Skip to main content
JHEP Reports logoLink to JHEP Reports
. 2022 Jan 26;4(3):100439. doi: 10.1016/j.jhepr.2022.100439

Nucleic acid biomarkers to assess graft injury after liver transplantation

Elissa Bardhi 1, Jennifer McDaniels 1, Thomas Rousselle 1, Daniel G Maluf 1,2, Valeria R Mas 1,
PMCID: PMC8856989  PMID: 35243279

Summary

Many risk factors and complications impact the success of liver transplantation, such as ischaemia-reperfusion injury, acute rejection, and primary graft dysfunction. Molecular biomarkers have the potential to accurately diagnose, predict, and monitor injury progression or organ failure. There is a critical opportunity for reliable and non-invasive biomarkers to reduce the organ shortage by enabling i) the assessment of donor organ quality, ii) the monitoring of short- and long-term graft function, and iii) the prediction of acute and chronic disease development. To date, no established molecular biomarkers have been used to guide clinical decision-making in transplantation. In this review, we outline the recent advances in cell-free nucleic acid biomarkers for monitoring graft injury in liver transplant recipients. Prior work in this area can be divided into two categories: biomarker discovery and validation studies. Circulating nucleic acids (CNAs) can be found in the extracellular environment pertaining to different biological fluids such as bile, blood, urine, and perfusate. CNAs that are packaged into extracellular vesicles may facilitate intercellular and interorgan communication. Thus, decoding their biological function, cellular origins and molecular composition is imperative for diagnosing causes of graft injury, guiding immunosuppression and improving overall patient survival. Herein, we discuss the most promising molecular biomarkers, their state of development, and the critical aspects of study design in biomarker research for early detection of post-transplant liver injury. Future advances in biomarker studies are expected to personalise post-transplant therapy, leading to improved patient care and outcomes.

Keywords: liver transplantation, biomarkers, molecular diagnostics, transcriptomics, microRNAs, extracellular vesicles, donor-derived DNA, personalized therapy

Abbreviations: ACR, acute cellular rejection; CIT, cold ischaemia time; CNA(s), circulating nucleic acid; dd-cfDNA, donor-derived cell-free DNA; ddPCR, digital droplet PCR; EV(s), extracellular vesicles; HCC, hepatocellular carcinoma; HLA, human leukocyte antigen; LFTs, liver function tests; LT, liver transplantation; miRNA, microRNA; RT-qPCR, reverse-transcription quantitative PCR; TCMR, T cell-mediated rejection


Key points.

  • Molecular evaluation of biofluids (i.e., blood, urine, perfusate, and bile) are providing new opportunities to understand how specific perturbations in nucleic acids can signal the early stages of disease.

  • Circulating nucleic acid (CNA) biomarkers (i.e., cell-free RNA/DNA) confer significantly more information than current prognostic and diagnostic methods, emerging as early and sensitive measures of liver injury and function.

  • Extracellular vesicles (EVs) carry a heterogeneous composition of RNAs, DNAs, lipids, and proteins, and have been found to protect molecular biomarkers from degradation in the extracellular environment.

  • Advances in EV analytic approaches have enabled the investigation of donor-specific EVs in recipient biofluids, demonstrating the capacity for these structures to facilitate intercellular and interorgan communication following liver transplantation.

  • Circulating miRNAs provide information on causal transcriptional processes occurring at a molecular level, leading to the identification of dysfunctional canonical pathways and novel therapeutic targets.

  • Donor-derived cell-free DNA (dd-cfDNA) is released from necrotic or apoptotic cells in the transplanted organ and may consequently be useful as an early predictor of post-transplant injury or rejection.

  • Large multicentre clinical trials have generated vast amounts of data and information at various molecular levels, demonstrating a promising opportunity for cell-free biomarkers to be introduced into clinical care.

Introduction

The field of liver transplantation (LT) has experienced great advances over the last few decades, largely due to the development of novel immunosuppressive drugs, technical refinements in surgery, and effective postoperative management of transplant recipients. Overall, adult and paediatric liver transplant rates continue to increase at a significant rate, reaching nearly 9,000 liver transplants performed in the US alone each year. This represents a 40.8% increase over the past decade as reported by the 2019 Annual OPTN/SRTR Data.1 Such progress is accompanied by increased rigor in translational and clinical research efforts, resulting in significant improvements in patient outcomes. For instance, with the recent advent of new antiviral treatments, recurrence of hepatitis C after transplantation is no longer the leading cause of graft loss.1,2 Nonetheless, long-term allograft failure continues to occur, owing to both immunologic and non-immunologic factors, with immunosuppressive toxicity contributing to patient morbidity and mortality.

Finding the right balance between under- and over-immunosuppression remains a holy grail in transplantation. Currently, core biopsies remain the “gold standard” for the diagnosis of post-transplant abnormalities despite increased concerns regarding interobserver variability between histological evaluations.3,4 Considering the large physical size and high complexity of the liver, core biopsies only capture a small fraction of a single lobe, making up less than 0.002% of the total mass and often leading to inaccurate diagnoses.5,6 In the absence of reliable tools to measure immunosuppressive burden, clinicians also depend on non-invasive measurements of traditional, non-specific laboratory biomarkers (liver function tests [LFTs]) to guide immunosuppression dosage.5,7 Thus, there remains a significant need for specific, non-invasive biomarkers that can accurately determine the degree and cause of liver injury post-LT. Recently, cell-free molecular biomarkers have emerged as potential early and sensitive markers of tissue injury and function.

Multi-omics approach to identifying biomarker signatures

The prognostic and diagnostic applications of molecular biomarkers have been widely studied throughout the transplant community. High-throughput technologies allow for the generation of large amounts of data and information at different molecular levels.8 Recent studies using multi-omics techniques have identified unique molecular signatures that confer significantly more information than the microscopy and staining measures currently used for medical diagnostics.[9], [10], [11], [12], [13], [14], [15] Such advanced technology paired with extensive patient databases has allowed for the discovery of a diverse set of biomarkers, shifting the focus away from a single marker to a combination of various biomarkers. Because of the complexity of the aetiology of post-transplant liver injury, prognostic and diagnostic tests will likely need to integrate multiple layers of biological information (e.g., transcriptomics, epigenetics, proteomics) in order to be both sensitive and specific (especially in the presence of coexisting conditions). By definition, biomarkers are measurable and reproducible objective indicators of medical state, observed externally from the patient, which can give information on normal biological mechanisms, pathogenic processes, or responses to an exposure or intervention.16

Common molecular biomarkers include nucleic acids (e.g., microRNAs [miRNAs] and cell-free DNA), epigenetic patterns (e.g., DNA methylation and histone modifications), and the more traditional protein-based markers (e.g., glycoproteins, antigens, and antibodies) that correspond to disease progression and severity. Less common markers may include proteomic and metabolomic biomarkers, which are explored using multi-analyte technologies that assess the complete set of proteins or metabolites of a biological sample.9 However, findings from proteomic and metabolomic biomarker discovery studies have not been readily transferred to clinical trials due to their high complexity, relative novelty, and the intricate analytical techniques required to interpret such data.17,18 Therefore, this review will focus on the biomarkers that have been described using an important number of clinical samples and that are thus more likely to be transferred to the clinical setting promptly. Principal applications of these biomarkers in transplantation include: i) predicting the progression or development of disease, ii) diagnosing specific post-transplant conditions, iii) assessing the severity or extent of graft injury, and iv) monitoring the response to therapeutic interventions such as immunosuppressants and steroids.19 Overall, this article describes the recent scientific advances in the discovery and implementation of circulating molecular biomarkers in LT.

Extracellular nucleic acid markers

Since the discovery of cell-free nucleic acids by Mandel in 1948, researchers have demonstrated that nucleic acids in body fluids have the potential to detect, track, and discriminate between patients with and without disease.20 Circulating biomarkers offer a viable non-invasive approach to early disease diagnosis. While most nucleic acids are located within cells, a small quantity termed “circulating nucleic acids” (CNAs) can be found in the extracellular environment. In the more studied cancer field, researchers have found traces of tumoural-shed nucleic acids in the blood, urine, and stool.21,22 The information stored within CNAs has various benefits related to cancer detection and treatment. These circulating biomarkers offer additional information regarding methylation or transcriptional and translational status via non-coding RNA, which may be suitable for the early detection of cancer.23

Despite the robust presence of CNA in biological fluids, the origin and function of these biomarkers in the extracellular environment remain poorly understood. Given the high levels of nucleases within circulating biofluids, it has been demonstrated that nucleic acids packaged into membranous structures are protected from immediate degradation upon release.24 These structures often contain a heterogenous composition of RNAs, DNAs, lipids, and proteins, which vary depending on cellular origin and physiological state.25 In the last decade, a novel method of cell-to-cell communication mediated by extracellular vesicles (EVs) has emerged. These 0.05–1 μm membrane-bound vesicles are involved in normal physiological processes, as well as being implicated in pathological conditions.26 They are released by cells into the bloodstream and other bodily fluids, shuttling bioactive molecules from their cell of origin to recipient cells via phagocytic or non-phagocytic pathways and delivering their contents directly into the cytoplasm of target cells.27 This emerging field of interest has spurred a plethora of EV studies that employ high-throughput technologies including proteomics, transcriptomics, and lipidomics.28

Recent studies have demonstrated that EVs may facilitate intercellular and interorgan communication.[29], [30], [31], [32] It has also been suggested that through the internalisation of EVs, bioactive contents such as miRNAs can alter gene expression and mediate a physiological response in target cells. Thus, decoding the composition, biogenesis, and biological functions of EV cargo can further elucidate cellular crosstalk within transplant organs. Such information can be combined with physiological data to elucidate the cellular pathways responsible for differential liver graft outcomes. Fig. 1 depicts EVs and their pertinent molecular contents, which can function as liver-specific biomarkers.

Fig. 1.

Fig. 1

EVs released from graft tissue function as well-protected messenger complexes that allow for the transfer of lipids, proteins, and nucleic acids to neighbouring or distant targets.

By carrying biomolecules from tissues to biofluids, EVs may serve as non-invasive sources of clinical biomarkers. Characterising the cellular sources using surface markers can provide biological information about the functional state of liver-specific parent cells. dsDNA, donor-specific DNA; EV, extracellular vesicle; miRNA, microRNA.

The pursuit of non-invasive biomarkers of allograft rejection has led to the evaluation of EVs in a range of biofluids. Specifically, using bulk analyses to evaluate EV cargo, researchers have been able to identify markers of varying specificity, sensitivity, and usefulness. Recently, EVs bearing donor-human lymphocyte antigen (HLA) have garnered interest as potential biomarkers of allograft function. Mastoridis et al. used ImageStream flow cytometry to detect and characterise circulating organ-specific EVs.33 The EVs from 3 liver transplant recipients were labelled with a pan-EV marker (CFSE [carboxyfluorescein diacetate succinimidyl ester]), a bona fide marker of exosomes (CD63), and probes for donor and recipient HLA (such as HLA-B27 and HLA-B8). The investigators confirmed that donor-specific EVs were detectable in the circulation after LT. Further multiparametric analyses were employed to interrogate donor EV cargo for co-stimulatory/inhibitory molecules, thereby providing additional support for the application’s potential to characterise disease and impart functional insights. Recently, the same group employed advanced imaging flow cytometry to explore the kinetics of EV release and the extent to which donor EVs might induce cross-dressing (a phenomenon where recipient cells display membrane proteins from the transplant donor) following LT.34 Typically, the cross-dressing process occurs after organ transplantation when donor leukocytes and/or parenchymal cells release vast amounts of small EVs, including exosomes (30–150 nm in diameter).34 These EVs are taken up and displayed as patches on the surface of host dendritic cells, causing alloreactive T cell stimulation or tolerization. The authors conclude that cross-dressing can be transiently detected in the circulation shortly after LT in association with the release of donor-HLA EVs. In LT recipients, most circulating cells exhibiting donor-HLAs are indeed cross-dressed cells and not passenger leukocytes.34

Such advances in EV analytic approaches have enabled the investigation of donor-specific allograft-derived EV release following LT and demonstrated the capacity for these to cross-dress recipient cells through the transfer of donor major histocompatibility complexes. Given the presence of cross-dressed cells in experimental and clinical transplantation and the recognised impact of these cells on the overall alloresponse, these pathways could be critical for the design of tolerance-promoting protocols.35

In addition to functioning as “biomarker carriers,” EVs may hold therapeutic potential based on their drug delivery capabilities and regenerative properties.36 A recent report showed human liver stem cell-derived EVs can reduce liver injury and promote regeneration during normothermic machine perfusion in a donation after circulatory death donor model with prolonged warm ischaemia.37 Further identification of the cellular origins of EVs (i.e., stellate cells, hepatocytes, endothelial cells) and their intended recipient cells will be critical for targeted interventions.

MiRNAs as markers of injury

Ongoing investigations into the transcriptomic profiles associated with biological fluids have revealed the potential for non-coding RNAs to serve as biomarkers of graft injury. Non-coding RNAs (e.g., miRNAs, long non-coding RNAs, transfer RNAs, small-interfering RNAs) represent specific, sensitive, and stable markers of liver injury. MiRNAs have garnered heightened interest because of their upstream roles in modulating transcriptional programmes and orchestrating both physiological and pathological processes. Given the regulatory role of miRNAs in mediating gene expression on the post-transcriptional level, it is not surprising that physiological and pathological changes can induce alterations in the circulating miRNA expression profiles. MiRNA sequences are highly conserved across species, allowing for tissue-specificity and easy detection through reverse-transcription quantitative PCR (RT-qPCR).38 Additionally, circulating miRNAs are rapidly released from cells in response to hypoxic stress and injury, and can enable real-time assessments of the dynamic conditions related to transplant injury.39,40 It has also been demonstrated that critical facets of adult tissue repair mechanisms are subject to control by miRNAs.41 The release of shedding vesicles containing miRNA is known to be highly regulated and depends on the activation state of the source cells.26 These features establish miRNAs as ideal biomarkers for a variety of clinical applications. Circulating miRNAs’ potential as sensitive markers of liver injury has been reported in many studies. Table 1 summarises the differentially expressed miRNAs found to be associated with acute rejection, HCV, alcohol-related liver disease, non-alcoholic fatty liver disease, hepatocellular carcinoma (HCC), or drug-mediated hepatoxicity.[42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79] Of note, miR-122, miR-34a, miR-223, miR-192, miR-885-5p, miR-155, and miR-21 were related to 3 or more of these conditions, demonstrating their broad potential to assess multiple forms of liver injury. Mir-122 is the only miRNA that was found to be associated with each of these conditions.[42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53]

Table 1.

miRNAs associated with various post-transplant liver conditions.[42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79]

graphic file with name fx1.jpg

As one of the earliest examples of tissue-specific miRNAs, miR-122 is of heightened interest as a marker of liver injury.80,81 Upon its discovery, investigators found that miR-122 accounted for 72% of all miRNAs cloned from the livers of mice.82 Other miRNAs such as miR-21, miR-29, miR-192, and miR-194 are also highly enriched in the liver and may function as specific markers of post-transplant injury.[83], [84], [85], [86], [87]

Investigators have used clinical models to explore the quantitative relationship between miRNA expression and degree of liver injury. Changes in liver-specific miRNAs in the plasma of mice are both dose- and exposure duration-dependent.88,89 Wang et al. found that expression of miR-122 and miR-192 increased more rapidly than LFTs after drug overdose (≤1 hour), and progressively increased as the dose of acetaminophen increased.83 Similarly, Chang et al. reported that patients with non-alcoholic fatty liver disease and advanced fibrosis (grade 3-4) had significantly lower levels of serum miR-122 than those with lower grade fibrosis (grade 1-2), demonstrating an inverse relationship between the severity of fibrosis and miR-122.90 In contrast, most studies have demonstrated that elevated miR-122 serum levels correlate with hepatic injury and inflammatory activity in patients with HCV, acute rejection, alcohol-related liver disease, and non-alcoholic fatty liver disease.[42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53] These discrepancies might be due to differences in the aetiology of liver disease, thus, the relationship between miR-122 and fibrotic injury needs to be further explored.

MiRNAs can also provide information on causal transcriptional processes occurring at a molecular level, leading to the identification of dysfunctional canonical pathways and novel therapeutic targets. Of note, Shaked et al. investigated the miRNA profiles of longitudinal serum biopsies to identify early predictors of acute rejection.91 Using a prospective study design, the investigators demonstrate that a two-miRNA (miR-483-3p and miR-885-5p) combination model could predict acute cellular rejection (ACR) up to 40 days before biopsy-proven rejection. The findings of this study suggest that serum miRNAs could identify ACR without the need for a liver biopsy. Consequently, these markers could replace or, more likely, contribute to the diagnostic value of liver biopsies. Critically, immunosuppression could also be personalised for individual patients based on miRNA measurements during protocolised immunosuppression minimisation. Validated, non-invasive markers of rejection could be used to predict clinical rejection episodes, thereby providing a unique tool for physicians to optimise immunosuppression levels while avoiding ACR. In summary, these findings may give rise to more precise postoperative immunosuppression regimens, addressing the critical need to mitigate the toxic effects of excess immunosuppression, which is associated with system-wide comorbidities.

In a recent report, Ruiz et al. reported that 3 upregulated plasma miRNAs (miR-155-5p, miR-122-5p, and miR-181a-5p) can accurately diagnose T cell-mediated rejection (TCMR) with AUROCs of 0.87, 0.91, and 0.89, respectively.43 Furthermore, they report that miR-155-5p was able to discriminate all patients who presented TCMR. This study demonstrates that miRNA expression levels in plasma can differentiate TCMR from other causes of graft dysfunction and may serve as a useful tool in clinical practice. Because this is a single-centre study with a limited sample size, results should be interpreted cautiously and must be validated in a larger patient population. In addition, only a few miRNAs were examined, and the methodologies used for their analysis vary from other works.

Similarly, Muthukumar et al. reported a miRNA signature in serum that can diagnose and differentiate acute rejection from other forms of liver injury.42 The group first identified a panel of 9 miRNAs that is diagnostic of acute rejection of human liver when compared to a cohort without signs of rejection. To reduce confounding variables this analysis was limited to patients who received transplants for non-immunological and non-viral liver diseases. Remarkably, the authors found that a diagnostic model using a combination of miR-210 and miR-34a grouped all the patients correctly. These results suggest that serum miRNAs could identify acute rejection without the need for a liver biopsy. The investigators then tested the specificity of these serum markers by comparing the acute rejection cohort to recurrent HCV samples. While HCV rarely poses an ongoing threat to post-transplant outcomes, the investigators still demonstrate the ability of circulating miRNAs to successfully differentiate liver injury caused by different aetiologies. These miRNA signatures can be detected significantly earlier than the current established diagnostic methods, allowing for more timely intervention and guided treatment. Ultimately, these studies support the utility of cell-free miRNA as diagnostic non-invasive biomarkers and also demonstrate the applicability of new bioinformatics tools (i.e., logistic regression models and AUROCs) in the discovery of biomarkers.

Furthermore, as the study of machine perfusion accelerates, there is an even greater demand to identify and validate markers of liver viability and functionality during perfusion. Perfusate is an especially advantageous choice as a source of biomarkers for 3 critical reasons: i) there is a large volume readily available, ii) collection is non-invasive, and iii) it can be rapidly analysed while the liver is awaiting transplantation and/or being perfused. Therefore, as scientists promptly work to extend the limits of cold ischaemia time (CIT), there is a growing window of opportunity in which graft quality can be non-invasively examined in real-time. During CIT the liver is under significant hypoxic stress and releases uniquely packed EVs in response to extreme conditions. Few groups have explored the prognostic potential of circulating miRNAs released during CIT. Using advanced molecular techniques, it takes about 1 hour to isolate RNA, 1 hour for cDNA conversion, and 1 hour for qPCR set up and analysis. Given the CIT is 6-9 hours for most of the liver donors (range: 5-13 hours), there is ample time to collect perfusate and run prognostic tests while the patient awaits implantation.92 Selten et al. set out to determine whether miRNA expression in perfusate correlated with early allograft dysfunction after LT.93 The investigators found that hepatocyte-specific miR-122 and the miR-122/miR-222 ratios in perfusate served as potential biomarkers of early allograft dysfunction. The two-miRNA ratio model may aid in the development of therapies during graft preservation, thereby reducing post-transplant complications. This work demonstrates the maladaptive physiological and pathological processes that may begin manifesting in the donor organ under ischaemic conditions prior to transplantation. Such findings can be used to assemble a panel of miRNAs that can be tested at pre-implantation and may predict future graft function and outcomes. Decoding these early signals that are constitutively released into the perfusate may also allow for a better understanding of the role of cell-to-cell communication in the propagation of graft injury.

While the use of miRNA biomarkers holds great promise, the identification of robust controls for data normalisation remains a challenge in miRNA biomarker research, and the use of an exogenous “spike-in” control during RNA isolation remains essential. During RNA extraction, exogenous oligonucleotides (e.g., cel-miR-39, cel-miR-54, cel-miR-238) are added at known concentrations in order to assess RNA yield, signal the presence of nucleases, and monitor reverse-transcription efficiency.94 After conducting qPCR, spike-in expression values between patient samples can be compared, and outlier samples may be identified and considered for exclusion from further data analysis. In a recent publication, Roest et al. systematically investigated the challenges associated with the isolation of RNA from a range of human biofluids related to LT, including serum, urine, bile, and perfusate.95 Using 4 different isolation techniques and complete sample sets from matched patients, the investigators demonstrate i) the large variability in RNA yield between biological samples, ii) the significant contamination of anticoagulant RT-qPCR inhibitors detected in perfusate samples, and iii) the benefits of using a spike-in control to determine RNA loss during workup. However, the investigators also warn against using spike-in controls to normalise qPCR data, instead confirming that it serves as a reliable indicator of RNA yield.95 Still, investigators are left without a set of standards for data normalisation in miRNA discovery research. There are limited consistent “housekeeping” miRNAs in human biofluids, and the heterogeneity of these samples makes the identification of one increasingly challenging.96 Recently, Faraldi et al. investigated the efficiency of 4 different normalisation strategies for analysing circulating miRNAs, including i) the global mean of miRNA expression, ii) the mean of endogenous controls (33 stable miRNAs ranked by NormFinder97 algorithm), iii) exogenous oligonucleotides (3 “spike-in” miRNAs), and iv) the single most stable endogenous miRNAs.94 The investigators found that global, endogenous, and single-miRNA methods performed similarly in reducing the technical variability among their experimental replicates. Interestingly, they report that exogenous spike-in normalisation methods (using both individual and mean Ct values) generated marked differences in the fold-change expression levels (opposite trend), suggesting that this strategy may lead to misinterpretation of results.94 Other groups have also demonstrated that the relative logarithmic differences between two endogenous miRNAs can be used to create “self-normalising miRNA pairs,” which circumvent the need for a reference miRNA in circulating biofluids.[98], [99], [100] Currently, it is clear that different normalisation methods significantly influence experimental outcomes and thus, the lack of an accepted standardised control strongly limits the clinical applicability of miRNA biomarkers.

Beyond identifying expression patterns that are significantly associated with graft survival and recurrent disease, miRNAs can be used to explore the molecular mechanisms and specific cell types involved in mediating post-transplant injury. Technologies based on immunoaffinity-capture surfaces and beads, such as flow cytometry and ELISA, have enabled the characterisation of EV populations expressing cell-specific antigens.101,102 Thus, liver-specific EV populations can be studied for their protein (e.g., western blotting), lipid (e.g., mass spectrometry), and nucleic acid (e.g., RT-qPCR, DNA, and RNA sequencing) content, allowing for the continued study of novel circulating molecular markers.102

However, it is important to note that miRNAs have a variate distribution in biofluids, which may subsequently affect findings. Some are known to be associated with proteins, others with vesicles, and others with both. For example, in alcohol-related liver disease and inflammatory liver injury, miR-122 and miR-155 are mainly associated with EVs, whereas in drug-induced liver injury, these miRNAs predominate in protein-rich fractions.18 Such variations are important to consider when investigating the origin, function, and target of pertinent miRNA markers from differing biological fluids.62

Lastly, it may be useful to discern the mechanisms and pathways by which master miRNA regulators drive liver injury by quantifying the expression of novel downstream targets. Studying the molecular pathways and biological functions associated with significant miRNAs could lead to translational and therapeutic advances in LT. In future work, gain- and loss-of-function approaches could be used to target top miRNA candidates and thereby to identify significant molecular interactions that orchestrate liver injury in clinical models.103,104

Cell-free DNA as a marker of graft injury

In the current era of immunosuppression, acute rejection rates in LT remain acceptably low, while immunosuppressant toxicity continues to have severe consequences on long-term outcomes. Thus, a non-invasive marker to personalise immunosuppression remains a critical need. The transplant field is in need of biomarkers that are practical, cost-effective, and reproducible, with a rapid (same-day) turnaround time. Analytic performance shows high specificity and sensitivity for graft complications, and such biomarkers should have diagnostic or prognostic utility at the earliest disease stages. Donor-derived cell-free DNA (dd-cfDNA) is a promising new biomarker for the detection of graft injury.92 One of the earliest studies concluded that plasma dd-cfDNA levels can function as markers of cell death, released from necrotic or apoptotic cells in the transplanted organ, and may consequently be useful in predicting rejection.105

There are various approaches for detecting dd-cfDNA; for instance, the use of preselected single nucleotide polymorphisms and high-throughput sequencing for readouts have been reported.[94], [95], [96] Currently, the most common methods include shotgun sequencing or droplet digital PCR (ddPCR). This data is expressed either as GcfDNA percentage (graft cfDNA/total cfDNA) or absolute quantification in copies/millilitre.104,106,107 In a recent multicentre study, the detection of dd-cfDNA in plasma by ddPCR allowed for earlier and more sensitive discrimination of acute rejection compared with conventional LFTs.108 Specifically, LFTs and plasma dd-cfDNA were both monitored in 115 adult liver transplant recipients at 3 different transplant centres as part of a prospective, observational, multicentre cohort trial. Multivariable logistic regression modelling demonstrated that GcfDNA provided additional information on graft integrity beyond LFT measurements. Diagnostic sensitivity and specificity were 90.3% (95% CI 74.2%-98.0%) and 92.9% (95% CI 89.3%-95.6%), respectively, for dd-cfDNA at a threshold value of 10%.

Additional studies using different biomarker detection methods have explored the potential of dd-cfDNA biomarkers. Using probe-based ddPCR, Beck et al. have demonstrated that the plasma dd-cfDNA fraction was between 5%-10% at day 10 post-transplant in stable liver transplant recipients, whereas in cases of rejection it reached approximately 20% and gradually increased to 55–60%.103 Macher et al., who instead quantified cfDNA by RT-qPCR, also showed higher total cfDNA and dd-cfDNA serum levels in patients with liver transplant injury (acute rejection, hepatic arterial and venous thrombosis, and profound cholestasis ending in multiple organ failure) compared to recipients with stable graft function.109 In contrast, increases in total cfDNA levels but not dd-cfDNA were observed in patients with complications that did not compromise the donated organ (biliary peritonitis and surgical wound infection).

A recent study aimed to evaluate the feasibility of measuring dd-cfDNA through short tandem repeats analysis by quantitative fluorescent-PCR and, to assess the role of the concentration and fragment size of total cfDNA as biomarkers of acute rejection.110 Short tandem repeat amplification by quantitative fluorescent-PCR may be an alternative, easily implementable, strategy for rapid dd-cfDNA quantification in clinical laboratories. The results of this pilot study indicate that dd-cfDNA increases very early, even 1-2 weeks before the diagnosis of acute rejection, so it could be useful as a prognostic biomarker to improve patient risk stratification.

In summary, these studies show that dd-cfDNA is a biomarker with promising clinical utility in LT. In cases of acute rejection, dd-cfDNA has repeatedly outperformed LFTs, allowing for the earlier and more sensitive discrimination of this complication. Currently, the main limitation associated with cfDNA is specificity. However, it has been reported that unique methylation patterns on DNA fragments can serve as fundamental determinants of cell identity. Lehmann-Werman et al. recently described a method to detect cfDNA carrying hepatocyte-specific methylation patterns.111 The authors identify 3 genomic loci that are specifically unmethylated in hepatocytes. Probing cfDNA methylation sites allows for the i) identification of the cellular origins of markers, ii) quantification of injury in specific cell types, and iii) identification of novel therapeutic targets in transplant injury.

Advancing precision medicine: the now and the future

The prognostic and diagnostic value of non-invasive biomarkers has been widely explored in LT. However, findings have not yet been translated into routine clinical use, owing to small sample sizes, and the lack of proper control groups or independent validations. The main limitations of CNA analysis include its low quantity and compromised quality (potential degradation) in biological fluids. The current challenges associated with biomarker research are summarised in Table 2. Many recent analyses are also limited to tissue-derived biomarkers, which are useful for integrative analyses but have limited implications for clinical practice. While the use of tissue miRNA expression for diagnosis is not as practical as non-invasive markers, significant miRNA discoveries in liver tissue can be used to guide future investigations in biological fluids. Within the liver, several microRNAs including miR-21, miR-221/222, and miR-181b promote liver fibrosis through the TGF-β or NF-κB pathways, whereas miR-29b, miR-101, miR-122, and miR-214-3p prevent fibrosis by inhibiting collagen synthesis or suppressing activation of the TGF-β pathway.112 Serum miR-34a was found to be upregulated in patients with fibrosis in a stage-dependent manner, and miR-571 and miR-513-3p were elevated in patients with cirrhosis.74,86 MiR-29a was downregulated in patients with fibrosis in an inversely stage-dependent manner.113 Also, while miR-122 is significantly downregulated in HCC tissue samples, many groups have reported the upregulation of circulating miR-122 in the serum of patients with HCC compared to that of controls.54,114,115 More research is needed to understand the correlation between tissue and peripheral biomarker signatures.

Table 2.

Current challenges associated with biomarker discovery research.

Challenge
Working with human samples
  • Recruitment: enrolling patients with diverse backgrounds from multiple transplant centres remains a challenge

  • Small sample size: acquiring a large number of study participants is necessary for significant findings

  • Clinical data organisation: collecting, storing, and sorting a significant volume of longitudinal patient data can be difficult to manage

  • Study timeline: retention of patients during follow-up is costly, time-consuming, and difficult

  • Variable post-transplant drug regimens: immunosuppressive treatment, dosage, and management may change over time, presenting a confounding variable that is difficult to adjust for

Study Design
  • Lack of standardised study endpoints: patient/graft survival, acute rejection episodes, biochemical data (i.e., liver enzymes), disease recurrence and development, and non-immunological injury (nephrotoxicity) must all be assessed in relation to post-transplant outcomes

  • Lack of control group: post-transplant control groups are difficult to define

  • Identification of best markers: there is no standardised method for narrowing down the top candidate markers when using high-throughput approaches

Technical challenges
  • Isolating nucleic acids: circulating nucleic acids are characterised by low concentration and high degradation rates

  • Experimental methods: utilising high throughput technologies can be costly and time-consuming

  • Normalisation methods: multiple approaches can be used to normalise cell-free nucleic acids

Building predictive models
  • Team science: biostatisticians are needed to aid in the development and implementation of statistical and mathematical methods

  • Overfitting: over-testing the training data can result in a model that appears very accurate but has memorised the key points in the data set rather than learned how to generalise, requiring an independent dataset to validate findings.

Statistical significance does not readily translate into clinical utility in biomarker research. The pipeline from discovery to clinical practice for biomarkers is complex, and independent validations of biomarker-guided practice using a large population of human patients with longitudinal follow ups are needed. Table 3 outlines the pertinent biomarker clinical trials identified in LT in chronological order (retrieved from the clinicaltrials.gov database). Of note, the Liver Immunosuppression Free Trial (LIFT) [NCT02498977] is currently studying the utility of transcriptional tissue biomarkers to guide immunosuppression withdrawals and is in phase IV of clinical testing. Such biomarkers improve the accuracy and speed by which physicians can detect post-transplant complications. While this work is undoubtedly restricted by the need for tissue biopsies, it represents an important starting point for the use of molecular diagnostics in transplantation. The LiverCare Kit [NCT04793360] utilises Streck and PAX gene tubes to assess dd-cfDNA and gene expression patterns in the blood of transplant patients, specifically monitoring clinical events related to rejection, recurrent disease, and biliary obstruction. Typically, many discovery biomarker studies utilise plasma or serum for analysis; however, the utilisation of whole blood for biomarker surveillance is more transferrable to clinical settings where physicians may not have access to large centrifuges. Thus, the reproducibility and efficacy of biomarkers need to be validated using clinically relevant biological samples.

Table 3.

Biomarkers to assess graft injury after liver transplantation.

Clinical trial title and identifier Institution Enrolment Start date End date Description
Serum Markers of Ischemia-Reperfusion Injury in Liver Transplant Patients [NCT00698399] Vanderbilt University Medical Center 20 Mar 2008 Feb 2010 Observational trial of markers (TMAO, NGAL, cystatin-C, and allantoin) in the serum of patients who are undergoing LT surgery.
Gene Expression in Liver Allograft Rejection and Recurrent Hepatitis C [NCT01428700] University of Pennsylvania 275 Aug 2011 Jan 2013 Observational study evaluating whether certain patterns of biomarkers in blood post-LT can be used to determine if the transplanted liver is being rejected or sustaining injury.
Pilot Study of Immunosuppression Drug Weaning in Liver Recipients Exhibiting Biomarkers of High Likelihood of Tolerance Hospital Clinic of Barcelona 25 Sep 2011 Jan 2013 Non-randomised prospective study in which gradual weaning of immunosuppressive drugs will be offered to LT recipients exhibiting a favourable peripheral blood gene expression profile.
Discovery and Validation of Proteogenomic Biomarker Panels in Liver Transplant Recipients [NCT01672164] Northwestern University Feinberg School of Medicine 202 Aug 2012 Dec 2015 The main focus of this study is to develop blood and/or urine tests that will help to detect early signs of rejection in LT patients.
The Relationship of Hepatobiliary microRNA Expression Profile and Clinical Outcome in Liver Transplantation [NCT02307890] University of Edinburgh 100 Aug 2014 Aug 2020 Observational study correlating miRNA levels in bile duct biopsies taken during LT with the incidence of IC following LT.
Liver Immunosuppression Free Trial (LIFT) [NCT02498977] King's College London 148 Oct 2015 Oct 2021 Prospective interventional study analysing the risk/benefit ratio of employing transcriptional biomarkers to guide immunosuppression withdrawal post-LT.
Plasmatic Factor V as a Predictor of Graft Dysfunction After Liver Transplantation [NCT03396016] University Health Network, Toronto 140 Apr 2018 Apr 2021 Observational study validating the use of coagulation cofactor Factor V as a predictive biomarker of graft function after LT.
Enteric Microbiome and Liver Transplantation [NCT03666312 IRCCS Ospedale San Raffaele 275 Sep 2018 Aug 2021 Observational study of the faecal microbiome of LT patients in combination with a large panel of clinical, lab and functional parameters correlated to different clinical outcomes.
Liquid Biopsy-based Monitoring System for Relapse of HCC After Liver Transplantation: A Multi-center and Prospective Study [NCT03708705] Zhejiang University 500 Nov 2018 Jul 2020 Prospective study aiming to develop a liquid biopsy-based biomarker system for relapse of HCC tumour post-LT using genomic and proteomic information.
Monocytic Expression of HLA-DR After Liver Transplantation (EdMonHG) [NCT03995537] Hospices Civils de Lyon 100 Feb 2020 Sep 2023 Observational study examining the expression of monocytic surface markers (HLA-DR) in blood and their association with post-LT immune dysfunction (acute cell rejection and sepsis).
Non-invasive Rapid Assessment of Patients with Liver Transplants Using Magnetic Resonance Imaging With LiverMultiScan. Leiden University Medical Centre; King's College Hospital NHS Trust 131 Aug 2020 Oct 2020 Prospective biomarker trial comparing the accuracy of a new test (LiverMultiScan) against an existing test (liver biopsy) in the assessment of LT recipients.
Role of Fecal Microbiota in Predicting Graft Rejection and Sepsis Among Recipients of Living Donor Liver Transplant in First Year. [NCT04621812] The Institute of Liver and Biliary Sciences 100 Nov 2020 Oct 2022 Observational study analysing the role of gut microflora an early biomarker for graft dysfunction and its influence on immune remodelling for the prediction of post-LT infection or rejection.
Molecular Assessment and Profiling of Liver Transplant Recipients [NCT04793360] Icahn School of Medicine at Mount Sinai 1500 May 2021 Dec 2025 Prospective observational study to assess the correlation between clinical events (e.g., rejection, recurrent disease, biliary obstruction), graft histology, dd-cfDNA levels, and gene expression profiling

dd-cfDNA, deceased donor cell-free DNA; HCC, hepatocellular carcinoma; HLA, human leukocyte antigen; IC, ischaemic cholangiopathy; LT, liver transplantation; NGAL, neutrophil gelatinase-associated lipocalin; TMAO, trimethylamine N-oxide.

Limitations in study design can also significantly delay the utilisation of molecular diagnostic markers in clinical settings. Biomarker studies can be retrospective (i.e., using previous data) or prospective (i.e., following a large number of patients). Prospective designs allow for the longitudinal collection of pre-diagnostic samples, making this the optimal study design for identifying early, prognostic markers of injury.116 Marker levels likely increase as liver injury progresses, thus, samples collected closer to the occurrence of physical symptoms will be more informative than those taken after a clinical diagnosis.

Furthermore, since current diagnostic methods and cohort groupings are often based on histological findings of core biopsies, predictive biomarkers for post-transplant conditions are limited by biopsy findings. Establishing clinically meaningful surrogate endpoints for biomarker discovery research is an ongoing challenge in LT. Compared to related fields, it is less clear which outcomes are most useful in the prediction of long-term liver graft function. This issue emphasises the need for more diverse surrogate markers of injury to improve study design and predictive models.

Despite a decade of research, no LT biomarkers are currently available for use in clinical practice. Large multicentre clinical trials have generated vast amounts of data and information at various molecular levels, demonstrating a promising opportunity for cell-free biomarkers to be introduced into clinical care. This article summarised how recent advances in sequencing and biotechnological methodologies are contributing to the identification of new transcriptomic and genomic biomarkers that could be used in post-transplant management. Data-driven approaches have allowed for the integration and interpretation of the structural and functional features of EVs, miRNA, and cf-ddDNA. Molecular profiles of biofluids such as blood, urine, perfusate, and bile are being integrated with post-LT challenges, providing new opportunities to understand how specific perturbations in biological fluids can signal the early stages of disease.

Financial support

This review was partially supported by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) of the National Institutes of Health under awards numbers: R01DK109581 and R01DK122682 (PI: VRM).

Authors’ contributions

EB (Literature review, Writing –original draft, review & editing), JM (Writing – review & editing), TR (Writing – review & editing), DM (Supervision; Writing – review & editing), VM (Literature review; Writing – original draft, review & editing).

Conflict of interest

The authors of this manuscript have no conflicts of interest to disclose.

Please refer to the accompanying ICMJE disclosure forms for further details.

Footnotes

Author names in bold designate shared co-first authorship

Supplementary data to this article can be found online at https://doi.org/10.1016/j.jhepr.2022.100439.

Supplementary data

The following is the supplementary data to this article:

Multimedia component 1
mmc1.pdf (390.5KB, pdf)

References

  • 1.Kwong A.J., Kim W.R., Lake J.R., Smith J.M., Schladt D.P., Skeans M.A., et al. OPTN/SRTR 2019 annual data report: liver. Am J Transpl. 2021;21(Suppl 2):208–315. doi: 10.1111/ajt.16494. [DOI] [PubMed] [Google Scholar]
  • 2.Pearlman B.L., Traub N. Sustained virologic response to antiviral therapy for chronic hepatitis C virus infection: a cure and so much more. Clin Infect Dis. 2011;52(7):889–900. doi: 10.1093/cid/cir076. [DOI] [PubMed] [Google Scholar]
  • 3.Van Ha T.G. Liver biopsy in liver transplant recipients. Semin Interv Radiol. 2004;21(4):271–274. doi: 10.1055/s-2004-861561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Boyd A., Cain O., Chauhan A., Webb G.J. Medical liver biopsy: background, indications, procedure and histopathology. Frontline Gastroenterol. 2020;11(1):40–47. doi: 10.1136/flgastro-2018-101139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bedossa P., Dargère D., Paradis V. Sampling variability of liver fibrosis in chronic hepatitis C. Hepatol Baltim Md. 2003;38(6):1449–1457. doi: 10.1016/j.hep.2003.09.022. [DOI] [PubMed] [Google Scholar]
  • 6.Guido M., Rugge M. Liver biopsy sampling in chronic viral hepatitis. Semin Liver Dis. 2004;24(1):89–97. doi: 10.1055/s-2004-823103. [DOI] [PubMed] [Google Scholar]
  • 7.Ratziu V., Charlotte F., Heurtier A., Gombert S., Giral P., Bruckert E., et al. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology. 2005;128(7):1898–1906. doi: 10.1053/j.gastro.2005.03.084. [DOI] [PubMed] [Google Scholar]
  • 8.Montaner J., Ramiro L., Simats A., Tiedt S., Makris K., Jickling G.C., et al. Multilevel omics for the discovery of biomarkers and therapeutic targets for stroke. Nat Rev Neurol. 2020;16(5):247–264. doi: 10.1038/s41582-020-0350-6. [DOI] [PubMed] [Google Scholar]
  • 9.Christians U., Klawitter J., Klawitter J. Biomarkers in transplantation--proteomics and metabolomics. Ther Drug Monit. 2016;38(Suppl 1):S70–74. doi: 10.1097/FTD.0000000000000243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cui D., Li W., Jiang D., Wu J., Xie J., Wu Y. Advances in multi-omics applications in HBV-associated hepatocellular carcinoma. Front Med. 2021;8:754709. doi: 10.3389/fmed.2021.754709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hu C., Jia W. Multi-omics profiling: the way towards precision medicine in metabolic diseases [published online ahead of print, 2021 Aug 18] J Mol Cell Biol. 2021 doi: 10.1093/jmcb/mjab051. mjab051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wu Y., Liu Z., Xu X. Molecular subtyping of hepatocellular carcinoma: a step toward precision medicine. Cancer Commun Lond Engl. 2020;40(12):681–693. doi: 10.1002/cac2.12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Löffler M.W., Mohr C., Bichmann L., Freudenmann L.K., Walzer M., Schroeder C.M., et al. Multi-omics discovery of exome-derived neoantigens in hepatocellular carcinoma. Genome Med. 2019;11(1):28. doi: 10.1186/s13073-019-0636-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cavalli M., Diamanti K., Pan G., Spalinskas R., Kumar C., Deshmukh A.S., et al. A multi-omics approach to liver diseases: integration of single nuclei transcriptomics with proteomics and HiCap bulk data in human liver. Omics J Integr Biol. 2020;24(4):180–194. doi: 10.1089/omi.2019.0215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Diels S., Cuypers B., Tvarijonaviciute A., Derudas B., Van Dijck E., Verrijken A., et al. A targeted multi-omics approach reveals paraoxonase-1 as a determinant of obesity-associated fatty liver disease. Clin Epigenetics. 2021;13(1):158. doi: 10.1186/s13148-021-01142-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Strimbu K., Tavel J.A. What are biomarkers? Curr Opin HIV AIDS. 2010;5(6):463–466. doi: 10.1097/COH.0b013e32833ed177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Füzéry A.K., Levin J., Chan M.M., Chan D.W. Translation of proteomic biomarkers into FDA approved cancer diagnostics: issues and challenges. Clin Proteomics. 2013;10(1):13. doi: 10.1186/1559-0275-10-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Tolstikov V., Moser A.J., Sarangarajan R., Narain N.R., Kiebish M.A. Current status of metabolomic biomarker discovery: impact of study design and demographic characteristics. Metabolites. 2020;10(6):E224. doi: 10.3390/metabo10060224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Salvadori M., Tsalouchos A. Biomarkers in renal transplantation: an updated review. World J Transpl. 2017;7(3):161–178. doi: 10.5500/wjt.v7.i3.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mandel P., Metais P. Nuclear acids in human blood plasma. C R Seances Soc Biol Fil. 1948;142(3-4):241–243. [PubMed] [Google Scholar]
  • 21.Laugsand E.A., Brenne S.S., Skorpen F. DNA methylation markers detected in blood, stool, urine, and tissue in colorectal cancer: a systematic review of paired samples. Int J Colorectal Dis. 2021;36(2):239–251. doi: 10.1007/s00384-020-03757-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Loktionov A. Biomarkers for detecting colorectal cancer non-invasively: DNA, RNA or proteins? World J Gastrointest Oncol. 2020;12(2):124–148. doi: 10.4251/wjgo.v12.i2.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Locke W.J., Guanzon D., Ma C., Liew Y.J., Duesing K.R., Fung K.Y.C., et al. DNA methylation cancer biomarkers: translation to the clinic. Front Genet. 2019;10:1150. doi: 10.3389/fgene.2019.01150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gruner H.N., McManus M.T. Examining the evidence for extracellular RNA function in mammals. Nat Rev Genet. 2021;22(7):448–458. doi: 10.1038/s41576-021-00346-8. [DOI] [PubMed] [Google Scholar]
  • 25.Enache L.S., Enache E.L., Ramière C., Diaz O., Bancu L., Sin A., et al. Circulating RNA molecules as biomarkers in liver disease. Int J Mol Sci. 2014;15(10):17644–17666. doi: 10.3390/ijms151017644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bollati V., Motta V., Iodice S., Carugno M. In: Epigenomics in Health and Disease. Fraga M.F., Fernández A.F., editors. Academic Press; 2016. Chapter 8 - epigenomic studies in epidemiology; pp. 163–182. [DOI] [Google Scholar]
  • 27.Wetmore B.A., Brees D.J., Singh R., Watkins P.B., Andersen M.E., Loy J., et al. Quantitative analyses and transcriptomic profiling of circulating messenger RNAs as biomarkers of rat liver injury. Hepatol Baltim Md. 2010;51(6):2127–2139. doi: 10.1002/hep.23574. [DOI] [PubMed] [Google Scholar]
  • 28.Ahmadzada T., Kao S., Reid G., Clarke S., Grau G.E., Hosseini-Beheshti E. Extracellular vesicles as biomarkers in malignant pleural mesothelioma: a review. Crit Rev Oncol Hematol. 2020;150:102949. doi: 10.1016/j.critrevonc.2020.102949. [DOI] [PubMed] [Google Scholar]
  • 29.Turchinovich A., Samatov T.R., Tonevitsky A.G., Burwinkel B. Circulating miRNAs: cell-cell communication function? Front Genet. 2013;4:119. doi: 10.3389/fgene.2013.00119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Gieseler F., Ender F. Extracellular vesicles and cell-cell communication: new insights and new therapeutic strategies not only in oncology. Int J Mol Sci. 2020;21(12):E4331. doi: 10.3390/ijms21124331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.McMasters A., McMasters K.M., Hao H. Exosome to promote cancer progression via its bioactive cargoes. Arch Cancer Biol Ther. 2021;2(2):29–34. [PMC free article] [PubMed] [Google Scholar]
  • 32.Mathieu M., Martin-Jaular L., Lavieu G., Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17. doi: 10.1038/s41556-018-0250-9. [DOI] [PubMed] [Google Scholar]
  • 33.Mastoridis S., Bertolino G.M., Whitehouse G., Dazzi F., Sanchez-Fueyo A., Martinez-Llordella M. Multiparametric analysis of circulating exosomes and other small extracellular vesicles by advanced imaging flow cytometry. Front Immunol. 2018;9:1583. doi: 10.3389/fimmu.2018.01583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mastoridis S., Londoño M.C., Kurt A., Kodela E., Crespo E., Mason J., et al. Impact of donor extracellular vesicle release on recipient cell “cross-dressing” following clinical liver and kidney transplantation. Am J Transpl. 2021;21(7):2387–2398. doi: 10.1111/ajt.16123. [DOI] [PubMed] [Google Scholar]
  • 35.Mastoridis S., Martinez-Llordella M., Sanchez-Fueyo A. Extracellular vesicles as mediators of alloimmunity and their therapeutic potential in liver transplantation. World J Transpl. 2020;10(11):330–344. doi: 10.5500/wjt.v10.i11.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Borrelli D.A., Yankson K., Shukla N., Vilanilam G., Ticer T., Wolfram J. Extracellular vesicle therapeutics for liver disease. J Contr Release. 2018;273:86–98. doi: 10.1016/j.jconrel.2018.01.022. [DOI] [PubMed] [Google Scholar]
  • 37.De Stefano N., Navarro-Tableros V., Roggio D., Calleri A., Rigo F., David E., et al. Human liver stem cell-derived extracellular vesicles reduce injury in a model of normothermic machine perfusion of rat livers previously exposed to a prolonged warm ischemia. Transpl Int. 2021;34(9):1607–1617. doi: 10.1111/tri.13980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Balasubramanian S., Rajasingh S., Thangavel J., Dawn B., Rajasingh J. In: Mesenchymal Stem Cell Derived Exosomes. Tang Y., Dawn B., editors. Academic Press; 2015. Chapter 3 - exosome function in miRNA-mediated paracrine effects; pp. 37–62. [DOI] [Google Scholar]
  • 39.Turunen T.A., Roberts T.C., Laitinen P., Väänänen M.A., Korhonen P., Malm T., et al. Changes in nuclear and cytoplasmic microRNA distribution in response to hypoxic stress. Sci Rep. 2019;9(1):10332. doi: 10.1038/s41598-019-46841-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gorospe M., Tominaga K., Wu X., Fähling M., Ivan M. Post-transcriptional control of the hypoxic response by RNA-binding proteins and MicroRNAs. Front Mol Neurosci. 2011;4:7. doi: 10.3389/fnmol.2011.00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sen C.K., Ghatak S. miRNA control of tissue repair and regeneration. Am J Pathol. 2015;185(10):2629–2640. doi: 10.1016/j.ajpath.2015.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Muthukumar T., Akat K.M., Yang H., Schwartz J.E., Li C., Bang H., et al. Serum MicroRNA transcriptomics and acute rejection or recurrent hepatitis C virus in human liver allograft recipients: a pilot study. Transplantation. 2021 doi: 10.1097/TP.0000000000003815. Published online May 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ruiz P., Millán O., Ríos J., Díaz A., Sastre L., Colmenero J., et al. MicroRNAs 155-5p, 122-5p, and 181a-5p identify patients with graft dysfunction due to T cell-mediated rejection after liver transplantation. Liver Transpl. 2020;26(10):1275–1286. doi: 10.1002/lt.25842. [DOI] [PubMed] [Google Scholar]
  • 44.Millán O., Ruiz P., Orts L., Ferré P., Crespo G., Santana M., et al. Monitoring of miR-181a-5p and miR-155-5p Plasmatic Expression as Prognostic Biomarkers for Acute and Subclinical Rejection in de novo Adult Liver Transplant Recipients. Front Immunol. 2019;10:873. doi: 10.3389/fimmu.2019.00873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Verhoeven C.J., Farid W.R.R., Roest H.P., Ramakrishnaiah V., de Ruiter P.E., de Jonge J., et al. Polarized release of hepatic microRNAs into bile and serum in response to cellular injury and impaired liver function. Liver Int. 2016;36(6):883–892. doi: 10.1111/liv.12955. [DOI] [PubMed] [Google Scholar]
  • 46.Moghoofei M., Najafipour S., Mostafaei S., Tavakoli A., Bokharaei-Salim F., Ghorbani S., et al. MicroRNAs profiling in HIV, HCV, and HIV/HCV Co-infected patients. Curr HIV Res. 2021;19(1):27–34. doi: 10.2174/1570162X18666200908112113. [DOI] [PubMed] [Google Scholar]
  • 47.Morishita A., Oura K., Tadokoro T., Fujita K., Tani J., Masaki T. MicroRNAs in the pathogenesis of hepatocellular carcinoma: a review. Cancers. 2021;13(3):514. doi: 10.3390/cancers13030514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.McCrae J.C., Sharkey N., Webb D.J., Vliegenthart A.D.B., Dear J.W. Ethanol consumption produces a small increase in circulating miR-122 in healthy individuals. Clin Toxicol Phila Pa. 2016;54(1):53–55. doi: 10.3109/15563650.2015.1112015. [DOI] [PubMed] [Google Scholar]
  • 49.Church R.J., Kullak-Ublick G.A., Aubrecht J., Bonkovsky H.L., Chalasani N., Fontana R.J., et al. Candidate biomarkers for the diagnosis and prognosis of drug-induced liver injury: an international collaborative effort. Hepatol Baltim Md. 2019;69(2):760–773. doi: 10.1002/hep.29802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ng R., Wu H., Xiao H., Chen X., Willenbring H., Steer C.J., et al. Inhibition of microRNA-24 expression in liver prevents hepatic lipid accumulation and hyperlipidemia. Hepatol Baltim Md. 2014;60(2):554–564. doi: 10.1002/hep.27153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Zarrinpar A., Gupta S., Maurya M.R., Subramaniam S., Loomba R. Serum microRNAs explain discordance of non-alcoholic fatty liver disease in monozygotic and dizygotic twins: a prospective study. Gut. 2016;65(9):1546–1554. doi: 10.1136/gutjnl-2015-309456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ward J., Kanchagar C., Veksler-Lublinsky I., Lee R.C., McGill M.R., Jaeschke H., et al. Circulating microRNA profiles in human patients with acetaminophen hepatotoxicity or ischemic hepatitis. Proc Natl Acad Sci U S A. 2014;111(33):12169–12174. doi: 10.1073/pnas.1412608111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Krauskopf J., Caiment F., Claessen S.M., Johnson K.J., Warner R.L., Schomaker S.J., et al. Application of high-throughput sequencing to circulating microRNAs reveals novel biomarkers for drug-induced liver injury. Toxicol Sci. 2015;143(2):268–276. doi: 10.1093/toxsci/kfu232. [DOI] [PubMed] [Google Scholar]
  • 54.Jin Y., Wong Y.S., Goh B.K.P., Chan C.Y., Cheow P.C., Chow P.K.H., et al. Circulating microRNAs as potential diagnostic and prognostic biomarkers in hepatocellular carcinoma. Sci Rep. 2019;9(1):10464. doi: 10.1038/s41598-019-46872-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Nakano T., Chen I.H., Goto S., Lai C.Y., Tseng H.P., Hsu L.W., et al. Hepatic miR-301a as a liver transplant rejection biomarker? And its role for interleukin-6 production in hepatocytes. Omics J Integr Biol. 2017;21(1):55–66. doi: 10.1089/omi.2016.0164. [DOI] [PubMed] [Google Scholar]
  • 56.Zhang P., Guo Z., Zhong K., Li Q., Ouyang J., Chen M., et al. Evaluation of immune profiles and MicroRNA expression profiles in peripheral blood mononuclear cells of long-term stable liver transplant recipients and recipients with acute rejection episodes. Transpl Proc. 2015;47(10):2907–2915. doi: 10.1016/j.transproceed.2015.10.048. [DOI] [PubMed] [Google Scholar]
  • 57.Sarma N.J., Tiriveedhi V., Subramanian V., Shenoy S., Crippin J.S., Chapman W.C., et al. Hepatitis C virus mediated changes in miRNA-449a modulates inflammatory biomarker YKL40 through components of the NOTCH signaling pathway. PLoS One. 2012;7(11) doi: 10.1371/journal.pone.0050826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nasser M.Z., Zayed N.A., Mohamed A.M., Attia D., Esmat G., Khairy A. Circulating microRNAs (miR-21, miR-223, miR-885-5p) along the clinical spectrum of HCV-related chronic liver disease in Egyptian patients. Arab J Gastroenterol. 2019;20(4):198–204. doi: 10.1016/j.ajg.2019.11.003. [DOI] [PubMed] [Google Scholar]
  • 59.Zhou X., Fang S., Wang M., Xiong A., Zheng C., Wang J., et al. Diagnostic value of circulating miRNA-122 for hepatitis B virus and/or hepatitis C virus-associated chronic viral hepatitis. Biosci Rep. 2019;39(9) doi: 10.1042/BSR20190900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Nakashiki S., Miuma S., Mishima H., Masumoto H., Hidaka M., Soyama A., et al. Bile extracellular vesicles from end-stage liver disease patients show altered microRNA content. Hepatol Int. 2021;15(3):821–830. doi: 10.1007/s12072-021-10196-5. [DOI] [PubMed] [Google Scholar]
  • 61.Momen-Heravi F., Saha B., Kodys K., Catalano D., Satishchandran A., Szabo G. Increased number of circulating exosomes and their microRNA cargos are potential novel biomarkers in alcoholic hepatitis. J Transl Med. 2015;13:261. doi: 10.1186/s12967-015-0623-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bala S., Petrasek J., Mundkur S., Catalano D., Levin I., Ward J., et al. Circulating microRNAs in exosomes indicate hepatocyte injury and inflammation in alcoholic, drug-induced, and inflammatory liver diseases. Hepatol Baltim Md. 2012;56(5):1946–1957. doi: 10.1002/hep.25873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Trajkovski M., Hausser J., Soutschek J., Bhat B., Akin A., Zavolan M., et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature. 2011;474(7353):649–653. doi: 10.1038/nature10112. [DOI] [PubMed] [Google Scholar]
  • 64.Meng F., Glaser S.S., Francis H., Yang F., Han Y., Stokes A., et al. Epigenetic regulation of miR-34a expression in alcoholic liver injury. Am J Pathol. 2012;181(3):804–817. doi: 10.1016/j.ajpath.2012.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Blaya D., Coll M., Rodrigo-Torres D., Vila-Casadesús M., Altamirano J., Llopis M., et al. Integrative microRNA profiling in alcoholic hepatitis reveals a role for microRNA-182 in liver injury and inflammation. Gut. 2016;65(9):1535–1545. doi: 10.1136/gutjnl-2015-311314. [DOI] [PubMed] [Google Scholar]
  • 66.Saha B., Momen-Heravi F., Kodys K., Szabo G. MicroRNA cargo of extracellular vesicles from alcohol-exposed monocytes signals naive monocytes to differentiate into M2 macrophages. J Biol Chem. 2016;291(1):149–159. doi: 10.1074/jbc.M115.694133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tang Y., Banan A., Forsyth C.B., Fields J.Z., Lau C.K., Zhang L.J., et al. Effect of alcohol on miR-212 expression in intestinal epithelial cells and its potential role in alcoholic liver disease. Alcohol Clin Exp Res. 2008;32(2):355–364. doi: 10.1111/j.1530-0277.2007.00584.x. [DOI] [PubMed] [Google Scholar]
  • 68.Saikia P., Roychowdhury S., Bellos D., Pollard K.A., McMullen M.R., McCullough R.L., et al. Hyaluronic acid 35 normalizes TLR4 signaling in Kupffer cells from ethanol-fed rats via regulation of microRNA291b and its target Tollip. Sci Rep. 2017;7(1):15671. doi: 10.1038/s41598-017-15760-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Li M., He Y., Zhou Z., Ramirez T., Gao Y., Gao Y., et al. MicroRNA-223 ameliorates alcoholic liver injury by inhibiting the IL-6-p47phox-oxidative stress pathway in neutrophils. Gut. 2017;66(4):705–715. doi: 10.1136/gutjnl-2016-311861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Leti F., Malenica I., Doshi M., Courtright A., Van Keuren-Jensen K., Legendre C., et al. High-throughput sequencing reveals altered expression of hepatic microRNAs in nonalcoholic fatty liver disease-related fibrosis. Transl Res J Lab Clin Med. 2015;166(3):304–314. doi: 10.1016/j.trsl.2015.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Ao R., Wang Y., Tong J., Wang B.F. Altered microRNA-9 expression level is directly correlated with pathogenesis of nonalcoholic fatty liver disease by targeting Onecut2 and SIRT1. Med Sci Monit Int Med J Exp Clin Res. 2016;22:3804–3819. doi: 10.12659/msm.897207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zheng L., Lv G.C., Sheng J., Yang Y.D. Effect of miRNA-10b in regulating cellular steatosis level by targeting PPAR-alpha expression, a novel mechanism for the pathogenesis of NAFLD. J Gastroenterol Hepatol. 2010;25(1):156–163. doi: 10.1111/j.1440-1746.2009.05949.x. [DOI] [PubMed] [Google Scholar]
  • 73.Du J., Niu X., Wang Y., Kong L., Wang R., Zhang Y., et al. MiR-146a-5p suppresses activation and proliferation of hepatic stellate cells in nonalcoholic fibrosing steatohepatitis through directly targeting Wnt1 and Wnt5a. Sci Rep. 2015;5:16163. doi: 10.1038/srep16163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cermelli S., Ruggieri A., Marrero J.A., Ioannou G.N., Beretta L. Circulating microRNAs in patients with chronic hepatitis C and non-alcoholic fatty liver disease. PLoS One. 2011;6(8) doi: 10.1371/journal.pone.0023937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pirola C.J., Fernández Gianotti T., Castaño G.O., Mallardi P., San Martino J., Mora Gonzalez Lopez Ledesma M., et al. Circulating microRNA signature in non-alcoholic fatty liver disease: from serum non-coding RNAs to liver histology and disease pathogenesis. Gut. 2015;64(5):800–812. doi: 10.1136/gutjnl-2014-306996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tan Y., Ge G., Pan T., Wen D., Gan J. A pilot study of serum microRNAs panel as potential biomarkers for diagnosis of nonalcoholic fatty liver disease. PLoS One. 2014;9(8) doi: 10.1371/journal.pone.0105192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Latorre J., Moreno-Navarrete J.M., Mercader J.M., Sabater M., Rovira Ò., Gironès J., et al. Decreased lipid metabolism but increased FA biosynthesis are coupled with changes in liver microRNAs in obese subjects with NAFLD. Int J Obes 2005. 2017;41(4):620–630. doi: 10.1038/ijo.2017.21. [DOI] [PubMed] [Google Scholar]
  • 78.Świtlik W.Z., Bielecka-Kowalska A., Karbownik M.S., Kordek R., Jabłkowski M., Szemraj J. Forms of diagnostic material as sources of miRNA biomarkers in hepatocellular carcinoma: a preliminary study. Biomark Med. 2019;13(7):523–534. doi: 10.2217/bmm-2018-0485. [DOI] [PubMed] [Google Scholar]
  • 79.Pelizzaro F., Cardin R., Sartori A., Imondi A., Penzo B., Aliberti C., et al. Circulating MicroRNA-21 and MicroRNA-122 as prognostic biomarkers in hepatocellular carcinoma patients treated with transarterial chemoembolization. Biomedicines. 2021;9(8):890. doi: 10.3390/biomedicines9080890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jopling C. Liver-specific microRNA-122. RNA Biol. 2012;9(2):137–142. doi: 10.4161/rna.18827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lewis A.P., Jopling C.L. Regulation and biological function of the liver-specific miR-122. Biochem Soc Trans. 2010;38(6):1553–1557. doi: 10.1042/BST0381553. [DOI] [PubMed] [Google Scholar]
  • 82.Lagos-Quintana M., Rauhut R., Yalcin A., Meyer J., Lendeckel W., Tuschl T. Identification of tissue-specific microRNAs from mouse. Curr Biol CB. 2002;12(9):735–739. doi: 10.1016/s0960-9822(02)00809-6. [DOI] [PubMed] [Google Scholar]
  • 83.Wang X., He Y., Mackowiak B., Gao B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut. 2021;70(4):784–795. doi: 10.1136/gutjnl-2020-322526. [DOI] [PubMed] [Google Scholar]
  • 84.Morimoto A., Kannari M., Tsuchida Y., Sasaki S., Saito C., Matsuta T., et al. An HNF4α-microRNA-194/192 signaling axis maintains hepatic cell function. J Biol Chem. 2017;292(25):10574–10585. doi: 10.1074/jbc.M117.785592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Krützfeldt J., Rösch N., Hausser J., Manoharan M., Zavolan M., Stoffel M. MicroRNA-194 is a target of transcription factor 1 (Tcf1, HNF1α) in adult liver and controls expression of frizzled-6. Hepatol Baltim Md. 2012;55(1):98–107. doi: 10.1002/hep.24658. [DOI] [PubMed] [Google Scholar]
  • 86.Roderburg C., Urban G.W., Bettermann K., Vucur M., Zimmermann H., Schmidt S., et al. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatol Baltim Md. 2011;53(1):209–218. doi: 10.1002/hep.23922. [DOI] [PubMed] [Google Scholar]
  • 87.Caviglia J.M., Yan J., Jang M.K., Gwak G.Y., Affo S., Yu L., et al. MicroRNA-21 and Dicer are dispensable for hepatic stellate cell activation and the development of liver fibrosis. Hepatol Baltim Md. 2018;67(6):2414–2429. doi: 10.1002/hep.29627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang K., Zhang S., Marzolf B., Troisch P., Brightman A., Hu Z., et al. Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc Natl Acad Sci U S A. 2009;106(11):4402–4407. doi: 10.1073/pnas.0813371106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Howell L.S., Ireland L., Park B.K., Goldring C.E. MiR-122 and other microRNAs as potential circulating biomarkers of drug-induced liver injury. Expert Rev Mol Diagn. 2018;18(1):47–54. doi: 10.1080/14737159.2018.1415145. [DOI] [PubMed] [Google Scholar]
  • 90.Chang Y., Han J.A., Kang S.M., Jeong S.W., Ryu T., Park H.S., et al. Clinical impact of serum exosomal microRNA in liver fibrosis. PLoS One. 2021;16(9) doi: 10.1371/journal.pone.0255672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Shaked A., Chang B.L., Barnes M.R., Sayre P., Li Y.R., Asare S., et al. An ectopically expressed serum miRNA signature is prognostic, diagnostic, and biologically related to liver allograft rejection. Hepatol Baltim Md. 2017;65(1):269–280. doi: 10.1002/hep.28786. [DOI] [PubMed] [Google Scholar]
  • 92.Jochmans I., Fieuws S., Tieken I., Samuel U., Pirenne J. The impact of implantation time during liver transplantation on outcome: a eurotransplant cohort study. Transpl Direct. 2018;4(6):e356. doi: 10.1097/TXD.0000000000000793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Selten J.W., Verhoeven C.J., Heedfeld V., Roest H.P., 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. doi: 10.1002/lt.24766. [DOI] [PubMed] [Google Scholar]
  • 94.Faraldi M., Gomarasca M., Sansoni V., Perego S., Banfi G., Lombardi G. Normalization strategies differently affect circulating miRNA profile associated with the training status. Sci Rep. 2019;9(1):1584. doi: 10.1038/s41598-019-38505-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Roest H.P., IJzermans J.N.M., van der Laan L.J.W. Evaluation of RNA isolation methods for microRNA quantification in a range of clinical biofluids. BMC Biotechnol. 2021;21(1):48. doi: 10.1186/s12896-021-00706-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Chorley B.N., Atabakhsh E., Doran G., Gautier J.C., Ellinger-Ziegelbauer H., Jackson D., et al. Methodological considerations for measuring biofluid-based microRNA biomarkers. Crit Rev Toxicol. 2021;51(3):264–282. doi: 10.1080/10408444.2021.1907530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Andersen C.L., Jensen J.L., Ørntoft T.F. Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets. Cancer Res. 2004;64(15):5245–5250. doi: 10.1158/0008-5472.CAN-04-0496. [DOI] [PubMed] [Google Scholar]
  • 98.Sheinerman K.S., Tsivinsky V.G., Abdullah L., Crawford F., Umansky S.R. Plasma microRNA biomarkers for detection of mild cognitive impairment: biomarker validation study. Aging. 2013;5(12):925–938. doi: 10.18632/aging.100624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Starlinger P., Hackl H., Pereyra D., Skalicky S., Geiger E., Finsterbusch M., et al. Predicting postoperative liver dysfunction based on blood-derived MicroRNA signatures. Hepatol Baltim Md. 2019;69(6):2636–2651. doi: 10.1002/hep.30572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sheinerman K.S., Tsivinsky V.G., Umansky S.R. Analysis of organ-enriched microRNAs in plasma as an approach to development of Universal Screening Test: feasibility study. J Transl Med. 2013;11:304. doi: 10.1186/1479-5876-11-304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kowal J., Arras G., Colombo M., Jouve M., Morath J.P., Primdal-Bengtson B., et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113(8):E968–E977. doi: 10.1073/pnas.1521230113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Maia J., Batista S., Couto N., Gregório A.C., Bodo C., Elzanowska J., et al. Employing flow cytometry to extracellular vesicles sample microvolume Analysis and quality control. Front Cell Dev Biol. 2020;8:593750. doi: 10.3389/fcell.2020.593750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Beck J., Bierau S., Balzer S., Andag R., Kanzow P., Schmitz J., et al. Digital droplet PCR for rapid quantification of donor DNA in the circulation of transplant recipients as a potential universal biomarker of graft injury. Clin Chem. 2013;59(12):1732–1741. doi: 10.1373/clinchem.2013.210328. [DOI] [PubMed] [Google Scholar]
  • 104.Gielis E.M., Ledeganck K.J., De Winter B.Y., Del Favero J., Bosmans J.L., Claas F.H., et al. Cell-free DNA: an upcoming biomarker in transplantation. Am J Transpl. 2015;15(10):2541–2551. doi: 10.1111/ajt.13387. [DOI] [PubMed] [Google Scholar]
  • 105.Lo Y.M., Tein M.S., Pang C.C., Yeung C.K., Tong K.L., Hjelm N.M. Presence of donor-specific DNA in plasma of kidney and liver-transplant recipients. Lancet Lond Engl. 1998;351(9112):1329–1330. doi: 10.1016/s0140-6736(05)79055-3. [DOI] [PubMed] [Google Scholar]
  • 106.De Vlaminck I., Valantine H.A., Snyder T.M., Strehl C., Cohen G., Luikart H., et al. Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection. Sci Transl Med. 2014;6(241):241ra77. doi: 10.1126/scitranslmed.3007803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Snyder T.M., Khush K.K., Valantine H.A., Quake S.R. Universal noninvasive detection of solid organ transplant rejection. Proc Natl Acad Sci U S A. 2011;108(15):6229–6234. doi: 10.1073/pnas.1013924108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.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) doi: 10.1371/journal.pmed.1002286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Macher H.C., Suárez-Artacho G., Guerrero J.M., Gómez-Bravo M.A., Álvarez-Gómez S., Bernal-Bellido C., et al. Monitoring of transplanted liver health by quantification of organ-specific genomic marker in circulating DNA from receptor. PLoS One. 2014;9(12) doi: 10.1371/journal.pone.0113987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Fernández-Galán E., Badenas C., Fondevila C., Jiménez W., Navasa M., Puig-Butillé J.A., et al. Monitoring of donor-derived cell-free DNA by short tandem repeats: concentration of total cell-free DNA and fragment size for acute rejection risk assessment in liver transplantation. Liver Transpl. 2021 doi: 10.1002/lt.26272. Published online August 18. [DOI] [PubMed] [Google Scholar]
  • 111.Lehmann-Werman R., Magenheim J., Moss J., Neiman D., Abraham O., Piyanzin S., et al. Monitoring liver damage using hepatocyte-specific methylation markers in cell-free circulating DNA. JCI Insight. 2018;3(12):120687. doi: 10.1172/jci.insight.120687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bataller R., Brenner D.A. Liver fibrosis. J Clin Invest. 2005;115(2):209–218. doi: 10.1172/JCI24282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Iacob D.G., Rosca A., Ruta S.M. Circulating microRNAs as non-invasive biomarkers for hepatitis B virus liver fibrosis. World J Gastroenterol. 2020;26(11):1113–1127. doi: 10.3748/wjg.v26.i11.1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Xu J., Wu C., Che X., Wang L., Yu D., Zhang T., et al. Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol Carcinog. 2011;50(2):136–142. doi: 10.1002/mc.20712. [DOI] [PubMed] [Google Scholar]
  • 115.Qi P., Cheng S.Q., Wang H., Li N., Chen Y.F., Gao C.F. Serum microRNAs as biomarkers for hepatocellular carcinoma in Chinese patients with chronic hepatitis B virus infection. PLoS One. 2011;6(12) doi: 10.1371/journal.pone.0028486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Pesch B., Brüning T., Johnen G., Casjens S., Bonberg N., Taeger D., et al. Biomarker research with prospective study designs for the early detection of cancer. Biochim Biophys Acta. 2014;1844(5):874–883. doi: 10.1016/j.bbapap.2013.12.007. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.pdf (390.5KB, pdf)

Articles from JHEP Reports are provided here courtesy of Elsevier

RESOURCES