Skip to main content

EDITORIAL article

Front. Transplant., 14 June 2024
Sec. Transplantation Immunology
This article is part of the Research Topic Immunosenescence in Organ Transplantation View all 5 articles

Editorial: Immunosenescence in organ transplantation

\r\nJasper Iske,,,,
Jasper Iske1,2,3,4,5*Hao Zhou
\r\nHao Zhou5*
  • 1Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
  • 2Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
  • 3German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
  • 4Berlin Institutes of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
  • 5Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States

Editorial on the Research Topic
Immunosenescence in organ transplantation

Introduction

Aging is a complex biological phenomenon that manifests at the cellular, tissue, and organismal levels. It is increasingly clear that rich dynamics underlie biological aging, yet the fundamental nature of this process remains poorly understood. Specifically, inter-tissue dynamics of biological age, and how this may change over the lifespan, have received little attention.

Organ transplantation represents an exceptional, clinically relevant example, with donor/recipient age-discrepant combinations being a clinical routine to meet the ever-increasing demands. Age limits in organ transplantation have been raised drastically for both donors and recipients. Increasing amounts of organs are being utilized to meet the ever-growing demand, and older patients have shown the greatest proportional increase as transplant recipients over the last decade. With patients desperately awaiting a life-saving organ, these procedures are frequently performed heterochronic, i.e., with disparate chronological ages of the donor. Notably, increasing donor age, independent of recipient age, has been correlated with a higher rate of acute allograft rejection (1, 2).

Cellular senescence constitutes a molecular underpinning of aging characterized by irreversible growth arrest and cellular dysfunction. A cardinal feature of senescent cells constitutes a distinct, pro-inflammatory secretome consisting of cytokines (IL-6, IL-8, TNF-α), chemokines (CCL2, CCL20) and matrix remodeling enzymes termed the “Senescence-associated Secretory Phenotype” (SASP). The SASP has been delineated to promote age-related tissue dysfunction, age-associated chronic diseases and organismal aging, impairing tissue homeostasis and impeding neighboring cell function. In recent years, the accumulation of senescent cells in old donor organs has been characterized as crucial driver of ischemia-reperfusion injury and allo-immune responses, while also impairing recipient outcomes and altering the efficacy of immunosuppressive regimens.

The significance of cellular senescence in organ transplantation

The accumulation of senescent cells has been identified as a critical driver for promoting the immunogenicity of older organs. Mechanistically, ischemia reperfusion injury leads to a systemic release of cell-free mitochondrial DNA (cf-mt-DNA) deriving from senescent cells, which in turn triggers IL-17-dominated alloimmune responses in transplant recipients (3). Hereby, dendritic cells were found as crucial drivers communicating augmented immunogenicity by promoting CD4+ T cells derived IL-17 production leading to compromised survival of older donor organs (4). Moreover, senescence of intragraft T cells has been characterized as an instigator of age-specific organ dysfunction through augmented SASP-related signaling accelerating organ injury and potentially contributing to the limited lifetime of organ transplants (59).

More recent studies furthermore deciphered mechanisms by which the transplantation of older organs induces senescence in younger recipients, promoting age-related pathologies (10). Notably, 30 days after transplantation of an older donor organ, augmented frequencies of senescent cells in various recipient organs including draining lymph nodes, livers and hindlimb muscles could be observed associated with a systemic increase of SASP factors including mt-DNA. These findings went along with compromised physical performance and impaired spatial learning and memory abilities which may be of translational relevance not only for future age-mismatched organ transplantations but also for those that have already been performed (11).

Since transplantation of an allogeneic donor organ exposes the recipient immune system to repetitive antigen stimulation, it may not only promote somatic cell senescence but also accelerate immunosenescence (12). Indeed, various studies have demonstrated augmented numbers of senescent T cells linked to compromised telomere lengths following kidney transplantation (13, 14). Of translational relevance, higher frequencies of both, senescent CD4+ and CD8+ T cells correlated tightly to infections (13) and exhibited inflammatory profiles (14).

In clinical transplantation, T cell immunosenescence has been the main driver of less frequent acute rejections in older transplant recipients. However, accelerated T cell immunosenescence has also been linked to an altered responsiveness to immunosuppressive therapy. Thus, enhanced immunosuppressive effects have been observed for tacrolimus (15) and rapamycin (16) while CTLA4-Ig appeared to be less effective in aged recipients (17).

In clinical practice, older and younger transplant patients are usually treated with the same immunosuppressants and comparable drug levels (18) while strategies of “just” lowering dosages of established immunosuppressants in older patients have not always been successful (19, 20).

Recent evidence has furthermore suggested that age-associated hormone changes in recipients may affect allo-immune responses and transplant outcomes, highlighting the importance of considering age- and sex-specific factors in transplant medicine (21).

Future perspectives

A promising therapeutic approach targeting the augmented immunogenicity of older donor organs while impeding transplantation-derived senescence induction, lies in the application of drugs that selectively clear senescent cells, known as senolytics (22). Noteworthy, senolytics are already undergoing several clinical trials with encouraging outcomes. Consequently, transplantation emerges as a captivating field for further exploration and application. In experimental models, treating old donor animals with senolytics, prior to transplantation, cleared senescent cells and reduced cf-mt-DNA release, thereby attenuating immune responses and improving the survival of old cardiac allografts, comparable to young donor organs (3). Moreover, senolytic treatment improved physical function of young recipients receiving old donor organs (11).

Although therapeutic benefits of senolytics have accelerated their journey from discovery to clinical trials, cell type specific efficacy, bioavailability and the off-target side effects require further investigation particularly with a view on long-term outcomes. Novel senolytics drugs with higher specificity, selectivity and even greater efficacy are currently under investigation while research for novel compounds is ongoing (23).

For their application in transplantation, the most efficacious deployment of senolytics needs to be evaluated. Hereby, treatment of the donor, the recipient, and the organ itself by recruiting ex vivo perfusion systems (24, 25) need to be compared to ensure most efficient treatment while sparing side effects.

To date, transplantation studies have mainly focused on the graft's effect on the recipient's environment. However, studying how the recipient's environment modulates the graft may uncover further mechanisms regulating long term graft outcomes and chronic rejection. Since the transplantation of an old donor organ into a young recipient was sufficient to accelerate aging processes, transplanting a young donor organ into an old recipient is likely to induce senescence in the donor organ (10).

In contrast, the effect of young recipient age on old graft function, may have beneficial effects. Supporting this concept, studies on age-mismatched muscle transplantation in mouse models revealed that the regenerative capacity of skeletal muscle grafts aligns with the age of the recipient (26). Muscle grafts from elderly donors namely exhibited enhanced regenerative abilities when transplanted into young recipients (26). Furthermore, heterochronic parabiosis (HPB) models have provided evidence for rejuvenating aged organ function through the infusion of young blood into older organisms affecting a broad range of organs (27, 28), while the actual identities of rejuvenating factors remain a matter of debate and need further exploration.

Conclusion

Collectively, the accumulation of senescent cells poses a risk towards utilizing older organs as an untapped source for expanding the donor pool. Utilizing senolytics may provide a novel approach to face this challenge but is currently in the initial phase of clinical realization. Investigating molecular patterns underlying age-mismatched transplantation may furthermore reveal novel mechanisms driving or impeding the aging process.

Author contributions

JI: Conceptualization, Writing – original draft, Writing – review & editing. HZ: Conceptualization, Writing – original draft, Writing – review & editing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Colvin MM, Smith CA, Tullius SG, Goldstein DR. Aging and the immune response to organ transplantation. J Clin Invest. (2017) 127:2523–9. doi: 10.1172/JCI90601

PubMed Abstract | Crossref Full Text | Google Scholar

2. Tullius SG, Milford E. Kidney allocation and the aging immune response. N Engl J Med. (2011) 364:1369–70. doi: 10.1056/NEJMc1103007

PubMed Abstract | Crossref Full Text | Google Scholar

3. Iske J, Seyda M, Heinbokel T, Maenosono R, Minami K, Nian Y, et al. Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat Commun. (2020) 11:4289. doi: 10.1038/s41467-020-18039-x

PubMed Abstract | Crossref Full Text | Google Scholar

4. Oberhuber R, Heinbokel T, Cetina Biefer HR, Boenisch O, Hock K, Bronson RT, et al. CD11c+ dendritic cells accelerate the rejection of older cardiac transplants via interleukin-17A. Circulation. (2015) 132:122–31. doi: 10.1161/CIRCULATIONAHA.114.014917

PubMed Abstract | Crossref Full Text | Google Scholar

5. Mogilenko DA, Shpynov O, Andhey PS, Arthur L, Swain A, Esaulova E, et al. Comprehensive profiling of an aging immune system reveals clonal GZMK(+) CD8(+) T cells as conserved hallmark of inflammaging. Immunity. (2021) 54:99–115.e112. doi: 10.1016/j.immuni.2020.11.005

PubMed Abstract | Crossref Full Text | Google Scholar

6. Pereira BI, De Maeyer RPH, Covre LP, Nehar-Belaid D, Lanna A, Ward S, et al. Sestrins induce natural killer function in senescent-like CD8(+) T cells. Nat Immunol. (2020) 21:684–94. doi: 10.1038/s41590-020-0643-3

PubMed Abstract | Crossref Full Text | Google Scholar

7. He A, Sarwar A, Thole LML, Siegle J, Sattler A, Ashraf MI, et al. Renal inflamm-aging provokes intra-graft inflammation following experimental kidney transplantation. Am J Transplant. (2022) 22:2529–47. doi: 10.1111/ajt.17154

PubMed Abstract | Crossref Full Text | Google Scholar

8. Iske J, Dedeilia A, Xiao Y, Martin F, Emmert MY, Sage PT, et al. The impact of T-cell aging on alloimmunity and inflammaging. Transplantation. (2024) 108:634–42. doi: 10.1097/TP.0000000000004715

PubMed Abstract | Crossref Full Text | Google Scholar

9. Nian Y, Minami K, Maenesono R, Iske J, Yang J, Azuma H, et al. Changes of T-cell immunity over a lifetime. Transplantation. (2019) 103:2227–33. doi: 10.1097/TP.0000000000002786

PubMed Abstract | Crossref Full Text | Google Scholar

10. Iske J, Matsunaga T, Zhou H, Tullius SG. Donor and recipient age-mismatches: the potential of transferring senescence. Front Immunol. (2021) 12:671479. doi: 10.3389/fimmu.2021.671479

PubMed Abstract | Crossref Full Text | Google Scholar

11. Iske J, Roesel MJ, Martin F, Schroeter A, Matsunaga T, Maenosono R, et al. Transplanting old organs promotes senescence in young recipients. Am J Transplant. (2024) 24:391–405. doi: 10.1016/j.ajt.2023.10.013

PubMed Abstract | Crossref Full Text | Google Scholar

12. Heinbokel T, Elkhal A, Liu G, Edtinger K, Tullius SG. Immunosenescence and organ transplantation. Transplant Rev (Orlando). (2013) 27:65–75. doi: 10.1016/j.trre.2013.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

13. Higdon LE, Gustafson CE, Ji X, Sahoo MK, Pinsky BA, Margulies KB, et al. Association of premature immune aging and cytomegalovirus after solid organ transplant. Front Immunol. (2021) 12:661551. doi: 10.3389/fimmu.2021.661551

PubMed Abstract | Crossref Full Text | Google Scholar

14. Daniel L, Tassery M, Lateur C, Thierry A, Herbelin A, Gombert JM, et al. Allotransplantation is associated with exacerbation of CD8T-cell senescence: the particular place of the innate CD8T-cell component. Front Immunol. (2021) 12:674016. doi: 10.3389/fimmu.2021.674016

PubMed Abstract | Crossref Full Text | Google Scholar

15. Krenzien F, Quante M, Heinbokel T, Seyda M, Minami K, Uehara H, et al. Age-dependent metabolic and immunosuppressive effects of tacrolimus. Am J Transplant. (2017) 17:1242–54. doi: 10.1111/ajt.14087

PubMed Abstract | Crossref Full Text | Google Scholar

16. Quante M, Heinbokel T, Edtinger K, Minami K, Uehara H, Nian Y, et al. Rapamycin prolongs graft survival and induces CD4+IFN-γ+IL-10+ regulatory type 1 cells in old recipient mice. Transplantation. (2018) 102:59–69. doi: 10.1097/TP.0000000000001902

PubMed Abstract | Crossref Full Text | Google Scholar

17. Heinbokel T, Quante M, Iske J, Nian Y, Maenosono R, Minami K, et al. CTLA4-Ig prolongs graft survival specifically in young but not old mice. Am J Transplant. (2020) 21(2):488–502. doi: 10.1111/ajt.16218

PubMed Abstract | Crossref Full Text | Google Scholar

18. Knoll GA. Kidney transplantation in the older adult. Am J Kidney Dis. (2013) 61:790–7. doi: 10.1053/j.ajkd.2012.08.049

PubMed Abstract | Crossref Full Text | Google Scholar

19. Andrés A, Budde K, Clavien PA, Becker T, Kessler M, Pisarski P, et al. A randomized trial comparing renal function in older kidney transplant patients following delayed versus immediate tacrolimus administration. Transplantation. (2009) 88:1101–8. doi: 10.1097/TP.0b013e3181ba06ee

Crossref Full Text | Google Scholar

20. Lehner LJ, Staeck O, Halleck F, Liefeldt L, Bamoulid J, Budde K. Need for optimized immunosuppression in elderly kidney transplant recipients. Transplant Rev (Orlando). (2015) 29:237–9. doi: 10.1016/j.trre.2015.08.001

PubMed Abstract | Crossref Full Text | Google Scholar

21. Maenosono R, Nian Y, Iske J, Liu Y, Minami K, Rommel T, et al. Recipient sex and estradiol levels affect transplant outcomes in an age-specific fashion. Am J Transplant. (2021) 21(10):3239–55. doi: 10.1111/ajt.16611

PubMed Abstract | Crossref Full Text | Google Scholar

22. Matsunaga T, Iske J, Schroeter A, Azuma H, Zhou H, Tullius SG. The potential of senolytics in transplantation. Mech Ageing Dev. (2021) 200:111582. doi: 10.1016/j.mad.2021.111582

PubMed Abstract | Crossref Full Text | Google Scholar

23. Smer-Barreto V, Quintanilla A, Elliott RJR, Dawson JC, Sun J, Campa VM, et al. Discovery of senolytics using machine learning. Nat Commun. (2023) 14:3445. doi: 10.1038/s41467-023-39120-1

PubMed Abstract | Crossref Full Text | Google Scholar

24. Iske J, Schroeter A, Knoedler S, Nazari-Shafti TZ, Wert L, Roesel MJ, et al. Pushing the boundaries of innovation: the potential of ex vivo organ perfusion from an interdisciplinary point of view. Front Cardiovasc Med. (2023) 10:1272945. doi: 10.3389/fcvm.2023.1272945

PubMed Abstract | Crossref Full Text | Google Scholar

25. Roesel MJ, Wiegmann B, Ius F, Knosalla C, Iske J. The role of ex-situ perfusion for thoracic organs. Curr Opin Organ Transplant. (2022) 27(5):466–73. doi: 10.1097/MOT.0000000000001008

PubMed Abstract | Crossref Full Text | Google Scholar

26. Carlson BM, Faulkner JA. Muscle transplantation between young and old rats: age of host determines recovery. Am J Physiol. (1989) 256:C1262–6. doi: 10.1152/ajpcell.1989.256.6.C1262

PubMed Abstract | Crossref Full Text | Google Scholar

27. Zhang B, Lee DE, Trapp A, Tyshkovskiy A, Lu AT, Bareja A, et al. Multi-omic rejuvenation and life span extension on exposure to youthful circulation. Nat Aging. (2023) 3(8):948–64. doi: 10.1038/s43587-023-00451-9

Google Scholar

28. Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. (2005) 433:760–4. doi: 10.1038/nature03260

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: senescence, senolytic, Senescence-associated Secretory Phenotype (SASP), transplantation, alloimmunity, aging, senescent cell, solid organ transplantation

Citation: Iske J and Zhou H (2024) Editorial: Immunosenescence in organ transplantation. Front. Transplant. 3:1422358. doi: 10.3389/frtra.2024.1422358

Received: 23 April 2024; Accepted: 28 May 2024;
Published: 14 June 2024.

Edited and Reviewed by: Gavin Pettigrew, University of Cambridge, United Kingdom

Reviewed by:

Denise Lo, Emory University, United States

© 2024 Iske and Zhou. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jasper Iske, jasper.iske@outlook.com; Hao Zhou, hzhou21@bwh.harvard.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.