Home Variability in, variability out: best practice recommendations to standardize pre-analytical variables in the detection of circulating and tissue microRNAs
Article Publicly Available

Variability in, variability out: best practice recommendations to standardize pre-analytical variables in the detection of circulating and tissue microRNAs

  • Jenna Khan EMAIL logo , Joshua A. Lieberman and Christina M. Lockwood
Published/Copyright: March 17, 2017

Abstract:

microRNAs (miRNAs) hold promise as biomarkers for a variety of disease processes and for determining cell differentiation. These short RNA species are robust, survive harsh treatment and storage conditions and may be extracted from blood and tissue. Pre-analytical variables are critical confounders in the analysis of miRNAs: we elucidate these and identify best practices for minimizing sample variation in blood and tissue specimens. Pre-analytical variables addressed include patient-intrinsic variation, time and temperature from sample collection to storage or processing, processing methods, contamination by cells and blood components, RNA extraction method, normalization, and storage time/conditions. For circulating miRNAs, hemolysis and blood cell contamination significantly affect profiles; samples should be processed within 2 h of collection; ethylene diamine tetraacetic acid (EDTA) is preferred while heparin should be avoided; samples should be “double spun” or filtered; room temperature or 4 °C storage for up to 24 h is preferred; miRNAs are stable for at least 1 year at –20 °C or –80 °C. For tissue-based analysis, warm ischemic time should be <1 h; cold ischemic time (4 °C) <24 h; common fixative used for all specimens; formalin fix up to 72 h prior to processing; enrich for cells of interest; validate candidate biomarkers with in situ visualization. Most importantly, all specimen types should have standard and common workflows with careful documentation of relevant pre-analytical variables.

Introduction

MicroRNAs (miRNAs) are short (~18–25 nucleotides) non-coding RNAs involved in regulating cellular functions in a wide range of health and disease states by repressing gene expression through physical interaction with messenger RNAs (mRNAs). A single miRNA is capable of regulating hundreds of mRNAs [1]. Due to their relative high stability, miRNAs are actively being investigated as biomarkers for diagnostic and prognostic purposes. However, studies to date have yielded inconsistent results and clinical assays remain elusive. A number of recent reviews [1], [2], [3], [4], [5], [6], [7], [8], [9] have thoroughly discussed the challenges and pitfalls involved in accurately quantifying miRNAs, including pre-analytical, analytical, and methodological factors that contribute to result heterogeneity. There is clearly an urgent need for a global consensus on study design and laboratory procedures.

Pre-analytical variables are a well-established source of bias in laboratory testing [10], [11], [12]. This bias is particularly pronounced in the study of miRNAs due to inconsistent specimen collection, handling, processing, extraction and normalization. In addition, the small size, complex biosynthesis and ubiquitous presence of miRNAs in essentially all tissue types make them inherently difficult to consistently and specifically quantify. We provide a summary of current best practices with the intent to standardize pre-analytical variables in the detection of both circulating and tissue miRNAs (Figure 1).

Figure 1: Sources of pre-analytical variation in miRNA analysis.
Figure 1:

Sources of pre-analytical variation in miRNA analysis.

Pre-analytical variables for miRNAs

The goal of standardized procedures with regard to pre-analytical variables is to collect samples with a consistent composition for reproducible results. Not properly accounting for pre-analytical variables yields inaccurate results or systematic biases [13], [14]. Standardization requires minimizing multiple confounding factors, including prior to collection, within specimens, and during handling, processing, and storage. It is often not possible to control all of these variables, particularly in research studies that rely on banked specimens. In these cases, it is important to consider potentially confounding factors and account for them in result interpretation. Accurate assessment of the impact of pre-analytic variables requires collecting relevant information at the time of specimen procurement. To aid in this process, the College of American Pathologists Diagnostic Intelligence and Health Information Technology Committee sponsored a Biorepository Working Group to develop ranked lists of pre-analytic variables for annotating biospecimens [15]. These lists provide helpful guidelines for prioritizing the most relevant (and practically implementable) variables to record based on the specific needs and capabilities of an institution and study (Table 1).

Table 1:

Common best practice recommendations.

– Study design
 – Develop standard procedures for specimen selection and handling
 – Review miRNA databases to select appropriate miRNAs for study
  – Avoid blood cell miRNAs
 – Account for environmental and biological preanalytic variables with appropriate case and control subjects
 – Document relevant preanalytic variables and deviations from standard procedures
– Extraction
 – Common extraction method for all specimens
– Normalization
 – Use exogenous “spike in” miRNA or validated endogenous miRNA

Common pre-analytical variables

Common biological/environmental variables

Pre-analytical variables can be subdivided into biological, environmental, and technical factors [13]. Biological and environmental factors are predominantly common to both tissue and circulating miRNAs. Multiple studies have highlighted that both external and patient-intrinsic factors such as age, sex, race, body mass index (BMI), diet, fasting state, underlying illnesses/organ dysfunction, blood cell count, exercise, vitamin supplementation, medications, smoking, diurnal variation, chemical exposure and even altitude can influence miRNA concentrations and are often difficult to standardize [2], [13], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27]. Appropriate case and control subjects should be selected and specimen collection protocols designed and consistently implemented to minimize effects of confounding variables. Repeated sampling from the same individual can be considered as an additional method to attenuate the effects of pre-analytical variables [13]. Several recent reviews provide planning guides or outlines for circulating miRNA studies [1], [7].

Common technical variables

Technical factors include collection, processing, transport and storage and can also be controlled for in study design. Technical variables common to both tissue and circulating miRNAs are discussed below with detailed recommendations outlined in specimen-specific sections.

Hemolysis/blood cell contamination

Contamination from blood cells is a significant source of miRNAs and its impact has historically been underappreciated. This has led to the inappropriate use of blood cell-miRNAs as endogenous controls (miR-16) and their incorrect assignment as potential cancer biomarkers in non-blood cell diseases [3], [19]. A recent study demonstrated that blood cell counts and hemolysis can alter plasma miRNA concentrations by up to 50-fold and that 58% of reported tumor-associated circulating miRNAs are highly expressed in blood cells [19]. Among the miRNAs examined, the observed variation stemming solely from blood cells was greater than many literature-reported differences for potential cancer biomarkers, raising a concern that reported findings are related to the secondary effect of blood cell miRNA contamination. Blood cell counts and/or hemolysis also likely contribute to variable results observed between different methodologies (extraction methods, specimen type, etc.) and certainly hinder the establishment of a standard practice for miRNA profiling.

Although this phenomenon has been rigorously studied for circulating miRNAs, blood cell contamination is also relevant for tissue specimens and deserves special consideration. Tumor tissue can be highly vascularized and complete removal of blood cells from tissue is neither common practice nor practical. A more detailed discussion of the challenges associated with selecting cells or regions of interest in tissue is presented below (see “Tissue variability”).

To address this issue, there has been a collective effort to compile blood cell-miRNAs and their purported disease and biologic pathway associations [3], [6], [7], [28], [29], [30], [31]. Existing miRNA-disease relationship databases were recently reviewed and a selection guide created for specific research interests [32]. Investigators should carefully review available resources prior to designing or interpreting studies and eliminate candidate miRNAs that are highly expressed in blood cells. The development of a single comprehensive database of cellular expression patterns for miRNAs would also facilitate better selection of candidate miRNAs [7].

Specimen collection and processing

Collection and processing procedures significantly influence miRNA concentrations. As they are generally specific to either circulating or tissue-derived miRNAs, they are discussed in the corresponding sections below. A recent review provides helpful flow charts for miRNA profiling study design in both body fluids and tissue specimens, including factors to consider based on specimen type, purification requirements and study goals [1].

miRNA extraction and detection

After specimen collection and processing, miRNAs must be extracted. Extraction methods are broadly grouped into two categories: guanidine/phenol/chloroform (GPC)-based extraction and column- or bead-based commercial isolation kits. Most techniques are comparable within a single method category, but demonstrate significant variation between different methods [2]. A recent evaluation of extraction kits demonstrated good comparability between three of five tested kits [20]. In contrast to this study, miRNeasy and mirVana kits have shown increased RNA recovery in studies comparing extraction kits [33], [34], [35], [36], [37]. Despite some inconsistencies, there is evidence that column-based methods perform better than GPC extraction due to organic and phenolic contaminants in GPC-extracted nucleic acids [34], [37]. Size fractionation and gel purification via polyacrylamide gels can subsequently enrich for small RNAs (18–24 nucleotide range) [1]. This process specifically selects for mature miRNAs and reduces miRNA precursors. One proposal is direct miRNA detection (without extraction) as a superior method because it eliminates miRNA loss during extraction, removes the need to normalize to total RNA, and is easier to perform routinely in a clinical laboratory [9]. Overall, the lack of consensus regarding miRNA extraction highlights the importance of using the same extraction method throughout a study to minimize the introduction of confounding variables.

Normalization

Various detection methods [quantitative reverse transcription PCR (qRT-PCR), microarray, RNA sequencing (RNAseq), next generation sequencing (NGS)] are available for miRNA quantification from circulation and tissue alike [1], [9], [38]. All of these methods require normalization to account and correct for variation across samples. Varying normalization methods have likely also contributed to inconsistent miRNA results. Although a normalization consensus has not been established, exogenous miRNA “spike-in” [such as Caenorhabditis elegans (cel-miR-39, cel-miR-54, cel-miR-238) or Arabidopsis thaliana miRNAs] during extraction can assess extraction efficiency [3], [20], [35]. In one study, normalization to a spike in control miRNA reduced extraction bias between five different kits and improved correlation of qRT-PCR results [20]. A ubiquitously expressed endogenous miRNA can also be used to normalize; however, as discussed in the hemolysis section, great care must be taken to select an appropriate miRNA(s) whose concentration is largely unaffected by pre-analytic variables. A study on lung cancer biomarkers outlined an empirical approach to identify endogenous circulating miRNAs for normalization [39]. Another study categorized sets of reference miRNAs for tissue-based analysis of colorectal and pancreatic adenocarcinoma [40]. When a relatively large number of miRNAs are measured, endogenous control methods could be supplemented with a broad normalization approach such as global median normalization (the average expression of each miRNA relative to itself) [3], [5], [41]. Normalization to plasma or serum volume can also correct for variations in extraction efficiency since RNA concentration does not appear to correlate with the number of miRNAs detected [3], [9], [22], [35].

RNA preservation reagents

Multiple technologies have been developed to preserve circulating and tissue-based RNA species, including RNAlater, Tempus, PAXgene, and others. In general, miRNA species are much more stable than mRNA and most studies have not used these methods. Several studies have demonstrated successful isolation and quantitation of specific miRNAs or whole miRNomes from whole blood [42], [43], [44], [45] or tissue [46]. However, few studies have systematically compared RNA stabilization media with a control method. One study directly compared Tempus and PAXgene whole blood stabilization systems and identified small differences in miR30b quantitation [43]. Another detected variation in miRNA quantitation isolated from whole blood preserved in either RNAlater or PAXgene, but noted both systems were appropriate for miRNA analysis [45]. A different study of the PAXgene Tissue stabilization system identified high-concordance in miRNA quantitation between frozen and stabilized specimens (R2=0.95) and reduced concordance between stabilized and formalin-fixed, paraffin embedded (FFPE) tissues (R2=0.81). In summary, RNA stabilization systems are compatible with miRNA analysis, choice of reagent influences total RNA yield and miRNA quantitation, and represents a poorly studied pre-analytical variable. Insufficient data is available to offer a definitive recommendation on the use of RNA preservation reagents. Available data suggests these are compatible with miRNA analysis but we emphasize the need for consistency in study design.

Pre-analytical variables for circulating miRNAs

Measurement of circulating miRNAs holds promise as a non-invasive means of monitoring progression, treatment response, and/or screening for disease. Although appealing to consider circulating miRNAs surrogate markers (“liquid biopsy”) for tissue biopsy-based sampling, it must be emphasized that studies evaluating the population of circulating miRNAs suggest that only ~10–30% of tissue miRNAs are detectable in serum or plasma [28], [47]. In addition, a tumor biomarker model based on placental kinetics suggests that >0.3 g of tumor mass is necessary for detectable miRNA concentrations in circulation [48]. Due to the inherent assumption that circulating miRNAs are altered in a specific manner for a particular disease, it is beneficial to correlate with tissue concentrations. One recent review reported that only 49% of published circulating miRNA signatures in cancer have an accompanying study of miRNA expression in corresponding tissue and only 7% show perfect correlation in a discovery setting [4]. The challenge of matching circulating and tissue miRNAs is highlighted by a recent comparison of tissue and plasma miRNAs in pancreatic adenocarcinoma that noted concordant overexpression of only two of eight studied miRNAs [49].

In summary, the ubiquitous presence of miRNAs and their high concentration in cells compared to fluid makes reducing fluid/cell contamination one of the primary goals of pre-analytical standardization for evaluation of circulating miRNAs. Any step in specimen procurement from within-patient (such as tumor cell fragility) to specimen collection, processing, and storage that can cause cell leakage or lysis can significantly impact miRNAs [50]. Thus, potential variables to control for in study design should be considered broadly and not based solely on miRNA literature (Table 2).

Table 2:

Circulating-specific best practice recommendations.

– Specimen
 – Plasma or serum
 – “Double-spun” platelet-poor or filtered
 – EDTA preferred, avoid heparin
– Collection/processing – minimize hemolysis and blood cell contamination
 – ≥22-gage needle
 – Discard first 1–2 mL of blood (skin plug)
 – Measure concurrent CBC
 – Evaluate specimens for hemolysis as per study design
 – Process blood as quickly as possible (≤2 h)
– Acceptable storage conditions
 – Serum/plasma for 24 h at room temperature or 4 °C
  – At least 1 year at −20 °C or −80 °C
  – All specimens should be stored at the same temperature
  – For longer storage use control specimens matched for storage time
 – Minimize freeze/thaw cycles
– Clearly document preanalytic variables

Specimen type

Although miRNAs can be detected in many fluid types [51], we specifically focus on blood as the most widely studied source. Blood cell-derived miRNAs are substantial contributors to circulating miRNA concentrations, which makes whole blood a suboptimal specimen type [19], [52]. Comparisons of plasma versus serum for miRNA detection have yielded conflicting results as to which is the superior specimen type [5], [20], [30], [35]. It has been suggested that the observed increased plasma-derived miRNA yields are due to the presence of cellular contaminants [33]. Serum, in contrast, has less baseline platelet contamination than plasma; however, clot formation may result in variable release of confounding miRNAs from blood cells [20], [22]. As there is not overwhelming evidence for one specimen type, the choice of serum versus plasma should be based on the specific study goals with miRNAs of interest being cross-referenced to published literature of differential expression in serum versus plasma [30]. For standardization, it is important to use the same specimen type consistently throughout a study and to explicitly define both the specimen type and handling.

Fluid specimens likely contain miRNAs from different sources in different forms: excreted freely by cells, packaged in micro, or exosomes, or passively leaked from damaged cells. Although they exist in circulation primarily bound to Argonaute-containing protein complexes, the relative abundance of each miRNA phase within a sample can vary biologically, be influenced by specimen processing conditions, and/or be differentially detected. It is therefore necessary to address whether distinguishing mature miRNAs from precursor forms is feasible in study design. For example, some extraction methods do not capture exosomes. Some suggest that focusing on specific carriers in study design, such as extracellular vesicles, may be a means to not only generate more reproducible miRNA profiles, but also more clinically relevant profiles. Studies suggest that exosomal miRNA expression patterns correlate with tumor characteristics and could serve well as tumor markers, however evaluation has been suboptimal due limited available methods for isolation [53]. In addition, implementation into clinical practice will not be practical until isolation methods that require less time, labor and specimen volume have been optimized [6], [51].

Specimen collection

Multiple aspects of specimen collection can influence miRNA concentrations. Simple venipuncture can contaminate the specimen with epithelial cells and traumatic phlebotomy can cause hemolysis. Current recommendations are to use, at minimum, a 22-gage needle and to discard the first 1–2 mL of blood drawn [3], [54]. Tourniquet use and differences in posture at sampling can cause hemoconcentration and should be standardized [13]. Various anticoagulants and blood stabilizers commonly used for collection of plasma and serum can also affect miRNA concentrations. A direct comparison of four different anticoagulants [EDTA, heparin, sodium citrate, or sodium fluoride/potassium oxalate (NaF/KOx)] demonstrated the most reproducible quantitation using NaF/KOx [55]. As EDTA and citrate are more commonly used in clinical practice, EDTA is recommended because citrate may trigger hemolysis [5]. Heparin should generally be avoided due to its ability to inhibit enzymes used in amplification-based quantitation assays. If, however, specimens have already been collected in heparin tubes, at least one study demonstrated that adequate treatment with heparinase facilitated miRNA detection [5].

Overall, efforts should be made during collection and processing to minimize hemolysis, release of miRNAs from platelets, and contamination from skin or WBCs to eliminate potential systematic bias from blood cell contamination.

Specimen composition

Blood cell counts, plasma volume, and plasma components can significantly alter the profile of circulating miRNAs [2]. Multiple studies have correlated blood cell counts and measured miRNA concentrations, even without hemolysis present [19], [30], [52]. It is therefore important to minimize in vitro hemolysis and blood cell contamination and also to document the extent of blood cell contamination in each specimen.

For study of miRNAs that are know to be expressed in blood cells, measurement of a concurrent CBC should be considered to allow for appropriate interpretation of the miRNA levels and potential exclusion of patients with blood cell counts outside of a predefined range (Table 2) [19]. For all specimens a visual, spectrophotometric, or alternative hemolysis cutoff should be established in the specimen processing inclusion criteria for circulating miRNA studies (Table 2). Based on variability in measured concentrations of miR-451 and miR-16 (both highly expressed in red blood cells), some authors suggest measuring free hemoglobin at an absorbance of 414 nm with a cutoff of 0.2 or higher to identify hemolyzed specimens [56]. Since lipemia can falsely elevate absorbance at 414 nm, additional spectrophotometric assessment at 385 nm can be a lipemia indicator [5]. In cases where only purified RNA is available, a ratio of hemolysis-affected miRNAs to non-hemolysis affected miRNAs can determine the likelihood of erythrocyte miRNA contamination in the original specimen. For example, a delta quantification cycle value (Cq) (miR-23amiR-451) of more than five can indicate possible red cell contamination whereas a delta Cq of >7 indicates a high risk of hemolysis [28]. Although it would be ideal to completely exclude all hemolyzed specimens, this may not be feasible or practical. A modified workflow for hemolyzed specimens has been proposed that excludes hemolyzed specimens from initial biomarker discovery, but that then conditionally allows for their inclusion in the final analysis if the miRNAs of interest are not affected by hemolysis [5].

The evaluation of hemolysis alone, however, is inadequate to account for blood cell-derived changes in miRNA concentrations. A recent study demonstrated hemolysis-independent release of primarily vesicle-associated blood cell miRNAs miR-16 and miR-21. This release occurred during the first 5 h of room temperature storage of whole blood, prior to separation of plasma or serum from cellular components [52]. Notably, increased concentrations were absent for miRNAs not associated with blood cell expression, such as liver-specific miR-122. Thus, there is a need for meticulous specimen processing and storage documentation as well as evaluation of blood cell associated miRNAs in each specimen.

Another underappreciated variable that can affect miRNA quantification is plasma volume. Plasma inherently contains polymerase inhibitors such as hemoglobin, lactoferrin, and IgG, which can co-purify with nucleic acids. A comprehensive analysis demonstrated that optimizing plasma volume is critical for accurate quantification: too little plasma will have insufficient miRNA for quantification and too much plasma can increase polymerase inhibitors. One study suggested 50 μL of serum or plasma optimized miRNA detection using SYBR Green or TaqMan qRT-PCR (11-fold and three-fold, respectively) compared to 200 and 10 μL [55]. Another study demonstrated better recovery with 200 μL of plasma compared to higher volumes of up to 500 μL [37]. Whether putative polymerase inhibitors interfere with miRNA detection depends on multiple factors including both extraction and quantification methods. Additional processing, such as enriching small RNAs and silica adsorption can be used to effectively remove inhibitors [55]. Depending on miRNA detection methods, a combination of polymerases resistant to blood-borne inhibitors could be an alternative workaround.

Specimen processing

Blood sample processing requires centrifugation to remove cellular components. Although plasma is considered the cell-free fluid portion of blood, it typically contains residual platelets and microparticles. It is essential to recognize that even trace amounts of contaminating platelets or microparticles will artificially increase miRNAs, and potentially obscure disease-related miRNA expression profiles because cellular miRNA concentrations are so much higher than fluid concentrations. Centrifugation speed can significantly impact miRNA concentrations either by inadequately removing cellular components (slow spin) or by causing cell lysis (fast spin). The influence of platelets and microparticles on miRNAs in matched fresh plasma and serum was systematically evaluated by measuring miRNAs after two-step centrifugation (slower 1940 g with no brake for 10 min followed by faster 3400 g with high brake for 10 min) and 0.22 μm filtration [30]. Significant differences were identified in 72% of the measured miRNAs due to processing (4× to >1000× variation in expression). Notably, the miRNAs most affected were those with the highest expression in platelets. Another study examined the effects of two-step centrifugation (1000 g followed by either 1000 g, 2000 g, or 10,000 g) and found that much faster spins of 10,000 g substantially reduced platelet-associated miRNAs [36]. A separate study showed that two-step centrifugation effectively removed platelets from frozen plasma and serum specimens stored for up to 6 years [30]. Incorporation of an additional centrifugation step during specimen processing is highly recommended to remove confounding platelet miRNAs [3], [5], [30]. Alternatively, filtration can remove platelets and cell debris. Although platelet-derived miRNAs accounted for the majority of differences observed, filtration additionally removed microparticle miRNAs [30]. The use of filtration should be considered based on the relevance of microparticle miRNAs to the study objectives. A post-processing platelet count with rejection of specimens above a predefined threshold can also be incorporated to further improve standardization [30].

In laboratory medicine, it is well established that delayed processing can cause significant changes in many measured analytes [13]. It is therefore not unexpected that delayed specimen processing affects miRNAs. For example, a delay in centrifugation of 6 h compared to 2 h caused significant variation in six miRNA profiles, particularly miR-15b and miR-191 [36]. An additional study investigating the effect of delayed processing showed an initial selective increase of vesicle-associated miRNAs at 1–3 h, followed by a steady decline in all miRNAs measured up to 24 h [52]. They also demonstrated that the decrease in miRNA concentrations could be ameliorated by the addition of an RNaseA inhibitor. These studies illustrate the need for rapid specimen processing that may include freezing separated plasma/serum or using a stabilizing agent such as an RNase inhibitor to maximize miRNA yield.

Specimen storage

Circulating miRNAs are typically preserved by storage at temperatures low enough to significantly reduce RNase activity rather than by chemical fixation. Fortunately, studies to date suggest that samples can be stored as processed/separated plasma/serum for at least 24 h either at room temperature or 4 °C [2], [33], [37], [47]. Furthermore, miRNAs are relatively stable and can be stored for at least 1 year at –20 °C or –70/80 °C [37]. A recent study compared multiple storage conditions on miR-134 and miR-346 concentrations in whole blood specimens measured by qRT-PCR [57]. There was no significant change due to processing delay, storage conditions, or storage duration; however, increased freeze-thaw cycles significantly decreased miRNA concentrations. Another storage study at −80 °C and −20 °C showed good stability of miRNAs for up to 6 years [58]. Interestingly, they found overall similar amounts of miRNAs when comparing storage at −80 °C and −20 °C but differential expression of miRNAs depending on the storage temperature, suggesting that storage temperatures should be consistent throughout a study. A separate study demonstrated similar miRNA profiles and expression following storage at −80 °C for 12 years [36]. Since there is minimal and occasionally conflicting data about the effects of long-term storage (multiple years), control specimens matched for storage time should be considered [35].

Freeze-thaw cycles can potentially affect multiple analytes and conflicting results have been reported on the impact of freeze-thaw cycles on miRNAs [47], [57], [58], [59]. Thus, minimizing freeze-thaw cycles is recommended.

Pre-analytical variables for tissue-based analysis of miRNAs

Human tissue, most often fixed in formalin and embedded in paraffin (FFPE), is the cornerstone of diagnostic pathology. Pathology laboratories archive clinical tissue samples as FFPE “blocks” and these stored specimens are an invaluable resource for basic, translational, and clinical science. Although not routinely used for clinical diagnostic purposes, analysis of miRNAs in such specimens is increasingly common, with the hope that miRNA signatures will supplement morphology, along with protein expression, transcriptional profiles, and DNA sequence analysis as important adjuncts for clinicians and anatomic pathologists. Analysis in situ provides important advantages, including direct association of miRNA signatures with histologically confirmed disease process. Thus, analysis of miRNAs in tissue not only may discover signatures associated with specific disease states, particularly solid neoplasms, but is also a necessary step to validate miRNA species identified as diagnostic, predictive, or prognostic markers.

Two themes related to pre-analytical variables emerge from review of the tissue miRNA literature. First, miRNAs are robust, and, when compared to other nucleic acids, better tolerate both direct and tissue insults. For example, various markers of total RNA integrity including electropherograms [55], [56], [59], [60], [61] and RNA integrity number (RIN) [57], [61] do not correlate with miRNA integrity, which consistently remains detectable despite overall specimen decay. Second, while standardized pre-analytical conditions for tissue samples are ideal, it is not always practical and variation in the above variables should be recorded and analyzed as potential confounders.

Ischemic time after sample procurement

A notable variable in clinical tissue samples is time the tissue spends at either room temperature or at 4 °C following excision and the resulting loss of blood supply prior to freezing or fixing. The warm and cold ischemic time, respectively, vary significantly from sample to sample and both have profound and deleterious effects on downstream recovery of nucleic acids and protein antigens [62], [63]. In addition, hypoxic/ischemic injury is a major biological stress that can induce significant transcriptional changes; the degree to which hypoxia alters miRNA expression profiles is an open area of research. The degree of ischemic effects, particularly tissue autolysis, varies significantly with tissue type. For example, pancreatic tissue is particularly sensitive given the high concentration of intracellular peptidases [64]. Unfortunately, the ischemic time is not readily controllable for most clinical samples and is generally not routinely recorded. Few studies have systematically examined the effect of ischemic time on miRNA recovery, further confounding attempts to establish best practices.

A recent study carefully assessed the effects of ischemic time preceding fixation or freezing on miRNA expression of autopsy cardiac tissue following myocardial infarct (also known as the post mortem interval) [60]. In this study, the post mortem interval ranged from 10 h to 154 h; cadavers were stored at room temperature for 3 h–24 h prior to cold storage at 4 °C. High quality miRNAs were detectable from cardiac tissue up to a week following death and the concentration of miRNAs varied less than that of other small RNA species. Despite the ability of miRNAs to withstand harsh treatment, the authors suggest prompt transfer to 4 °C to better preserve miRNA integrity. They further note that electrophoretic profiling of total RNA likely underestimates the population of intact miRNA since longer RNA species are more readily degraded than shorter molecules, which can be visualized as a smear on gel electrophoresis.

Multiple studies have documented changes in miRNA expression profiles following even short ischemic events. One study demonstrated time-dependent changes in the expression of 56 miRNAs and 1788 mRNAs during warm ischemic time [65]. The most prominent effects (particularly for miRNAs) occurred after 1 h of warm ischemic time; thus, the authors recommend freezing tissue at 30 min of warm ischemic time [65]. A more recent study using a gerbil model of cerebral ischemia examined the effects of 2 min carotid artery occlusion on miRNA expression in the hippocampus [66]. They identified seven miRNAs upregulated following transient carotid artery ligation and concluded that these changes are a neuroprotective mechanism against ischemic injury.

Although miRNAs tend to remain stable after tissue is removed from a patient, ischemic time can nonetheless potentially change miRNA expression or, to a lesser degree, cause sample degradation. Confounding effects are that time dependent and warm ischemia are more potent than cold ischemia. Uncertainty about total ischemic time is of particular concern in retrospective analysis of archived tissue samples, whereas prospective studies may incorporate time and/or recording standards. In addition to recording and recognizing warm and cold ischemic times as sources of variability, we offer specific recommendations for prospective studies in Table 3. For studies of archived tissue, we recommend collecting ischemic time if available. If unavailable, a common miRNA control whose expression is not affected by ischemia is critical.

Table 3:

Tissue-specific best practice recommendations.

– Ischemic time <1 h at room temperature and <24 h at 4 °C
– Common fixative for all specimens
– Common internal control miRNA for different specimens (i.e. paired formalin-fixed and frozen)
– Formalin fixation times common in clinical practice (<72 h, often <24 h) are unlikely to have significant confounding effects
– Select regions of interest for enrichment/focused analysis
– Validate candidate miRNA is present in tissue type by in situ visualization if individual cells were not carefully selected for in discovery process

Fixative, fixation time and tissue storage

For clinical samples, neutral buffered formalin is the fixative of choice for optimal evaluation by pathologists. Fixation is a biochemically complex process, with significant variation over time, temperature, and tissue thickness and has notable implications for tissue histology, antigen and nucleic acid recovery [61], [62], [63]. Tissue samples may also be frozen without fixative or occasionally fixed in an alternative agent for a specific purpose (e.g. immunofluorescence or electron microscopy). Following fixation, specimens are “processed” and undergo a series of buffer exchanges through alcohols of increasing hydrophobicity, xylene, and ultimately paraffin wax (or less commonly plastic). The resulting FFPE “tissue blocks” are stored for many years, serving as reference material in the case of recurrent disease and as biospecimens for research. Therefore the choice of fixative, effects of freezing, and the total fixation time prior to processing are additional variables worth considering.

Routine FFPE specimens vs. frozen specimens

Multiple studies have compared recovery of miRNAs from FFPE tissue [60], [61], [67], [68]. Early work demonstrated high concordance between fresh frozen and FFPE tissue samples with correlation coefficients (r2) of 0.86–0.89 [67]. Similarly, more recent work compared miRNAs recovered from cardiac tissue sampled at autopsy following a standardized protocol with paired samples that were frozen or fixed [60]. Despite different extraction methods, there was close concordance between the concentrations of five candidate miRNAs: miR-1, miR-26b, miR-191, miR-208b, and miR-499a extracted from frozen and FFPE tissue within the first 3 months of storage.

This has also been investigated in matched samples of breast tissue that were cryopreserved, cryopreserved with a significant freeze-thaw cycle, or FFPE samples analyzed for multiple miRNAs as well as small nucleolar RNA and mRNA species [61]. The miRNAs were detected at nearly identical concentrations across all conditions. In contrast, the small nucleolar and mRNAs were closely correlated in only freeze-thawed and FFPE specimens, highlighting the propensity for fixation and tissue damage to degrade most nucleic acids [61]. One study limitation is that miRNAs were extracted with different methods for frozen and FFPE tissue, as is the case for other studies [60], [61], [69]. A separate study demonstrated similar findings for two miRNAs in snap frozen and FFPE specimens from multiple mouse and human tissue specimens [68]. They reported strong concordance between miR-122 in FFPE and frozen specimens from multiple mouse tissues. In human samples, miR-16 concentrations were more variable, but the difference was not statistically significant. The study did not address whether this variability could have been related to contaminating blood cells. In contrast to these reports, a recent study observed decreased global mean miRNA expression with formalin fixation as compared to paired frozen specimens by qRT-PCR [40]. This effect grew stronger as fixation time increased to 2, 3, and 6 days. For tissue fixed for 2 days, the decrease in global mean miRNA expression was similar to the standard deviation of miRNA expression across five biological replicates (±0.52 Cq).

Multiple studies emphasize caution in using either electropherograms of total RNA or RIN in predicting the quality of miRNA isolated [60], [61], [70]. Although these metrics show apparent degradation following formalin fixation or other tissue insults, such as freeze-thaw events, miRNAs are readily detected without significant variation from well-preserved, snap-frozen tissue [61].

These findings demonstrate that miRNAs are consistently detectable regardless of immediate freezing or formalin-fixing tissue, provided the formalin fixation prior to embedding in paraffin is kept short (<2–3 days). Small variations between FFPE and frozen tissue may exist for specific miRNAs. Storage time may account for some of this variation (described below). In addition, different RNA extraction methods for fixed versus frozen tissue may account in part for these differences. The variation, although slight, emphasizes the importance of validation for each miRNA species when comparing tissues preserved through different means.

Effects of different fixatives on miRNA expression levels

Several studies have examined the effects of different fixatives on miRNA stability and recovery. For example, different fixation protocols impact the relative expression of miR-122 in liver tissue [68]. Tissue was fixed in neutral buffered formalin, formalin at pH 3 (to simulate decalcification conditions), and ethanol-formalin mixture (Schaefer solution) prior to measurement of miR-122. Consistently lower miR-122 concentrations were detected in tissue fixed in formalin at pH 3 versus neutral buffered formalin, while higher miR-122 concentrations were detected in tissue fixed in ethanol-formalin. A second study aimed to identify miRNA markers of mesothelioma and studied mesothelial cell lines, cytology specimens, and malignant mesothelioma tissue specimens [69]. The cell lines and cytology specimens were ethanol fixed, whereas the tissue specimens were formalin fixed. The authors found limited concordance in miRNA concentrations for a subset of five examined candidate miRNAs. Caution should thus be used in comparing samples exposed to different fixatives, and care should be taken to be consistent in study design.

Formalin exposure and long-term storage

Longer ribonucleic acids such as mRNAs degrade as a function of time in formalin [62], [68], [71]. In contrast, miRNAs appear to tolerate a wide range of fixation times [60], [61], [68]. One study demonstrated consistent recovery of miRNAs from autopsy samples fixed for up to 3 months in neutral buffered formalin [60]. These results corroborate earlier work that reported similar miRNA relative expression from tissue fixed in neutral buffered formalin from 12 to 72 h, and typical fixation times during routine clinical practice [68].

It is possible to recover miRNAs from FFPE stored for many years, potentially several decades. Multiple studies have demonstrated robust isolation of miRNAs from archived FFPE tissue stored for <8 years [61], [68], [72]. An early study examined 88 samples and demonstrated that miRNA expression recovered from archived FFPE tissue blocks decreased with time, particularly after storage of tissue for >7–8 years [68]. Surprisingly, miRNAs were detectable in blocks stored for as long as 28 years [68]. These findings were reproduced for two miRNAs in both human and mouse specimens: miR-122 from liver tissue and miR-16 in diverse tissue types, including bowel, bone marrow, liver, lymph node, and breast tissue [68]. A much larger study suggested the reduction in miRNA concentration over time may not be a significant problem [72]. For six miRNAs examined in 345 colorectal adenocarcinoma FFPE specimens stored 6–28 years, these miRNAs were detectable at statistically similar expression levels relative to an endogenous reference small RNA across all time points.

Taken together, these studies illustrate that miRNAs are resilient and sufficiently robust to withstand routine clinical processing and storage conditions; however, standardized formalin fixation times are preferred [60], [61], [68], [70] The data suggest that variation in formalin fixation times on the scale routinely practiced in clinical laboratories (up to several days) likely do not significantly impact miRNA quantity or quality. Nonetheless, we caution that the ratio of formalin to tissue, tissue thickness, and temperature additionally impact the fixation process and prospective clinical research protocols should attempt to standardize these variables. For miRNA analysis in archived FFPE tissue, it is important to maintain a standard control miRNA for determining relative expression and to recognize that miRNA concentrations may decrease over time, especially for samples stored longer than 7 years.

Specimen processing

Once specimens are fixed and submitted for histology, they are “processed” through a series of incubations in increasingly hydrophobic solutions (alcohols) and finally into xylene prior to being embedded in paraffin. Larger resections are typically processed for 10 h, while biopsies and rush specimens may go through a 2 or 4 h processing time. The question of whether different processing times/protocols influences miRNA recovery or downstream analysis has not been rigorously studied. Given the general robustness of miRNAs following formalin fixation, we hypothesize that a large difference is unlikely, but this remains a potential confounding variable. Thus, we reiterate the importance of standardizing sample types and selection of an appropriate control miRNA species.

Tissue variability

An clear advantage of tissue-based miRNA analysis is the ability to define specific cell populations for miRNA analysis and confirm the biological of interest. However, tissue sections are inherently heterogenous with widely variable amounts of pathologic tissue. Thus, careful cell population selection is a vital pre-analytical variable in tissue-based miRNA analysis. Further, tissue-specificity of miRNAs is an active area of research that requires correlation with histomorphology or cell lineage markers. If tissue is analyzed without careful selection of cell population, study results may be unduly biased due to varying density of the cells of interest admixing with other cell types. Companion histological analysis by an experienced pathologist is thus recommended prior to miRNA tissue analysis. A routine hematoxylin & eosin (H&E) stained section allows rapid confirmation/identification of diseased tissue, reactive tissue around the disease process including the presence of blood elements, and potential contaminating tissue from a different specimen.

Selected cell populations of interest

In addition to cells of interest, neoplastic tissue sections invariably contain reactive cells as well as uninvolved normal tissue. For tissue-based miRNA analysis, FFPE blocks are selected based on arbitrary concentrations of diseased tissue. Thresholds are variable and may be qualitative [49] or semi-quantitative, ranging from at least 70%–80% neoplastic cells [73], [74] to as low as >30% neoplastic cells [75]. Tissue sections of neoplasms are complex and contain multiple cell types such that an estimate of 80% diseased cells likely includes a substantial population of stromal/mesenchymal cells, including activated stromal cells responding to the disease process. Contamination has the potential to severely bias results, as demonstrated by recent work where two miRNAs, miR-143/145, proposed to function as colorectal adenocarcinoma tumor suppressors, are not expressed by colonic epithelial cells at all [76]. Rather, the authors discovered that mesenchymal cells (fibroblasts and smooth muscle cells) express these miRNAs, which are expressed 33-fold less in non-neoplastic epithelial cells.

Several solutions to this sampling problem are regularly employed to enrich for cells of interest. One approach is to macrodissect regions containing apparently homogenous populations of the cell type of interest based on histopathological evaluation of H&E stained slides [77]. A second approach adopted in multiple studies is to use laser capture microdissection to isolate cells of interest [69], [78] Additionally, flow cytometry has been successfully used to select specific cell subtypes, such as epithelial cells, in samples of dissociated, intact cells [76]. Finally, post hoc validation can be performed by detection of candidate miRNAs of interest in neoplastic cells by fluorescence in situ hybridization (FISH) [79], [80].

It must be noted that there could be biological significance to altered miRNA profiles in reactive/adjacent cells as neighboring cells may have profound roles in the pathologic process; however, we stress that determining the cells from which the miRNA signal originates is a critical factor. We emphasize that each of the above validation/selection mechanisms requires histopathologic examination to confirm the cell population examined and that while the mechanism to select/validate the cells studied may vary by disease process and available tissue, it should be explicitly stated. Finally, we suggest that erratic, suspect, or unexpected results be analyzed to confirm whether the miRNA signals detected originate with pathologic cells or bystander/reactive cells.

Contamination by blood components

As discussed above, multiple blood components can confound miRNA analysis [29], [30], underscoring the importance of carefully selecting regions of interest by macro- or microdissection and, ideally, determining what cell types express the candidate miRNA(s) by in situ visualization. Both tissue resident immune cells and blood components are always present in tissue samples, sometimes with prognostic significance (i.e. tumor-infiltrating lymphocytes in colonic adenocarcinoma and melanoma) [81], [82], [83], [84] The challenge of avoiding confounding cells from any hematopoietic lineage may be particularly acute in disease processes with a robust immunological response, bloody specimens, friable tissue, highly vascularized tissue, or neoplasms with a prominent vascular component.

Variation in miRNA extraction

As previously reviewed and discussed above, multiple miRNA extraction protocols are available but do not always yield identical results [2]. Unfortunately, many studies extract miRNA from tissue using different methods depending upon the specimen type (i.e. FFPE tissue, cytology specimens, cell culture, or frozen tissue) [60], [61], [68], [69], [74]. A full review of the performance of different miRNA isolation techniques is beyond the scope. However, we emphasize that this is a notable and frequently ill-considered pre-analytical variable, particularly when specimen type is changed simultaneously.

Pre-analytical variables associated with in situ analysis

As highlighted above, it is often both informative and a helpful ancillary validation step to correlate miRNA expression with specific cell types through dissection, FISH/chromogenic in situ hybridization, or immunohistochemical (IHC) phenotype of cells from which RNA is extracted. Already, miRNAs are being advocated to distinguish diseased (typically neoplastic) tissue or characterize cell differentiation. For example, recent work has suggested that miR-211 may be aid in distinguishing invasive melanoma from dysplastic nevi, a potentially challenging diagnostic decision in anatomic pathology [85]. Whether for biomarker discovery or clinical diagnostic assays, such in situ techniques are becoming more commonplace and several important pre-analytic variables are worth noting.

For laser capture microdissection, one recent study reported enhanced miRNA yields from microdissected tissues by using crystal violet for histologic staining (as opposed to the more routine H&E), silicon carbide (SiC) matrix in RNA-binding columns, and overnight treatment with proteinase K [86]. Storing stained slides at room temperature prior to laser capture microdissection for up to 7 days did not adversely impact miRNA recovery, nor did storage at room temperature for up to a day between dissection and RNA extraction. These findings may simplify workflow in the laboratory.

IHC staining is frequently performed for pathological classification and is useful in identifying specific cells for microdissection. One study demonstrated sensitive quantitation of miRNAs by qRT-PCR in as few as 20 cells isolated from FFPE tissue and tested the impact of temperature in IHC staining protocols on miRNA recovery [78]. Relative expression of five miRNAs analyzed within each specimen remained consistent regardless of whether the IHC protocol avoided heating the tissue (hemalaun) or required heating to 98 °C or 90 °C.

Detection of miRNAs by FISH poses several technical challenges, including diffusion of miRNAs out of tissue and suboptimal hybridization efficiency. It is feasible and potentially useful to perform multicolor miRNA FISH and several solutions to these challenges have been demonstrated [79], [87]. To minimize diffusion of miRNAs, tissue sections may be incubated with 1-ethyl-3-[3-dimethylaminopropyl] carbodiimide (EDC) during or post-fixation [79], [87]. A longer linker region between a fluorophore-binding hapten (biotin) and a target-specific locked nucleic acid (LNA) probe has been demonstrated to improve probe-target hybridization efficiency [79]. Multiple mechanisms have been identified to boost fluorescence signal, particularly tyramide amplification [79], [87]. In addition, not all FISH protocols are amenable to subsequent or concurrent IHC [79], [87]. Methods aimed at combined detection of miRNAs and protein targets are an area of active research [87].

Conclusions

Successful biomarker discovery requires careful consideration of pre-analytical variation (see Figure 1). Ideally the effect of pre-analytical factors such as sample type, assay choice, individual variation, stability, and hemolysis should be experimentally determined for each candidate miRNA to assure that any observed differences between control and disease groups exceeds the range of observed assay variances [55]. When this is not possible, the use of standard operating procedures and meticulous documentation is critical to minimize the impact of controllable and uncontrollable confounding factors. We have outlined some practical best practice standards for incorporation into study design to improve the consistency of generated results (see Tables 13). In addition to improved study design, detailed documentation of the pre-analytic variables specific to each study and inclusion of this information in publications is critical for comprehensive comparison and evaluation of miRNA literature. Some have even suggested that scientific journals should improve standardization of scientific data reporting by clearly defining and requiring specific parameters in the “Methods” sections [9]. Overall, developing and documenting standard procedures related to pre-analytic variables is fundamental to biomarker discovery. Collection of relevant demographic, lifestyle, and clinical/laboratory data, as well as internal consistency within study design, are critical to minimize bias from pre-analytic variables. Although there has been a focus in the literature on discovery of new miRNA biomarkers, until there is a better understanding of cell specific physiology associated with preanalytic variables, the ability to interpret the clinical significance of miRNA profiles will be limited.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Pritchard CC, Cheng HH, Tewari M. MicroRNA profiling: approaches and considerations. Nat Rev Genet 2012;13:358–69.10.1038/nrg3198Search in Google Scholar PubMed PubMed Central

2. Becker N, Lockwood CM. Pre-analytical variables in miRNA analysis. Clin Biochem 2013;46:861–8.10.1016/j.clinbiochem.2013.02.015Search in Google Scholar PubMed

3. Nair VS, Pritchard CC, Tewari M, Ioannidis JP. Design and analysis for studying microRNAs in human disease: a primer on -Omic technologies. Am J Epidemiol 2014;180:140–52.10.1093/aje/kwu135Search in Google Scholar PubMed PubMed Central

4. Jarry J, Schadendorf D, Greenwood C, Spatz A, van Kempen LC. The validity of circulating microRNAs in oncology: five years of challenges and contradictions. Mol Oncol 2014;8:819–29.10.1016/j.molonc.2014.02.009Search in Google Scholar PubMed PubMed Central

5. Tiberio P, Callari M, Angeloni V, Daidone MG, Appierto V. Challenges in using circulating miRNAs as cancer biomarkers. BioMed Res Int 2015;2015:731479.10.1155/2015/731479Search in Google Scholar PubMed PubMed Central

6. He Y, Lin J, Kong D, Huang M, Xu C, Kim T-K, et al. Current state of circulating microRNAs as cancer biomarkers. Clin Chem Lab Med 2015;61:1138–55.10.1373/clinchem.2015.241190Search in Google Scholar PubMed

7. Haider BA, Baras AS, McCall MN, Hertel JA, Cornish TC, Halushka MK. A critical evaluation of microRNA biomarkers in non-neoplastic disease. PLoS One 2014;9:e89565.10.1371/journal.pone.0089565Search in Google Scholar PubMed PubMed Central

8. Butz H, Patócs A. Technical aspects related to the analysis of circulating microRNAs. In: Igaz P, editor. Circulating microRNAs in Disease Diagnostics and their Potential Biological Relevance. Switzerland: Springer Basel, 2015:55–71.10.1007/978-3-0348-0955-9_3Search in Google Scholar PubMed

9. Ono S, Lam S, Nagahara M, Hoon DS. Circulating microRNA biomarkers as liquid biopsy for cancer patients: pros and cons of current assays. J Clin Med 2015;4:1890–907.10.3390/jcm4101890Search in Google Scholar PubMed PubMed Central

10. Bonini P, Plebani M, Ceriotti F, Rubboli F. Errors in laboratory medicine. Clin Chem Lab Med 2002;48:691–8.10.1093/clinchem/48.5.691Search in Google Scholar

11. Lippi G, Guidi GC, Mattiuzzi C, Plebani M. Preanalytical variability: the dark side of the moon in laboratory testing. Clin Chem Lab Med 2006;44:358–65.10.1515/CCLM.2006.073Search in Google Scholar PubMed

12. Ottomano C. Errors in medicine and errors in laboratory medicine: what is the difference? Blood Transfus Trasfus Sangue 2010;8:79–81.Search in Google Scholar

13. Ellervik C, Vaught J. Preanalytical variables affecting the integrity of human biospecimens in biobanking. Clin Chem Lab Med 2015;61:914–34.10.1373/clinchem.2014.228783Search in Google Scholar PubMed

14. Vaught J, Lockhart N. The evolution of biobanking best practices. Clin Chim Acta Int J Clin Chem 2012;413:1569–75.10.1016/j.cca.2012.04.030Search in Google Scholar PubMed PubMed Central

15. Robb JA, Gulley ML, Fitzgibbons PL, Kennedy MF, Cosentino LM, Washington K, et al. A call to standardize preanalytic data elements for biospecimens. Arch Pathol Lab Med 2014;138:526–37.10.5858/arpa.2013-0250-CPSearch in Google Scholar PubMed PubMed Central

16. Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. Aging differentially affects human skeletal muscle microRNA expression at rest and after an anabolic stimulus of resistance exercise and essential amino acids. Am J Physiol – Endocrinol Metab 2008;295:E1333–40.10.1152/ajpendo.90562.2008Search in Google Scholar PubMed PubMed Central

17. Te JL, Dozmorov IM, Guthridge JM, Nguyen KL, Cavett JW, Kelly JA, et al. Identification of unique MicroRNA signature associated with lupus nephritis. PLoS One 2010;5. doi: 10.1371/journal.pone.0010344.10.1371/journal.pone.0010344Search in Google Scholar PubMed PubMed Central

18. Chen F, Zhang W, Liang Y, Huang J, Li K, Green CD, et al. Transcriptome and network changes in climbers at extreme altitudes. PLoS One 2012;7:e31645.10.1371/journal.pone.0031645Search in Google Scholar PubMed PubMed Central

19. Pritchard CC, Kroh E, Wood B, Arroyo JD, Dougherty KJ, Miyaji MM, et al. Blood cell origin of circulating micrornas: a cautionary note for cancer biomarker studies. Cancer Prev Res (Phila Pa) 2012;5:492–7.10.1158/1940-6207.CAPR-11-0370Search in Google Scholar PubMed PubMed Central

20. Tan GW, Khoo AS, Tan LP. Evaluation of extraction kits and RT-qPCR systems adapted to high-throughput platform for circulating miRNAs. Sci Rep 2015;5:9430.10.1038/srep09430Search in Google Scholar PubMed PubMed Central

21. Ameling S, Kacprowski T, Chilukoti RK, Malsch C, Liebscher V, Suhre K, et al. Associations of circulating plasma microRNAs with age, body mass index and sex in a population-based study. BMC Med Genomics 2015;8:61.10.1186/s12920-015-0136-7Search in Google Scholar PubMed PubMed Central

22. Wang K, Yuan Y, Cho J-H, McClarty S, Baxter D, Galas DJ. Comparing the microRNA spectrum between serum and plasma. PLoS One 2012;7. doi: 10.1371/journal.pone.0041561.10.1371/journal.pone.0041561Search in Google Scholar PubMed PubMed Central

23. Flowers E, Won GY, Fukuoka Y. MicroRNAs associated with exercise and diet: a systematic review. Physiol Genomics 2015;47:1–11.10.1152/physiolgenomics.00095.2014Search in Google Scholar PubMed PubMed Central

24. Gontero P, Marra G, Soria F, Oderda M, Zitella A, Baratta F, et al. A randomized double-blind placebo controlled phase I–II study on clinical and molecular effects of dietary supplements in men with precancerous prostatic lesions. Chemoprevention or “chemopromotion”? Prostate 2015;75:1177–86.Search in Google Scholar

25. de Boer HC, van Solingen C, Prins J, Duijs JM, Huisman MV, Rabelink TJ, et al. Aspirin treatment hampers the use of plasma microRNA-126 as a biomarker for the progression of vascular disease. Eur Heart J 2013;34:3451–7.10.1093/eurheartj/eht007Search in Google Scholar PubMed

26. Takahashi K, Yokota S, Tatsumi N, Fukami T, Yokoi T, Nakajima M. Cigarette smoking substantially alters plasma microRNA profiles in healthy subjects. Toxicol Appl Pharmacol 2013;272:154–60.10.1016/j.taap.2013.05.018Search in Google Scholar PubMed

27. Witwer KW. XenomiRs and miRNA homeostasis in health and disease. RNA Biol 2012;9:1147–54.10.4161/rna.21619Search in Google Scholar PubMed PubMed Central

28. Blondal T, Jensby Nielsen S, Baker A, Andreasen D, Mouritzen P, Wrang Teilum M, et al. Assessing sample and miRNA profile quality in serum and plasma or other biofluids. Methods 2013;59:S1–6.10.1016/j.ymeth.2012.09.015Search in Google Scholar PubMed

29. Duttagupta R, Jiang R, Gollub J, Getts RC, Jones KW. Impact of cellular miRNAs on circulating miRNA biomarker signatures. PLoS One 2011;6. doi: 10.1371/journal.pone.0020769.10.1371/journal.pone.0020769Search in Google Scholar PubMed PubMed Central

30. Cheng HH, Yi HS, Kim Y, Kroh EM, Chien JW, Eaton KD, et al. Plasma processing conditions substantially influence circulating microRNA biomarker levels. PLoS One 2013;8:e64795. doi: 10.1371/journal.pone.0064795.10.1371/journal.pone.0064795Search in Google Scholar PubMed PubMed Central

31. Kirschner MB, Edelman JJ, Kao SC, Vallely MP, van Zandwijk N, Reid G. The impact of hemolysis on cell-free microRNA biomarkers. Front Genet 2013;4:94.10.3389/fgene.2013.00094Search in Google Scholar PubMed PubMed Central

32. Wang Y, Cai Y. A survey on database resources for microRNA–disease relationships. Brief Funct Genomics 2016. doi: 10.1093/bfgp/elw015. [Epub ahead of print].10.1093/bfgp/elw015Search in Google Scholar PubMed

33. McDonald JS, Milosevic D, Reddi HV, Grebe SK, Algeciras-Schimnich A. Analysis of circulating microRNA: preanalytical and analytical challenges. Clin Chem Lab Med 2011;57:833–40.10.1373/clinchem.2010.157198Search in Google Scholar PubMed

34. Moret I, Sánchez-Izquierdo D, Iborra M, Tortosa L, Navarro-Puche A, Nos P, et al. Assessing an improved protocol for plasma microRNA extraction. PLoS One 2013;8. doi: 10.1371/journal.pone.0082753.10.1371/journal.pone.0082753Search in Google Scholar PubMed PubMed Central

35. Kroh EM, Parkin RK, Mitchell PS, Tewari M. Analysis of circulating microRNA biomarkers in plasma and serum using quantitative reverse transcription-PCR (qRT-PCR). Methods 2010;50:298–301.10.1016/j.ymeth.2010.01.032Search in Google Scholar PubMed PubMed Central

36. Page K, Guttery DS, Zahra N, Primrose L, Elshaw SR, Pringle JH, et al. Influence of plasma processing on recovery and analysis of circulating nucleic acids. PLoS One 2013;8. doi: 10.1371/journal.pone.0077963.10.1371/journal.pone.0077963Search in Google Scholar PubMed PubMed Central

37. Sourvinou IS, Markou A, Lianidou ES. Quantification of circulating miRNAs in plasma: effect of preanalytical and analytical parameters on their isolation and stability. J Mol Diagn 2013;15:827–34.10.1016/j.jmoldx.2013.07.005Search in Google Scholar PubMed

38. Mestdagh P, Hartmann N, Baeriswyl L, Andreasen D, Bernard N, Chen C, et al. Evaluation of quantitative miRNA expression platforms in the microRNA quality control (miRQC) study. Nat Methods 2014;11:809–15.10.1038/nmeth.3014Search in Google Scholar PubMed

39. Bianchi F, Nicassio F, Marzi M, Belloni E, Dall’olio V, Bernard L, et al. A serum circulating miRNA diagnostic test to identify asymptomatic high-risk individuals with early stage lung cancer. EMBO Mol Med 2011;3:495–503.10.1002/emmm.201100154Search in Google Scholar PubMed PubMed Central

40. Boisen MK, Dehlendorff C, Linnemann D, Schultz NA, Jensen BV, Høgdall EV, et al. MicroRNA expression in formalin-fixed paraffin-embedded cancer tissue: identifying reference microRNAs and variability. BMC Cancer 2015;15:1024.10.1186/s12885-015-2030-2Search in Google Scholar PubMed PubMed Central

41. Yuan T, Huang X, Woodcock M, Du M, Dittmar R, Wang Y, et al. Plasma extracellular RNA profiles in healthy and cancer patients. Sci Rep 2016;6:19413.10.1038/srep19413Search in Google Scholar PubMed PubMed Central

42. Backes C, Leidinger P, Altmann G, Wuerstle M, Meder B, Galata V, et al. Influence of next-generation sequencing and storage conditions on miRNA patterns generated from PAXgene blood. Anal Chem 2015;87:8910–6.10.1021/acs.analchem.5b02043Search in Google Scholar PubMed

43. Häntzsch M, Tolios A, Beutner F, Nagel D, Thiery J, Teupser D, et al. Comparison of whole blood RNA preservation tubes and novel generation RNA extraction kits for analysis of mRNA and miRNA profiles. PLoS One 2014;9. doi: 10.1371/journal.pone.0113298.10.1371/journal.pone.0113298Search in Google Scholar PubMed PubMed Central

44. Kruhøffer M, Dyrskjøt L, Voss T, Lindberg RL, Wyrich R, Thykjaer T, et al. Isolation of microarray-grade total RNA, microRNA, and DNA from a single PAXgene blood RNA tube. J Mol Diagn 2007;9:452–8.10.2353/jmoldx.2007.060175Search in Google Scholar PubMed PubMed Central

45. Weber DG, Casjens S, Rozynek P, Lehnert M, Zilch-Schöneweis S, Bryk O, et al. Assessment of mRNA and microRNA stabilization in peripheral human blood for multicenter studies and biobanks. Biomark Insights 2010;5:95–102.10.4137/BMI.S5522Search in Google Scholar

46. Viertler C, Groelz D, Gündisch S, Kashofer K, Reischauer B, Riegman PH, et al. A new technology for stabilization of biomolecules in tissues for combined histological and molecular analyses. J Mol Diagn 2012;14:458–66.10.1016/j.jmoldx.2012.05.002Search in Google Scholar PubMed

47. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A 2008;105:10513–8.10.1073/pnas.0804549105Search in Google Scholar PubMed PubMed Central

48. Williams Z, Ben-Dov IZ, Elias R, Mihailovic A, Brown M, Rosenwaks Z, et al. Comprehensive profiling of circulating microRNA via small RNA sequencing of cDNA libraries reveals biomarker potential and limitations. Proc Natl Acad Sci U S A 2013;110:4255–60.10.1073/pnas.1214046110Search in Google Scholar PubMed PubMed Central

49. Ali S, Dubaybo H, Brand RE, Sarkar FH. Differential expression of microRNAs in tissues and plasma co-exists as a biomarker for pancreatic cancer. J Cancer Sci Ther 2015;7:336–46.10.4172/1948-5956.1000372Search in Google Scholar PubMed PubMed Central

50. Meng QH, Wagar EA. Pseudohyperkalemia: a new twist on an old phenomenon. Crit Rev Clin Lab Sci 2015;52:45–55.10.3109/10408363.2014.966898Search in Google Scholar PubMed

51. Witwer KW. Circulating microRNA biomarker studies: pitfalls and potential solutions. Clin Chem Lab Med 2015;61:56–63.10.1373/clinchem.2014.221341Search in Google Scholar PubMed

52. Köberle V, Kakoschky B, Ibrahim AA, Schmithals C, Peveling-Oberhag J, Zeuzem S, et al. Vesicle-associated microRNAs are released from blood cells on incubation of blood samples. Transl Res 2016;169:40–6.10.1016/j.trsl.2015.10.010Search in Google Scholar PubMed

53. Thind A, Wilson C. Exosomal miRNAs as cancer biomarkers and therapeutic targets. J Extracell Vesicles 2016;5:31292.10.3402/jev.v5.31292Search in Google Scholar PubMed PubMed Central

54. Witwer KW, Buzás EI, Bemis LT, Bora A, Lässer C, Lötvall J, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles 2013;2. doi: 10.3402/jev.v2i0.20360.10.3402/jev.v2i0.20360Search in Google Scholar PubMed PubMed Central

55. Kim D-J, Linnstaedt S, Palma J, Park JC, Ntrivalas E, Kwak-Kim JY, et al. Plasma components affect accuracy of circulating cancer-related microRNA quantitation. J Mol Diagn 2012;14:71–80.10.1016/j.jmoldx.2011.09.002Search in Google Scholar PubMed PubMed Central

56. Kirschner MB, Kao SC, Edelman JJ, Armstrong NJ, Vallely MP, van Zandwijk N, et al. Haemolysis during sample preparation alters microRNA content of plasma. PLoS One 2011;6. doi: 10.1371/journal.pone.0024145.10.1371/journal.pone.0024145Search in Google Scholar PubMed PubMed Central

57. Zhao H, Shen J, Hu Q, Davis W, Medico L, Wang D, et al. Effects of preanalytic variables on circulating microRNAs in whole blood. Cancer Epidemiol Biomarkers Prev 2014;23:2643–8.10.1158/1055-9965.EPI-14-0550Search in Google Scholar PubMed PubMed Central

58. Grasedieck S, Schöler N, Bommer M, Niess JH, Tumani H, Rouhi A, et al. Impact of serum storage conditions on microRNA stability. Leukemia 2012;26:2414–6.10.1038/leu.2012.106Search in Google Scholar PubMed

59. Farina NH, Wood ME, Perrapato SD, Francklyn CS, Stein GS, Stein JL, et al. Standardizing analysis of circulating microRNA: clinical and biological relevance. J Cell Biochem 2014;115:805–11.10.1002/jcb.24745Search in Google Scholar PubMed PubMed Central

60. Kakimoto Y, Kamiguchi H, Ochiai E, Satoh F, Osawa M. MicroRNA stability in postmortem FFPE tissues: quantitative analysis using autoptic samples from acute myocardial infarction patients. PLoS One 2015;10. doi: 10.1371/journal.pone.0129338.10.1371/journal.pone.0129338Search in Google Scholar PubMed PubMed Central

61. Peiró-Chova L, Peña-Chilet M, López-Guerrero JA, García-Giménez JL, Alonso-Yuste E, Burgues O, et al. High stability of microRNAs in tissue samples of compromised quality. Virchows Arch Int J Pathol 2013;463:765–74.10.1007/s00428-013-1485-2Search in Google Scholar PubMed

62. Masuda N, Ohnishi T, Kawamoto S, Monden M, Okubo K. Analysis of chemical modification of RNA from formalin-fixed samples and optimization of molecular biology applications for such samples. Nucleic Acids Res 1999;27:4436–43.10.1093/nar/27.22.4436Search in Google Scholar PubMed PubMed Central

63. Chafin D, Theiss A, Roberts E, Borlee G, Otter M, Baird GS. Rapid two-temperature formalin fixation. PLoS One 2013;8. doi: 10.1371/journal.pone.0054138.10.1371/journal.pone.0054138.Search in Google Scholar

64. Tomita Y, Nihira M, Ohno Y, Sato S. Ultrastructural changes during in situ early postmortem autolysis in kidney, pancreas, liver, heart and skeletal muscle of rats. Leg Med Tokyo Jpn 2004;6:25–31.10.1016/j.legalmed.2003.09.001Search in Google Scholar PubMed

65. Borgan E, Navon R, Vollan HK, Schlichting E, Sauer T, Yakhini Z, et al. Ischemia caused by time to freezing induces systematic microRNA and mRNA responses in cancer tissue. Mol Oncol 2011;5:564–76.10.1016/j.molonc.2011.08.004Search in Google Scholar PubMed PubMed Central

66. Sun M, Yamashita T, Shang J, Liu N, Deguchi K, Feng J, et al. Time-dependent profiles of microRNA expression induced by ischemic preconditioning in the gerbil hippocampus. Cell Transplant 2015;24:367–76.10.3727/096368915X686869Search in Google Scholar PubMed

67. Xi Y, Nakajima G, Gavin E, Morris CG, Kudo K, Hayashi K, et al. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA N Y N 2007;13:1668–74.10.1261/rna.642907Search in Google Scholar PubMed PubMed Central

68. Siebolts U, Varnholt H, Drebber U, Dienes H-P, Wickenhauser C, Odenthal M. Tissues from routine pathology archives are suitable for microRNA analyses by quantitative PCR. J Clin Pathol 2009;62:84–8.10.1136/jcp.2008.058339Search in Google Scholar PubMed PubMed Central

69. Cappellesso R, Nicolè L, Caroccia B, Guzzardo V, Ventura L, Fassan M, et al. Young investigator challenge: microRNA-21/MicroRNA-126 profiling as a novel tool for the diagnosis of malignant mesothelioma in pleural effusion cytology. Cancer Cytopathol 2016;124:28–37.10.1002/cncy.21646Search in Google Scholar PubMed

70. Jung M, Schaefer A, Steiner I, Kempkensteffen C, Stephan C, Erbersdobler A, et al. Robust microRNA stability in degraded RNA preparations from human tissue and cell samples. Clin Chem Lab Med 2010;56:998–1006.10.1373/clinchem.2009.141580Search in Google Scholar PubMed

71. Lou JJ, Mirsadraei L, Sanchez DE, Wilson RW, Shabihkhani M, Lucey GM, et al. A review of room temperature storage of biospecimen tissue and nucleic acids for anatomic pathology laboratories and biorepositories. Clin Biochem 2014;47:267–73.10.1016/j.clinbiochem.2013.12.011Search in Google Scholar PubMed PubMed Central

72. Bovell L, Shanmugam C, Katkoori VR, Zhang B, Vogtmann E, Grizzle WE, et al. MiRNAs are stable in colorectal cancer archival tissue blocks. Front Biosci Elite Ed 2012;4:1937–40.10.2741/e514Search in Google Scholar

73. Buitrago DH, Patnaik SK, Kadota K, Kannisto E, Jones DR, Adusumilli PS. Small RNA sequencing for profiling microRNAs in long-term preserved formalin-fixed and paraffin-embedded non-small cell lung cancer tumor specimens. PLoS One 2015;10. doi: 10.1371/journal.pone.0121521.10.1371/journal.pone.0121521.Search in Google Scholar

74. Wang Y, Chen J, Lin Z, Cao J, Huang H, Jiang Y, et al. Role of deregulated microRNAs in non-small cell lung cancer progression using fresh-frozen and formalin-fixed, paraffin-embedded samples. Oncol Lett 2016;11:801–8.10.3892/ol.2015.3976Search in Google Scholar PubMed PubMed Central

75. Feiersinger F, Nolte E, Wach S, Rau TT, Vassos N, Geppert C, et al. MiRNA-21 expression decreases from primary tumors to liver metastases in colorectal carcinoma. PLoS One 2016;11. doi: 10.1371/journal.pone.0148580.10.1371/journal.pone.0148580Search in Google Scholar PubMed PubMed Central

76. Kent OA, McCall MN, Cornish TC, Halushka MK. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res 2014;42:7528–38.10.1093/nar/gku461Search in Google Scholar PubMed PubMed Central

77. Tetzlaff MT, Curry JL, Yin V, Pattanaprichakul P, Manonukul J, Uiprasertkul M, et al. Distinct pathways in the pathogenesis of sebaceous carcinomas implicated by differentially expressed microRNAs. JAMA Ophthalmol 2015;133:1109–16.10.1001/jamaophthalmol.2015.2310Search in Google Scholar PubMed

78. Schuster C, Budczies J, Faber C, Kirchner T, Hlubek F. MicroRNA expression profiling of specific cells in complex archival tissue stained by immunohistochemistry. Lab Investig J Tech Methods Pathol 2011;91:157–65.10.1038/labinvest.2010.134Search in Google Scholar PubMed

79. Renwick N, Cekan P, Masry PA, McGeary SE, Miller JB, Hafner M, et al. Multicolor microRNA FISH effectively differentiates tumor types. J Clin Invest 2013;123:2694–702.10.1172/JCI68760Search in Google Scholar PubMed PubMed Central

80. Lin M, Shi C, Lin X, Pan J, Shen S, Xu Z, et al. sMicroRNA-1290 inhibits cells proliferation and migration by targeting FOXA1 in gastric cancer cells. Gene 2016;582:137–42.10.1016/j.gene.2016.02.001Search in Google Scholar PubMed

81. Pagès F, Kirilovsky A, Mlecnik B, Asslaber M, Tosolini M, Bindea G, et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J Clin Oncol Off J Am Soc Clin Oncol 2009;27:5944–51.10.1200/JCO.2008.19.6147Search in Google Scholar PubMed

82. Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006;313:1960–4.10.1126/science.1129139Search in Google Scholar PubMed

83. Wang E, Miller LD, Ohnmacht GA, Mocellin S, Perez-Diez A, Petersen D, et al. Prospective molecular profiling of melanoma metastases suggests classifiers of immune responsiveness. Cancer Res 2002;62:3581–6.Search in Google Scholar

84. Greenson JK, Bonner JD, Ben-Yzhak O, Cohen HI, Miselevich I, Resnick MB, et al. Phenotype of microsatellite unstable colorectal carcinomas: well-differentiated and focally mucinous tumors and the absence of dirty necrosis correlate with microsatellite instability. Am J Surg Pathol 2003;27:563–70.10.1097/00000478-200305000-00001Search in Google Scholar PubMed

85. Babapoor S, Horwich M, Wu R, Levinson S, Gandhi M, Makkar H, et al. MicroRNA in situ hybridization for miR-211 detection as an ancillary test in melanoma diagnosis. Mod Pathol Off J U S Can Acad Pathol Inc 2016;29:461–75.10.1038/modpathol.2016.44Search in Google Scholar PubMed

86. Patnaik SK, Kannisto E, Yendamuri S. Factors affecting the yield of microRNAs from laser microdissectates of formalin-fixed tissue sections. BMC Res Notes 2012;5:40.10.1186/1756-0500-5-40Search in Google Scholar PubMed PubMed Central

87. Chaudhuri AD, Yelamanchili SV, Fox HS. Combined fluorescent in situ hybridization for detection of microRNAs and immunofluorescent labeling for cell-type markers. Front Cell Neurosci 2013;7:160.10.3389/fncel.2013.00160Search in Google Scholar PubMed PubMed Central

Received: 2016-6-1
Accepted: 2016-9-5
Published Online: 2017-3-17
Published in Print: 2017-5-1

©2017 Walter de Gruyter GmbH, Berlin/Boston

Articles in the same Issue

  1. Frontmatter
  2. Editorial
  3. Not all good things come in big packages
  4. Pre-analytical and Analytical Issues of miRNA Measurement
  5. Variability in, variability out: best practice recommendations to standardize pre-analytical variables in the detection of circulating and tissue microRNAs
  6. Pitfalls of analysis of circulating miRNA: role of hematocrit
  7. Intraindividual variation of microRNA expression levels in plasma and peripheral blood mononuclear cells and the associations of these levels with the pathogenesis of autoimmune thyroid diseases
  8. miRNA assays in the clinical laboratory: workflow, detection technologies and automation aspects
  9. Clinical Applications of the Different Circulating Forms of miRNAs
  10. The role of extracellular vesicle microRNAs in cancer biology
  11. The clinical significance of platelet microparticle-associated microRNAs
  12. microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application
  13. microRNAs in cardiovascular disease – clinical application
  14. miRNAs in Cancer
  15. Non-coding RNAs: the cancer genome dark matter that matters!
  16. miRNAs as novel biomarkers in the management of prostate cancer
  17. Upregulated miR-16 expression is an independent indicator of relapse and poor overall survival of colorectal adenocarcinoma patients
  18. Identification of a novel microRNA, miR-4449, as a potential blood based marker in multiple myeloma
  19. miRNA analysis in pancreatic cancer: the Dartmouth experience
  20. miRNAs and Genomic
  21. miRNAs, single nucleotide polymorphisms (SNPs) and age-related macular degeneration (AMD)
Downloaded on 28.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/cclm-2016-0471/html?lang=en
Scroll to top button