Skip to main content
Molecular Oncology logoLink to Molecular Oncology
. 2017 Oct 26;11(12):1673–1686. doi: 10.1002/1878-0261.12144

Cell‐to‐cell communication: microRNAs as hormones

Recep Bayraktar 1, Katrien Van Roosbroeck 1, George A Calin 1,2,3,
PMCID: PMC5709614  PMID: 29024380

Abstract

Mammalian cells can release different types of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies. Accumulating evidence suggests that EVs play a role in cell‐to‐cell communication within the tumor microenvironment. EVs’ components, such as proteins, noncoding RNAs [microRNAs (miRNAs), and long noncoding RNAs (lncRNAs)], messenger RNAs (mRNAs), DNA, and lipids, can mediate paracrine signaling in the tumor microenvironment. Recently, miRNAs encapsulated in secreted EVs have been identified in the extracellular space. Mature miRNAs that participate in intercellular communication are released from most cells, often within EVs, and disseminate through the extracellular fluid to reach remote target cells, including tumor cells, whose phenotypes they can influence by regulating mRNA and protein expression either as tumor suppressors or as oncogenes, depending on their targets. In this review, we discuss the roles of miRNAs in intercellular communication, the biological function of extracellular miRNAs, and their potential applications for diagnosis and therapeutics. We will give examples of miRNAs that behave as hormones.

Keywords: cell–cell communication, circulating miRNAs, exosomes, extracellular vesicles, microRNAs, tumor microenvironment


Abbreviations

CAAs

cancer‐associated adipocytes

CAFs

cancer‐associated fibroblasts

CLL

chronic lymphocytic leukemia

ECM

extracellular matrix

EVs

extracellular vesicles

FGF2

fibroblast growth factor 2

HDL

high‐density lipoproteins

LDL

low‐density lipoproteins

miRNAs

microRNAs

MVs

microvesicles

NFs

normal fibroblasts

RISC

RNA‐induced silencing complex

TAMs

tumor‐associated macrophages

TGF‐β

transforming growth factor‐β

TLDA

TaqMan Low‐Density Arrays

TLRs

toll‐like receptors

TME

tumor microenvironment

TMVs

tumor‐derived MVs

VEGF

vascular endothelial growth factor

Introduction

MicroRNAs (miRNAs) are a class of short noncoding RNAs that play key roles in almost all biological pathways in mammalian and other organisms (Ambros, 2004; Bartel, 2004). miRNAs are 19–25 nucleotides in length and can regulate gene expression both at the transcriptional and at translational level by repressing target genes (Ambros, 2004; Bartel, 2004). Initially, miRNAs are transcribed as thousand‐base‐long primary transcripts by RNA polymerase II and are called precursor miRNAs (Ambros, 2004; Bartel, 2004). Precursor miRNAs are exported to the cytoplasm via exportin 5, where they are integrated into DICER and RNA‐induced silencing complex (RISC), which includes argonaute proteins. Finally, mature miRNA strands specifically bind to partially complementary sequence motifs in the 3′ untranslated region of target mRNA (Ambros, 2004; Bartel, 2004). In 2002, we demonstrated that a coding gene on a region of the long arm of chromosome 13, at 13q14, was frequently deleted in chronic lymphocytic leukemia (CLL) and that this deletion caused the loss of two miRNAs (miR‐15a and miR‐16‐1) (Bullrich et al., 2001). Subsequently, we reported that the chromosome 13q14 region was deleted in more than 65% of CLL patients and that allelic loss in this region correlates with downregulation of both miR‐15 and miR‐16 expression (Calin et al., 2002). This finding opened new opportunities for noncoding RNAs in cancer research.

Extracellular vesicles (EVs) are small membrane vesicles, such as ectosomes, microparticles, microvesicles (MVs), tumor‐derived MVs (TMVs), exosomes, and oncosomes that are produced by different mechanisms and can be released from almost all cell types (Cocucci and Meldolesi, 2011; Raposo and Stoorvogel, 2013; Redis et al., 2012; Valadi et al., 2007). EVs participate in cell–cell communication and can typically be classified based on their size (from 4 to 10 microns), intracellular origin, and density (Cocucci and Meldolesi, 2011; Raposo and Stoorvogel, 2013; Valadi et al., 2007). EVs can be found in all different body fluids, such as blood, serum, plasma, saliva, urine, and pleural effusions (Fernandez‐Mercado et al., 2015; Liu et al., 2017a; Mitchell et al., 2008; Ortiz‐Quintero, 2016; Weber et al., 2010). In the last decade, several studies have shown that EVs are enriched for various proteins, such as cytokines, messenger RNAs, lipids, and noncoding RNAs, such as miRNAs and long noncoding RNAs (lncRNAs) (Colombo et al., 2013; Valadi et al., 2007). The content of EVs, which are shed by almost all cell types under both physiological and pathological conditions, can be transported from a parental cell to neighboring cells (Arita et al., 2008; Cocucci and Meldolesi, 2011; Thery et al., 2009; Trajkovic et al., 2008). Through their protein, RNA, and DNA cargoes, EVs can regulate important biological functions of recipient cells, such as proliferation, angiogenesis, and apoptosis, processes that are deregulated in human cancers (Clayton et al., 2004; Kanlikilicer et al., 2016; Rashed et al., 2017; Valadi et al., 2007).

In this review, we summarize the current knowledge regarding the contributions of miRNAs in secreted EVs, their potential clinical and therapeutic applications, biological significance, relationship with tumors, and roles in cell–cell communication.

How cells communicate

The best‐known mechanisms of cell–cell communication are chemical receptor‐mediated events, direct cell–cell interaction, and cell–cell synapses (Valadi et al., 2007). In recent years, an additional mechanism has been identified via extracellular vesicles (Fig. 1), which will be described in the following sections.

Figure 1.

Figure 1

MicroRNAs release and uptake mechanism between donor and recipient cells. Biogenesis of exosomes. Early endosomes originate from the cell membrane via endocytosis. Multivesicular bodies originate by invagination of the plasma membrane. Multivesicular bodies fuse with the plasma membrane and exosomes are released into the extracellular space. Some types of miRNAs are generally localized in membrane‐derived vesicles (exosomes, microvesicles, apoptotic bodies), while some miRNAs are found mainly in miRNA‐binding protein complexes, such as Ago‐2, or high‐density lipoproteins (HDL). Finally, miRNAs enter into recipient cells and interact with specific target genes.

EVs: an alternative type of cell–cell communication

Exosomes are the best characterized class of EVs; they are secreted by almost all cell types, are 40 to 100 nm in diameter, and are enriched in a 1.13–1.19 g·mL−1 fraction in a sucrose density gradient (Kanada et al., 2015; Turturici et al., 2014). The protein and lipid composition of exosomes reflects their cellular sources (Staubach et al., 2009; Yim et al., 2016). The most common exosomal proteins are annexins, tetraspanins (CD63, CD81, CD82, and CD9), and heat‐shock proteins (Hsp60, Hsp70, and Hsp90). In 2007, Valadi et al. showed a new mechanism of cell–cell communication: delivery of RNA by transfer through exosomes. They demonstrated that exosomes carry both mRNAs and miRNAs when released from mouse and human mast cells and that many of these RNAs seem to be packaged exclusively into exosomes. Zhang et al. (2010) have been reported that cells can selectively package miRNAs into MVs and secreted miRNAs can act as signaling molecules mediating intercellular communication. In their study, it has been demonstrated that miR‐150 (a leukocyte and lymphocyte‐specific miRNA) was selectively packaged into MVs and exosomal miR‐150 entered the recipient cells (Zhang et al., 2010). Exosomal miR‐150 derived from THP1 cells can enter and deliver into human microvascular endothelial HMEC‐1 cells, and delivered miR‐150 dramatically reduced c‐Myb expression level and promoted cell migration (Zhang et al., 2010).

High‐density lipoproteins as carriers of extracellular miRNAs

Circulating miRNAs can be transported from donor cells to recipient cells by exosomes, microvesicles, apoptotic bodies, lipoproteins, and ribonucleoproteins (such as nucleophosmin 1 and argonaute 2) (Boon and Vickers, 2013; Vickers and Remaley, 2012). New studies have revealed that high‐density lipoproteins (HDL) and low‐density lipoproteins (LDL) can transport miRNAs into cells as well (Vickers et al., 2011). In 2011, Vickers et al. demonstrated that several types of RNAs, such as miRNAs and tRNAs, could be detected in HDL and LDL particles. They profiled approximately 700 miRNAs in pure human HDL particles using real‐time PCR‐based TaqMan Low‐Density Arrays (TLDA) (Vickers et al., 2011) and found that miR‐135a*, miR‐188‐5p, and miR‐877 were upregulated in healthy control HDL particles, while miR‐223, miR‐105, and miR‐106a were upregulated in HDL particles from patients with familial hypercholesterolemia. In addition, they showed that HDL‐mediated miRNA‐223 transport represses cholesterol uptake by direct binding to the 3′‐UTR of the scavenger receptor BI (SR‐BI) mRNA and controls its expression and function (Vickers et al., 2011). Tabet et al. (2014) demonstrated that miR‐223 is transferred from native HDL to endothelial cells and HDL inhibits expression of intercellular adhesion molecule 1 (ICAM‐1) through the transfer of miR‐223 to endothelial cells. All these findings support that HDL‐miRNA delivery is a new mechanism of miRNA transport.

Transfer of biological information between tumor microenvironment and malignant cells via miRNAs

It has long been known that small molecules, cytokines, and growth factors mediate functional interactions between cancer cells and the tumor microenvironment (Schoepp et al., 2017). However, recent studies have revealed that cancer cells also transfer functional information at the paracrine level using EVs. In fact, it appears that the cargo of EVs can influence the stroma by activating molecular pathways, which are at least partially different from those modulated by soluble factors. The tumor microenvironment (TME) plays critical roles in the initiation, development, and progression of cancer. The TME includes extracellular matrix as well as different types of cells, including cancer‐associated fibroblasts (CAFs), tumor‐associated macrophages (TAMs), endothelial cells, pericytes, and immune cells (Berindan‐Neagoe and Calin, 2014; Friedl and Alexander, 2011; Laberge et al., 2012).

miRNAs and cancer‐associated fibroblasts

Fibroblasts are the most common constituent in human tissues and tumors, and they regulate tissue repair and inflammation during wound healing (Kalluri and Zeisberg, 2006). Previous studies have reported that activated tumor fibroblasts or cancer‐associated fibroblasts are phenotypically and functionally different from normal fibroblasts (Zhang et al., 2017). CAFs have been shown to stimulate cancer progression and proliferation through helping create the extracellular matrix (ECM) and through the secretion of a variety of cytokines, chemokines, and growth factors, such as vascular endothelial growth factor (VEGF), transforming growth factor‐β (TGF‐β), and fibroblast growth factor 2 (FGF2) (Kalluri, 2016; Kalluri and Zeisberg, 2006; Orimo et al., 2005; Shen et al., 2016). Accumulating evidence suggests that miRNAs play key roles in the activation and transition of fibroblasts (Aprelikova et al., 2013; Mitra et al., 2012).

Recent studies have shown deregulated miRNA expression in CAFs from clinical specimens. For instance, in 2012, Mitra et al. demonstrated that miR‐155 was upregulated, while miR‐31 and miR‐214 were downregulated, in ovarian CAFs when compared with primary human normal omental fibroblasts or primary human CAFs. They found that triple transfection of these miRNAs with mimics activated tumor‐promoting functions, such as migration, invasion, and colony formation, in primary human normal omental fibroblasts (Mitra et al., 2012). In 2014, Yeung et al. used next‐generation sequencing to show that miR‐21 expression is upregulated in exosomes and tissue isolated from cancer‐associated adipocytes (CAAs) and CAFs (Au Yeung et al., 2016). Moreover, it has been shown that exosomes can be secreted by CAFs and taken up by cancer cells, as isolated exosomes from primary CAFs transfected with labeled miR‐21 successfully entered ovarian cancer cells (Au Yeung et al., 2016). They confirmed transfer of miR‐21 from primary CAFs to ovarian cancer cells by co‐culturing CAFs transfected with labeled miR‐21 with ovarian cancer cells (Au Yeung et al., 2016). In addition, miR‐21 was frequently upregulated in CAFs from both human pancreatic ductal adenocarcinoma samples and primary cell cultures, and higher expression of miR‐21 was significantly correlated with decreased overall survival in pancreatic ductal adenocarcinoma patients (Donahue et al., 2014; Kadera et al., 2013). In esophageal squamous cell carcinoma, miR‐21 expression in tumor tissues was found to be primarily localized in the stroma adjacent to the cancer cells (Nouraee et al., 2013). We further showed that miR‐21 could be secreted by fibroblasts in the microenvironment and taken up by cancer cells, resulting in increased migration and invasion potential of esophageal tumor cells (Nouraee et al., 2013). However, the exact mechanism of miR‐21 secretion and transportation was not explored. Finally, miR‐21 was found to induce the expression of CAF markers, suggesting its involvement in the ‘activation’ of fibroblasts to CAFs (Nouraee et al., 2013).

Yang et al. (2014) investigated the miRNA signatures of CAFs and normal fibroblasts isolated from human gastric cancer tissue, and they reported that miR‐34b, miR‐93, miR‐301a, and miR‐106b were significantly upregulated, while miR‐483‐3p, miR‐26a, let‐7g, miR‐148a, miR‐145, miR‐424, and miR‐214 were significantly downregulated in CAFs compared with corresponding normal fibroblasts. To understand the clinical significance of miR‐106b expression, they analyzed a subset of patients with gastric cancer and found that overall survival was dramatically higher in patients with low miR‐106b expression than in patients with high miR‐106b expression (Yang et al., 2014). Baroni et al. (2016) revealed that in patients with different breast cancer subtypes (luminal‐A, luminal‐B, HER2, and triple negative), miR‐9 was upregulated in primary triple‐negative breast CAFs when compared to normal fibroblasts (NFs). They also demonstrated that miR‐9 secreted from tumors can be transferred to recipient normal fibroblasts via exosomes and that miR‐9 uptake leads to increased cell motility in normal fibroblast cells that overexpress this miRNA (Baroni et al., 2016). They co‐injected MDA‐MB‐468 triple‐negative breast cancer cells with either normal fibroblasts or normal fibroblasts that overexpressed miR‐9 into the mammary fat pad of 8‐week‐old female SCID mice, and found that tumor volume was significantly increased in mice co‐injected with MDA‐MB‐468 breast cancer cells and normal fibroblasts overexpressing miR‐9, compared with the control group (Baroni et al., 2016). These findings showed that higher expression levels of miR‐9 in fibroblasts affect breast cancer progression and provide new insights into the role of miR‐9 in the crosstalk between cancer cells and stroma (Baroni et al., 2016). (Donnarumma et al. (2017) showed that the release of specific miRNAs from CAF exosomes can promote oncogenic signaling in breast cancer. Their results demonstrated that three miRNAs (miR‐21, miR‐143, and miR‐378e) were significantly upregulated in CAF exosomes in respect to NF exosomes (Donnarumma et al., 2017). When T47D luminal‐A breast cancer cells were treated with exosomes isolated from NFs and CAFs and labeled with PKH26, they found that T47D cells are able to take up exosomes derived from CAFs and NFs (Donnarumma et al., 2017). Furthermore, they observed that CAF exosomes treatment significantly promoted stemness, EMT, invasiveness capacity, and anchorage‐independent cell growth (Donnarumma et al., 2017). Pang et al. (2015) showed that miR‐155 was upregulated in exosomes derived from pancreatic cancer cells. They co‐cultured primary mouse pancreatic fibroblast cells with BxPC‐3 and SW1990 pancreatic cancer cells and showed that normal fibroblasts could convert into CAFs after miR‐155 had been taken up (Pang et al., 2015). They demonstrated that miR‐155 targets the TP53INP1 protein, leading to increased alpha‐SMA protein expression in normal fibroblasts (Pang et al., 2015). Altogether, their results indicated that miR‐155 in exosomes secreted from pancreatic cancer cells might activate normal fibroblasts by targeting TP53INP1 (Pang et al., 2015). In conclusion, CAFs regulate tumorigenesis in many cancer types through exosome‐mediated delivery of specific miRNAs (Fig. 2).

Figure 2.

Figure 2

Schematic illustration of the interaction between primary tumor cells and tumor microenvironment through miRNAs. miRNAs can play a key role in cell–cell communication in several physiological and pathophysiological processes associated with many human diseases, including cancer. Selected examples of paracrine miRNA signaling between primary tumor cells, immune cells, and endothelial cells are shown.

miRNAs and tumor‐associated macrophages

Macrophages originate from precursor blood monocytes and play key roles in the immune system. Depending on their polarization state, they can perform distinct functions in different biological processes. Traditionally, macrophages can be categorized as either classically activated (M1) macrophages or alternatively activated (M2) macrophages (Biswas and Mantovani, 2010; Lawrence and Natoli, 2011; Mosser and Edwards, 2008). M1 macrophages play key roles in the pro‐inflammatory and antitumor responses and are induced by toll‐like receptors (TLRs), IFN‐γ, and lipopolysaccharide (LPS), while M2 macrophages are activated by IL‐4 and IL‐13 and are responsible for angiogenesis, tissue remodeling, and tumor progression (Mantovani et al., 2007; Zhang et al., 2012).

Tumor‐associated macrophages (TAMs) are generally composed of M2‐polarized macrophages and associated with poor clinical outcomes in patient with various cancer types (Noy and Pollard, 2014; Steidl et al., 2010). TAMs share many similarities with the M2‐polarized macrophages population and can affect different protumoral functions, such as promotion of angiogenesis, matrix remodeling, secretion of growth factors, and suppression of inflammation and adaptive immunity (Binnemars‐Postma et al., 2017; Sica et al., 2006). Recent studies showed that new molecular players, miRNAs, have an important role in the polarization of TAMs and on monocyte/macrophage lineage development (Baer et al., 2016; Li et al., 2015; Wang et al., 2015). (Baer et al. (2016) showed that deletion of the miRNA‐processing enzyme DICER regulates TAM programming. They generated Dicer1‐knockout mice and employed orthotropic mammary carcinoma, subcutaneous lung and colorectal carcinoma models. Macrophage‐specific Dicer1 deletion significantly delayed tumor progression. They demonstrate that let‐7‐5p family members were upregulated in Dicer‐knockout and wild‐type TAMs (Baer et al., 2016). These findings suggest that Dicer1 deletion can regulate the antitumor phenotype of M2 TAMs to M1‐like TAMs in different types of tumor models (Baer et al., 2016). Squadrito et al. (2012) described that the human and mouse mannose receptor MRC1 gene encodes miR‐511‐3p, an intronic miRNA, and showed that miR‐511‐3p and MRC1 are transcriptionally co‐regulated. They overexpressed miR‐511‐3p in BM‐derived hematopoietic cells and injected Lewis lung carcinoma (LLC) cells in mice and were able to show that overexpression of miR‐511‐3p inhibited tumor growth in the LLC model (Squadrito et al., 2012). (Sonda et al. (2013) identified 79 differentially expressed miRNAs in either tumor‐infiltrating versus normal CD11b+ cells purified from the spleen of healthy mice. They found that miR‐142‐3p and miR‐150 were the most downregulated miRNAs in tumor‐infiltrating CD11b+ cells and computationally determined genes that are involved in monocyte–macrophage commitment, as well as potential target genes of miR‐142‐3p (Sonda et al., 2013). They further showed that miR‐142‐3p directly regulates gp130, which is a common subunit of the interleukin‐6 cytokine receptor family that regulates macrophage generation (Sonda et al., 2013). A comprehensive list of extracellular miRNAs shuttling between cancer cells and their neighboring cells can be found in Table 1.

Table 1.

MicroRNAs as hormones: extracellular miRNAs shuttling between cancer cells and their neighboring cells

miRNA Donor cells Accepting cells Target/Pathway Function/impact References
miR‐21,‐143,‐378 CAFs Breast cancer cells EMT Promotes the stemness and EMT Donnarumma et al. (2017)
miR‐21 CAFs Ovarian cancer cells APAF1 Stimulates cancer cell motility and invasion Au Yeung et al. (2016)
miR‐146a CAFs Pancreatic cells Snail Increases proliferation Richards et al. (2017)
miR‐9 CAFs Normal fibroblasts CDH1 Enhances cell motility Baroni et al. (2016)
miR‐29b CAFs Breast cancer cells CCL11 and CXCL14 Inhibits breast cancer cellular viability and metastasis Liu et al. (2017b)
miR‐409 CAFs Prostate cancer cells RSU1 and STAG2 Induces cell growth and EMT Josson et al. (2015)
miR‐133b CAFs Normal fibroblasts IL6 and TGF‐β Promotes fibroblast activation Doldi et al. (2015)
miR‐320a CAFs Hepatocellular carcinoma cells PBX3 Inhibits tumor progression Zhang et al. (2017)
miR‐221 CAFs Breast cancer cells IL6/Stat3 Promotes hormonal therapy resistance Sansone et al. (2017)
miR‐124 CAFs Oral cancer cells CCL2 and IL‐8 Promotes cell growth and migration Li et al. (2017)
miR‐7 CAFs Head and neck cancer cells RASSF2 Enhances cell proliferation and migration Shen et al. (2017)
miR‐101 CAFs Hepatocellular cancer cells TGF‐βR1, Smad2, and VE‐cadherin Inhibits vascular mimicry formation Yang et al. (2016)
miR‐141,‐146b‐5p CAFs Breast cancer cells p16 Represses the migration and invasiveness Al‐Khalaf and Aboussekhra (2017)
miR‐1, ‐206, ‐31 CAFs Lung cancer cells FOXO3a/VEGFA/CCL2 Promotes migration and tumor growth Shen et al. (2016)
miR‐214 CAFs Ovarian cancer cells CCL5 Activate tumor‐promoting functions Mitra et al. (2012)
miR‐940 TAMs Ovarian cancer cells CD206 and CD163 Tumor‐promoting function Chen et al. (2017)
miR‐21 TAMs Gastric cancer cells PTEN Suppresses cell apoptosis and enhances activation of PI3K/AKT Zheng et al. (2017)
miR‐720 TAMs Breast cancer cells GATA3 M2 polarization Zhong and Yi (2016)
miR‐155 TAMs Hepatocellular cancer cells C/EBPbeta Suppresses cytokine production He et al. (2009)
miR‐511‐3p TAMs Macrophage expressing MRC1 cells ROCK2 Inhibits tumor growth and alters tumor blood vessel morphology Squadrito et al. (2012)
let‐7b TAMs Prostate cancer TAMs IL‐12, IL‐23, IL‐10 TNF‐α, and CXCL12 Inhibits macrophage migration and tumor growth Li et al. (2015)
miR‐125a TAMs Epithelial cells FIH1 and IRF4 Enhances M1 and attenuates M2 polarization Zhao et al. (2016)

miRNAs function as toll‐like receptors ligands

Toll‐like receptors (TLRs) are highly conserved pathogen recognition receptors that play a key role in the active innate immune system of all multicellular organisms (Takeda et al., 2003). TLRs are widely expressed in different cell types, including macrophages, dendritic cells (DCs), neutrophils, natural killer cells, and fibroblasts (Batool et al., 2016; Visintin et al., 2001). TLRs recognize conserved structural motifs of pathogens (pathogen‐associated molecular patterns, PAMPs) including flagellins, nucleic acids, lipoproteins, saccharides, and peptidoglycans (Akira, 2009). In addition, TLRs induce chronic inflammation and regulate the maintenance of tissue homeostasis. Despite their structural and functional similarities, each member of the TLR family recognizes specific ligands and has a different cellular localization. TLR1, 2, 4, 5, 6, and 10 are expressed on cell surfaces that are involved in the recognition of cell surface molecules, while TLR3, 7, 8, and 9 are located intracellularly and recognize nucleic acids.

Previous studies showed that TLRs, especially TLR7 and TLR8, recognize GU‐rich single‐stranded RNA (ssRNA) derived from human immunodeficiency virus‐1 (HIV‐1) in certain cell types, such as dendritic cells and B lymphocytes, which leads to cell activation and IFN‐α release (Heil et al., 2004; Lund et al., 2004). In 2012, Fabbri et al. showed that tumor‐secreted miRNAs are involved in intercellular communication in the tumor microenvironment as they can bind and activate TLR7 and TLR8 (Fabbri et al., 2012). They found that miR‐16, miR‐21, miR‐27b, and miR‐29a were highly expressed in exosomes (Fabbri et al., 2012). Interestingly, miR‐16 was upregulated in exosomes from derived HEK cells, while miR‐21 and miR‐29a were upregulated in exosomes from derived non‐small‐cell lung cancer cells (Fabbri et al., 2012). They observed that lung cancer‐specific miR‐21 and miR‐29a bind to TLR7 and TLR8 and activate TLR‐mediated NF‐κB signaling, as well as increase secretion of the prometastatic and pro‐inflammatory cytokines TNF‐α and IL‐6 (Fabbri et al., 2012). Lehmann and colleagues showed that extracellular let‐7 is a potential activator of TLR7 signaling in both immune cells and neurons (Lehmann et al., 2012). They found that extracellular let‐7 interacts with TLR7, which is expressed on central nervous system (CNS) neurons, and they detected TLR7 mRNA in the mouse brain cortex (Lehmann et al., 2012). These findings suggest that in addition to cancer‐specific exosomal miRNAs, let‐7 can bind to TLR7 and plays a key role in neurodegenerative diseases such as Alzheimer disease.

Circulating miRNAs as biomarkers

Several types of malignancies require early and sensitive detection in order to ensure effective treatment and management of the patients. Abnormal expression of specific miRNAs was observed during the progression of several types of cancer (Fabbri et al., 2008). Accumulating evidence demonstrates that specific miRNAs hold great promise as novel biomarkers for clinical diagnosis of many types of diseases, including cancer (Berindan‐Neagoe et al., 2014). Especially, circulating miRNAs are remarkably stable in the blood. Therefore, they can be extracted from blood and measured in blood or other bodily fluids for each patient (Cheng, 2015; Larrea et al., 2016). To measure circulating miRNAs, methods such as qRT‐PCR (LNA‐based, TaqMan, or other proprietary technologies), digital PCR (dPCR), microarrays, and next‐generation sequencing (NGS) are used (Larrea et al., 2016; Van Roosbroeck et al., 2013).

Hematological cancers

As circulating miRNAs represent a class of ideal biomarkers for hematological cancer diagnosis, many studies have been performed to determine their potential and are reviewed elsewhere (Chen et al., 2014a; Lawrie, 2013). In this section, we give a few examples of specific circulating miRNAs that have been shown to be altered in the plasma of hematological cancer patients. The oncogenic role of especially miR‐21, miR‐155, miR‐150, and miR‐210 are well established in hematological malignancies (Fabbri et al., 2008; Fernandez‐Mercado et al., 2015; Munker and Calin, 2011). In 2008, Lawrie et al. compared miR‐155, miR‐21, and miR‐210 expression levels in serum samples from diffuse large B‐cell lymphoma (DLBCL) patients with those of healthy controls, and found that circulating miR‐155, miR‐21, and miR‐210 levels were significantly upregulated in DLBCL patients and that serum levels of miR‐21 are associated with relapse‐free survival in DLBCL. Additionally, levels of miR‐21 were upregulated in DLBCL cell lines and clinical specimens of DLBCL (Chen et al., 2014b). These findings suggest that miR‐21 might function as a biomarker for the diagnosis of DLBCL. Our previous studies showed that miR‐155 was significantly overexpressed in monoclonal B‐cell lymphocytosis (MBL) when compared with normal B cells and that miR‐155 overexpression in plasma is a predictor of poor response to therapy (Ferrajoli et al., 2013). Similar studies have reported that miR‐150 and miR‐342 were significantly downregulated in the plasma of acute myeloid leukemia (AML) patients at diagnosis compared to healthy controls and that these microRNAs are candidate biomarkers and potential predictors of relapse in AML (Fayyad‐Kazan et al., 2013). In addition, circulating miR‐192 has been found to be significantly downregulated in patients with chronic lymphocytic leukemia (CLL) compared with healthy individuals (Fathullahzadeh et al., 2016). Finally, Filip et al. (2017) showed that miR‐34a‐5p, miR31‐5p, miR‐155‐5p, miR‐150‐5p, miR‐15a‐3p, and miR‐29a‐3p were upregulated in serum samples of CLL patients compared with healthy individuals.

Solid cancers

Several studies using microarray platforms to investigate miRNA biomarker potential in solid tumors were published, among them the study of Volinia and coworkers, who described a large‐scale detailed analysis of the miRNA profiles in 540 samples from six solid tumors: breast, colon, lung, pancreas, prostate, and stomach (Volinia et al., 2006). This screening showed that miR‐21, miR‐191, and miR‐17‐5p are significantly upregulated in all of the tumor types and that miR‐29b‐2, miR‐223, miR‐128b, miR‐199a‐1, miR‐24‐1, miR‐24‐2, miR‐146, miR‐155, miR‐181b‐1, miR‐20a, miR‐107, miR‐32, miR‐92, miR‐214, miR‐30c, miR‐25, miR‐221, and miR‐106a were overexpressed in three or more types of solid cancers (Volinia et al., 2006). After identifying the presence of miRNAs in solid tumors, circulating miRNA signatures have been widely described in many cancer types (Chen et al., 2008; Lawrie et al., 2008; Mitchell et al., 2008). For instance, Hannafon et al. showed that miR‐1246 was upregulated in exosomes from breast cancer cells compared to normal mammary epithelial cells and mouse plasma. When they analyzed human plasma samples, miR‐1246 and miR‐21 were detected at significantly higher levels in the plasma exosomes of 16 breast cancer patients as compared to the plasma exosomes of healthy control samples (Hannafon et al., 2016). In another example, Zhang et al. (2015) identified 66 circulating miRNAs that were upregulated, while 32 circulating miRNAs were downregulated in serum samples of ovarian cancer patients compared with serum samples from healthy controls. More specifically, miR‐497, miR‐16‐2*, miR‐195, and miR‐2861 were expressed at significantly higher levels in serum samples of ovarian cancer patients compared to cervical intraepithelial neoplasia patients and healthy control subjects (Zhang et al., 2015). Xu et al. (2016) identified 13 miRNAs deregulated in plasma in patients with PDAC: miR‐106b‐3p, miR‐126‐3p, miR‐1271, miR‐1285, miR‐19b‐3p, miR‐26b‐3p, miR‐296‐5p, miR‐486‐5p, miR‐663B, miR‐7–5p, miR‐938, miR‐942, and miR‐181c‐5p. Of particular interest was miR‐486‐5p, which showed diagnostic value in pancreatic cancer patients when compared with healthy controls and patients with chronic pancreatitis (Xu et al., 2016). In addition, Zhang et al. (2016) report that miR‐1246 and miR‐1290 were overexpressed in serum samples of patients with non‐small‐cell lung cancer (NSCLC) when compared to healthy individuals. After overexpression of these miRNAs in lung epithelial cells, ectopic overexpression of miR‐1246 repressed PRL36A, GLIPR1, HAS2, NCKAP5, MT1G, and CYP4F11, whereas overexpression of miR‐1290 repressed MT1G, MT1H, GLIPR1, CYP4F11, and NCKAP5 (Zhang et al., 2016). Furthermore, they identified the putative tumor suppressor MT1G as a direct target of miR‐1246 and miR‐1290 [76]. Their observations indicate that miR‐1246 and miR‐1290 can behave as noninvasive biomarkers that may be used for the early detection of lung cancer (Zhang et al., 2016). Finally, Zhu et al. (2016) showed that the serum levels of miR‐182, miR‐183, and miR‐210 were significantly upregulated and that miR‐126 levels were significantly downregulated in NSCLC patients compared with healthy controls.

Novel miRNA‐based therapeutic approaches

As cancer‐specific miRNAs are master regulators of many critical oncogenic pathways, their utilization for cancer diagnosis and as promising potential therapeutic target for clinical treatment strategies or personalized medicine is becoming more and more clear (Ling et al., 2013). After discovering the role of miRNAs in cell–cell signaling and TME, targeting miRNAs as anticancer therapeutic strategy is becoming more realistic and promising. Several groups designed miRNA overexpression or inhibition systems as a therapeutic strategy, and this approach led to promising outcomes in vivo (Bayraktar et al., 2017; Hydbring et al., 2017; Mangala et al., 2016). For instance, Mangala et al. showed that miR‐106b‐5p, miR‐30c‐5p, and miR‐141‐3p were significantly upregulated in ovarian cancer‐associated endothelial cells compared to normal endothelial cells. Silencing of these miRNAs leads to restoration of tight junction function and in turn decreases angiogenesis. To evaluate the in vivo biological effects of miR‐106b‐5p and miR‐30c‐5p silencing, they established an ovarian cancer orthotopic mouse model and found that miR‐106b‐5p inhibitor treatment resulted in 50% reduction in tumor growth, while miR‐30c‐5p inhibitor treatment resulted in ~25% reduction in tumor growth in ovarian cancer orthotopic mouse model (Mangala et al., 2016). The combination of miRNA mimic or inhibitor delivery with chemotherapeutic drugs has recently been used to enhance the efficiency of treatment in several types of tumor. Recently, we showed that overexpression of miR‐155 induces chemoresistance in lung cancer cells and acute lymphoblastic leukemia cells (Van Roosbroeck et al., 2017). Additionally, miR‐155‐overexpressing tumors became resistant to cisplatin treatment in an orthotopic lung cancer mouse model (Van Roosbroeck et al., 2017). Administration of miR‐155 inhibitor alone significantly reduced number of tumors, tumor size, and aggregate mass of metastasis, but when we combined miR‐155 inhibitor with cisplatin treatment, the chemotherapy resistance was almost completely reversed (Van Roosbroeck et al., 2017).

Another novel approach is the combination of small interfering RNA (siRNA) and miRNA‐based treatment that can allow a ‘boosting’ effect for targeting oncogenic pathways and repressing cancer growth (Nishimura et al., 2013). We proved that combined inhibition of EphA2, an ovarian cancer oncogene, with EphA2 siRNA and miR‐520d‐3p mimic exhibits synergistic effects (Nishimura et al., 2013). We identified miR‐520d‐3p as a tumor suppressor upstream of EphA2, which is associated with longer overall and relapse‐free survival time in ovarian cancer patients (Nishimura et al., 2013). In this study, miR‐520d‐3p was shown to directly target EphA2 and EphB2 and inhibit their protein expression in ovarian cancer cells. Treatment with the combination of miR‐520d‐3p mimic and EphA2 siRNA resulted in less invasion/migration by HeyA8 and SKOV3ip1 cells than either miR‐520d‐3p or EphA2 siRNA alone or the controls. Moreover, in vivo therapeutic delivery of miR‐520d‐3p mimic and EphA2 siRNA induced potent synergy, resulting in substantial inhibition of tumor growth when compared with individual treatments in ovarian cancer tumor xenograft models (Nishimura et al., 2013).

Currently, some miRNA‐associated therapies are being evaluated in ongoing Phase I clinical trials in cancer (Van Roosbroeck and Calin, 2017). The first miRNA‐based cancer therapy was a liposome‐formulated synthetic miR‐34a mimic (MRX34) (ClinicalTrials.gov, ID: NCT01829971). Because of multiple immune‐related severe adverse events observed in cancer patients receiving MRX34, this study has been terminated early (Van Roosbroeck and Calin, 2017). miRNAs can target multiple genes simultaneously and lead to unexpected side effects and unwanted toxicities. However, combination therapies of miRNAs with siRNA and/or chemotherapy can reduce the risks of adverse events and increase therapeutic synergy as compared with monotherapy.

Conclusion

Since the discovery of the first miRNA, several thousands of miRNAs have been identified in humans, and studies on miRNAs have increased, remarkably during the last decade. Furthermore, miRNAs are frequently deregulated in human diseases including cancer, which offers many opportunities for diagnosis, prognosis, and treatment of human diseases. Recently, it was found that miRNAs are released by donor cells, play a key role in the process of cell‐to‐cell communication, influence the phenotype of recipient cells, and likely reach many distant tissues. Taken together, these miRNAs can serve as valuable biomarkers for various pathological conditions.

Acknowledgements

Work in Dr. Calin's laboratory is supported by National Institutes of Health (NIH/NCATS) Grant UH3TR00943‐01 through the NIH Common Fund, Office of Strategic Coordination (OSC), the NIH/NCI Grant 1 R01 CA182905‐01, a U54 Grant—UPR/MDACC Partnership for Excellence in Cancer Research 2016 Pilot Project, a Team DOD (CA160445P1) Grant, a Ladies Leukemia League Grant, a CLL Moonshot Flagship Project, a SINF 2017 Grant, and the Estate of C. G. Johnson, Jr. Dr. Van Roosbroeck, is supported by the Lauri Strauss Leukemia Foundation.

References

  1. Akira S (2009) Pathogen recognition by innate immunity and its signaling. Proceed Japan Acad Series B Phys Biol Sci 85, 143–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Al‐Khalaf HH and Aboussekhra A (2017) p16INK4A induces senescence and inhibits EMT through microRNA‐141/microRNA‐146b‐5p‐dependent repression of AUF1. Mol Carcinog 56, 985–999. [DOI] [PubMed] [Google Scholar]
  3. Ambros V (2004) The functions of animal microRNAs. Nature 431, 350–355. [DOI] [PubMed] [Google Scholar]
  4. Aprelikova O, Palla J, Hibler B, Yu X, Greer YE, Yi M, Stephens R, Maxwell GL, Jazaeri A, Risinger JI et al (2013) Silencing of miR‐148a in cancer‐associated fibroblasts results in WNT10B‐mediated stimulation of tumor cell motility. Oncogene 32, 3246–3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Arita S, Baba E, Shibata Y, Niiro H, Shimoda S, Isobe T, Kusaba H, Nakano S and Harada M (2008) B cell activation regulates exosomal HLA production. Eur J Immunol 38, 1423–1434. [DOI] [PubMed] [Google Scholar]
  6. Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, Li Y, Lu ES, Kwan K, Wong KK et al (2016) Exosomal transfer of stroma‐derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun 7, 11150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baer C, Squadrito ML, Laoui D, Thompson D, Hansen SK, Kiialainen A, Hoves S, Ries CH, Ooi CH and De Palma M (2016) Suppression of microRNA activity amplifies IFN‐gamma‐induced macrophage activation and promotes anti‐tumour immunity. Nat Cell Biol 18, 790–802. [DOI] [PubMed] [Google Scholar]
  8. Baroni S, Romero‐Cordoba S, Plantamura I, Dugo M, D'Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG et al (2016) Exosome‐mediated delivery of miR‐9 induces cancer‐associated fibroblast‐like properties in human breast fibroblasts. Cell Death Dis 7, e2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bartel DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297. [DOI] [PubMed] [Google Scholar]
  10. Batool M, Anwar MA and Choi S (2016) Toll‐like receptors targeting technology for the treatment of lymphoma. Expert Opin Drug Discov 11, 1047–1059. [DOI] [PubMed] [Google Scholar]
  11. Bayraktar R, Pichler M, Kanlikilicer P, Ivan C, Bayraktar E, Kahraman N, Aslan B, Oguztuzun S, Ulasli M, Arslan A et al (2017) MicroRNA 603 acts as a tumor suppressor and inhibits triple‐negative breast cancer tumorigenesis by targeting elongation factor 2 kinase. Oncotarget 8, 11641–11658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Berindan‐Neagoe I and Calin GA (2014) Molecular pathways: microRNAs, cancer cells, and microenvironment. Clin Cancer Res 20, 6247–6253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Berindan‐Neagoe I, Monroig Pdel C, Pasculli B and Calin GA (2014) MicroRNAome genome: a treasure for cancer diagnosis and therapy. CA Cancer J Clin 64, 311–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Binnemars‐Postma K, Storm G and Prakash J (2017) Nanomedicine strategies to target tumor‐associated macrophages. Int J Mol Sci 18, 979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Biswas SK and Mantovani A (2010) Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 11, 889–896. [DOI] [PubMed] [Google Scholar]
  16. Boon RA and Vickers KC (2013) Intercellular transport of microRNAs. Arterioscler Thromb Vasc Biol 33, 186–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bullrich F, Fujii H, Calin G, Mabuchi H, Negrini M, Pekarsky Y, Rassenti L, Alder H, Reed JC, Keating MJ et al (2001) Characterization of the 13q14 tumor suppressor locus in CLL: identification of ALT1, an alternative splice variant of the LEU2 gene. Can Res 61, 6640–6648. [PubMed] [Google Scholar]
  18. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M, Rai K et al (2002) 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X et al (2008) Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 18, 997–1006. [DOI] [PubMed] [Google Scholar]
  20. Chen M, Calin GA and Meng QH (2014a) Circulating microRNAs as promising tumor biomarkers. Adv Clin Chem 67, 189–214. [DOI] [PubMed] [Google Scholar]
  21. Chen W, Wang H, Chen H, Liu S, Lu H, Kong D, Huang X, Kong Q and Lu Z (2014b) Clinical significance and detection of microRNA‐21 in serum of patients with diffuse large B‐cell lymphoma in Chinese population. Eur J Haematol 92, 407–412. [DOI] [PubMed] [Google Scholar]
  22. Chen X, Ying X, Wang X, Wu X, Zhu Q and Wang X (2017) Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA‐940 to induce macrophage M2 polarization. Oncol Rep 38, 522–528. [DOI] [PubMed] [Google Scholar]
  23. Cheng G (2015) Circulating miRNAs: roles in cancer diagnosis, prognosis and therapy. Adv Drug Deliv Rev 81, 75–93. [DOI] [PubMed] [Google Scholar]
  24. Clayton A, Turkes A, Dewitt S, Steadman R, Mason MD and Hallett MB (2004) Adhesion and signaling by B cell‐derived exosomes: the role of integrins. FASEB J 18, 977–979. [DOI] [PubMed] [Google Scholar]
  25. Cocucci E and Meldolesi J (2011) Ectosomes. Curr Biol 21, R940–R941. [DOI] [PubMed] [Google Scholar]
  26. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Thery C and Raposo G (2013) Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J Cell Sci 126, 5553–5565. [DOI] [PubMed] [Google Scholar]
  27. Doldi V, Callari M, Giannoni E, D'Aiuto F, Maffezzini M, Valdagni R, Chiarugi P, Gandellini P and Zaffaroni N (2015) Integrated gene and miRNA expression analysis of prostate cancer associated fibroblasts supports a prominent role for interleukin‐6 in fibroblast activation. Oncotarget 6, 31441–31460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Donahue TR, Nguyen AH, Moughan J, Li L, Tatishchev S, Toste P and Farrell JJ (2014) Stromal microRNA‐21 levels predict response to 5‐fluorouracil in patients with pancreatic cancer. J Surg Oncol 110, 952–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Donnarumma E, Fiore D, Nappa M, Roscigno G, Adamo A, Iaboni M, Russo V, Affinito A, Puoti I, Quintavalle C et al (2017) Cancer‐associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 8, 19592–19608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Fabbri M, Garzon R, Andreeff M, Kantarjian HM, Garcia‐Manero G and Calin GA (2008) MicroRNAs and noncoding RNAs in hematological malignancies: molecular, clinical and therapeutic implications. Leukemia 22, 1095–1105. [DOI] [PubMed] [Google Scholar]
  31. Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ et al (2012) MicroRNAs bind to toll‐like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA 109, E2110–E2116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Fathullahzadeh S, Mirzaei H, Honardoost MA, Sahebkar A and Salehi M (2016) Circulating microRNA‐192 as a diagnostic biomarker in human chronic lymphocytic leukemia. Cancer Gene Ther 23, 327–332. [DOI] [PubMed] [Google Scholar]
  33. Fayyad‐Kazan H, Bitar N, Najar M, Lewalle P, Fayyad‐Kazan M, Badran R, Hamade E, Daher A, Hussein N, ElDirani R et al (2013) Circulating miR‐150 and miR‐342 in plasma are novel potential biomarkers for acute myeloid leukemia. J Transl Med 11, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Fernandez‐Mercado M, Manterola L, Larrea E, Goicoechea I, Arestin M, Armesto M, Otaegui D and Lawrie CH (2015) The circulating transcriptome as a source of non‐invasive cancer biomarkers: concepts and controversies of non‐coding and coding RNA in body fluids. J Cell Mol Med 19, 2307–2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Ferrajoli A, Shanafelt TD, Ivan C, Shimizu M, Rabe KG, Nouraee N, Ikuo M, Ghosh AK, Lerner S, Rassenti LZ et al (2013) Prognostic value of miR‐155 in individuals with monoclonal B‐cell lymphocytosis and patients with B chronic lymphocytic leukemia. Blood 122, 1891–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Filip AA, Grenda A, Popek S, Koczkodaj D, Michalak‐Wojnowska M, Budzynski M, Wasik‐Szczepanek E, Zmorzynski S, Karczmarczyk A and Giannopoulos K (2017) Expression of circulating miRNAs associated with lymphocyte differentiation and activation in CLL‐another piece in the puzzle. Ann Hematol 96, 33–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Friedl P and Alexander S (2011) Cancer invasion and the microenvironment: plasticity and reciprocity. Cell 147, 992–1009. [DOI] [PubMed] [Google Scholar]
  38. Hannafon BN, Trigoso YD, Calloway CL, Zhao YD, Lum DH, Welm AL, Zhao ZJ, Blick KE, Dooley WC and Ding WQ (2016) Plasma exosome microRNAs are indicative of breast cancer. Breast Cancer Res 18, 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. He M, Xu Z, Ding T, Kuang DM and Zheng L (2009) MicroRNA‐155 regulates inflammatory cytokine production in tumor‐associated macrophages via targeting C/EBPbeta. Cell Mol Immunol 6, 343–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, Lipford G, Wagner H and Bauer S (2004) Species‐specific recognition of single‐stranded RNA via toll‐like receptor 7 and 8. Science 303, 1526–1529. [DOI] [PubMed] [Google Scholar]
  41. Hydbring P, Wang Y, Fassl A, Li X, Matia V, Otto T, Choi YJ, Sweeney KE, Suski JM, Yin H et al (2017) Cell‐cycle‐targeting microRNAs as therapeutic tools against refractory cancers. Cancer Cell 31, 576–590 e578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Josson S, Gururajan M, Sung SY, Hu P, Shao C, Zhau HE, Liu C, Lichterman J, Duan P, Li Q et al (2015) Stromal fibroblast‐derived miR‐409 promotes epithelial‐to‐mesenchymal transition and prostate tumorigenesis. Oncogene 34, 2690–2699. [DOI] [PubMed] [Google Scholar]
  43. Kadera BE, Li L, Toste PA, Wu N, Adams C, Dawson DW and Donahue TR (2013) MicroRNA‐21 in pancreatic ductal adenocarcinoma tumor‐associated fibroblasts promotes metastasis. PLoS ONE 8, e71978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Kalluri R (2016) The biology and function of fibroblasts in cancer. Nat Rev Cancer 16, 582–598. [DOI] [PubMed] [Google Scholar]
  45. Kalluri R and Zeisberg M (2006) Fibroblasts in cancer. Nat Rev Cancer 6, 392–401. [DOI] [PubMed] [Google Scholar]
  46. Kanada M, Bachmann MH, Hardy JW, Frimannson DO, Bronsart L, Wang A, Sylvester MD, Schmidt TL, Kaspar RL, Butte MJ et al (2015) Differential fates of biomolecules delivered to target cells via extracellular vesicles. Proc Natl Acad Sci USA 112, E1433–E1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kanlikilicer P, Rashed MH, Bayraktar R, Mitra R, Ivan C, Aslan B, Zhang X, Filant J, Silva AM, Rodriguez‐Aguayo C et al (2016) Ubiquitous release of exosomal tumor suppressor miR‐6126 from ovarian cancer cells. Can Res 76, 7194–7207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Laberge RM, Awad P, Campisi J and Desprez PY (2012) Epithelial‐mesenchymal transition induced by senescent fibroblasts. Cancer Microenviron 5, 39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Larrea E, Sole C, Manterola L, Goicoechea I, Armesto M, Arestin M, Caffarel MM, Araujo AM, Araiz M, Fernandez‐Mercado M et al (2016) New concepts in cancer biomarkers: circulating miRNAs in liquid biopsies. Int J Mol Sci 17, 627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Lawrence T and Natoli G (2011) Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol 11, 750–761. [DOI] [PubMed] [Google Scholar]
  51. Lawrie CH (2013) MicroRNAs in hematological malignancies. Blood Rev 27, 143–154. [DOI] [PubMed] [Google Scholar]
  52. Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, Banham AH, Pezzella F, Boultwood J, Wainscoat JS et al (2008) Detection of elevated levels of tumour‐associated microRNAs in serum of patients with diffuse large B‐cell lymphoma. Br J Haematol 141, 672–675. [DOI] [PubMed] [Google Scholar]
  53. Lehmann SM, Kruger C, Park B, Derkow K, Rosenberger K, Baumgart J, Trimbuch T, Eom G, Hinz M, Kaul D et al (2012) An unconventional role for miRNA: let‐7 activates toll‐like receptor 7 and causes neurodegeneration. Nat Neurosci 15, 827–835. [DOI] [PubMed] [Google Scholar]
  54. Li X, Fan Q, Li J, Song J and Gu Y (2017) MiR‐124 down‐regulation is critical for cancer associated fibroblasts‐enhanced tumor growth of oral carcinoma. Exp Cell Res 351, 100–108. [DOI] [PubMed] [Google Scholar]
  55. Li Y, Zhao L, Shi B, Ma S, Xu Z, Ge Y, Liu Y, Zheng D and Shi J (2015) Functions of miR‐146a and miR‐222 in tumor‐associated macrophages in breast cancer. Sci Rep 5, 18648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Ling H, Fabbri M and Calin GA (2013) MicroRNAs and other non‐coding RNAs as targets for anticancer drug development. Nat Rev Drug Discovery 12, 847–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Liu X, Liu X, Wu Y, Wu Q, Wang Q, Yang Z and Li L (2017a) MicroRNAs in biofluids are novel tools for bladder cancer screening. Oncotarget 8, 32370–32379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Liu Y, Zhang J, Sun X, Su Q and You C (2017b) Down‐regulation of miR‐29b in carcinoma associated fibroblasts promotes cell growth and metastasis of breast cancer. Oncotarget 8, 39559–39570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, Iwasaki A and Flavell RA (2004) Recognition of single‐stranded RNA viruses by toll‐like receptor 7. Proc Natl Acad Sci USA 101, 5598–5603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Mangala LS, Wang H, Jiang D, Wu SY, Somasunderam A, Volk DE, Lokesh GL, Li X, Pradeep S, Yang X et al (2016) Improving vascular maturation using noncoding RNAs increases antitumor effect of chemotherapy. JCI Insight 1, e87754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Mantovani A, Sica A and Locati M (2007) New vistas on macrophage differentiation and activation. Eur J Immunol 37, 14–16. [DOI] [PubMed] [Google Scholar]
  62. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova‐Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A et al (2008) Circulating microRNAs as stable blood‐based markers for cancer detection. Proc Natl Acad Sci USA 105, 10513–10518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Mitra AK, Zillhardt M, Hua Y, Tiwari P, Murmann AE, Peter ME and Lengyel E (2012) MicroRNAs reprogram normal fibroblasts into cancer‐associated fibroblasts in ovarian cancer. Cancer Discov 2, 1100–1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Mosser DM and Edwards JP (2008) Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8, 958–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Munker R and Calin GA (2011) MicroRNA profiling in cancer. Clin Sci 121, 141–158. [DOI] [PubMed] [Google Scholar]
  66. Nishimura M, Jung EJ, Shah MY, Lu C, Spizzo R, Shimizu M, Han HD, Ivan C, Rossi S, Zhang X et al (2013) Therapeutic synergy between microRNA and siRNA in ovarian cancer treatment. Cancer Discov 3, 1302–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Nouraee N, Van Roosbroeck K, Vasei M, Semnani S, Samaei NM, Naghshvar F, Omidi AA, Calin GA and Mowla SJ (2013) Expression, tissue distribution and function of miR‐21 in esophageal squamous cell carcinoma. PLoS ONE 8, e73009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Noy R and Pollard JW (2014) Tumor‐associated macrophages: from mechanisms to therapy. Immunity 41, 49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Orimo A, Gupta PB, Sgroi DC, Arenzana‐Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL and Weinberg RA (2005) Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF‐1/CXCL12 secretion. Cell 121, 335–348. [DOI] [PubMed] [Google Scholar]
  70. Ortiz‐Quintero B (2016) Cell‐free microRNAs in blood and other body fluids, as cancer biomarkers. Cell Prolif 49, 281–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Pang W, Su J, Wang Y, Feng H, Dai X, Yuan Y, Chen X and Yao W (2015) Pancreatic cancer‐secreted miR‐155 implicates in the conversion from normal fibroblasts to cancer‐associated fibroblasts. Cancer Sci 106, 1362–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Raposo G and Stoorvogel W (2013) Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 200, 373–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Rashed MH, Kanlikilicer P, Rodriguez‐Aguayo C, Pichler M, Bayraktar R, Bayraktar E, Ivan C, Filant J, Silva A, Aslan B et al (2017) Exosomal miR‐940 maintains SRC‐mediated oncogenic activity in cancer cells: a possible role for exosomal disposal of tumor suppressor miRNAs. Oncotarget 8, 20145–20164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Redis RS, Calin S, Yang Y, You MJ and Calin GA (2012) Cell‐to‐cell miRNA transfer: from body homeostasis to therapy. Pharmacol Ther 136, 169–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE and Hill R (2017) Cancer‐associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 36, 1770–1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Sansone P, Berishaj M, Rajasekhar VK, Ceccarelli C, Chang Q, Strillacci A, Savini C, Shapiro L, Bowman RL, Mastroleo C et al (2017) Evolution of cancer stem‐like cells in endocrine‐resistant metastatic breast cancers is mediated by stromal microvesicles. Can Res 77, 1927–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Schoepp M, Strose AJ and Haier J (2017) Dysregulation of miRNA Expression in Cancer Associated Fibroblasts (CAFs) and its consequences on the tumor microenvironment. Cancers 9, 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Shen Z, Qin X, Yan M, Li R, Chen G, Zhang J and Chen W (2017) Cancer‐associated fibroblasts promote cancer cell growth through a miR‐7‐RASSF2‐PAR‐4 axis in the tumor microenvironment. Oncotarget 8, 1290–1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Shen H, Yu X, Yang F, Zhang Z, Shen J, Sun J, Choksi S, Jitkaew S and Shu Y (2016) Reprogramming of normal fibroblasts into cancer‐associated fibroblasts by miRNAs‐mediated CCL2/VEGFA signaling. PLoS Genet 12, e1006244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Sica A, Schioppa T, Mantovani A and Allavena P (2006) Tumour‐associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti‐cancer therapy. Eur J Cancer 42, 717–727. [DOI] [PubMed] [Google Scholar]
  81. Sonda N, Simonato F, Peranzoni E, Cali B, Bortoluzzi S, Bisognin A, Wang E, Marincola FM, Naldini L, Gentner B et al (2013) miR‐142‐3p prevents macrophage differentiation during cancer‐induced myelopoiesis. Immunity 38, 1236–1249. [DOI] [PubMed] [Google Scholar]
  82. Squadrito ML, Pucci F, Magri L, Moi D, Gilfillan GD, Ranghetti A, Casazza A, Mazzone M, Lyle R, Naldini L et al (2012) miR‐511‐3p modulates genetic programs of tumor‐associated macrophages. Cell Rep 1, 141–154. [DOI] [PubMed] [Google Scholar]
  83. Staubach S, Razawi H and Hanisch FG (2009) Proteomics of MUC1‐containing lipid rafts from plasma membranes and exosomes of human breast carcinoma cells MCF‐7. Proteomics 9, 2820–2835. [DOI] [PubMed] [Google Scholar]
  84. Steidl C, Lee T, Shah SP, Farinha P, Han G, Nayar T, Delaney A, Jones SJ, Iqbal J, Weisenburger DD et al (2010) Tumor‐associated macrophages and survival in classic Hodgkin's lymphoma. New Engl J Med 362, 875–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Tabet F, Vickers KC, Cuesta Torres LF, Wiese CB, Shoucri BM, Lambert G, Catherinet C, Prado‐Lourenco L, Levin MG, Thacker S et al (2014) HDL‐transferred microRNA‐223 regulates ICAM‐1 expression in endothelial cells. Nat Commun 5, 3292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Takeda K, Kaisho T and Akira S (2003) Toll‐like receptors. Annu Rev Immunol 21, 335–376. [DOI] [PubMed] [Google Scholar]
  87. Thery C, Ostrowski M and Segura E (2009) Membrane vesicles as conveyors of immune responses. Nat Rev Immunol 9, 581–593. [DOI] [PubMed] [Google Scholar]
  88. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, Schwille P, Brugger B and Simons M (2008) Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247. [DOI] [PubMed] [Google Scholar]
  89. Turturici G, Tinnirello R, Sconzo G and Geraci F (2014) Extracellular membrane vesicles as a mechanism of cell‐to‐cell communication: advantages and disadvantages. Am J Physiol Cell Physiol 306, C621–C633. [DOI] [PubMed] [Google Scholar]
  90. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ and Lotvall JO (2007) Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9, 654–659. [DOI] [PubMed] [Google Scholar]
  91. Van Roosbroeck K and Calin GA (2017) Cancer hallmarks and MicroRNAs: the therapeutic connection. Adv Cancer Res 135, 119–149. [DOI] [PubMed] [Google Scholar]
  92. Van Roosbroeck K, Fanini F, Setoyama T, Ivan C, Rodriguez‐Aguayo C, Vannini I, Redis RS, D'Abundo L, Zhang X, Xiao L et al (2017) Combining anti‐Mir‐155 with chemotherapy for the treatment of lung cancers. Clin Cancer Res 23, 2891–2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Van Roosbroeck K, Pollet J and Calin GA (2013) miRNAs and long noncoding RNAs as biomarkers in human diseases. Exp Rev Mol Diagnost 13, 183–204. [DOI] [PubMed] [Google Scholar]
  94. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD and Remaley AT (2011) MicroRNAs are transported in plasma and delivered to recipient cells by high‐density lipoproteins. Nat Cell Biol 13, 423–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Vickers KC and Remaley AT (2012) Lipid‐based carriers of microRNAs and intercellular communication. Curr Opin Lipidol 23, 91–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Visintin A, Mazzoni A, Spitzer JH, Wyllie DH, Dower SK and Segal DM (2001) Regulation of toll‐like receptors in human monocytes and dendritic cells. J Immunol 166, 249–255. [DOI] [PubMed] [Google Scholar]
  97. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M et al (2006) A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 103, 2257–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Wang Z, Brandt S, Medeiros A, Wang S, Wu H, Dent A and Serezani CH (2015) MicroRNA 21 is a homeostatic regulator of macrophage polarization and prevents prostaglandin E2‐mediated M2 generation. PLoS ONE 10, e0115855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ and Wang K (2010) The microRNA spectrum in 12 body fluids. Clin Chem 56, 1733–1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Xu J, Cao Z, Liu W, You L, Zhou L, Wang C, Lou W, Sun B, Miao Y, Liu X et al (2016) Plasma miRNAs effectively distinguish patients with pancreatic cancer from controls: a multicenter study. Ann Surg 263, 1173–1179. [DOI] [PubMed] [Google Scholar]
  101. Yang J, Lu Y, Lin YY, Zheng ZY, Fang JH, He S and Zhuang SM (2016) Vascular mimicry formation is promoted by paracrine TGF‐beta and SDF1 of cancer‐associated fibroblasts and inhibited by miR‐101 in hepatocellular carcinoma. Cancer Lett 383, 18–27. [DOI] [PubMed] [Google Scholar]
  102. Yang TS, Yang XH, Chen X, Wang XD, Hua J, Zhou DL, Zhou B and Song ZS (2014) MicroRNA‐106b in cancer‐associated fibroblasts from gastric cancer promotes cell migration and invasion by targeting PTEN. FEBS Lett 588, 2162–2169. [DOI] [PubMed] [Google Scholar]
  103. Yim N, Ryu SW, Choi K, Lee KR, Lee S, Choi H, Kim J, Shaker MR, Sun W, Park JH et al (2016) Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein‐protein interaction module. Nat Commun 7, 12277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Zhang WC, Chin TM, Yang H, Nga ME, Lunny DP, Lim EK, Sun LL, Pang YH, Leow YN, Malusay SR et al (2016) Tumour‐initiating cell‐specific miR‐1246 and miR‐1290 expression converge to promote non‐small cell lung cancer progression. Nat Commun 7, 11702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Zhang Z, Li X, Sun W, Yue S, Yang J, Li J, Ma B, Wang J, Yang X, Pu M et al (2017) Loss of exosomal miR‐320a from cancer‐associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Lett 397, 33–42. [DOI] [PubMed] [Google Scholar]
  106. Zhang Y, Liu D, Chen X, Li J, Li L, Bian Z, Sun F, Lu J, Yin Y, Cai X et al (2010) Secreted monocytic miR‐150 enhances targeted endothelial cell migration. Mol Cell 39, 133–144. [DOI] [PubMed] [Google Scholar]
  107. Zhang QW, Liu L, Gong CY, Shi HS, Zeng YH, Wang XZ, Zhao YW and Wei YQ (2012) Prognostic significance of tumor‐associated macrophages in solid tumor: a meta‐analysis of the literature. PLoS ONE 7, e50946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Zhang Y, Zhang D, Wang F, Xu D, Guo Y and Cui W (2015) Serum miRNAs panel (miR‐16‐2*, miR‐195, miR‐2861, miR‐497) as novel non‐invasive biomarkers for detection of cervical cancer. Sci Rep 5, 17942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Zhao JL, Huang F, He F, Gao CC, Liang SQ, Ma PF, Dong GY, Han H and Qin HY (2016) Forced activation of Notch in macrophages represses tumor growth by upregulating miR‐125a and disabling tumor‐associated macrophages. Can Res 76, 1403–1415. [DOI] [PubMed] [Google Scholar]
  110. Zheng P, Chen L, Yuan X, Luo Q, Liu Y, Xie G, Ma Y and Shen L (2017) Exosomal transfer of tumor‐associated macrophage‐derived miR‐21 confers cisplatin resistance in gastric cancer cells. J Exp Clin Cancer Res 36, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Zhong Y and Yi C (2016) MicroRNA‐720 suppresses M2 macrophage polarization by targeting GATA3. Biosci Rep 36, e00363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Zhu W, Zhou K, Zha Y, Chen D, He J, Ma H, Liu X, Le H and Zhang Y (2016) Diagnostic value of serum miR‐182, miR‐183, miR‐210, and miR‐126 levels in patients with early‐stage non‐small cell lung cancer. PLoS ONE 11, e0153046. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Oncology are provided here courtesy of Wiley

RESOURCES