The complex microRNA (miRNA) network plays an important role in the regulation of cellular processes such as development, differentiation, and apoptosis. Recently, the presence of cell-free miRNAs that circulate in body fluids was discovered. The ability of these circulating miRNAs to mirror physiological and pathophysiological conditions as well as their high stability in stored patient samples underlines the potential of these molecules to serve as biomarkers for various diseases. In this review, we describe recent findings in miRNA-mediated cell-to-cell communication and the functions of circulating miRNAs in the field of hematology. Furthermore, we discuss current approaches to design biomarker studies with circulating miRNAs. This article critically reviews the novel field of circulating miRNAs and highlights their suitability for clinical and basic research in addition to their potential as a novel class of biomarkers.

Tissue-bound miRNAs classify hematological diseases

MicroRNAs (miRNAs) are 19 to 25–nucleotide-long, noncoding RNA molecules that were recently identified to play key roles in regulating gene expression by inhibiting translation and/or triggering degradation of target mRNAs.1  Already at their time of discovery, miRNAs such as lin-4 were found to coordinate the temporal transition of tissues between developmental stages.2 ,3  The results of extensive profiling approaches further supported the idea that miRNA expression patterns encode the developmental history of both human tissues and cancers.4  Despite the relatively small number of miRNA species, they seem to contain a large amount of diagnostic information. In a pivotal study, Lu et al4  demonstrated that miRNAs are able to discriminate leukemias of different origin. The potential of miRNAs to distinguish between acute leukemias was further emphasized by Mi et al,5  showing that 4 miRNAs (miR-223, miR-128a, miR-128b, and let-7b) were able to discriminate between acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). In fact, a combination of any 2 of these miRNAs could differentiate ALL from AML cases with an overall diagnostic accuracy of 97% to 99%. Subsequently, researchers further stratified lymphoid and myeloid disease entities according to cytogenetics, molecular genetics, and prognostic information based on their tissue-bound miRNA expression patterns6 -10  (see supplemental Table 1 on the Blood website).

Circulating miRNAs, from intracellular to extracellular and back

Recently, circulating miRNAs were discovered in a variety of body fluids11 -13  that can be obtained in a noninvasive or minimally invasive manner. Especially in blood, the levels and composition of circulating miRNAs were found to reflect the presence of malignant and nonmalignant diseases, even of localized tumors such as glioblastomas and lung cancer.14 ,15  Based on their origin, cell-free miRNAs are referred to in the current literature as, for example, blood-, serum- or plasma-borne. Here, we will refer to cell-free miRNAs as “circulating miRNAs” regardless of their origin or the body fluid they are derived from.

Due to their accessibility, the most common miRNA sources reported in literature are whole blood, serum, and plasma (Table 1). Despite the omnipresence of ribonucleases, miRNAs were found to be highly stable in blood and other body fluids. In fact, serum-borne miRNAs were documented to be stable for more than 10 days at room temperature and more than 10 years if stored at −20°C.16  Two distinct mechanisms have been shown to protect circulating miRNAs from degradation. First, miRNAs form complexes with proteins such as AGO-2,17  a component of the RNA-induced silencing complex, or NPM-1,18  which is involved in the biogenesis of ribosomes as well as with high-density lipoproteins (HDLs).19  Second, precursor miRNAs20  and mature miRNAs are packaged into small vesicles (Figure 1)21 ,22  that are either derived from the endosomal membrane compartment or shed directly from the plasma membrane17 ,23  (Figure 1A-B). Although it has been reported that the majority of miRNAs is present in both packaging types, there is growing evidence that extracellular miRNAs are sorted selectively either into vesicles24 -26  or into ribonucleoprotein complexes,17 ,27  and that the association of circulating miRNAs with the exosome or protein compartment is disease dependent.28  In 2012, Palma et al29  discovered that breast cancer cells selectively release miRNAs into anomalous vesicles, which are larger than conventional exosomes and exhibit metastasis-relevant surface markers such as CD44. The identification of such cancer-associated exosomes based on their expression of specific marker proteins like EpCAM, CD2430  or CD44 could possibly serve as a new diagnostic tool for minimal residual disease in solid tumors as well as hematological neoplasias.

Table 1

Advantages and disadvantages of using whole blood, plasma, and serum for profiling circulating miRNAs

Blood mediumAdvantagesDisadvantages
Whole blood It is not biased by cell lysis. High levels of blood cell miRNAs might conceal disease-specific patterns. 
It is possible to use small nuclear RNAs as housekeeping genes (eg, RNU6). 
There is direct use of the primary sample; no further work-up is necessary. Cell counts have to be considered for biological variation. 
Blood serum There are slightly higher miRNA yields compared to plasma.31 ,32  The coagulation process in serum production might increase sample-to-sample variations.31  
Removal of cells decreases the heterogeneity of the readout. 
The majority of archived samples is stored in the form of serum. Further work-up is necessary. 
 It is biased by cell lysis.35  
Blood plasma Removal of cells decreases the heterogeneity of the readout. Frequent contamination with platelets leading to elevates platelet-specific miRNAs.31 ,33  
It is not biased by the coagulation process. 
 Further work-up is necessary. 
 It is biased by cell lysis.34  
Blood mediumAdvantagesDisadvantages
Whole blood It is not biased by cell lysis. High levels of blood cell miRNAs might conceal disease-specific patterns. 
It is possible to use small nuclear RNAs as housekeeping genes (eg, RNU6). 
There is direct use of the primary sample; no further work-up is necessary. Cell counts have to be considered for biological variation. 
Blood serum There are slightly higher miRNA yields compared to plasma.31 ,32  The coagulation process in serum production might increase sample-to-sample variations.31  
Removal of cells decreases the heterogeneity of the readout. 
The majority of archived samples is stored in the form of serum. Further work-up is necessary. 
 It is biased by cell lysis.35  
Blood plasma Removal of cells decreases the heterogeneity of the readout. Frequent contamination with platelets leading to elevates platelet-specific miRNAs.31 ,33  
It is not biased by the coagulation process. 
 Further work-up is necessary. 
 It is biased by cell lysis.34  
Figure 1

Long-distance intercellular miRNA transfer. In addition to short-distance miRNA exchange, for example, via gap junctions or during immunological synapse formation,21 ,22  miRNAs can also be released by cells either into extracellular vesicles or complexed to proteins for long-distance communication. These packaged, mature miRNAs and pre-miRNAs are protected from extracellular ribonuclease activity and can be taken up by recipient cells, where they influence cellular processes. (A) During “plasma membrane budding,” ectosomes containing cytoplasmic components like miRNAs are released into the extracellular surrounding. (B) The fusion of exocytic multivesicular bodies (MVBs) with the plasma membrane releases miRNA-containing intraluminal vesicles called exosomes. When derived from healthy cells, exosomes reach a diameter of 60 to 90 nm. Exosomes released by cancer cells were reported to differ in size or to display metastasis-associated surface markers.29 ,30  (C) The ABCA1 transporter mediates the release of HDL-complexed miRNAs.19  (D) Apoptotic bodies are released from a cell that undergoes apoptosis. These large vesicles contain fragmented DNA and cytoplasmic components including miRNAs. General mechanisms of vesicle uptake in recipient cells involve (E) endocytosis, (F) phagocytosis, and (G) fusion with the plasma membrane. The uptake of HDL-complexed miRNAs in recipient cells is mediated by (H) SR-B1 receptors.19  Release and uptake mechanisms of extravesicular AGO- or NPM-1–complexed miRNAs are not described yet.

Figure 1

Long-distance intercellular miRNA transfer. In addition to short-distance miRNA exchange, for example, via gap junctions or during immunological synapse formation,21 ,22  miRNAs can also be released by cells either into extracellular vesicles or complexed to proteins for long-distance communication. These packaged, mature miRNAs and pre-miRNAs are protected from extracellular ribonuclease activity and can be taken up by recipient cells, where they influence cellular processes. (A) During “plasma membrane budding,” ectosomes containing cytoplasmic components like miRNAs are released into the extracellular surrounding. (B) The fusion of exocytic multivesicular bodies (MVBs) with the plasma membrane releases miRNA-containing intraluminal vesicles called exosomes. When derived from healthy cells, exosomes reach a diameter of 60 to 90 nm. Exosomes released by cancer cells were reported to differ in size or to display metastasis-associated surface markers.29 ,30  (C) The ABCA1 transporter mediates the release of HDL-complexed miRNAs.19  (D) Apoptotic bodies are released from a cell that undergoes apoptosis. These large vesicles contain fragmented DNA and cytoplasmic components including miRNAs. General mechanisms of vesicle uptake in recipient cells involve (E) endocytosis, (F) phagocytosis, and (G) fusion with the plasma membrane. The uptake of HDL-complexed miRNAs in recipient cells is mediated by (H) SR-B1 receptors.19  Release and uptake mechanisms of extravesicular AGO- or NPM-1–complexed miRNAs are not described yet.

Close modal

Circulating miRNAs require specific considerations for their detection and quantification.

Whole blood vs plasma vs serum vs exosomes: what is the best source? Challenges in the analysis of circulating miRNAs include preanalytical decisions, such as sample storage, sample processing, the profiling method, and postanalytical decisions concerning, for example, data normalization. So far, a comparison of circulating miRNA levels among plasma, serum, and whole blood is still missing. However, a detailed analysis of miRNA spectra in serum and plasma was performed by Wang et al,31  who investigated potential biases with regard to the preparation of blood samples on miRNA quantification. In their analysis, they detected higher RNA concentrations in serum compared with the corresponding plasma samples.31  The authors31  suggest that this difference in concentration is attributed to the release of RNA during the coagulation process and is not caused by cell lysis, as the amount of blood-cell associated miRNAs such as miR-150, miR-16, and miR-126 did not change in their experiments. Based on these findings, the authors recommend plasma over serum to reduce sample-to-sample variations induced by serum coagulation. In contrast, van Schooneveld et al32  highlighted the advantages of serum for miRNA expression analyses, due to its more consistent and higher yield of small RNAs. Hence, the choice between plasma and serum for miRNA analysis may depend on more practical reasons, such as availability and handling. More recent approaches include the analysis of purified exosome-associated miRNAs (exosomal miRNAs). Considering the protein-complexed nature of circulating miRNAs,17 ,19 ,27  it does not seem generally advisable to focus on exosomal miRNAs for biomarker studies. However, exosomal miRNA profiling could turn out to be superior for detecting abnormalities in secretory cell types and for targeted analyses of cancer-related vesicles. Other issues in the choice of sample material concern the influence of red blood cell counts and hemolysis as well as the frequent presence of platelets in blood plasma and partly in serum samples upon miRNA quantification.31 ,33 -36  These problems can only be addressed through standardized protocols.

Current approaches and postanalytical processing to quantify circulating miRNAs.

The lack of standardized protocols for small RNA extraction combined with a growing variety of miRNA profiling approaches particularly complicates the comparison of independent data sets. In addition, postanalysis decisions such as data normalization have the potential to bias results and conclusions. An overview of currently employed methods reported in recent literature (2011-2012), which were used for miRNA extraction and quantification in serum, plasma, whole blood, and cerebrospinal fluid (CSF) and for data normalization, is shown in supplemental Table 2.

Despite reasonable enrichment of small RNAs by RNA extraction protocols, little is known about the composition of the extracted RNA. Do RNA subgroups behave differently in isolation protocols based on their size or sequence? This issue has been addressed recently by Kim et al,37  who demonstrated that small RNAs with low guanine-cytosine content are recovered inefficiently from small cell numbers in RNA extractions using TRIzol. In addition, theses authors found that some pre-miRNAs, small interfering RNAs, and transfer RNAs are also affected by this phenomenon.37  A possible explanation for this finding is that small RNAs need to base pair with carrier molecules such as longer RNAs for effective precipitation, thus resulting in a biased RNA extraction. It remains to be shown whether this is true for other situations, for example, affinity-based RNA extraction protocols using columns.

Because of its high sensitivity, fast readout, and universal application, quantitative realtime polymerase chain reaction (qRT-PCR) is widely used for miRNA quantification. Improvements of this technology include the incorporation of locked nucleic acids into the PCR primers, which further increased specificity and sensitivity and circumvented the need for input RNA preamplification due to enhanced base stacking, RNA backbone stabilization, and the balancing of differences in melting temperatures caused by variances in RNA guanine-cytosine content.38  Other approaches to quantify circulating miRNAs include next-generation sequencing (NGS) and hybridization-based methods such as miRNA microarray technologies, NanoString,39  or Scano-miR.40  Older methods like northern blot analysis are not suitable due to the necessity of high amounts of RNA input and the difficulty of accurately quantifying miRNAs. A shared limitation is the lack of absolute miRNA quantification41 ,42  and their insufficient comparability due to platform-dependent detection biases.41 ,43 

In contrast to qRT-PCR and microarray technology, NGS allows the identification of novel miRNAs, miRNA sequence changes, and isomiRs.44  Notably, not all novel miRNAs discovered by NGS are indeed true miRNAs. For example, Jones et al45  proposed miR-720 and miR-1308 as tools to differentiate myeloma patients from healthy controls. However, in 2010, miR-720 was deleted from miRBase, a reference database for miRNAs, as miR-720 was validated to be a transfer RNA fragment.45  The major advantages and disadvantages of the currently used miRNA quantification methods are presented in supplemental Table 3 and were extensively reviewed in 2012 by Pritchard et al.46 

A further challenge remains the bioinformatical processing of large miRNA expression data sets, including data normalization. The lack of endogenous normalization controls for circulating miRNAs, in particular, is still discussed controversially. Frequently used genes for normalizing qRT-PCR data include small nuclear RNAs like RNU6B or RNU43. However, these molecules are considered inadequate due to their low or varying abundance in body fluids47 -49  and a nonexcludable disease-specificity, as shown by Baraniskin et al for RNU2 in colorectal cancer.50  The ideal case for normalization and quantification of circulating miRNAs would be a universal housekeeping gene. Until such a gene (or RNA) is identified, fluid-specific housekeeping genes will have to be identified individually for each analysis. In general, all ubiquitously expressed miRNAs are candidates for normalization controls, although a systematic screen for endogenous controls in various body fluids has not yet been performed. Because of this drawback, many studies report the use of the mean expression value of all expressed miRNAs (global mean normalization) to normalize their data sets. This method has been reported to exhibit superior performance over the normalization with reference genes in large and unbiased profiling approaches.51 ,52 

Current design of circulating miRNA biomarker studies

Numerous published biomarker studies (Table 2 53 -56  and supplemental Table 4) use a set of preselected miRNAs, often with known tissue-based expression, and correlate their circulating levels to healthy control vs disease or different disease subgroups. Regarding diffuse large B-cell lymphoma (DLBCL), Lawrie et al57  were the first to investigate whether circulating miRNAs can be detected in serum and if their expression levels differ between DLBCL patients and healthy individuals. In their retrospective study, they found deregulated levels of all 3 examined miRNAs (miR-155, miR-210, and miR-21), but only increased miR-21 levels were associated with an increased relapse-free survival. A similar study was published in 2012 by Fang et al,58  who confirmed elevated levels of miR-21 and miR-155 in DLBCL patients but could not demonstrate a significant association with any clinical parameters. It is not clear if the closed study design, the sample numbers, or an inhomogeneity of the profiled DLBCL samples led to different results.

Table 2

Studies involving circulating miRNAs in hematological diseases

DiseaseAnalyzed aspectNumber of deregulated/analyzed miRNAsMost important candidate miRNAsProfiling methodReference
Serum/plasma-based studies 
 ATL Identification of miRNA markers for the diagnosis and classification of acute and chronic ATL 5/>200 miR-155 ↑pp Microarray; qRT-PCR 63 
let-7g 
miR-126 ↓pp 
miR-130a 
let-7b 
 CLL Identification of clinically relevant plasma miRNAs in CLL 27/333 miR-150/150* Multiplexing qRT-PCR (Applied Biosystems) 65 
miR-20a 
miR-29a 
miR-135a* 
DLBCL Utility of cancer-associated miRNAs for DLBCL diagnosis 3/3 miR-21 ↑gp TaqMan qRT-PCR 57 
 Suitability of serum miRNAs for DLBCL detection 5/7 (1↓, 4↑) miR-15a, -16-1 ↑ TaqMan qRT-PCR 58 
miR-29c, -155 ↑ 
miR-34a ↓ 
 MDS Suitability of 2 plasma miRNAs to predict survival in MDS patients 2/2 let-7a ↓gp qRT-PCR (Applied Biosystems) 59 
miR-16 ↓gp 
 MM Identification of diagnostic miRNA markers in MM 3/9detected miR-720 Human miRNA microarray (Agilent), TaqMan qRT-PCR 60 
miR-1308 
miR-1246 
 Expression dynamics of miR-17-92a in MM 25/381 miR-17-92a ↓ TaqMan Low Density Arrays, qRT-PCR 53 
(20↓, 5↑) miR-223 ↓ 
 Prognostic utility of plasma miRNAs 5/667 miR-20a ↑pp TaqMan Low Density Arrays, qRT-PCR 61 
miR-148a ↑pp 
miR-99b ↑t(4;14) 
miR-221 ↑del(13q) 
 NHL Suitability of miR-92a for diagnosis and NHL treatment monitoring 1/470 miR-92a ↓ Agilent miRNA Microarrays version 1.5, TaqMan qRT-PCR 54 
 NKTL Diagnostic and prognostic value of circulating miR-221 NA miR-221 Direct qPCR without RNA extraction 55 
CSF-based studies 
 DLBCL Identification of diagnostic miRNAs in CSF of DLBCL patients 3/6 miR-21 ↑ TaqMan qRT-PCR 56 
miR-19 ↑ 
miR-92a ↑ 
DiseaseAnalyzed aspectNumber of deregulated/analyzed miRNAsMost important candidate miRNAsProfiling methodReference
Serum/plasma-based studies 
 ATL Identification of miRNA markers for the diagnosis and classification of acute and chronic ATL 5/>200 miR-155 ↑pp Microarray; qRT-PCR 63 
let-7g 
miR-126 ↓pp 
miR-130a 
let-7b 
 CLL Identification of clinically relevant plasma miRNAs in CLL 27/333 miR-150/150* Multiplexing qRT-PCR (Applied Biosystems) 65 
miR-20a 
miR-29a 
miR-135a* 
DLBCL Utility of cancer-associated miRNAs for DLBCL diagnosis 3/3 miR-21 ↑gp TaqMan qRT-PCR 57 
 Suitability of serum miRNAs for DLBCL detection 5/7 (1↓, 4↑) miR-15a, -16-1 ↑ TaqMan qRT-PCR 58 
miR-29c, -155 ↑ 
miR-34a ↓ 
 MDS Suitability of 2 plasma miRNAs to predict survival in MDS patients 2/2 let-7a ↓gp qRT-PCR (Applied Biosystems) 59 
miR-16 ↓gp 
 MM Identification of diagnostic miRNA markers in MM 3/9detected miR-720 Human miRNA microarray (Agilent), TaqMan qRT-PCR 60 
miR-1308 
miR-1246 
 Expression dynamics of miR-17-92a in MM 25/381 miR-17-92a ↓ TaqMan Low Density Arrays, qRT-PCR 53 
(20↓, 5↑) miR-223 ↓ 
 Prognostic utility of plasma miRNAs 5/667 miR-20a ↑pp TaqMan Low Density Arrays, qRT-PCR 61 
miR-148a ↑pp 
miR-99b ↑t(4;14) 
miR-221 ↑del(13q) 
 NHL Suitability of miR-92a for diagnosis and NHL treatment monitoring 1/470 miR-92a ↓ Agilent miRNA Microarrays version 1.5, TaqMan qRT-PCR 54 
 NKTL Diagnostic and prognostic value of circulating miR-221 NA miR-221 Direct qPCR without RNA extraction 55 
CSF-based studies 
 DLBCL Identification of diagnostic miRNAs in CSF of DLBCL patients 3/6 miR-21 ↑ TaqMan qRT-PCR 56 
miR-19 ↑ 
miR-92a ↑ 

ATL, adult T-cell leukemia; BL, Burkitt lymphoma; gp, associated with good prognosis; NHL, non-Hodgkin lymphoma; NKTL, extranodal natural killer/T-cell lymphoma; pp, associated with poor prognosis; NA, lacked access to the paper; CLL, chronic lymphocytic lymphoma; DLBCL, diffuse large B-cell lymphoma; MM, multiple myeloma; CSF, cerebrospinal fluid; Arrows, up or down regulation.

This has been identified as not a true miRNA (dead miRBase entry).

Zuo et al59  measured 2 myelodysplastic syndrome (MDS)–related miRNAs, miR-16 and let-7a, in the plasma of 50 patients with MDS and showed that levels of these miRNAs had significant prognostic relevance. In a multivariate analysis, the authors demonstrated that reduced plasma levels of let-7a were an independent predictor for overall survival.59  Other studies also proposed the prognostic validity of serum and plasma miRNAs, for example, in multiple myeloma (MM),60 ,61  AML,62  and T-cell leukemia63  (see Table 2).

So far, the most comprehensive study of circulating miRNAs was published by Keller et al,14  who investigated the miRNA transcriptome of whole blood samples in 14 selected diseases, including multiple cancers, using miRNA microarrays.64  The resulting miRNA profiles allowed the identification of 2 miRNAs that discriminated between healthy controls and each profiled disease with an average accuracy of 88.5% or 72.5%, respectively. Despite the high discriminative potential, the predictive value of the total miRNome (the full complement of miRNAs in a genome) amounted on average to only 67.45%. Confounding variables for such an experimental approach in whole blood include existing known and unknown comorbidities and varying blood counts, which might distort disease-specific miRNA profiles.

But how do circulating miRNAs compare with established biomarkers? Can they complement existing markers to improve their sensitivity and specificity? These questions were partially addressed in a study by Moussay et al,65  who investigated the potential of circulating miRNAs as biomarkers in the plasma of patients with chronic lymphocytic leukemia (CLL) and compared their presence to the ZAP-70 status, which is an established clinical risk stratifier.66  A small set of 3 miRNAs, miR-195, miR-29a, and miR-222, allowed the separation of patients with CLL from healthy controls. Furthermore, a signature of 6 miRNAs (miR-29a, miR-483-5p, miR-195, miR-185, miR-135a*, and miR-15a) could segregate ZAP-70+ and ZAP-70 samples. In contrast, IgVH mutation status did not correlate with circulating miRNA levels, whereas plasma levels of miR-20a positively correlated with a longer time to treatment interval. These initial studies strengthen the concept of circulating miRNAs as potential biomarkers. However, to understand the connection of circulating miRNAs with the pathology of hematological diseases, comprehensive signatures that compare circulating and tissue-bound miRNA levels within the same individuals are necessary. Such prospective profiling approaches would not only identify disease-specific miRNA signatures, but they would create new opportunities to monitor treatment responses and identify early relapses. Nevertheless, to establish circulating miRNAs as biomarkers, well-designed clinical trials are required. So far, there is only 1 trial registered that is relevant to hematology and investigates circulating miR-155 as a predictive marker in acute graft-versus-host disease (clinicaltrials.gov identifier NCT01521039).

Functional aspects of circulating miRNAs in hematopoietic cells

The discovery of circulating miRNAs raised several questions about their function. Are these molecules mere degradation products without any functional relevance, or are they actively secreted to serve as messengers in cell-to-cell communication? Based on multiple studies, endothelial cells in particular seem to have the ability to communicate through miRNA trafficking in physiological and pathophysiological conditions. For instance, Zhang et al24  reported the active secretion of miR-150 by THP-1, a cell line derived from human acute myelomonocytic leukemia, as well as by plasma cells. Secreted miR-150 was taken up by cocultured human microvascular endothelial cells in which miR-150 regulated the protein level of c-MYB, a bona fide miR-150-target.24  A similar setup was used by Umezu et al,67  who visualized the transport of fluorescently labeled miR-92a via exosomes from K562, a human chronic myeloid leukemia cell line, to human umbilical vein endothelial cells (HUVECs). This led to the repression of miR-92a targets like integrin α5 in the recipient cells.67  A very different mechanism of cellular miRNA communication was recently identified by Fabbri et al,68  who showed that 2 tumor-secreted exosomal miRNAs, miR-21 and miR-29b, can bind and thereby activate Toll-like receptors on immune cells. This activation triggered an inflammatory response in the recipient cells and ultimately resulted in a benefit for tumor growth and metastasis in vivo.68  These and similar studies69 -78  (Table 3) indicate that exosome-shuttled miRNAs can serve as in vivo messengers that are able to regulate signaling pathways in recipient cells and might represent a novel platform for tumor cells to modulate their environment.

Table 3

Studies investigating long-distance intercellular miRNA communication

AuthorsAnalyzed aspectCells/cell linestransported miRNAsMethodsReference
Yuan et al, 2009 Transfer of miRNAs by embryonic stem cell microvesicles Murine ES cells miR-290 Coculture, qRT-PCR, visual confirmation of GFP-labeled MV uptake 60 
MEFs miR-291-3p 
 miR-292-3p 
 miR-294, -295 
Zhang et al, 2010 Influence of secreted monocytic miR-150 on endothelial cell migration THP-1 primary PBMCs miR-150 and 26 others Endothelial cell migration and various reporter assays, quantification of miRNA target via western blot 24 
HMEC-1 
Collino et al, 2010 miRNA content of MSC and HLSC vesicles Primary MSCs and HLSCs miR-21, -451, qRT-PCR of producer cells, MVs, and target cells; visual confirmation of MV and miRNA uptake, quantification of miRNA targets 70 
mTEC miR-100, -99a 
 miR-223, -564 
Vickers et al, 2011 Cellular export and uptake of functional HDL-bound miRNAs Huh7, HEK293, J774, BHK cells, human plasma miR-135a* miRNA microarray and luciferase reporter assays, loss/gain of function studies 19 
miR-188-5p 
miR-877, -223 
Grange et al, 2011 Exchange of genetic information between tumor and stromal cells HUVECs miR-29a/c, -19b, MV treatment of SCID mice, in vitro + in vivo angiogenesis + metastasis assays, mRNA/miRNA profiling 71 
CD105+ cancer stem cells miR-151, -650, 
 miR-200c, -141 
 miR-92 
Hergenreider et al, 2012 Atheroprotective communication between endothelial cells and SMCs through miRNAs HUVECs miR-143 Coculture and reporter assays, qRT-PCR, injection of MVs into ApoE-/- mice + readout, antagomiR experiments 72 
HAECs miR-145 
HASMCs  
Umezu et al, 2012 Leukemia cell to endothelial cell communication via exosomal miRNAs K562 miR-92a Coculture and reporter assays, miRNA expression profiling 67 
HUVECs 
Fonsato et al, 2012 Antitumor effect of miRNA-containing vesicles on hepatoma cells HLSCs miR-451 Intratumor injection of miRNA and miRNA inhibitor–containing MVs into SCID mice 73 
HepG2 miR-223 
Primary hepatoma miR-24 
 miR-31 
Xin et al, 2012 Communication between MSCs and neural cells via exosomal miRNAs Primary rat MSCs, neurons, and astrocytes miR-133b Coculture, knockdown, and luciferase reporter assays, in vitro treatment of neurons with MVs 74 
Ismail et al, 2013 Transport of macrophage-derived MVs to target cells Primary PBMCs miR-223 AntagomiR transfection, fluorescent MV labeling + microscopy, luciferase reporter assay, qRT-PCR 75 
THP-1 
CCL-204 
A549 
Cantaluppi et al, 2012 miRNA-containing MVs released by endothelial cells preventing kidney injury Primary PBMCs miR-126 Injection of MVs into rats following IRI (also Dicer knocked-down/antagomiR MV application) 76 
TepCs miR-296 
Montecalvo et al, 2012 Transfer of functional miRNAs between murine DCs via exosomes Murine BM-derived DCs and 1H3.1 and OT-II T cells miR-451, miR148a Fluorescence- and microscopy-based analysis of MV uptake and cytosolic content release, miRNA profiling + reporter assays 77 
Katsman et al, 2012 Effect of ES cell MVs on retinal progenitor cells Murine ES cells miR-292 qRT-PCR 78 
MIO-M1 miR-295 
AuthorsAnalyzed aspectCells/cell linestransported miRNAsMethodsReference
Yuan et al, 2009 Transfer of miRNAs by embryonic stem cell microvesicles Murine ES cells miR-290 Coculture, qRT-PCR, visual confirmation of GFP-labeled MV uptake 60 
MEFs miR-291-3p 
 miR-292-3p 
 miR-294, -295 
Zhang et al, 2010 Influence of secreted monocytic miR-150 on endothelial cell migration THP-1 primary PBMCs miR-150 and 26 others Endothelial cell migration and various reporter assays, quantification of miRNA target via western blot 24 
HMEC-1 
Collino et al, 2010 miRNA content of MSC and HLSC vesicles Primary MSCs and HLSCs miR-21, -451, qRT-PCR of producer cells, MVs, and target cells; visual confirmation of MV and miRNA uptake, quantification of miRNA targets 70 
mTEC miR-100, -99a 
 miR-223, -564 
Vickers et al, 2011 Cellular export and uptake of functional HDL-bound miRNAs Huh7, HEK293, J774, BHK cells, human plasma miR-135a* miRNA microarray and luciferase reporter assays, loss/gain of function studies 19 
miR-188-5p 
miR-877, -223 
Grange et al, 2011 Exchange of genetic information between tumor and stromal cells HUVECs miR-29a/c, -19b, MV treatment of SCID mice, in vitro + in vivo angiogenesis + metastasis assays, mRNA/miRNA profiling 71 
CD105+ cancer stem cells miR-151, -650, 
 miR-200c, -141 
 miR-92 
Hergenreider et al, 2012 Atheroprotective communication between endothelial cells and SMCs through miRNAs HUVECs miR-143 Coculture and reporter assays, qRT-PCR, injection of MVs into ApoE-/- mice + readout, antagomiR experiments 72 
HAECs miR-145 
HASMCs  
Umezu et al, 2012 Leukemia cell to endothelial cell communication via exosomal miRNAs K562 miR-92a Coculture and reporter assays, miRNA expression profiling 67 
HUVECs 
Fonsato et al, 2012 Antitumor effect of miRNA-containing vesicles on hepatoma cells HLSCs miR-451 Intratumor injection of miRNA and miRNA inhibitor–containing MVs into SCID mice 73 
HepG2 miR-223 
Primary hepatoma miR-24 
 miR-31 
Xin et al, 2012 Communication between MSCs and neural cells via exosomal miRNAs Primary rat MSCs, neurons, and astrocytes miR-133b Coculture, knockdown, and luciferase reporter assays, in vitro treatment of neurons with MVs 74 
Ismail et al, 2013 Transport of macrophage-derived MVs to target cells Primary PBMCs miR-223 AntagomiR transfection, fluorescent MV labeling + microscopy, luciferase reporter assay, qRT-PCR 75 
THP-1 
CCL-204 
A549 
Cantaluppi et al, 2012 miRNA-containing MVs released by endothelial cells preventing kidney injury Primary PBMCs miR-126 Injection of MVs into rats following IRI (also Dicer knocked-down/antagomiR MV application) 76 
TepCs miR-296 
Montecalvo et al, 2012 Transfer of functional miRNAs between murine DCs via exosomes Murine BM-derived DCs and 1H3.1 and OT-II T cells miR-451, miR148a Fluorescence- and microscopy-based analysis of MV uptake and cytosolic content release, miRNA profiling + reporter assays 77 
Katsman et al, 2012 Effect of ES cell MVs on retinal progenitor cells Murine ES cells miR-292 qRT-PCR 78 
MIO-M1 miR-295 

BM, bone marrow; DC, dendritic cell; IRI, renal ischemia-reperfusion injury; MSC, mesenchymal stem cell; MV, microvesicle; PBMC, peripheral blood mononuclear cell; SCID, severe combined immunodeficiency; GFP, green fluorescent protein.

Regarding cell-to-cell transfer of protein-complexed miRNAs, Vickers et al19  demonstrated that cellular release of HDL-complexed miRNAs is mediated by the adenosine triphosphate–binding cassette transporter A1 (ABCA1) (Figure 1C) and that their uptake is guided by the scavenger receptor class B type I (SR-BI) (Figure 1H). Furthermore, they could show that HDL can carry distinct miRNA populations depending on the health status of the tested individual and that HDL-associated miRNA profiles significantly differ from exosome-associated miRNAs.19  Another class of microvesicles that is involved in miRNA trafficking represent apoptotic bodies, which derive from the cellular membrane of apoptotic cells (Figure 1D) and contain cytoplasmic compounds such as miRNAs. Zernecke et al79  were the first to show that miR-126–enriched apoptotic bodies shed by endothelial cells are taken up by neighboring vascular cells, where they promote proliferation as well as production of CXCL12, a chemokine known to counteract apoptosis. Similarly, the relevance of miRNA trafficking in the bone marrow compartment was highlighted by Salvucci et al,80  who demonstrated that granulocyte colony-stimulating factor promotes the accumulation of miR-126–containing microvesicles in the bone marrow and thereby targets the expression of surface vascular cell adhesion molecule-1 on early hematopoietic cells.80  This novel regulatory layer underlines the growing impact of circulating miRNAs on stem cell physiology.

Naturally, the concept of circulating miRNAs inspired new therapeutic approaches such as the use of nanotechnology-based carrier strategies to enrich or antagonize miRNAs in diseased cells.81  Both strategies proved to be effective in different murine lymphoma models.82 ,83  However, both the clinical administration of exosomes84  and the drug-induced inhibition of exosomal release68 ,85  from, for example, leukemia cells remain highly speculative.

It is a fascinating idea that cells can trigger or repress pathways in recipient cells via miRNAs during or even after cell death. Whether similar transport mechanisms account for the exchange of circulating miRNAs between the blood-brain and the blood-CSF barriers, as recently shown for central nervous system lymphomas,86  still needs to be investigated. Despite all the novel insights in this field, it is not yet clear whether cell-mediated miRNA transfer is a highly specialized fine-tuning mechanism or a substantial pathway and whether it can be targeted therapeutically to counter, for example, leukemogenesis.

Despite the recognized potential of circulating miRNAs as biomarkers, only little is known about their origin in physiological and pathological conditions or about the factors that influence their levels and composition. Circulating miRNAs are profiled in an increasing number of diseases and body fluids, but to introduce them as biomarkers into clinical routine, carefully conducted clinical studies are needed. Importantly, preanalytic factors such as the collection, processing, and storage of samples have to be monitored and standardized (supplemental Tables 2 and 3). Although the idea of functional circulating miRNAs is intriguing, their relevance is still a matter of debate. It remains to be seen whether circulating miRNAs are mere fine-tuners or central players in hematological diseases, a question that functional studies only recently started to focus on. However, judged by their advantageous properties and the continuously increasing amount of studies, circulating miRNAs have the potential to become reasonable diagnostic tools once their infancy has passed.

The online version of this article contains a data supplement.

This work was supported by grants from Deutsche Krebshilfe (109420), fellowship 2010/04 from the European Hematology Association, and grants from Deutsche Forschungsgemeinschaft (D.3955 (SFB 1074)).

Contribution: S.G., A.S., C.L., C.B., H.D., D.M., and F.K. wrote the paper.

Conflict-of-interest disclosure: A.S. is employed by Exiqon. The remaining authors declare no competing financial interests.

Correspondence: Florian Kuchenbauer, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany; e-mail: florian.kuchenbauer@uni-ulm.de.

1
Bartel
 
DP
MicroRNAs: genomics, biogenesis, mechanism, and function.
Cell
2004
, vol. 
116
 
2
(pg. 
281
-
297
)
2
Lee
 
RC
Feinbaum
 
RL
Ambros
 
V
The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14.
Cell
1993
, vol. 
75
 
5
(pg. 
843
-
854
)
3
Reinhart
 
BJ
Slack
 
FJ
Basson
 
M
, et al. 
The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans.
Nature
2000
, vol. 
403
 
6772
(pg. 
901
-
906
)
4
Lu
 
J
Getz
 
G
Miska
 
EA
, et al. 
MicroRNA expression profiles classify human cancers.
Nature
2005
, vol. 
435
 
7043
(pg. 
834
-
838
)
5
Mi
 
S
Lu
 
J
Sun
 
M
, et al. 
MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia.
Proc Natl Acad Sci USA
2007
, vol. 
104
 
50
(pg. 
19971
-
19976
)
6
Fulci
 
V
Colombo
 
T
Chiaretti
 
S
, et al. 
Characterization of B- and T-lineage acute lymphoblastic leukemia by integrated analysis of MicroRNA and mRNA expression profiles.
Genes Chromosomes Cancer
2009
, vol. 
48
 
12
(pg. 
1069
-
1082
)
7
Lawrie
 
CH
Soneji
 
S
Marafioti
 
T
, et al. 
MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma.
Int J Cancer
2007
, vol. 
121
 
5
(pg. 
1156
-
1161
)
8
Calin
 
GA
Liu
 
CG
Sevignani
 
C
, et al. 
MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias.
Proc Natl Acad Sci USA
2004
, vol. 
101
 
32
(pg. 
11755
-
11760
)
9
Jongen-Lavrencic
 
M
Sun
 
SM
Dijkstra
 
MK
Valk
 
PJ
Löwenberg
 
B
MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia.
Blood
2008
, vol. 
111
 
10
(pg. 
5078
-
5085
)
10
Schwind
 
S
Maharry
 
K
Radmacher
 
MD
, et al. 
Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2010
, vol. 
28
 
36
(pg. 
5257
-
5264
)
11
Weber
 
JA
Baxter
 
DH
Zhang
 
S
, et al. 
The microRNA spectrum in 12 body fluids.
Clin Chem
2010
, vol. 
56
 
11
(pg. 
1733
-
1741
)
12
Hanson
 
EK
Lubenow
 
H
Ballantyne
 
J
Identification of forensically relevant body fluids using a panel of differentially expressed microRNAs.
Anal Biochem
2009
, vol. 
387
 
2
(pg. 
303
-
314
)
13
Koga
 
Y
Yasunaga
 
M
Moriya
 
Y
Akasu
 
T
Fujita
 
S
Yamamoto
 
S
Matsumura
 
Y
Exosome can prevent RNase from degrading microRNA in feces.
J Gastrointest Oncol
2011
, vol. 
2
 
4
(pg. 
215
-
222
)
14
Keller
 
A
Leidinger
 
P
Bauer
 
A
, et al. 
Toward the blood-borne miRNome of human diseases.
Nat Methods
2011
, vol. 
8
 
10
(pg. 
841
-
843
)
15
Niyazi
 
M
Zehentmayr
 
F
Niemöller
 
OM
, et al. 
MiRNA expression patterns predict survival in glioblastoma.
Radiat Oncol
2011
, vol. 
6
 pg. 
153
 
16
Grasedieck
 
S
Schöler
 
N
Bommer
 
M
, et al. 
Impact of serum storage conditions on microRNA stability.
Leukemia
2012
, vol. 
26
 
11
(pg. 
2414
-
2416
)
17
Arroyo
 
JD
Chevillet
 
JR
Kroh
 
EM
, et al. 
Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
12
(pg. 
5003
-
5008
)
18
Wang
 
K
Zhang
 
S
Weber
 
J
Baxter
 
D
Galas
 
DJ
Export of microRNAs and microRNA-protective protein by mammalian cells.
Nucleic Acids Res
2010
, vol. 
38
 
20
(pg. 
7248
-
7259
)
19
Vickers
 
KC
Palmisano
 
BT
Shoucri
 
BM
Shamburek
 
RD
Remaley
 
AT
MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins.
Nat Cell Biol
2011
, vol. 
13
 
4
(pg. 
423
-
433
)
20
Chen
 
TS
Lai
 
RC
Lee
 
MM
Choo
 
AB
Lee
 
CN
Lim
 
SK
Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs.
Nucleic Acids Res
2010
, vol. 
38
 
1
(pg. 
215
-
224
)
21
Mittelbrunn
 
M
Gutiérrez-Vázquez
 
C
Villarroya-Beltri
 
C
, et al. 
Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells.
Nat Commun
2011
, vol. 
2
 pg. 
282
 
22
Katakowski
 
M
Buller
 
B
Wang
 
X
Rogers
 
T
Chopp
 
M
Functional microRNA is transferred between glioma cells.
Cancer Res
2010
, vol. 
70
 
21
(pg. 
8259
-
8263
)
23
Camussi
 
G
Deregibus
 
MC
Bruno
 
S
Grange
 
C
Fonsato
 
V
Tetta
 
C
Exosome/microvesicle-mediated epigenetic reprogramming of cells.
Am J Cancer Res
2011
, vol. 
1
 
1
(pg. 
98
-
110
)
24
Zhang
 
Y
Liu
 
D
Chen
 
X
, et al. 
Secreted monocytic miR-150 enhances targeted endothelial cell migration.
Mol Cell
2010
, vol. 
39
 
1
(pg. 
133
-
144
)
25
Nolte-’t Hoen
 
EN
Buermans
 
HP
Waasdorp
 
M
Stoorvogel
 
W
Wauben
 
MH
’t Hoen
 
PA
Deep sequencing of RNA from immune cell-derived vesicles uncovers the selective incorporation of small non-coding RNA biotypes with potential regulatory functions.
Nucleic Acids Res
2012
, vol. 
40
 
18
(pg. 
9272
-
9285
)
26
Jaiswal
 
R
Luk
 
F
Gong
 
J
Mathys
 
JM
Grau
 
GE
Bebawy
 
M
Microparticle conferred microRNA profiles—implications in the transfer and dominance of cancer traits.
Mol Cancer
2012
, vol. 
11
 
1
pg. 
37
 
27
Li
 
H
Huang
 
S
Guo
 
C
Guan
 
H
Xiong
 
C
Cell-free seminal mRNA and microRNA exist in different forms.
PLoS ONE
2012
, vol. 
7
 
4
pg. 
e34566
 
28
Bala
 
S
Petrasek
 
J
Mundkur
 
S
, et al. 
Circulating microRNAs in exosomes indicate hepatocyte injury and inflammation in alcoholic, drug-induced, and inflammatory liver diseases.
Hepatology
2012
, vol. 
56
 
5
(pg. 
1946
-
1957
)
29
Palma
 
J
Yaddanapudi
 
SC
Pigati
 
L
, et al. 
MicroRNAs are exported from malignant cells in customized particles.
Nucleic Acids Res
2012
, vol. 
40
 
18
(pg. 
9125
-
9138
)
30
Rupp
 
AK
Rupp
 
C
Keller
 
S
, et al. 
Loss of EpCAM expression in breast cancer derived serum exosomes: role of proteolytic cleavage.
Gynecol Oncol
2011
, vol. 
122
 
2
(pg. 
437
-
446
)
31
Wang
 
K
Yuan
 
Y
Cho
 
JH
McClarty
 
S
Baxter
 
D
Galas
 
DJ
Comparing the MicroRNA spectrum between serum and plasma.
PLoS ONE
2012
, vol. 
7
 
7
pg. 
e41561
 
32
van Schooneveld
 
E
Wouters
 
MC
Van der Auwera
 
I
, et al. 
Expression profiling of cancerous and normal breast tissues identifies microRNAs that are differentially expressed in serum from patients with (metastatic) breast cancer and healthy volunteers.
Breast Cancer Res
2012
, vol. 
14
 
1
pg. 
R34
 
33
McDonald
 
JS
Milosevic
 
D
Reddi
 
HV
Grebe
 
SK
Algeciras-Schimnich
 
A
Analysis of circulating microRNA: preanalytical and analytical challenges.
Clin Chem
2011
, vol. 
57
 
6
(pg. 
833
-
840
)
34
Kirschner
 
MB
Kao
 
SC
Edelman
 
JJ
Armstrong
 
NJ
Vallely
 
MP
van Zandwijk
 
N
Reid
 
G
Haemolysis during sample preparation alters microRNA content of plasma.
PLoS ONE
2011
, vol. 
6
 
9
pg. 
e24145
 
35
Pritchard
 
CC
Kroh
 
E
Wood
 
B
, et al. 
Blood cell origin of circulating microRNAs: a cautionary note for cancer biomarker studies.
Cancer Prev Res (Phila)
2012
, vol. 
5
 
3
(pg. 
492
-
497
)
36
Blondal
 
T
Jensby Nielsen
 
S
Baker
 
A
Andreasen
 
D
Mouritzen
 
P
Wrang Teilum
 
M
Dahlsveen
 
IK
Assessing sample and miRNA profile quality in serum and plasma or other biofluids.
Methods
2013
, vol. 
59
 
1
(pg. 
S1
-
S6
)
37
Kim
 
YK
Yeo
 
J
Kim
 
B
Ha
 
M
Kim
 
VN
Short structured RNAs with low GC content are selectively lost during extraction from a small number of cells.
Mol Cell
2012
, vol. 
46
 
6
(pg. 
893
-
895
)
38
Kumar
 
R
Singh
 
SK
Koshkin
 
AA
Rajwanshi
 
VK
Meldgaard
 
M
Wengel
 
J
The first analogues of LNA (locked nucleic acids): phosphorothioate-LNA and 2′-thio-LNA.
Bioorg Med Chem Lett
1998
, vol. 
8
 
16
(pg. 
2219
-
2222
)
39
Geiss
 
GK
Bumgarner
 
RE
Birditt
 
B
, et al. 
Direct multiplexed measurement of gene expression with color-coded probe pairs.
Nat Biotechnol
2008
, vol. 
26
 
3
(pg. 
317
-
325
)
40
Alhasan
 
AH
Kim
 
DY
Daniel
 
WL
Watson
 
E
Meeks
 
JJ
Thaxton
 
CS
Mirkin
 
CA
Scanometric microRNA array profiling of prostate cancer markers using spherical nucleic acid-gold nanoparticle conjugates.
Anal Chem
2012
, vol. 
84
 
9
(pg. 
4153
-
4160
)
41
Linsen
 
SE
de Wit
 
E
Janssens
 
G
, et al. 
Limitations and possibilities of small RNA digital gene expression profiling.
Nat Methods
2009
, vol. 
6
 
7
(pg. 
474
-
476
)
42
Jensen
 
SG
Lamy
 
P
Rasmussen
 
MH
Ostenfeld
 
MS
Dyrskjøt
 
L
Orntoft
 
TF
Andersen
 
CL
Evaluation of two commercial global miRNA expression profiling platforms for detection of less abundant miRNAs.
BMC Genomics
2011
, vol. 
12
 pg. 
435
 
43
Leshkowitz
 
D
Horn-Saban
 
S
Parmet
 
Y
Feldmesser
 
E
Differences in microRNA detection levels are technology and sequence dependent.
RNA
2013
, vol. 
19
 
4
(pg. 
527
-
538
)
44
Kuchenbauer
 
F
Morin
 
RD
Argiropoulos
 
B
, et al. 
In-depth characterization of the microRNA transcriptome in a leukemia progression model.
Genome Res
2008
, vol. 
18
 
11
(pg. 
1787
-
1797
)
45
Schopman
 
NC
Heynen
 
S
Haasnoot
 
J
Berkhout
 
B
A miRNA-tRNA mix-up: tRNA origin of proposed miRNA.
RNA Biol
2010
, vol. 
7
 
5
(pg. 
573
-
576
)
46
Pritchard
 
CC
Cheng
 
HH
Tewari
 
M
MicroRNA profiling: approaches and considerations.
Nat Rev Genet
2012
, vol. 
13
 
5
(pg. 
358
-
369
)
47
Turchinovich
 
A
Weiz
 
L
Langheinz
 
A
Burwinkel
 
B
Characterization of extracellular circulating microRNA.
Nucleic Acids Res
2011
, vol. 
39
 
16
(pg. 
7223
-
7233
)
48
Mitchell
 
PS
Parkin
 
RK
Kroh
 
EM
, et al. 
Circulating microRNAs as stable blood-based markers for cancer detection.
Proc Natl Acad Sci USA
2008
, vol. 
105
 
30
(pg. 
10513
-
10518
)
49
Qi
 
R
Weiland
 
M
Gao
 
XH
Zhou
 
L
Mi
 
QS
Identification of endogenous normalizers for serum microRNAs by microarray profiling: U6 small nuclear RNA is not a reliable normalizer.
Hepatology
2012
, vol. 
55
 
5
(pg. 
1640
-
1642, author reply 1642-1643
)
50
Baraniskin
 
A
Nöpel-Dünnebacke
 
S
Ahrens
 
M
, et al. 
Circulating U2 small nuclear RNA fragments as a novel diagnostic biomarker for pancreatic and colorectal adenocarcinoma.
Int J Cancer
2013
, vol. 
132
 
2
(pg. 
E48
-
E57
)
51
D’haene
 
B
Mestdagh
 
P
Hellemans
 
J
Vandesompele
 
J
miRNA expression profiling: from reference genes to global mean normalization.
Methods Mol Biol
2012
, vol. 
822
 (pg. 
261
-
272
)
52
Cookson
 
VJ
Bentley
 
MA
Hogan
 
BV
Horgan
 
K
Hayward
 
BE
Hazelwood
 
LD
Hughes
 
TA
Circulating microRNA profiles reflect the presence of breast tumours but not the profiles of microRNAs within the tumours.
Cell Oncol (Dordr)
2012
, vol. 
35
 
4
(pg. 
301
-
308
)
53
Yoshizawa
 
S
Ohyashiki
 
JH
Ohyashiki
 
M
, et al. 
Downregulated plasma miR-92a levels have clinical impact on multiple myeloma and related disorders.
Blood Cancer J
2012
, vol. 
2
 
1
pg. 
e53
 
54
Ohyashiki
 
K
Umezu
 
T
Yoshizawa
 
S
, et al. 
Clinical impact of down-regulated plasma miR-92a levels in non-Hodgkin’s lymphoma.
PLoS ONE
2011
, vol. 
6
 
2
pg. 
e16408
 
55
Guo
 
HQ
Huang
 
GL
Guo
 
CC
Pu
 
XX
Lin
 
TY
Diagnostic and prognostic value of circulating miR-221 for extranodal natural killer/T-cell lymphoma.
Dis Markers
2010
, vol. 
29
 
5
(pg. 
251
-
258
)
56
Baraniskin
 
A
Kuhnhenn
 
J
Schlegel
 
U
, et al. 
Identification of microRNAs in the cerebrospinal fluid as marker for primary diffuse large B-cell lymphoma of the central nervous system.
Blood
2011
, vol. 
117
 
11
(pg. 
3140
-
3146
)
57
Lawrie
 
CH
Gal
 
S
Dunlop
 
HM
, et al. 
Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma.
Br J Haematol
2008
, vol. 
141
 
5
(pg. 
672
-
675
)
58
Fang
 
C
Zhu
 
DX
Dong
 
HJ
, et al. 
Serum microRNAs are promising novel biomarkers for diffuse large B cell lymphoma.
Ann Hematol
2012
, vol. 
91
 
4
(pg. 
553
-
559
)
59
Zuo
 
Z
Calin
 
GA
de Paula
 
HM
, et al. 
Circulating microRNAs let-7a and miR-16 predict progression-free survival and overall survival in patients with myelodysplastic syndrome.
Blood
2011
, vol. 
118
 
2
(pg. 
413
-
415
)
60
Jones
 
CI
Zabolotskaya
 
MV
King
 
AJ
Stewart
 
HJ
Horne
 
GA
Chevassut
 
TJ
Newbury
 
SF
Identification of circulating microRNAs as diagnostic biomarkers for use in multiple myeloma.
Br J Cancer
2012
, vol. 
107
 
12
(pg. 
1987
-
1996
)
61
Huang
 
JJ
Yu
 
J
Li
 
JY
Liu
 
YT
Zhong
 
RQ
Circulating microRNA expression is associated with genetic subtype and survival of multiple myeloma.
Med Oncol
2012
, vol. 
29
 
4
(pg. 
2402
-
2408
)
62
Tanaka
 
M
Oikawa
 
K
Takanashi
 
M
Kudo
 
M
Ohyashiki
 
J
Ohyashiki
 
K
Kuroda
 
M
Down-regulation of miR-92 in human plasma is a novel marker for acute leukemia patients.
PLoS ONE
2009
, vol. 
4
 
5
pg. 
e5532
 
63
Ishihara
 
K
Sasaki
 
D
Tsuruda
 
K
, et al. 
Impact of miR-155 and miR-126 as novel biomarkers on the assessment of disease progression and prognosis in adult T-cell leukemia.
Cancer Epidemiol
2012
, vol. 
36
 
6
(pg. 
560
-
565
)
64
Schöler
 
N
Langer
 
C
Kuchenbauer
 
F
Circulating microRNAs as biomarkers - True Blood?
Genome Med
2011
, vol. 
3
 
11
pg. 
72
 
65
Moussay
 
E
Wang
 
K
Cho
 
JH
, et al. 
MicroRNA as biomarkers and regulators in B-cell chronic lymphocytic leukemia.
Proc Natl Acad Sci USA
2011
, vol. 
108
 
16
(pg. 
6573
-
6578
)
66
Zenz
 
T
Mertens
 
D
Küppers
 
R
Döhner
 
H
Stilgenbauer
 
S
From pathogenesis to treatment of chronic lymphocytic leukaemia.
Nat Rev Cancer
2010
, vol. 
10
 
1
(pg. 
37
-
50
)
67
Umezu
 
T
Ohyashiki
 
K
Kuroda
 
M
Ohyashiki
 
JH
Leukemia cell to endothelial cell communication via exosomal miRNAs [published online ahead of print July 16, 2012].
Oncogene
68
Fabbri
 
M
Paone
 
A
Calore
 
F
, et al. 
MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
31
(pg. 
E2110
-
E2116
)
69
Yuan
 
A
Farber
 
EL
Rapoport
 
AL
Tejada
 
D
Deniskin
 
R
Akhmedov
 
NB
Farber
 
DB
Transfer of microRNAs by embryonic stem cell microvesicles.
PLoS ONE
2009
, vol. 
4
 
3
pg. 
e4722
 
70
Collino
 
F
Deregibus
 
MC
Bruno
 
S
, et al. 
Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs.
PLoS ONE
2010
, vol. 
5
 
7
pg. 
e11803
 
71
Grange
 
C
Tapparo
 
M
Collino
 
F
, et al. 
Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche.
Cancer Res
2011
, vol. 
71
 
15
(pg. 
5346
-
5356
)
72
Hergenreider
 
E
Heydt
 
S
Tréguer
 
K
, et al. 
Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs.
Nat Cell Biol
2012
, vol. 
14
 
3
(pg. 
249
-
256
)
73
Fonsato
 
V
Collino
 
F
Herrera
 
MB
, et al. 
Human liver stem cell-derived microvesicles inhibit hepatoma growth in SCID mice by delivering antitumor microRNAs.
Stem Cells
2012
, vol. 
30
 
9
(pg. 
1985
-
1998
)
74
Xin
 
H
Li
 
Y
Buller
 
B
, et al. 
Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth.
Stem Cells
2012
, vol. 
30
 
7
(pg. 
1556
-
1564
)
75
Ismail
 
N
Wang
 
Y
Dakhlallah
 
D
, et al. 
Macrophage microvesicles induce macrophage differentiation and miR-223 transfer.
Blood
2013
, vol. 
121
 
6
(pg. 
984
-
995
)
76
Cantaluppi
 
V
Gatti
 
S
Medica
 
D
, et al. 
Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells.
Kidney Int
2012
, vol. 
82
 
4
(pg. 
412
-
427
)
77
Montecalvo
 
A
Larregina
 
AT
Shufesky
 
WJ
, et al. 
Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes.
Blood
2012
, vol. 
119
 
3
(pg. 
756
-
766
)
78
Katsman
 
D
Stackpole
 
EJ
Domin
 
DR
Farber
 
DB
Embryonic stem cell-derived microvesicles induce gene expression changes in Müller cells of the retina.
PLoS ONE
2012
, vol. 
7
 
11
pg. 
e50417
 
79
Zernecke
 
A
Bidzhekov
 
K
Noels
 
H
, et al. 
Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection.
Sci Signal
2009
, vol. 
2
 
100
pg. 
ra81
 
80
Salvucci
 
O
Jiang
 
K
Gasperini
 
P
, et al. 
MicroRNA126 contributes to granulocyte colony-stimulating factor-induced hematopoietic progenitor cell mobilization by reducing the expression of vascular cell adhesion molecule 1.
Haematologica
2012
, vol. 
97
 
6
(pg. 
818
-
826
)
81
Langer
 
C
Rücker
 
FG
Buske
 
C
Döhner
 
H
Kuchenbauer
 
F
Targeted therapies through microRNAs: pulp or fiction?
Ther Adv Hematol
2012
, vol. 
3
 
2
(pg. 
97
-
104
)
82
Leone
 
E
Morelli
 
E
Di Martino
 
MT
, et al. 
Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growth[published online ahead of print February 27, 2013].
Clin Cancer Res
83
Babar
 
IA
Cheng
 
CJ
Booth
 
CJ
Liang
 
X
Weidhaas
 
JB
Saltzman
 
WM
Slack
 
FJ
Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma.
Proc Natl Acad Sci USA
2012
, vol. 
109
 
26
(pg. 
E1695
-
E1704
)
84
Katakowski
 
M
Buller
 
B
Zheng
 
X
, et al. 
Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth[published online ahead of print February 16, 2013].
Cancer Lett
85
Tabatadze
 
N
Savonenko
 
A
Song
 
H
Bandaru
 
VV
Chu
 
M
Haughey
 
NJ
Inhibition of neutral sphingomyelinase-2 perturbs brain sphingolipid balance and spatial memory in mice.
J Neurosci Res
2010
, vol. 
88
 
13
(pg. 
2940
-
2951
)
86
Baraniskin
 
A
Kuhnhenn
 
J
Schlegel
 
U
Schmiegel
 
W
Hahn
 
S
Schroers
 
R
MicroRNAs in cerebrospinal fluid as biomarker for disease course monitoring in primary central nervous system lymphoma.
J Neurooncol
2012
, vol. 
109
 
2
(pg. 
239
-
244
)

Supplemental data

Sign in via your Institution