Meijers RWJ, Klepper M, Hesselink DA, Baan CC, Litjens NHR, Betjes MGH. Uremia-Associated Premature Aging of T Cells Does Not Predict Infectious Complications After Renal Transplantation. Am J Transplant 2016; 16:2324-33. [PMID: 26914971 DOI: 10.1111/ajt.13759] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 01/26/2016] [Accepted: 02/13/2016] [Indexed: 01/25/2023]
Abstract
Patients with end-stage renal disease have prematurely aged T cell systems. We tested whether T cell aging parameters were associated with the risk of infections after renal transplantation (RTx). We studied 188 patients over 1 year. Peripheral T cells were analyzed before and at 3 and 6 mo after RTx for frequency of recent thymic emigrants, relative telomere length and differentiation status. These parameters were related to the occurrence of opportunistic and serious infections. Overall, 84 patients developed an infection. In this group, 50 developed an opportunistic infection and 53 developed a serious infection. T cell aging parameters assessed before RTx were not associated with infection risk. The memory T cells showed a decrease within the first 3 mo in both groups (p < 0.001). The CD4(+) memory T cells increased between 3 and 6 mo within the infection group (p = 0.015). The number of CD8(+) memory T cells increased in both groups (p < 0.001) but reached baseline levels only in the infection group. In the infection group, the CD8(+) CD28(null) T cell percentage increased between 3 and 6 mo (p = 0.024), tending to be higher than at baseline (p = 0.061). These differences in post-RTx dynamics resulted from infections. Parameters of uremia-associated premature aging of peripheral T cells do not predict posttransplant infections.
Collapse
Affiliation(s)
- B Dedeoglu
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - R W J Meijers
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - M Klepper
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - D A Hesselink
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - C C Baan
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - N H R Litjens
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - M G H Betjes
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
37
|
Crepin T, Gaiffe E, Courivaud C, Roubiou C, Laheurte C, Moulin B, Frimat L, Rieu P, Mousson C, Durrbach A, Heng AE, Saas P, Bamoulid J, Ducloux D. Pre-transplant end-stage renal disease-related immune risk profile in kidney transplant recipients predicts post-transplant infections. Transpl Infect Dis 2016; 18:415-22. [PMID: 27027787 DOI: 10.1111/tid.12534] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 11/26/2015] [Accepted: 01/31/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND End-stage renal disease (ESRD) is associated with premature aging of the T-cell system. Nevertheless, the clinical significance of pre-transplant ESRD-related immune senescence is unknown. METHODS We studied whether immune risk phenotype (IRP), a typical feature of immune senescence, may affect post-transplant infectious complications. A total of 486 patients were prospectively studied during the first year post transplant. IRP was defined as positive cytomegalovirus serology with at least 1 of the following criteria: CD4/CD8 ratio <1 and/or CD8 T-cell count >90th percentile. RESULTS We found that 47 patients (9.7%) had pre-transplant IRP. IRP+ patients did not differ from IRP- patients for any clinical characteristics, but exhibited more pronounced immune senescence. Both opportunistic infections (43% vs. 6%, P < 0.001) and severe bacterial infection (SBI) (40% vs. 25%, P = 0.028) were more frequent in IRP(+) patients. In multivariate analysis, IRP was predictive of both opportunistic infection (hazard ratio [HR] 2.97 [95% confidence interval {CI} 1.53-5.76], P = 0.001), and SBI (HR 2.33 [95% CI 1.34-3.92], P = 0.008). Acute rejection rates were numerically much lower in IRP+ patients. A total of 418 patients (86%) had biological evaluation 1 year post transplant. Among 41 IRP+ patients, 35 (85%) remained IRP+ 1 year post transplant. CONCLUSION Pre-transplant IRP is associated with an increased risk of post-transplant infection.
Collapse
Affiliation(s)
- T Crepin
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - E Gaiffe
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France.,CIC Biothérapie, INSERM CIC1431, CHU Besançon, Besançon, France
| | - C Courivaud
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - C Roubiou
- Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - C Laheurte
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Plateforme de Biomonitoring, EFS Bourgogne Franche-Comté, CIC 1431/UMR1098, Besançon, France
| | - B Moulin
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Strasbourg, Strasbourg, France
| | - L Frimat
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Nancy, Nancy, France
| | - P Rieu
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Reims, Reims, France
| | - C Mousson
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Dijon, Dijon, France
| | - A Durrbach
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Kremlin-Bicêtre, Le Kremlin-Bicêtre, France
| | - A-E Heng
- Department of Nephrology, Dialysis, and Renal Transplantation, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - P Saas
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CIC Biothérapie, INSERM CIC1431, CHU Besançon, Besançon, France.,Plateforme de Biomonitoring, EFS Bourgogne Franche-Comté, CIC 1431/UMR1098, Besançon, France
| | - J Bamoulid
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France
| | - D Ducloux
- INSERM, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Faculté de Médecine et de Pharmacie, Université de Franche-Comté, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,Department of Nephrology, Dialysis, and Renal Transplantation, CHU Besançon, Besançon, France.,CIC Biothérapie, INSERM CIC1431, CHU Besançon, Besançon, France
| |
Collapse
|
38
|
Dedeoglu B, Meijers RWJ, Klepper M, Hesselink DA, Baan CC, Litjens NHR, Betjes MGH. Loss of CD28 on Peripheral T Cells Decreases the Risk for Early Acute Rejection after Kidney Transplantation. PLoS One 2016; 11:e0150826. [PMID: 26950734 PMCID: PMC4780739 DOI: 10.1371/journal.pone.0150826] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 02/20/2016] [Indexed: 01/26/2023] Open
Abstract
Background End-stage renal disease patients have a dysfunctional, prematurely aged peripheral T-cell system. Here we hypothesized that the degree of premature T-cell ageing before kidney transplantation predicts the risk for early acute allograft rejection (EAR). Methods 222 living donor kidney transplant recipients were prospectively analyzed. EAR was defined as biopsy proven acute allograft rejection within 3 months after kidney transplantation. The differentiation status of circulating T cells, the relative telomere length and the number of CD31+ naive T cells were determined as T-cell ageing parameters. Results Of the 222 patients analyzed, 30 (14%) developed an EAR. The donor age and the historical panel reactive antibody score were significantly higher (p = 0.024 and p = 0.039 respectively) and the number of related donor kidney transplantation was significantly lower (p = 0.018) in the EAR group. EAR-patients showed lower CD4+CD28null T-cell numbers (p<0.01) and the same trend was observed for CD8+CD28null T-cell numbers (p = 0.08). No differences regarding the other ageing parameters were found. A multivariate Cox regression analysis showed that higher CD4+CD28null T-cell numbers was associated with a lower risk for EAR (HR: 0.65, p = 0.028). In vitro, a significant lower percentage of alloreactive T cells was observed within CD28null T cells (p<0.001). Conclusion Immunological ageing-related expansion of highly differentiated CD28null T cells is associated with a lower risk for EAR.
Collapse
Affiliation(s)
- Burç Dedeoglu
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
- * E-mail:
| | - Ruud W. J. Meijers
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - Mariska Klepper
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - Dennis A. Hesselink
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - Carla C. Baan
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - Nicolle H. R. Litjens
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| | - Michiel G. H. Betjes
- Department of Internal Medicine, section Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, South Holland, the Netherlands
| |
Collapse
|
39
|
Yap M, Tilly G, Giral M, Brouard S, Degauque N. Benefits of Using CD45RA and CD28 to Investigate CD8 Subsets in Kidney Transplant Recipients. Am J Transplant 2016; 16:999-1006. [PMID: 26820487 DOI: 10.1111/ajt.13581] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/06/2015] [Accepted: 10/12/2015] [Indexed: 01/25/2023]
Abstract
The deleterious role of CD8 T cells in kidney graft outcome has regained interest over the years, and memory T cells are considered as one of the main hurdles to achieve transplantation success. Monitoring the CD8 immune response in transplant recipients involved a heterogeneous combination of markers, but the justification of their choice is rarely stated. Whereas the number of parameters is not an issue in phenotypic analysis, functional assays have to accommodate the cell number with the narrowing of the subset. The aim of the study was to investigate the similarities and differences of the subsets identified using three nomenclatures (CD45RA and CCR7/CD27/CD28) in kidney transplant recipients with stable graft function. We found that all three nomenclatures can identify naïve and effector memory (EM) rheumatoid arthritis T cell CD8 with similar features. Whereas CM CD8 could only be documented using CCR7 and CD45RA, the characteristics of EM CD8 will differ according to the nomenclature. We found that the use of the CD45RA and CD28 gives the benefit of examining two EM populations at early and late differentiation states. This systematic comparison provides a cohesive layout of the advantages of using these nomenclature strategies in kidney transplant recipients to guide the choice of their use.
Collapse
Affiliation(s)
- M Yap
- INSERM, UMR, 1064, Nantes, France
- Université de Nantes, Faculté de Médecine, Nantes, France
| | - G Tilly
- INSERM, UMR, 1064, Nantes, France
- Université de Nantes, Faculté de Médecine, Nantes, France
| | - M Giral
- INSERM, UMR, 1064, Nantes, France
- Université de Nantes, Faculté de Médecine, Nantes, France
- CHU de Nantes, ITUN, Nantes, France
- CIC biothérapie, Nantes, France
- CHU Nantes, CRB, Nantes, France
| | - S Brouard
- INSERM, UMR, 1064, Nantes, France
- CHU de Nantes, ITUN, Nantes, France
- CIC biothérapie, Nantes, France
- CHU Nantes, CRB, Nantes, France
| | - N Degauque
- INSERM, UMR, 1064, Nantes, France
- CHU de Nantes, ITUN, Nantes, France
| |
Collapse
|
40
|
T Cell Response in Patients with Implanted Biological and Mechanical Prosthetic Heart Valves. Mediators Inflamm 2016; 2016:1937564. [PMID: 26989331 PMCID: PMC4773556 DOI: 10.1155/2016/1937564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/30/2022] Open
Abstract
The study was aimed at assessing T cell subsets of peripheral blood from recipients of long-term functioning (more than 60 months) biological and mechanical heart valve prostheses. The absolute and relative number of CD4 and CD8 T cell subsets was analyzed: naïve (N, CD45RA+CD62L+), central memory (CM, CD45RA−CD62L+), effector memory (EM, CD45RA−CD62L−), and terminally differentiated CD45RA-positive effector memory (TEMRA, CD45RA+CD62L−) in 25 persons with biological and 7 with mechanical prosthesis compared with 48 apparently healthy volunteers. The relative and absolute number of central memory and naïve CD3+CD8+ in patients with biological prosthesis was decreased (p < 0.001). Meanwhile the number of CD45RA+CD62L−CD3+CD8+ and CD3+CD4+ was increased (p < 0.001). Patients with mechanical prosthesis had increased absolute and relative number of CD45RA+CD62L−CD3+CD8+ cells (p = 0.006). Also the relative number of CD3+CD4+ cells was reduced (p = 0.04). We assume that altered composition of T cell subsets points at development of xenograft rejection reaction against both mechanical and biological heart valve prostheses.
Collapse
|
41
|
Bottomley MJ, Harden PN, Wood KJ. CD8+ Immunosenescence Predicts Post-Transplant Cutaneous Squamous Cell Carcinoma in High-Risk Patients. J Am Soc Nephrol 2015; 27:1505-15. [PMID: 26563386 DOI: 10.1681/asn.2015030250] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/05/2015] [Indexed: 01/19/2023] Open
Abstract
Most morbidity associated with malignancy in long-term renal transplant recipients is due to cutaneous squamous cell carcinoma (SCC). Previously identified measures to stratify SCC risk have limited use, however. We hypothesized that an increased proportion of senescent, terminally differentiated CD8(+) T cells would identify renal transplant recipients at elevated SCC risk. Peripheral blood lymphocytes were isolated from 117 stable transplant recipients at high risk of SCC and analyzed phenotypically by flow cytometry. Participants were followed up prospectively for SCC development. The predictive value of variables was assessed using Cox regression. Age at transplant and enrollment, dialysis duration, and previous disease were predictive of SCC development during follow-up. Previously published clinical phenotype-based risk scores lost predictive value with the removal of age as a covariate. The percentage of CD57-expressing CD8(+) T cells was the strongest immunologic predictor of future SCC and correlated with increasing CD8(+) T cell differentiation. We dichotomized participants into those with a majority (CD57hi) and a minority (CD57lo) of CD8(+) T cells expressing CD57; CD57hi participants were more likely to develop SCC during follow-up (hazard ratio, 2.9; 95% confidence interval, 1.0 to 8.0), independent of potential confounders, and tended to develop earlier recurrence. The CD57hi phenotype was stable with time and associated with increasing age and cytomegalovirus seropositivity. Our results show that the CD57hi phenotype is a strong predictor of SCC development and recurrence in this cohort of long-term, high-risk renal transplant recipients. This information may allow identification of recipients who may benefit from intensive dermatologic screening and immunosuppression reduction.
Collapse
Affiliation(s)
- Matthew J Bottomley
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom; and Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom
| | - Paul N Harden
- Oxford Kidney Unit, Churchill Hospital, Oxford, United Kingdom
| | - Kathryn J Wood
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom; and
| |
Collapse
|
42
|
Yap M, Brouard S, Pecqueur C, Degauque N. Targeting CD8 T-Cell Metabolism in Transplantation. Front Immunol 2015; 6:547. [PMID: 26557123 PMCID: PMC4617050 DOI: 10.3389/fimmu.2015.00547] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/12/2015] [Indexed: 12/19/2022] Open
Abstract
Infiltration of effector CD8 T cells plays a major role in allograft rejection, and increases in memory and terminally differentiated effector memory CD8 T cells are associated with long-term allograft dysfunction. Alternatively, CD8 regulatory T cells suppress the inflammatory responses of effector lymphocytes and induce allograft tolerance in animal models. Recently, there has been a renewed interest in the field of immunometabolics and its important role in CD8 function and differentiation. The purpose of this review is to highlight the key metabolic pathways involved in CD8 T cells and to discuss how manipulating these metabolic pathways could lead to new immunosuppressive strategies for the transplantation field.
Collapse
Affiliation(s)
- Michelle Yap
- UMR 1064, INSERM , Nantes , France ; Faculté de Médecine, Université de Nantes , Nantes , France
| | - Sophie Brouard
- UMR 1064, INSERM , Nantes , France ; CHU de Nantes, ITUN , Nantes , France ; CIC Biothérapie , Nantes , France ; CHU Nantes, CRB , Nantes , France
| | - Claire Pecqueur
- Faculté de Médecine, Université de Nantes , Nantes , France ; UMR 892, INSERM , Nantes , France
| | - Nicolas Degauque
- UMR 1064, INSERM , Nantes , France ; CHU de Nantes, ITUN , Nantes , France
| |
Collapse
|
43
|
Betjes MGH. Clinical consequences of circulating CD28-negative T cells for solid organ transplantation. Transpl Int 2015; 29:274-84. [DOI: 10.1111/tri.12658] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/06/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Michiel G. H. Betjes
- Department of Nephrology and Transplantation; Erasmus Medical Center; Rotterdam the Netherlands
| |
Collapse
|
44
|
Shi XL, de Mare-Bredemeijer ELD, Tapirdamaz Ö, Hansen BE, van Gent R, van Campenhout MJH, Mancham S, Litjens NHR, Betjes MGH, van der Eijk AA, Xia Q, van der Laan LJW, de Jonge J, Metselaar HJ, Kwekkeboom J. CMV Primary Infection Is Associated With Donor-Specific T Cell Hyporesponsiveness and Fewer Late Acute Rejections After Liver Transplantation. Am J Transplant 2015; 15:2431-42. [PMID: 25943855 DOI: 10.1111/ajt.13288] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 02/17/2015] [Accepted: 02/22/2015] [Indexed: 01/25/2023]
Abstract
Viral infections, including cytomegalovirus (CMV), abrogate transplantation tolerance in animal models. Whether this also occurs in humans remains elusive. We investigated how CMV affects T cells and rejection episodes after liver transplantation (LT). Phenotype and alloreactivity of peripheral and allograft-infiltrating T cells from LT patients with different CMV status were analyzed by flow cytometry. The association of CMV status with early and late acute rejection was retrospectively analyzed in a cohort of 639 LT patients. CMV-positivity was associated with expansion of peripheral effector memory T cell subsets after LT. Patients with CMV primary infection showed donor-specific CD8(+) T cell hyporesponsiveness. While terminally differentiated effector memory cells comprised the majority of peripheral donor-specific CD8(+) T cells in CMV primary infection patients, they were rarely present in liver allografts. Retrospective analysis showed that R(-) D(+) serostatus was an independent protective factor for late acute rejection by multivariate Cox regression analysis (hazard ratio [HR] = 0.18, 95% CI = 0.04-0.86, p = 0.015). Additionally, CMV primary infection patients showed the highest Vδ1/Vδ2 γδ T cell ratio, which has been shown to be associated with operational tolerance after LT. In conclusion, our data suggest that CMV primary infection may promote tolerance to liver allografts, and CMV status should be considered when tapering or withdrawing immunosuppression.
Collapse
Affiliation(s)
- X-L Shi
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands.,Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - E L D de Mare-Bredemeijer
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Ö Tapirdamaz
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - B E Hansen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - R van Gent
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - M J H van Campenhout
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - S Mancham
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - N H R Litjens
- Department of Internal Medicine, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - M G H Betjes
- Department of Internal Medicine, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - A A van der Eijk
- Department of Virology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Q Xia
- Department of Liver Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - J de Jonge
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - H J Metselaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - J Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
45
|
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]
|
46
|
Betjes MGH, Litjens NHR. Chronic kidney disease and premature ageing of the adaptive immune response. Curr Urol Rep 2015; 16:471. [PMID: 25404185 DOI: 10.1007/s11934-014-0471-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Los of renal function is associated with uremia-associated immune deficiency, which contributes significantly to the mortality and morbidity of end-stage renal disease (ESRD) patients. In this review, the effect of ESRD on the adaptive cellular immune system is discussed. Progressive loss of renal function causes a preferential loss of number and function of lymphoid cells. More in depth analysis of these changes reveals a loss of thymic function, attrition of telomeres, and expanded memory T cell population, which is compatible with the concept of premature immunological ageing. Latency for cytomegalovirus is associated with more profound changes and the expansion of a unique pro-inflammatory, cytotoxic subset of CD4-positive CD28null T cells. Epigenetically, modifications in hematopoietic stem cells may underlie uremia-associated immunological ageing, which is not reversed by kidney transplantation. Possible therapeutic options to reverse or halt uremia-associated immunological ageing are discussed.
Collapse
Affiliation(s)
- Michiel G H Betjes
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Centre, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands,
| | | |
Collapse
|
47
|
Beloki L, Ciaurriz M, Mansilla C, Zabalza A, Perez-Valderrama E, Samuel ER, Lowdell MW, Ramirez N, Olavarria E. Assessment of the effector function of CMV-specific CTLs isolated using MHC-multimers from granulocyte-colony stimulating factor mobilized peripheral blood. J Transl Med 2015; 13:165. [PMID: 25990023 PMCID: PMC4458005 DOI: 10.1186/s12967-015-0515-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 05/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adoptive transfer of CMV-specific T cells has shown promising results in preventing pathological effects caused by opportunistic CMV infection in immunocompromised patients following allogeneic hematopoietic stem cell transplantation. The majority of studies have used steady-state leukapheresis for CMV-reactive product manufacture, a collection obtained prior to or months after G-CSF mobilization, but the procurement of this additional sample is often not available in the unrelated donor setting. If the cellular product for adoptive immunotherapy could be generated from the same G-CSF mobilized collection, the problems associated with the additional harvest could be overcome. Despite the tolerogenic effects associated with G-CSF mobilization, recent studies described that CMV-primed T cells generated from mobilized donors remain functional. METHODS MHC-multimers are potent tools that allow the rapid production of antigen-specific CTLs. Therefore, in the present study we have assessed the feasibility and efficacy of CMV-specific CTL manufacture from G-CSF mobilized apheresis using MHC-multimers. RESULTS CMV-specific CTLs can be efficiently isolated from G-CSF mobilized samples with Streptamers and are able to express activation markers and produce cytokines in response to antigenic stimulation. However, this anti-viral functionality is moderately reduced when compared to non-mobilized products. CONCLUSIONS The translation of Streptamer technology for the isolation of anti-viral CTLs from G-CSF mobilized PBMCs into clinical practice would widen the number of patients that could benefit from this therapeutic strategy, although our results need to be taken into consideration before the infusion of antigen-specific T cells obtained from G-CSF mobilized samples.
Collapse
Affiliation(s)
- Lorea Beloki
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Miriam Ciaurriz
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Amaya Zabalza
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Estela Perez-Valderrama
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Edward R Samuel
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Mark W Lowdell
- Department of Haematology, University College London Medical School, University College London, London, UK.
| | - Natalia Ramirez
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain.
| | - Eduardo Olavarria
- Oncohematology Research Group, Navarrabiomed - Miguel Servet Foundation, IDISNA (Navarra's Health Research Institute), Irunlarrea 3, 31008, Pamplona, Spain. .,Department of Haematology, Complejo Hospitalario de Navarra, Navarra Health Service, IDISNA (Navarra's Health Research Institute), Pamplona, Spain.
| |
Collapse
|
48
|
Meijers RWJ, Betjes MGH, Baan CC, Litjens NHR. T-cell ageing in end-stage renal disease patients: Assessment and clinical relevance. World J Nephrol 2014; 3:268-276. [PMID: 25374821 PMCID: PMC4220360 DOI: 10.5527/wjn.v3.i4.268] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/08/2014] [Accepted: 08/31/2014] [Indexed: 02/06/2023] Open
Abstract
End-stage renal disease (ESRD) patients have a defective T-cell-mediated immune system which is related to excessive premature ageing of the T-cell compartment. This is likely to be caused by the uremia-associated pro-inflammatory milieu, created by loss of renal function. Therefore, ESRD patients are highly susceptible for infections, have an increased risk for virus-associated cancers, respond poorly to vaccination and have an increased risk for atherosclerotic diseases. Three ageing parameters can be used to assess an immunological T-cell age. First, thymic output can be determined by assessing the T-cell receptor excision circles-content together with CD31 expression within the naïve T cells. Second, the telomere length of T cells and third the T-cell differentiation status are also indicators of T-cell ageing. Analyses based on these parameters in ESRD patients revealed that the immunological T-cell age is increased by on average 20 years compared to the chronological age. After kidney transplantation (KTx) the aged T-cell phenotype persists although the pro-inflammatory milieu is diminished. This might be explained by epigenetic modifications at hematopoietic stem cells level. Assessment of an immunological T-cell age could be an important tool to identify KTx recipients who are at risk for allograft rejection or to prevent over-immunosuppression.
Collapse
|
49
|
Meijers RWJ, Litjens NHR, de Wit EA, Langerak AW, Baan CC, Betjes MGH. Uremia-associated immunological aging is stably imprinted in the T-cell system and not reversed by kidney transplantation. Transpl Int 2014; 27:1272-84. [DOI: 10.1111/tri.12416] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/21/2014] [Accepted: 07/26/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Ruud W. J. Meijers
- Department of Internal Medicine; Section Nephrology and Transplantation; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Nicolle H. R. Litjens
- Department of Internal Medicine; Section Nephrology and Transplantation; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Elly A. de Wit
- Department of Internal Medicine; Section Nephrology and Transplantation; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Anton W. Langerak
- Department of Immunology; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Carla C. Baan
- Department of Internal Medicine; Section Nephrology and Transplantation; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| | - Michiel G. H. Betjes
- Department of Internal Medicine; Section Nephrology and Transplantation; Erasmus MC, University Medical Center; Rotterdam The Netherlands
| |
Collapse
|
50
|
Kwekkeboom J, van der Laan LJW, Betjes MGH, Manintveld OC, Hoek RAS, Cransberg K, de Bruin RWF, Dor FJMF, de Jonge J, Boor PPC, van Gent R, van Besouw NM, Boer K, Litjens NHR, Hesselink DA, Hoogduijn MJ, Massey E, Rowshani AT, van de Wetering J, de Jong H, Hendriks RW, Metselaar HJ, van Gelder T, Weimar W, IJzermans JNM, Baan CC. Rotterdam: main port for organ transplantation research in the Netherlands. Transpl Immunol 2014; 31:200-6. [PMID: 25240732 DOI: 10.1016/j.trim.2014.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/08/2014] [Indexed: 12/25/2022]
Abstract
This overview describes the full spectrum of current pre-clinical and clinical kidney-, liver-, heart- and lung transplantation research performed in Erasmus MC - University Medical Centre in Rotterdam, The Netherlands. An update is provided on the development of a large living donor kidney transplantation program and on optimization of kidney allocation, including the implementation of a domino kidney-donation program. Our current research efforts to optimize immunosuppressive regimens and find novel targets for immunosuppressive therapy, our recent studies on prevention of ischemia-reperfusion-induced graft injury, our newest findings on stimulation of tissue regeneration, our novel approaches to prevent rejection and viral infection, and our latest insights in the regulation of allograft rejection, are summarized.
Collapse
Affiliation(s)
- Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands.
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Olivier C Manintveld
- Department of Cardiology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Rogier A S Hoek
- Department of Pulmonary Diseases, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Karlien Cransberg
- Department of Pediatric Nephrology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Ron W F de Bruin
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Frank J M F Dor
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Rogier van Gent
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Nicole M van Besouw
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Karin Boer
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Nicolle H R Litjens
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Martin J Hoogduijn
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Emma Massey
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Ajda T Rowshani
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | | | - Huib de Jong
- Department of Pediatric Nephrology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Diseases, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Herold J Metselaar
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Teun van Gelder
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands; Department of Clinical Pharmacology, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Willem Weimar
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Jan N M IJzermans
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus MC-University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|