Mechanisms and Consequences of Injury and Repair in Older Organ Transplants : Transplantation

Journal Logo

Editorials and Perspectives: Overview

Mechanisms and Consequences of Injury and Repair in Older Organ Transplants

Slegtenhorst, Bendix R.1,2; Dor, Frank J.M.F.2; Elkhal, Abdala1; Rodriguez, Hector1,3; Yang, Xiaoyong1,4; Edtinger, Karoline1,5; Quante, Markus1,6; Chong, Anita S.7; Tullius, Stefan G.1,8

Author Information
Transplantation 97(11):p 1091-1099, June 15, 2014. | DOI: 10.1097/TP.0000000000000072
  • Free

Abstract

Donor organ scarcity remains a significant clinical challenge in transplantation. Older organs, increasingly utilized to meet the growing demand for donor organs, have been linked to inferior transplant outcomes. Susceptibility to organ injury, reduced repair capacity, and increased immunogenicity are interrelated and impacted by physiological and pathological aging processes. Insights into the underlying mechanisms are needed to develop age-specific interventional strategies with regards to organ preservation, immunosuppression, and allocation. In this overview, we summarize current knowledge of injury and repair mechanisms and the effects of aging relevant to transplantation.

In an effort to meet the increasing demand for supply in transplantation, the utilization of organs from older donors has become commonplace. Indeed, donor age has steadily increased over the last decades. Some countries have initiated efforts to optimize allocation of older donor organs. The Eurotransplant Senior Program (ESP), for example, was established in 1999 with the aim of increasing the utilization of organs from donors older than 65 years and allocating them to older recipients. This program has focused on limiting ischemic times, thereby recognizing age-specific injury and repair processes. Consequently, the utilization of older organs for transplantation has risen in this program. Although transplantation of older organs has proven to be effective in expanding the donor pool, advanced donor age has been linked to inferior transplant outcomes. A thorough understanding of age-related injury and repair mechanisms is needed to develop effective interventional strategies with the goals of improving transplant outcomes while increasing the utilization of older organs.

EFFECTS OF DONOR AGE ON CLINICAL TRANSPLANT OUTCOMES

In kidney transplantation, donor age is a limiting factor of long-term graft survival for organs from both deceased and living donors (1, 2). Likewise, donor age has been linked to delayed graft function (DGF) (1, 2) and higher rates of acute rejection (3, 4). At the same time, DGF has been associated with higher rates of acute rejection episodes (5). Both DGF (5) and acute rejection episodes (6), in turn, are independent predictors of graft survival. Of note, frequencies of acute rejection (4) and graft survival (7) improve when older organs are transplanted into older recipients. Age does not only impact outcomes in renal transplantation, and organ-specific aspects are of relevance. In liver transplantation, donor age is a strong risk factor of graft failure (8). In addition, donor age increases the risk of HCV recurrence (9) and fibrosis progression (10), and antiviral therapies are less efficient (11). Donor age has also been shown to predict patient survival in heart transplantation (12). Moreover, donor age was associated with the highest increase in graft failure for pancreas transplantation (13). In lung transplantation, donor age older than 65 years correlated with compromised patient survival in a large retrospective series (14). Of note, however, other studies demonstrated comparable outcomes of donors older than 55 years with those of younger donors (15, 16).

PHYSIOLOGICAL AGING

Aging is characterized by processes of cellular senescence and low-grade inflammation. Cellular senescence refers to a cellular state of growth arrest that can be induced by endogenous or exogenous stressors. These stressors comprise critical telomere shortening, DNA damage, oncogene mutations, and oxidative stress, all activating the p53 and p16INK4a tumor suppressor pathways and resulting in cell cycle arrest (17). During the aging process, senescent cells accumulate (18). Although senescent cells remain viable, they exhibit an altered phenotype. While remaining metabolically active, senescent cells are characterized by compromised replication and resistance to apoptosis. The accumulation of senescent cells induced by telomere shortening impairs an organ’s ability to repair and to regenerate subsequent to injury (19, 20). Interestingly, elimination of senescent cells has experimentally been shown to delay onset and progression of age-related diseases (21).

Chronic inflammation is a hallmark of aging and has been termed inflamm-aging (22). Chronic antigenic stress originating from a compromised clearance of self-antigens and exogenous antigens, such as microbes, is thought to contribute to inflammation (23). Moreover, age is related to increased levels of reactive oxygen species (ROS) (24). ROS, in turn, activate MAPK and NF-κB pathways, leading to the expression of inflammatory cytokines and chemokines (25). Additionally, ROS induce formation of the NLRP3 inflammasome, leading to the expression of IL-1β (26). More recently, a role for senescent cells in chronic inflammation has been established. Senescent cells maintain a state of low-grade inflammation by secreting proinflammatory cytokines, chemokines, and proteases in response to DNA damage, which has been labeled as the senescence-associated secretory phenotype (27). DNA injury induces the activation of DNA damage response genes that ultimately activate NF-κB pathways, leading to cell activation and death (28).

INJURY AND REPAIR IN THE AGING ORGAN

During transplantation, temporary deprivation of oxygen and nutrients is inevitable, which, in combination with subsequent reperfusion events, results in organ damage. As a consequence to aging processes, older organs are more prone to deleterious insults (Fig. 1).

F1-5
FIGURE 1:
Organ age contributes to an augmented susceptibility to injury and to an impaired repair and regenerative capacity. Recipient immune responses are augmented subsequent to an inflammatory environment of the donor organ, increased antigenic burden, epigenetic DNA alterations, and an enhanced antigen presentation by resident dendritic cells. Additionally, the old organ is more prone to acute injuries such as ischemia reperfusion injury. Degenerative processes of the organ such as reduced functional mass, vasculopathies, steatosis, and fibrosis advance the injury furthermore, whereas impaired cellular stress responses and diminished growth signaling and receptor responsiveness debilitate repair function.

Organ-Specific Age-Related Injuries

The aging process and related diseases affect organs in distinct ways and have organ-specific effects relevant to transplantation. All organs experience a decline in functional mass with age (29–31). For the kidney, these processes have been linked to a reduced ability to recovery from graft injury and compromised transplant outcomes. A reduced nephron mass is known to be a predictive factor for renal disease progression and graft outcome (32). Moreover, reduced numbers of nephrons have been linked to hyperfiltration and subsequent damage (33). Additionally, donor age correlates with interstitial fibrosis, tubular atrophy, and glomerulosclerosis (34), possibly mediated by mechanisms of cellular senescence (20). In the liver, aging is associated with nonalcoholic steatohepatitis and fibrosis (35). Consequences of steatosis on microcirculatory flow, energy homeostasis, and inflammation have been implicated as injury mechanisms (36). In heart transplantation, development of cardiac allograft vasculopathy (CAV) is a significant factor of long-term graft survival (37) and donor age has been shown to be the strongest predictor for the onset of CAV (38). CAV is characterized by diffuse intimal thickening and luminal narrowing, caused by both alloimmune-dependent and -independent factors (39). The prevalence of coronary lesions varies from 17% in individuals less than 20 years old to 85% in individuals greater than 50 years old (40), with pre-existing lesions predicting the development of CAV (41). Taken together, there is strong evidence that age imprints an organ-specific propensity of graft injury and a compromise in the ability to recover, leading to reduced allograft function and survival.

Ischemia Reperfusion Injury

Ischemia reperfusion injury (IRI) encompasses the combined injury to an organ subsequent to ischemia and deprivation of nutrients and oxygen, with subsequent reperfusion injury caused by mitochondrial failure, sterile inflammation, and endothelial cell dysfunction. During the ischemic phase, cellular maintenance of pH, ion homeostasis, and cell integrity are compromised by ATP depletion, leading to a process called oncosis that comprises cell swelling, rupture, and cell death (42). Moreover, apoptotic pathways are activated and result in further cell loss. The capacity to maintain adequate levels of ATP is reduced with aging, possibly linked to the downregulation of electron transport enzymes (43) or ROS-induced damage of mitochondrial DNA, impairing mitochondrial function (44).

The reperfusion phase is characterized by a burst of ROS, induction of a sterile inflammatory response, congestion of the microcirculation known as the no-reflow phenomenon, and endothelial dysfunction. Under physiologic conditions, mitochondria generate ROS by leakage of electrons from the oxidative chain. It is well established that aging is associated with an increase in basal levels of ROS linked to mitochondrial DNA mutations and consequent respiratory chain dysfunction (45). After ischemia and reperfusion, older organs generate higher levels of ROS (46), which are directly cytotoxic and increase the production of inflammatory cytokines (25). Induction of the mitochondrial permeability transition pore (mPTP) is a critical pathological process leading to cell death (47). The mPTP constitutes a nonspecific pore that opens the inner mitochondrial membrane and allows entry of molecules less than 1.5 kDa into the mitochondrion. Influx of ions disrupts the electrochemical gradient and uncouples oxidative phosphorylation, resulting in ATP depletion. These mechanisms lead to mitochondrial swelling, membrane rupture, and release of apoptotic proteins, resulting in apoptotic cell death or necrosis (48). Induction of mPTP has been linked to an accumulation of matrix calcium (49) and to oxidative stress during IRI (50). Of note, aging increases mPTP activity subsequent to increased ROS production and impaired calcium homeostasis (51, 52).

After reperfusion, a sterile inflammatory response is initiated by endogenous molecules that are released by necrotic and injured cells. These molecules have been characterized as danger associated molecular patterns (DAMPs) that activate pattern recognition receptors such as the Toll-like receptors (TLR), C-type lectin receptors, and NOD-like receptors. These events, in turn, stimulate an augmented expression of inflammatory cytokines (53) and upregulate MHC and costimulatory molecules (54), thus promoting the induction of alloimmune responses. In kidneys that expressed TLR4 mutations, graft function immediately posttransplantation was improved (55) and acute rejection rates were reduced (56). Aging may enhance the inflammatory response through multiple mechanisms. Phagocytic capacities of dendritic cells (DC) (57) and other phagocytic cells (58, 59) decline with aging, resulting in diminished clearance of apoptotic cells (60). Although the clearance of apoptotic cells is associated with the induction of tolerance (61), apoptotic cells undergoing secondary necrosis induce the release of DAMPs that activate DCs through C-type lectin receptors (62), leading to dendritic cell maturation and proinflammatory cytokine secretion. Furthermore, it is conceivable that increased ischemia-induced injury and necrotic cell death in aged organs lead to an augmentation of DAMP signaling, which further enhances the inflammatory response.

Organ Age Augments Immunogenicity

Clinically, organ age has been linked to higher acute rejection rates (4), providing another means by which older organs may suffer injury after transplantation. Age-associated epigenetic changes that result in hypermethylation of the CpG regions or hypomethylation of the non-CpG regions (63, 64) may increase the immunogenicity of the DNA. In particular, hypomethylation of aged DNA has been reported to elicit a stronger activation of DCs compared to DNA from young donors (65). Furthermore, old DCs have been shown to secrete larger amounts of inflammatory cytokines upon stimulation, possibly as a result of a decreased activation of PI3K-signaling pathways and a reduced suppression of p38-MAPK activation (57). Although immunosenescence leads to an overall decline of immune function, enhanced antigen-presenting capacities have been reported (66, 67). Experimental data from our laboratory confirm that older intragraft DCs stimulate the alloimmune response more potently compared to DCs from younger grafts (unpublished data). Graft endothelial cells may be another contributor to an enhanced immunogenicity as older endothelial cells express higher levels of VCAM-1 and MCP-1, thereby facilitating leukocyte adhesion and infiltration (68). Taken together, these observations support the hypothesis that the magnitude of the alloreactive T-cell responses directed against older grafts will be enhanced relative to younger, better-preserved allografts. Furthermore, the magnitude of the alloantibody response and the breadth of the specificity of this response may be enlarged to include cryptic self-antigens exposed during necrotic cell death after the transplantation of older compared to younger allografts.

Cellular Stress Responses

Heat Shock Response

Heat, oxidative stress, osmotic stress, and infection all induce heat shock responses, through the expression of heat shock proteins (HSP) that are regulated by heat shock factors (HSF). HSPs are involved in protein homeostasis and serve as molecular chaperones that aid the folding and repair of damaged proteins, thereby preventing protein aggregation and cell apoptosis (69). HSF1 also promotes ubiquitination and proteolysis (70). After an acute injury, such as IRI, HSPs assist in restoring normal organ function (71). Aging is associated with elevated basal levels of HSPs, possibly resulting from continuous stress signals (72). However, induction of HSPs during stress diminishes with aging (73) and is associated with increased injury (74). The protective capacity of HSP-32, or hemeoxygenase 1 (HO-1), has been comprehensively reviewed elsewhere (75). Briefly, while not having a chaperoning function, HO-1 exhibits anti-inflammatory, anti-oxidant, and anti-apoptotic properties (76). During acute injury and transplantation of older kidneys, HO-1 induction is diminished while the pharmacological induction of HO-1 ameliorates injury (77, 78). Although HSPs have been regarded as immunostimulatory because of their property to activate TLRs, this paradigm has recently been disputed (79), and their immunosuppressive functions have been described and confirmed (80). In experimental transplantation models, HSP treatment resulted in improved graft outcomes (81). Thus, impaired induction of HSP may contribute to inferior outcomes when transplanting older organs.

Heat Shock Response

The endoplasmic reticulum (ER) is the cell organelle responsible for protein synthesis and folding. As a quality control mechanism, only correctly folded proteins are released from the ER, whereas incorrectly folded proteins are retained for refolding or targeted for degradation. When ER function is disturbed by stressors such as IRI, unfolded proteins accumulate, inducing an adaptive response termed the unfolded protein response (UPR) (82). This response comprises the upregulation of the ER folding machinery, shutdown of protein synthesis, and upregulation of ER-associated degradation through the proteasome (83). Mitochondria also synthesize a limited amount of proteins and induce a similar mitochondrion-specific UPR upon stress (84). Apoptosis is the ultimate response if repair is inadequate (85). Stressors from ischemia and reperfusion, such as hypoxia, glucose depletion, and ROS, cause ER stress and activate the UPR (82). ER stress is involved in renal (86), hepatic (87), and cardiac (88) IRI. With age, the molecular chaperones and enzymes involved in the UPR exhibit reduced expression, leading to increased cell loss through apoptosis (89). One might speculate that a dysfunctional UPR caused by aging impairs organ repair in transplantation; however, evidence for this phenomenon remains lacking.

Ubiquitin-Proteasome Function

The ubiquitin-proteasome system (UPS) is a proteolytic mechanism for proteins marked for degradation. This system is involved in normal protein turnover and in the removal of damaged proteins during repair processes. The target protein is ubiquitinated and subsequently degraded by the proteasome. Proteasome activity declines with aging, while the ubiquitination system appears not to be affected (72, 90). Compromised protein clearance, in turn, leads to accumulation of damaged proteins during cardiac ischemia (91), conceivably augmenting organ injury. Indeed, proteasome inhibition has been shown to aggravate injury in cardiac (92) and renal (93) IRI. However, reduction of proteasomal activity also suppresses NF-κB activity. Other studies have reported on the protection by pharmacological proteasomal inhibition against IRI, possibly through NF-κB inhibition (94). Cell specificity of UPS function appears to be an important factor in mediating protection from or aggravation of IRI (95). The only age-specific study on UPS function and acute injury concluded that an age-dependent reduction of UPS activity resulted into both NF-κB reduction and augmented tissue injury, possibly through increased apoptosis. In transplantation, proteasome inhibition has successfully been used to counter antibody-mediated rejection, possibly by inducing apoptosis of plasma cells (96). These contradicting data make it difficult at this time to assess the net effect of an age-mediated reduction in proteasomal activity.

Autophagy

Autophagy is a process by which dysfunctional organelles and cytoplasmic proteins are degraded. During this process, part of the cytoplasm is isolated in a double-membrane vesicle, denoted by the term autophagosome. The autophagosome fuses with a lysosome, which is followed by enzymatic degradation of the enclosed material. Autophagy is essential for normal cell homeostasis and also plays an important role during periods of cellular stress, for example, ischemia, oxidative stress, and nutrient deprivation (97). By degrading nonessential cell constituents, metabolic demand is reduced, and substrates for ATP generation are provided. At the same time, oxidative damage may be reduced by maintaining Tricarboxylic acid cycle function. The relevance of autophagy in transplantation is demonstrated by IRI experiments providing evidence of the protective role of autophagy (98–100). Of note, autophagy function declines with age (101) and may contribute to an increased IRI sensitivity of older organs (99, 102).

Organ Age-Dependent Regulation of Repair and Regeneration

Epithelial proliferation is regulated by the interaction of epithelial and supporting cells, such as monocytes and macrophages, endothelial cells, stellate cells, and mesenchymal stem cells (103–105). These cells exert autocrine and paracrine functions by producing growth factors subsequent to injury. Genes that encode insulin-like growth factor 1, hepatocyte growth factor (HGF), epidermal growth factor (EGF), heparin-binding EGF-like growth factor (HB-EGF), and fibroblast growth factor (FGF) are upregulated and stimulate epithelial proliferation in response to kidney injury (106). Similarly, HGF, EGF, HB-EGF, TGF-α, and amphiregulin stimulate hepatocyte growth (107). The injured endothelium is also in need of repair after ischemic injury (108) and damaged tissue requires neovascularization for recovery. Angiogenesis is mediated by hypoxia-inducible factor-1 (HIF-1) and the subsequent transcription of angiogenic growth factors. Endothelial cells and pericytes are stimulated by their cognate receptors to form new capillaries (109). Proliferation of endothelial cells to restore the sinusoid network in liver regeneration is stimulated by vascular endothelial growth factor (VEGF), FGF, stem cell factor, angiopoietins, and TGF-α (107). VEGF (110) and angiopoietin (111) are known to be involved in restoring vasculature in renal IRI. Aging is associated with a downregulation of growth factors, receptors, and receptor signal transduction (112–114). Moreover, a compromised induction of mitogenic signals may impair the initiation of repair and regeneration.

Bone marrow–derived circulating cells also assist in repair. Endothelial progenitor cells (EPCs) are now recognized to encompass two distinct populations: early and late outgrowth EPCs (115). Circulating EPCs assist in vascular recovery directly by incorporation (late outgrowth EPCs) or indirectly through the secretion of paracrine factors (early outgrowth EPCs) (116). These cells are recruited to the site of injury through chemotaxis, mediated by stromal-derived factor (SDF), a ligand for chemokine receptor CXCR4 (117) and CXCR7 (118), or by angiopoietin (119), a ligand for Tie-2. Mobilization of these cells occurs after ischemic injury (115, 117). With aging, there is a reduced expression of chemokines and angiogenic growth factors, caused by an impaired induction of HIF-1 that regulates SDF, angiopoietin, and VEGF (120), which may result in inhibited recruitment of EPCs by older organs. In an experimental model, homing of mesenchymal stem cells was impaired in older animals through SDF-1- and CXCR4-dependent mechanisms (121).

Immune cells do not only cause tissue injury but play also a critical role in the resolution of inflammation and promotion of repair. Depending on microenvironmental cues, macrophages can be activated to a proinflammatory or an anti-inflammatory state, a phenomenon called macrophage polarization. Based on in vitro studies, macrophages are classified in subsets where M1 macrophages represent a proinflammatory type and M2 macrophages an anti-inflammatory, tissue repair type. M1 macrophages can be induced by IFN-γ, LPS, and TLR ligation, whereas IL-4 and IL-13 promote M2 skewing (122). Clearance of apoptotic neutrophils and epithelial cells also induces M2 polarization with relevance in resolving inflammation (123). M2 macrophages produce IL-10, IL-1Ra, and TGF-β that suppress inflammation. M2 macrophages have also been shown to promote tubular proliferation subsequent to IRI (124). Macrophages may act through the production of growth factors (125) and secretion of Wnt protein (126), which is involved in promoting epithelial cell-cycle progression. Aging impairs macrophage polarization, subsequent to age-related changes of the microenvironment, including increased expression of inflammatory cytokines (127). Macrophages from the recipient are therefore likely to have an impaired ability to polarize to a M2 phenotype, which may inhibit resolution of inflammation and initiation of repair subsequent to the transplantation of old organs.

Reparative and Regenerative Capacity

Organs have distinct reparative and regenerative capabilities. The liver, for instance, regenerates excellently (128), demonstrated by a remarkable compensatory growth after surgical resection, although hepatic regenerative capacity diminishes with aging (129). Cardiac injury tends to result in fibrotic scarring (130). New heart cells can be generated, predominantly by cardiomyocytes during normal cell homeostasis as well as post-injury (131), but may also be generated by resident progenitor cells (132) or bone marrow cells (133). Both cardiomyocyte renewal in normal cell homeostasis (134) and the number of stem cells and progenitor cells decline with aging (135), suggesting an impaired regenerative capacity after injury. During the recovering phase of acute kidney injury, resident epithelial cells repopulate the zones of injury. This process involves epithelial de-differentiation of the renal tubular cells, spreading of the cells over the uncovered basement membrane, and proliferation and differentiation to complete repair (136). Of note, epithelial proliferation is impaired by aging (137), thus impacting the kidney’s ability to recover from injury. Cellular senescence has been implicated with regards to this impairment in IRI (19, 20). Although most of the newly generated cells appear to derive from surviving epithelial cells (138), resident progenitor cells and stem cells also have been identified as participants in the regenerative process (139). These are thought to contribute to renal repair by epithelial differentiation and proliferation. Moreover, there is an age-associated decline of progenitor cells (140), with cellular senescence assumed to be the main driver (141). Cellular senescence also affects endothelial cells and impairs their ability to proliferate, reducing angiogenic potential (142).

ORGAN PRESERVATION

As older organs are more prone to injury, optimal preservation appears of critical clinical significance. Cold static preservation has long been the standard method of preservation, but hypothermic machine perfusion is gaining ground. Machine perfusion has been implemented for kidney transplantation, and experimental and clinical pilot studies in machine perfusion for extrarenal organs appear promising (142–145). For cardiac transplantation, warm machine perfusion with oxygenated blood is currently being studied in a randomized trial (NCT00855712). A large trial has demonstrated that machine perfusion results in better outcomes of all deceased-donor kidneys (146, 147) with reduced rates and intensities for DGF and improved graft survival. Much attention has been given in the past years to machine perfusion of marginal organs, namely, for organs from expanded criteria kidney donors (ECD) or organs from donation after circulatory death (DCD). Three-year graft survival improved in renal transplantation, particularly when transplanting ECD kidneys (147–149). Donor age is a key component for the assessment of organ quality and a constituent of the ECD criteria. One-year graft survival rates for older grafts (>65 years) that incurred DGF demonstrated improvements with pulsatile perfusion, although an overall survival benefit of older grafts could not be established in this small cohort study (150). A phase 2 trial exploring the potential of machine perfusion of livers from extended criteria donors, defined by age greater than 65 years, macrosteatosis, impaired liver function, and hepatitis C with macrosteatosis, is currently in progress (NCT01274520). Preliminary data showed a low incidence of allograft dysfunction (151). A lung machine perfusion study was designed to include only high-risk donor lungs, defined by the following criteria: PaO2-FIO2 ratio less than 300, pulmonary edema, poor lung deflation and inflation, blood transfusions exceeding 10 units, and DCD (143). In this small, nonrandomized study, outcomes of those marginal lungs were comparable to standard criteria lungs. These data suggest that marginal organs, frequently those from older donors, may benefit most from machine perfusion.

Our understanding of the mechanistic aspects of pulsatile perfusion is currently limited. The rationale behind machine perfusion has typically been linked to the supply with nutrients and substrates, thereby preserving ATP levels and ameliorating subsequent injury (152). More recently, the importance of flow for maintaining endothelial cell function has been described (153). Shear stress-dependent expression of Kruppel-like factor 2 (KLF2) decays rapidly after flow cessation. KLF2 is necessary for the inhibition of proinflammatory cytokines, adhesion molecules, and prothrombotic genes expressed by endothelial cells. Sustaining endothelial homeostasis and the suppression of inflammation may therefore be another mechanism by which machine perfusion mediates superior graft outcomes (154). Statins have been shown to induce KLF2 expression and reversed the detrimental effects of flow cessation when added to the preservation solution (153).

ORGAN ALLOCATION

Optimal allocation of organs appears imperative during times of limited supply, thus the current discard of approximately 50% of ECD kidneys in the United States is alarming (155). The recently approved revised kidney allocation system attempts to address the high discard rate by improving allocation efficiency. Under this newly implemented allocation system, 20% of the highest quality kidneys, determined by the Kidney Donor Profile Index, will be allocated to candidates with the highest estimated posttransplant survival. The remaining 80% will be allocated to candidates within 15 years of the donor range. The introduction of the ESP in Europe has already resulted in increased utilization of older organs. In 2012, 25% of deceased donors in the Eurotransplant area were greater than 65 years old (Eurotransplant Annual Report 2012). Moreover, outcomes when utilizing older kidneys in the ESP were comparable to those of younger kidneys transplanted into older recipients outside the program (156). Of note, older recipients have been shown to mount a less potent immune response (4), which may offset the increased immunogenicity of older donor organs. Levels of cellular senescence in pretransplant renal biopsies have been shown to more accurately predict postoperative function than donor age alone (157) and have thus been proposed as an additional component assessing transplant outcome (158).

FUTURE PERSPECTIVE

Developing strategies minimizing organ damage and improving outcomes of older organs should receive high priority. Adaptations of practical nature, such as an optimization of allocation protocols to limit cold ischemic times, have already shown good outcomes in the ESP and could be implemented on a broader scale. Moreover, widespread implementation of machine perfusion could also aid in better preserving damage-prone older organs. Particularly appealing, however, is the prospect of directly targeting age-dependent deficiencies, and pharmacological interventions could be deployed to address impaired cellular stress responses. Ex vivo conditioning may provide an additional avenue of interest. Presently, most interventions remain on an experimental level, but as age-specific research progresses, we expect a clinical demand to implement age- and injury-specific preservation methods.

CONCLUDING REMARKS

Aging affects a majority of organs currently utilized for transplantation. Organ damage caused by physiological aging or pathological processes accumulate and impact transplant outcomes. An increased sensitivity to IRI and enhanced immunogenicity aggravate graft injury. Additionally, defective cellular stress and maintenance systems and a diminished cell proliferation debilitate the capacity for repair and regeneration. It is to be expected that the rate of older donors will furthermore increase in the future. At the same time, discard rates of organs, particularly from older donors, are alarming. Age-specific research will therefore become even more clinically relevant to guide policies for organ recovery, distribution, and to optimize treatments.

REFERENCES

1. Noppakun K, Cosio FG, Dean PG, et al. Living donor age and kidney transplant outcomes. Am J Transplant 2011; 11: 1279.
2. Moers C, Kornmann NS, Leuvenink HG, et al. The influence of deceased donor age and old-for-old allocation on kidney transplant outcome. Transplantation 2009; 88: 542.
3. de Fijter JW, Mallat MJ, Doxiadis II, et al. Increased immunogenicity and cause of graft loss of old donor kidneys. J Am Soc Nephrol 2001; 12: 1538.
4. Tullius SG, Milford E. Kidney allocation and the aging immune response. N Engl J Med 2011; 364: 1369.
5. Yarlagadda SG, Coca SG, Formica RN Jr, et al. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis. Nephrol Dial Transplant 2009; 24: 1039.
6. Guedes AM, Malheiro J, Fonseca I, et al. Over ten-year kidney graft survival determinants. Int J Nephrol 2012; 2012: 302974.
7. Chavalitdhamrong D, Gill J, Takemoto S, et al. Patient and graft outcomes from deceased kidney donors age 70 years and older: an analysis of the organ procurement transplant Network/United network of organ sharing database. Transplantation 2008; 85: 1573.
8. Feng S, Goodrich NP, Bragg-Gresham JL, et al. Characteristics associated with liver graft failure: the concept of a donor risk index. Am J Transplant 2006; 6: 783.
9. Cameron AM, Ghobrial RM, Hiatt JR, et al. Effect of nonviral factors on hepatitis C recurrence after liver transplantation. Ann Surg 2006; 244: 563.
10. Machicao VI, Bonatti H, Krishna M, et al. Donor age affects fibrosis progression and graft survival after liver transplantation for hepatitis C. Transplantation 2004; 77: 84.
11. Berenguer M, Aguilera V, Prieto M, et al. Worse recent efficacy of antiviral therapy in liver transplant recipients with recurrent hepatitis C: impact of donor age and baseline cirrhosis. Liver Transpl 2009; 15: 738.
12. Stehlik J, Feldman DS, Brown RN, et al. Interactions among donor characteristics influence post-transplant survival: a multi-institutional analysis. J Heart Lung Transplant 2010; 29: 291.
13. Axelrod DA, Sung RS, Meyer KH, et al. Systematic evaluation of pancreas allograft quality, outcomes and geographic variation in utilization. Am J Transplant 2010; 10: 837.
14. Bittle GJ, Sanchez PG, Kon ZN, et al. The use of lung donors older than 55 years: a review of the United Network of Organ Sharing database. J Heart Lung Transplant 2013; 32: 760.
15. Dahlman S, Jeppsson A, Schersten H, et al. Expanding the donor pool: lung transplantation with donors 55 years and older. Transplant Proc 2006; 38: 2691.
16. Pizanis N, Heckmann J, Tsagakis K, et al. Lung transplantation using donors 55 years and older: is it safe or just a way out of organ shortage? Eur J Cardiothorac Surg 2010; 38: 192.
17. Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol 2007; 8: 729.
18. Jeyapalan JC, Ferreira M, Sedivy JM, et al. Accumulation of senescent cells in mitotic tissue of aging primates. Mech Ageing Dev 2007; 128: 36.
19. Westhoff JH, Schildhorn C, Jacobi C, et al. Telomere shortening reduces regenerative capacity after acute kidney injury. J Am Soc Nephrol 2010; 21: 327.
20. Braun H, Schmidt BM, Raiss M, et al. Cellular senescence limits regenerative capacity and allograft survival. J Am Soc Nephrol 2012; 23: 1467.
21. Baker DJ, Wijshake T, Tchkonia T, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011; 479: 232.
22. Franceschi C, Bonafe M, Valensin S, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 2000; 908: 244.
23. Vasto S, Candore G, Balistreri CR, et al. Inflammatory networks in ageing, age-related diseases and longevity. Mech Ageing Dev 2007; 128: 83.
24. Kregel KC, Zhang HJ. An integrated view of oxidative stress in aging: basic mechanisms, functional effects, and pathological considerations. Am J Physiol Regul Integr Comp Physiol 2007; 292: R18.
25. Chung HY, Sung B, Jung KJ, et al. The molecular inflammatory process in aging. Antioxid Redox Signal 2006; 8: 572.
26. Tschopp J, Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat Rev Immunol 2010; 10: 210.
27. Rodier F, Coppe JP, Patil CK, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol 2009; 11: 973.
28. Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal 2012; 24: 835.
29. Olivetti G, Melissari M, Capasso JM, et al. Cardiomyopathy of the aging human heart. myocyte loss and reactive cellular hypertrophy. Circ Res 1991; 68: 1560.
30. Wynne HA, Cope LH, Mutch E, et al. The effect of age upon liver volume and apparent liver blood flow in healthy man. Hepatology 1989; 9: 297.
31. Nyengaard JR, Bendtsen TF. Glomerular number and size in relation to age, kidney weight, and body surface in normal man. Anat Rec 1992; 232: 194.
32. Barbari A, Stephan A, Masri MA, et al. Nephron mass in kidney transplantation. Transplant Proc 2002; 34: 2401.
33. Terasaki PI, Koyama H, Cecka JM, et al. The hyperfiltration hypothesis in human renal transplantation. Transplantation 1994; 57: 1450.
34. De Vusser K, Lerut E, Kuypers D, et al. The predictive value of kidney allograft baseline biopsies for long-term graft survival. J Am Soc Nephrol 2013; 24: 1913.
35. Argo CK, Northup PG, Al-Osaimi AM, et al. Systematic review of risk factors for fibrosis progression in non-alcoholic steatohepatitis. J Hepatol 2009; 51: 371.
36. Vetelainen R, van Vliet A, Gouma DJ, et al. Steatosis as a risk factor in liver surgery. Ann Surg 2007; 245: 20.
37. Stehlik J, Edwards LB, Kucheryavaya AY, et al. The registry of the International Society for Heart and Lung Transplantation: 29th official adult heart transplant report—2012. J Heart Lung Transplant 2012; 31: 1052.
38. Nagji AS, Hranjec T, Swenson BR, et al. Donor age is associated with chronic allograft vasculopathy after adult heart transplantation: implications for donor allocation. Ann Thorac Surg 2010; 90: 168.
39. Schmauss D, Weis M. Cardiac allograft vasculopathy: recent developments. Circulation 2008; 117: 2131.
40. Tuzcu EM, Kapadia SR, Tutar E, et al. High prevalence of coronary atherosclerosis in asymptomatic teenagers and young adults: evidence from intravascular ultrasound. Circulation 2001; 103: 2705.
41. Li H, Tanaka K, Anzai H, et al. Influence of pre-existing donor atherosclerosis on the development of cardiac allograft vasculopathy and outcomes in heart transplant recipients. J Am Coll Cardiol 2006; 47: 2470.
42. Kalogeris T, Baines CP, Krenz M, et al. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 2012; 298: 229.
43. Navarro A, Boveris A. The mitochondrial energy transduction system and the aging process. Am J Physiol Cell Physiol 2007; 292: C670.
44. Short KR, Bigelow ML, Kahl J, et al. Decline in skeletal muscle mitochondrial function with aging in humans. Proc Natl Acad Sci U S A 2005; 102: 5618.
45. Lagouge M, Larsson NG. The role of mitochondrial DNA mutations and free radicals in disease and ageing. J Intern Med 2013; 273: 529.
46. Lesnefsky EJ, Hoppel CL. Ischemia-reperfusion injury in the aged heart: role of mitochondria. Arch Biochem Biophys 2003; 420: 287.
47. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med 2007; 357: 1121.
48. Halestrap AP. What is the mitochondrial permeability transition pore? J Mol Cell Cardiol 2009; 46: 821.
49. Haworth RA, Hunter DR. The Ca2+-induced membrane transition in mitochondria. II. Nature of the Ca2+ trigger site. Arch Biochem Biophys 1979; 195: 460.
50. Kim JS, Jin Y, Lemasters JJ. Reactive oxygen species, but not Ca2+ overloading, trigger pH- and mitochondrial permeability transition-dependent death of adult rat myocytes after ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2006; 290: H2024.
51. Ananthakrishnan R, Kaneko M, Hwang YC, et al. Aldose reductase mediates myocardial ischemia-reperfusion injury in part by opening mitochondrial permeability transition pore. Am J Physiol Heart Circ Physiol 2009; 296: H333.
52. Pi Y, Goldenthal MJ, Marin-Garcia J. Mitochondrial channelopathies in aging. J Mol Med (Berl) 2007; 85: 937.
53. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 2010; 10: 826.
54. Mills KH. TLR-dependent T cell activation in autoimmunity. Nat Rev Immunol. 2011; 11: 807.
55. Kruger B, Krick S, Dhillon N, et al. Donor toll-like receptor 4 contributes to ischemia and reperfusion injury following human kidney transplantation. Proc Natl Acad Sci U S A 2009; 106: 3390.
56. Palmer SM, Burch LH, Mir S, et al. Donor polymorphisms in toll-like receptor-4 influence the development of rejection after renal transplantation. Clin Transplant 2006; 20: 30.
57. Agrawal A, Agrawal S, Cao JN, et al. Altered innate immune functioning of dendritic cells in elderly humans: a role of phosphoinositide 3-kinase-signaling pathway. J Immunol 2007; 178: 6912.
58. Butcher SK, Chahal H, Nayak L, et al. Senescence in innate immune responses: reduced neutrophil phagocytic capacity and CD16 expression in elderly humans. J Leukoc Biol 2001; 70: 881.
59. Swift ME, Burns AL, Gray KL, et al. Age-related alterations in the inflammatory response to dermal injury. J Invest Dermatol 2001; 117: 1027.
60. Aprahamian T, Takemura Y, Goukassian D, et al. Ageing is associated with diminished apoptotic cell clearance in vivo. Clin Exp Immunol 2008; 152: 448.
61. Morelli AE, Larregina AT. Apoptotic cell-based therapies against transplant rejection: role of recipient’s dendritic cells. Apoptosis 2010; 15: 1083.
62. Sancho D, Joffre OP, Keller AM, et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 2009; 458: 899.
63. Maegawa S, Hinkal G, Kim HS, et al. Widespread and tissue specific age-related DNA methylation changes in mice. Genome Res 2010; 20: 332.
64. Christensen BC, Houseman EA, Marsit CJ, et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CpG island context. PLoS Genet 2009; 5: e1000602.
65. Agrawal A, Tay J, Yang GE, et al. Age-associated epigenetic modifications in human DNA increase its immunogenicity. Aging (Albany NY) 2010; 2: 93.
66. Castle SC, Uyemura K, Crawford W, et al. Antigen presenting cell function is enhanced in healthy elderly. Mech Ageing Dev 1999; 107: 137.
67. Ordemann R, Hutchinson R, Friedman J, et al. Enhanced allostimulatory activity of host antigen-presenting cells in old mice intensifies acute graft-versus-host disease. J Clin Invest 2002; 109: 1249.
68. Rippe C, Blimline M, Magerko KA, et al. MicroRNA changes in human arterial endothelial cells with senescence: relation to apoptosis, eNOS and inflammation. Exp Gerontol 2012; 47: 45.
69. Calabrese V, Cornelius C, Cuzzocrea S, et al. Hormesis, cellular stress response and vitagenes as critical determinants in aging and longevity. Mol Aspects Med 2011; 32: 279.
70. Hahn JS, Hu Z, Thiele DJ, et al. Genome-wide analysis of the biology of stress responses through heat shock transcription factor. Mol Cell Biol 2004; 24: 5249.
71. Kelly KJ. Heat shock (stress response) proteins and renal ischemia/reperfusion injury. Contrib Nephrol 2005; 148: 86.
72. Kourtis N, Tavernarakis N. Cellular stress response pathways and ageing: intricate molecular relationships. EMBO J 2011; 30: 2520.
73. Macario AJ, Conway de Macario E. Sick chaperones, cellular stress, and disease. N Engl J Med 2005; 353: 1489.
74. Okaya T, Blanchard J, Schuster R, et al. Age-dependent responses to hepatic ischemia/reperfusion injury. Shock 2005; 24: 421.
75. Ollinger R, Pratschke J. Role of heme oxygenase-1 in transplantation. Transpl Int 2010; 23: 1071.
76. Kotsch K, Martins PN, Klemz R, et al. Heme oxygenase-1 ameliorates ischemia/reperfusion injury by targeting dendritic cell maturation and migration. Antioxid Redox Signal 2007; 9: 2049.
77. Ferenbach DA, Nkejabega NC, McKay J, et al. The induction of macrophage hemeoxygenase-1 is protective during acute kidney injury in aging mice. Kidney Int 2011; 79: 966.
78. Tullius SG, Nieminen-Kelha M, Reutzel-Selke A, et al. Improvement of long-term function in renal allografts from ‘marginal donors’ following the induction of heme-oxygenase-1. Transplant Proc 2001; 33: 1160.
79. Broere F, van der Zee R, van Eden W. Heat shock proteins are no DAMPs, rather ‘DAMPERs’. Nat Rev Immunol 2011; 11: 565; author reply 565.
80. Wachstein J, Tischer S, Figueiredo C, et al. HSP70 enhances immunosuppressive function of CD4(+)CD25(+)FoxP3(+) T regulatory cells and cytotoxicity in CD4(+)CD25(−) T cells. PLoS One 2012; 7: e51747.
81. Borges TJ, Porto BN, Teixeira CA, et al. Prolonged survival of allografts induced by mycobacterial Hsp70 is dependent on CD4+CD25+ regulatory T cells. PLoS One 2010; 5: e14264.
82. Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest 2005; 115: 2656.
83. Brodsky JL. Cleaning up: ER-associated degradation to the rescue. Cell 2012; 151: 1163.
84. Haynes CM, Ron D. The mitochondrial UPR—protecting organelle protein homeostasis. J Cell Sci 2010; 123 (Pt 22): 3849.
85. Szegezdi E, Logue SE, Gorman AM, et al. Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO Rep 2006; 7: 880.
86. Inagi R. Endoplasmic reticulum stress in the kidney as a novel mediator of kidney injury. Nephron Exp Nephrol 2009; 112: e1.
87. Kaplowitz N, Than TA, Shinohara M, et al. Endoplasmic reticulum stress and liver injury. Semin Liver Dis 2007; 27: 367.
88. Toth A, Nickson P, Mandl A, et al. Endoplasmic reticulum stress as a novel therapeutic target in heart diseases. Cardiovasc Hematol Disord Drug Targets 2007; 7: 205.
89. Naidoo N. ER and aging-protein folding and the ER stress response. Ageing Res Rev 2009; 8: 150.
90. Breusing N, Grune T. Regulation of proteasome-mediated protein degradation during oxidative stress and aging. Biol Chem 2008; 389: 203.
91. Ishihara Y, Hamaguchi A, Sekine M, et al. Accumulation of cytochrome P450 induced by proteasome inhibition during cardiac ischemia. Arch Biochem Biophys 2012; 527: 16.
92. Tian Z, Zheng H, Li J, et al. Genetically induced moderate inhibition of the proteasome in cardiomyocytes exacerbates myocardial ischemia-reperfusion injury in mice. Circ Res 2012; 111: 532.
93. Huber JM, Tagwerker A, Heininger D, et al. The proteasome inhibitor bortezomib aggravates renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2009; 297: F451.
94. Pye J, Ardeshirpour F, McCain A, et al. Proteasome inhibition ablates activation of NF-kappa B in myocardial reperfusion and reduces reperfusion injury. Am J Physiol Heart Circ Physiol 2003; 284: H919.
95. Glembotski CC. Clarifying the cardiac proteasome paradox: protein quality control. Circ Res 2012; 111: 509.
96. Walsh RC, Alloway RR, Girnita AL, et al. Proteasome inhibitor-based therapy for antibody-mediated rejection. Kidney Int 2012; 81: 1067.
97. Murrow L, Debnath J. Autophagy as a stress-response and quality-control mechanism: implications for cell injury and human disease. Annu Rev Pathol 2013; 8: 105.
98. Jiang M, Wei Q, Dong G, et al. Autophagy in proximal tubules protects against acute kidney injury. Kidney Int 2012; 82: 1271.
99. Wang JH, Ahn IS, Fischer TD, et al. Autophagy suppresses age-dependent ischemia and reperfusion injury in livers of mice. Gastroenterology 2011; 141: 2188,2199.e6.
100. Sala-Mercado JA, Wider J, Undyala VV, et al. Profound cardioprotection with chloramphenicol succinate in the swine model of myocardial ischemia-reperfusion injury. Circulation 2010; 122 (11 Suppl): S179.
101. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy–inflammation–cell death axis in organismal aging. Science 2011; 333: 1109.
102. Wang JH, Behrns KE, Leeuwenburgh C, et al. Critical role of autophage in ischemia/reperfusion injury to aged livers. Autophagy 2012; 8: 140.
103. Karp SJ. Clinical implications of advances in the basic science of liver repair and regeneration. Am J Transplant 2009; 9: 1973.
104. Nelson PJ, Rees AJ, Griffin MD, et al. The renal mononuclear phagocytic system. J Am Soc Nephrol 2012; 23: 194.
105. Humphreys BD, Bonventre JV. Mesenchymal stem cells in acute kidney injury. Annu Rev Med 2008; 59: 311.
106. Liu KD, Brakeman PR. Renal repair and recovery. Crit Care Med 2008; 36 (4 Suppl): S187.
107. Michalopoulos GK. Liver regeneration. J Cell Physiol 2007; 213: 286.
108. Basile DP. The endothelial cell in ischemic acute kidney injury: implications for acute and chronic function. Kidney Int 2007; 72: 151.
109. Rey S, Semenza GL. Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling. Cardiovasc Res 2010; 86: 236.
110. Leonard EC, Friedrich JL, Basile DP. VEGF-121 preserves renal microvessel structure and ameliorates secondary renal disease following acute kidney injury. Am J Physiol Renal Physiol 2008; 295: F1648.
111. Regner KR, Roman RJ. Role of medullary blood flow in the pathogenesis of renal ischemia-reperfusion injury. Curr Opin Nephrol Hypertens 2012; 21: 33.
112. Shiraha H, Gupta K, Drabik K, et al. Aging fibroblasts present reduced epidermal growth factor (EGF) responsiveness due to preferential loss of EGF receptors. J Biol Chem 2000; 275: 19343.
113. Tran KT, Rusu SD, Satish L, et al. Aging-related attenuation of EGF receptor signaling is mediated in part by increased protein tyrosine phosphatase activity. Exp Cell Res 2003; 289: 359.
114. Komi-Kuramochi A, Kawano M, Oda Y, et al. Expression of fibroblast growth factors and their receptors during full-thickness skin wound healing in young and aged mice. J Endocrinol 2005; 186: 273.
115. Patschan D, Patschan S, Muller GA. Endothelial progenitor cells in acute ischemic kidney injury: strategies for increasing the cells’ renoprotective competence. Int J Nephrol 2011; 2011: 828369.
116. Yoder MC, Mead LE, Prater D, et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood 2007; 109: 1801.
117. Togel F, Isaac J, Hu Z, et al. Renal SDF-1 signals mobilization and homing of CXCR4-positive cells to the kidney after ischemic injury. Kidney Int 2005; 67: 1772.
118. Mazzinghi B, Ronconi E, Lazzeri E, et al. Essential but differential role for CXCR4 and CXCR7 in the therapeutic homing of human renal progenitor cells. J Exp Med 2008; 205: 479.
119. Patschan D, Backhaus R, Elle HJ, et al. Angiopoietin-2 modulates eEOC-mediated renoprotection in AKI in a dose-dependent manner. J Nephrol 2013; 26: 667.
120. Bosch-Marce M, Okuyama H, Wesley JB, et al. Effects of aging and hypoxia-inducible factor-1 activity on angiogenic cell mobilization and recovery of perfusion after limb ischemia. Circ Res 2007; 101: 1310.
121. Kyriakou C, Rabin N, Pizzey A, et al. Factors that influence short-term homing of human bone marrow-derived mesenchymal stem cells in a xenogeneic animal model. Haematologica 2008; 93: 1457.
122. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012; 122: 787.
123. Lech M, Anders HJ. Macrophages and fibrosis: how resident and infiltrating mononuclear phagocytes orchestrate all phases of tissue injury and repair. Biochim Biophys Acta 2013; 1832: 989.
124. Lee S, Huen S, Nishio H, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol 2011; 22: 317.
125. Menke J, Iwata Y, Rabacal WA, et al. CSF-1 signals directly to renal tubular epithelial cells to mediate repair in mice. J Clin Invest 2009; 119: 2330.
126. Lin SL, Li B, Rao S, et al. Macrophage Wnt7b is critical for kidney repair and regeneration. Proc Natl Acad Sci U S A 2010; 107: 4194.
127. Mahbub S, Deburghgraeve CR, Kovacs EJ. Advanced age impairs macrophage polarization. J Interferon Cytokine Res 2012; 32: 18.
128. Diehl AM, Chute J. Underlying potential: cellular and molecular determinants of adult liver repair. J Clin Invest 2013; 123: 1858.
129. Timchenko NA. Aging and liver regeneration. Trends Endocrinol Metab 2009; 20: 171.
130. Baddour JA, Sousounis K, Tsonis PA. Organ repair and regeneration: an overview. Birth Defects Res C Embryo Today 2012; 96: 1.
131. Senyo SE, Steinhauser ML, Pizzimenti CL, et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 2013; 493: 433.
132. Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003; 114: 763.
133. Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001; 410: 701.
134. Bergmann O, Bhardwaj RD, Bernard S, et al. Evidence for cardiomyocyte renewal in humans. Science 2009; 324: 98.
135. Ballard VL, Edelberg JM. Stem cells for cardiovascular repair—the challenges of the aging heart. J Mol Cell Cardiol 2008; 45: 582.
136. Bonventre JV. Dedifferentiation and proliferation of surviving epithelial cells in acute renal failure. J Am Soc Nephrol 2003; 14 (Suppl 1): S55.
137. Schmitt R, Marlier A, Cantley LG. Zag expression during aging suppresses proliferation after kidney injury. J Am Soc Nephrol 2008; 19: 2375.
138. Humphreys BD, Valerius MT, Kobayashi A, et al. Intrinsic epithelial cells repair the kidney after injury. Cell Stem Cell 2008; 2: 284.
139. Oliver JA, Klinakis A, Cheema FH, et al. Proliferation and migration of label-retaining cells of the kidney papilla. J Am Soc Nephrol 2009; 20: 2315.
140. Miya M, Maeshima A, Mishima K, et al. Age-related decline in label-retaining tubular cells: implication for reduced regenerative capacity after injury in the aging kidney. Am J Physiol Renal Physiol 2012; 302: F694.
141. Schmitt R, Cantley LG. The impact of aging on kidney repair. Am J Physiol Renal Physiol 2008; 294: F1265.
142. Lahteenvuo J, Rosenzweig A. Effects of aging on angiogenesis. Circ Res 2012; 110: 1252.
143. Cypel M, Yeung JC, Liu M, et al. Normothermic ex vivo lung perfusion in clinical lung transplantation. N Engl J Med 2011; 364: 1431.
144. Taylor MJ, Baicu S, Leman B, et al. Twenty-four hour hypothermic machine perfusion preservation of porcine pancreas facilitates processing for islet isolation. Transplant Proc 2008; 40: 480.
145. Guarrera JV, Henry SD, Samstein B, et al. Hypothermic machine preservation in human liver transplantation: the first clinical series. Am J Transplant 2010; 10: 372.
146. Moers C, Smits JM, Maathuis MH, et al. Machine perfusion or cold storage in deceased-donor kidney transplantation. N Engl J Med 2009; 360: 7.
147. Moers C, Pirenne J, Paul A, et al., Machine Preservation Trial Study Group. Machine perfusion or cold storage in deceased-donor kidney transplantation. N Engl J Med 2012; 366: 770.
148. Treckmann J, Moers C, Smits JM, et al. Machine perfusion versus cold storage for preservation of kidneys from expanded criteria donors after brain death. Transpl Int 2011; 24: 548.
149. Gallinat A, Moers C, Smits JM, et al. Machine perfusion versus static cold storage in expanded criteria donor kidney transplantation: 3-year follow-up data. Transpl Int 2013; 26: E52.
150. Gallinat A, Moers C, Treckmann J, et al. Machine perfusion versus cold storage for the preservation of kidneys from donors ≥ 65 years allocated in the Eurotransplant Senior Programme. Nephrol Dial Transplant 2012; 27: 4458.
151. Guarrera JV. Assist devices: machine preservation of extended criteria donors. Liver Transpl 2012; 18 (Suppl 2): S31.
152. St Peter SD, Imber CJ, Friend PJ. Liver and kidney preservation by perfusion. Lancet 2002; 359: 604.
153. Gracia-Sancho J, Villarreal G Jr, Zhang Y, et al. Flow cessation triggers endothelial dysfunction during organ cold storage conditions: strategies for pharmacologic intervention. Transplantation 2010; 90: 142.
154. Gallinat A, Fox M, Luer B, et al. Role of pulsatility in hypothermic reconditioning of porcine kidney grafts by machine perfusion after cold storage. Transplantation 2013; 96: 142.
155. Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2011 annual data report: kidney. Am J Transplant 2013; 13 (Suppl 1): 11.
156. Giessing M, Fuller TF, Friedersdorff F, et al. Outcomes of transplanting deceased-donor kidneys between elderly donors and recipients. J Am Soc Nephrol 2009; 20: 37.
157. Koppelstaetter C, Schratzberger G, Perco P, et al. Markers of cellular senescence in zero hour biopsies predict outcome in renal transplantation. Aging Cell 2008; 7: 491.
158. Gingell-Littlejohn M, McGuinness D, McGlynn LM, et al. Pre-transplant CDKN2A expression in kidney biopsies predicts renal function and is a future component of donor scoring criteria. PLoS One 2013; 8: e68133.
Keywords:

Aging; Transplantation; Injury; Repair

© 2014 by Lippincott Williams & Wilkins