Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Oct 1.
Published in final edited form as: Curr Opin Organ Transplant. 2022 Aug 9;27(5):481–487. doi: 10.1097/MOT.0000000000001019

Preserving and rejuvenating old organs for transplantation: Novel treatments including the potential of senolytics

Tomohisa Matsunaga 1,2,#, Maximilian J Roesel 1,3,#, Andreas Schroeter 1,4, Yao Xiao 1, Hao Zhou 1, Stefan G Tullius 1
PMCID: PMC9490781  NIHMSID: NIHMS1826174  PMID: 35950886

Abstract

Purpose of review

Older donors have the potential to close the gap between demand and supply in solid organs transplantation. Utilizing older organs, at the same time, has been associated with worse short- and long-term outcomes. Here, we introduce potential mechanisms on how treatments during machine perfusion (MP) may safely improve the utilization of older organs.

Recent findings

Consequences of ischemia reperfusion injury (IRI), a process of acute, sterile inflammation leading to organ injury are more prominent in older organs. Of relevance, organ age and IRI seem to act synergistically, leading to an increase of damage associated molecular patterns that trigger innate and adaptive immune responses. While cold storage has traditionally been considered the standard of care in organ preservation, accumulating data support that both hypothermic and normothermic MP improve organ quality, particularly in older organs. Furthermore, MP provides the opportunity to assess the quality of organs while adding therapeutic agents. Experimental data have already demonstrated the potential of applying treatments during MP. New experimental show that the depletion of senescent cells that accumulate in old organs improves organ quality and transplant outcomes.

Summary

As the importance of expanding the donor pool is increasing, MP and novel treatments bear the potential to assess and regenerate older organs, narrowing the gap between demand and supply.

Keywords: transplantation, old donors, preservation, machine perfusion, senolytics

Introduction

In parallel to changing demographics, donor age has increased for all solid organ transplants over the past decade. (optn.transplant.hrsa.gov/data/view-data-reports/national-data, accessed on 6.30.2022). Donor age, at the same time, significantly increases the risk of adverse outcomes linked to an advanced organ damage in addition to an organ-age related augmented immunogenicity (1-3). As demand still faces limited supply, optimal utilization of old organs seems critical to reduce waiting times, morbidity, and mortality rates of patients awaiting organ transplantation (2, 3).

The consequences of ischemia-reperfusion injury (IRI) have been proven to advance synergistically with rising donor age and prolonged cold ischemic time, clinically manifested by more frequent rates of delayed graft function (DGF) (4-7). Moreover, DGF appears to be associated with an elevated risk of acute rejections following transplantation, impairing both short- and long-term outcomes (8, 9).

In recent years, machine perfusion (MP) has gained clinical relevance, yielding superior short-term outcomes with lower DGF rates compared to static cold storage (CS). MP not only contributes to improved preservation of the graft but may also facilitate assessment and potentially treatment during the preservation period (10*, 11).

Here, we present both experimental and clinical data on age-specific aspects of organ damage and assess potential treatment methods within MP when using older organs for transplantation.

Donor age effects on transplant outcomes

Globally changing demographics with a rising older population have impacted the availability of organs for transplantation. As older organs bear the potential of closing the gap between demand and supply, optimal utilization of those organs is urgently needed to expand the donor pool (12-14). At the same time, donor age has been associated with increased rejection rates and more advanced organ injuries subsequent to IRI, all leading to compromised transplant outcomes (2, 3, 15).

Clinically, donor age seems to negatively affect outcomes across all solid organ transplantations. In lung transplantation, grafts from donors age > 65 years have shown increased 1-year graft failure rates. Interestingly, inferior outcomes of lungs procured from donors aged 56-65 were only observed when transplanted into severely ill recipients, emphasizing on the importance of donor recipient matching (16). An extensive analysis of the United Network for Organ Sharing (UNOS) registry demonstrated poorer survival rates with incrementally increasing older donor organs (>40 years) (17). Comparably seen in liver transplantation, an analysis of almost 35.000 transplants listed with the European Liver Transplant Registry demonstrated progressively worsening survival outcomes at both 3- and 12-months with increasing donor age (18). In renal transplantation, donor age and diabetes as the cause of renal failure have been identified as the main drivers of recipient death-censored survival in an analysis of > 50.000 deceased kidney transplantations (19, 20). Delineating the relevance and consequences of recipient and donor age among > than 300.000 renal transplant recipients listed in the UNOS data base, older kidneys demonstrated to have higher rates of acute rejections within the first post-transplant year. This finding is in line with our own observations that older organs have an augmented immunogenicity (2, 3). Indeed, recipients of organs from donors 60-69 years experienced a 24.5% rejection rate which was significantly higher compared to rates of younger donors (3).

Accordingly supported by experimental data, older organs appear to be more immunogenic due to an augmented T-cell alloreactivity, cytokine production, and elevated numbers of peripheral T- and B-cells(21).

Additional experimental data from our group have recently delineated the detrimental role of old dendritic cells (DCs) in augmenting alloimmune responses (22). In contrast young DCs have been able to enhance allograft survival while depleting intra-graft DCs prolonged allograft survival in old mice (23, 24). Mechanistically, we found that old organs contained increased numbers of senescent cells serving as a source of cell-free-mt-DNA (cf-mt-DNA). Increased cf-mt-DNA levels, in turn, promote DC-mediated amplification of Th1/Th17 alloimmune responses (22, 24). Those data have provided a rationale seeking ways to deplete senescent cells in old organs prior to transplantation to improve organ quality and transplant outcomes.

Aging increases the susceptibility of grafts to ischemia-reperfusion injury

IRI represents a process of acute, sterile inflammation occurring with the restoration of blood flow after an initial phase of ischemia (25). This multi-factorial process leads to a morphological changes involving broad molecular and cellular mechanisms (26). As a result of a complex interaction of different channels and transporters, calcium overload has been proposed to be an initial step in the pathogenesis of the injury (27, 28). Moreover, both ischemia itself and reperfusion lead to an overload of Reactive Oxygen Species (ROS) (29). Mechanistically, ROS disturb cellular functions and signaling pathways in addition to augmenting innate immune responses (30). Both, calcium- and ROS-overload initiate the formation of the mitochondrial permeability transition pore (mPTP) (31). This process leads to structural damage through mitochondrial swelling, allowing the influx of hydrogen ions while uncoupling the electron transport chain and accelerating ROS release upon reperfusion (32, 33). These changes also trigger both apoptosis and necrosis causing a further release of ROS, proinflammatory cytokines and damage-associated molecular patterns (34). Finally, disturbed cell and tissue integrity triggers an inflammatory cascade activating players of both, the innate and the adaptive immune system (35).

As an inevitable consequence of solid organ transplantation, IRI affects both short- and long-term outcomes (19, 36-39). Although effects on clinical outcomes have not been entirely delineated, there is strong experimental evidence that aging enhances the susceptibility of IRI in clinical transplantation (40, 41). Despite the minimal morphologic changes with age, older livers have significantly reduced intracellular energy content after IRI linked to a reduced capacity of mitochondrial ATP production (42). Recent investigations have been able to link the augmented IRI-susceptibility of older livers to a depletion of both sirtuin-1 and mitofusin-2 accompanied with defective autophagy and mPTP onset (43). In addition, an age-dependent mitophagy dysfunction may be at play. Mechanistically, defective mitochondria are removed via autophagy (=mitophagy) with the critical involvement of parkin, an E3 ubiquitin ligase, priming impaired mitochondria for removal. Immunohistochemistry analysis of DCD-graft biopsies performed 2-hours after revascularization revealed a low parkin expression with increasing donor age, a process linked to an augmented IRI-sensitivity (44). Of additional interest, pretreating old rats with pooled young plasma restored hepatic autophagic activity and reduced age-dependent liver IRI (45).

Comparable data are available for older kidneys showing an ineffective autophagy following IRI (46). Additionally, changes in blood urea nitrogen and creatinine appear more severe with aging including mitochondrial morphology, tubular epithelial loss and hemorrhage being significantly more pronounced in older kidneys (47, 48). Recent experimental data have shown that organ age and IRI act synergistically, leading to an increased DAMP release, triggering innate immune activation. Cf-mt-DNA levels acting as a DAMP were 15x higher in old mice after IRI compared to young mice (22). Of clinical relevance, several studies have shown increased levels of urinary mt-DNA in patients with acute kidney injury, often induced by renal IRI (49-51). Moreover, there seems to be a synergistic relationship between donor age and prolonged ischemia in regard to organ damage leading to both functional and morphological deterioration after transplantation as shown in experimental renal transplant models (52).

The potential of Machine Perfusion to preserve older organs

Although CS has been the gold standard preservation method for many years, MP bears critical advantages, allowing for longer preservation times, evaluation of organ quality and the potential of organ reconditioning to maximize the utilization of organs for transplantation (53-56). MP per-se is not novel. As early as at the beginning of the twentieth century, efforts have been made to develop such techniques with the first success achieved by Carrel and Lindbergh, developing a normothermic MP method.(57). Belzer's hypothermic MP technique in the early 1960s then replaced normothermic MP. Nevertheless, CS prevailed and became the gold standard for many years as organs of higher quality had predominantly been utilized without the need for MP (58, 59).

A renaissance of MP then occurred with an augmented demand for the utilization of sub-optimal organs, predominantly those from older and DCD donors. In a first international randomized controlled trial (RCT), 336 consecutive deceased donors were randomly assigned to MP or to CS opening a new era in MP (60). Indeed, hypothermic MP decreased the incidence of DGF in renal transplantation while improving 1-year graft survival rate (61, 62). Moreover, avoiding cold ischemia periods altogether appeared advantageous. Indeed, livers preserved by normothermic MP demonstrated significantly reduced levels of hepatocellular enzyme release, despite a 50% lower rate of organ discard and a 54% longer mean preservation time in a RCT (54). Moreover, normothermic MP was shown to inhibit inflammation and promote graft regeneration (63). RCTs comparing different perfusion techniques are still needed but a clinical trial comparing hypothermic and normothermic MP is already underway in liver transplantation (ClinicalTrials.gov Identifier: NCT04644744).

In addition to improving outcomes, MP facilitates the assessment of organ quality, allowing transplant teams to assess the viability of the graft prior to transplantation (11, 64, 65). Although both hypothermic and normothermic MP seem to be beneficial for transplant outcomes, the advantageous of either approach regarding organ-assessment, and treatment has not been compared yet.

Different markers assessing organ quality have been evaluated, including transaminases, glucose metabolism, lactate clearance, and acid-base balance to access the viability of high-risk livers during normothermic MP (64, 66). In renal transplantation, measuring glutathione S-transferase, lanine-aminopeptidase, N-acetyl-β-D-glucosaminidase, and heart-type fatty acid binding protein during hypothermic MP have been found to be associated with DGF, allowing clinicians to adjust posttransplant recipient management (67). Novel approaches are being developed to further improve the assessment of organs, for example, adding creatinine to the perfusate and calculating creatinine clearance from the perfusate over time (68*).

MP also provides a platform for the administration of therapeutic agents (10, 69, 70) . Experimentally, pharmacotherapy, genetic treatment, mesenchymal stromal cells (MSC), and nanoparticles, have all been successfully administered directly during ex vivo MP (10). For instance, adding human MSCs to porcine kidneys under normothermic MP increases the release of immunomodulatory cytokines including human hepatocyte growth factor, interleukin (IL) −6, and IL-8. Conversely, MSC treatment decreased the release of lactate dehydrogenase and neutrophil gelatinase-associated lipocalin (71). In clinical ex-vivo studies, pairs of DCD kidneys were used, demonstrating ameliorated inflammatory responses and an augmented synthesis of adenosine triphosphate and growth factors, including endothelial growth factor, fibroblast growth factor 2, transforming growth factor α when treated with MSCs during MP. Moreover, normalization of the cytoskeleton and mitosis have been observed in MSC-treated organs. Taken together, the regeneration of human renal tissue ex vivo seems possible, providing a rationale for further clinical application (72). Further support of MP as a platform for organ targeted therapy comes from a recent ex vivo kidney perfusion study providing evidence for an augmented avidity of monoclonal antibodies during ex vivo MP. Administration of CD47-blocking antibody (αCD47Ab), an inhibitor of thrombospondin mediated IRI-signaling during CS resulted in no antibody binding whereas one hour of ex vivo MP, in turn, yielded widely spread abundancy of αCD47Ab through the tissue regions of interest (73). Of clinical relevance, the concept of selectively treating the organ isolated ex-vivo allows for high dosing and cost-effective application while avoiding side effects.

The potential of senolytics

With aging, senescent cells accumulate and tissue regeneration is becoming limited (74-76). Although there is currently not a single marker for senescent cells, they can be characterized by the expression of p21CIP1 and/or p16INK4a, DNA segments with chromatin alterations reinforcing senescence, senescence-associated heterochromatin foci, senescence-associated β-galactosidase, and short telomeres (77-80). Of pathophysiological relevance, senescent cells secrete a myriad of pro-inflammatory factors termed the senescence-associated secretory phenotype (SASP) that both disrupt tissue homeostasis, affecting functions of neighbor cells resulting in age-related tissue dysfunction, chronic age-related diseases, and advanced biological aging (81**-84). As senescence represents a cell-autonomous mechanism, preventing the transformation into malignant cells (85), approaches to inhibit cellular senescence by targeting p16INK4a, Rb, p53, or p21CIP1 must be well balanced in order to avoid the development of malignancies (86, 87).

Promising evidence exists for the novel drug class of senolytics with the capacity to selectively deplete senescent cells. (69, 88). Several types of potential targets for anti-aging agents have recently been identified in senescent cells, including the BCL family, HSP90 inhibitors, and PI3K/AKT (89-92) (Fig1). Indeed, the accumulation of senescent cells in cardiomyocytes, endothelial cells, smooth muscle cells, cardiac fibroblasts, and cardiac progenitor cells has been successfully decreased by senolytics leading to diminished amounts of SASP, reduced cardiac hypertrophy and decreased size of the left ventricle (93-95). In preclinical studies, senolytics have been able to alleviate age-related dysfunction, inflammation, and disease in multiple organs, potentially making them effective agents if using organs from older donors for transplantation (88). Notably, several clinical trials evaluating the clinical potential of senolytics are currently underway, with first promising results, showing improved physical function in patients with idiopathic pulmonary fibrosis (88, 96). Moreover, another early clinical trial found the treatment with senolytics to reduce senescent cell burden in adipose tissue of diabetic kidney disease patients, further supporting the concept of targeting senescent cells in humans by senolytics (97). In addition, SToMP-AD, an open label pilot phase 1 trial of senolytics for Alzheimer’s disease has been completed (98**, 99).

Figure 1: Mechanisms through which senolytics interfere with aging.

Figure 1:

Mechanisms of actions of different senolytics (Dasatinib, Fisetin, Quercetin, Curcumin, PCC1, 17-AAG, 17-DMAG, Geldanamycin, UBC-1325, A-1331852, A1155463) are shown. Interfering with distinct signaling pathways, senolytics facilitate the apoptosis of senescent cells, thereby reducing the secretion of the senescence-associated secretory phenotype. Arrows represent activation; bars represent inhibition; yellow/red particles symbolize the senescence-associated secretory phenotype. Abbreviations: RTK, receptor tyrosine kinase; PI3K, Phosphatidylinositol 3-kinase; Akt, Protein kinase B; HSP90, heat shock protein 90; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; bad, BCL2 associated agonist of cell death; Bax, Bcl-2-associated X protein; Bcl-xL, B-cell lymphoma-extra-large; FoxO4, Forkhead box protein O4; p21, cyclin-dependent kinase inhibitor 1; p53, tumor suppressor p53; PCC1, procyanidin C1; 17-AAG, 17-N-allylamino-17-demethoxygeldanamycin; 17-DMAG, 17-Dimethylaminoethylamino- 17-demethoxygeldanamycin; A-1331852 & A-1155463, both selective Bcl-xL inibitors.

Senolytics may also play a beneficial role in organ transplantation. Experimentally, we have shown that senolytics applied prior to experimental IRI reduced senescent cell burden significantly, alleviated systemic inflammation while reducing levels of cf-mt-DNA, Th17, and IFNγ+ T cells. Moreover, administering senolytics prolonged the survival of old murine cardiac transplants significantly even beyond that of young hearts (22).

Preliminary data have also shown that senolytics can deplete senescent cells from discarded human kidneys perfused by cold MP (unpublished own data). Thus, perfusing old organs with senolytics may help to expand the donor pool and improve outcomes of transplants specifically from older donors.

Conclusion

Old organs have an augmented susceptibility to IRI together with an amplified immunogenicity impacting both short -and long-term transplant outcomes. MP has not only the potential to ameliorate the consequences of IRI but can also serve as a platform for preoperative assessment and organ treatment. Senolytics represent a novel class of agents that deplete senescent cells. Thus far only tested experimentally, senolytics represent a novel approach to improve the quality of older organs, thereby increasing organ availability, and improving transplant outcomes.

Acknowledgements

This work was supported by the Osaka Medical College Foundation (to TM), the Biomedical Education Program (BMEP) of the German Academic Exchange Service (to MJR and AS), NIH grants R01AG064165, R56 AGO39449 and UO-1 AI132898 (to SGT) and a grant by the Pepper Foundation (to HZ).

References

  • 1.Messina M, Diena D, Dellepiane S, Guzzo G, Lo Sardo L, Fop F, et al. Long-Term Outcomes and Discard Rate of Kidneys by Decade of Extended Criteria Donor Age. Clin J Am Soc Nephrol. 2017;12(2):323–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Tullius SG, Milford E. Kidney allocation and the aging immune response. N Engl J Med. 2011;364(14):1369–70. [DOI] [PubMed] [Google Scholar]
  • 3.Tullius SG, Tran H, Guleria I, Malek SK, Tilney NL, Milford E. The combination of donor and recipient age is critical in determining host immunoresponsiveness and renal transplant outcome. Ann Surg. 2010;252(4):662–74. [DOI] [PubMed] [Google Scholar]
  • 4.Helantera I, Ibrahim HN, Lempinen M, Finne P. Donor Age, Cold Ischemia Time, and Delayed Graft Function. Clin J Am Soc Nephrol. 2020;15(6):813–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chapal M, Le Borgne F, Legendre C, Kreis H, Mourad G, Garrigue V, et al. A useful scoring system for the prediction and management of delayed graft function following kidney transplantation from cadaveric donors. Kidney Int. 2014;86(6):1130–9. [DOI] [PubMed] [Google Scholar]
  • 6.Siedlecki A, Irish W, Brennan DC. Delayed graft function in the kidney transplant. Am J Transplant. 2011;11(11):2279–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Irish WD, McCollum DA, Tesi RJ, Owen AB, Brennan DC, Bailly JE, et al. Nomogram for predicting the likelihood of delayed graft function in adult cadaveric renal transplant recipients. J Am Soc Nephrol. 2003;14(11):2967–74. [DOI] [PubMed] [Google Scholar]
  • 8.Irish WD, Ilsley JN, Schnitzler MA, Feng S, Brennan DC. A risk prediction model for delayed graft function in the current era of deceased donor renal transplantation. Am J Transplant. 2010;10(10):2279–86. [DOI] [PubMed] [Google Scholar]
  • 9.Yarlagadda SG, Coca SG, Formica RN Jr., Poggio ED, Parikh CR. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis. Nephrol Dial Transplant. 2009;24(3):1039–47. [DOI] [PubMed] [Google Scholar]
  • 10.*. Zulpaite R, Miknevicius P, Leber B, Strupas K, Stiegler P, Schemmer P. Ex-vivo Kidney Machine Perfusion: Therapeutic Potential. Front Med (Lausanne). 2021;8:808719. Review about ex vivo kidney perfusion focusing on current perfusion techniques and therapeutic options inluding newest experimental and clinical studies.
  • 11.Adham M, Peyrol S, Chevallier M, Ducerf C, Vernet M, Barakat C, et al. The isolated perfused porcine liver: assessment of viability during and after six hours of perfusion. Transpl Int. 1997;10(4):299–311. [DOI] [PubMed] [Google Scholar]
  • 12.Tullius SG, Rabb H. Improving the Supply and Quality of Deceased-Donor Organs for Transplantation. N Engl J Med. 2018;378(20):1920–9. [DOI] [PubMed] [Google Scholar]
  • 13.Klassen DK, Edwards LB, Stewart DE, Glazier AK, Orlowski JP, Berg CL. The OPTN Deceased Donor Potential Study: Implications for Policy and Practice. Am J Transplant. 2016;16(6):1707–14. [DOI] [PubMed] [Google Scholar]
  • 14.Saidi RF, Hejazii Kenari SK. Challenges of organ shortage for transplantation: solutions and opportunities. Int J Organ Transplant Med. 2014;5(3):87–96. [PMC free article] [PubMed] [Google Scholar]
  • 15.Gerbase-DeLima M, de Marco R, Monteiro F, Tedesco-Silva H, Medina-Pestana JO, Mine KL. Impact of Combinations of Donor and Recipient Ages and Other Factors on Kidney Graft Outcomes. Front Immunol. 2020;11:954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Baldwin MR, Peterson ER, Easthausen I, Quintanilla I, Colago E, Sonett JR, et al. Donor age and early graft failure after lung transplantation: a cohort study. Am J Transplant. 2013;13(10):2685–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Weber DJ, Wang IW, Gracon AS, Hellman YM, Hormuth DA, Wozniak TC, et al. Impact of donor age on survival after heart transplantation: an analysis of the United Network for Organ Sharing (UNOS) registry. J Card Surg. 2014;29(5):723–8. [DOI] [PubMed] [Google Scholar]
  • 18.Burroughs AK, Sabin CA, Rolles K, Delvart V, Karam V, Buckels J, et al. 3-month and 12-month mortality after first liver transplant in adults in Europe: predictive models for outcome. Lancet. 2006;367(9506):225–32. [DOI] [PubMed] [Google Scholar]
  • 19.Dayoub JC, Cortese F, Anzic A, Grum T, de Magalhaes JP. The effects of donor age on organ transplants: A review and implications for aging research. Exp Gerontol. 2018;110:230–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Keith DS, Demattos A, Golconda M, Prather J, Norman D. Effect of donor recipient age match on survival after first deceased donor renal transplantation. J Am Soc Nephrol. 2004;15(4):1086–91. [DOI] [PubMed] [Google Scholar]
  • 21.Reutzel-Selke A, Jurisch A, Denecke C, Pascher A, Martins PN, Kessler H, et al. Donor age intensifies the early immune response after transplantation. Kidney Int. 2007;71(7):629–36. [DOI] [PubMed] [Google Scholar]
  • 22.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(1):4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Min WP, Gorczynski R, Huang XY, Kushida M, Kim P, Obataki M, et al. Dendritic cells genetically engineered to express Fas ligand induce donor-specific hyporesponsiveness and prolong allograft survival. J Immunol. 2000;164(1):161–7. [DOI] [PubMed] [Google Scholar]
  • 24.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(2):122–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.In: Fitridge R, Thompson M, editors. Mechanisms of Vascular Disease: A Reference Book for Vascular Specialists. Adelaide (AU) 2011. [PubMed] [Google Scholar]
  • 26.Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW, et al. Current Mechanistic Concepts in Ischemia and Reperfusion Injury. Cell Physiol Biochem. 2018;46(4):1650–67. [DOI] [PubMed] [Google Scholar]
  • 27.Wang R, Wang M, He S, Sun G, Sun X. Targeting Calcium Homeostasis in Myocardial Ischemia/Reperfusion Injury: An Overview of Regulatory Mechanisms and Therapeutic Reagents. Front Pharmacol. 2020;11:872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pittas K, Vrachatis DA, Angelidis C, Tsoucala S, Giannopoulos G, Deftereos S. The Role of Calcium Handling Mechanisms in Reperfusion Injury. Curr Pharm Des. 2018;24(34):4077–89. [DOI] [PubMed] [Google Scholar]
  • 29.Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol. 2015;6:524–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012;298:229–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bauer TM, Murphy E. Role of Mitochondrial Calcium and the Permeability Transition Pore in Regulating Cell Death. Circ Res. 2020;126(2):280–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Morciano G, Giorgi C, Bonora M, Punzetti S, Pavasini R, Wieckowski MR, et al. Molecular identity of the mitochondrial permeability transition pore and its role in ischemia-reperfusion injury. J Mol Cell Cardiol. 2015;78:142–53. [DOI] [PubMed] [Google Scholar]
  • 33.Baines CP. The mitochondrial permeability transition pore and ischemia-reperfusion injury. Basic Res Cardiol. 2009;104(2):181–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sanada S, Kitakaze M. Ischemic preconditioning: emerging evidence, controversy, and translational trials. Int J Cardiol. 2004;97(2):263–76. [DOI] [PubMed] [Google Scholar]
  • 35.Salvadori M, Rosso G, Bertoni E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant. 2015;5(2):52–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rampes S, Ma D. Hepatic ischemia-reperfusion injury in liver transplant setting: mechanisms and protective strategies. J Biomed Res. 2019;33(4):221–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gelman AE, Fisher AJ, Huang HJ, Baz MA, Shaver CM, Egan TM, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction Part III: Mechanisms: A 2016 Consensus Group Statement of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant. 2017;36(10):1114–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Saat TC, van den Akker EK, JN IJ, Dor FJ, de Bruin RW. Improving the outcome of kidney transplantation by ameliorating renal ischemia reperfusion injury: lost in translation? J Transl Med. 2016;14:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li S, Guan Q, Chen Z, Gleave ME, Nguan CY, Du C. Reduction of cold ischemia-reperfusion injury by graft-expressing clusterin in heart transplantation. J Heart Lung Transplant. 2011;30(7):819–26. [DOI] [PubMed] [Google Scholar]
  • 40.Roesel MJ, Sharma NS, Schroeter A, Matsunaga T, Xiao Y, Zhou H, et al. Primary Graft Dysfunction: The Role of Aging in Lung Ischemia-Reperfusion Injury. Front Immunol. 2022;13:891564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Dickson KM, Martins PN. Implications of liver donor age on ischemia reperfusion injury and clinical outcomes. Transplant Rev (Orlando). 2020;34(3):100549. [DOI] [PubMed] [Google Scholar]
  • 42.Selzner M, Selzner N, Jochum W, Graf R, Clavien PA. Increased ischemic injury in old mouse liver: an ATP-dependent mechanism. Liver Transpl. 2007;13(3):382–90. [DOI] [PubMed] [Google Scholar]
  • 43.Chun SK, Lee S, Flores-Toro J, U RY, Yang MJ, Go KL, et al. Loss of sirtuin 1 and mitofusin 2 contributes to enhanced ischemia/reperfusion injury in aged livers. Aging Cell. 2018;17(4):e12761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Li Y, Ruan DY, Jia CC, Zheng J, Wang GY, Zhao H, et al. Aging aggravates hepatic ischemia-reperfusion injury in mice by impairing mitophagy with the involvement of the EIF2alpha-parkin pathway. Aging (Albany NY). 2018;10(8):1902–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Liu A, Yang J, Hu Q, Dirsch O, Dahmen U, Zhang C, et al. Young plasma attenuates age-dependent liver ischemia reperfusion injury. FASEB J. 2019;33(2):3063–73. [DOI] [PubMed] [Google Scholar]
  • 46.Diao C, Wang L, Liu H, Du Y, Liu X. Aged kidneys are refractory to autophagy activation in a rat model of renal ischemia-reperfusion injury. Clin Interv Aging. 2019;14:525–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Xu X, Fan M, He X, Liu J, Qin J, Ye J. Aging aggravates long-term renal ischemia-reperfusion injury in a rat model. J Surg Res. 2014;187(1):289–96. [DOI] [PubMed] [Google Scholar]
  • 48.Kusaka J, Koga H, Hagiwara S, Hasegawa A, Kudo K, Noguchi T. Age-dependent responses to renal ischemia-reperfusion injury. J Surg Res. 2012;172(1):153–8. [DOI] [PubMed] [Google Scholar]
  • 49.Hu Q, Ren J, Wu J, Li G, Wu X, Liu S, et al. Urinary Mitochondrial DNA Levels Identify Acute Kidney Injury in Surgical Critical Illness Patients. Shock. 2017;48(1):11–7. [DOI] [PubMed] [Google Scholar]
  • 50.Whitaker RM, Stallons LJ, Kneff JE, Alge JL, Harmon JL, Rahn JJ, et al. Urinary mitochondrial DNA is a biomarker of mitochondrial disruption and renal dysfunction in acute kidney injury. Kidney Int. 2015;88(6):1336–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Malek M, Nematbakhsh M. Renal ischemia/reperfusion injury; from pathophysiology to treatment. J Renal Inj Prev. 2015;4(2):20–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tullius SG, Reutzel-Selke A, Egermann F, Nieminen-Kelha M, Jonas S, Bechstein WO, et al. Contribution of prolonged ischemia and donor age to chronic renal allograft dysfunction. J Am Soc Nephrol. 2000;11(7):1317–24. [DOI] [PubMed] [Google Scholar]
  • 53.Chandak P, Phillips BL, Uwechue R, Thompson E, Bates L, Ibrahim I, et al. Dissemination of a novel organ perfusion technique: ex vivo normothermic perfusion of deceased donor kidneys. Artif Organs. 2019;43(11):E308–E19. [DOI] [PubMed] [Google Scholar]
  • 54.Nasralla D, Coussios CC, Mergental H, Akhtar MZ, Butler AJ, Ceresa CDL, et al. A randomized trial of normothermic preservation in liver transplantation. Nature. 2018;557(7703):50–6. [DOI] [PubMed] [Google Scholar]
  • 55.Van Raemdonck D, Neyrinck A, Cypel M, Keshavjee S. Ex-vivo lung perfusion. Transpl Int. 2015;28(6):643–56. [DOI] [PubMed] [Google Scholar]
  • 56.Reddy SP, Brockmann J, Friend PJ. Normothermic perfusion: a mini-review. Transplantation. 2009;87(5):631–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Carrel A, Lindbergh CA. The Culture of Whole Organs. Science. 1935;81(2112):621–3. [DOI] [PubMed] [Google Scholar]
  • 58.Weissenbacher A, Vrakas G, Nasralla D, Ceresa CDL. The future of organ perfusion and re-conditioning. Transpl Int. 2019;32(6):586–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Belzer FO, Ashby BS, Gulyassy PF, Powell M. Successful seventeen-hour preservation and transplantation of human-cadaver kidney. N Engl J Med. 1968;278(11):608–10. [DOI] [PubMed] [Google Scholar]
  • 60.Moers C, Smits JM, Maathuis MH, Treckmann J, van Gelder F, Napieralski BP, et al. Machine perfusion or cold storage in deceased-donor kidney transplantation. N Engl J Med. 2009;360(1):7–19. [DOI] [PubMed] [Google Scholar]
  • 61.Jiao B, Liu S, Liu H, Cheng D, Cheng Y, Liu Y. Hypothermic machine perfusion reduces delayed graft function and improves one-year graft survival of kidneys from expanded criteria donors: a meta-analysis. PLoS One. 2013;8(12):e81826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Treckmann J, Moers C, Smits JM, Gallinat A, Maathuis MH, van Kasterop-Kutz M, et al. Machine perfusion versus cold storage for preservation of kidneys from expanded criteria donors after brain death. Transpl Int. 2011;24(6):548–54. [DOI] [PubMed] [Google Scholar]
  • 63.Jassem W, Xystrakis E, Ghnewa YG, Yuksel M, Pop O, Martinez-Llordella M, et al. Normothermic Machine Perfusion (NMP) Inhibits Proinflammatory Responses in the Liver and Promotes Regeneration. Hepatology. 2019;70(2):682–95. [DOI] [PubMed] [Google Scholar]
  • 64.Martins PN, Buchwald JE, Mergental H, Vargas L, Quintini C. The role of normothermic machine perfusion in liver transplantation. Int J Surg. 2020;82S:52–60. [DOI] [PubMed] [Google Scholar]
  • 65.op den Dries S, Karimian N, Sutton ME, Westerkamp AC, Nijsten MW, Gouw AS, et al. Ex vivo normothermic machine perfusion and viability testing of discarded human donor livers. Am J Transplant. 2013;13(5):1327–35. [DOI] [PubMed] [Google Scholar]
  • 66.Watson CJE, Kosmoliaptsis V, Pley C, Randle L, Fear C, Crick K, et al. Observations on the ex situ perfusion of livers for transplantation. Am J Transplant. 2018;18(8):2005–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Moers C, Varnav OC, van Heurn E, Jochmans I, Kirste GR, Rahmel A, et al. The value of machine perfusion perfusate biomarkers for predicting kidney transplant outcome. Transplantation. 2010;90(9):966–73. [DOI] [PubMed] [Google Scholar]
  • 68.*. Verstraeten L, Jochmans I. Sense and Sensibilities of Organ Perfusion as a Kidney and Liver Viability Assessment Platform. Transpl Int. 2022;35:10312. Overview of the available evidence and challenges of predicting posttransplant outcomes using injury markers and perfusion parameters when utilizing ex vivo machine perfusion systems.
  • 69.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] [PMC free article] [PubMed] [Google Scholar]
  • 70.Karimian N, Yeh H. Opportunities for Therapeutic Intervention During Machine Perfusion. Curr Transplant Rep. 2017;4(2):141–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pool MBF, Vos J, Eijken M, van Pel M, Reinders MEJ, Ploeg RJ, et al. Treating Ischemically Damaged Porcine Kidneys with Human Bone Marrow- and Adipose Tissue-Derived Mesenchymal Stromal Cells During Ex Vivo Normothermic Machine Perfusion. Stem Cells Dev. 2020;29(20):1320–30. [DOI] [PubMed] [Google Scholar]
  • 72.Brasile L, Henry N, Orlando G, Stubenitsky B. Potentiating Renal Regeneration Using Mesenchymal Stem Cells. Transplantation. 2019;103(2):307–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hameed AM, Lu DB, Burns H, Byrne N, Chew YV, Julovi S, et al. Pharmacologic targeting of renal ischemia-reperfusion injury using a normothermic machine perfusion platform. Sci Rep. 2020;10(1):6930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Korolchuk VI, Miwa S, Carroll B, von Zglinicki T. Mitochondria in Cell Senescence: Is Mitophagy the Weakest Link? EBioMedicine. 2017;21:7–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Jurk D, Wilson C, Passos JF, Oakley F, Correia-Melo C, Greaves L, et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat Commun. 2014;2:4172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995;92(20):9363–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bernadotte A, Mikhelson VM, Spivak IM. Markers of cellular senescence. Telomere shortening as a marker of cellular senescence. Aging (Albany NY). 2016;8(1):3–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rodier F, Munoz DP, Teachenor R, Chu V, Le O, Bhaumik D, et al. DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion. J Cell Sci. 2011;124(Pt 1):68–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee BY, Han JA, Im JS, Morrone A, Johung K, Goodwin EC, et al. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell. 2006;5(2):187–95. [DOI] [PubMed] [Google Scholar]
  • 80.Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113(6):703–16. [DOI] [PubMed] [Google Scholar]
  • 81.**. Gasek NS, Kuchel GA, Kirkland JL, Xu M. Strategies for Targeting Senescent Cells in Human Disease. Nat Aging. 2021;1(10):870–9. Key publication reviewing features of senescensce including intracellular signaling pathways, and strategies for targeting senescent cells.
  • 82.Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The Clinical Potential of Senolytic Drugs. J Am Geriatr Soc. 2017;65(10):2297–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Roesel MJ, Matsunaga T, Tullius SG. Opportunities and Challenges of Targeting an Aging Immune System. Transplantation. 2021;105(12):2515–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wissler Gerdes EO, Zhu Y, Weigand BM, Tripathi U, Burns TC, Tchkonia T, et al. Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. Int Rev Neurobiol. 2020;155:203–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Campisi J. Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol. 2001;11(11):S27–31. [DOI] [PubMed] [Google Scholar]
  • 86.Takeuchi S, Takahashi A, Motoi N, Yoshimoto S, Tajima T, Yamakoshi K, et al. Intrinsic cooperation between p16INK4a and p21Waf1/Cip1 in the onset of cellular senescence and tumor suppression in vivo. Cancer Res. 2010;70(22):9381–90. [DOI] [PubMed] [Google Scholar]
  • 87.Larsen CJ. [pRB, p53, p16INK4a, senescence and malignant transformation]. Bull Cancer. 2004;91(5):399–402. [PubMed] [Google Scholar]
  • 88.Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288(5):518–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: Eliminating Senescent Cells and Alleviating Intervertebral Disc Degeneration. Front Bioeng Biotechnol. 2022;10:823945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yosef R, Pilpel N, Tokarsky-Amiel R, Biran A, Ovadya Y, Cohen S, et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun. 2016;7:11190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Fuhrmann-Stroissnigg H, Ling YY, Zhao J, McGowan SJ, Zhu Y, Brooks RW, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8(1):422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wagner V, Gil J. Senescence as a therapeutically relevant response to CDK4/6 inhibitors. Oncogene. 2020;39(29):5165–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Owens WA, Walaszczyk A, Spyridopoulos I, Dookun E, Richardson GD. Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev. 2021;198:111540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, et al. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell. 2019;18(3):e12945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Anderson R, Lagnado A, Maggiorani D, Walaszczyk A, Dookun E, Chapman J, et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019;38(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine. 2019;40:554–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Corrigendum to 'Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease' EBioMedicine 47 (2019) 446-456. EBioMedicine. 2020;52:102595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.**. Wissler Gerdes EO, Misra A, Netto JME, Tchkonia T, Kirkland JL. Strategies for late phase preclinical and early clinical trials of senolytics. Mech Ageing Dev. 2021;200:111591. Key review disussing the efficacy of senolytics in pre-clinical and early clinical trials, showing the potenital of these novel drugs and the need for more clinical trials.
  • 99.Gonzales MM, Garbarino VR, Marques Zilli E, Petersen RC, Kirkland JL, Tchkonia T, et al. Senolytic Therapy to Modulate the Progression of Alzheimer's Disease (SToMP-AD): A Pilot Clinical Trial. J Prev Alzheimers Dis. 2022;9(1):22–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES