v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

Assessing Donor Liver Quality and Restoring Graft Function in the Era of Extended Criteria Donors

  • Yimou Lin1,
  • Haitao Huang1,
  • Lifeng Chen2,
  • Ruihan Chen1,
  • Jimin Liu3,
  • Shusen Zheng1,4 and
  • Qi Ling1,4,* 
 Author information
Journal of Clinical and Translational Hepatology   2023;11(1):219-230

doi: 10.14218/JCTH.2022.00194

Abstract

Liver transplantation (LT) is the final treatment option for patients with end-stage liver disease. The increasing donor shortage results in the wide usage of grafts from extended criteria donors across the world. Using such grafts is associated with the elevated incidences of post-transplant complications including initial nonfunction and ischemic biliary tract diseases, which significantly reduce recipient survival. Although several clinical factors have been demonstrated to impact donor liver quality, accurate, comprehensive, and effective assessment systems to guide decision-making for organ usage, restoration or discard are lacking. In addition, the development of biochemical technologies and bioinformatic analysis in recent years helps us better understand graft injury during the perioperative period and find potential ways to restore graft function. Moreover, such advances reveal the molecular profiles of grafts or perfusate that are susceptible to poor graft function and provide insight into finding novel biomarkers for graft quality assessment. Focusing on donors and grafts, we updated potential biomarkers in donor blood, liver tissue, or perfusates that predict graft quality following LT, and summarized strategies for restoring graft function in the era of extended criteria donors. In this review, we also discuss the advantages and drawbacks of these potential biomarkers and offer suggestions for future research.

Graphical Abstract

Keywords

Liver transplantation, Extended criteria donors, Graft quality, Assessment, Biomarkers, Restoration

Introduction

Liver transplantation (LT) is a life-saving treatment option for patients with end-stage liver disease. In recent decades, good short- and long-term outcomes after LT have been achieved because of improvements in surgical technologies and organ preservation.1 Graft quality is believed to play a dominant role in early graft function and thereby dramatically influences graft survival and mortality after LT.2–4 Over the last decade, the disparity between the need for LT and the organ shortage is widening, which leads to the expanded usage of grafts from the extended criteria donors (ECDs).1 Traditionally, ECDs are donors with underlying medical diseases such as diabetes, or hypertension, advanced age, high-degree liver steatosis, prolonged ischemia time, pathogenic infection, prolonged intensive care unit stay, hypernatremia, and donation after circulatory death (DCD).5–7 ECD graft quality is routinely considered inferior because of their increased rate of post-transplant complications, such as primary graft nonfunction (PNF),2 early allograft dysfunction (EAD),4 and ischemic-type biliary lesions (ITBLs)8,9

PNF is early graft loss after LT and requires emergency regrafting, which occurs following 2–10% of LTs.10–12 ECDs include DCD donors13 and those with severe steatosis,14 prolonged ischemia time,15–17 and high donor bilirubin level18 sharply increase the risk of PNF, thereby reducing patient and graft survival. Unlike PNF, EAD represents marginal, usually reversible, graft function during the first postoperative week, and results in a higher morbidity and mortality.4 Compared with 1–10% seen in donation after brain death (DBD) LT, the incidence of biliary complications after DCD LT is approximately 10–30%,19–22 in which the time from asystole to cross-clamp is considered as a major risk factor.23 Moreover, advanced donor age, prolonged ischemia time, microvascular thrombosis, bile salt toxicity and immune injury may be the underlying mechanisms of the development of biliary complications.24,25

Therefore, ECDs should be well defined and precisely allocated to appropriate recipients. More importantly, in the era of ECD, effective systems need to be established to assess donor liver quality and guide the decision for organ usage or discard. Based on clinical risk parameters (Fig. 1), models like donor risk index,2 Eurotransplant donor risk index,26 and discard risk index18 were constructed to evaluate the risk of graft failure or discard, serving as useful tools to make decisions for organ allocation.2,26 However, those scoring models mainly focus on donor characteristics and cannot assess the degree of liver injury.27 Furthermore, combining clinical parameters with advanced molecular profiles, imaging, or histopathology may contribute to the development of better systems. In recent years, with the rapid development of multi-omics, single cell technology, and bioinformatic analysis, significant achievements have been made in revealing the molecular profiles that are closely related to poor graft outcomes, and which can provide novel biomarkers for evaluation of graft viability.

Clinical factors that influence graft quality during the entire process of liver transplantation.
Fig. 1  Clinical factors that influence graft quality during the entire process of liver transplantation.

Herein, we provide a review of potentially useful biomarkers in donor blood, liver tissue, and graft perfusate, which have been associated with impaired graft quality or predictive for the occurrence of EAD, PNF, and biliary complications after LT. In this review, we mainly focus on studies using human liver grafts. Given that the available biomarkers were insufficient in the field of LT, we also include experimental studies that have been performed in animal models. Furthermore, we summarize potential therapies for graft repairment during LT. Finally, we describe the pros and cons of the potential biomarkers, accompanied with suggestions for future graft assessment and restoration.

Potential biomarkers in donor blood

Donor serum alanine transferase (ALT), aspartate transferase (AST), total bilirubin, gamma glutamyl transpeptidase, and sodium concentration may reveal the underlying liver dysfunction and ischemic injury prior to graft procurement. Over the past decades, numerous studies have demonstrated that such laboratory disorders in donor blood are independent risk factors for early graft dysfunction following LT.18,26,28 In recent years, novel biomarkers in donor blood have been found to useful for predicting graft outcomes. By analyzing data from over 10,000 nondiabetic donors, Ezekian et al.29 showed that elevated donor serum hemoglobin A1c (HbA1c) >6.5% was associated with increased rate of PNF and decreased graft and patient survival. HbA1c is known to be a useful biomarker representing the average plasma glucose concentration within the last 3 months, serving as an early warning of diabetes. The liver undergoes glycogen deposition and hepatic steatosis resulting from diabetes.30,31 Therefore, it is worth noting that HbA1c may be a valuable marker for further stratifying marginal graft quality. In a large prospective study of 815 participants, Piemonti et al.32 identified increased serum donor interleukin 6 (IL6) and C-X-C motif chemokine ligand 10 (CXCL10) concentration as predictors of poor early graft function, graft failure and inferior graft survival after DBD LT. IL6 is responsible for transforming naïve B cells into mature plasma cells, as well as activating the production of IL17 to inhibit regulatory T lymphocyte (Treg) function.32 Alternatively, CXCL10 is a useful chemoattractant for macrophages, natural killer (NK) cells and dendritic cells (DCs), thereby shaping initial immunity.32 More interestingly, Pollara et al.33 found that elevated circulating mitochondria-derived damage-associated molecular patterns (mtDAMPs) in donor plasma were associated with severe inflammation response and the development of EAD following DBD LT in a group of 55 recipients. The major source of mtDAMPs may be the mitochondria released from graft tissue or cell death during organ procurement, suggesting that mtDAMPs might quantitatively assess graft injury.

Potential biomarkers in donor grafts

The liver, a multifunctional organ in the body, is mainly engaged in metabolism, synthesis, storage, detoxification, and complex immune activities. After implantation, the donor graft becomes the new center of the recipient to perform those functions.34 Therefore, the graft features could significantly regulate hepatic homeostasis and influence outcomes after LT (Table 1).35–55 Donor grafts could be gained for histological assessment and quantification of liver injury during LT. Histopathology is the gold standard for the diagnosis of steatosis, fibrosis, necrosis, inflammation, and cellular infiltration in liver grafts. In our center, pretransplant, and post-reperfusion liver biopsies are routinely performed, offering valuable clues for graft quality assessment (Supplementary Table 1).56 In addition, bile duct biopsies could provide valuable information to evaluate bile duct injury and predict graft outcomes. Dries et al.57 proposed a scoring system (Supplementary Table 2), including biliary epithelium, mural stroma, peribiliary vascular plexus, thrombosis, intramural bleeding, peribiliary gland, and inflammation, to quantify bile duct injury.

Table 1

Biomarkers from donor livers potentially useful for the prediction of graft outcomes following transplantation

BiomarkerStudySampleModel(s)GroupKey point
Genetic variantHeme oxygenase-1 A/T-allele genotypeBuis et al. (2008)35Pretransplant biopsiesHuman LTA-allele genotype (n=245) vs. TT-genotype (n=61)Graft with TT-genotype had elevated serum transaminases after LT and a higher incidence of PNF
HLA-C2 alleleHanvesakul et al. (2008)36Pretransplant biopsiesHuman LT459 livers biopsiesDonor grafts with HLA-C2 allele were associated with less incidence of graft dysfunction
GcfDNALevitsky et al. (2021)37Recipient bloodHuman LTNormal function (n=94) vs. Acute dysfunction (n=68)Elevated GcfDNA represented early graft injury after LT
GcfDNASchutz et al. (2017)38Recipient bloodHuman LT/Elevated GcfDNA could predict early graft injury
RNANrf2 mRNAZaman et al. (2007)39Pretransplant biopsiesHuman LT14 donor liver biopsiesHigher Nrf2 mRNA expression before IRI were associated with lower liver injury
MICA mRNAResch et al. (2021)40Pretransplant biopsiesHuman LT88 liver biopsiesHigh expression of MICA mRNA could reduce graft injury
MiR-22Khorsandi et al. (2015)41Post-reperfusion biopsiesHuman DCD LTPNF (n=7) vs. non-PNF (n=7)Graft miR-22 was associated with PNF
MiR-146b-5pLi et al. (2017)421.5 hours after LTHuman LTEAD (n=22) vs. non-EAD (n=20)Graft miR-146b-5p was associated with EAD
MiR-103 and miR-181Ling et al. (2017)43Pretransplant biopsiesHuman LTNODM (n=15) vs. non-NODM (n=15)Graft miR-103 and miR-181 were significantly associated with the development of NODM
CircFOXN2 and circNEXTIN3Wang et al. (2021)44Pretransplant biopsiesHuman LTEAD (n=29) vs. non-EAD (n=86)Two circRNAs were associated with EAD
LncRNA LOC103692832Chen et al. (2019)4512 hours after LTRat DBD LT model/Graft lncRNA LOC103692832 was related to early graft injury
ProteinSirtuin 1Nakamura et al. (2017)462 hours after LTHuman LT51 liver biopsiesHigh graft Sirtuin 1 was associated with superior liver function
Heme oxygenase-1Nakamura et al. (2018)472 hours after LTHuman LT51 liver biopsiesEnhanced Sirtuin 1 expression and protected against IRI
YAPLiu et al. (2019)483 hours after LTHuman LT60 liver biopsiesImproved early liver function
FGF15Gulfo et al. (2020)49Post-reperfusion biopsiesRat DBD LT model/Low graft FGF15 was associated with more severe hepatic damage and inhibited regeneration
CEACAM1Nakamura et al. (2020)50Pretransplant biopsiesHuman LT60 liver biopsiesHepatic CEACAM1 could prevent early graft injury
Hepatic occult collagen depositionHirao et al. (2021)51Pretransplant biopsiesHuman LTLow level (n=140) vs. High level (n=54)Increased risks of severe IRI and EAD
MetaboliteLysophospholipids, bile acids, phospholipids, sphingomyelins, and histidine metabolism productsCortes et al. (2014)52Pretransplant biopsiesHuman LTEAD (n=48) vs. non-EAD (n=48)Predictors for EAD
Lactate and phosphocholineFaitot et al. (2018)53Pretransplant biopsiesHuman LTEAD (n=7) vs. non-EAD (n=35)Predictors for EAD
single cell RNA sequencingA pro-inflammatory phenotype of KCs and a subset of DCsYang et al. (2021)5424 hours after LTRat steatotic LT modelFatty graft (n=3) vs. Control graft (n=3)A pro-inflammatory phenotype of KCs that highly expressed colony-stimulating factor 3 and a subset of DCs with high expression of XCR1 were enriched in the steatotic grafts
A dynamic transcription profileWang et al. (2021)55Grafts gained from preprocurement, at the end of organ preservation and 2 h after reperfusionHuman DBD LTn=1Showed a dynamic transcription profile of intrahepatic cells during LT

Genetic variants

With the advent of genome-wide association studies and pretransplant genetic analysis, a series of genes and variants have been found to be susceptible to graft injury.58 Heme oxygenase-1 (HO-1), a regulator of immune response, is considered to be cytoprotective gene of ischemia-reperfusion injury (IRI) during LT and is modulated by a single-nucleotide polymorphism A (-413) T.35 Buis et al.35 reported that, compared with recipients of a liver with an A-allele genotype (n=245), recipients of livers with an HO-1 TT-genotype (n=61) had dramatically elevated serum hepatic transaminases after LT and a higher incidence of PNF. HLA-C, which is the major inhibitory ligand for immunoglobulin-like receptors, inhibit the cytotoxic activity of NK cells, and therefore reduced liver inflammatory damage.59 In a large LT cohort of 459 patients, Hanvesakul et al.36 found that donor grafts with at least one HLA-C2 allele were associated with less incidence of graft dysfunction and rejection.

After LT, graft-derived cell-free DNA (GcfDNA), which is continuously released into recipient circulation because of cellular turnover, is a promising noninvasive biomarker to assess graft quality. Previous studies have showed that the elevated GcfDNA was a signal of early graft injury after LT, particularly acute cellular rejection.37,60,61,38 For example, a prospective study conducted by Schutz et al.38 demonstrated that GcfDNA increased by more than 50% 1 day following LT, probably because of the IRI. However, GcfDNA rapidly decreased to a median of <10% within 7–10 days without the recipient experiencing early graft injury over a 1 year observation period.38 This suggested that GcfDNA may be a precise and superior biomarker to predict early graft dysfunction compared with conventional liver function tests.

RNAs

Protein-coding associated RNAs, for example messenger RNA (mRNA) and noncoding RNAs including microRNAs (miRNAs), circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) are believed to be reliable markers to evaluate graft injury because of their organ specificity. Nrf2 transcription factor, which is activated by reactive oxygen species, is known to protector against liver IRI via activating phase II antioxidants.62 Zaman et al.39 demonstrated that grafts (n=6) with increased Nrf2 mRNA expression before IRI were associated with lower liver injury. Interestingly, donors with low Nrf2 mRNA levels (n=8) were significantly older than those with high levels, suggesting that older grafts experienced severe IRI39 and inferior graft quality. Additionally, Resch et al.40 reported that high gene expression of the major histocompatibility complex class 1 related chain A (MICA) mRNA in zero hour biopsies (n=88) was associated with mild graft injury and prolonged graft survival. During LT, MICA had an important role in linking the innate and adaptive immune responses via interacting with NK cells, mucosal-associated invariant T, CD8+T cells, et al.40 miR-22, a regulator of a series of pathways such as cell cycle, metabolism and kinase signaling, is relevant to cell survival, glucose metabolism, and protein translation.41 Khorsandi et al.41 rereported that low expression of graft miR-22 was associated with the incidence of PNF after DCD LT (n=21). Another study of 42 human LTs showed that high expression of donor graft miR-146b-5p was associated with the development of EAD.42 Downregulation of miR-146b increased the production of tumor necrosis factor receptor-associated factor 6, which activated the nuclear factor-kappa B (NF-κB) pathway, and in turn enhanced Treg function.42,63 In our previous study, we found that elevated donor graft miR-103 and miR-181 were significantly associated with the development of new-onset diabetes mellitus (NODM) in recipients following LT (n=30).43 NODM not only increased the risk of biliary stricture and cholangitis but also resulted in poor graft survival, serving as an indicator of poor graft quality as well.64 The two miRNAs targeted several genes related to glucose homeostasis and insulin signal transduction, which may have been the underlying mechanism.43

In a cohort of 115 human LTs, Wang et al.44 reported that low levels of donor graft circFOXN2 and circNEXTIN3 that regulated miR-135b-5p and miR-149-5p and had roles in hepatic IRI were associated with the incidence of EAD. In a mice model of IRI (Qu et al.65 identified 13 differentially expressed circRNAs (e.g., Chr3:83031528|83031748, Chr10:89473752|89483524) in postperfusion livers that were involved in more severe IRI in steatotic livers. In a rat LT model, Chen et al.45 demonstrated that lncRNA LOC103692832 in rat grafts was related to early graft injury following LT that was mediated by the expression of apoptosis-related genes like HMOX1 and ATF3. Nevertheless, the mechanisms of these potentially involved circRNAs and lncRNAs are still unclear, and further prospective or multicenter studies with larger samples are needed to verify the results.

Proteins

Sirtuin1, a histone/protein deacetylase that regulates inflammatory responses, cellular aging, and stress resistance, has an important role in autophagy induction involved in liver IRI.66 A previous study showed that high Sirtuin1 expression in grafts post-reperfusion sharply inhibited proinflammatory cytokine levels accompanied by superior liver function and improved patient survival.46 HO-1 is a rate-limiting enzyme that converts heme to biliverdin, free iron, carbon monoxide, and has anti-inflammatory and anti-oxidative activitiy.47 In addition, Nakamura et al.47 showed that high HO-1 levels in post-reperfusion liver biopsies (n=51) were associated with good liver function, dramatically enhanced Sirtuin1/LC3B expression, and protected against hepatic IRI by inducing autophagy. Notch1, a highly conserved transmembrane receptor, has been shown to reduce cellular apoptosis or necrosis and inflammatory response.55 Kageyama et al.44 demonstrated high Notch1 expression in grafts was correlated with low serum ALT levels, consistent with alleviated liver damage. In addition, Liu et al.48 found that high graft YAP expression after LT was linked with well-preserved histopathology and improved liver function at 1–7 days following LT. YAP is an effector of Hippo pathway and regulates cell proliferation and apoptosis and maintains hepatic homeostasis. FGF15, which is secreted from the ileum following inflammatory stimulation, binds to Fgfr4/Klb, which is followed by downregulation of CYP7A1 expression and inhibition of bile acid synthesis and activation of the Hippo pathway to upregulate YAP levels.49 In a rat DBD LT model, Gulfo et al.49 reported ed that low FGF15 levels in grafts was associated with more severe hepatic damage and inhibited regeneration that was mediated by increased CYP7A1 and decreased YAP levels.

The use of ECD grafts has raised the incidence of graft dysfunction, which ranges from reversible dysfunction, known as EAD, to irreversible dysfunction or PNF. Therefore, biomarkers to predict EAD and PNF are necessary in the era of ECD. CEACAM1 is a glycoprotein involved in hepatocyte differentiation and regeneration and regulation of insulin clearance, serving as a bridge between hepatic injury and metabolic homeostasis.50 Low CEACAM1 expression in human donor liver biopsies (n=60) was recognized as an independent predictor of EAD.50 In a large study cohort (n=194), Hirao et al.51 found that liver grafts with high occult collagen deposition were of increased risk of severe IRI and EAD, highlighting the effect of occult fibrosis on post-transplant outcome. In addition, Kurian et al.68 investigated several upregulated signaling pathways including NF-κB and targets such as CXCL1, IL1, TRAF6, TIPARP, TNFRSF1B, as predictors of EAD. Kornasiewicz et al.69 used graft proteomics to identify 21 significantly differentially expressed proteins in patients with (n=3) and without PNF (n=6). The proteins were mainly associated with mitochondrial oxidative phosphorylation or vital for the adenosine triphosphate-dependent turnover of proteins.

Metabolites

Cortes et al.52 used metabolomic profiling of 124 graft biopsies to identify significantly increased lysophospholipids, bile acids, phospholipids, sphingomyelins, and histidine metabolism products that were predictors for EAD. Based on the metabolic features, an EAD predictive model was established and further determined in a validation set (n=24) to have 91% sensitivity and 82% specificity. Likewise, Faitot et al.53 reported that lactate concentrations >8.3 mmol/g and phosphocholine concentrations >0.646 mmol/g were significantly associated with EAD. In our previous study, we identified metabolic profiles containing 57 dramatically differentially expressed metabolic features that were enriched in 24 common pathways including fatty acid, alanine, aspartate, thiamine, and riboflavin metabolism, the urea cycle, and ammonia recycling in PNF grafts.28 Graft metabolites and clinical characteristics were combined to develop a PNF predictive model derived from eight selected metabolic variations including achillicin, 3-hydroxypropanal, 3-oxododecanoic acid glycerides, and dopexamine in combination with clinical parameters including donor total bilirubin >2 ng/mL, graft weight >1.5 kg, cold ischemia time >10 h, graft warm ischemia time >60 m. The model had an area under curve of 0.930 for predicting PNF.28

Single cell technology

Recent advances in single cell RNA sequencing (scRNA-Seq) allow investigation of the transcriptomic landscape of single cells in organisms and have increased our understanding of the heterogeneity and relevance between cells. In a rat LT model, Yang et al.54 identified 11 kinds of cells in grafts and drew a single cell map of IRI after steatotic LT by scRNA-Seq. More importantly, they found a pro-inflammatory phenotype of Kupffer cells (KCs) that highly expressed colony-stimulating factor 3 and a subset of DCs with high expression of XCR1 that were enriched in steatotic grafts, suggesting their participation in fatty graft IRI.54 In addition, Wang et al.55 described a dynamic transcription profile of intrahepatic cells during LT by performing scRNA-Seq of grafts at preprocurement, at the end of organ preservation, and 2 h after reperfusion. They also found that a cluster of KCs that highly expressed TNFAIP3 interacting protein 3 after reperfusion, protected grafts against liver IRI.55 We believe that as research on scRNA-Seq deepens, it may provide a deeper understanding of mechanisms related to liver IRI during LT, identify grafts at increased risk of IRI and develop strategies to protect organ against liver damage. In a study published on BioRxiv, we established a graft-tolerant mouse LT model and identified two stages of graft recovery, which included an acute and stable phases.70 We also found that the interaction between CD206+MerTK+ macrophages and CD49a+CD49b NK cells regulated metabolic and immune remodeling of the graft.70

Potential biomarkers in perfusate

The donor graft and perfusate keep interplaying during preservation. Molecules including nucleic acids, proteins, and metabolites in perfusate may be associated with graft outcomes. In a review by Verhoeven et al.71 in 2014, ALT, AST, lactate dehydrogenase, lactate, adenine nucleotide level, hyaluronic acid, thrombomodulin and inflammatory markers (e.g., hypoxia-inducible factor-1α, and tumor necrosis factor-α) in perfusate and perfusate pH were useful biomarkers to assess graft quality. Machine perfusion (MP) such as hypothermic machine perfusion (HMP), hypothermic oxygenated perfusion (HOPE), and normothermic machine perfusion (NMP) continuously inject the perfusion fluid into the graft blood vessels to form a circuit, mitigating IRI and maintaining cellular metabolism in graft.72 So far, a series of current and ongoing clinical trials have shown that they were superior in reducing ischemic complications compared with static cold storage (SCS).73–76 In addition, the development of detection technology and MP have facilitated the discovery of a series of novel perfusate biomarkers for graft viability evaluation and are summarized as below and in Table 2.5,77–83

Table 2

Potential biomarkers of graft function that are found in graft perfusates

BiomarkerStudyModelGroupKey point
Bile productionPavel et al. (2019)77NMP5 discarded human DCD liversEarlier production of bile and higher bile flows during NMP were linked to better bile duct histology
Biliary bicarbonate, pH, and glucoseMatton et al. (2019)78NMP23 human donor liversHigh biliary bicarbonate and pH and low glucose were associated with bile duct injury
Bile/perfusate glucose ratio and bile/ perfusate Na+ ratioLinares-Cervantes et al. (2019)5A porcine DCD LT model; NMP/Bile/perfusate glucose ratio≤0.7 and bile/ perfusate Na+ ratio ≥1.1 were correlated with successful LT
CDmiRsVerhoeven et al. (2013)79Human LT; SCSGrafts developed ITBL (n=20) vs. Grafts without biliary strictures (n=37)CDmiRs could be predictive of bile duct injury and ITBL
miR-122Selten et al. (2017)80Human DCD/DBD LT; SCSEAD (n=35) vs. non-EAD (n=48)High miR-122 level could predict EAD
FMNMuller et al. (2019)81Human DCD/DBD LT; HOPE53 donor liversHigh FMN level could predict severe graft dysfunction following LT
D-dimerKarangwa et al. (2017)82NMP12 discarded human liversHigh D-dimer level was associated with graft damage
NGA2FVerhelst et al. (2018)83Human DCD/DBD LT; SCSPNF (n=3) vs. non-PNF (n=63)Increased NGA2F level could predict PNF

Bile production and bile composition (e.g., bile glucose and Na+) during NMP are useful biomarkers for graft synthesis function. Pavel et al.77 restored five discarded DCD livers with NMP for 12 h and found that earlier production of bile and higher bile flows during NMP contributed to better bile duct histology. In addition, Matton et al.78 showed that high biliary bicarbonate and pH, and low biliary glucose in human liver grafts (n=23) during NMP were significantly associated with high risk of bile duct injury. In a porcine LT model, Linares-Cervantes et al.5 demonstrated that a bile/perfusate glucose ratio ≤0.7 and a bile/perfusate Na+ ratio ≥1.1 within 4 h of NMP predicted graft survival after LT. Given that the role of donor graft miRNAs in predicting post-transplant outcomes, perfusate miRNAs may serve similarly. Furthermore, miRNAs have been shown to be stable in perfusate for at least 1 day.79 Verhoeven et al.79 showed that cholangiocyte-derived miRNAs (CDmiRs) in perfusate were predictive of bile duct injury and the development of ITBL. They also found that a significantly elevated hepatocyte-derived miRNA to CDmiRs ratio was associated with the incidence of ITBL. Moreover, Selten et al.80 reported that both high miR-122 levels and a high miR-122/miR-222 ratio in SCS perfusate predicted the development of EAD and poor graft survival after LT in 83 recipients.

Flavin mononucleotide (FMN), a critical molecular of generating electrons for ubiquinone reduction in mitochondrial complex 1, was shown to be associated with mitochondrial injury.81 Muller et al.81 preserved 53 grafts with HOPE and demonstrated that a high perfusate FMN level after 30 m of HOPE was strongly linked to severe graft dysfunction. Wang et al.84 infused 23 DCD livers with normothermic regional perfusion and found that the levels of perfusate FMN in transplantable grafts (n=15) were dramatically lower than those in nontransplantable grafts (n=8). D-dimer, a product of fibrin degradation, is a small protein fragment released during fibrinolysis. Karangwa et al.82 preserved 12 discard donor livers with NMP and showed that D-dimer levels >3,500 ng/mL were significantly associated with graft liver injury, suggesting that it was predictive of poor graft function.

In a multicenter cohort study, Verhelst et al.83 compared the glycome patterns in SCS perfusate in PNF (n=3) and non-PNF (n=63) groups and found that increased NGA2F, a single under galactosylated biantennary glycan, predicted the development of PNF with 100% accuracy. That highlighted the essential role of omics, especially the metabolomics, in discovering potential perfusate markers of poor graft function during LT.

Potential strategies for restoring graft function

In recent years, in vivo and ex vivo potential protective interventions that have been used to restore graft function are listed in Table 3.85–102 During the process of ex vivo therapies, the role of MP is apparent because it provides a platform for graft preconditioning.

Table 3

Potential therapies to restore donor liver function

TherapyStudyTargetModelOutcome
Gene therapyJiang et al. (2011)85Toll-like receptor 4 siRNAMice-IRI in vivoReduce liver IRI
Zhao et al. (2017)86High-mobility group box 1 siRNAMice-IRI in vivoReduce liver IRI
Gillooly et al. (2019)87siRNA against the Fas receptorRats HMP and NMPAbsorbed by rat donor livers during HMP and NMP
Goldaracena et al. (2017)88Antisense oligonucleotidePorcine LT NMPPrevent HCV replication or reinfection after LT
Cell therapyPeng et al. (2018)89DC+ apoptotic lymphocytesRat LT in vivoProlong rat survival
Sanchez-Fueyo A et al. (2020)90TregsHuman LT in vivoReduce antidonor T cell responses and play the potential role of graft rejection
Shi et al. (2017)91MSCsHuman LT in vivoSuppress acute rejection and improve graft histology
Verstegen et al. (2020)92MSCsPorcine LT HOPEAbsorbed by porcine grafts and continue to maintain paracrine activity after distribution
Extracellular vesiclesZheng et al. (2018)93EVs deprived from DCsRat IRI in vivoModulate differentiation of Tregs and protect liver against IRI
Chen et al. (2019)94EVs deprived from TregsRat LT in vivoProlong liver graft survival
Rigo et al. (2018)95EVs deprived from human liver stem cellsRats NMPAbsorbed by hepatocytes and reduce liver injury
Anti-inflammatory agentsGoldaracena et al. (2016)96Alprostadil, n-acetylcysteine, carbon monoxide, and sevofluranePorcine LT NMPReduce liver injury
Yu et al. (2019)97mcc950Porcine LT HMPReduce liver injury
VasodilatorsHara et al. (2016)98Prostaglandin E1Rat LT NMPReduce liver injury and improve bile production, energy status, and rat survival
Nassar et al. (2014)99Prostacyclin analog (epoprostenol)Porcine LT NMPHigh bile production and good histopathology
Echeverri et al. (2018)100Endothelin1 antagonist (BQ123), epoprostenol, verapamilPorcine LT NMPHigh hepatic artery flow and reduce hepatocyte injury
DefattingNagrath et al. (2009)101A cocktail*Rat NMPDecrease the intracellular lipid content of liver by 50% during 3 h perfusion
Boteon et al. (2019)102A cocktail* + L-carnitineHuman NMPDecrease liver triglycerides by 38% and macrosteatosis by 40% over 6 h perfusion

Gene therapy

Previous in vivo studies were performed to treat liver IRI by using small interfering RNA (siRNA). Jiang et al.85 silenced toll-like receptor 4, a critical mediator of inflammation, in a hepatic IRI mouse model, resulting is significant reduction of serum transferases and histological injury. In another study, Zhao et al.86 downregulated nuclear high-mobility group box 1 by transfecting mice with siRNA and found that it effectively inhibited the expression of serum inflammatory cytokines and protected the liver against IRI. Although the efficacy of hydrodynamic injection has been shown in these animal models, it is difficult to use in the clinic because of off-target effects. Recent studies of graft perfusates showed a potential to solve this problem. For example, Gillooly et al.87 found that Fas siRNA directly added to the perfusate was successfully delivered to rat livers during HMP and NMP. This technology ensured that the siRNA only targeted the grafts, opening a new door for graft reconditioning. Antisense oligonucleotide, another gene modulation agent, was demonstrated to significantly reduce miR-122 expression and inhibit hepatitis C virus replication or reinfection after LT in a porcine LT model with NMP, further confirming the possibility of ex vivo gene therapy in grafts.88

Cell therapy

In vivo cell therapies such as tolerogenic DCs, Tregs, and mesenchymal stem cells (MSCs) have a role in immunomodulation. In a rat LT model, we innovatively treated acute rejection with a combination of galectin-1-induced tolerogenic DCs and apoptotic lymphocytes, which resulted in prolonged survival of the treated rats, with 37.5% surviving over 100 days, compared with untreated, all of which died within 14 days.90 In a phase I clinical trial, Sanchez-Fueyo et al.90 demonstrated that autologous Tregs transfer was safe and effective in reducing antidonor T cell responses after LT by intravenously administering autologous Tregs to the LT candidates. In addition, Shi et al.91 found that human MSCs injection in LT recipients suppressed acute rejection and improved graft histology by upregulating the Treg/T help 17 cell ratio. Compared with in vivo treatment, ex vivo technology provides novel strategies for graft restoration. For instance, Verstegen et al.92 showed in a porcine LT model that MSCs directly added to the perfusate during HOPE were effectively distributed to the porcine grafts, which continued to maintain their paracrine activity after distribution.

Extracellular vesicles

It has been reported that the above tolerogenic cells had the potential to undergo spontaneous malignant transformation.103 Therefore, some investigators began to use MSC-, DC- and trig-derived extracellular vesicles (EVs) as alternatives to cell therapy. In in vivo mice and rat IRI models, MSC-derived EVs had a diverse set of functions including mitochondrial autophagy,104,105 inhibition of immune response106,107 and liver regeneration.108,109 Zheng et al.93 found in a rat IRI model that DC-derived EVs could protect liver against IRI through modulating differentiation of Tregs. In a rat LT model, Chen et al.94 demonstrated that injection with Tregs-derived EVs after LT suppressed the proliferation of CD8+ cytotoxic T cells and prolonged liver graft survival. Compared to the in vivo injection, the ex vivo technology has the potential to directly target donor grafts without concern for off-target effect. Rigo et al.95 successfully delivered human liver stem cells-derived EVs into the rat livers during NMP, leading to less histological damage and lower levels of AST and lactate dehydrogenase in the treated group.

Anti-inflammatory agents

Liver IRI is characterized by the activation of pro-inflammatory responses. Therefore, adding anti-inflammatory agents to perfusate may regulate immune response and alleviate graft damage. In a porcine LT model, Goldaracena et al.96 put alprostadil, n-acetylcysteine, carbon monoxide, and sevoflurane into the NMP perfusate, showing significantly decreased interleukin-6, tumor necrosis factor-α, and AST during NMP, and lower AST and bilirubin levels in serum after LT in the treated group.96 In addition, Yu et al.97 used Mcc950, which strongly inhibited the nucleotide-binding domain leucine-rich repeat containing family pyrin domain containing 3 inflammasome, as an addition to the HMP perfusate in a porcine LT model. They found that Mcc950 significantly reduced inflammatory cytokines and histological injury, and prolonged long-term survival after LT.

Vasodilators

During the ischemic phase of LT, rapid adenosine triphosphate depletion and lack of blood flow result in mitochondrial dysfunction and liver sinusoidal endothelial cell (LSEC) injury.110 After reperfusion, the injured LSECs not only produce insufficient vasodilators but also expressed P-selectin to accumulate platelets, which resulted in microcirculation disorder.110 In a rat LT model, Hara et al.98 inhibited the accumulation of platelets by adding prostaglandin E1 (PGE1) to the perfusate under normothermic conditions. PGE1 ameliorated serum liver enzymes and histologic necrosis, and significantly improved bile production and energy status. In addition, Nassar et al.99 added a prostacyclin analog (epoprostenol) to NMP perfusate to preserve porcine livers and found that the use of prostacyclin analog led to high bile production and good histopathology. Furthermore, Echeverri et al.100 compared the effects of endothelin1 antagonist (BQ123), prostacyclin analog (epoprostenol) and calcium channel antagonist (verapamil) to treat hepatic artery vasospasm induced by IRI in a porcine LT model. They demonstrated that grafts with BQ123 and verapamil treatment had higher hepatic artery flow and less hepatocyte injury compared with those treated with epoprostenol.

Defatting agents

Moderate to severe (>30%) macrosteatosis is a well-known risk factor for poor graft quality, making it necessary to defat prior to LT.14 Nagrath et al.101 treated rat fatty livers with a combination of six defatting agents normothermically and showed that the treatment could decrease the intracellular lipid content of rat liver by 50% after 3 h perfusion. Furthermore, Boteon et al.102 assessed the efficacy of the above six agents combined with additional L-carnitine in defatting human livers with severe steatosis. They found that this method reduced liver triglycerides and macrosteatosis by 38% and 40% over 6 h NMP, enhanced metabolic parameters including increased urea and bile production, and downregulated biomarkers of liver injury (e.g., lower ALT and reduced inflammatory cytokines).

Other agents

In addition to the above agents, human atrial natriuretic peptide (hANP), heavy water, marine worm super hemoglobin (M101), glycine, relaxin, and polyethylene glycols have been found to alleviate liver injury.111–116 Nigmet et al.111 added hANP, a protective cardiovascular hormone for vascular endothelia, to SCS perfusate to preserve rat livers, showing that hANP supplementation decreased transaminase release, increased bile production, and protected sinusoidal architecture. In a porcine LT model, Alix et al.113 added M101 to SCS perfusate and demonstrated that M101 significantly reduced blood levels of ALT, AST, and tumor necrosis factor α in recipients 3 days following LT. Moreover, Gassner et al.114 used glycine, a simple amino acid that protected sinusoidal cells and hepatocytes, as an addition to NMP rat liver perfusate. They found less sinusoidal dilatation and tissue damage in the treated group.

Conclusions and perspectives

This review summarized and updated biomarkers in donor blood, liver tissue or graft perfusate to evaluate early graft injury (e.g., EAD, and PNF) and ITBL, and to identify potential therapies for graft repairment during the era of ECD. We focused on studies using human liver grafts and investigations of potential biomarkers involved in anti- or pro-inflammatory processes, which in turn shape immunity, regulate graft IRI, and further influence the development of EAD, PNF, or ITBL following LT. Given that relevant mechanisms of some molecules are lacking, further prospective studies and experiments are urgently needed to clearly understand their roles.

Although various biomarkers with available prognostic and diagnostic value in graft quality assessment have been widely explored, few are currently used in clinical practice. Current challenges associated with biomarker discovery research are as follows. Firstly, the sample sizes of these studies were small and mainly limited to single centers, suggesting that large multicenter cohorts or prospective randomized clinical trials are greatly necessary. Another problem is that the studies lack standardized endpoints and control groups.117 Graft quality is commonly considered to be associated with early graft dysfunction or ITBL, yet other complications after LT (e.g., ACR, metabolic disorders, and graft steatosis or fibrosis) are still a matter of substantial debate. Therefore, we primarily summarized biomarkers predictive of EAD, PNF, and ITBL. Current studies mainly focus on finding biomarkers related to early graft injury, do not have prolonged follow-up and overlook long-term complications like ITBL. Importantly, the measurement of biomarkers should be rapid and easy and have high predictive specificity and sensitivity for graft quality. However, detection of potential biomarkers is costly and time consuming. Moreover, biomarkers need to be stable and measurable during graft procurement, preservation, and implantation.

Despite the availability of liver biopsies for histological assessment and quantification of liver injury during LT, they are invasive and only represent specific parts of the grafts. On the contrary, perfusates can be collected in large volumes and contain markers from the whole graft. In recent years, MP has constantly advanced, and it use in evaluation of graft viability has gradually increased. Nevertheless, different regions or centers have their own standards to determine graft quality.78,118 More clear international guidelines that could guide the decision for organ usage, discard, or restoration prior to LT are recommended. In addition, we believe that MP could provide a platform for graft preconditioning, making it convenient to explore novel strategies for graft repair. Although high cost and the technical complexity limit wide usage of MP at its current stage, recently completed and ongoing clinical trials will make it an indispensable part of LT.72,73

Supporting information

Supplementary Table 1

Histological scoring of hepatocellular damage.56

(DOCX)

Supplementary Table 2

Histological scoring system for assessing injury of the distal common bile duct.57

(DOCX)

Abbreviations

ACR: 

acute cellular rejection

ALT: 

alanine transferase

AST: 

aspartate transferase

circRNAs: 

circular RNAs

DBD: 

donation after brain death

DCD: 

donation after circulatory death

DCs: 

dendritic cells

EAD: 

early allograft dysfunction

ECD: 

extended criteria donor

EV: 

extracellular vesicle

FMN: 

flavin mononucleotide

GcfDNA: 

graft-derived cell-free DNA

HbA1c: 

hemoglobin A1c

HCV: 

hepatitis C virus

HMP: 

hypothermic machine perfusion

HO-1: 

Heme oxygenase-1

HOPE: 

hypothermic oxygenated perfusion

IRI: 

ischemia-reperfusion injury

ITBL: 

ischemic-type biliary lesion

KC: 

Kupffer cell

lncRNA: 

long noncoding RNA

LSEC: 

liver sinusoidal endothelial cell

LT: 

liver transplantation

MICA: 

major histocompatibility complex class 1 related chain A

miRNA: 

microRNA

MP: 

machine perfusion

mRNA: 

messenger RNA

MSC: 

mesenchymal stem cell

mtDAMP: 

mitochondria-derived damage-associated molecular pattern

NF-κB: 

nuclear factor-kappa B

NK: 

natural killer

NMP: 

normothermic machine perfusion

NODM: 

new-onset diabetes mellitus

PGE1: 

Prostaglandin E1

PNF: 

primary graft nonfunction

scRNA-Seq: 

single cell RNA sequencing

SCS: 

static cold storage

siRNA: 

small interfering RNA

Treg: 

regulatory T lymphocyte

Declarations

Funding

This article was funded by the National Natural Science Foundation of China (No. 82171757) and the Zhejiang Province Natural Science Foundation of China (No. LZ22H030004).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Conceived of the paper (QL), wrote the original draft (YL), generated the figures (YL, HH), reviewed and edited the paper (QL, LC, RC, JL, SZ). All the authors agreed to the published version of the manuscript.

References

  1. Bodzin AS, Baker TB. Liver Transplantation Today: Where We Are Now and Where We Are Going. Liver Transpl 2018;24(10):1470-1475 View Article PubMed/NCBI
  2. Feng S, Goodrich NP, Bragg-Gresham JL, Dykstra DM, Punch JD, DebRoy MA, et al. Characteristics associated with liver graft failure: the concept of a donor risk index. Am J Transplant 2006;6(4):783-790 View Article PubMed/NCBI
  3. Agopian VG, Petrowsky H, Kaldas FM, Zarrinpar A, Farmer DG, Yersiz H, et al. The evolution of liver transplantation during 3 decades: analysis of 5347 consecutive liver transplants at a single center. Ann Surg 2013;258(3):409-421 View Article PubMed/NCBI
  4. Lee DD, Singh A, Burns JM, Perry DK, Nguyen JH, Taner CB. Early allograft dysfunction in liver transplantation with donation after cardiac death donors results in inferior survival. Liver Transpl 2014;20(12):1447-1453 View Article PubMed/NCBI
  5. Linares-Cervantes I, Echeverri J, Cleland S, Kaths JM, Rosales R, Goto T, et al. Predictor parameters of liver viability during porcine normothermic ex situ liver perfusion in a model of liver transplantation with marginal grafts. Am J Transplant 2019;19(11):2991-3005 View Article PubMed/NCBI
  6. Nair A, Hashimoto K. Extended criteria donors in liver transplantation-from marginality to mainstream. Hepatobiliary Surg Nutr 2018;7(5):386-388 View Article PubMed/NCBI
  7. Zheng J, Xiang J, Zhou J, Li Z, Hu Z, Lo CM, et al. Liver grafts for transplantation from donors with diabetes: an analysis of the Scientific Registry of Transplant Recipients database. PLoS One 2014;9(5):e98104 View Article PubMed/NCBI
  8. Foley DP, Fernandez LA, Leverson G, Anderson M, Mezrich J, Sollinger HW, et al. Biliary complications after liver transplantation from donation after cardiac death donors: an analysis of risk factors and long-term outcomes from a single center. Ann Surg 2011;253(4):817-825 View Article PubMed/NCBI
  9. Brunner SM, Junger H, Ruemmele P, Schnitzbauer AA, Doenecke A, Kirchner GI, et al. Bile duct damage after cold storage of deceased donor livers predicts biliary complications after liver transplantation. J Hepatol 2013;58(6):1133-1139 View Article PubMed/NCBI
  10. D’Alessandro AM, Ploeg RJ, Knechtle SJ, Pirsch JD, Stegall MD, Hoffmann R, et al. Retransplantation of the liver—a seven-year experience. Transplantation 1993;55(5):1083-1087 View Article PubMed/NCBI
  11. Ploeg RJ, D’Alessandro AM, Knechtle SJ, Stegall MD, Pirsch JD, Hoffmann RM, et al. Risk factors for primary dysfunction after liver transplantation—a multivariate analysis. Transplantation 1993;55(4):807-813 View Article PubMed/NCBI
  12. Al-Freah MAB, McPhail MJW, Dionigi E, Foxton MR, Auzinger G, Rela M, et al. Improving the Diagnostic Criteria for Primary Liver Graft Nonfunction in Adults Utilizing Standard and Transportable Laboratory Parameters: An Outcome-Based Analysis. Am J Transplant 2017;17(5):1255-1266 View Article PubMed/NCBI
  13. Coll E, Miñambres E, Sánchez-Fructuoso A, Fondevila C, Campo-Cañaveral de la Cruz JL, Domínguez-Gil B. Uncontrolled Donation After Circulatory Death: A Unique Opportunity. Transplantation 2020;104(8):1542-1552 View Article PubMed/NCBI
  14. Croome KP, Mathur AK, Mao S, Aqel B, Piatt J, Senada P, et al. Perioperative and long-term outcomes of utilizing donation after circulatory death liver grafts with macrosteatosis: A multicenter analysis. Am J Transplant 2020;20(9):2449-2456 View Article PubMed/NCBI
  15. De Carlis R, Di Sandro S, Lauterio A, Botta F, Ferla F, Andorno E, et al. Liver Grafts From Donors After Circulatory Death on Regional Perfusion With Extended Warm Ischemia Compared With Donors After Brain Death. Liver Transpl 2018;24(11):1523-1535 View Article PubMed/NCBI
  16. Farid SG, Attia MS, Vijayanand D, Upasani V, Barlow AD, Willis S, et al. Impact of Donor Hepatectomy Time During Organ Procurement in Donation After Circulatory Death Liver Transplantation: The United Kingdom Experience. Transplantation 2019;103(4):e79-e88 View Article PubMed/NCBI
  17. Paterno F, Guarrera JV, Wima K, Diwan T, Cuffy MC, Anwar N, et al. Clinical Implications of Donor Warm and Cold Ischemia Time in Donor After Circulatory Death Liver Transplantation. Liver Transpl 2019;25(9):1342-1352 View Article PubMed/NCBI
  18. Rana A, Sigireddi RR, Halazun KJ, Kothare A, Wu MF, Liu H, et al. Predicting Liver Allograft Discard: The Discard Risk Index. Transplantation 2018;102(9):1520-1529 View Article PubMed/NCBI
  19. Dubbeld J, Hoekstra H, Farid W, Ringers J, Porte RJ, Metselaar HJ, et al. Similar liver transplantation survival with selected cardiac death donors and brain death donors. Br J Surg 2010;97(5):744-753 View Article PubMed/NCBI
  20. Chan EY, Olson LC, Kisthard JA, Perkins JD, Bakthavatsalam R, Halldorson JB, et al. Ischemic cholangiopathy following liver transplantation from donation after cardiac death donors. Liver Transpl 2008;14(5):604-610 View Article PubMed/NCBI
  21. Abt P, Crawford M, Desai N, Markmann J, Olthoff K, Shaked A. Liver transplantation from controlled non-heart-beating donors: an increased incidence of biliary complications. Transplantation 2003;75(10):1659-1663 View Article PubMed/NCBI
  22. Pine JK, Aldouri A, Young AL, Davies MH, Attia M, Toogood GJ, et al. Liver transplantation following donation after cardiac death: an analysis using matched pairs. Liver Transpl 2009;15(9):1072-1082 View Article PubMed/NCBI
  23. Taner CB, Bulatao IG, Perry DK, Sibulesky L, Willingham DL, Kramer DJ, et al. Asystole to cross-clamp period predicts development of biliary complications in liver transplantation using donation after cardiac death donors. Transpl Int 2012;25(8):838-846 View Article PubMed/NCBI
  24. Hessheimer AJ, Cárdenas A, García-Valdecasas JC, Fondevila C. Can we prevent ischemic-type biliary lesions in donation after circulatory determination of death liver transplantation?. Liver Transpl 2016;22(7):1025-1033 View Article PubMed/NCBI
  25. de Vries Y, von Meijenfeldt FA, Porte RJ. Post-transplant cholangiopathy: Classification, pathogenesis, and preventive strategies. Biochim Biophys Acta Mol Basis Dis 2018;1864(4 Pt B):1507-1515 View Article PubMed/NCBI
  26. Braat AE, Blok JJ, Putter H, Adam R, Burroughs AK, Rahmel AO, et al. The Eurotransplant donor risk index in liver transplantation: ET-DRI. Am J Transplant 2012;12(10):2789-2796 View Article PubMed/NCBI
  27. Flores A, Asrani SK. The donor risk index: A decade of experience. Liver Transpl 2017;23(9):1216-1225 View Article PubMed/NCBI
  28. Zhang X, Zhang C, Huang H, Chen R, Lin Y, Chen L, et al. Primary nonfunction following liver transplantation: Learning of graft metabolites and building a predictive model. Clin Transl Med 2021;11(7):e483 View Article PubMed/NCBI
  29. Ezekian B, Mulvihill MS, Freischlag K, Yerokun BA, Davis RP, Hartwig MG, et al. Elevated HbA1c in donor organs from patients without a diagnosis of diabetes portends worse liver allograft survival. Clin Transplant 2017;31(9):e13047 View Article PubMed/NCBI
  30. Stone BG, Van Thiel DH. Diabetes mellitus and the liver. Semin Liver Dis 1985;5(1):8-28 View Article PubMed/NCBI
  31. Kotronen A, Juurinen L, Tiikkainen M, Vehkavaara S, Yki-Järvinen H. Increased liver fat, impaired insulin clearance, and hepatic and adipose tissue insulin resistance in type 2 diabetes. Gastroenterology 2008;135(1):122-130 View Article PubMed/NCBI
  32. Piemonti L, Sordi V, Pellegrini S, Scotti GM, Scavini M, Sioli V, et al. Circulating CXCL10 and IL-6 in solid organ donors after brain death predict graft outcomes. Sci Rep 2021;11(1):6624 View Article PubMed/NCBI
  33. Pollara J, Edwards RW, Lin L, Bendersky VA, Brennan TV. Circulating mitochondria in deceased organ donors are associated with immune activation and early allograft dysfunction. JCI Insight 2018;3(15):121622 View Article PubMed/NCBI
  34. Ling Q, Xu X, Wang B, Li L, Zheng S. The Origin of New-Onset Diabetes After Liver Transplantation: Liver, Islets, or Gut?. Transplantation 2016;100(4):808-813 View Article PubMed/NCBI
  35. Buis CI, van der Steege G, Visser DS, Nolte IM, Hepkema BG, Nijsten M, et al. Heme oxygenase-1 genotype of the donor is associated with graft survival after liver transplantation. Am J Transplant 2008;8(2):377-385 View Article PubMed/NCBI
  36. Hanvesakul R, Spencer N, Cook M, Gunson B, Hathaway M, Brown R, et al. Donor HLA-C genotype has a profound impact on the clinical outcome following liver transplantation. Am J Transplant 2008;8(9):1931-1941 View Article PubMed/NCBI
  37. Levitsky J, Kandpal M, Guo K, Kleiboeker S, Sinha R, Abecassis M. Donor-derived cell-free DNA levels predict graft injury in liver transplant recipients. Am J Transplant 2022;22(2):532-540 View Article PubMed/NCBI
  38. Schütz E, Fischer A, Beck J, Harden M, Koch M, Wuensch T, et al. Graft-derived cell-free DNA, a noninvasive early rejection and graft damage marker in liver transplantation: A prospective, observational, multicenter cohort study. PLoS Med 2017;14(4):e1002286 View Article PubMed/NCBI
  39. Zaman MB, Leonard MO, Ryan EJ, Nolan NP, Hoti E, Maguire D, et al. Lower expression of Nrf2 mRNA in older donor livers: a possible contributor to increased ischemia-reperfusion injury?. Transplantation 2007;84(10):1272-1278 View Article PubMed/NCBI
  40. Resch T, Hackl H, Esser H, Günther J, Schwelberger H, Ritschl PV, et al. Expression of MICA in Zero Hour Biopsies Predicts Graft Survival After Liver Transplantation. Front Immunol 2021;12:606146 View Article PubMed/NCBI
  41. Khorsandi SE, Quaglia A, Salehi S, Jassem W, Vilca-Melendez H, Prachalias A, et al. The microRNA Expression Profile in Donation after Cardiac Death (DCD) Livers and Its Ability to Identify Primary Non Function. PLoS One 2015;10(5):e0127073 View Article PubMed/NCBI
  42. Li C, Zhao Q, Zhang W, Chen M, Ju W, Wu L, et al. MicroRNA-146b-5p Identified in Porcine Liver Donation Model is Associated with Early Allograft Dysfunction in Human Liver Transplantation. Med Sci Monit 2017;23:5876-5884 View Article PubMed/NCBI
  43. Ling Q, Xie H, Li J, Liu J, Cao J, Yang F, et al. Donor Graft MicroRNAs: A Newly Identified Player in the Development of New-onset Diabetes After Liver Transplantation. Am J Transplant 2017;17(1):255-264 View Article PubMed/NCBI
  44. Wang K, Wei X, Wei Q, Lu D, Li W, Pan B, et al. A two-circular RNA signature of donor circFOXN2 and circNECTIN3 predicts early allograft dysfunction after liver transplantation. Ann Transl Med 2020;8(4):94 View Article PubMed/NCBI
  45. Chen S, Fang H, Li J, Shi J, Zhang J, Wen P, et al. Microarray Analysis For Expression Profiles of lncRNAs and circRNAs in Rat Liver after Brain-Dead Donor Liver Transplantation. Biomed Res Int 2019;2019:5604843 View Article PubMed/NCBI
  46. Nakamura K, Kageyama S, Ke B, Fujii T, Sosa RA, Reed EF, et al. Sirtuin 1 attenuates inflammation and hepatocellular damage in liver transplant ischemia/Reperfusion: From mouse to human. Liver Transpl 2017;23(10):1282-1293 View Article PubMed/NCBI
  47. Nakamura K, Kageyama S, Yue S, Huang J, Fujii T, Ke B, et al. Heme oxygenase-1 regulates sirtuin-1-autophagy pathway in liver transplantation: From mouse to human. Am J Transplant 2018;18(5):1110-1121 View Article PubMed/NCBI
  48. Liu Y, Lu T, Zhang C, Xu J, Xue Z, Busuttil RW, et al. Activation of YAP attenuates hepatic damage and fibrosis in liver ischemia-reperfusion injury. J Hepatol 2019;71(4):719-730 View Article PubMed/NCBI
  49. Gulfo J, Rotondo F, Ávalos de León CG, Cornide-Petronio ME, Fuster C, Gracia-Sancho J, et al. FGF15 improves outcomes after brain dead donor liver transplantation with steatotic and non-steatotic grafts in rats. J Hepatol 2020;73(5):1131-1143 View Article PubMed/NCBI
  50. Nakamura K, Kageyama S, Kaldas FM, Hirao H, Ito T, Kadono K, et al. Hepatic CEACAM1 expression indicates donor liver quality and prevents early transplantation injury. J Clin Invest 2020;130(5):2689-2704 View Article PubMed/NCBI
  51. Hirao H, Ito T, Kadono K, Kojima H, Naini BV, Nakamura K, et al. Donor Hepatic Occult Collagen Deposition Predisposes to Peritransplant Stress and Impacts Human Liver Transplantation. Hepatology 2021;74(5):2759-2773 View Article PubMed/NCBI
  52. Cortes M, Pareja E, García-Cañaveras JC, Donato MT, Montero S, Mir J, et al. Metabolomics discloses donor liver biomarkers associated with early allograft dysfunction. J Hepatol 2014;61(3):564-574 View Article PubMed/NCBI
  53. Faitot F, Besch C, Battini S, Ruhland E, Onea M, Addeo P, et al. Impact of real-time metabolomics in liver transplantation: Graft evaluation and donor-recipient matching. J Hepatol 2018;68(4):699-706 View Article PubMed/NCBI
  54. Yang X, Lu D, Wang R, Lian Z, Lin Z, Zhuo J, et al. Single-cell profiling reveals distinct immune phenotypes that contribute to ischaemia-reperfusion injury after steatotic liver transplantation. Cell Prolif 2021;54(10):e13116 View Article PubMed/NCBI
  55. Wang L, Li J, He S, Liu Y, Chen H, He S, et al. Resolving the graft ischemia-reperfusion injury during liver transplantation at the single cell resolution. Cell Death Dis 2021;12(6):589 View Article PubMed/NCBI
  56. Xia W, Ke Q, Wang Y, Feng X, Guo H, Wang W, et al. Donation after cardiac death liver transplantation: Graft quality evaluation based on pretransplant liver biopsy. Liver Transpl 2015;21(6):838-846 View Article PubMed/NCBI
  57. op den Dries S, Westerkamp AC, Karimian N, Gouw AS, Bruinsma BG, Markmann JF, et al. Injury to peribiliary glands and vascular plexus before liver transplantation predicts formation of non-anastomotic biliary strictures. J Hepatol 2014;60(6):1172-1179 View Article PubMed/NCBI
  58. Yang JY, Sarwal MM. Transplant genetics and genomics. Nat Rev Genet 2017;18(5):309-326 View Article PubMed/NCBI
  59. Parham P. MHC class I molecules and KIRs in human history, health and survival. Nat Rev Immunol 2005;5(3):201-214 View Article PubMed/NCBI
  60. Zhao D, Zhou T, Luo Y, Wu C, Xu D, Zhong C, et al. Preliminary clinical experience applying donor-derived cell-free DNA to discern rejection in pediatric liver transplant recipients. Sci Rep 2021;11(1):1138 View Article PubMed/NCBI
  61. Kataria A, Kumar D, Gupta G. Donor-derived Cell-free DNA in Solid-organ Transplant Diagnostics: Indications, Limitations, and Future Directions. Transplantation 2021;105(6):1203-1211 View Article PubMed/NCBI
  62. Leonard MO, Kieran NE, Howell K, Burne MJ, Varadarajan R, Dhakshinamoorthy S, et al. Reoxygenation-specific activation of the antioxidant transcription factor Nrf2 mediates cytoprotective gene expression in ischemia-reperfusion injury. FASEB J 2006;20(14):2624-2626 View Article PubMed/NCBI
  63. Lu Y, Hippen KL, Lemire AL, Gu J, Wang W, Ni X, et al. miR-146b antagomir-treated human Tregs acquire increased GVHD inhibitory potency. Blood 2016;128(10):1424-1435 View Article PubMed/NCBI
  64. Ling Q, Xie H, Lu D, Wei X, Gao F, Zhou L, et al. Association between donor and recipient TCF7L2 gene polymorphisms and the risk of new-onset diabetes mellitus after liver transplantation in a Han Chinese population. J Hepatol 2013;58(2):271-277 View Article PubMed/NCBI
  65. Qu X, Zheng C, Wang B, Wang F, Sun X, Gao Y, et al. Comprehensive analysis of circular RNAs from steatotic livers after ischemia and reperfusion injury by next-generation RNA sequencing. FEBS Lett 2021;595(1):99-109 View Article PubMed/NCBI
  66. Finkel T, Deng CX, Mostoslavsky R. Recent progress in the biology and physiology of sirtuins. Nature 2009;460(7255):587-591 View Article PubMed/NCBI
  67. Kageyama S, Nakamura K, Ke B, Busuttil RW, Kupiec-Weglinski JW. Serelaxin induces Notch1 signaling and alleviates hepatocellular damage in orthotopic liver transplantation. Am J Transplant 2018;18(7):1755-1763 View Article PubMed/NCBI
  68. Kurian SM, Fouraschen SM, Langfelder P, Horvath S, Shaked A, Salomon DR, et al. Genomic profiles and predictors of early allograft dysfunction after human liver transplantation. Am J Transplant 2015;15(6):1605-1614 View Article PubMed/NCBI
  69. Kornasiewicz O, Bojarczuk K, Bugajski M, Golab J, Krawczyk M. Application of a proteomic approach to identify proteins associated with primary graft non-function after liver transplantation. Int J Mol Med 2012;30(4):755-764 View Article PubMed/NCBI
  70. Huang H, Zhang X, Chen H, Feng S, Zhang C, Chen R, et al. Decoding the single-cell landscape and intercellular crosstalk in the transplanted liver: a 4-dimension mouse model. bioRxiv 2021 View Article
  71. Verhoeven CJ, Farid WR, de Jonge J, Metselaar HJ, Kazemier G, van der Laan LJ. Biomarkers to assess graft quality during conventional and machine preservation in liver transplantation. J Hepatol 2014;61(3):672-684 View Article PubMed/NCBI
  72. MacConmara M, Vagefi PA. Machine Perfusion in Liver Transplantation. Adv Surg 2021;55:175-195 View Article PubMed/NCBI
  73. Nasralla D, Coussios CC, Mergental H, Akhtar MZ, Butler AJ, Ceresa CDL, et al. A randomized trial of normothermic preservation in liver transplantation. Nature 2018;557(7703):50-56 View Article PubMed/NCBI
  74. de Meijer VE, Fujiyoshi M, Porte RJ. Ex situ machine perfusion strategies in liver transplantation. J Hepatol 2019;70(1):203-205 View Article PubMed/NCBI
  75. Marecki H, Bozorgzadeh A, Porte RJ, Leuvenink HG, Uygun K, Martins PN. Liver ex situ machine perfusion preservation: A review of the methodology and results of large animal studies and clinical trials. Liver Transpl 2017;23(5):679-695 View Article PubMed/NCBI
  76. Nickkholgh A, Nikdad M, Shafie S, Abbasi Dezfouli S, Mehrabi A, Eason JD, et al. Ex Situ Liver Machine Perfusion as an Emerging Graft Protective Strategy in Clinical Liver Transplantation: the Dawn of a New Era. Transplantation 2019;103(10):2003-2011 View Article PubMed/NCBI
  77. Pavel MC, Reyner E, Molina V, Garcia R, Ruiz A, Roque R, et al. Evolution Under Normothermic Machine Perfusion of Type 2 Donation After Cardiac Death Livers Discarded as Nontransplantable. J Surg Res 2019;235:383-394 View Article PubMed/NCBI
  78. Matton APM, de Vries Y, Burlage LC, van Rijn R, Fujiyoshi M, de Meijer VE, et al. Biliary Bicarbonate, pH, and Glucose Are Suitable Biomarkers of Biliary Viability During Ex Situ Normothermic Machine Perfusion of Human Donor Livers. Transplantation 2019;103(7):1405-1413 View Article PubMed/NCBI
  79. Verhoeven CJ, Farid WR, de Ruiter PE, Hansen BE, Roest HP, de Jonge J, et al. MicroRNA profiles in graft preservation solution are predictive of ischemic-type biliary lesions after liver transplantation. J Hepatol 2013;59(6):1231-1238 View Article PubMed/NCBI
  80. Selten JW, Verhoeven CJ, Heedfeld V, Roest HP, de Jonge J, Pirenne J, et al. The release of microRNA-122 during liver preservation is associated with early allograft dysfunction and graft survival after transplantation. Liver Transpl 2017;23(7):946-956 View Article PubMed/NCBI
  81. Muller X, Schlegel A, Kron P, Eshmuminov D, Würdinger M, Meierhofer D, et al. Novel Real-time Prediction of Liver Graft Function During Hypothermic Oxygenated Machine Perfusion Before Liver Transplantation. Ann Surg 2019;270(5):783-790 View Article PubMed/NCBI
  82. Karangwa SA, Burlage LC, Adelmeijer J, Karimian N, Westerkamp AC, Matton AP, et al. Activation of Fibrinolysis, But Not Coagulation, During End-Ischemic Ex Situ Normothermic Machine Perfusion of Human Donor Livers. Transplantation 2017;101(2):e42-e48 View Article PubMed/NCBI
  83. Verhelst X, Geerts A, Jochmans I, Vanderschaeghe D, Paradissis A, Vanlander A, et al. Glycome Patterns of Perfusate in Livers Before Transplantation Associate With Primary Nonfunction. Gastroenterology 2018;154(5):1361-1368 View Article PubMed/NCBI
  84. Wang L, Thompson E, Bates L, Pither TL, Hosgood SA, Nicholson ML, et al. Flavin Mononucleotide as a Biomarker of Organ Quality-A Pilot Study. Transplant Direct 2020;6(9):e600 View Article PubMed/NCBI
  85. Jiang N, Zhang X, Zheng X, Chen D, Zhang Y, Siu LK, et al. Targeted gene silencing of TLR4 using liposomal nanoparticles for preventing liver ischemia reperfusion injury. Am J Transplant 2011;11(9):1835-1844 View Article PubMed/NCBI
  86. Zhao G, Fu C, Wang L, Zhu L, Yan Y, Xiang Y, et al. Down-regulation of nuclear HMGB1 reduces ischemia-induced HMGB1 translocation and release and protects against liver ischemia-reperfusion injury. Sci Rep 2017;7:46272 View Article PubMed/NCBI
  87. Gillooly AR, Perry J, Martins PN. First Report of siRNA Uptake (for RNA Interference) During Ex Vivo Hypothermic and Normothermic Liver Machine Perfusion. Transplantation 2019;103(3):e56-e57 View Article PubMed/NCBI
  88. Goldaracena N, Spetzler VN, Echeverri J, Kaths JM, Cherepanov V, Persson R, et al. Inducing Hepatitis C Virus Resistance After Pig Liver Transplantation-A Proof of Concept of Liver Graft Modification Using Warm Ex Vivo Perfusion. Am J Transplant 2017;17(4):970-978 View Article PubMed/NCBI
  89. Peng Y, Ye Y, Jia J, He Y, Yang Z, Zhu X, et al. Galectin-1-induced tolerogenic dendritic cells combined with apoptotic lymphocytes prolong liver allograft survival. Int Immunopharmacol 2018;65:470-482 View Article PubMed/NCBI
  90. Sánchez-Fueyo A, Whitehouse G, Grageda N, Cramp ME, Lim TY, Romano M, et al. Applicability, safety, and biological activity of regulatory T cell therapy in liver transplantation. Am J Transplant 2020;20(4):1125-1136 View Article PubMed/NCBI
  91. Shi M, Liu Z, Wang Y, Xu R, Sun Y, Zhang M, et al. A Pilot Study of Mesenchymal Stem Cell Therapy for Acute Liver Allograft Rejection. Stem Cells Transl Med 2017;6(12):2053-2061 View Article PubMed/NCBI
  92. Verstegen MMA, Mezzanotte L, Ridwan RY, Wang K, de Haan J, Schurink IJ, et al. First Report on Ex Vivo Delivery of Paracrine Active Human Mesenchymal Stromal Cells to Liver Grafts During Machine Perfusion. Transplantation 2020;104(1):e5-e7 View Article PubMed/NCBI
  93. Zheng L, Li Z, Ling W, Zhu D, Feng Z, Kong L. Exosomes Derived from Dendritic Cells Attenuate Liver Injury by Modulating the Balance of Treg and Th17 Cells After Ischemia Reperfusion. Cell Physiol Biochem 2018;46(2):740-756 View Article PubMed/NCBI
  94. Chen L, Huang H, Zhang W, Ding F, Fan Z, Zeng Z. Exosomes Derived From T Regulatory Cells Suppress CD8+ Cytotoxic T Lymphocyte Proliferation and Prolong Liver Allograft Survival. Med Sci Monit 2019;25:4877-4884 View Article PubMed/NCBI
  95. Rigo F, De Stefano N, Navarro-Tableros V, David E, Rizza G, Catalano G, et al. Extracellular Vesicles from Human Liver Stem Cells Reduce Injury in an Ex Vivo Normothermic Hypoxic Rat Liver Perfusion Model. Transplantation 2018;102(5):e205-e210 View Article PubMed/NCBI
  96. Goldaracena N, Echeverri J, Spetzler VN, Kaths JM, Barbas AS, Louis KS, et al. Anti-inflammatory signaling during ex vivo liver perfusion improves the preservation of pig liver grafts before transplantation. Liver Transpl 2016;22(11):1573-1583 View Article PubMed/NCBI
  97. Yu Y, Cheng Y, Pan Q, Zhang YJ, Jia DG, Liu YF. Effect of the Selective NLRP3 Inflammasome Inhibitor mcc950 on Transplantation Outcome in a Pig Liver Transplantation Model With Organs From Donors After Circulatory Death Preserved by Hypothermic Machine Perfusion. Transplantation 2019;103(2):353-362 View Article PubMed/NCBI
  98. Maida K, Akamatsu Y, Hara Y, Tokodai K, Miyagi S, Kashiwadate T, et al. Short Oxygenated Warm Perfusion With Prostaglandin E1 Administration Before Cold Preservation as a Novel Resuscitation Method for Liver Grafts From Donors After Cardiac Death in a Rat In Vivo Model. Transplantation 2016;100(5):1052-1058 View Article PubMed/NCBI
  99. Nassar A, Liu Q, Farias K, D’Amico G, Buccini L, Urcuyo D, et al. Role of vasodilation during normothermic machine perfusion of DCD porcine livers. Int J Artif Organs 2014;37(2):165-172 View Article PubMed/NCBI
  100. Echeverri J, Goldaracena N, Kaths JM, Linares I, Roizales R, Kollmann D, et al. Comparison of BQ123, Epoprostenol, and Verapamil as Vasodilators During Normothermic Ex Vivo Liver Machine Perfusion. Transplantation 2018;102(4):601-608 View Article PubMed/NCBI
  101. Nagrath D, Xu H, Tanimura Y, Zuo R, Berthiaume F, Avila M, et al. Metabolic preconditioning of donor organs: defatting fatty livers by normothermic perfusion ex vivo. Metab Eng 2009;11(4-5):274-283 View Article PubMed/NCBI
  102. Boteon YL, Attard J, Boteon APCS, Wallace L, Reynolds G, Hubscher S, et al. Manipulation of Lipid Metabolism During Normothermic Machine Perfusion: Effect of Defatting Therapies on Donor Liver Functional Recovery. Liver Transpl 2019;25(7):1007-1022 View Article PubMed/NCBI
  103. Carlson K, Kink J, Hematti P, Al-Adra DP. Extracellular Vesicles as a Novel Therapeutic Option in Liver Transplantation. Liver Transpl 2020;26(11):1522-1531 View Article PubMed/NCBI
  104. Yang B, Duan W, Wei L, Zhao Y, Han Z, Wang J, et al. Bone Marrow Mesenchymal Stem Cell-Derived Hepatocyte-Like Cell Exosomes Reduce Hepatic Ischemia/Reperfusion Injury by Enhancing Autophagy. Stem Cells Dev 2020;29(6):372-379 View Article PubMed/NCBI
  105. Zhang L, Song Y, Chen L, Li D, Feng H, Lu Z, et al. MiR-20a-containing exosomes from umbilical cord mesenchymal stem cells alleviates liver ischemia/reperfusion injury. J Cell Physiol 2020;235(4):3698-3710 View Article PubMed/NCBI
  106. Haga H, Yan IK, Borrelli DA, Matsuda A, Parasramka M, Shukla N, et al. Extracellular vesicles from bone marrow-derived mesenchymal stem cells protect against murine hepatic ischemia/reperfusion injury. Liver Transpl 2017;23(6):791-803 View Article PubMed/NCBI
  107. Yao J, Zheng J, Cai J, Zeng K, Zhou C, Zhang J, et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J 2019;33(2):1695-1710 View Article PubMed/NCBI
  108. Nong K, Wang W, Niu X, Hu B, Ma C, Bai Y, et al. Hepatoprotective effect of exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells against hepatic ischemia-reperfusion injury in rats. Cytotherapy 2016;18(12):1548-1559 View Article PubMed/NCBI
  109. Anger F, Camara M, Ellinger E, Germer CT, Schlegel N, Otto C, et al. Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Liver Regeneration After Ischemia Reperfusion Injury in Mice. Stem Cells Dev 2019;28(21):1451-1462 View Article PubMed/NCBI
  110. Dar WA, Sullivan E, Bynon JS, Eltzschig H, Ju C. Ischaemia reperfusion injury in liver transplantation: Cellular and molecular mechanisms. Liver Int 2019;39(5):788-801 View Article PubMed/NCBI
  111. Nigmet Y, Hata K, Tamaki I, Okamura Y, Tsuruyama T, Miyauchi H, et al. Human Atrial Natriuretic Peptide in Cold Storage of Donation After Circulatory Death Rat Livers: An Old but New Agent for Protecting Vascular Endothelia?. Transplantation 2019;103(3):512-521 View Article PubMed/NCBI
  112. Shimada S, Fukai M, Shibata K, Sakamoto S, Wakayama K, Ishikawa T, et al. Heavy Water (D2O) Containing Preservation Solution Reduces Hepatic Cold Preservation and Reperfusion Injury in an Isolated Perfused Rat Liver (IPRL) Model. J Clin Med 2019;8(11):E1818 View Article PubMed/NCBI
  113. Alix P, Val-Laillet D, Turlin B, Ben Mosbah I, Burel A, Bobillier E, et al. Adding the oxygen carrier M101 to a cold-storage solution could be an alternative to HOPE for liver graft preservation. JHEP Rep 2020;2(4):100119 View Article PubMed/NCBI
  114. Gassner JMGV, Nösser M, Moosburner S, Horner R, Tang P, Wegener L, et al. Improvement of Normothermic Ex Vivo Machine Perfusion of Rat Liver Grafts by Dialysis and Kupffer Cell Inhibition With Glycine. Liver Transpl 2019;25(2):275-287 View Article PubMed/NCBI
  115. Boehnert MU, Hilbig H, Armbruster FP. Relaxin as an additional protective substance in preserving and reperfusion solution for liver transplantation, shown in a model of isolated perfused rat liver. Ann N Y Acad Sci 2005;1041:434-440 View Article PubMed/NCBI
  116. Panisello Rosello A, Teixeira da Silva R, Castro C, G Bardallo R, Calvo M, Folch-Puy E, et al. Polyethylene Glycol 35 as a Perfusate Additive for Mitochondrial and Glycocalyx Protection in HOPE Liver Preservation. Int J Mol Sci 2020;21(16):E5703 View Article PubMed/NCBI
  117. Bardhi E, McDaniels J, Rousselle T, Maluf DG, Mas VR. Nucleic acid biomarkers to assess graft injury after liver transplantation. JHEP Rep 2022;4(3):100439 View Article PubMed/NCBI
  118. Watson CJE, Kosmoliaptsis V, Pley C, Randle L, Fear C, Crick K, et al. Observations on the ex situ perfusion of livers for transplantation. Am J Transplant 2018;18(8):2005-2020 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Assessing Donor Liver Quality and Restoring Graft Function in the Era of Extended Criteria Donors

Yimou Lin, Haitao Huang, Lifeng Chen, Ruihan Chen, Jimin Liu, Shusen Zheng, Qi Ling
  • Reset Zoom
  • Download TIFF