The fusion of two worlds: Non-coding RNAs and extracellular vesicles - diagnostic and therapeutic implications (Review)

  • Authors:
    • Yukie Sato-Kuwabara
    • Sonia A. Melo
    • Fernando A. Soares
    • George A. Calin
  • View Affiliations

  • Published online on: October 17, 2014     https://doi.org/10.3892/ijo.2014.2712
  • Pages: 17-27
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The role of the extracellular non-coding RNAs, particularly microRNAs present in tumor-derived extravesicles, has been intensively exploited in human cancer as a promising tool for diagnostic and prognostic purposes. Current knowledge on exosomes shows an important role not only as vehicles in the intercellular communication, but the transfer of their content can specifically modulate the surrounding microenvironment, leading to tumor development and progression and affecting therapy response. Based on this, much effort has focused on understanding the mechanisms behind the biology of exosomes and their closely interaction with non-coding RNAs as an efficient tool in tumor diagnostic and therapy. Here we summarize the current knowledge on extracellular and exosomes-enclosed non-coding RNAs, and their importance as potential biomarkers and mediators of intercellular communication in tumor biology.

1. Introduction

Data from genome-wide transcriptional analysis in humans have shown that the amount of protein coding transcripts accounts for approximately 2% of the entire genome, while the non-coding RNAs (ncRNAs) correspond to around 98% of all the genomic output (1,2). Interestingly, it has been reported that the proportion of non-coding regions in the genome increases according to the complexity of the organism, suggesting a important role for these sequences in physiology and development of the organisms (3,4). For this reason, much attention has been given to the studies on these non-protein-coding RNAs in many fields, especially in cancer, leading to new hypothesis about cancer biology (5). Additionally, the identification of the circulating microRNAs (miRNAs) in bodily fluids makes them potential non-invasive biomarkers for cancer diagnosis and prognosis.

The comprehension of the mechanisms involved in the interactions between tumor cells and the surrounding environment is relevant for tumor biology elucidation and for the improvement of innovative and more efficient therapy approaches (6). The role of extracellular vesicles in cell-to-cell communication in cancer has been the focus of several studies. MiRNAs are one of the most studied exosomal cargos due to their potential role in tumor diagnosis, prognosis and therapy.

In this review, we summarize the role of ncRNAs in cancer, focusing on miRNAs. Additionally, we focus on the role of exosomes in intercellular communication and their potential use in providing diagnostic opportunities, unraveling new therapeutic targets and predicting therapeutic responses.

2. World of non-coding RNAs

The ncRNAs can be divided in two main groups, according to their sizes: long non-coding RNAs (lncRNAs), which are greater than 200 nucleotides and small non-coding RNAs with no more than 200 nucleotides (7,8). These two main categories also show some subgroups, based on the structure and biological function of the transcripts, as long intergenic ncRNAs, pseudogenes, enhancer RNAs, transcribed ultra-conserved region, repeated-associated ncRNAs and antisense RNA in the lncRNAs group. In the small ncRNAs, miRNAs, tiny transcription initiation RNAs, small interfering RNAs, promoter-associated short RNAs, antisense termini associates short RNAs and retrotransposon-derived RNAs have been reported in the literature (5,8).

The miRNAs are the most widely described ncRNA in the literature, since the first small ncRNA lin-4 was described in C. elegans more than 20 years ago (9,10). The synthesis of these evolutionarily conserved endogenous short single-stranded RNAs (18–20 nucleotides in length) begins in the nucleus, when the transcription of miRNA-coding genes generate long primary transcript (pri-miRNA) with stem-loop, which will be detached by the RNase III Drosha/Pasha/DGCR8 complex, and then producing a 70-nucleotide precursor (pre-miRNA). After being transported to the cytoplasm by the protein Exportin-5 (XPO5), the pre-miRNA will be converted in mature miRNA by the action of the Dicer and binding to Argonaute 2 (Ago2) to form the RNA-induced silencing complex (RISC) (11,12). Overall, miRNAs regulate gene expression post-transcriptionally, most commonly through the binding to a specific sequence at the 3′-untranslated region (3′-UTR) of a target protein-coding mRNA, causing a translational repression or cleavage of the target transcripts (13). Thus, miRNAs have a relevant role in many pathological and physiological processes, such as cell proliferation, differentiation, development and apoptosis, acting as oncogenes or tumor suppressors, depending on which genes they regulate (14).

The involvement of miRNA genes in cancer was first described in 2002, when the authors reported that two miRNAs (miR-15a and miR-16-1) are mapped at 13q14, a chromosomal region frequently deleted in B-cell chronic lymphocytic leukemia (B-CLL) and that both genes are down-regulated in a high proportion of the cases (15). Since then, the number of studies on miRNAs and cancer has been increasing considerably, adding novel insights into the role of the miRNAs in human tumor such as in hematological malignancies (1619), colorectal (2023), breast (2428), head and neck (2932) and gastric cancer (3336).

The lncRNAs are transcripts longer than 200 nucleotides, a cutoff based on RNA purification protocols that exclude small RNAs rather than for its functional role (37). The lncRNAs were first described in a study involving large-scale sequencing and annotation of full-length cDNA libraries in mouse (38), and the number of reports about characterization and functions of the lncRNAs has been constantly increasing in the literature (39). The lncRNAs have many features in common with mRNAs, as transcription by RNA polymerase II, polyadenilation and splicing mechanism. This category of ncRNAs composes a heterogeneous group, which makes the lncRNAs classification difficult (40). Most commonly, the lncRNAs can be classified as sense or antisense, divergent or convergent and intronic or intergenic, depending on their position relative to the neighboring protein-coding genes (7,41). Due to lncRNA structure heterogeneity, it is also difficult to assign a specific function to this group and still requires further studies. Evidences suggest that lncRNAs act mainly in regulation of protein-coding genes transcription, but in more complex ways than the miRNAs (42). lncRNAs can repress the transcription of target genes involving epigenetic modifications like chromatin remodeling, since some lncRNAs have been reported to interact with many chromatin modifiers (43). Additionally, lncRNAs can either play a role as putative gene enhancers or decoy RNAs in transcriptional control (41). Some lncRNAs (antisense ncRNA) also play a role in post-transcriptional regulation by interfering with the RNA splicing mechanism (44).

Due to their roles in the functions mentioned above, the lncRNAs have been related to many human cancers, contributing to tumor development and progress (40). Many lncRNAs have been mapped at cancer risk loci in the human genome, such as PTCSC3 (14q13.3) in thyroid cancer (45,46), PCA3 (9q21–22) in prostate cancer (47,48), ANRIL (9p21) in prostate and breast cancers, leukemia and melanoma (4952), MALAT1 (11q13) in liver, colorectal, prostate, bladder and lung cancers (5356).

The role of ncRNAs in many human tumor types has been exhaustingly studied in the past few years and its relevance in mechanisms involved in cancer development and progress is unquestionable. Additionally, the discovery of stable miRNAs in bodily fluids introduced new insights in the ncRNAs comprehension and can represent a new diagnostic approach using less invasive methods (57). The use of circulating miRNAs as tumor biomarkers has many advantages since these transcripts are conserved across species, shows tissue or disease-specific expression and their levels can be quantified by various methods, as microarray profiling, northern blot analysis, in situ hybridization, high-throughput sequencing and qRT-PCR, which is the most used method due to its sensitivity, specificity and reliability (5861). The first evidence of the presence of miRNAs in serum was reported by Lawrie et al (2008), who showed the higher serum levels of miR-21 in large B-cell lymphoma (62). Since then, many studies have reported the presence of different circulating miRNAs in various tumor types, such as in colorectum (63,64), esophagus (65,66), breast (67,68), stomach (69,70) and ovary (71,72). Also, the circulating miRNAs have been reported in many other fluids such as plasma, urine, saliva and cerebrospinal fluid (58,73). Considering this discovery, it is possible that other types of ncRNAs similar to lncRNAs can also be identified in bodily fluids (40).

3. World of extracellular vesicles

The intercellular communication can occur by a direct cell-to-cell contact including the adhesion junctions, or by the releasing of soluble signaling molecules or by the exchange of cellular fragments as extracellular vesicle (EV) (74,75). The EVs are bilayered membrane vesicles secreted by all cell types, and released in the interstitial space or into circulating bodily fluids, where they can travel long distances until they are up taken by receptor cells (76). Different terminology is used to describe EVs based on their morphology and diameter. Exosomes, microvesicles, ectosomes, microparticles and others, are classified based on their size, shape and membrane surface composition (77). The most accepted classification in the literature shows two major groups of EVs, based on their mechanism of biogenesis and sizes: exosomes and microvesicles (or ectosomes). Additionally, apoptotic bodies have been considered by some as a third category of EVs (7880). In this review, we will focus on the exosomes and microvesicles.

Exosomes are 40–140 nm diameter bilayered-membrane vesicles of endocytic origin, with a cup-shaped morphology, showing densities ranging between 1.13–1.19 g/ml (81). The exosomes are originated by the inward budding of clathrin-coated domains in the plasma membrane, generating the multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs) in the late endosome. The formation of ILVs occurs during the endosome maturation, when specific cytosolic proteins are incorporated into these vesicles inside de MVBs. These initial steps occur under control of the ESCRT (endosomal sorting complex required for transport) machinery. Later, the MVBs fuse with lysosomes for degradation or with the cell membrane releasing the exosomes to the extracellular space, process regulated by the RAB family (76,8284). Microvesicles (or ectosomes) are larger than exosomes, with size ranging between 100 and 1,000 nm in diameter and heterogeneous in morphology. Differently from the exosomes, microvesicles (MVs) are originated from the plasma membrane through direct outward budding into the extracellular space. During this process, the newly originated vesicle captures the donor cellular cytosolic content and the receptors on the plasma membrane (Fig. 1). The regulation of MVs biogenesis is intracellular calcium-dependent and it is the result of the activation of cell surface receptors, phospholipid redistribution and cytoskeletal protein contraction (84,85). The apoptotic bodies (ABs) are membrane vesicles, heterogeneous in shape, showing sizes ranging between 50–500 nm in diameter. The ABs are released from the outward protrusion of the plasma membrane during the late phase of cell death by apoptosis and are featured by the presence of organelles inside the vesicles (85,86).

The EV cargo specificity

The interaction between the EVs and the target cells can occur by different mechanisms, as direct interaction of the surface proteins of the EVs with the receptors on the target cells, triggering the activation of the intracellular pathways. EVs can also be engulfed by the target cells through membrane fusion or by endocytosis/phagocytosis, with transfer and release of their cargo. Transcripts as mRNAs and miRNAs contained inside de EVs can be transferred to the target cells and be functional (6).

The EVs carry specific contents (cargo) as membrane receptors, ligands, proteins, nucleic acids and infectious agents, depending on the cell of origin and how they were originated from the donor cell (75). There is no consensus regarding the specific content of different EVs, but it seems that MVs are characterized by the presence of cell-surface proteins from the donor cells such as receptors and adhesion proteins. In turn, exosomes have been found to be characterized by proteins associated to their endosomal origin and MVBs formation (84). Some specific markers have been associated to exosomes as tetraspanins (CD9, CD63, CD81 and CD82), major histocompatibility complex class I and II, LAMP1 and LAMP2, flotilins, annexins, TSG101 and heat shock proteins (83,87,88). The protein content of MVs seems to be more heterogeneous, containing cell membrane markers, phosphatidylserine (PS) residues, integrins, selectins and CD40 ligands (84), and high levels of cholesterol and signaling complexes known as lipid rafts (76). Most importantly, it has been shown that the population of exosomes secreted by cancer cells contains a representation of the entire genome of the cell of origin, providing exciting opportunities of using exosomes as a liquid biopsy (89).

Tumor-derived exosomes

The exosomes are by far the most extensively studied due to their characteristics (as presence in bodily fluids and expression of specific markers) that can contribute not only to intercellular communication but also to their potential role in diagnosis (82). The release of exosomes can be a response to different cellular stress conditions common in cancer, such as hypoxia, acidic pH, heat shock and oxidative stress, resulting in alterations of the tumor microenvironment and distal organs activating angiogenesis and promoting migration and leading to metastasis (9092).

An important step before considering using the exosomal content for study or diagnosis purposes is a reliable exosome isolation method, to insure the quality of the results. Exosomes can be collected from fluids or cell supernatant by a series of sequential centrifugations to remove larger cellular debris and filtration through 100–220 nm filters to exclude larger EVs, including MVs and apoptotic bodies. Then, the exosomes are pelleted by ultracentrifugation and/or suspension in a sucrose gradient for the completely remove of protein contamination (77,93,94). The exosomes isolation can also be performed using specific filters, immune isolation by magnetic beads or microfluidic separation (95). Recently, some commercial isolation kits are available based on polymer-based precipitation and on the magnetic bead isolation. Then, some additional procedures are necessary to confirm the purity of the isolated exosomes. One of them is to verify the size and shape of the exosomes by electron microscopy analysis. Vesicles diameter and morphology can also be assessed by specific instruments that can visualize, characterize and measure small vesicles. Another important factor that must be evaluated is the protein content, that can be assessed by flow cytometry and western blot analysis for markers as CD63, CD81, Tsg101 and flotilin (77,96).

4. The fusion of the two worlds

Exosomal miRNAs

In the bodily fluids, the miRNAs have been reported to play a role at intercellular communication, and can act at short and long distant sites in a hormone-like behavior (14,87). The transport of circulating miRNAs can be carried by protein transporters or by exosomes. It is known that serum contains ribonucleases, suggesting that the circulating miRNAs are protected from the RNase action within extravesicles. The miRNA recruitment to the exosomes depends on the attachment of RNA-induced silencing complexes (RISCs) to the ESCRT components. However, the release of exosomal miRNAs is under control of a ceramide-dependent machinery, as reported by Kosaka et al (73). These authors showed that the inhibition of neutral sphingomyelinase 2 (a regulator of the ceramide biosynthesis) resulted in lower levels of miRNA secretion (73).

The first evidence of the existence of miRNAs in exosomes was reported by Valadi et al, showing that these vesicles contain both mRNA and miRNAs, which can be transferable to another cell, where the transcripts can be functional (97). Since then, the number of studies regarding the identification of exosomal miRNAs in cancer has been increasing in the literature. A summary of some studies in the literature in this field is in Table I. Most of these reports are based on in vitro studies involving a variety of cancer cell lines, identifying the exosomal miRNA content as in breast cancer (98,99), leukemia (100), melanoma (101,102), prostate cancer (103), ovarian (104) and gastric cancer (105). The transcripts content of the exosomes usually can differ from that in the donor cells, and the exosomal miRNA profile of tumor cells can also differ from that released by normal controls (106).

Table I

Summary of the studies reporting the identification of exosomal miRNAs in cancer.

Table I

Summary of the studies reporting the identification of exosomal miRNAs in cancer.

TumorSampleExosome extractionmiRNARefs.
Breast cancerCell lineSC, 0.22 μm filtering and UCmiR-233(108)
Breast cancerCell lineSC and UC or ExoQuick (System Biosciences)miR-210(116)
Breast cancerCell lineSC, 0.22 μm filtering and UCmiR-100, miR-17, miR-222, miR-342-3p, miR-451, miR-30a(114)
Breast cancerCell lineSC, 0.22 μm filtering, UC and sucrose gradientmiR-198, miR-26a, miR-34a, miR-49a, let-7a, miR-328, miR-130a, miR-149, miR-602 and miR-92b(99)
Breast cancerSerum samples, tumor samples, cell line, animal modelsSC, UCmiR-105(113)
Cervical cancerCervicovaginal lavage fluidSC and UCmiR-21 and miR-146a(126)
Cholangiocarcinoma (biliary tree)Bile sampleSC and 0.22 μm filteringmiR-222, miR-126, miR-486-3p, miR-484, miR-19a, miR-19b, miR-16, miR-191, miR-31, miR-1274b, miR-618, miR-486-3p, miR-16, miR-1274b, miR-484, miR-191(125)
Colorectal cancerCell lineSC and 0.22 μm filteringmiR-21, miR-192, miR-221(107)
Colorectal cancerSerum samplesSC, 0.22 μm filtering and UClet-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223 and miR-23(121)
Esophageal cancerSerum samplesSC, 0.45 μm filtering and ExoQuick (System Biosciences)miR-21(120)
Gastric cancerCell lineSC, 0.1 μm filtering and UClet-7 family (a, b, c, d, e, f, g, i)(105)
GlioblastomaTumor samples, serum samplesSC, 0.22 μm filtering and UClet-7a, miR-15b, mR-16, miR-19b, miR-21, miR-26a, miR-27a, miR-92, miR-93, miR-320, miR-20(118)
GlioblastomaTumor samples, cell lines, animal modelsSC, 0.22 μm filtering, and UCmiR-1(140)
LeukemiaCell lineExoQuick (System Biosciences)miR-19a, miR-146-5p, miR-454, miR-18b, miR-574-3p, miR-21, miR-431, miR-345, miR-210, miR-197, miR-20a, miR-24, miR-19b, miR-130b, miR-106b, miR-224, miR-210, miR-652, miR-379, miR-185(117)
LeukemiaCell lineSC, 0.22 μm filtering and ExoQuick (System Biosciences)miR-17–92 cluster, miR-24, miR-222(109)
LeukemiaCell lineSC, 0.22 μm filtering and UCmiR-1908 and miR-298(100)
Lung adenocarcinomaPlasma samplesSize exclusion by chromatography, magnetic beads (EpCAM)miR-17-3p, miR-21, miR-106a, miR-146, miR-155, miR-191, miR-192, miR-203, miR-205, miR-210, miR-212, miR-214(122)
Lung adenocarcinomaPlasma samplesExoQuick (System Biosciences)miR-378a, miR-379, miR-139-5p, miR-200-5p, miR-151-5p, miR-30a-3p, miR-200b-5p, miR-629, miR-100 and miR-154-3p(123)
Lung adenocarcinomaCell lineSC, 0.22 μm filtering, and UCmiR-192(112)
Lung cancerCell lineUC and ExoQuick (System Biosciences)miR-21, miR-98, miR-133b, miR-138, miR-181a, miR-200c(115)
Lung cancerPlasma sample, bronchoalveolar lavage fluidSC, 0.22 μm filtering, and UCmiR-222, miR-126, miR-144, miR-302a, miR-302c(124)
MelanomaCell lineSC, 0.22 μm filtering and UC; SC and ExoQuick (System Biosciences)miR-181b, miR-181a, miR-4802-3p, miR-23b, miR22, miR-107, miR-103a, miR-9, miR-338-3p(101)
Melanona and colon carcinomaCell lineSC, 0.22 μm filtering, prominin-1 based immuno-magnetic selectionmiR-216b, miR-889, miR-4307, miR-4272, miR-203, miR-4289, miR-3149, miR-203, miR-3145, miR-1911, miR-513a-3p, miR-3916, miR-886-3p, miR-1182, miR-3613-5p, let-7i, miR-3132, miR-3914, miR-3618, miR-1307, miR-3614-3p, miR-519c-3p, miR-3160, miR-3153, miR-4278, miR-3646, miR-3926, miR-515-5p, miR-3169, miR-590-3p, miR-525-5p, miR-548g, miR-365, miR-525-3p, miR-320d(102)
Multiple myelomaCell line0.22 μm filtering, UC and ExoQuick (System Biosciences)miR-125q-3p, miR-128, miR-15a, miR-185, miR-192, miR-212, miR-324-3p, miR-331-5p, miR-345, miR-422a, miR-429, miR-511, miR-576-3p, miR-618, miR-9, miR-1271, miR-139, miR-148, miR-151-3p, miR-15b, miR-19b-1, miR-21, miR-34b, miR-378, miR-589, miR-592, miR-625, miR-93(111)
Ovarian cancerSerum samples, cell lineMagnetic activated cell sorting (MACS) - EpCAMmiR-21, miR-141, miR-200a, miR-200c, miR-200b, miR-203, miR-205, miR-214(119)
Ovarian cancerCell lineSC, UC and sucrose gradientlet-7(104)
Prostate cancerCell lineSC, 0.22 μm filtering, and UCmiR-143(110)
Protaste cancerCell lineSC and UCmiR-1280, miR-720 and miR-1260b(103)

[i] SC, sequential centrifugation; UC, ultracentrifugation; MV, microvesicle.

Studies have reported evidence of the intercellular transfer of the exosomal content between different cells. Chiba et al showed that exosomes derived from colorectal cancer cells can be transferred to hepatoma and lung cancer cells (107). In addition, some of these reports have demonstrated that the transferred exosomal content can be functional in the receptor cells. Yang et al reported the presence of a specific miRNA for IL-4-activated macrophage - miR-233, in the co-cultured breast cancer cells and it can enhance the invasiveness potential of the receptor cells (108). The transfer of the leukemia cell line-derived exosomal miR-92a to the endothelial cells affected the endothelial cell migration and tube formation in the receptor cells (109). Kosaka et al showed that exosomal miR-143 derived from a normal prostate cell line act as a tumor suppressor by inhibiting the growth in the target prostate cancer cells (110). Similar results were reported by Roccaro et al in a study demonstrating that the exosomal miR-15 from the normal bone marrow mesenchymal stromal cells causes a tumor suppressor effect when transferred to tumor cells, where this miRNA is downregulated (111). In a recent study, Valencia et al demonstrated that the exosomal miR-192 derived from lung adenocarcinoma cell lines repressed the angiogenic activity in the co-cultured endothelial cells by the inhibition of the proangiogenic factors (112). More recently, Zhou et al showed that the transfer of exosomal miR-105 to non-metastatic breast cancer cells induces metastasis and vascular permeability by targeting the cellular tight junction protein ZO-1 (113).

The intercellular transfer of the exosome cargo can also affect the resistance or sensitivity of cancer cells to therapy. The transfer of the exosomes derived from chemoresistant breast cancer cell lines can also spread the resistance potential to receptor chemosensitive cell lines, possibly due to the action of the exosomal content as miR-100, miR-222 and miR-30a (114). Similarly, in lung cancer, Xiao et al reported that after miR-21- and miR-133-enriched exosome transfer from the chemoresistant tumor cells, the chemosensitive target cells acquire resistance to the drug exposure (115).

It is well known that hypoxia is an important factor that triggers angiogenesis and metastasis formation and evidence has been presented for the involvement of the exosome in this mechanism. King et al found an increased concentration of released exosomes and higher expression of exosomal miR-210 secreted by breast cancer cells under hypoxic conditions when compared to normoxic cells (116). In leukemia, the miR-210 can also be found in a subset of miRNAs upregulated in exosomes released by tumor cells under hypoxic conditions (117).

The exosomal miRNA expression profiling from serum and plasma samples has also been assessed in glioblastoma, where the expression of 11 miRNAs known to be upregulated was slightly lower in exosomes than in the donor cells but still reflecting the tumor profile (118). When the serum samples from ovarian cancer patients were evaluated, a distinct exosomal miRNA profile was identified from that of benign disease (119). Another study reporting a potential use of exosomal miRNAs as diagnostic markers showed a higher expression of miR-21 released from esophageal cancer serum samples when compared to non-tumoral samples, and it correlated with advanced tumor stages, lymph node involvement and metastasis (120). Exosomal let-7a, miR-1229, miR-1246, miR-150, miR-21, miR-223 and miR-23a from colorectal tumor samples and cancer cell lines are more highly expressed than those from healthy controls samples and normal colon cell lines, and the expression levels of these miRNAs are significantly decreased in exosomes samples collected after tumor resection, indicating the cancer status (121).

The exosomal miRNA profiling from plasma samples was assessed to develop a diagnostic screening method for lung adenocarcinoma. The expression pattern of 12 specific upregulated miRNAs (miR-17-3p, miR-21, miR-106a, miR-146, miR-155, miR-191, miR-192, miR-203, miR-205, miR-210, miR-212, miR-214) in tumor samples was similar in the tumor plasma-derived exosomes and distinct from the control samples, indicating exosomal miRNAs could be relevant as a screening method for this tumor (122). In another report, the exosomal miRNAs miR-378a, miR-379, miR-139-5p and miR-200-5p were identified as possible markers to distinguish tumor from normal samples in lung adenocarcinoma (123). In addition, the miRNAs miR-151-5p, miR-30a-3p, miR-200b-5p, miR-629, miR-100 and miR-154-3p are possible markers to discriminate lung adenocarcinoma from granulomas (123). More recently, Rodríguez et al evaluated the exosomes derived from bronchoalveolar lavage (BAL) and plasma samples from lung cancer, in which the exosomal miRNA content derived from tumor plasma samples is more elevated than in the BAL, suggesting that the a higher concentration of exosomal miRNAs are released in the plasma than in the bronchoalveolar fluid (124).

The use of exosome as a diagnostic tool has also been evaluated in other fluids than plasma and serum samples. Recently, a report identified the exosomal miRNAs in bile from cholangiocarcinoma patients and a potential diagnostic panel that includes miR-486-3p, miR-16, miR-1274b, miR-484 and miR-191 as predictive markers (125). In a study involving cervicovaginal lavage fluids, the miR-21 and miR-146a were highly expressed in fluids from cervical cancer samples when compared to those from HPV(+) and HPV(−) normal samples (126).

Exosomal lncRNAs

The study of lncRNAs is a relatively new field on cancer research, and many questions about their expression and functions remain unclear, like the presence of these ncRNAs in the bodily fluids. Since the use of circulating miRNAs in diagnostic screening methods and therapeutics have been intensively evaluated in many tumor types, the presence of released lncRNAs in bodily fluids, specially within extravesicles as exosomes, could be a source of novel potential biomarkers for diagnosis, prognosis and therapeutics purposes. Of our knowledge, there are only few data about this particular aspect of the lncRNAs (Table II). The ucRNA (ultranconserved lncRNA) TUC399 was identified in exosomes derived from hepatocellular cancer cell lines, and the intercellular transfer of exosomal TUC399 can contribute to tumor growth and progression (127). More recently, the same group demonstrated that the expression of lncRNAs linc-RoR (long intergenic non-coding RNA, regulator of reprogramming) in hepatocellular cancer is responsive to hypoxic conditions and the transfer of exosomal linc-RoR can modulate the intercellular response to hypoxia (128).

Table II

Summary of the reports of the circulating lncRNAs in cancer.

Table II

Summary of the reports of the circulating lncRNAs in cancer.

TumorSampleExtravesicles isolationLong ncRNARefs.
Gastric cancerPlasma samples, cell linesNAH19, HOTAIR, MALAT1(129)
Hepatocellular cancerCell lineUC and density gradient separationTUC399(127)
Hepatocellular cancerTissue samples, plasma samplesNAHULC(132)
Hepatocellular cancerCell line, animal modelSC and UClinc-RoR(128)
Leukemia and multiple myelomaPlasma samplesNATUG1, MALAT1, HOTAIR, lincRNA-p21, GAS5(133)
Prostate cancerTissue samples, plasma samplesNAMALAT-1 and PCA3(130)

[i] SC, sequential centrifugation; UC, ultracentrifugation; NA, not applied.

At present, reports regarding the circulating lncRNAs in bodily fluids are also scant in the literature. In a study evaluating the expression of the lncRNAs H19, HOTAIR and MALAT1 in gastric cancer plasma samples, Arita et al (2013) showed that only H19 is higher expressed in tumor samples than in the controls, and reported significanly decreased expression levels in post-operative tumor samples, indicating that the release of lncRNAs into the plasma can reflect the disease status (129). In another report, Ren et al identified the MALAT1-derived mini-RNA (MD-miniRNA) as potential novel plasma biomarker in prostate cancer (130). Some reports have demonstrated the use of lncRNA PCA3 as a specific and reliable marker detectable in urine samples from patients of prostate cancer, instead of the standardized use of the prostate-specific antigen (PSA). The evidence that highly upregulated in liver cancer (HULC) lncRNA expression is significantly higher in plasma tumor samples than in the healthy controls indicates the use of this lncRNA as potential circulating biomarker for diagnosis in hepatocellular cancer (131,132). The evaluation of lncRNAs expression in plasma samples from leukemia and multiple myeloma showed that TUG1, MALAT1, HOTAIR and GAS5 are more highly expressed in leukemia than in the control samples, and only lincRNA-p21 is upregulated in multiple myeloma (133).

However, some limitations in using exosomal ncRNAs in diagnostics have been pointed out by many authors. The specificity and sensitivity of exosomal tumor marker detection in bodily fluids is still challenging. For example, serum and plasma-derived extravesicles as exosomes can be released by other than tumor cells, such as different blood cell types, affecting the purity of the tumor-derived exosome samples. In addition, the release of these exosomes depends on the age of the patient, infection or inflammation status of the disease, possibly introducing a bias in comparison analysis if not appropriately normalized to these conditions. Another issue that must be considered is the need of a standardized protocol for collecting and handling of the samples, as well as the exosomes isolation method (95,106).

Relevance in therapy

Since the miRNA is able to target multiple genes and signaling networks simultaneously, acting like oncogenes or tumor suppressor factors, it makes them a suitable tool for therapeutics interventions. A well and highly specific design is necessary for a successful result and to prevent undesirable targets. However, one of the principal limitations of this approach is the nuclease activity, causing the degradation before the miRNAs can achieve the targets. The use of vesicles for the delivery of exogenous therapeutic molecules to the targets has been intensively considered as a new promising therapeutic intervention. As mentioned before, the exosomes have the ability to transfer functional proteins and transcripts as perfect non-immunogenic carriers of therapeutic agents to target cells, making them suitable as therapeutic tool (134).

Considering that the exosome content can act as modulator of the microenvironment, facilitating tumor growth and metastasis, the blockade of the production, release and uptake by receptor cells could reverse the influence of the increased levels of exosomes in tumor progression (95). Based on this, focusing on the inhibition of key components of the extravesicle production and release, such as the members of the ESCRT machinery, could be a useful strategy for therapy (6).

A third possible direction is represented by the drug or gene delivery by extravesicles to the target sites. Considering the elucidation of the intercellular transfer by exosomes, many reports have demonstrated the use of the extravesicles as small RNA carriers (135,136). Intercellular transfer by exosomes can be used as miRNA carriers to restore miRNA expression in the target cells, where they play a therapeutic role as tumor suppressor factors. The targeted delivery of miRNAs by exosomes was demonstrated in a study in breast cancer cells expressing high levels of EGFR. This was achieved by the engineering of the donor cells to modify the surface of the exosomes to express the transmembrane domain of the PDGFR fused to the GE11 peptide. Then, the modified exosomes can deliver the let-7a miRNA after intravenous injection to EGFR-expressing xenograft breast cancer tissue in immunodeficient mice (137). The ability of the miRNAs to target multiple genes can be a limitation for the specificity of this method as a selective approach for targeted therapy. The use of synthetic siRNA has been exploited as a more selective therapeutic tool. In an interesting study reported by Alvarez-Erviti et al, dendritic cells expressing a specific protein of the exosomal membrane Lamp2b fused to a neuron-penetrating RVG peptide were isolated from mice, and the exosomes derived from these cells were loaded with exogenous siRNA to GAPDH by electroporation. Subsequently, these RVG-targeted exosomes were intravenously injected, which delivered GAPDH siRNA to specific cells in the brain, leading to a selective gene knockdown (138). Similarly, another report showed the delivery of siRNA into monocytes and lymphocytes by exosomes as gene delivery vector, reflecting a selective gene silencing of MAPK1 (139).

5. Concluding remarks

The discovery of the intercellular communication by the extravesicles has opened a new field for tumor biology. Exosomes can be found in the bodily fluids in a variety of tumor types and many reports have proved that the exosomal content as proteins, mRNA, miRNA and DNA can reflect the disease status, making them suitable for biomarkers for non-invasive diagnostic and prognosis purposes. With the advance of the engineering that permits the manipulation of the exosomal content and surface markers, many studies have been focusing on the development of therapeutic approaches in various tumor types, involving more specific delivery to the target tumor cells with more selective and efficient results. However, despite the efforts focusing on the study of the extracellular vesicles, specially exosomes, there are many aspects of the their biology that still need to be elucidated so that it would improve the advantages of the use of this promising approach in tumors.

Acknowledgements

G.A.C. is The Alan M. Gewirtz Leukemia & Lymphoma Society Scholar. Study in G.A.C.’s laboratory is supported in part by the NIH/NCI grants 1UH2TR00943-01 and 1 R01 CA182905-01, Developmental Research Awards in Prostate Cancer, Multiple Myeloma, Leukemia (P50 CA100632) and Head and Neck (P50 CA097007) SPOREs, a SINF MDACC_ DKFZ grant in CLL, a SINF grant in colon cancer, a Kidney Cancer Pilot Project, the Duncan Family Institutional Seed Funds, The Blanton-Davis Ovarian Cancer - 2013 Sprint for Life Research Award, the Laura and John Arnold Foundation, the RGK Foundation and the Estate of C. G. Johnson, Jr and by the CLL Global Research Foundation.

References

1 

Mattick JS: Non-coding RNAs: the architects of eukaryotic complexity. EMBO Rep. 2:986–991. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Morceau F, Chateauvieux S, Gaigneaux A, Dicato M and Diederich M: Long and short non-coding RNAs as regulators of hematopoietic differentiation. Int J Mol Sci. 14:14744–14770. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Mattick JS and Makunin IV: Non-coding RNA. Hum Mol Genet. 15(Spec 1): R17–R29. 2006. View Article : Google Scholar

4 

Amaral PP and Mattick JS: Noncoding RNA in development. Mamm Genome. 19:454–492. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Setoyama T, Ling H, Natsugoe S and Calin GA: Non-coding RNAs for medical practice in oncology. Keio J Med. 60:106–113. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Vader P, Breakefield XO and Wood MJA: Extracellular vesicles: emerging targets for cancer therapy. Trends Mol Med. 20:385–393. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: insights into functions. Nat Rev Genet. 10:155–159. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Prensner JR and Chinnaiyan AM: The emergence of lncRNAs in cancer biology. Cancer Discov. 1:391–407. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Lee RC, Feinbaum RL and Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 75:843–854. 1993. View Article : Google Scholar

10 

Wightman B, Ha I and Ruvkun G: Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 75:855–862. 1993. View Article : Google Scholar : PubMed/NCBI

11 

Zhang W, Dahlberg JE and Tam W: MicroRNAs in tumorigenesis: a primer. Am J Pathol. 171:728–738. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Siomi H and Siomi MC: Posttranscriptional regulation of microRNA biogenesis in animals. Mol Cell. 38:323–332. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Raisch J, Darfeuille-Michaud A and Nguyen HTT: Role of microRNAs in the immune system, inflammation and cancer. World J Gastroenterol. 19:2985–2996. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Ling H, Fabbri M and Calin GA: MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 12:847–865. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Calin GA, Dumitru CD, Shimizu M, et al: Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 99:15524–15529. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Yendamuri S and Calin GA: The role of microRNA in human leukemia: a review. Leukemia. 23:1257–1263. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Balatti V, Pekarky Y, Rizzotto L and Croce CM: miR deregulation in CLL. Adv Exp Med Biol. 792:309–325. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Gordon JEA, Wong JJ-L and Rasko JEJ: MicroRNAs in myeloid malignancies. Br J Haematol. 162:162–176. 2013. View Article : Google Scholar

19 

Srivastava S, Tsongalis GJ and Kaur P: Recent advances in microRNA-mediated gene regulation in chronic lymphocytic leukemia. Clin Biochem. 46:901–908. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Rossi S, Kopetz S, Davuluri R, Hamilton SR and Calin GA: MicroRNAs, ultraconserved genes and colorectal cancers. Int J Biochem Cell Biol. 42:1291–1297. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Hong L, Han Y, Zhou Y and Nita A: Angiogenesis-related microRNAs in colon cancer. Expert Opin Biol Ther. 13:77–84. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Hutchison J, Cohen Z, Onyeagucha BC, Funk J and Nelson MA: How microRNAs influence both hereditary and inflammatory-mediated colon cancers. Cancer Genet. 206:309–316. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Menéndez P, Villarejo P, Padilla D, Menéndez JM and Rodríguez-Montes JA: Implications of the histological determination of microRNAs in the screening, diagnosis and prognosis of colorectal cancer. J Surg Oncol. 108:70–73. 2013.PubMed/NCBI

24 

Ferracin M, Querzoli P, Calin GA and Negrini M: MicroRNAs: toward the clinic for breast cancer patients. Semin Oncol. 38:764–775. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Harquail J, Benzina S and Robichaud GA: MicroRNAs and breast cancer malignancy: an overview of miRNA-regulated cancer processes leading to metastasis. Cancer Biomark. 11:269–280. 2012.PubMed/NCBI

26 

Zhang ZJ and Ma SL: miRNAs in breast cancer tumorigenesis (Review). Oncol Rep. 27:903–910. 2012.PubMed/NCBI

27 

Mulrane L, McGee SF, Gallagher WM and O’Connor DP: miRNA dysregulation in breast cancer. Cancer Res. 73:6554–6562. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Singh R and Mo Y-Y: Role of microRNAs in breast cancer. Cancer Biol Ther. 14:201–212. 2013. View Article : Google Scholar : PubMed/NCBI

29 

John K, Wu J, Lee B-W and Farah CS: MicroRNAs in head and neck cancer. Int J Dent. 2013:6502182013. View Article : Google Scholar : PubMed/NCBI

30 

Nagadia R, Pandit P, Coman WB, Cooper-White J and Punyadeera C: miRNAs in head and neck cancer revisited. Cell Oncol (Dordr). 36:1–7. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Nohata N, Hanazawa T, Kinoshita T, Okamoto Y and Seki N: MicroRNAs function as tumor suppressors or oncogenes: aberrant expression of microRNAs in head and neck squamous cell carcinoma. Auris Nasus Larynx. 40:143–149. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Tu HF, Lin SC and Chang KW: MicroRNA aberrances in head and neck cancer: pathogenetic and clinical significance. Curr Opin Otolaryngol Head Neck Surg. 21:104–111. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Song JH and Meltzer SJ: MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 143:35–47.e2. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Wang F, Sun GP, Zou YF, Hao JQ, Zhong F and Ren WJ: MicroRNAs as promising biomarkers for gastric cancer. Cancer Biomark. 11:259–267. 2012.PubMed/NCBI

35 

Gao M, Yin H and Fei ZW: Clinical application of microRNA in gastric cancer in Eastern Asian area. World J Gastroenterol. 19:2019–2027. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Song S and Ajani JA: The role of microRNAs in cancers of the upper gastrointestinal tract. Nat Rev Gastroenterol Hepatol. 10:109–118. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Kapranov P, Cheng J, Dike S, et al: RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science. 316:1484–1488. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Okazaki Y, Furuno M, Kasukawa T, et al: Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 420:563–573. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011. View Article : Google Scholar

40 

Spizzo R, Almeida MI, Colombatti A and Calin GA: Long non-coding RNAs and cancer: a new frontier of translational research? Oncogene. 31:4577–4587. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Tang JY, Lee JC, Chang YT, Hou MF, Huang HW, Liaw CC and Chang HW: Long noncoding RNAs-related diseases, cancers, and drugs. Sci World J. 2013:9435392013.PubMed/NCBI

42 

Van Roosbroeck K, Pollet J and Calin GA: miRNAs and long noncoding RNAs as biomarkers in human diseases. Expert Rev Mol Diagn. 13:183–204. 2013.PubMed/NCBI

43 

Shi X, Sun M, Liu H, Yao Y and Song Y: Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett. 339:159–166. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Fitzgerald KA and Caffrey DR: Long noncoding RNAs in innate and adaptive immunity. Curr Opin Immunol. 26:140–146. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Jendrzejewski J, He H, Radomska HS, et al: The polymorphism rs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type. Proc Natl Acad Sci USA. 109:8646–8651. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Fan M, Li X, Jiang W, Huang Y, Li J and Wang Z: A long non-coding RNA, PTCSC3, as a tumor suppressor and a target of miRNAs in thyroid cancer cells. Exp Ther Med. 5:1143–1146. 2013.PubMed/NCBI

47 

Hessels D and Schalken JA: The use of PCA3 in the diagnosis of prostate cancer. Nat Rev Urol. 6:255–261. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Day JR, Jost M, Reynolds MA, Groskopf J and Rittenhouse H: PCA3: from basic molecular science to the clinical lab. Cancer Lett. 301:1–6. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Pasmant E, Laurendeau I, Héron D, Vidaud M, Vidaud D and Bièche I: Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 67:3963–3969. 2007. View Article : Google Scholar

50 

Iacobucci I, Sazzini M, Garagnani P, et al: A polymorphism in the chromosome 9p21 ANRIL locus is associated to Philadelphia positive acute lymphoblastic leukemia. Leuk Res. 35:1052–1059. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Cheetham SW, Gruhl F, Mattick JS and Dinger ME: Long noncoding RNAs and the genetics of cancer. Br J Cancer. 108:2419–2425. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Cheng W, Zhang Z and Wang J: Long noncoding RNAs: new players in prostate cancer. Cancer Lett. 339:8–14. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Ji P, Diederichs S, Wang W, et al: MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 22:8031–8041. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Gutschner T and Diederichs S: The hallmarks of cancer: a long non-coding RNA point of view. RNA Biol. 9:703–719. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Li CH and Chen Y: Targeting long non-coding RNAs in cancers: progress and prospects. Int J Biochem Cell Biol. 45:1895–1910. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Martens-Uzunova ES, Olvedy M and Jenster G: Beyond microRNA-novel RNAs derived from small non-coding RNA and their implication in cancer. Cancer Lett. 340:201–211. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Chen X, Ba Y, Ma L, et al: Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 18:997–1006. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Gilad S, Meiri E, Yogev Y, et al: Serum microRNAs are promising novel biomarkers. PLoS One. 3:e31482008. View Article : Google Scholar : PubMed/NCBI

59 

De Planell-Saguer M and Rodicio MC: Analytical aspects of microRNA in diagnostics: a review. Anal Chim Acta. 699:134–152. 2011.PubMed/NCBI

60 

Ajit SK: Circulating microRNAs as biomarkers, therapeutic targets, and signaling molecules. Sensors (Basel). 12:3359–3369. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Schwarzenbach H, Nishida N, Calin GA and Pantel K: Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol. 11:145–156. 2014. View Article : Google Scholar : PubMed/NCBI

62 

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. 141:672–675. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Luo X, Stock C, Burwinkel B and Brenner H: Identification and evaluation of plasma microRNAs for early detection of colorectal cancer. PLoS One. 8:e628802013. View Article : Google Scholar : PubMed/NCBI

64 

Wang J, Huang S-K, Zhao M, et al: Identification of a circulating microRNA signature for colorectal cancer detection. PLoS One. 9:e874512014. View Article : Google Scholar : PubMed/NCBI

65 

Komatsu S, Ichikawa D, Takeshita H, et al: Circulating microRNAs in plasma of patients with oesophageal squamous cell carcinoma. Br J Cancer. 105:104–111. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Wu C, Wang C, Guan X, et al: Diagnostic and prognostic implications of a serum miRNA panel in oesophageal squamous cell carcinoma. PLoS One. 9:e922922014. View Article : Google Scholar : PubMed/NCBI

67 

Chan M, Liaw CS, Ji SM, et al: Identification of circulating microRNA signatures for breast cancer detection. Clin Cancer Res. 19:4477–4487. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Zearo S, Kim E, Zhu Y, Zhao JT, Sidhu SB, Robinson BG and Soon PS: MicroRNA-484 is more highly expressed in serum of early breast cancer patients compared to healthy volunteers. BMC Cancer. 14:2002014. View Article : Google Scholar : PubMed/NCBI

69 

Tsujiura M, Ichikawa D, Komatsu S, et al: Circulating microRNAs in plasma of patients with gastric cancers. Br J Cancer. 102:1174–1179. 2010. View Article : Google Scholar : PubMed/NCBI

70 

Zhu C, Ren C, Han J, et al: A five-microRNA panel in plasma was identified as potential biomarker for early detection of gastric cancer. Br J Cancer. 110:2291–2299. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Zheng H, Liu JY, Song FJ and Chen KX: Advances in circulating microRNAs as diagnostic and prognostic markers for ovarian cancer. Cancer Biol Med. 10:123–130. 2013.PubMed/NCBI

72 

Shapira I, Oswald M, Lovecchio J, et al: Circulating biomarkers for detection of ovarian cancer and predicting cancer outcomes. Br J Cancer. 110:976–983. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Kosaka N, Iguchi H and Ochiya T: Circulating microRNA in body fluid: a new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 101:2087–2092. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Ahmed KA and Xiang J: Mechanisms of cellular communication through intercellular protein transfer. J Cell Mol Med. 15:1458–1473. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Ogorevc E, Kralj-Iglic V and Veranic P: The role of extracellular vesicles in phenotypic cancer transformation. Radiol Oncol. 47:197–205. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Lee TH, D’Asti E, Magnus N, Al-Nedawi K, Meehan B and Rak J: Microvesicles as mediators of intercellular communication in cancer - the emerging science of cellular ‘debris’. Semin Immunopathol. 33:455–467. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Zhang HG and Grizzle WE: Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am J Pathol. 184:28–41. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Choi DS, Kim DK, Kim YK and Gho YS: Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 13:1554–1571. 2013. View Article : Google Scholar : PubMed/NCBI

79 

El Andaloussi S, Mäger I, Breakefield XO and Wood MJA: Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 12:347–357. 2013.PubMed/NCBI

80 

Raposo G and Stoorvogel W: Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Van der Pol E, Böing AN, Harrison P, Sturk A and Nieuwland R: Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. 64:676–705. 2012.PubMed/NCBI

82 

Simpson RJ, Jensen SS and Lim JWE: Proteomic profiling of exosomes: current perspectives. Proteomics. 8:4083–4099. 2008. View Article : Google Scholar : PubMed/NCBI

83 

Nazarenko I, Rupp A-K and Altevogt P: Exosomes as a potential tool for a specific delivery of functional molecules. Methods Mol Biol. 1049:495–511. 2013. View Article : Google Scholar : PubMed/NCBI

84 

Principe S, Hui ABY, Bruce J, Sinha A, Liu FF and Kislinger T: Tumor-derived exosomes and microvesicles in head and neck cancer: implications for tumor biology and biomarker discovery. Proteomics. 13:1608–1623. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Akers JC, Gonda D, Kim R, Carter BS and Chen CC: Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retro-virus-like vesicles, and apoptotic bodies. J Neurooncol. 113:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Mathivanan S, Ji H and Simpson RJ: Exosomes: extracellular organelles important in intercellular communication. J Proteomics. 73:1907–1920. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Kosaka N, Yoshioka Y, Hagiwara K, Tominaga N, Katsuda T and Ochiya T: Trash or treasure: extracellular microRNAs and cell-to-cell communication. Front Genet. 4:1732013. View Article : Google Scholar : PubMed/NCBI

88 

Martins VR, Dias MS and Hainaut P: Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol. 25:66–75. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Kahlert C, Melo SA, Protopopov A, et al: Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 289:3869–3875. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Fang DY, King HW, Li JY and Gleadle JM: Exosomes and the kidney: blaming the messenger. Nephrology (Carlton). 18:1–10. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Gajos-Michniewicz A, Duechler M and Czyz M: MiRNA in melanoma-derived exosomes. Cancer Lett. 347:29–37. 2014. View Article : Google Scholar

92 

Peinado H, Lavotshkin S and Lyden D: The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 21:139–146. 2011. View Article : Google Scholar : PubMed/NCBI

93 

Théry C, Amigorena S, Raposo G and Clayton A: Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. Chapter 3(Unit 3): 222006.PubMed/NCBI

94 

Lässer C: Identification and analysis of circulating exosomal microRNA in human body fluids. Methods Mol Biol. 1024:109–128. 2013.PubMed/NCBI

95 

Gonda DD, Akers JC, Kim R, Kalkanis SN, Hochberg FH, Chen CC and Carter BS: Neuro-oncologic applications of exosomes, microvesicles, and other nano-sized extracellular particles. Neurosurgery. 72:501–510. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Lässer C, Eldh M and Lötvall J: Isolation and characterization of RNA-containing exosomes. J Vis Exp. e30372012.

97 

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

98 

Jenjaroenpun P, Kremenska Y, Nair VM, Kremenskoy M, Joseph B and Kurochkin IV: Characterization of RNA in exosomes secreted by human breast cancer cell lines using next-generation sequencing. Peer J. 1:e2012013. View Article : Google Scholar : PubMed/NCBI

99 

Kruger S, Abd Elmageed ZY, Hawke DH, et al: Molecular characterization of exosome-like vesicles from breast cancer cells. BMC Cancer. 14:442014. View Article : Google Scholar : PubMed/NCBI

100 

Feng DQ, Huang B, Li J, et al: Selective miRNA expression profile in chronic myeloid leukemia K562 cell-derived exosomes. Asian Pac J Cancer Prev. 14:7501–7508. 2013. View Article : Google Scholar : PubMed/NCBI

101 

Xiao D, Ohlendorf J, Chen Y, et al: Identifying mRNA, microRNA and protein profiles of melanoma exosomes. PLoS ONE. 7:e468742012. View Article : Google Scholar : PubMed/NCBI

102 

Rappa G, Mercapide J, Anzanello F, Pope RM and Lorico A: Biochemical and biological characterization of exosomes containing prominin-1/CD133. Mol Cancer. 12:622013. View Article : Google Scholar : PubMed/NCBI

103 

Hessvik NP, Phuyal S, Brech A, Sandvig K and Llorente A: Profiling of microRNAs in exosomes released from PC-3 prostate cancer cells. Biochim Biophys Acta. 1819.1154–1163. 2012.PubMed/NCBI

104 

Kobayashi M, Salomon C, Tapia J, Illanes SE, Mitchell MD and Rice GE: Ovarian cancer cell invasiveness is associated with discordant exosomal sequestration of Let-7 miRNA and miR-200. J Transl Med. 12:42014. View Article : Google Scholar : PubMed/NCBI

105 

Ohshima K, Inoue K, Fujiwara A, et al: Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 5:e132472010. View Article : Google Scholar : PubMed/NCBI

106 

Zöller M: Pancreatic cancer diagnosis by free and exosomal miRNA. World J Gastrointest Pathophysiol. 4:74–90. 2013.PubMed/NCBI

107 

Chiba M, Kimura M and Asari S: Exosomes secreted from human colorectal cancer cell lines contain mRNAs, microRNAs and natural antisense RNAs, that can transfer into the human hepatoma HepG2 and lung cancer A549 cell lines. Oncol Rep. 28:1551–1558. 2012.

108 

Yang M, Chen J, Su F, et al: Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol Cancer. 10:1172011. View Article : Google Scholar : PubMed/NCBI

109 

Umezu T, Ohyashiki K, Kuroda M and Ohyashiki JH: Leukemia cell to endothelial cell communication via exosomal miRNAs. Oncogene. 32:2747–2755. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Kosaka N, Iguchi H, Yoshioka Y, Hagiwara K, Takeshita F and Ochiya T: Competitive interactions of cancer cells and normal cells via secretory microRNAs. J Biol Chem. 287:1397–1405. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Roccaro AM, Sacco A, Maiso P, et al: BM mesenchymal stromal cell-derived exosomes facilitate multiple myeloma progression. J Clin Invest. 123:1542–1555. 2013. View Article : Google Scholar : PubMed/NCBI

112 

Valencia K, Luis-Ravelo D, Bovy N, et al: miRNA cargo within exosome-like vesicle transfer influences metastatic bone colonization. Mol Oncol. 8:689–703. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Zhou W, Fong MY, Min Y, et al: Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell. 25:501–515. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Chen W, Zhong S, Ji M, et al: MicroRNAs delivered by extracellular vesicles: an emerging resistance mechanism for breast cancer. Tumour Biol. 35:2883–2892. 2014. View Article : Google Scholar : PubMed/NCBI

115 

Xiao X, Yu S, Li S, et al: Exosomes: decreased sensitivity of lung cancer A549 cells to cisplatin. PLoS One. 9:e895342014. View Article : Google Scholar : PubMed/NCBI

116 

King HW, Michael MZ and Gleadle JM: Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer. 12:4212012. View Article : Google Scholar : PubMed/NCBI

117 

Tadokoro H, Umezu T, Ohyashiki K, Hirano T and Ohyashiki JH: Exosomes derived from hypoxic leukemia cells enhance tube formation in endothelial cells. J Biol Chem. 288:34343–34351. 2013. View Article : Google Scholar : PubMed/NCBI

118 

Skog J, Würdinger T, van Rijn S, et al: Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 10:1470–1476. 2008. View Article : Google Scholar : PubMed/NCBI

119 

Taylor DD and Gercel-Taylor C: MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 110:13–21. 2008. View Article : Google Scholar : PubMed/NCBI

120 

Tanaka Y, Kamohara H, Kinoshita K, et al: Clinical impact of serum exosomal microRNA-21 as a clinical biomarker in human esophageal squamous cell carcinoma. Cancer. 119:1159–1167. 2013. View Article : Google Scholar : PubMed/NCBI

121 

Ogata-Kawata H, Izumiya M, Kurioka D, et al: Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS One. 9:e929212014. View Article : Google Scholar : PubMed/NCBI

122 

Rabinowits G, Gerçel-Taylor C, Day JM, Taylor DD and Kloecker GH: Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 10:42–46. 2009. View Article : Google Scholar : PubMed/NCBI

123 

Cazzoli R, Buttitta F, Di Nicola M, Malatesta S, Marchetti A, Rom WN and Pass HI: microRNAs derived from circulating exosomes as noninvasive biomarkers for screening and diagnosing lung cancer. J Thorac Oncol. 8:1156–1162. 2013. View Article : Google Scholar : PubMed/NCBI

124 

Rodríguez M, Silva J, López-Alfonso A, et al: Different exosome cargo from plasma/bronchoalveolar lavage in non-small-cell lung cancer. Genes Chromosomes Cancer. Apr 25–2014.(Epub ahead of print).

125 

Li L, Masica D, Ishida M, et al: Human bile contains microRNA-laden extracellular vesicles that can be used for cholangiocarcinoma diagnosis. Hepatology. Feb 4–2014.(Epub ahead of print).

126 

Liu J, Sun H, Wang X, Yu Q, Li S, Yu X and Gong W: Increased exosomal microRNA-21 and microRNA-146a levels in the cervicovaginal lavage specimens of patients with cervical cancer. Int J Mol Sci. 15:758–773. 2014. View Article : Google Scholar : PubMed/NCBI

127 

Kogure T, Yan IK, Lin W-L and Patel T: Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: A mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer. 4:261–272. 2013. View Article : Google Scholar : PubMed/NCBI

128 

Takahashi K, Yan IK, Haga H and Patel T: Modulation of hypoxia-signaling pathways by extracellular linc-RoR. J Cell Sci. 127:1585–1594. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Arita T, Ichikawa D, Konishi H, et al: Circulating long non-coding RNAs in plasma of patients with gastric cancer. Anticancer Res. 33:3185–3193. 2013.PubMed/NCBI

130 

Ren S, Wang F, Shen J, et al: Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur J Cancer. 49:2949–2959. 2013. View Article : Google Scholar : PubMed/NCBI

131 

Panzitt K, Tschernatsch MMO, Guelly C, et al: Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 132:330–342. 2007. View Article : Google Scholar : PubMed/NCBI

132 

Xie H, Ma H and Zhou D: Plasma HULC as a promising novel biomarker for the detection of hepatocellular carcinoma. Biomed Res Int. 2013:1361062013.PubMed/NCBI

133 

Isin M, Ozgur E, Cetin G, Erten N, Aktan M, Gezer U and Dalay N: Investigation of circulating lncRNAs in B-cell neoplasms. Clin Chim Acta. 431:255–259. 2014. View Article : Google Scholar : PubMed/NCBI

134 

Lässer C: Exosomal RNA as biomarkers and the therapeutic potential of exosome vectors. Expert Opin Biol Ther. 12(Suppl 1): S189–S197. 2012.PubMed/NCBI

135 

Lee Y, El Andaloussi S and Wood MJA: Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum Mol Genet. 21:R125–R134. 2012. View Article : Google Scholar : PubMed/NCBI

136 

Kosaka N, Takeshita F, Yoshioka Y, Hagiwara K, Katsuda T, Ono M and Ochiya T: Exosomal tumor-suppressive microRNAs as novel cancer therapy: ‘exocure’ is another choice for cancer treatment. Adv Drug Deliv Rev. 65:376–382. 2013. View Article : Google Scholar : PubMed/NCBI

137 

Ohno S, Takanashi M, Sudo K, et al: Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther. 21:185–191. 2013. View Article : Google Scholar : PubMed/NCBI

138 

Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S and Wood MJA: Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 29:341–345. 2011. View Article : Google Scholar : PubMed/NCBI

139 

Wahlgren J, De Karlson LT, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen P and Valadi H: Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 40:e1302012. View Article : Google Scholar : PubMed/NCBI

140 

Bronisz A, Wang Y, Nowicki MO, et al: Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 74:738–750. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 46 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sato-Kuwabara Y, Melo SA, Soares FA and Calin GA: The fusion of two worlds: Non-coding RNAs and extracellular vesicles - diagnostic and therapeutic implications (Review). Int J Oncol 46: 17-27, 2015
APA
Sato-Kuwabara, Y., Melo, S.A., Soares, F.A., & Calin, G.A. (2015). The fusion of two worlds: Non-coding RNAs and extracellular vesicles - diagnostic and therapeutic implications (Review). International Journal of Oncology, 46, 17-27. https://doi.org/10.3892/ijo.2014.2712
MLA
Sato-Kuwabara, Y., Melo, S. A., Soares, F. A., Calin, G. A."The fusion of two worlds: Non-coding RNAs and extracellular vesicles - diagnostic and therapeutic implications (Review)". International Journal of Oncology 46.1 (2015): 17-27.
Chicago
Sato-Kuwabara, Y., Melo, S. A., Soares, F. A., Calin, G. A."The fusion of two worlds: Non-coding RNAs and extracellular vesicles - diagnostic and therapeutic implications (Review)". International Journal of Oncology 46, no. 1 (2015): 17-27. https://doi.org/10.3892/ijo.2014.2712