Abstract
microRNAs (miRNAs) constitute a large family of small, approximately 20–22 nucleotide non-coding RNAs that regulate the expression of target genes, mainly at the post-transcriptional level. Multiple studies report that miRNAs are involved in homeostatic maintenance and that aberrant expression of miRNAs is often observed in various types of diseases, including cancer. In cancer biology, miRNAs exert functional roles in tumor initiation, drug resistance, and metastasis. miRNAs are also secreted through small vesicles called exosomes, which are endosome-derived vesicles derived from various cell types including immune and tumor cells. In addition to cellular miRNAs (ce-miRNAs), secreted miRNAs (se-miRNAs) play important roles in cancer development and metastasis. Therefore, se-miRNAs in body fluids have been investigated as a promising biomarkers and therapeutic targets for cancer treatment. In this review, we summarize the current knowledge of miRNA functions in cancer development and discuss the potential clinical applications of se-miRNAs, e.g. as diagnostic markers and therapeutic targets.
Introduction
Several lines of evidence indicate that microRNA (miRNAs) are critical regulators of global messenger RNA (mRNA) expression in normal and abnormal biological processes [1], [2], [3]. Aberrant expression of miRNAs has been observed in multiple types of cancers and is associated with malignant cancer phenotypes, including metastasis and drug resistance. Therefore, manipulation of miRNA expression represents a promising approach to cancer therapy. Several recent studies report that miRNAs are secreted through exosomes, and that the levels of circulating miRNAs packaged in exosomes differ between cancer patients and healthy donors [4], [5], [6]. More importantly, the contents and amount of secreted microRNA (se-miRNAs) are associated with disease stage and regulation of the malignant phenotype [7], [8], [9]. In this review, we summarize the major findings regarding se-miRNA function in tumor development, and discuss how detection of se-miRNAs and removal of cancer-derived exosomes could be applied to clinical diagnosis and treatment.
Biosynthesis and functions of miRNAs
miRNAs are approximately 20–22-nucleotide non-coding RNAs that regulate gene expression at the post-transcriptional level by binding to the 3′-untranslated regions (3′UTRs) of target genes or to imperfectly complementary sequences called miRNA response elements (MREs). The binding of miRNAs to target genes mainly leads to the degradation or translational inhibition [10] (Figure 1). miRNAs are transcribed, mainly by RNA polymerase II, as long primary transcripts containing hairpin structures (pri-miRNAs). In the nucleus, these transcripts are cleaved into 70–100 nucleotide precursor miRNAs (pre-miRNAs) by a microprocessor complex containing RNase III Drosha and DGCR8. The pre-miRNAs are exported to the cytoplasm by exportin-5, a member of the Ran-dependent nuclear transport receptor family [11], and further processed into a miRNA:miRNA* complex by a complex containing RNase III Dicer and the transactivating response RNA-binding protein (TRBP).

Biogenesis and function of cellular and exosomal microRNA.
miRNAs are transcribed by RNA polymerase II or III as pri-miRNA, and then processed in the nucleus by Drosha-DGCR8 into pre-miRNA. The pre-miRNA is exported to the cytoplasm by exportin-5 and further cleaved by a complex composed of Dicer and TRBP. The functional strand of mature miRNA is incorporated into the RNA-induced silencing complex (RISC), which contains GW182 and Argonaute protein. As a component of this complex, the mature miRNA regulates gene expression by binding to partially complementary sequences in the 3′UTRs or coding regions of target mRNAs, leading to mRNA degradation or translational repression.
One of the two strands (the miRNA) is selected as a guide strand, and the complementary strand (miRNA*) is usually degraded [12]. However, recent evidence suggests that miRNA* can also serve as a functional strand, and may play significant biological roles [13]. The mature miRNA is incorporated into the RNA-induced silencing complex (RISC), which contains the GW182 and Argonaute (AGO) proteins. As a component of this complex, the mature miRNA regulates gene expression by binding to partially complementary sequences in the 3′UTRs or MREs of target mRNAs, leading to mRNA degradation or translational repression [12].
Secretion of miRNAs
In 1979, Taylor and Doellgast first reported that several types of tumor cells release or shed intact microvesicles composed of membrane proteins [14]. Subsequently, a number of studies reported that mRNA, miRNAs, and long non-coding RNAs are secreted from different types of cells through extracellular vesicles (EVs) [15] and play important roles in cell-to-cell communications such as inflammatory response, tissue regeneration, and tumor progression [16], [17], [18], [19], [20].
EVs are mainly classified into the following three types: exosomes (30–100 nm), microvesicles (100–1000 nm), and oncosomes (1–10 μm); each of these three vesicle types has a unique size and function in intracellular communication [21], [22], [23], [24]. Although the molecular mechanisms of EV biogenesis and secretion are not fully understood and depend on cell type [25], neutral sphingomyelinase 2 and some Rab GTPases, such as Rab11, Rab27a and Rab27b, have been reported to regulate EV secretion [26], [27], [28]. Because EVs are secreted from various types of cells under both physiological and pathological conditions, and their RNA and protein contents differ between patients and healthy donors [29], [30], [31], [32], these vesicles are increasingly recognized as promising biomarkers for the diagnosis and prognosis of various diseases.
Se-mRNAs in cancer
Cellular microRNAs (ce-miRNAs) can be classified as tumor-suppressive or oncogenic and play important roles in tumor development such as tumor initiation, acquisition of drug resistance, and metastasis [33], [34], [35]. Likewise, se-miRNAs in exosomes (exosomal miRNAs) are also important for tumor progression and metastasis in multiple cancers [7], [9], [20], [36], [37] (Figure 2). In addition to these findings, Melo et al. [8] reported that exosomes derived from breast cancer cells contain pre-miRNAs that interact with the RISC complex and exhibit cell-independent generation of mature miRNAs from pre-miRNAs. In addition, McKenzie et al. [42] reported that the secretion of AGO2 into exosomes depends on the KRAS mutation status of colon cancer cells. Since AGO2 is important for ce-miRNA-mediated gene expression, these findings suggest that se-miRNA function is also regulated by the activity and mutation status of oncogenes in donor cells. Therefore, many cancer researchers are trying to remove or antagonize tumor-derived exosomes as a novel approach to cancer therapy or to develop sensitive systems for detecting se-miRNAs in patients’ body fluids.

Biogenesis and function of exosomal microRNA.
The endosomal system is composed of primary endocytic vesicles, early endosomes, and multivesicular bodies (MVBs). Primary endocytosed vesicles are recycled to the plasma membrane or transferred to MVBs. The proteins and miRNAs that are entrapped into the limiting membrane of MVBs can be selectively packaged into intraluminal vesicles (ILVs) by invagination of the MVB membrane. MVBs can fuse either with the lysosome for degradation or with the plasma membrane for the release of exosomes into the extracellular space. Exosomal miRNAs are transferred from donor cells to recipient cells and function as cellular miRNAs for gene regulation.
The roles of se-miRNAs in cancer development
Tumorigenesis
Antonyak et al. [38] demonstrated that EVs from MDA-MB-231 breast cancer and U87 glioblastoma cell lines induce normal fibroblast and epithelial cells to acquire the transformed characteristics of cancer cells, such as anchorage-independent growth and improved survival. Exosomal miRNAs also promote the acquisition of tumor-seeding ability in non-tumorigenic human breast cells [8]. In contrast to exosomes from healthy donors, those from breast cancer patient serum induce MCF10A cells to form tumors in immunodeficient mice. Interestingly, Dicer protein is more abundant in exosomes from cancer patients. Recently, Gutkin et al. [43] reported that hTERT mRNA is also packaged into exosomes and supports the transformation of fibroblasts into telomerase positive cells. Considering these findings, exosomal miRNAs and mRNAs might play an important role in the transformation of normal cells into malignant cells.
Tumor metastasis
Several studies reported that exosomes are among of the most important factors involved in pre-metastatic niche formation and organotropism in metastasis [36], [44], [45]. In pancreatic cancer, exosomes from pancreatic ductal adenocarcinomas (PDACs) induce liver pre-metastatic niche formation in mice [45]. Exosomes derived form PDACs promote Kupffer cell-mediated TGF-β secretion, which enhances fibronectin production in hepatic stellate cells. Fibronectin accumulation in hepatic stellate cells induces the formation of the pre-metastatic niche by recruiting bone marrow-derived macrophages to the liver.
The organotropisms of metastatic cancer cells are determined by the combination of integrin family proteins displayed on exosomes [44]. While exosomes expressing integrin αvβ5 specifically bind Kupffer cells and support the liver tropism of colon and pancreatic cancer cells, exosomes expressing integrin α6β4 and α6β1 support the lung metastasis of breast cancer cells via binding to lung-resident fibroblasts and epithelial cells.
Exosomal miRNAs are also important for tumor metastasis [7], [9], [20]. Zhou et al. [9] reported that high levels of miR-105 are present in metastatic breast cancer cells and their exosomes, and that exosomal miR-105 promotes tumor metastasis by directly targeting zonula occludens 1 (ZO-1), which regulates tight junction and supports the barrier function of endothelial monolayers. Exosomal miR-181c also promotes the brain metastasis of breast cancer cells by destroying the blood-brain barrier [20]. MiR-181 induces the delocalization of actin fibers via downregulation of 3-phosphoinositide-dependent protein kinase-1, leading to the disruption of intercellular junctions in brain endothelial cells. Fong et al. [7] reported that miR-122 promotes breast cancer metastasis by regulating glucose metabolism in pre-metastatic sites. Interestingly, in contrast to the intracellular level of miR-122, which does not significantly differ between non-metastatic and metastatic cancer cells, the level of exosomal miR-122 is correlated with the metastatic capacity of breast cancer cells. Exosomal miR-122 regulates glucose metabolism through the direct target of pyruvate kinase isozymes M2 (PKM2) in pre-metastatic sites, resulting in the formation of the pre-metastatic niche.
Drug resistance
The main mechanisms underlying the acquisition of drug resistance can be classified into three categories: (1) up-regulation of drug transporters that export anti-cancer agents [46]; (2) up-regulation of anti-apoptotic genes [47]; and (3) activation of cell survival pathways [48]. In addition to these important pathways related to drug resistance, the epithelial-to-mesenchymal transition (EMT) was originally considered to be important for both tumor metastasis and drug resistance; however, more recent evidence suggests that EMT contributes mainly to drug resistance [49], [50]. Several miRNAs have been identified as critical regulators of drug resistance in various types of cancer through the pathways described above [33], [51], [52], [53], [54], [55], [56].
Se-miRNAs are also important for the regulation of drug resistance [39], [40], [41]. Chen et al. reported that the level of miR-222 is elevated in exosomes derived from docetaxel-resistant breast cancer cells, and that exosomal miR-222 is transferred into drug-sensitive breast cancer cells [41]. Challagundla et al. [40] reported that exosomal miR-21 and miR-155 promote the acquisition of chemoresistance by human neuroblastoma (NBL) cells. Transfer of exosomal miR-21 from NBL cells to human monocytes and exosomal miR-155 from human monocytes to NBL cells is important for the tumor microenvironment, and such crosstalk promotes resistance to chemotherapy. Exosomal miR-21 from NBL cells induced Toll-like receptor 8 and nuclear factor-κB (NF-κB)-dependent up-regulation of miR-155 in monocytes, leading to the production of miR-155–containing exosomes. Exosomal miR-155 promotes CDDP resistance in NBL cells through the direct suppression of TERF1, a component of the shelterin complex and inhibitor of telomerase. Au Yeung et al. [39] reported that exosomal miR-21 from cancer-associated adipocytes (CAAs) and fibroblasts (CAFs) promotes paclitaxel resistance in ovarian cancer cells. Exosomal miR-21 from CAAs and CAFs is transferred into ovarian cancer cells, where it inhibits apoptosis and confers resistance to paclitaxel via direct suppression of APAF1 [39], [57].
Exosome-based strategies for cancer diagnosis and therapy
Cancer diagnosis and therapy targeting exosomes
Given that exosomes and exosomal miRNAs from cancer tissues are important for tumor development processes such as tumor seeding, drug resistance, and metastasis (see Section “The roles of se-miRNAs in cancer development”), many clinical oncologists and cancer researchers are currently trying to develop novel methods for cancer diagnosis and therapies based on detection and removal (or antagonism) of tumor-derived exosomes.
Diagnosis
Because exosomal miRNAs and circulating RNAs are associated with secreted proteins such as CD63, Ago proteins, and HDL [17], [58], [59], they are stable in body fluids and easily detectable using quantitative PCR methods. Several studies have shown that exosomal miRNAs can be detected in a variety of body fluids, from blood to breast milk, and that their levels are correlated with patient status [4], [5], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73] (Table 1). On January 21, 2016, cancer diagnosis using exosomes from blood became commercially available in the US (http://www.exosomedx.com) [6].
Profiles of se-miRNAs in cancer patients.
Cancer | Body fluids | microRNAs | Reference |
---|---|---|---|
Glioma | Cerebrospinal fluid (exosome) | ↑ miR-21 | [73] |
Oral cancer | Blood (plasma) Blood (plasma) Blood (plasma) | ↑ miR-181c ↑ miR-21 ↑ miR-196 | [61], [62], [63] |
Lung cancer | Blood (plasma/exosome) Blood (exosome) Blood (plasma) | ↑ miR-205, -19a, -19b, -30b, and -20a ↑ miR-378a, miR-379, miR-139-5p, and miR-200b-5p Down-regulation after RT ↑miR-29a and miR-150 | [64], [65], [66] |
Colon cancer | Blood (exosome) Blood (Serum) Blood (Serum) | ↑ let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23a ↑ miR-23a, miR-27a, miR-142-5p and miR-376c ↑ miR-203 | [67], [68], [69] |
Ovarian cancer | Blood (exosome) Blood (exosome) | ↑ miR-21, miR-141, miR-200a, miR-200b, miR-203, miR-205 and miR-214 ↑ miR-200b and 200c | [4], [5] |
Breast Cancer | Blood (Serum) | Combination of miRNA profile![]() | [60] |
Gastric cancer | Blood (plasma) | 1 miR-26a, miR-142-3p, miR-148a, and miR-195 | [71] |
Prostate cancer | Urine (exosome) | T miR-574-3p, miR-141-5p and miR-21-5p | [72] |
RT, radiation therapy.
Like cellular miRNA expression profiles, the expression profiles of cell-free and circulating miRNAs are also correlated with the pathological condition of cancer patients [4], [60]. Therefore, a considerable amount of research has been expended in trying to develop novel methods for the detection and characterization of microvesicles in the blood of patients classified according to clinical stage [74], [75], [76]. Im et al. [74] developed a surface plasmon resonance-based system for the detection of exosomal proteins. Using this system, they identified CD24 and EpCAM as specific markers for tumor-derived exosomes derived from ovarian cancers. Yoshioka et al. [75] also established a highly sensitive and rapid analytical method, ExoScreen, for detecting and characterizing exosomes from blood samples of colon cancer patients. These reports suggest that exosomes are promising biomarkers for the early detection of cancers as well as for monitoring therapy response and patient condition.
Several studies have reported that specific exosomal miRNAs are associated with the condition of cancer patients [5], [67], [73]. In epithelial ovarian cancer (EOC), serum exosome levels are elevated, and high levels of exosomal miR-200b and miR-200c are observed mainly in patients with advanced EOC [5]. Ogata-Kawata et al. [67] found that in comparison to healthy controls, colon cancer patients, even in the early stages, exhibited significantly higher levels of seven exosomal miRNAs (let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223, and miR-23a). Shi et al. [73] reported that increases in the level of exosomal miR-21 in cerebrospinal fluid correlate with poor prognosis and cancer recurrence in glioma patients.
Exosomal lincRNA and miRNA are also detected in the urine of prostate and bladder cancer patients [77], [78], [79]. Bryzgunova et al. [77] reported that the level of miR-19b, normalized to that of miR-16, in urine-derived exosomes was significantly lower in prostate cancer patients than in healthy donors . Armstrong et al. also reported that in bladder cancer, high levels of miR-21 and miR-4454 are commonly detected in whole blood cell, tumor, and urine. Recently, lncRNA HOX transcript antisense RNA (HOTAIR), which is associated with tumor progression and poor prognosis in several types of cancers [80], [81], [82], was reported in urinary exosomes from bladder cancer patients. Since urinary exosomes are a non-invasive source, combinatorial methods designed to detect simultaneously urinary miRNAs and lncRNA might provide a more precise and simple approach for the diagnosis of prostate and bladder cancers [83].
Exosome targeting therapy
Several studies report that exosomes from cancer cells attenuate the anti-tumor effects of immunotherapeutic drugs such as rituximab and trastuzumab by acting as a decoy for cancer cells [84], [85]. Therefore, cancer researchers are attempting to remove such exosomes from patient blood. Ciravolo et al. [84] also reported that the exosomes from HER2-positive breast cancer cells display HER2 protein on their surface, thereby competitively inhibiting the interaction between breast cancer cells and trastuzumab, and ultimately leading to tumor cell proliferation and poor prognosis. Aung et al. [85] found that lymphoma-derived exosomes that display CD20 protein on their surface inhibit the anti-tumor effects of the anti-CD20 chimeric antibody rituximab by direct binding. These findings suggest that reduction or removal of cancer exosomes would support immunotherapy against cancer-specific antigens.
Another approach is to trap metastatic cells using a 3D scaffold device, called an “M-Trap” [86]. Cancer exosomes from ovarian cancer patient ascites are embedded on the M-Trap, acting as a pre-metastatic niche. Therefore, this device effectively impairs the interaction between metastatic ovarian cancer cells and their niche. In addition to the exciting findings about the roles of exosomes in cancer metastasis described in subsection “Tumor metastasis” [20], [44], [45], these results suggest that exosomes are critical mediators in the crosstalk and homing of metastatic tumor cells to the niche in various types of cancers.
Conclusions
In this review, we summarized the roles of exosomal miRNA in cancer biology, including tumor initiation, drug resistance, and metastasis. Given that several lines of evidence demonstrate that miRNA profiles, not only in tumor tissues but also in body fluids, reflect the condition and tumor stage of cancer patients, development of highly sensitive and rapid systems for detecting exosomes and their RNAs should contribute to improvements in cancer diagnosis. In addition, targeting and removal of cancer exosomes represents a novel and promising approach for treatment of metastatic cancer. Accordingly, increasing our understanding of the molecular mechanisms of miRNA and exosome secretion, along with profiling of exosomal miRNAs in each tissues and body fluid, is an important goal for both basic and clinical cancer research.
Acknowledgments
This study was supported in part by a Grant-in-Aid for Scientific Research C (16K08261), the Japan Agency for Medical Research and Development (AMED), and the NOVARTIS Foundation (Japan) for the Promotion of Science.
Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.
Research funding: None declared.
Employment or leadership: None declared.
Honorarium: None declared.
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. Croce CM, Calin GA. miRNAs, cancer, and stem cell division. Cell 2005;122:6–7.10.1016/j.cell.2005.06.036Search in Google Scholar
2. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 2006;103:2257–61.10.1073/pnas.0510565103Search in Google Scholar
3. Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, et al. Relation between microRNA expression and progression and prognosis of gastric cancer: a microRNA expression analysis. Lancet Oncol 2010;11:136–46.10.1016/S1470-2045(09)70343-2Search in Google Scholar
4. Taylor DD, Gercel-Taylor C. MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol 2008;110:13–21.10.1016/j.ygyno.2008.04.033Search in Google Scholar PubMed
5. Meng X, Muller V, Milde-Langosch K, Trillsch F, Pantel K, Schwarzenbach H. Diagnostic and prognostic relevance of circulating exosomal miR-373, miR-200a, miR-200b and miR-200c in patients with epithelial ovarian cancer. Oncotarget 2016;7:16923–35.10.18632/oncotarget.7850Search in Google Scholar PubMed PubMed Central
6. Sheridan C. Exosome cancer diagnostic reaches market. Nat Biotechnol 2016;34:359–60.10.1038/nbt0416-359Search in Google Scholar PubMed
7. Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol 2015;17:183–94.10.1038/ncb3094Search in Google Scholar PubMed PubMed Central
8. Melo SA, Sugimoto H, O’Connell JT, Kato N, Villanueva A, Vidal A, et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell 2014;26:707–21.10.1016/j.ccell.2014.09.005Search in Google Scholar PubMed PubMed Central
9. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014;25:501–15.10.1016/j.ccr.2014.03.007Search in Google Scholar PubMed PubMed Central
10. Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, et al. The Microprocessor complex mediates the genesis of microRNAs. Nature 2004;432:235–40.10.1038/nature03120Search in Google Scholar PubMed
11. Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science 2004;303:95–8.10.1126/science.1090599Search in Google Scholar
12. Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 2012;4:143–59.10.1002/emmm.201100209Search in Google Scholar
13. Uchino K, Takeshita F, Takahashi RU, Kosaka N, Fujiwara K, Naruoka H, et al. Therapeutic effects of microRNA-582-5p and -3p on the inhibition of bladder cancer progression. Mol Ther 2013;21:610–9.10.1038/mt.2012.269Search in Google Scholar
14. Taylor DD, Doellgast GJ. Quantitation of peroxidase-antibody binding to membrane fragments using column chromatography. Anal Biochem 1979;98:53–9.10.1016/0003-2697(79)90704-8Search in Google Scholar
15. Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell 2016;29:653–68.10.1016/j.ccell.2016.03.004Search in Google Scholar PubMed
16. Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol 2009;11:1143–9.10.1038/ncb1929Search in Google Scholar PubMed
17. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654–9.10.1038/ncb1596Search in Google Scholar PubMed
18. Alexander M, Hu R, Runtsch MC, Kagele DA, Mosbruger TL, Tolmachova T, et al. Exosome-delivered microRNAs modulate the inflammatory response to endotoxin. Nat Commun 2015;6:7321.10.1038/ncomms8321Search in Google Scholar PubMed PubMed Central
19. Khan M, Nickoloff E, Abramova T, Johnson J, Verma SK, Krishnamurthy P, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res 2015;117:52–64.10.1161/CIRCRESAHA.117.305990Search in Google Scholar PubMed PubMed Central
20. Tominaga N, Kosaka N, Ono M, Katsuda T, Yoshioka Y, Tamura K, et al. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat Commun 2015;6:6716.10.1038/ncomms7716Search in Google Scholar PubMed PubMed Central
21. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200:373–83.10.1083/jcb.201211138Search in Google Scholar PubMed PubMed Central
22. Di Vizio D, Kim J, Hager MH, Morello M, Yang W, Lafargue CJ, et al. Oncosome formation in prostate cancer: association with a region of frequent chromosomal deletion in metastatic disease. Cancer Res 2009;69:5601–9.10.1158/0008-5472.CAN-08-3860Search in Google Scholar PubMed PubMed Central
23. Minciacchi VR, You S, Spinelli C, Morley S, Zandian M, Aspuria PJ, et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget 2015;6:11327–41.10.18632/oncotarget.3598Search in Google Scholar PubMed PubMed Central
24. Headley MB, Bins A, Nip A, Roberts EW, Looney MR, Gerard A, et al. Visualization of immediate immune responses to pioneer metastatic cells in the lung. Nature 2016;531:513–7.10.1038/nature16985Search in Google Scholar PubMed PubMed Central
25. Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: from biogenesis and secretion to biological function. Immunol Lett 2006;107:102–8.10.1016/j.imlet.2006.09.005Search in Google Scholar PubMed
26. Kosaka N, Iguchi H, Hagiwara K, Yoshioka Y, Takeshita F, Ochiya T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J Biol Chem 2013;288:10849–59.10.1074/jbc.M112.446831Search in Google Scholar PubMed PubMed Central
27. Savina A, Fader CM, Damiani MT, Colombo MI. Rab11 promotes docking and fusion of multivesicular bodies in a calcium-dependent manner. Traffic 2005;6:131–43.10.1111/j.1600-0854.2004.00257.xSearch in Google Scholar PubMed
28. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol 2010;12:19–30; sup pp 1–13.10.1038/ncb2000Search in Google Scholar PubMed
29. Taylor DD, Gercel-Taylor C. The origin, function, and diagnostic potential of RNA within extracellular vesicles present in human biological fluids. Front Genet 2013;4:142.10.3389/fgene.2013.00142Search in Google Scholar PubMed PubMed Central
30. Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, Gonzalez S, Sanchez-Cabo F, Gonzalez MA, et al. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nat Commun 2011;2:282.10.1038/ncomms1285Search in Google Scholar PubMed PubMed Central
31. McDonald MK, Tian Y, Qureshi RA, Gormley M, Ertel A, Gao R, et al. Functional significance of macrophage-derived exosomes in inflammation and pain. Pain 2014;155:1527–39.10.1016/j.pain.2014.04.029Search in Google Scholar PubMed PubMed Central
32. Putz U, Howitt J, Doan A, Goh CP, Low LH, Silke J, et al. The tumor suppressor PTEN is exported in exosomes and has phosphatase activity in recipient cells. Sci Signal 2012;5:ra70.10.1126/scisignal.2003084Search in Google Scholar PubMed
33. Takahashi RU, Miyazaki H, Takeshita F, Yamamoto Y, Minoura K, Ono M, et al. Loss of microRNA-27b contributes to breast cancer stem cell generation by activating ENPP1. Nat Commun 2015;6:7318.10.1038/ncomms8318Search in Google Scholar PubMed PubMed Central
34. Bu P, Wang L, Chen KY, Srinivasan T, Murthy PK, Tung KL, et al. A miR-34a-numb feedforward loop triggered by inflammation regulates asymmetric stem cell division in intestine and colon cancer. Cell Stem Cell 2016;18:189–202.10.1016/j.stem.2016.01.006Search in Google Scholar PubMed PubMed Central
35. Ryu S, McDonnell K, Choi H, Gao D, Hahn M, Joshi N, et al. Suppression of miRNA-708 by polycomb group promotes metastases by calcium-induced cell migration. Cancer Cell 2013;23:63–76.10.1016/j.ccr.2012.11.019Search in Google Scholar PubMed
36. Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012;18:883–91.10.1038/nm.2753Search in Google Scholar PubMed PubMed Central
37. Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012;151:1542–56.10.1016/j.cell.2012.11.024Search in Google Scholar PubMed
38. Antonyak MA, Li B, Boroughs LK, Johnson JL, Druso JE, Bryant KL, et al. Cancer cell-derived microvesicles induce transformation by transferring tissue transglutaminase and fibronectin to recipient cells. Proc Natl Acad Sci USA 2011;108:4852–7.10.1073/pnas.1017667108Search in Google Scholar PubMed PubMed Central
39. Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun 2016;7:11150.10.1038/ncomms11150Search in Google Scholar PubMed PubMed Central
40. Challagundla KB, Wise PM, Neviani P, Chava H, Murtadha M, Xu T, et al. Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. J Natl Cancer Inst 2015;107:13.10.1093/jnci/djv135Search in Google Scholar PubMed PubMed Central
41. Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, et al. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS One 2014;9:e95240.10.1371/journal.pone.0095240Search in Google Scholar PubMed PubMed Central
42. McKenzie AJ, Hoshino D, Hong NH, Cha DJ, Franklin JL, Coffey RJ, et al. KRAS-MEK signaling controls Ago2 sorting into exosomes. Cell Rep 2016;15:978–87.10.1016/j.celrep.2016.03.085Search in Google Scholar PubMed PubMed Central
43. Gutkin A, Uziel O, Beery E, Nordenberg J, Pinchasi M, Goldvaser H, et al. Tumor cells derived exosomes contain hTERT mRNA and transform nonmalignant fibroblasts into telomerase positive cells. Oncotarget 2016;7:59173–88.10.18632/oncotarget.10384Search in Google Scholar PubMed PubMed Central
44. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015;527:329–35.10.1038/nature15756Search in Google Scholar PubMed PubMed Central
45. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol 2015;17:816–26.10.1038/ncb3169Search in Google Scholar PubMed PubMed Central
46. Honma K, Iwao-Koizumi K, Takeshita F, Yamamoto Y, Yoshida T, Nishio K, et al. RPN2 gene confers docetaxel resistance in breast cancer. Nat Med 2008;14:939–48.10.1038/nm.1858Search in Google Scholar PubMed
47. Janzen DM, Tiourin E, Salehi JA, Paik DY, Lu J, Pellegrini M, et al. An apoptosis-enhancing drug overcomes platinum resistance in a tumour-initiating subpopulation of ovarian cancer. Nat Commun 2015;6:7956.10.1038/ncomms8956Search in Google Scholar PubMed PubMed Central
48. Hashimoto A, Oikawa T, Hashimoto S, Sugino H, Yoshikawa A, Otsuka Y, et al. P53- and mevalonate pathway-driven malignancies require Arf6 for metastasis and drug resistance. J Cell Biol 2016;213:81–95.10.1083/jcb.201510002Search in Google Scholar PubMed PubMed Central
49. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015;527:472–6.10.1038/nature15748Search in Google Scholar PubMed PubMed Central
50. Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015;527:525–30.10.1038/nature16064Search in Google Scholar PubMed PubMed Central
51. Fujita Y, Yagishita S, Hagiwara K, Yoshioka Y, Kosaka N, Takeshita F, et al. The clinical relevance of the miR-197/CKS1B/STAT3-mediated PD-L1 network in chemoresistant non-small-cell lung cancer. Mol Ther 2015;23:717–27.10.1038/mt.2015.10Search in Google Scholar PubMed PubMed Central
52. Chen X, Wang YW, Xing AY, Xiang S, Shi DB, Liu L, et al. Suppression of SPIN1-mediated PI3K-Akt pathway by miR-489 increases chemosensitivity in breast cancer. J Pathol 2016;239:459–72.10.1002/path.4743Search in Google Scholar PubMed
53. Lv L, An X, Li H, Ma L. Effect of miR-155 knockdown on the reversal of doxorubicin resistance in human lung cancer A549/dox cells. Oncol Lett 2016;11:1161–6.10.3892/ol.2015.3995Search in Google Scholar PubMed PubMed Central
54. Garofalo M, Romano G, Di Leva G, Nuovo G, Jeon YJ, Ngankeu A, et al. EGFR and MET receptor tyrosine kinase-altered microRNA expression induces tumorigenesis and gefitinib resistance in lung cancers. Nat Med 2012;18:74–82.10.1038/nm.2577Search in Google Scholar PubMed PubMed Central
55. Zhao JJ, Chu ZB, Hu Y, Lin J, Wang Z, Jiang M, et al. Targeting the miR-221-222/PUMA/BAK/BAX pathway abrogates dexamethasone resistance in multiple myeloma. Cancer Res 2015;75: 4384–97.10.1158/0008-5472.CAN-15-0457Search in Google Scholar PubMed PubMed Central
56. Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun 2013;4:1393.10.1038/ncomms2393Search in Google Scholar PubMed PubMed Central
57. Gabriely G, Wurdinger T, Kesari S, Esau CC, Burchard J, Linsley PS, et al. MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol 2008;28:5369–80.10.1128/MCB.00479-08Search in Google Scholar PubMed PubMed Central
58. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA 2011;108:5003–8.10.1073/pnas.1019055108Search in Google Scholar PubMed PubMed Central
59. 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;13: 423–33.10.1038/ncb2210Search in Google Scholar PubMed PubMed Central
60. Shimomura A, Shiino S, Kawauchi J, Takizawa S, Sakamoto H, Matsuzaki J, et al. Novel combination of serum microRNA for detecting breast cancer in the early stage. Cancer Sci 2016;107:326–34.10.1111/cas.12880Search in Google Scholar PubMed PubMed Central
61. Yang CC, Hung PS, Wang PW, Liu CJ, Chu TH, Cheng HW, et al. miR-181 as a putative biomarker for lymph-node metastasis of oral squamous cell carcinoma. J Oral Pathol Med 2011;40:397–404.10.1111/j.1600-0714.2010.01003.xSearch in Google Scholar PubMed
62. Ren W, Qiang C, Gao L, Li SM, Zhang LM, Wang XL, et al. Circulating microRNA-21 (MIR-21) and phosphatase and tensin homolog (PTEN) are promising novel biomarkers for detection of oral squamous cell carcinoma. Biomarkers 2014;19:590–6.10.3109/1354750X.2014.955059Search in Google Scholar PubMed
63. Lu YC, Chang JT, Huang YC, Huang CC, Chen WH, Lee LY, et al. Combined determination of circulating miR-196a and miR-196b levels produces high sensitivity and specificity for early detection of oral cancer. Clin Biochem 2015;48:115–21.10.1016/j.clinbiochem.2014.11.020Search in Google Scholar PubMed
64. Aushev VN, Zborovskaya IB, Laktionov KK, Girard N, Cros MP, Herceg Z, et al. Comparisons of microRNA patterns in plasma before and after tumor removal reveal new biomarkers of lung squamous cell carcinoma. PLoS One 2013;8:e78649.10.1371/journal.pone.0078649Search in Google Scholar PubMed PubMed Central
65. Cazzoli R, Buttitta F, Di Nicola M, Malatesta S, Marchetti A, Rom WN, et al. microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. J Thorac Oncol 2013;8:1156–62.10.1097/JTO.0b013e318299ac32Search in Google Scholar PubMed PubMed Central
66. Dinh TK, Fendler W, Chalubinska-Fendler J, Acharya SS, O’Leary C, Deraska PV, et al. Circulating miR-29a and miR-150 correlate with delivered dose during thoracic radiation therapy for non-small cell lung cancer. Radiat Oncol 2016;11:61.10.1186/s13014-016-0636-4Search in Google Scholar PubMed PubMed Central
67. Ogata-Kawata H, Izumiya M, Kurioka D, Honma Y, Yamada Y, Furuta K, et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One 2014;9:e92921.10.1371/journal.pone.0092921Search in Google Scholar PubMed PubMed Central
68. Vychytilova-Faltejskova P, Radova L, Sachlova M, Kosarova Z, Slaba K, Fabian P, et al. Serum-based microRNA signatures in early diagnosis and prognosis prediction of colon cancer. Carcinogenesis 2016;37:941–50.10.1093/carcin/bgw078Search in Google Scholar PubMed
69. Hur K, Toiyama Y, Okugawa Y, Ide S, Imaoka H, Boland CR, et al. Circulating microRNA-203 predicts prognosis and metastasis in human colorectal cancer. Gut 2015;1–11.10.1136/gutjnl-2014-308737Search in Google Scholar PubMed PubMed Central
70. Perez-Carbonell L, Sinicrope FA, Alberts SR, Oberg AL, Balaguer F, Castells A, et al. MiR-320e is a novel prognostic biomarker in colorectal cancer. Br J Cancer 2015;113:83–90.10.1038/bjc.2015.168Search in Google Scholar PubMed PubMed Central
71. Qiu X, Zhang J, Shi W, Liu S, Kang M, Chu H, et al. Circulating microRNA-26a in plasma and its potential diagnostic value in gastric cancer. PLoS One 2016;11:e0151345.10.1371/journal.pone.0151345Search in Google Scholar PubMed PubMed Central
72. Samsonov R, Shtam T, Burdakov V, Glotov A, Tsyrlina E, Berstein L, et al. Lectin-induced agglutination method of urinary exosomes isolation followed by mi-RNA analysis: application for prostate cancer diagnostic. Prostate 2016;76:68–79.10.1002/pros.23101Search in Google Scholar PubMed
73. Shi R, Wang PY, Li XY, Chen JX, Li Y, Zhang XZ, et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget 2015;6:26971–81.10.18632/oncotarget.4699Search in Google Scholar PubMed PubMed Central
74. Im H, Shao H, Park YI, Peterson VM, Castro CM, Weissleder R, et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat Biotechnol 2014;32:490–5.10.1038/nbt.2886Search in Google Scholar PubMed PubMed Central
75. Yoshioka Y, Kosaka N, Konishi Y, Ohta H, Okamoto H, Sonoda H, et al. Ultra-sensitive liquid biopsy of circulating extracellular vesicles using ExoScreen. Nat Commun 2014;5:3591.10.1038/ncomms4591Search in Google Scholar PubMed PubMed Central
76. Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun 2015;6:6999.10.1038/ncomms7999Search in Google Scholar PubMed PubMed Central
77. Bryzgunova OE, Zaripov MM, Skvortsova TE, Lekchnov EA, Grigor’eva AE, Zaporozhchenko IA, et al. Comparative study of extracellular vesicles from the urine of healthy individuals and prostate cancer patients. PLoS One 2016;11:e0157566.10.1371/journal.pone.0157566Search in Google Scholar PubMed PubMed Central
78. Armstrong DA, Green BB, Seigne JD, Schned AR, Marsit CJ. MicroRNA molecular profiling from matched tumor and bio-fluids in bladder cancer. Mol Cancer 2015;14:194.10.1186/s12943-015-0466-2Search in Google Scholar PubMed PubMed Central
79. Berrondo C, Flax J, Kucherov V, Siebert A, Osinski T, Rosenberg A, et al. Expression of the long non-coding RNA HOTAIR correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes. PLoS One 2016;11:e0147236.10.1371/journal.pone.0147236Search in Google Scholar PubMed PubMed Central
80. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010;464:1071–6.10.1038/nature08975Search in Google Scholar PubMed PubMed Central
81. Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, et al. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene 2013;32:1616–25.10.1038/onc.2012.193Search in Google Scholar PubMed PubMed Central
82. Wang H, Li Q, Tang S, Li M, Feng A, Qin L, et al. The role of long noncoding RNA HOTAIR in the acquired multidrug resistance to imatinib in chronic myeloid leukemia cells. Hematology 2016:1–9. Available at: http://dx.doi.org/10.1080/10245332.2016.1258152.10.1080/10245332.2016.1258152Search in Google Scholar PubMed
83. Motamedinia P, Scott AN, Bate KL, Sadeghi N, Salazar G, Shapiro E, et al. Urine exosomes for non-invasive assessment of gene expression and mutations of prostate cancer. PLoS One 2016;11:e0154507.10.1371/journal.pone.0154507Search in Google Scholar PubMed PubMed Central
84. Ciravolo V, Huber V, Ghedini GC, Venturelli E, Bianchi F, Campiglio M, et al. Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy. J Cell Physiol 2012;227:658–67.10.1002/jcp.22773Search in Google Scholar PubMed
85. Aung T, Chapuy B, Vogel D, Wenzel D, Oppermann M, Lahmann M, et al. Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3. Proc Natl Acad Sci USA 2011;108:15336–41.10.1073/pnas.1102855108Search in Google Scholar PubMed PubMed Central
86. de la Fuente A, Alonso-Alconada L, Costa C, Cueva J, Garcia-Caballero T, Lopez-Lopez R, et al. M-Trap: exosome-based capture of tumor cells as a new technology in peritoneal metastasis. J Natl Cancer Inst 2015;107:10.10.1093/jnci/djv184Search in Google Scholar PubMed PubMed Central
©2017 Walter de Gruyter GmbH, Berlin/Boston
Articles in the same Issue
- Frontmatter
- Editorial
- Not all good things come in big packages
- Pre-analytical and Analytical Issues of miRNA Measurement
- Variability in, variability out: best practice recommendations to standardize pre-analytical variables in the detection of circulating and tissue microRNAs
- Pitfalls of analysis of circulating miRNA: role of hematocrit
- 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
- miRNA assays in the clinical laboratory: workflow, detection technologies and automation aspects
- Clinical Applications of the Different Circulating Forms of miRNAs
- The role of extracellular vesicle microRNAs in cancer biology
- The clinical significance of platelet microparticle-associated microRNAs
- microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application
- microRNAs in cardiovascular disease – clinical application
- miRNAs in Cancer
- Non-coding RNAs: the cancer genome dark matter that matters!
- miRNAs as novel biomarkers in the management of prostate cancer
- Upregulated miR-16 expression is an independent indicator of relapse and poor overall survival of colorectal adenocarcinoma patients
- Identification of a novel microRNA, miR-4449, as a potential blood based marker in multiple myeloma
- miRNA analysis in pancreatic cancer: the Dartmouth experience
- miRNAs and Genomic
- miRNAs, single nucleotide polymorphisms (SNPs) and age-related macular degeneration (AMD)
Articles in the same Issue
- Frontmatter
- Editorial
- Not all good things come in big packages
- Pre-analytical and Analytical Issues of miRNA Measurement
- Variability in, variability out: best practice recommendations to standardize pre-analytical variables in the detection of circulating and tissue microRNAs
- Pitfalls of analysis of circulating miRNA: role of hematocrit
- 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
- miRNA assays in the clinical laboratory: workflow, detection technologies and automation aspects
- Clinical Applications of the Different Circulating Forms of miRNAs
- The role of extracellular vesicle microRNAs in cancer biology
- The clinical significance of platelet microparticle-associated microRNAs
- microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application
- microRNAs in cardiovascular disease – clinical application
- miRNAs in Cancer
- Non-coding RNAs: the cancer genome dark matter that matters!
- miRNAs as novel biomarkers in the management of prostate cancer
- Upregulated miR-16 expression is an independent indicator of relapse and poor overall survival of colorectal adenocarcinoma patients
- Identification of a novel microRNA, miR-4449, as a potential blood based marker in multiple myeloma
- miRNA analysis in pancreatic cancer: the Dartmouth experience
- miRNAs and Genomic
- miRNAs, single nucleotide polymorphisms (SNPs) and age-related macular degeneration (AMD)