Open Access

Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review)

  • Authors:
    • Maksymilian Baryła
    • Michał Skrzycki
    • Roman Danielewicz
    • Maciej Kosieradzki
    • Marta Struga
  • View Affiliations

  • Published online on: September 25, 2024     https://doi.org/10.3892/ijmm.2024.5431
  • Article Number: 107
  • Copyright: © Baryła et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To meet the demand for kidney transplants (KTx), organs are frequently retrieved not only from standard criteria donors (SCD; a donor who is aged <50 years and suffered brain death from any number of causes, such as traumatic injuries or a stroke) but also from expanded criteria donors (any donor aged >60 years or donors aged >50 years with two of the following: A history of high blood pressure, a creatinine serum level ≥1.5 mg/dl or death resulting from a stroke). This comes at the cost of a higher risk of primary non‑function (the permanent hyperkalemia, hyperuremia and fluid overload that result in the need for continuous dialysis after KTx), delayed graft function (the need for dialysis session at least once during the first week after KTx), earlier graft loss and urinary complications (vesico‑ureteral reflux, obstruction of the vesico‑ureteral anastomosis, urine leakage). At present, there are no commercially available diagnostic tools for assessing kidney quality prior to KTx. Currently available predictive models based on clinical data, such as the Kidney Donor Profile Index, are insufficient. One promising option is the application of perfusion solutions for protein biomarkers of kidney quality and predictors of short‑ and long‑term outcomes. However, to date, protein markers that can be detected with ELISA, western blotting and cytotoxic assays have not been identified to be a beneficial predictors of kidney quality. These include lactate dehydrogenases, glutathione S‑transferases, fatty acid binding proteins, extracellular histones, IL‑18, neutrophil gelatinase‑associated lipocalin, MMPs and kidney injury molecule‑1. However, novel methods, including liquid chromatography‑mass spectrometry (LC‑MS) and microarrays, allow the analysis of all renal proteins suspended/dissolved in the acellular preservation solution used for kidney storage before KTx (including hypothermic machine perfusion as one of kidney storage methods) e.g. Belzer University of Wisconsin. Recent proteomic studies utilizing LC‑MS have identified complement pathway elements (C3, C1QB, C4BPA, C1S, C1R and C1RL), desmoplakin, blood coagulation pathway elements and immunoglobulin heavy variable 2‑26 to be novel predictors of kidney quality before transplantation. This was because they were found to correlate with estimated glomerular filtration rate at 3 and 12 months after kidney transplantation. However, further proteomic studies focusing on distinct markers obtained from hypothermic and normothermic machine perfusion are needed to confirm their predictive value and to improve kidney storage methods. Therefore, the present literature review from PubMed, Scopus, Embase and Web of Science was performed with the aims of summarizing the current knowledge on the most frequently studied single protein biomarkers. In addition, novel analytical methods and insights into organ injury during preservation were documented, where future directions in assessing organ quality before kidney transplantation were also discussed.

Background

To meet the demand for kidney transplants (KTx), organs are frequently retrieved not only from standard criteria donors (SCD; a donor who is aged <50 years and suffered brain death from any number of causes, such as traumatic injuries or a stroke) but also from expanded criteria donors [ECDs; any donor aged >60 years or a donors aged ≥50 years with two of the following: a history of high blood pressure, a creatinine serum level ≥1.5 mg/dl or death resulting from a stroke]. This comes at the cost of a higher risk of primary non-function (PNF; the permanent hyperkalemia, hyperuremia and fluid overload that result in the need for continuous dialysis after KTx), delayed graft function (DGF; the need for dialysis session at least once during the first week after KTx), earlier graft loss and urinary complications (vesico-ureteral reflux, obstruction of the vesico-ureteral anastomosis, urine leakage) (1-3). An increased risk of graft failure and graftectomy can result in recipient immunization, leading to increasingly difficult donor selection and higher risk of consecutive graft failure (4,5). However, there is also evidence that graft survival is unaffected by the procurement of kidneys from ECDs compared to kidneys retrieved from SCDs (6,7).

Currently, there is no commercially available diagnostic tool for the quality assessment of the kidney before KTx (8). To this end, the majority of medical centers will typically perform a 0-h biopsy (9,10), but the results only become available days after surgery and assessment is limited to a limited part of the kidney. A 0-h biopsy does not provide an insight into the condition of the entire organ and cannot be used to predict its function after KTx. The period of kidney storage should be as short as possible and the decision on KTx should be made during this period. However, predictive models based on clinical data, such as the Kidney Donor Profile Index (KDPI) (11,12), the continuous kidney donor risk index (13) and deceased donor score (14), are insufficient for assessing the condition of potentially transplanted kidneys. A number of preservation and perfusion solutions have been assessed as possible novel biomarkers of kidney quality and predictors of short- and long-term outcomes. These include metabolites, enzymes, complement-related molecules, antibodies, histones and cytoskeletal elements released by tubular, interstitial, immunological and epithelial cells (15-17). However, none of these previous studies have contributed to the identification of biomarkers that are beneficial for assessing the value of the kidney. This is due to the lack of randomized controlled trials in this area to confirm the utility of potential biomarkers. Some of the markers, such as lactate dehydrogenase (LDH) or neutrophil gelatinase-associated lipocalin (NGAL), are not tissue specific (18,19). In addition, the methods used to detect the majority of the candidates are time-consuming, requiring qualified personnel, expensive equipments and reagents. Therefore, there remain to be a demand for a rapid, non-invasive diagnostic tool and reliable predictive model for assessing the post-transplant function of a sub-optimal donor kidney.

Studies on single protein biomarkers

The analysis of biological samples inevitably involves the detection of a wide range of proteins. In such scenarios, two primary pathways for analysis emerge, namely targeted and untargeted. The former involves targeting specific groups of proteins, such as immunoglobulins or individual proteins based on existing knowledge, assessing their relevance to the phenomena in question. To detect these known individual particles, targeted analytical methods were used in previous studies. These included ELISA, western blotting and enzymatic/cytotoxic assays.

Proteins released after cell disruption
LDH

LDH was one of the first proteins to be studied in the renal perfusate. This cytosolic enzyme is responsible for the conversion of pyruvate to lactate by NADH under normoxic and hypoxic conditions (20). In total, five tetrameric isoenzymes consisting of LDHA and LDHB subunits have been previously described (18,21,22). They differ in their degree of substrate affinity, electrophoretic mobility, inhibition constants and tissue distribution (18). In an experimental rat model, LDHA was previously found to be mainly present in the proximal renal tubules, whilst LDHB was found to be mainly expressed in the distal part of the nephron (23).

In relation to KTx, LDH has been documented to serve as a non-specific marker of cell injury (18). Perfusate LDH activity is not applicable in the assessment of renal function prior to KTx. Despite the studies showing a positive association between LDH concentration and DGF (15,24-26), there are no randomized controlled trials that have confirmed the viability of LDH measurements for the assessment of kidney quality. Furthermore, LDH is not a kidney-specific biomarker (18,27,28). Guzzi et al (15) previously found that besides glutathione S-transferase (GST), LDH levels were the most commonly reported parameter in the renal perfusate. Elevated LDH levels have been reported to associate positively with DGF in cases of donation after cardiac death (DCDs) (24-26), but not in all cases of donations after brain death (DBDs) (29,30). According to previous studies, (15,24-26,31,32) where LDH levels were measured only in perfusate from DCDs, LDH levels were found to be positively associated with DGF in all cases. By contrast, in studies describing the measurement of LDH levels in the perfusate of kidneys from DBDs, a positive association between DGF and LDH levels was only found in two (29,30) of the four selected studies found (15). In addition, this meta-analysis (15) also showed that LDH activity should not be considered as a marker of PNF in patients with DCDs grafts. Further analysis of cases of DBDs and DCDs demonstrated that although the LDH levels were associated positively with DGF in the majority of the included studies, following multivariable analysis only one study showed a weak association (32). In another meta-analysis previously conducted by Bhangoo et al (16), perfusate LDH activity was found to be a predictor of DGF, PNF and graft failure (GF). However, this previous analysis lacked a donor-type subgroup assessment (16). A number of the included studies revealed a significant association between LDH activity and DGF, PNF (31,33). Among the different types of donors, LDH activity was the highest in kidneys from DCD cases, which reflected the greatest extent of damage prior to KTx among the studied organs (34). Perfusate assessment has been used to estimate renal cell injury after ischemia in a previous study (29), where LDH levels were previously found to be increased during the ischemic period (29). This phenomenon also associated positively with damage to renal tubular cells detected by electron microscopy. The lesions found include mitochondrial swelling and loss of detail in the inner cristae, swelling and detachment of the basolateral tubular membrane connective tissue (29).

GSTs

GSTs form one of the most frequently studied groups of biomarkers obtained from the renal hypothermic machine perfusate (15,16). In humans, the GST family consists of three subpopulations according to their localization: Cytosolic, mitochondrial and microsomal (35). GSTs can be divided into subclasses, designated by Greek letters that differ in their levels of expression in humans. α-GST is abundantly expressed in liver and renal proximal tubular cells, whereas distal tubular cells mainly express the p-GST isoform (36,37). Although GSTs mainly function as catabolic agents to catalyze the detoxification of xenobiotics within glutathione, they have also been reported to mediate cell signaling. p-GST isoform can inhibit the function of kinases involved in the MAPK pathway, which regulates cell proliferation and cell death (38).

GSTs are considered one of the most valuable and reliable non-invasive markers for pre-transplant DGF prediction, but their utility in forecasting long-term outcomes, PNF and discarding kidneys from transplantation remains controversial (15,16). Significant differences were observed in the accuracy of the predictive values of different isoforms for DGF. Hall et al (39) previously found that p-GST measured at baseline and at the end of hypothermic machine perfused (HMP) kidneys from deceased donors was associated positively with DGF. The relative risk ratio of DGF for each log-unit increase at baseline and post-perfusion p-GST concentration after adjustment (for black ethnicity, male sex, history of previous kidney transplant, diabetes as the cause of end-stage renal disease, need for blood transfusion before KTx, number of human leukocyte antigen mismatches, body mass index in kg/m2 and duration of dialysis before transplant) was found to be 1.14 (95% CI, 1.00-1.28) and 1.33 (95% CI, 1.02-1.72), respectively. α-GST did not associate with DGF. In addition, the concentrations of both iso-enzymes did not differ between the transplanted and discarded organs at start and just before end of perfusion, showing that GSTs levels could not be used as a tool for discarding kidneys from KTx.

Perfusate GST levels have been previously compared with HMP parameters to improve the predictive value of GST. However, the value of this prospect remains controversial, where no clear association has yet been established. In a previous study by Moers et al (33), a correlation between the studied biomarkers [such as LDH, aspartate aminotransferase, N-Acetyl-/β-glucosaminidase, GSTs, alanine aminopeptidase (Ala-AP) and heart fatty acid binding proteins (H-FABP)] and pump parameters (such as cold ischemia time and renal vascular resistance) was not confirmed. Hall et al (39) also previously found no correlation between pump parameters (such as renal resistance at 4 h of perfusion and perfusate flow at 4 h) and concentrations of GST isoforms. By contrast, Qiao et al (40) found that the model combining end-of-perfusion vascular resistance and GST [area under the curve (AUC)=0.888; 95% CI, 0.842-0.933] significantly improved DGF predictability compared to using terminal resistance (AUC=0.756; 95% CI, 0.693-0.818) or GST alone (AUC= 0.729, 95% CI, 0.591-0.806).

Perfusate GST activity has also been investigated using other potential biomarkers of DGF. Gok et al (41) reported that GST with Ala-AP activity or GST activity with FABP concentration corresponded positively to renal injury during ischemia. However, its role in predicting the occurrence of PNF and in kidney discards for transplantation remains unclear (33).

Extracellular histones

Ischemic injury during HMP leads to the release of histones from disrupted cells (42), including extracellular histones H2A, H2B, H3 and H4. Each nucleated cell contains nucleosomes built from these four elements in an equimolar manner (43). Following the induction of pathological conditions, such as ischemia, histones are released from dying cells into the extracellular space (44,45). Using an experimental porcine model of warm ischemia and machine perfusion of retrieved kidneys, extracellular histones were reported to be cytotoxic to endothelial and renal epithelial cells in a temperature-dependent manner (46). Furthermore, histone release is higher during sub normothermic machine perfusion (SNMP) at 28°C compared with hypothermic machine perfusion (HMP) at 4°C (46). Under normal conditions, histones are responsible for nucleosome assembly (47), chromatin stability (47) and epigenetic modifications in transcription, replication and DNA repair (48). However, in the extracellular space, they function as damage-associated molecular pattern proteins (DAMPs) (49).

A previous retrospective study of 309 kidneys from DCDs was conducted on extracellular histones in the renal perfusate. van Smaalen et al (42) measured histone H3 levels at 1, 2 and 4 h of HMP. In uncontrolled DCD kidneys (Maastricht categories 1 and 2), no difference in H3 levels could be found compared with the PNF, DGF and IF (immediate function) groups. In the control cohort (Maastricht categories 3 and 4), a higher H3 concentration in perfusate was associated with DGF [univariable odds ratio (OR)=3.071; 95% CI, 1.588-5.941; multivariable OR=2.433; 95% CI, 1.253-4.723]. When comparing two groups of patients with histone H3 concentrations above and below the median, Kaplan-Meier analysis revealed a significant difference in graft survival in favor of lower concentrations. In particular, the 1- and 5-year survival rates were significantly superior in the lower H3 group (at 1 year, 84 vs. 72%; at 5 years, 79 vs. 66%) (50).

FABPs

Ischemia leads to the overloading of proximal tubular cells with free fatty acids (FFAs) and tubulo-interstitial damage (51,52). During oxidative stress, FFAs bind to fatty acid binding proteins (FABPs), thereby preventing their intracellular accumulation and further oxidation (53). In addition, they can regulate the expression of genes involved in FFA metabolism and promote their β-oxidation in peroxisomes (53). To date, two isoforms have been described in human proximal tubular cells, namely liver (L)-FABPs and heart (H)-FABPs, which are also present in distal tubular cells (54). Under normal conditions, FABPs are involved in the cellular uptake and utilization of FFAs from the plasma by rapid incorporation into triacylglycerols and phospholipids, promotion of FFA metabolism in mitochondria or peroxisomes, regulation of intracellular cholesterol metabolism, regulation of gene expression involved in lipid metabolism and modulation of cell proliferation (54,55).

Perfusate FABPs have been reported to associate with DGF. Moers et al (33) found that H-FABP was an independent predictor of DGF but only had a moderate prognostic value. In another previous study, L-FABP levels were reported to be associated with estimated glomerular filtration rate (eGFR) 6 months after KTx (56). To the best of our knowledge, the association between FABP levels in the renal perfusate and PNF has not been studied. Sun et al (57) could not confirm the predictive value of FABPs for DGF and 3-year post-transplant function (expressed by eGFR value) in a retrospective study of cases of DCDs. Perfusate FABPs levels have been shown to associate with high vascular resistance and early graft dysfunction (33). Fig. 1 presents a list of molecules that can be released after renal tubular cells disruption upon necrosis and apoptosis resulting from ischemia.

Proteins secreted during ischemia in HMP
NGAL

NGAL is a protein that is originally synthesized in the bone marrow and is stored in neutrophil granules (58). NGAL has been previously detected in the lung, colon, trachea and renal epithelium (19). NGAL is capable of binding siderophores. Therefore, it has been postulated that NGAL can serve as a bacteriostatic agent by sequestering iron (59) and as a regulator of extracellular iron-induced injury (60). Production in non-hematopoietic locations has been associated with systemic inflammation, where one such stimuli is IL-1β (61). NGAL is typically released in response to ischemia (62) and nephrotoxic agent (such as cisplatin) contact (63). In addition, ATP depletion induces NGAL mRNA expression, leading to the production of NGAL (62). In this context, necrosis and cell apoptosis is a factor that can trigger NGAL mRNA expression in renal tubular cells (63).

In a previous systematic review, Guzzi et al (15) found three studies on NGAL (29,32,56), but only one (56) showing a positive association between NGAL concentration in the HMP kidney perfusate and DGF occurrence. The study by Moser et al (29) found that NGAL levels are not significantly different between the DGF and non-DGF groups. In another study by Hoogland et al (32), NGAL concentrations measured 4 h after initiation of perfusion were not reported to be significantly associated with the risk of DGF or PNF. Furthermore, the post-HMP NGAL concentration was found to be associated with 6-month eGFR but not with PNF (56). In another study on discarded human kidneys, NGAL levels were found to be markedly higher after 6 h of normothermic machine perfusion (NMP) administration in organs from donors with higher serum creatinine levels (sCr) at the time of retrieval (64).

However, care must be taken when examining the perfusate for the presence of NGAL and interpreting the available data. NGAL is secreted in different forms, including monomeric, dimeric and conjugated with MMP-9 (65). Available assays utilizing antibodies have not been suitably adapted to measure the monomeric and heterodimeric forms released by injured or stressed tubular cells (19). Another important issue is the timing of sample collection from the perfusion machine. NGAL synthesis and release are considered to be altered by hypothermia (15). To determine the most appropriate time interval, measuring NGAL in the renal perfusate not only after 4 h, but also after 14-16 h of HMP, are of importance to confirm the effect of hypothermia and time of perfusion on its concentrations.

MMPs

MMPs have been recognized for their roles in ischemia and reperfusion processes. They have been studied in the context of ischemia-reperfusion injury in the central nervous system (66), lungs (67), liver (68), heart, (69,70), skeletal muscle (71), retina (72) and kidney (73-77). MMPs, also known as matrixins, are zinc-dependent enzymes that are located mainly in the extracellular space (78). However, MMP-1, MMP-2 and MMP-11 have been found to function intracellularly. To date, 23 MMPs have been identified in humans (79). They are first synthetized as pre-proenzymes and the majority are secreted into the extracellular space. MMPs are typically produced after stimuli, such as by inflammatory cytokines (TNF-α and IL-1), free radicals, reactive oxygen species (ROS), oncogenic cellular transformation, physical stress and chemical agents (78). In addition, a number of soluble factors can induce the expression of MMPs and molecules on the cell surface, including leukocyte function-associated antigen-1, intercellular adhesion molecule-1-mediated cell adhesion very late antigen 4 and vascular cell adhesion molecule (VCAM)-1 (80). The interaction between gp39-CD40 in monocytes and α5β1 integrin-fibronectin can also lead to MMP expression (80). Secreted pre-proenzymes are activated in the extracellular space by tissue, plasma or opportunistic bacterial proteinases. In particular, the urokinase-type plasminogen activator/plasmin system has been reported to serve a role in MMP activation (81). Pro-MMP-2 has been reported to be activated on the cell surface (82). Pro-MMP-2 is localized in podosomes or the invadopodia (83,84). This activation process requires the activation of the first membrane-type MMP (MT1-MMP), and the tissue inhibitor of MMP-2 (TIMP-2)-bound MT1-MMP (78). The TIMP-2 in the latter complex binds through its C-terminal domain to the hemopexin domain of pro-MMP-2, which is assumed to localize the zymogen close to the active MT1-MMP (78). It is also activated by plasmin produced by urokinase-type plasmin activator (from an inactive form), which is attached to a high-affinity cell membrane binding site by a specific N-terminal sequence of its non-catalytic chain (82).

MMP-2 and MMP-9, also known as gelatinases, are the most frequently investigated MMPs in KTx and organ preservation. MMP-2 activity contributes to the proteolysis of collagen types I, IV, V, VII, X, XI and XIV, gelatin, elastin, fibronectin, laminin and aggrecan (69,85). By contrast, MMP-9 targets include collagen types IV, V, VII, X and XIV, gelatin, aggrecan, elastin, entactin and fibronectin (86). Data on the presence of MMPs in the renal perfusate and their predictive value remain scarce. Moser et al (29) previously investigated the activity of NGAL, LDH and MMPs in renal perfusates from DBDs and donors after controlled cardiac death (cDCDs). MMPs could be detected in all samples and a significant positive association between perfusate MMP levels and the occurrence of DGF was reported. However, this association was not observed for NGAL and LDH levels. Additionally, higher concentrations of MMP-2 and MMP-9 were observed in the cDCDs group compared with those in the DBDs group. In another previous proteomic analysis on three types of donors, namely living kidney donors (LKDs), DBDs and cDCDs, the levels of MMPs were found to be ranked in the following manner: LKDs < DBDs < cDCDs (34).

Previous studies using animal models have shown that different storage methods may influence MMP secretion. In one such study, it was observed that HMP reduced MMP-9/NF-κB-dependent expression compared with static cold storage (SCS) in a rabbit model (87). Notably, HMP did not affect MMP-2 expression. In another study performed by Sulikowski et al (88) on a rat model, the authors investigated the impact of University of Wisconsin solution (UW) and EuroCollins solution (EC) on the gene expression of MMP-2 and tissue inhibitor of MMP-2 (TIMP-2). After 24 h of cold ischemia time (CIT), the gene expression levels of MMP-2 and TIMP-2 were found to be decreased in kidneys perfused with UW, whereas they were increased in kidneys perfused with EC. After warm ischemia, the levels of MMP-2 and TIMP-2 gene expression were increased in kidneys perfused with UW, whereas they were significantly lower in kidneys perfused with EC.

Kidney injury molecule-1 (KIM-1)

KIM-1, also known as hepatitis A virus cellular receptor 1 and T cell immunoglobulin- and mucin-domain-containing molecule, is considered to be a biomarker of ischemia-mediated renal injury during preservation (56). Its attractiveness stems from its specific expression pattern and function in renal proximal tubular cells. KIM-1 is a cell membrane glycoprotein (89), which is comprised of an extracellular portion, a six-cysteine immunoglobulin-like domain, two N-glycosylation sites and a thrombospondin-rich domain, which is characteristic of mucin-like O-glycosylated proteins (90). KIM-1 is predominantly localized on the apical surface of renal proximal tubular epithelial cells (91). It exhibits homology to a hepatitis A virus receptor (89). KIM-1 expression is typically absent under healthy conditions, but is activated under various conditions and diseases, such as cell dedifferentiation, ischemic injury (89), toxic injury (92), autosomal dominant polycystic kidney disease (93) and renal cell carcinoma (94). In the renal proximal epithelium, KIM-1 serves as a phosphatidylserine receptor that can recognize apoptotic cells and direct them into lysosomes (95). It can also bind to oxidized lipoproteins in injured cells. In addition, KIM-1 has been observed to mediate the transformation of epithelial cells into phagocytic cells (96).

A previous study on 671 kidneys retrieved from standard criteria donors (SCDs) and ECDs failed to demonstrate the utility of KIM-1 as a biomarker, predictor of DGF or long-term outcomes (56). KIM-1 concentrations were measured at the beginning and end of the perfusion. The mean difference between the KIM-1 concentrations at these points was not found to be associated with DGF. In an experimental model of NMP of discarded human kidneys, KIM-1 was found to be released at lower levels in kidneys with a higher urine output (64). In a previous study, in the DCDs kidney group, perfusate KIM-1 was proposed as an independent marker of DGF and was correlated with 3-month eGFR (57).

IL-18

IL-18 is a cytokine that belongs to the IL-1 family and has been detected during hypothermic machine perfusion (25,56). IL-18 precursors are present in monocytes, macrophages, dendritic cells, intestinal cells, keratocytes, epithelial cells and renal tubular cells (97), in addition to being readily released by dying cells (97). Transcription of the IL-18 precursor is stimulated by the binding of Toll-like receptors to PAMPs, which then signals through the NF-κB pathway (97). Activation of the IL-18 precursor leads to its intracellular conversion into its mature form by caspase-1 (98). Extracellular activation mechanisms involve various proteases, such as neutrophil proteinase 3, granzyme B and chymase (99,100). In renal epithelial cells, the IL-18 precursor can be activated and converted into its mature form by the MMP meprin B (101).

IL-18 was originally identified to be an IFN-γ-inducing factor. It serves an important role in both naive and adaptive immunity (102). Among IL-12 and IL-15, which can upregulate IL-18 receptor expression, IL-18 induces IFN-γ production in CD4, CD8 T cells and natural killer (NK) cells (103). In addition, IL-18 can directly upregulate perforin- and FasL-dependent cytotoxicity in NK and CD8 T cells (103). IL-1β/IL-18 signaling initiates the transcription of several inflammatory factors, including GM-CSF, IL-4, histamine and TNFα, which facilitate leukocyte infiltration (104). IL-18, in the absence of IL-12 and IL-15, is responsible for the differentiation of naive T cells into T helper 2 cells, which produce IL-4 and IL-13, (105,106). Upon IL-18 stimulation, mast cells and basophils can produce IL-4 and IL-13 (107), whilst γδ T-cells produce IL-17 (97).

Previous studies on kidneys during storage using perfusate IL-18 could not confirm its predictive value for DGF (25,56). Its concentration was higher in samples from kidneys developing DGF, but no statistical significance could be found. In addition, although IL-18 associated with PNF in kidneys from DCDs, the diagnostic accuracy of IL-18 alone was poor (56). Similarly, in kidneys with a KDPI >80, IL-18 provided no predictive value for PNF, DGF or 1-year post-transplant outcomes according to another previous study (108). Table I shows a list of protein biomarkers detected using untargeted analysis methods. Fig. 2 shows molecules secreted during ischemia in HMP.

Table I

Protein biomarkers of kidney quality detected in human perfusate.

Table I

Protein biomarkers of kidney quality detected in human perfusate.

Protein classNameType of donor(Refs.)
Cytoplasmic enzymesLactate dehydrogenaseDBDs, DCDs and LKDs(15,16,26,29,34)
Glutathione-S-transferases and its isoformsDBDs and DCDs(15,16)
Glutathione peroxidaseDBDs(145)
Peroxidoxin-2DBDs, DCDs and LKDs(34)
Alanine aminopeptidase (cytosolic form)DCDs(41)
Protein deglycase DJ-1DCDs, DBDs and LKDs(34)
Alcohol dehydrogenase, subunit βDBDs and DCDs(17)
Aldose reductaseDBDs and DCDs(17)
α-enolaseDBDs and DCDs(17)
AminoacylaseDBDs and DCDs(17)
Superoxide dismutaseDBDs(144)
Mitochondrial enzymesAspartate aminotransferaseDBDs and DCDs(33)
GAPDHDBDs and DCDs(17)
Superoxide dismutaseDBDs(144)
Lyzosomal enzymes N-Acetyl-/β-glucosaminidaseDBDs and DCDs(33)
Proteins secreted extracellularlyMMP-2DCDs, DBDs and LKDs(34)
Neutrophil gelatinase-associated lipocalinDBDs, DCDs and LKDs(34)
α1-antitrypsinDBDs, DCDs and LKDs(34)
Superoxide dismutaseDBDs(144)
Proteins attached to cellular membraneKidney injury molecule 1DBDs and DCDs(56)
Alanine aminopeptidase (membrane form)DCDs(41)
MMP-2 (membrane attached form)DBDs, DCDs and LKDs(29,34)
Nucleus componentsHistones H2A, H2B, H3 and H4DCDs(42)
Complement elementsC3a, c3d, and soluble c5b-9DCDs(128)
C1q subcomponent subunit B, C1s subcomponent, C1r subcomponent and C4b-binding protein α chainDBDs(116)
Cytoskeletal elementsActinin-1DBDs(116)
Talin-1DBDs(116)
DesmoplakinDBDs(116)
Collagen α-1(I) chainDCDs, DBDs and LKDs(34)
Proteasome elementsLiver-FABPDCDs, DBDs and LKDs(34)
Heart-FABPDBDs and DCDs(56)
Leucyl aminopeptidaseDCDs(143)
Pyroglutamyl aminopeptidaseDCDs(143)
Arginyl aminopeptidaseDCDs(143)
Dipeptidyl IV-aminopeptidaseDCDs(143)
Peroxisomal elementsCatalaseDBDs(144)
Immunological componentsImmunoglobulin heavy variable 2-26DBDs(116)
Immunoglobulin κ-chainDBDs and DCDs(17)
Immunoglobulin γ1-chainDBDs and DCDs(17)
Immunoglobulin γ2-chainDBDs and DCDs(17)
Immunoglobulin γ3-chainDBDs and DCDs(17)
Immunoglobulin γ3-chainDBDs and DCDs(17)
Vascular cell adhesion moleculeDBDs(141)
Intercellular adhesion moleculeDBDs(141)
TNF-αDBDs(141)
IFN-γDBDs(141)
IL-1β, -2, -6, -18DBDs and DCDs(56,141)
IL-4, -8, -10Not mentioned(145,146)
IL-1RαNot mentioned(145)
IL-6RDBDs(141)
Macrophage inflammatory protein-1αNot mentioned(145)
Monocyte chemoattractant protein-1Not mentioned(145)
Chemokine (C-X-C motif) ligand 1DBDs(141)
Cellular iron homeostasisTransferrinDBDs and DCDs(17)
HemopexinDBDs and DCDs(17)
HaptoglobinDBDs and DCDs(17)
OxyhemoglobinDCDs(117)
Blood coagulationFibrinogen γ-chainDBDs and DCDs(17)
Fibrinogen α-chainDCDs(117)
Annexin A5DBDs and DCDs(17)
Plasminogen activator inhibitor-1DBDs and DCDs(17)
Transport proteinsTransthyretinDBDs and DCDs(17)
AlbuminDBDs and DCDs(17)
Vitamin D binding proteinDBDs and DCDs(17)
High density lipoproteinDCDs(117)

[i] DBDs, donations after brain death; DCDs, donation after cardiac death; LKDs, living kidney donors; FABP, fatty acid binding protein.

Novel directions and highlights in proteomic studies

Whole-proteome analysis

Studies conducted over the past decade have provided novel insights and techniques into the proteomic analysis of the renal perfusate, where untargeted protein analysis techniques, including two-dimensional-polyacrylamide gel electrophoresis (2D-PAGE) and liquid chromatography-mass spectrometry (LC-MS), have been devised.

2D-PAGE (109) is a potent method for detecting nearly all protein elements in tested samples. According to this technique, proteins are separated according to their isoelectric points by isoelectric focusing, followed by SDS-PAGE, which segregates proteins based on their molecular weight and electrical charge (110). Staining with Coomassie Blue or silver helps to identify the target protein fractions, which can then be excised for further analysis (111).

LC-MS is currently the most accurate method for proteomic investigation (112). This tool involves digesting the protein sample with proteolytic enzymes, which is most commonly trypsin, to generate unique protein mass value. These peptides are then separated by high-performance liquid chromatography. After separation, MS is used to analyze each peak, converting the samples to the gas phase and transferring them to the ionization chamber. Electrospray ionization then ionizes the samples into cations, which are subsequently separated based on their mass-to-charge ratio using a mass analyzer. The detector identifies and quantifies each ion and the results are compared with database information (113). Proteomics most frequently uses the following two modifications of the MS element known as the ionizer: i) Matrix-assisted laser desorption/ionization (MALDI), which is laser desorption with the participation of a matrix; and ii) electrospray ionization. Other modifications can also be made to mass analyzers. They include quadrupole mass analyzers, Fourier transform ion cyclotron mass analyzer, Orbitrap mass analyzers, ion trap mass analyzer and TOF mass analyzer (114). At present, the most commonly applied method in proteome research is the time-of-flight (TOF) analyzer combined with a MALDI ionizer (113). TOF allows for the study of the flight time of ions between the ionizer and the detector, which depends on the ratio of the mass to the number of ion charges (112). The greatest advantage of MALDI-TOF is the direct detection of the composition of a complex mixture of proteins and other molecules (113,115). The underlying principles of both 2D-PAGE and LC-MS are shown in Fig. 3.

Complement elements

Previous studies on LC-MS analysis have highlighted the role of the complement system in renal injury during HMP and oxygenated-HMP (116,117). The complement system forms a part of the innate immune response that contributes to extensive endothelial cell damage (118). This system contains soluble proteins (such as C3a, C3b and C5a) and membrane-bound proteins (C5b, C6, C7, C8 and C9). Under physiological conditions, regulatory proteins control the degradation of C3 and C5 convertases to prevent overreactions (118-120). Different elements are involved in three pathways: Classical, alternative and lectin pathways. Both ischemia and reperfusion are important factors that can trigger the activation of the complement system (121). In addition, preformed donor-specific antibodies can activate the complement system by binding to C1q or C3d (122,123). Activation by DAMPs released during ischemia and reperfusion leads to the formation of a membrane-associated complex (MAC). This complex is comprised of C5b-9 subunits and has been implicated in apoptosis induction (124,125), direct cell injury, necrosis and neutrophil influx (126). MAC can also induce a pro-inflammatory response through the NF-κB signaling pathway in endothelial cells (127). Fig. 4 presents the different pathways of the complement system.

A previous study was performed on 22 kidneys retrieved from DBDs stored in HMP (116). Perfusate samples that were obtained after 15 min of HMP in patients with a good outcome (GO) at 1-year post-transplantation (eGFR ≥45 ml/min/1.73 m2 and sCr level ≤120 μmol/l) were found to contain more complement C1q subcomponent subunit B, complement C1s subcomponent (C1S), complement C1r subcomponent (C1R) and C4b binding protein α chain compared with those in perfusate samples of patients with a suboptimal outcome (SO) at 1-year post-transplantation (eGFR ≤45 ml/min/1.73 m2 and sCr level ≥120 μmol/l). Similar observations were made for samples collected after 4 h of HMP. In the GO group, complement C1r subcomponent-like protein, C1S and C1R were also found to be significantly upregulated compared with those in the SO group. The authors showed that the statistically significant difference in the concentration of complement elements can distinguish between GO and SO at 1-year post-transplantation (116).

Another study by Mulvey et al (117) focused on the proteomic investigation of the perfusate from 67 paired DCDs kidneys stored in oxygenated and non-oxygenated HMP, yielding 1,716 proteins including complement cascade, platelet aggregation and detoxification of reactive oxygen species. The most abundant proteins identified were complement C3, apolipoprotein A1 and fibrinogen α chain. One of the most abundant proteins was found to be C3. In addition, complement cascade proteins (such as C1R, C1QC, C3, C5, C7, C8A and C8G) were correlated with 12-month eGFR, where their abundance decreased during HMP (117). Oxygenation did not affect the proteome profile, including that of the complement cascade (117). Using porcine kidneys and discarded human kidneys (128), perfusion was previously conducted with a blood-based solution for 4 and 6 h. C3a, C3d and soluble C5b-9 (sC5b-9) were then measured in perfusate samples. In porcine kidneys, the presence of sC5b-9 was reported to contribute to lower creatinine clearance. It was then observed that the levels of complement products in the perfusate were higher in the kidneys from DBDs compared with those from DCDs.

Cytoskeletal structural elements

To identify novel biomarkers, cytoskeletal structural elements have been previously studied. These biomarkers tend to be associated with non-specific cellular damage (34,116,129), unlike the aforementioned markers associated with specific pathways or systems. During ischemia, metabolism switches from aerobic to anaerobic conditions (130). This results in increased lactate production and intracellular acidosis. These conditions inhibit Na+/K+-dependent ATPase activity, leading to the accumulation of sodium ions and cellular edema (131). Other consequences of ischemia include leakage of lysosomal enzymes, cytoskeletal breakdown and decreased calcium excretion (130). In addition, the excess calcium accumulation that ensues leads to the generation of ROS (132), but the quantity of ROS generated is not as potent as that during reperfusion. This is due to the reduction in the activity of cytochromes, nitric oxide synthases, xanthine oxidase and reduced nicotinamide adenine dinucleotide phosphate oxidase activity (130). Ischemia has been shown to trigger the expression and redistribution of microtubule cytoskeletal elements (such as actin, fodrin, tubulin and uvomorulin) (133,134). Cell integrity is then altered, leading to loss of cell polarity and redistribution of the basolateral membrane (135). These conditions can lead to various forms of cell death, including necrosis, apoptosis, autophagy and programmed necrosis (136).

A study by Coskun et al (129) revealed a group of structural proteins associated with cellular damage in the kidney. In particular, the preservation solution from 25 kidneys retrieved from DBDs stored in SCS was analyzed. Among the 206 proteins identified, a subset that associated significantly with donor characteristics was identified. Specifically, perlecan levels were correlated positively with sCr and blood urea nitrogen (BUN) levels, increased level of talin-1 was related positively to CIT, keratin type II cytoskeletal 8 was associated positively with recipient sCr, whilst profilin was associated positively with donor age (DA) and recipient BUN levels, myosin 6, 9 and 11 were correlated with DA, collagen VI from epithelial cells was associated positively with DA and recipient BUN levels, whereas microfibril-associated glycoprotein 4 was associated positively with DA and recipient sCr (113). Another study also to previously investigated HMP perfusates for the presences of structural components. van Leeuwen et al (116) studied 22 DBD kidneys and found a positive correlation between actinin-1 and talin-1 concentration in the perfusate and the 1-year post-transplant function expressed by eGFR. Upregulated cytoskeletal proteins actinin-1 and talin-1 in the HMP perfusate after 4 h of HMP discriminated kidneys with GO and SO after 1-year post-transplantation. It was therefore hypothesized that lower eGFR in the SO group was a result of the loss of cellular integrity and disruption in the podocyte architecture (116). In addition, this previous study (116) reported that the presence of desmoplakin quantified within LC-MS in the HMP perfusate after 4 h of perfusion was associated with SO. Desmoplakin is one of the most abundant components in desmosomes and cellular junctions (137). Together with immunoglobulin heavy variable 2-26, desmoplakin had a predictive value of 86% for SO occurrence in the first year after transplantation (116). The designs and results of whole-proteome studies are listed in Tables II and III.

Table II

Whole proteome analysis study designs.

Table II

Whole proteome analysis study designs.

Author, yearAnalytical methodType of donorKidney amountOutcomeTime of sample acquistion(Refs.)
Snoeijs et al, 2013i) 2D-DIGE; ii) LC-MS (spots identification)DCD and DBD18 (6 uncontrolled DCD; 6 controlled DCD; 6 DBD)Immediate function/DGF/PNF1 h of HMP(17)
Moser et al, 2017i) 2D-DIGE; ii) MS (spots identification); iii) Western blotting (results confirmation)DCD, DBD, LKD41 (9 DCD, 16 DBD, 16 LKD)DGF/SGF (SGF was defined as serum creatinine >265 μmol/l on post-transplant day 5 but no need for dialysis by day 5)End of HMP(34)
van Leeuwen et al, 2021LC-MSDBD22Suboptimal outcome at 1-year post-transplantation (kidney function with an eGFR ≥45 ml/min/1.73 m2 and a serum creatinine level of ≤120 μmol/l); good outcome at1-year post-transplantation (kidneys with an eGFR ≤45 ml/min/1.73 m2 and a serum creatinine level of ≥120 μmol/l 1-year post-transplantation)15 min of HMP (T1) and just before ending HMP (T2)(116)
Mulvey et al, 2023LC-MSDCD137eGFR measured one year after kidney transplant15 min of HMP; just before leaving donor's hospital; end of HMP(117)

[i] DBDs, donations after brain death; DCDs, donation after cardiac death; LKDs, living kidney donors; LC, liquid chromatography; MS, mass spectrometry; DIGE dimensional gel electrophoresis; eGFR, estimated glomerular filtration rate; DGF, delayed graft function; SGF, slow graft function; HMP, hypothermic machine perfusion.

Table III

Whole proteome analysis study results.

Table III

Whole proteome analysis study results.

Authors, yearNumber of detected proteinsNumber of selected proteinsClinical outcomeAnalytical methodSignificance(Refs.)
Snoeijs et al, 20136419Uncontrolled DCD, 4 DGF and 2 PNF; Controlled DCD, 3 IF and3 DGF; DBD, 3 IF and 3 DGFi) 2D-DIGE; ii) LC-MS (spots identification)i) α1-antitrypsin was upregulated in kidneys (considering all donor types) with DGF; ii) haptoglobin was downregulated in the perfusate of DCD kidneys (compared with DBD kidneys)(17)
Moser et al, 2017Not mentioned7Not mentionedi) 2D-DIGE; ii) MS (spots identification) Western Blot (results confirmation)i) total protein concentration-DCD > DBD > LKD; ii) 7 protein significantly different among the 3 groups, namely serum albumin, α-1-antitrypsin peroxiredoxin-2, heavy chain of immuno-globulin, fragment of collagen 1 and fatty acid binding protein, protein deglycase(34)
van Leeuwen et al, 20211,255T1, 28; T2, 48GO, 11 [12-month eGFR 67.4 (15.5)]; SO, 11 [12-month eGFR 27.6 (9.7)]LC-MST1: i) 18 proteins significantly upregulated in GO vs. SO.; ii) all significant proteins are located in extracellular regions, 5 of them belong to complement and coagulation cascades, including complement C1q subcomponent subunit B, complement C1s subcomponent, complement C1r subcomponent and C4b-binding protein α chain; iii) at T1, ATP-citrate synthase together with fatty acid binding protein 5 provided a predictive value of 91%, corresponding to a ROC with AUC of 0.97; and iv) 10 proteins significantly downregulated in GO vs. SO, 2 of them, fatty acid binding protein 4 and fatty acid binding protein 5, are involved in Peroxisome proliferator-activated receptor signalling. T2: i) 22 proteins significantly upregulated in GO vs. SO, 5 of them belong to complement and coagulation cascades, of which 4 are involved in complement activation of the classical pathway, namely complement C1r subcomponent-like protein, C1S and C1; ii) 26 proteins significantly downregulated in GO vs. SO, 14 of them are cytoskeletal proteins, including desmoplakin, talin-1 and α-actinin-1, 13 of the downregulated proteins are affiliated with the immune system; and iii) immunoglobulin heavy variable 2-26 togetherx heavy desmoplakine provided a predictive value of 86%, corresponding to ROC with AUC of 0.95 for 26 proteins.(116)
Mulvey et al, 20231,716137Mean eGFR in HMP + O2 group, 41 ml/min/1, 73 m2; In HMP without O2 group, 40 ml/min/1, 73 m2LC-MSi) Overall HMP duration associated with increasing levels of a subgroup of proteins; ii) HDL and complement cascade proteins are associated with 12-month eGFR; iii) blood-derived proteins are enriched in the perfusate of kidneys that developed acute rejection, these proteins belonged to 'regulation of response to wound healing' and 'metabolic processes contributed to ATP generation and nucleotide phosphorylation'; iv) the most abundant proteins in all samples were complement C3, apolipoprotein A1 and fibrinogen α-chain(117)

[i] DBDs, donations after brain death; DCDs, donation after cardiac death; LKDs, living kidney donors; DGF, delayed graft function; IF, immediate function; LC, liquid chromatography; MS, mass spectrometry; DIGE dimensional gel electrophoresis; eGFR, estimated glomerular filtration rate; GO, good outcome; SO, suboptimal outcome; ROC, receiver operating characteristic; AUC, area under the curve; HMP, hypothermic machine perfusion.

Other directions in proteomic investigations

Both 2D-PAGE and LC-MS have certain limitations. 2D-PAGE lacks specificity and frequently requires additional methods, such as western blotting, to confirm protein identity (111). LC-MS in contrast commands substantial financial investment, specialized equipment and trained personnel. Sample preparation for quantification is also time-consuming (112).

Modern methods, such as microarrays or protein chips, have emerged for the analysis of large protein panels (138). These microarrays contain predefined panels of proteins or antibodies tailored to the objectives of the experiment, such as analysis of specific protein fractions, cancer markers or virus antigens. Upon addition of the sample to the microarray plate, proteins will bind to the antibodies if present. Antibodies conjugated to fluorochromes are then introduced and bind to the previously formed complexes. This process distinguishes between capture, reverse and functional arrays, each designed for distinct analytical purposes. Fluorescent detectors then provide quantitative and qualitative results, enabling the simultaneous measurement of multiple compounds from a single sample (139). When the focus is on a limited number of proteins or a single protein, light scattering-based protein analysis techniques come into play. These include batch dynamic light scattering, static light scattering, charge and ζ potential measurements, in addition to circular dichroism spectrometry and isothermal titration calorimetry. These methods provide valuable information regarding protein size, abundance, charge, chirality, interactions and binding to other molecules (138,140). Perfusate analysis using a method that is a bridge between classical ELISA and microarrays, such as a multiplex immunoassay on a single platform, was performed by Baboudjian et al (141) in a prospective study of 74 kidneys from deceased donors (both ECDs and SCDs). VCAM obtained at the end of HMP was found to be a non-invasive predictor of early graft dysfunction at both 1 week (OR=3.57; 95% CI, 1.06-12.03) and 3 months (OR=4.039; 95% CI, 1.11-14.73) after transplantation (141). Table IV lists all the techniques utilized for proteomic analysis of renal perfusate. Fig. 5 shows a representative multiplex immunoassay on one platform and different types of ELISA.

Table IV

Analytical methods for protein detection in perfusate.

Table IV

Analytical methods for protein detection in perfusate.

MethodMechanism of actionAdvantagesDisadvantages
LC and high-performance LCSeparation and identification of proteins by mass transfer of a sample through a polar mobile phase and a non-polar stationary phase.i) High resolution and speed of analysis; ii) precise flow rate control; iii) senstive detection; and iv) low sample requirementi) Expensive solvents and columns; ii) regular maintenance and calibration is needed; and iii) sophisticated software is required
LC-mass spectrometryMixtures are separated according to their physical and chemical properties, then the components within each peak are identified and detected based on their mass spectrumi) The capability to quantify multiple analytes simultaneously; and ii) the method does not necessarily require immunological reagentsi) Regular maintenance and calibration is needed; ii) sophisticated software is required; iii) the instrument requires skilled personnel; and iv) expensive
2D-PAGESeparation of proteins in SDS gel by PAGE first by their charge then by their size, identifying each protein after staining for example within Coomassie Bluei) Enables the detection of hundreds of proteins on a single gel plate; and ii) enhanced resolution, detection, quantitation and reproducibilityi) Inability to resolve proteins that are too basic or too acidic, too large or too small; and ii) restricted to soluble and/or high abundant proteins
Enzymatic activity assayCarry out a reaction catalyzed by the protein under investigation and measure the concentration of the enzyme product using colorimetric, chemiluminescent or fluorescent methodi) High sensitivity and specificity; ii) rapid response; iii) easy to performi) Difficulties in assay standardization; ii) instability of the enzymesduring storage
ELISAProtein identification by binding a specific antibody to an antigen present on the desired molecule anddetecting a signal from staining agent conjugated with an antibody. It could be performed in 4 manners:Direct, indirect, Sandwich, competitivei high sensitivity and specificity; ii) high throughput; iii) easy to perform; and iv) it can determine the concentration of antigen in a samplei) Information limited to the amount or presence of the antigen in the sample; ii) detection is based on enzyme/substrate reactions
Western blottingSeparation of proteins by gel electrophoresis to resolve them by size and transfer to a membrane to immobilize the proteins. The proteins immobilized on the membrane are then identified in an immunoassay using colorimetric, chemiluminescent or fluorescent detectioni) The ability to identify specific target proteins; and ii) high sensitivityi) Time-consuming procedure; ii) problems in reproducibility; and iii) false-positive results when an antibody reacts with a non-intended protein
Multiplex immunoassay on one platformSimultaneous binding of proteins of interest to specific antibody conjugated beads, complex protein bead-antibody is conjugated to the fluorescent antibody, the specific signals of protein of interest are emitted by the fluorescent antibody and analyzed using a laser detectori) Good reproducibility; ii) rapid processing speed; iii) low sample volume; and iv) high sensitivityi) Cross-reactivity; ii) assay development time; iii) commitment to vendor-supplied assays; and iv) dynamic range

[i] LC, liquid chromatography.

Other potential protein biomarkers

In whole-proteome studies, other non-specific proteins associated with kidney injury have been detected in HMP perfusates. Proteins found at statistically significant levels include immunoglobulins κ, γ1, γ2 and γ3 chains (17). α1-Anti-trypsin with peroxidoxin-2 may discriminate between renal damage in DBDs, DCDs and LKDs (34).

Another group of proteins considered to be biomarkers and detected by whole-proteome analysis methods include proteins involved in transport (transthyretin, albumin and vitamin D-binding protein), blood coagulation (fibrinogen α-chain, fibrinogen γ-chain, annexin A5 and plasminogen activator inhibitor-1), energy metabolism (alcohol dehydrogenase subunit β, GAPDH, α-enolase, aldose reductase and aminoacylase), cellular iron homeostasis (transferrin, hemopexin, haptoglobin and oxyhemoglobin) and high-density lipoproteins (17,31). However, further studies are required to confirm their role in predicting short- and long-term outcomes in recipients.

A previous targeted proteomic analysis revealed the presence of Ala-AP (41,142), leucyl- and pyroglutamyl aminopeptidase (143), alanyl-arginyl- and dipeptidyl IV-aminopeptidase (143), aspartate aminotransferase (33) and N-acetyl-β-d-gluconamidase (33), glutathione peroxidase, catalase and superoxide dismutase (144), TNF-α, IFN-γ (141), IL-1β, -2, -4, -6, -8 and -10 (141,145,146), IL-1Rα, macrophage inflammatory protein-1, monocyte chemoattractant protein-1 (145), IL-6R and chemokine C-X-C motif ligand 1 (141). However, these proteins have been previously investigated, but none was found to be a reliable predictor of short- or long-term outcomes in recipients.

Conclusions and future perspectives

The number of patients with end-stage renal disease awaiting kidney transplantation is much higher compared with the number of transplanted organs (147). Therefore, an increasing number of kidneys have been retrieved from ECDs, which increases the risk of poor outcomes (148-150). However, organs in good condition, namely those not developing DGF or not requiring continuous dialysis therapy after KTx, are typically selected from this donor group.

Proteomic analysis of the perfusate remains to be a promising tool for the evaluation of kidneys before transplantation. This is especially true for the ECDs, DCDs and SCDs. Despite previous studies on this topic, protein markers that are sufficiently accurate for predicting the state of the kidney before KTx have remained elusive. However, the problem lies in the methodologies used in such studies. Searching for only one or a small group of proteins may not be the optimal approach. Changes in kidney metabolism during the entire transplantation procedure are extensive and must be monitored constantly. Therefore, biomarkers should include a large panel of proteins. Modern techniques of protein analysis will allow for such studies, whereas fast diagnostic methods (such as microarrays) will allow for the up-to-date supervision of kidney state during perfusion. To identify a reliable group of markers, profiling research must show the correlations of altered protein levels in the kidney and metabolic pathways activated during organ transplantation. Such pathways include ischemia-reperfusion, endothelial reticulum stress, oxidative stress and autophagy pathways. The optimal panel of markers should establish connections with all the proteins.

Another consideration should be the type of materials used for proteomic analysis. The present study has described proteins detected during HMP, which is a recognized method for kidney preservation, particularly for organs from ECDs (151,152). Continuous or pulsatile perfusion allows for the release and secretion of proteins into the perfusate. As a result, the dynamics of the processes during the HMP can be monitored. In addition, the quantity of proteins detected is likely to be more reliable compared with that detected during SCS. HMP ensures the removal of blood residues that may interfere with the final conclusions regarding the proteome profile.

Proteomic studies should be performed in different donor groups if the study aims to find an association between the detected protein and the selected endpoint. Various factors, such as CIT (153), diabetes and hypertension (154), cause of mortality (34), age (155,156) and terminal sCr level (157,158), can all affect the condition of the donated kidneys. Mixing organs from ECDs and SCDs will not lead to correct conclusions in the context of the selected endpoint due to the aforementioned factors. Another issue is defining the endpoint. In the majority of studies, DGF was defined as the need for dialysis within the first week after KTx (159-162). However, dialysis can be performed due to post-operative hyperkalemia or fluid overload that does not result from renal injury (163,164). Further studies should consider a more appropriate definition of DGF and other endpoints.

The quantification method is crucial for certain proteins. This applies to biomarkers that have isoforms, such as GSTs or form complexes, such as NGAL. Once a biomarker has been selected from the entire proteome, the next step should involve finding a suitable laboratory test or developing a novel method to detect the entire quantity of the desired molecule.

In summary, although numerous studies have been conducted with conflicting results, perfusate remains to be a promising source of protein biomarkers. Novel studies concerning the aforementioned issues may unravel other properties of well-known proteins, such as NGAL, KIM-1 or MMPs. Whole-proteome techniques will likely provide insights into non-specific kidney processes during preservation and complex protein profiles of numerous biological samples. Methods such as immunofixation, western blotting and ELISA enable the precise detection and quantification of specific proteins based on existing data. Immunofixation separates proteins electrophoretically and identifies them using specific antibodies, whilst western blotting employs primary and secondary antibodies for protein detection and ELISA quantifies proteins through spectrophotometric analysis.

Current advancements for studying proteins include microarrays and protein chips, which will allow for the simultaneous analysis of multiple targets. These techniques are particularly beneficial for studying specific protein fractions, cancer markers or viral antigens. Light scattering methods, such as dynamic light scattering and circular dichroism spectrometry, provide additional details on protein size, charge and interactions. Untargeted techniques, such as 2D-PAGE and LC-MS, enable comprehensive proteome profiling. 2D-PAGE separates proteins by isoelectric point and molecular mass, whereas LC-MS identifies proteins through peptide mass fingerprints and enzymatic digestion. Despite their complexity and cost, these methods will prove crucial for identifying novel proteins and understanding proteomic changes. Collectively, these techniques compose a robust framework for proteomic analysis, which is expected to advance the current understanding of protein function, interactions and roles in renal health and disease.

Recent studies on the role of the complement system and cytoskeletal elements show the association with short- and long-term outcomes (116,117,129). However, randomized controlled trials are required to establish their role in the assessment of kidney quality before transplantation. In addition, targeted methods for a protein of interest should be developed for further analysis. Despite novel storage protocols, such as NMP and SNMP, hypothermic methods remain in use in numerous countries and proteomic analysis of these perfusate is justified. SCS is the current storage standard for kidneys procured from DCDs in the UK (165). In the Netherlands, HMP is the standard method for kidney preservation in all donor types (166). To date, one randomized controlled trial has been performed in which SCS alone was compared with SCS plus a 1-h period of NMP at the end of storage (165). This previous study showed that SCS plus 1-h of NMP is not superior in reducing the DGF rate compared with kidney preservation by SCS alone, since this method did not improve the long-term results (135). However, additional studies comparing longer periods of NMP with HMP (165,167) are essential to confirm the safety and superiority of normothermic techniques over HMP or SCS. Nevertheless, proteomic analysis of the solutions used for normothermic perfusion should be performed as a source of novel biomarkers.

Availability of data and materials

Not applicable.

Authors' contributions

MB contributed to the design of the review. MB and MiS wrote the manuscript. MB and RD saw and verified all articles included in this review from the following four databases: Embase, PubMed (Medline), Web of Sscience and Scopus. MB designed the figures and tables. MiS, RD, MK and MaS revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

The authors would like to thank Ms Anna Palińska (Off Course School, Warsaw) for creating the figures with BioRender® Science Suite Inc.

Funding

No funding was received.

References

1 

Querard AH, Le Borgne F, Dion A, Giral M, Mourad G, Garrigue V, Rostaing L, Kamar N, Loupy A, Legendre C, et al: Propensity score-based comparison of the graft failure risk between kidney transplant recipients of standard and expanded criteria donor grafts: Toward increasing the pool of marginal donors. Am J Transplant. 18:1151–1157. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Querard AH, Foucher Y, Combescure C, Dantan E, Larmet D, Lorent M, Pouteau LM, Giral M and Gillaizeau F: Comparison of survival outcomes between expanded criteria donor and standard criteria donor kidney transplant recipients: A systematic review and meta-analysis. Transpl Int. 29:403–415. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Barba J, Zudaire JJ, Robles JE, Rosell D, Berian JM and Pascual I: Complications of kidney transplantation with grafts from expanded criteria donors. World J Urol. 31:893–900. 2013. View Article : Google Scholar

4 

Coupel S, Giral-Classe M, Karam G, Morcet JF, Dantal J, Cantarovich D, Blancho G, Bignon JD, Daguin P, Soulillou JP and Hourmant M: Ten-year survival of second kidney transplants: Impact of immunologic factors and renal function at 12 months. Kidney Int. 64:674–680. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Kousoulas L, Vondran FWR, Syryca P, Klempnauer J, Schrem H and Lehner F: Risk-adjusted analysis of relevant outcome drivers for patients after more than two kidney transplants. J Transplant. 2015:7120492015. View Article : Google Scholar : PubMed/NCBI

6 

Zádori G, Kovács DÁ, Fedor R, Kanyári Z, Zsom L, Asztalos L and Nemes B: Results of expanded-criteria donor kidneys: A single-center experience in hungary. Transplant Proc. 47:2189–2191. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Hwang JK, Park SC, Kwon KH, Choi BS, Kim JI, Yang CW, Kim YS and Moon IS: Long-term outcomes of kidney transplantation from expanded criteria deceased donors at a single center: Comparison with standard criteria deceased donors. Transplant Proc. 46:431–436. 2014. View Article : Google Scholar : PubMed/NCBI

8 

De Beule J and Jochmans I: Kidney perfusion as an organ quality assessment tool-are we counting our chickens before they have hatched? J Clin Med. 9:8792020. View Article : Google Scholar : PubMed/NCBI

9 

Mourão TB, Mine KL, Campos EF, Medina-Pestana JO, Tedesco-Silva H and Gerbase-DeLima M: Predicting delayed kidney graft function with gene expression in preimplantation biopsies and first-day posttransplant blood. Hum Immunol. 77:353–357. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Hall IE, Reese PP, Weng FL, Schröppel B, Doshi MD, Hasz RD, Reitsma W, Goldstein MJ, Hong K and Parikh CR: Preimplant histologic acute tubular necrosis and allograft outcomes. Clin J Am Soc Nephrol. 9:573–582. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Bachmann Q, Haberfellner F, Büttner-Herold M, Torrez C, Haller B, Assfalg V, Renders L, Amann K, Heemann U, Schmaderer C and Kemmner S: The kidney donor profile index (KDPI) correlates with histopathologic findings in post-reperfusion baseline biopsies and predicts kidney transplant outcome. Front Med (Lausanne). 9:8752062022. View Article : Google Scholar : PubMed/NCBI

12 

Rege A, Irish B, Castleberry A, Vikraman D, Sanoff S, Ravindra K, Collins B and Sudan D: Trends in usage and outcomes for expanded criteria donor kidney transplantation in the United States characterized by kidney donor profile index. Cureus. 8:e8872016.PubMed/NCBI

13 

Rao PS, Schaubel DE, Guidinger MK, Andreoni KA, Wolfe RA, Merion RM, Port FK and Sung RS: A comprehensive risk quantification score for deceased donor kidneys: The kidney donor risk index. Transplantation. 88:231–236. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Nyberg SL, Baskin-Bey ES, Kremers W, Prieto M, Henry ML and Stegall MD: Improving the prediction of donor kidney quality: Deceased donor score and resistive indices. Transplantation. 80:925–929. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Guzzi F, Knight SR, Ploeg RJ and Hunter JP: A systematic review to identify whether perfusate biomarkers produced during hypothermic machine perfusion can predict graft outcomes in kidney transplantation. Transpl Int. 33:590–602. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Bhangoo RS, Hall IE, Reese PP and Parikh CR: Deceased-donor kidney perfusate and urine biomarkers for kidney allograft outcomes: A systematic review. Nephrol Dial Transplant. 27:3305–3314. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Snoeijs MG, Pulinx B, van Dieijen-Visser MP, Buurman WA, van Heurn LW and Wodzig WK: Characterization of the perfusate proteome of human donor kidneys. Ann Clin Biochem. 50:140–146. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Khan AA, Allemailem KS, Alhumaydhi FA, Gowder SJT and Rahmani AH: The biochemical and clinical perspectives of lactate dehydrogenase: An enzyme of active metabolism. Endocr Metab Immune Disord Drug Targets. 20:855–868. 2020. View Article : Google Scholar

19 

Mårtensson J and Bellomo R: The rise and fall of NGAL in acute kidney injury. Blood Purif. 37:304–310. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Markert CL: Lactate dehydrogenase. Biochemistry and function of lactate dehydrogenase. Cell Biochem Funct. 2:131–134. 1984. View Article : Google Scholar : PubMed/NCBI

21 

Holmes RS and Goldberg E: Computational analyses of mammalian lactate dehydrogenases: Human, mouse, opossum and platypus LDHs. Comput Biol Chem. 33:379–385. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Dubach UC: On the origin of lactic dehydrogenase isoenzymes in urine. Helv Med Acta. 33:139–150. 1966.PubMed/NCBI

23 

Osis G, Traylor AM, Black LM, Spangler D, George JF, Zarjou A, Verlander JW and Agarwal A: Expression of lactate dehydrogenase A and B isoforms in the mouse kidney. Am J Physiol Renal Physiol. 320:F706–F718. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Kootstra G and Daemen JH: The non-heart-beating donor. Transplant Proc. 28:161996.PubMed/NCBI

25 

Daemen JW, Oomen AP, Janssen MA, van de Schoot L, van Kreel BK, Heineman E and Kootstra G: Glutathione S-transferase as predictor of functional outcome in transplantation of machine-preserved non-heart-beating donor kidneys. Transplantation. 63:89–93. 1997. View Article : Google Scholar : PubMed/NCBI

26 

Modgill VK, Wiggins PA, Rosenberg IL, Humphrey CS and Giles GR: An evaluation of viability tests of human cadaveric kidneys. Br J Surg. 64:548–553. 1977. View Article : Google Scholar : PubMed/NCBI

27 

Skillen AW: Clinical biochemistry of lactate dehydrogenase. Cell Biochem Funct. 2:140–144. 1984. View Article : Google Scholar : PubMed/NCBI

28 

Huijgen HJ, Sanders GT, Koster RW, Vreeken J and Bossuyt PM: The clinical value of lactate dehydrogenase in serum: A quantitative review. Eur J Clin Chem Clin Biochem. 35:569–579. 1997.PubMed/NCBI

29 

Moser MA, Arcand S, Lin HB, Wojnarowicz C, Sawicka J, Banerjee T, Luo Y, Beck GR, Luke PP and Sawicki G: Protection of the transplant kidney from preservation injury by inhibition of matrix metalloproteinases. PLoS One. 11:e01575082016. View Article : Google Scholar : PubMed/NCBI

30 

Nagelschmidt M, Minor T, Gallinat A, Moers C, Jochmans I, Pirenne J, Ploeg RJ, Paul A and Treckmann J: Lipid peroxidation products in machine perfusion of older donor kidneys. J Surg Res. 180:337–342. 2013. View Article : Google Scholar

31 

de Vries B, Snoeijs MGJ, von Bonsdorff L, Ernest van Heurn LW, Parkkinen J and Buurman WA: Redox-active iron released during machine perfusion predicts viability of ischemically injured deceased donor kidneys. Am J Transplant. 6:2686–2693. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Hoogland ER, de Vries EE, Christiaans MH, Winkens B, Snoeijs MG and van Heurn LW: The value of machine perfusion biomarker concentration in DCD kidney transplantations. Transplantation. 95:603–610. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Moers C, Varnav OC, van Heurn E, Jochmans I, Kirste GR, Rahmel A, Leuvenink HG, Squifflet JP, Paul A, Pirenne J, et al: The value of machine perfusion perfusate biomarkers for predicting kidney transplant outcome. Transplantation. 90:966–973. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Moser MAJ, Sawicka K, Arcand S, O'Brien P, Luke P, Beck G, Sawicka J, Cohen A and Sawicki G: Proteomic analysis of perfusate from machine cold perfusion of transplant kidneys: Insights into protection from injury. Ann Transplant. 22:730–739. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Udomsinprasert R, Pongjaroenkit S, Wongsantichon J, Oakley AJ, Prapanthadara LA, Wilce MC and Ketterman AJ: Identification, characterization and structure of a new Delta class glutathione transferase isoenzyme. Biochem J. 388:763–771. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Di Ilio C, Aceto A, Bucciarelli T, Angelucci S, Felaco M, Grilli A, Zezza A, Tenaglia R and Federici G: Glutathione transferase isoenzymes in normal and neoplastic human kidney tissue. Carcinogenesis. 12:1471–1475. 1991. View Article : Google Scholar : PubMed/NCBI

37 

Harrison DJ, Kharbanda R, Cunningham DS, McLellan LI and Hayes JD: Distribution of glutathione S-transferase isoenzymes in human kidney: Basis for possible markers of renal injury. J Clin Pathol. 42:624–628. 1989. View Article : Google Scholar : PubMed/NCBI

38 

Laborde E: Glutathione transferases as mediators of signaling pathways involved in cell proliferation and cell death. Cell Death Differ. 17:1373–1380. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Hall IE, Bhangoo RS, Reese PP, Doshi MD, Weng FL, Hong K, Lin H, Han G, Hasz RD, Goldstein MJ, et al: Glutathione S-transferase iso-enzymes in perfusate from pumped kidneys are associated with delayed graft function. Am J Transplant. 14:886–896. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Qiao Y, Ding C, Li Y, Tian X, Tian P, Ding X, Xiang H, Zheng J and Xue W: Predictive value of hypothermic machine perfusion parameters combined perfusate biomarkers in deceased donor kidney transplantation. Chin Med J (Engl). 135:181–186. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Gok MA, Pelsers M, Glatz JFC, Bhatti AA, Shenton BK, Peaston R, Cornell C, Mantle D and Talbot D: Comparison of perfusate activities of glutathione S-transferase, alanine aminopeptidase and fatty acid binding protein in the assessment of non-heart-beating donor kidneys. Ann Clin Biochem. 40:252–258. 2003. View Article : Google Scholar : PubMed/NCBI

42 

van Smaalen TC, Beurskens DMH, Hoogland ERP, Winkens B, Christiaans MHL, Reutelingsperger CP, van Heurn LWE and Nicolaes GAF: Presence of cytotoxic extracellular histones in machine perfusate of donation after circulatory death kidneys. Transplantation. 101:e93–e101. 2017. View Article : Google Scholar

43 

Felsenfeld G and Groudine M: Controlling the double helix. Nature. 421:448–453. 2003. View Article : Google Scholar : PubMed/NCBI

44 

Silk E, Zhao H, Weng H and Ma D: The role of extracellular histone in organ injury. Cell Death Dis. 8:e28122017. View Article : Google Scholar : PubMed/NCBI

45 

Wickman GR, Julian L, Mardilovich K, Schumacher S, Munro J, Rath N, Zander SA, Mleczak A, Sumpton D, Morrice N, et al: Blebs produced by actin-myosin contraction during apoptosis release damage-associated molecular pattern proteins before secondary necrosis occurs. Cell Death Differ. 20:1293–1305. 2013. View Article : Google Scholar : PubMed/NCBI

46 

van Smaalen TC, Beurskens DMH, Kox JJHFM, Polonia R, Vos R, Duimel H, van de Wetering WJ, López-Iglesias C, Reutelingsperger CP, Ernest van Heurn LW, et al: Extracellular histone release by renal cells after warm and cold ischemic kidney injury: Studies in an ex-vivo porcine kidney perfusion model. PLoS One. 18:e02799442023. View Article : Google Scholar : PubMed/NCBI

47 

Campos EI and Reinberg D: Histones: Annotating chromatin. Annu Rev Genet. 43:559–599. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Strahl BD and Allis CD: The language of covalent histone modifications. Nature. 403:41–45. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Kono H and Rock KL: How dying cells alert the immune system to danger. Nat Rev Immunol. 8:279–289. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Li B, Hao J, Zeng J and Sauter ER: SnapShot: FABP functions. Cell. 182:1066–1066.e1. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Zager RA, Johnson ACM and Hanson SY: Renal tubular triglyercide accumulation following endotoxic, toxic, and ischemic injury. Kidney Int. 67:111–121. 2005. View Article : Google Scholar

52 

Bobulescu IA: Renal lipid metabolism and lipotoxicity. Curr Opin Nephrol Hypertens. 19:393–402. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Storch J and Thumser AE: The fatty acid transport function of fatty acid-binding proteins. Biochim Biophys Acta. 1486:28–44. 2000. View Article : Google Scholar : PubMed/NCBI

54 

Pelsers MMAL: Fatty acid-binding protein as marker for renal injury. Scand J Clin Lab Invest Suppl. 241:73–77. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Yamamoto T, Noiri E, Ono Y, Doi K, Negishi K, Kamijo A, Kimura K, Fujita T, Kinukawa T, Taniguchi H, et al: Renal L-type fatty acid-binding protein in acute ischemic injury. J Am Soc Nephrol. 18:2894–2902. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Parikh CR, Hall IE, Bhangoo RS, Ficek J, Abt PL, Thiessen-Philbrook H, Lin H, Bimali M, Murray PT, Rao V, et al: Associations of perfusate biomarkers and pump parameters with delayed graft function and deceased donor kidney allograft function. Am J Transplant. 16:1526–1539. 2016. View Article : Google Scholar :

57 

Sun Z, Gao Z, Li X, Zheng X, Wang W and Qiao P: Perfusate neutrophil gelatinase-associated lipocalin, kidney injury molecular-1, liver-type fatty acid binding protein, and interleukin-18 as potential biomarkers to predict delayed graft function and long-term prognosis in kidney transplant recipients: A single-center retrospective study. Med Sci Monit. 29:e9387582023. View Article : Google Scholar

58 

Borregaard N and Cowland JB: Granules of the human neutrophilic polymorphonuclear leukocyte. Blood. 89:3503–3521. 1997. View Article : Google Scholar : PubMed/NCBI

59 

Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira S and Aderem A: Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature. 432:917–921. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Yang J, Goetz D, Li JY, Wang W, Mori K, Setlik D, Du T, Erdjument-Bromage H, Tempst P, Strong R and Barasch J: An iron delivery pathway mediated by a lipocalin. Mol Cell. 10:1045–1056. 2002. View Article : Google Scholar : PubMed/NCBI

61 

Cowland JB, Sørensen OE, Sehested M and Borregaard N: Neutrophil gelatinase-associated lipocalin is up-regulated in human epithelial cells by IL-1 beta, but not by TNF-alpha. J Immunol. 171:6630–6639. 2003. View Article : Google Scholar : PubMed/NCBI

62 

Mishra J, Ma Q, Prada A, Mitsnefes M, Zahedi K, Yang J, Barasch J and Devarajan P: Identification of neutrophil gelatinase-associated lipocalin as a novel early urinary biomarker for ischemic renal injury. J Am Soc Nephrol. 14:2534–2543. 2003. View Article : Google Scholar : PubMed/NCBI

63 

Mishra J, Mori K, Ma Q, Kelly C, Barasch J and Devarajan P: Neutrophil gelatinase-associated lipocalin: A novel early urinary biomarker for cisplatin nephrotoxicity. Am J Nephrol. 24:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Weissenbacher A, Stone JP, Lo Faro ML, Hunter JP, Ploeg RJ, Coussios CC, Fildes JE and Friend PJ: Hemodynamics and metabolic parameters in normothermic kidney preservation are linked with donor factors, perfusate cells, and cytokines. Front Med (Lausanne). 8:8010982022. View Article : Google Scholar : PubMed/NCBI

65 

Cai L, Rubin J, Han W, Venge P and Xu S: The origin of multiple molecular forms in urine of HNL/NGAL. Clin J Am Soc Nephrol. 5:2229–2235. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Rosell A and Lo EH: Multiphasic roles for matrix metalloproteinases after stroke. Curr Opin Pharmacol. 8:82–89. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Soccal PM, Gasche Y, Miniati DN, Hoyt G, Berry GJ, Doyle RL, Theodore J and Robbins RC: Matrix metalloproteinase inhibition decreases ischemia-reperfusion injury after lung transplantation. Am J Transplant. 4:41–50. 2004. View Article : Google Scholar

68 

Viappiani S, Sariahmetoglu M and Schulz R: The role of matrix metalloproteinase inhibitors in ischemia-reperfusion injury in the liver. Curr Pharm Des. 12:2923–2934. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Nagase H, Visse R and Murphy G: Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 69:562–573. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Chow AK, Cena J and Schulz R: Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br J Pharmacol. 152:189–205. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Roach DM, Fitridge RA, Laws PE, Millard SH, Varelias A and Cowled PA: Up-regulation of MMP-2 and MMP-9 leads to degradation of type IV collagen during skeletal muscle reperfusion injury; protection by the MMP inhibitor, doxycycline. Eur J Vasc Endovasc Surg. 23:260–269. 2002. View Article : Google Scholar : PubMed/NCBI

72 

Mathalone N, Lahat N, Rahat MA, Bahar-Shany K, Oron Y and Geyer O: The involvement of matrix metalloproteinases 2 and 9 in rat retinal ischemia. Graefes Arch Clin Exp Ophthalmol. 245:725–732. 2007. View Article : Google Scholar

73 

Cavdar Z, Ural C, Celik A, Arslan S, Terzioglu G, Ozbal S, Yildiz S, Ergur UB, Guneli E, Camsari T, et al: Protective effects of taurine against renal ischemia/reperfusion injury in rats by inhibition of gelatinases, MMP-2 and MMP-9, and p38 mitogen-activated protein kinase signaling. Biotech Histochem. 92:524–535. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Zhao H, Dong Y, Tian X, Tan TK, Liu Z, Zhao Y, Zhang Y, Harris DCh and Zheng G: Matrix metalloproteinases contribute to kidney fibrosis in chronic kidney diseases. World J Nephrol. 2:84–89. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Cavdar Z, Ozbal S, Celik A, Ergur BU, Guneli E, Ural C, Camsari T and Guner GA: The effects of alpha-lipoic acid on MMP-2 and MMP-9 activities in a rat renal ischemia and re-perfusion model. Biotech Histochem. 89:304–314. 2014. View Article : Google Scholar

76 

Kunugi S, Shimizu A, Kuwahara N, Du X, Takahashi M, Terasaki Y, Fujita E, Mii A, Nagasaka S, Akimoto T, et al: Inhibition of matrix metalloproteinases reduces ischemia-reperfusion acute kidney injury. Lab Invest. 91:170–180. 2011. View Article : Google Scholar

77 

Han WK, Waikar SS, Johnson A, Betensky RA, Dent CL, Devarajan P and Bonventre JV: Urinary biomarkers in the early diagnosis of acute kidney injury. Kidney Int. 73:863–869. 2008. View Article : Google Scholar

78 

Nagase H: Activation mechanisms of matrix metalloproteinases. Biol Chem. 378:151–160. 1997.PubMed/NCBI

79 

Visse R and Nagase H: Matrix metalloproteinases and tissue inhibitors of metalloproteinases: Structure, function, and biochemistry. Circ Res. 92:827–839. 2003. View Article : Google Scholar : PubMed/NCBI

80 

Nagase H and Woessner JF Jr: Matrix metalloproteinases. J Biol Chem. 274:21491–21494. 1999. View Article : Google Scholar : PubMed/NCBI

81 

Carmeliet P, Moons L, Lijnen R, Baes M, Lemaître V, Tipping P, Drew A, Eeckhout Y, Shapiro S, Lupu F and Collen D: Urokinase-generated plasmin activates matrix metalloproteinases during aneurysm formation. Nat Genet. 17:439–444. 1997. View Article : Google Scholar : PubMed/NCBI

82 

Mazzieri R, Masiero L, Zanetta L, Monea S, Onisto M, Garbisa S and Mignatti P: Control of type IV collagenase activity by components of the urokinase-plasmin system: A regulatory mechanism with cell-bound reactants. EMBO J. 16:2319–2332. 1997. View Article : Google Scholar : PubMed/NCBI

83 

Emonard HP, Remacle AG, Noël AC, Grimaud JA, Stetler-Stevenson WG and Foidart JM: Tumor cell surface-associated binding site for the M(r) 72,000 type IV collagenase. Cancer Res. 52:5845–5848. 1992.PubMed/NCBI

84 

Monsky WL, Kelly T, Lin CY, Yeh Y, Stetler-Stevenson WG, Mueller SC and Chen WT: Binding and localization of M(r) 72,000 matrix metalloproteinase at cell surface invadopodia. Cancer Res. 53:3159–3164. 1993.PubMed/NCBI

85 

Aimes RT and Quigley JP: Matrix metalloproteinase-2 is an interstitial collagenase. Inhibitor-free enzyme catalyzes the cleavage of collagen fibrils and soluble native type I collagen generating the specific 3/4- and 1/4-length fragments. J Biol Chem. 270:5872–5876. 1995. View Article : Google Scholar : PubMed/NCBI

86 

Yabluchanskiy A, Ma Y, Iyer RP, Hall ME and Lindsey ML: Matrix metalloproteinase-9: Many shades of function in cardiovascular disease. Physiology (Bethesda). 28:391–403. 2013.PubMed/NCBI

87 

Fu Z, Ye Q, Zhang Y, Zhong Z, Xiong Y, Wang Y, Hu L, Wang W, Huang W and Ko DS: Hypothermic machine perfusion reduced inflammatory reaction by downregulating the expression of matrix metalloproteinase 9 in a reperfusion model of donation after cardiac death. Artif Organs. 40:E102–E111. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Sulikowski T, Domanski L, Zietek Z, Adler G, Pawlik A, Ciechanowicz A, Ciechanowski K and Ostrowski M: Effect of preservation solutions UW and EC on the expression of matrix metalloproteinase II and tissue inhibitor of metalloproteinase II genes in rat kidney. Postepy Hig Med Dosw (Online). 66:45–50. 2012.PubMed/NCBI

89 

Ichimura T, Bonventre JV, Bailly V, Wei H, Hession CA, Cate RL and Sanicola M: Kidney injury molecule-1 (KIM-1), a putative epithelial cell adhesion molecule containing a novel immunoglobulin domain, is up-regulated in renal cells after injury. J Biol Chem. 273:4135–4142. 1998. View Article : Google Scholar : PubMed/NCBI

90 

Khan KNM, Hard GC and Alden CL: Chapter 47-Kidney. Haschek and Rousseaux's Handbook of Toxicologic Pathology. 3rd. Haschek WM, Rousseaux CG and Wallig MA: Academic Press; Boston: pp. 1667–1773. 2013, View Article : Google Scholar

91 

van Timmeren MM, van den Heuvel MC, Bailly V, Bakker SJL, van Goor H and Stegeman CA: Tubular kidney injury molecule-1 (KIM-1) in human renal disease. J Pathol. 212:209–217. 2007. View Article : Google Scholar : PubMed/NCBI

92 

Amin RP, Vickers AE, Sistare F, Thompson KL, Roman RJ, Lawton M, Kramer J, Hamadeh HK, Collins J, Grissom S, et al: Identification of putative gene based markers of renal toxicity. Environ Health Perspect. 112:465–479. 2004. View Article : Google Scholar : PubMed/NCBI

93 

Griffin BR, You Z, Noureddine L, Gitomer B, Perrenoud L, Wang W, Chonchol M and Jalal D; HALT Investigators: KIM-1 and kidney disease progression in autosomal dominant polycystic kidney disease: HALT-PKD results. Am J Nephrol. 51:473–479. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Han WK, Alinani A, Wu CL, Michaelson D, Loda M, McGovern FJ, Thadhani R and Bonventre JV: Human kidney injury molecule-1 is a tissue and urinary tumor marker of renal cell carcinoma. J Am Soc Nephrol. 16:1126–1134. 2005. View Article : Google Scholar : PubMed/NCBI

95 

Bonventre JV: Kidney injury molecule-1 (KIM-1): A urinary biomarker and much more. Nephrol Dial Transplant. 24:3265–3268. 2009. View Article : Google Scholar : PubMed/NCBI

96 

Ichimura T, Asseldonk EJPV, Humphreys BD, Gunaratnam L, Duffield JS and Bonventre JV: Kidney injury molecule-1 is a phosphatidylserine receptor that confers a phagocytic phenotype on epithelial cells. J Clin Invest. 118:1657–1668. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Kaplanski G: Interleukin-18: Biological properties and role in disease pathogenesis. Immunol Rev. 281:138–153. 2018. View Article : Google Scholar

98 

Ghayur T, Banerjee S, Hugunin M, Butler D, Herzog L, Carter A, Quintal L, Sekut L, Talanian R, Paskind M, et al: Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature. 386:619–623. 1997. View Article : Google Scholar : PubMed/NCBI

99 

Sugawara S, Uehara A, Nochi T, Yamaguchi T, Ueda H, Sugiyama A, Hanzawa K, Kumagai K, Okamura H and Takada H: Neutrophil proteinase 3-mediated induction of bioactive IL-18 secretion by human oral epithelial cells. J Immunol. 167:6568–6575. 2001. View Article : Google Scholar : PubMed/NCBI

100 

Ihim SA, Abubakar SD, Zian Z, Sasaki T, Saffarioun M, Maleknia S and Azizi G: Interleukin-18 cytokine in immunity, inflammation, and autoimmunity: Biological role in induction, regulation, and treatment. Front Immunol. 13:9199732022. View Article : Google Scholar : PubMed/NCBI

101 

Banerjee S and Bond JS: Prointerleukin-18 is activated by meprin beta in vitro and in vivo in intestinal inflammation. J Biol Chem. 283:31371–31377. 2008. View Article : Google Scholar : PubMed/NCBI

102 

Okamura H, Tsutsui H, Kashiwamura S, Yoshimoto T and Nakanishi K: Interleukin-18: A novel cytokine that augments both innate and acquired immunity. Adv Immunol. 70:281–312. 1998. View Article : Google Scholar : PubMed/NCBI

103 

Tsutsui H, Nakanishi K, Matsui K, Higashino K, Okamura H, Miyazawa Y and Kaneda K: IFN-gamma-inducing factor up-regulates Fas ligand-mediated cytotoxic activity of murine natural killer cell clones. J Immunol. 157:3967–3973. 1996. View Article : Google Scholar : PubMed/NCBI

104 

Li P, Li YL, Li ZY, Wu YN, Zhang CC, A X, Wang CX, Shi HT, Hui MZ, Xie B, et al: Cross talk between vascular smooth muscle cells and monocytes through interleukin-1β/interleukin-18 signaling promotes vein graft thickening. Arterioscler Thromb Vasc Biol. 34:2001–2011. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Hoshino T, Wiltrout RH and Young HA: IL-18 is a potent coinducer of IL-13 in NK and T cells: A new potential role for IL-18 in modulating the immune response. J Immunol. 162:5070–5077. 1999. View Article : Google Scholar : PubMed/NCBI

106 

Yoshimoto T, Mizutani H, Tsutsui H, Noben-Trauth N, Yamanaka K, Tanaka M, Izumi S, Okamura H, Paul WE and Nakanishi K: IL-18 induction of IgE: Dependence on CD4+ T cells, IL-4 and STAT6. Nat Immunol. 1:132–137. 2000. View Article : Google Scholar

107 

Yoshimoto T, Tsutsui H, Tominaga K, Hoshino K, Okamura H, Akira S, Paul WE and Nakanishi K: IL-18, although antiallergic when administered with IL-12, stimulates IL-4 and histamine release by basophils. Proc Natl Acad Sci USA. 96:13962–13966. 1999. View Article : Google Scholar : PubMed/NCBI

108 

Doshi MD, Reese PP, Hall IE, Schröppel B, Ficek J, Formica RN, Weng FL, Hasz RD, Thiessen-Philbrook H and Parikh CR: Utility of applying quality assessment tools for kidneys with KDPI ≥80. Transplantation. 101:1125–1133. 2017. View Article : Google Scholar

109 

Issaq H and Veenstra T: Two-dimensional polyacrylamide gel electrophoresis (2D-PAGE): Advances and perspectives. Biotechniques. 44:697–698. 7002008. View Article : Google Scholar : PubMed/NCBI

110 

Smith BJ: SDS polyacrylamide gel electrophoresis of proteins. Methods Mol Biol. 32:23–34. 1994.PubMed/NCBI

111 

Kielkopf CL, Bauer W and Urbatsch IL: Sodium dodecyl sulfate-polyacrylamide gel electrophoresis of proteins. Cold Spring Harb Protoc. 20212021.

112 

Pitt JJ: Principles and applications of liquid chromatography-mass spectrometry in clinical biochemistry. Clin Biochem Rev. 30:19–34. 2009.PubMed/NCBI

113 

Aslam B, Basit M, Nisar MA, Khurshid M and Rasool MH: Proteomics: Technologies and their applications. J Chromatogr Sci. 55:182–196. 2017. View Article : Google Scholar : PubMed/NCBI

114 

Li C, Chu S, Tan S, Yin X, Jiang Y, Dai X, Gong X, Fang X and Tian D: Towards higher sensitivity of mass spectrometry: A perspective from the mass analyzers. Front Chem. 9:8133592021. View Article : Google Scholar

115 

Wieser A, Schneider L, Jung J and Schubert S: MALDI-TOF MS in microbiological diagnostics-identification of microorganisms and beyond (mini review). Appl Microbiol Biotechnol. 93:965–974. 2012. View Article : Google Scholar

116 

van Leeuwen LL, Spraakman NA, Brat A, Huang H, Thorne AM, Bonham S, van Balkom BWM, Ploeg RJ, Kessler BM and Leuvenink HGD: Proteomic analysis of machine perfusion solution from brain dead donor kidneys reveals that elevated complement, cytoskeleton and lipid metabolism proteins are associated with 1-year outcome. Transpl Int. 34:1618–1629. 2021. View Article : Google Scholar : PubMed/NCBI

117 

Mulvey JF, Ul Shaheed S, Charles PD, Snashall C, Lo Faro ML, Sutton CW, Jochmans I, Pirenne J, van Kooten C, Leuvenink HGD, et al: Perfusate proteomes provide biological insight into oxygenated versus standard hypothermic machine perfusion in kidney transplantation. Ann Surg. 278:676–682. 2023.PubMed/NCBI

118 

Karpman D, Bekassy Z, Grunenwald A and Roumenina LT: A role for complement blockade in kidney transplantation. Cell Mol Immunol. 19:755–757. 2022. View Article : Google Scholar : PubMed/NCBI

119 

Yamanaka K, Kakuta Y, Miyagawa S, Nakazawa S, Kato T, Abe T, Imamura R, Okumi M, Maeda A, Okuyama H, et al: Depression of complement regulatory factors in rat and human renal grafts is associated with the progress of acute T-cell mediated rejection. PLoS One. 11:e01488812016. View Article : Google Scholar : PubMed/NCBI

120 

De Vries B, Matthijsen RA, Wolfs TGAM, Van Bijnen AAJHM, Heeringa P and Buurman WA: Inhibition of complement factor C5 protects against renal ischemia-reperfusion injury: Inhibition of late apoptosis and inflammation. Transplantation. 75:375–382. 2003. View Article : Google Scholar : PubMed/NCBI

121 

Biglarnia AR, Huber-Lang M, Mohlin C, Ekdahl KN and Nilsson B: The multifaceted role of complement in kidney transplantation. Nat Rev Nephrol. 14:767–781. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Zhang R: Donor-specific antibodies in kidney transplant recipients. Clin J Am Soc Nephrol. 13:182–192. 2018. View Article : Google Scholar :

123 

Nauser CL, Farrar CA and Sacks SH: Complement recognition pathways in renal transplantation. J Am Soc Nephrol. 28:2571–2578. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Sato T, Van Dixhoorn MG, Prins FA, Mooney A, Verhagen N, Muizert Y, Savill J, Van Es LA and Daha MR: The terminal sequence of complement plays an essential role in antibody-mediated renal cell apoptosis. J Am Soc Nephrol. 10:1242–1252. 1999. View Article : Google Scholar : PubMed/NCBI

125 

Shimizu A, Masuda Y, Kitamura H, Ishizaki M, Ohashi R, Sugisaki Y and Yamanaka N: Complement-mediated killing of mesangial cells in experimental glomerulonephritis: Cell death by a combination of apoptosis and necrosis. Nephron. 86:152–160. 2000. View Article : Google Scholar : PubMed/NCBI

126 

Zhou W, Farrar CA, Abe K, Pratt JR, Marsh JE, Wang Y, Stahl GL and Sacks SH: Predominant role for C5b-9 in renal ischemia/reperfusion injury. J Clin Invest. 105:1363–1371. 2000. View Article : Google Scholar : PubMed/NCBI

127 

Jane-wit D, Surovtseva YV, Qin L, Li G, Liu R, Clark P, Manes TD, Wang C, Kashgarian M, Kirkiles-Smith NC, et al: Complement membrane attack complexes activate noncanonical NF-κB by forming an Akt+ NIK+ signalosome on Rab5+ endosomes. Proc Natl Acad Sci USA. 112:9686–9691. 2015. View Article : Google Scholar

128 

Jager NM, Venema LH, Arykbaeva AS, Meter-Arkema AH, Ottens PJ, van Kooten C, Mollnes TE, Alwayn IPJ, Leuvenink HGD and Pischke SE; PROPER study consortium: Complement is activated during normothermic machine perfusion of porcine and human discarded kidneys. Front Immunol. 13:8313712022. View Article : Google Scholar : PubMed/NCBI

129 

Coskun A, Baykal AT, Kazan D, Akgoz M, Senal MO, Berber I, Titiz I, Bilsel G, Kilercik H, Karaosmanoglu K, et al: Proteomic analysis of kidney preservation solutions prior to renal transplantation. PLoS One. 11:e01687552016. View Article : Google Scholar : PubMed/NCBI

130 

Nieuwenhuijs-Moeke GJ, Pischke SE, Berger SP, Sanders JSF, Pol RA, Struys MMRF, Ploeg RJ and Leuvenink HGD: Ischemia and reperfusion injury in kidney transplantation: Relevant mechanisms in injury and repair. J Clin Med. 9:2532020. View Article : Google Scholar : PubMed/NCBI

131 

Kako K, Kato M, Matsuoka T and Mustapha A: Depression of membrane-bound Na+-K+-ATPase activity induced by free radicals and by ischemia of kidney. Am J Physiol. 254:C330–C337. 1988. View Article : Google Scholar : PubMed/NCBI

132 

Salvadori M, Rosso G and Bertoni E: Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant. 5:52–67. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Molitoris BA, Dahl R and Geerdes A: Cytoskeleton disruption and apical redistribution of proximal tubule Na(+)-K(+)-ATPase during ischemia. Am J Physiol. 263:F488–F495. 1992.PubMed/NCBI

134 

Caron A, Desrosiers RR and Béliveau R: Kidney ischemia-reperfusion regulates expression and distribution of tubulin subunits, beta-actin and rho GTPases in proximal tubules. Arch Biochem Biophys. 431:31–46. 2004. View Article : Google Scholar : PubMed/NCBI

135 

Genescà M, Sola A and Hotter G: Actin cytoskeleton derangement induces apoptosis in renal ischemia/reperfusion. Apoptosis. 11:563–571. 2006. View Article : Google Scholar : PubMed/NCBI

136 

Sanz AB, Sanchez-Niño MD, Ramos AM and Ortiz A: Regulated cell death pathways in kidney disease. Nat Rev Nephrol. 19:281–299. 2023. View Article : Google Scholar : PubMed/NCBI

137 

Garrod D and Chidgey M: Desmosome structure, composition and function. Biochim Biophys Acta. 1778:572–587. 2008. View Article : Google Scholar

138 

Chen CS and Zhu H: Protein microarrays. Biotechniques. 40:423–425, 427 passim. 2006. View Article : Google Scholar : PubMed/NCBI

139 

Uttamchandani M, Neo JL, Ong BNZ and Moochhala S: Applications of microarrays in pathogen detection and biodefence. Trends Biotechnol. 27:53–61. 2009. View Article : Google Scholar

140 

Shome M and Labaer J: Protein microarrays and their fabrication. Methods Mol Biol. 2597:131–142. 2023. View Article : Google Scholar

141 

Baboudjian M, Gondran-Tellier B, Boissier R, Ancel P, Marjollet J, Lyonnet L, François P, Sabatier F, Lechevallier E, Dutour A and Paul P: An enhanced level of VCAM in transplant preservation fluid is an independent predictor of early kidney allograft dysfunction. Front Immunol. 13:9669512022. View Article : Google Scholar : PubMed/NCBI

142 

Gok MA, Pelzers M, Glatz JFC, Shenton BK, Buckley PE, Peaston R, Cornell C, Mantle D, Soomro N, Jaques BC, et al: Do tissue damage biomarkers used to assess machine-perfused NHBD kidneys predict long-term renal function post-transplant? Clin Chim Acta. 338:33–43. 2003. View Article : Google Scholar : PubMed/NCBI

143 

Balupuri S, Talbot D, El-Sheikh M, Snowden C, Manas DM, Kirby J and Mantle D: Comparison of proteolytic enzymes and glutathione S-transferase levels in non-heart-beating donors' (NHBD) kidney perfusates. Clin Chem Lab Med. 38:1099–1102. 2000. View Article : Google Scholar

144 

Tejchman K, Sierocka A, Kotfis K, Kotowski M, Dolegowska B, Ostrowski M and Sienko J: Assessment of oxidative stress markers in hypothermic preservation of transplanted kidneys. Antioxidants (Basel). 10:12632021. View Article : Google Scholar : PubMed/NCBI

145 

Cohen J, Ratigan E, Shigeoka A, Steiner R, Stocks L and McKay D: Inflammatory profiling of hypothermic machine pumped kidney allografts. Am J Transplant. 15(Suppl 3): C2732015.

146 

Maritan E, Franchin M, Amico F, Ietto G, Villa F, Tozzi M, Ferraro S, Negri S, Alberio MG and Carcano G: Ischemia and reperfusion injury markers in kidney transplant: Mechanical perfusion vs cold storage. Preliminary experience: 536. Transplantation. 94(10S): S11592012. View Article : Google Scholar

147 

Boenink R, Astley ME, Huijben JA, Stel VS, Kerschbaum J, Ots-Rosenberg M, Åsberg AA, Lopot F, Golan E, Castro de la Nuez P, et al: The ERA registry annual report 2019: Summary and age comparisons. Clin Kidney J. 15:452–472. 2021. View Article : Google Scholar

148 

Aubert O, Kamar N, Vernerey D, Viglietti D, Martinez F, Duong-Van-Huyen JP, Eladari D, Empana JP, Rabant M, Verine J, et al: Long term outcomes of transplantation using kidneys from expanded criteria donors: Prospective, population based cohort study. BMJ. 351:h35572015. View Article : Google Scholar : PubMed/NCBI

149 

Tomita Y, Tojimbara T, Iwadoh K, Nakajima I and Fuchinoue S: Long-term outcomes in kidney transplantation from expanded-criteria donors after circulatory death. Transplant Proc. 49:45–48. 2017. View Article : Google Scholar : PubMed/NCBI

150 

Borda B, Németh T, Ottlakan A, Keresztes C, Kemény É and Lázár G: Post-transplantation morphological and functional changes in kidneys from expanded criteria donors. Physiol Int. 104:329–333. 2017. View Article : Google Scholar : PubMed/NCBI

151 

Tingle SJ, Figueiredo RS, Moir JA, Goodfellow M, Talbot D and Wilson CH: Machine perfusion preservation versus static cold storage for deceased donor kidney transplantation. Cochrane Database Syst Rev. 3:CD0116712019.PubMed/NCBI

152 

Tingle SJ, Thompson ER, Figueiredo RS, Moir JA, Goodfellow M, Talbot D and Wilson CH: Normothermic and hypothermic machine perfusion preservation versus static cold storage for deceased donor kidney transplantation. Cochrane Database Syst Rev. 7:CD0116712024.PubMed/NCBI

153 

Helanterä I, Ibrahim HN, Lempinen M and Finne P: Donor age, cold ischemia time, and delayed graft function. Clin J Am Soc Nephrol. 15:813–821. 2020. View Article : Google Scholar : PubMed/NCBI

154 

Heilman RL, Mathur A, Smith ML, Kaplan B and Reddy KS: Increasing the use of kidneys from unconventional and high-risk deceased donors. Am J Transplant. 16:3086–3092. 2016. View Article : Google Scholar : PubMed/NCBI

155 

Tasaki M, Saito K, Nakagawa Y, Ikeda M, Imai N, Narita I and Takahashi K: Effect of donor-recipient age difference on long-term graft survival in living kidney transplantation. Int Urol Nephrol. 46:1441–1446. 2014. View Article : Google Scholar : PubMed/NCBI

156 

Lim K, Lee YJ, Gwon JG, Jung CW, Yang J, Oh SW, Jo SK, Cho WY and Kim MG: Impact of donor age on the outcomes of kidney transplantation from deceased donors with histologic acute kidney injury. Transplant Proc. 51:2593–2597. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Scurt FG, Ernst A, Hammoud B, Wassermann T, Mertens PR, Schwarz A, Becker JU and Chatzikyrkou C: Effect of creatinine metrics on outcome after transplantation of marginal donor kidneys. Nephrology (Carlton). 27:973–982. 2022. View Article : Google Scholar : PubMed/NCBI

158 

Irish GL, Coates PT and Clayton PA: Association of admission, nadir, and terminal donor creatinine with kidney transplantation outcomes. Kidney Int Rep. 6:2075–2083. 2021. View Article : Google Scholar : PubMed/NCBI

159 

Perico N, Cattaneo D, Sayegh MH and Remuzzi G: Delayed graft function in kidney transplantation. Lancet. 364:1814–1827. 2004. View Article : Google Scholar : PubMed/NCBI

160 

Lai C, Yee SY, Ying T and Chadban S: Biomarkers as diagnostic tests for delayed graft function in kidney transplantation. Transpl Int. 34:2431–2441. 2021. View Article : Google Scholar : PubMed/NCBI

161 

Siedlecki A, Irish W and Brennan DC: Delayed graft function in the kidney transplant. Am J Transplant. 11:2279–2296. 2011. View Article : Google Scholar : PubMed/NCBI

162 

Schrezenmeier E, Müller M, Friedersdorff F, Khadzhynov D, Halleck F, Staeck O, Dürr M, Zhang K, Eckardt KU, Budde K and Lehner LJ: Evaluation of severity of delayed graft function in kidney transplant recipients. Nephrol Dial Transplant. 37:973–981. 2022. View Article : Google Scholar

163 

Parsons FM: Haemodialysis; indications and results. Postgrad Med J. 35:625–630, passim. 1959. View Article : Google Scholar : PubMed/NCBI

164 

Clark JE and Soricelli RR: Indications for dialysis. Med Clin North Am. 49:1213–1239. 1965. View Article : Google Scholar : PubMed/NCBI

165 

Hosgood SA, Callaghan CJ, Wilson CH, Smith L, Mullings J, Mehew J, Oniscu GC, Phillips BL, Bates L and Nicholson ML: Normothermic machine perfusion versus static cold storage in donation after circulatory death kidney transplantation: A randomized controlled trial. Nat Med. 29:1511–1519. 2023. View Article : Google Scholar : PubMed/NCBI

166 

Brat A, de Vries KM, van Heurn EWE, Huurman VAL, de Jongh W, Leuvenink HGD, van Zuilen AD, Haase-Kromwijk BJJM, de Jonge J, Berger SP and Hofker SH: Hypothermic machine perfusion as a national standard preservation method for deceased donor kidneys. Transplantation. 106:1043–1050. 2022. View Article : Google Scholar :

167 

Rijkse E, Bouari S, Kimenai HJAN, de Jonge J, de Bruin RWF, Slagter JS, van den Hoogen MWF, Ijzermans JNM, Hoogduijn MJ and Minnee RC: Additional Normothermic machine perfusion versus hypothermic machine perfusion in suboptimal donor kidney transplantation: Protocol of a randomized, controlled, open-label trial. Int J Surg Protoc. 25:227–237. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 54 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Baryła M, Skrzycki M, Danielewicz R, Kosieradzki M and Struga M: Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review). Int J Mol Med 54: 107, 2024.
APA
Baryła, M., Skrzycki, M., Danielewicz, R., Kosieradzki, M., & Struga, M. (2024). Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review). International Journal of Molecular Medicine, 54, 107. https://doi.org/10.3892/ijmm.2024.5431
MLA
Baryła, M., Skrzycki, M., Danielewicz, R., Kosieradzki, M., Struga, M."Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review)". International Journal of Molecular Medicine 54.6 (2024): 107.
Chicago
Baryła, M., Skrzycki, M., Danielewicz, R., Kosieradzki, M., Struga, M."Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review)". International Journal of Molecular Medicine 54, no. 6 (2024): 107. https://doi.org/10.3892/ijmm.2024.5431