Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 1.
Published in final edited form as: Cancer Metastasis Rev. 2012 Dec;31(0):653–662. doi: 10.1007/s10555-012-9368-6

MicroRNA control of epithelial–mesenchymal transition and metastasis

Jinsong Zhang 1, Li Ma 1,
PMCID: PMC3686549  NIHMSID: NIHMS481447  PMID: 22684369

Abstract

The great majority of cancer deaths are due to metastasis, which remains a poorly understood pathological process. The formation of a metastasis reflects a succession of complex steps leading to the macroscopic outgrowth of disseminated tumor cells at the secondary site. In the past 5 years, certain microRNAs (miRNAs) have been shown to regulate either a single step or multiple steps of metastasis, doing so by downregulating the expression of their target genes. In this review, we discuss recent studies on the functions and molecular mechanisms of miRNAs in regulating epithelial–mesenchymal transition (EMT) and cancer metastasis.

Keywords: MicroRNA (miRNA), Cancer, Metastasis, Epithelial, mesenchymal transition (EMT), Mesenchymal–epithelial transition (MET)

1 Introduction

Around 90% of cancer-related mortality is caused by metastasis, a multi-step process in which tumor cells disseminate from the primary site, enter lymphatic or blood circulation, arrest at distant anatomic sites, exit the vasculature, and colonize distant organs through metastatic outgrowth [1]. The current understanding of the molecular and cellular determinants of metastasis is largely limited.

miRNAs are endogenously expressed ~22 nt non-coding RNAs that negatively regulate the expression of their targeted genes [2]. Over the past decade, a growing list of miRNAs has been identified in various tissues and in different species. Recently, the miRNA database (www.mirbase.org) released the latest miRbase version (v18), which contains 18,226 entries representing hairpin precursor miRNAs, expressing 21,643 mature miRNA products, in 168 species. It is estimated that over one third of the human genome is targeted by miRNAs [3].

miRNAs bind to perfect or imperfect complementary sequences of target mRNAs at the miRNA recognition elements (MREs) of their 3′UTR through a “seed” region [3], leading to cleavage of target mRNAs and/or inhibition of their translation [4]. Interestingly, RNAs that compete with each other through common MREs, termed competing endogenous RNAs (ceRNAs), are proposed to regulate key oncogenes and tumor suppressor genes [5]. ceRNAs can be either mRNAs or RNAs produced from pseudogenes and other non-coding genes. For instance, RNAs that share miRNA binding sites with PTEN, such as its pseudogene PTENP1, positively regulate expression levels of PTEN by acting as endogenous miRNA decoys or sponges [68].

Numerous profiling and functional experiments have identified oncogenic and tumor-suppressing miRNAs, collectively termed “oncomirs” [9, 10]. Moreover, specific metastasis-regulating miRNAs, collectively termed “metastamirs” [11], have been found to be either positively or negatively associated with metastasis. In the following sections, we will review the roles of various metastamirs, as well as the functional targets and downstream pathways controlled by these non-coding RNAs.

2 miRNAs that regulate EMT/MET

Recently, it has been suggested that epithelial cancer cells may convert to motile mesenchymal ones by undergoing an epithelial–mesenchymal transition (EMT) [12, 13]. EMT is characterized by loss of cell adhesion, repression of E-cadherin expression, acquisition of mesenchymal markers (including N-cadherin, Vimentin, and Fibronectin), and increased cell motility and invasiveness [12]. Both EMT and mesenchymal–epithelial transition (MET), the reversion of EMT, are essential for developmental processes, including mesoderm formation, neural crest development, heart valve development, and secondary palate formation [14]. On the basis of recent studies, it is proposed that EMT can be resurrected by primary tumor cells in order to acquire motility and invasiveness, and that MET is important for the final stage of metastasis in which extravasated cancer cells revert to an epithelial state and proliferate into a macroscopic secondary tumor [14].

Interestingly, a seminal study by the Weinberg Lab demonstrated that induction of the EMT program can generate cells with properties of stem cells or cancer stem cells (CSCs) [15]—defined operationally as tumor-initiating cells [16]. Hence, the invasion step of the metastasis cascade may involve a cell-type conversion process which endows epithelial cancer cells with both motility and self-renewal ability, whereas the metastatic colonization step may reflect differentiation of CSCs into non-CSCs at the metastatic site. These steps have been shown to be orchestrated by pleiotropically acting molecules—transcription factors and miRNAs.

Several transcription factors, including Snail [17], Slug [18], Twist [19], ZEB1 [20], and ZEB2 [21], have been identified as inducers of EMT and tumor metastasis. More recently, miR-205 and the miR-200 family (miR-200a, miR-200b, miR-200c, miR-141, and miR-429, which share a consensus seed sequence) have emerged as new epithelial markers and repressors of EMT [22, 23] and stem cell properties [24]. The miR-200 family members function to promote MET and inhibit induction of EMT, doing so by directly targeting the mRNAs encoding ZEB1 and ZEB2 [22, 23]. Conversely, ZEB1 represses the transcription of miR-200 genes by directly binding to their promoter region, thereby forming a double-negative feedback loop [22, 25, 26]. Expression of the miR-200 family is lost in regions of metaplastic breast cancers lacking E-cadherin, whereas ZEB1 and ZEB2 are highly abundant in invasive mesenchymal cells [22].

On the other hand, the fact that the miR-200 family promotes the conversion of mesenchymal cells to epithelial-like cells suggests that these miRNAs may favor metastatic outgrowth. Expression of the miR-200 family members in the highly metastatic 4T1 mouse mammary tumor cell line is higher than that in its isogenic cell line, 4TO7, which is capable of forming micrometastases but not macrometastases [27, 28]. When ectopically expressed in 4TO7 cells, miR-200 promoted MET and enabled formation of macroscopic metastases in the lung and liver after these cells were injected intravenously into recipient mice [27, 28]. This effect was mediated, at least in part, by direct targeting of Sec23a, a COPII vesicle component that modulates the secretion of metastasis-suppressing proteins, such as Igfbp4 and Tinagl1 [28]. Collectively, these findings suggest a model in which the miR-200 miRNA family suppresses EMT and cancer cell dissemination, but promotes metastatic colonization after tumor cells have already disseminated to distant organs (Fig. 1).

Fig. 1.

Fig. 1

Schematic diagram of miR-200’s regulation of EMT/MET and metastasis. The miR-200 family members target ZEB1/ZEB2 and Sec23a, and play opposing roles at early and late steps of metastasis

Besides the miR-200 family, additional miRNAs have also been reported to regulate EMT (Table 1). miR-9, a MYC/MYCN-induced miRNA, directly targets the E-cadherin-encoding mRNA CDH1, leading to increased cell motility/invasiveness and a context-dependent EMT-like conversion [29]. Overexpression of miR-9 in otherwise non-metastatic epithelial breast tumor cells induced micro-metastasis formation in the lungs of recipient mice, whereas silencing of miR-9 in highly malignant cells inhibited metastasis [29]. The miR-103/107 family attenuates miRNA biosynthesis by targeting Dicer [30]. miR-103/107 can induce EMT by downregulating miR-200 levels, and empower metastatic dissemination of otherwise non-aggressive breast cancer cells in vivo [30]. The miR-221/222 miRNA cluster has been found to target ESR1 (estrogen receptor) [31], Dicer [32], and TRPS1 (trichorhinophalangeal syndrome type 1) [33], leading to EMT induction in breast cancer cells [33]. Treatment of the NMuMG mammary epithelial cells with TGF-β markedly induced miR-155, whose knockdown suppressed TGF-β-induced EMT, migration, and invasion, and this regulation has been attributed to the ability of this miRNA to target RHOA [34]. In non-small cell lung cancer cells, miR-30a inhibits EMT by directly targeting Snail, a transcription repressor of CDH1 [35]. In retinal pigment epithelium, miR-204 plays a critical role in maintaining epithelial barrier function and cell physiology by directly targeting TGFβR2 and SNAIL2 [36]. Taken together, cancer cells may exploit these miRNAs to acquire cellular plasticity and accomplish different steps of the metastatic process.

Table 1.

miRNAs involved in EMT/MET

miRNA Effect on EMT Target Reference
miR-9 Promote CDH1 [29]
miR-15b Suppress BMI1 [92]
miR-27 Promote APC [93]
miR-29a Promote TTP [94]
miR-30a Suppress Snail [35]
miR-103/107 Promote DICER1 [30]
miR-155 Promote RHOA [34]
miR-194 Suppress BMI1 [95]
miR-200 family Suppress ZEB1/ZEB2, Sec23a [23, 25, 28, 96, 97]
miR-205 Suppress ZEB1/ZEB2 [22]
miR-204 Suppress TGFβR2, SNAIL2 [36]
miR-221/222 Promote TRPS1, ESR1, DICER1 [3133]
miR-661 Promote StarD10, Nectin-1 [98]

3 Other miRNAs involved in metastasis

The link between miRNAs and metastasis was first provided by a study which reported that overexpression of miR-10b in otherwise non-metastatic breast tumors triggered tumor invasion and distant metastasis in xenotransplanation models [37]. Since then, a growing body of evidence has demonstrated the existence of pro-metastatic and anti-metastatic miRNAs (Table 2) (Fig. 2).

Table 2.

Additional miRNAs (besides those regulating EMT/MET) with functional roles in tumor invasion and metastasis

miRNA Role in invasion/metastasis Cancer type Target Reference
let-7 family Suppress Hepatocellular, colorectal, gastric, breast MYC, BCL2L1, RAS, HMGA2, MMP11, PBX3, COL1A2, MYH9, RAB40C [73, 99104]
miR-7 Suppress Glioblastoma, breast Pak1, EGFR [105, 106]
miR-10a Promote Pancreatic HOXB1, HOXB3 [107]
miR-10b Promote Breast, nasopharyngeal, esophageal, glioblastoma HOXD10, KLF4, LMP1, BCL2L11/Bim, TFAP2C/AP-2, CDKN1A/p21, CDKN2A/p16 [37, 4043, 87, 108]
miR-16 Suppress Prostate CDK1, CDK2 [109]
miR-17–92 Promote Breast, colorectal CTGF, Tsp1 [110, 111]
miR-21 Promote Breast, colorectal, gastric, lung, pancreatic, prostate, bladder, ovarian, hepatocellular PDCD4, PTEN, CDC25A, RHOB, TIAM1, TPM1, MARCKS, NF1B, SPRY2 [4655]
miR-22 Suppress Breast CDK6, SIRT1, SP1 [112]
miR-31 Suppress Breast RHOA, RDX, ITGA5 [60, 6668]
miR-122 Suppress Hepatocellular ADAM17, RHOA, RAC1 [113, 114]
miR-126 Suppress Breast, lung CRK, VEGF [61, 63, 64]
miR-146a/b Suppress Breast, pancreatic, glioma, prostate, gastric EGFR, ROCK1 IRAK1, NFKB1 [115120]
miR-194 Suppress Liver CDH2, DNMT3A, HBEGF [121]
miR-206 Suppress Breast, rhabdomyosarcoma ESR1, MET [122, 123]
miR-214 Promote Melanoma TFAP2C [124]
miR-335 Suppress Gastric, breast BCL2L2, SP1, SOX4, TNC [61, 125, 126]
miR-373 Promote Breast, testicular germ cell CD44, LATS2 [56, 57]
miR-378 Promote Breast, glioblastoma Sufu, Fus-1 [127]
miR-520c Promote Breast CD44 [57]

Fig. 2.

Fig. 2

miRNAs that regulate metastasis. Metastasis consists of multiple steps: epithelial–mesenchymal transition (EMT), local invasion, intravasation, extravasation, and colonization (as indicated by green boxes). miRNAs and their target genes are indicated in red/blue boxes and pink circles, respectively. Red box: metastasis-promoting miRNAs; blue box: metastasis-suppressing miRNAs

Twist, a potent inducer of EMT, can bind to the promoter region of mir-10b and activate its transcription [37]. The miR-10b miRNA directly targets the mRNA encoding HOXD10, a transcriptional repressor of several genes involved in cell migration and extracellular matrix (ECM) remodeling, including RHOC, α3 integrin, uPAR, and MT1-MMP (MMP-14) [37, 38]. In breast cancer cells, HOXD10 is also targeted by a metastasis-promoting, long non-coding RNA, HOTAIR [39]. Moreover, HOXD10, RHOC, uPAR, and MMP-14 are functional effectors of miR-10b in glioblastoma cells and mediate the effect of this miRNA on promoting invasiveness of such tumor cells [40, 41]. In human esophageal cancer cells, miR-10b promotes migration and invasion by targeting KLF4 [42]. Other targets of miR-10b include BCL2L11/Bim, TFAP2C/AP-2, CDKN1A/p21, and CDKN2A/p16 in glioblastoma [43].

Just like certain oncoproteins (e.g., HER2/ERBB2) which not only initiate tumor formation but also confer invasiveness and metastatic ability on cancer cells, several miRNAs, initially identified as oncomirs, have been found to promote migration, invasion, and metastasis. miR-21 is one of the best established oncomir that is overexpressed in most types of cancer analyzed [44]. In the Tet-Off miR-21 transgenic mice, 16-fold overexpression of miR-21 led to development of pre-B-cell lymphoma, which was reversed within a few days of doxycycline treatment, demonstrating that miR-21 is a bona fide oncogenic miRNA and that miR-21-driven tumors are addicted to this oncomir [45]. miR-21 targets a number of tumor suppressors, including PDCD4, PTEN, TPM1, and RHOB [4655], some of which have established inhibitory effects on cancer cell detachment, migration, and invasion steps of the metastatic cascade (Fig. 2). Consistent with this, miR-21 was found to promote invasion, intravasation, and metastasis in breast cancer and colon cancer [47, 49]. Another example is miR-373, which was initially identified in a forward genetic screen as an oncogenic miRNA acting to target the tumor suppressor LATS2 in testicular germ-cell tumors [56]. Later, miR-373 stood out again in a functional genomics screen as a miRNA that promoted cell migration. This miRNA also induced metastasis of otherwise non-metastatic MCF-7 breast cancer cells in vivo, which was mediated by targeting of CD44. In breast cancer patients, lymph node metastases exhibited upregulation of miR-373 compared with paired primary tumors [57].

Additional pro-metastatic miRNAs identified in a number of cancer types are listed in Table 2. Although each tumor type has its distinct miRNA signature, some of these miRNAs are deregulated in multiple types of cancer. For instance, miR-21 is aberrantly overexpressed in breast, colorectal, prostate, ovarian, and oral cancer, and it appears to target a group of tumor suppressor and metastasis suppressor genes involved in multiple tumor types [44]. miR-10b, which was initially shown to be highly expressed in breast tumors from metastasis-positive patients [37], has also been found to be upregulated in highly aggressive glioma [40, 41, 43] and pancreatic adenocarcinoma [58, 59]. These results suggest that some miRNAs may have a widespread role in tumor invasion and/or metastasis across different tumor types.

Besides the pro-metastatic roles played by miRNAs, a number of miRNAs act as anti-metastatic regulators. miR-335, miR-206, miR-126, and miR-31 are among the first metastasis-suppressing miRNAs discovered through expression profiling analysis of metastatic and non-metastatic cell lines [60, 61]. Mechanistically, miR-335 targets SOX4, TNC, and PTPRN2 to remodel the ECM of cancer cells and to inhibit cell migration [61], while miR-206 activates cancer cell apoptosis and inhibits cell migration through modulating expression of NOTCH3 and SRC1/3 [62]. In cancer cells, miR-126 targets the pro-angiogenic factor VEGF [63] and CRK [64], an adaptor protein involved in cell proliferation, adhesion, and migration. Interestingly, the action of miR-126 goes beyond cell-autonomous effects on tumor cells, as this miRNA also inhibits endothelial recruitment, angiogenesis, and colonization at the meta-static site by targeting IGFBP2, PITPNC1, and MERTK [65]. miR-31 is a multi-functional miRNA that regulates several steps of metastasis, including local invasion, anoikis, extravasation, and metastatic colonization; these effects can be explained by concomitant suppression of three pro-metastatic gene products, RHOA, radixin, and integrin α5 (Fig. 2) [60, 6668].

Expression of the let-7 miRNA family members is down-regulated in a variety of cancers and in stem cells [69, 70]. These miRNAs were initially found to inhibit tumor formation by downregulating oncogenic proteins, RAS and HMGA2 (Fig. 2) [71, 72]. Subsequently, it was reported that restoring let-7 expression in breast CSCs suppressed proliferative potential and mammosphere-forming ability in vitro, as well as tumorigenicity and metastatic ability in vivo [73]. These results are in consonance with recent findings that CSCs are responsible for the development of metastatic lesions [74, 75], and suggest that therapeutic strategies centered on restoration of let-7 miRNAs may not only shrink the primary tumor but also block dissemination of metastatic CSCs.

4 Implications of miRNAs in cancer diagnosis, prognosis, and therapeutics

Studies on miRNAs not only illuminate the molecular basis of metastasis but also have implications for diagnosis, prognosis, and treatment of cancer. Expression of 217 mammalian miRNAs and 16,000 mRNAs were profiled simultaneously in 334 normal tissues and cancer specimens. A number of miRNAs showed upregulation or downregulation in tumors, and the expression pattern of these miRNAs classified cancer types better than that of mRNAs [76]. Recently, it has been reported that cancer-associated miRNAs can be detected in serum or plasma of patients, and may effectively discriminate tumor-bearing individuals from healthy controls, which suggests the potential of using specific circulating miRNAs as non-invasive or minimally invasive cancer biomarkers [77, 78]. For instance, serum levels of miR-141 can distinguish between healthy individuals and patients with prostate cancer [77]. In colorectal cancer patients, the levels of miR-92a and miR-29a are significantly elevated in their plasma [79, 80]. These studies open new avenues for cancer detection and follow-up examination.

miRNAs that correlate with clinical outcomes provide promise for improved prognosis. In breast cancer patients, tumors with low expression of miR-335 and miR-126 have a higher probability of developing metastasis at distant sites compared with tumors expressing high levels of these two miRNAs [61]. miR-210, a hypoxia-induced miRNA, is an independent prognostic marker in breast cancer, and its expression levels show an inverse correlation with disease-free and overall survival outcomes [81, 82]. miR-21, the most studied oncomir, has been identified as an indicator of poor prognosis in multiple cancer types, including breast cancer [48], squamous cell carcinoma [83], astrocytoma [84], and gastric cancer [85].

miRNAs that are critical for tumor formation, maintenance, and progression might serve as targets for therapeutic intervention. Intensive efforts have been made to develop miRNA-based therapeutic strategies. For example, in an orthotopic model of pancreatic cancer, systemically delivered miR-34 or miR-143/145 mimics inhibited tumor growth without detectable toxicity [86], although the effect on metastasis remains unclear. On the other hand, miRNAs that promote tumor formation and/or metastasis can be targeted in vivo. For instance, therapeutic silencing of miR-10b using “antagomirs”—chemically modified, cholesterol-conjugated antisense miRNA inhibitors, resulted in sequence-specific inhibition of metastasis in a mouse mammary tumor model [87]. An attractive property of miRNAs is their ability to downregulate target genes in one or more pathways or networks at multiple levels [88]. Thus, targeting a single miRNA is expected to influence multiple miRNA target genes and their associated signaling pathways.

miRNA-based agents have been shown to modulate sensitivity to traditional cancer drugs. Treatment with miR-21 inhibitors can sensitize breast cancer cells to Topotecan and Taxol [89]. Expression of miR-205 in SKBR3 breast cancer cells directly targets HER3, leading to increased responsiveness to tyrosine kinase inhibitors Gefitinib and Lapatinib [90]. Although these results are based on in vitro experiments and need to be validated in vivo, they suggest that manipulating miRNA expression may improve cancer management, and that combination treatment with miRNA-based agents and other therapeutic drugs could be beneficial.

5 Concluding remarks

Combining expression profiling, functional characterization, mechanistic experiments, and clinical validation, studies in the past five years have identified a number of miRNAs that play diverse roles at multiple steps of tumor progression and metastasis. Although it is not fully understood how these miRNAs are deregulated in cancer, several mechanisms have been investigated. In particular, epigenetic silencing of cancer-implicated miRNAs by CpG island hypermethylation has been found in human tumor cells [91]. The miRNA–mRNA interactions are complex: each miRNA can potentially target hundreds of mRNAs, and each mRNA can be targeted by several distinct miRNAs. Thus, besides individual miRNA analysis, a comprehensive and systematic strategy will be necessary for future studies to elucidate the network of miRNAs and underlying pathways. Furthermore, genetically engineered animal models will shed new light on metastasis regulation by miRNAs. A survey of published miRNA knockout mouse models has revealed that many miRNAs are dispensable for normal development (likely due to functional redundancy under physiological conditions), but play essential roles under pathological conditions, such as cancer and cardiac disorder. Hence, generation and analysis of compound mouse mutants are expected to advance our understanding of the roles and mechanisms of miRNAs in metastatic progression, and to provide insight into clinical applications of miRNAs.

Acknowledgments

The miRNA research in the Ma Lab is supported by an NIH Pathway to Independence (K99/R00) Award CA138572, a CPRIT First-Time, Tenure-Track Faculty Award R1004, a University of Texas STARS Award, and a Faculty Development Award from MD Anderson’s Cancer Center Support Grant CA016672 from NIH.

References

  • 1.Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nature Reviews Cancer. 2003;3(6):453–458. doi: 10.1038/nrc1098. [DOI] [PubMed] [Google Scholar]
  • 2.Cai Y, Yu X, Hu S, Yu J. A brief review on the mechanisms of miRNA regulation. Genomics, Proteomics & Bioinformatics. 2009;7(4):147–154. doi: 10.1016/S1672-0229(08)60044-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20. doi: 10.1016/j.cell.2004.12.035. [DOI] [PubMed] [Google Scholar]
  • 4.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
  • 5.Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell. 2011;146(3):353–358. doi: 10.1016/j.cell.2011.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 2010;465(7301):1033–1038. doi: 10.1038/nature09144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Karreth FA, Tay Y, Perna D, Ala U, Tan SM, Rust AG, et al. In vivo identification of tumor-suppressive PTEN ceRNAs in an oncogenic BRAF-induced mouse model of melanoma. Cell. 2011;147(2):382–395. doi: 10.1016/j.cell.2011.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Tay Y, Kats L, Salmena L, Weiss D, Tan SM, Ala U, et al. Coding-independent regulation of the tumor suppressor PTEN by competing endogenous mRNAs. Cell. 2011;147(2):344–357. doi: 10.1016/j.cell.2011.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Calin GA, Croce CM. MicroRNA signatures in human cancers. Nature Reviews Cancer. 2006;6(11):857–866. doi: 10.1038/nrc1997. [DOI] [PubMed] [Google Scholar]
  • 10.Esquela-Kerscher A, Slack FJ. Oncomirs—microRNAs with a role in cancer. Nature Reviews Cancer. 2006;6(4):259–269. doi: 10.1038/nrc1840. [DOI] [PubMed] [Google Scholar]
  • 11.Hurst DR, Edmonds MD, Welch DR. Metastamir: the field of metastasis-regulatory microRNA is spreading. Cancer Research. 2009;69(19):7495–7498. doi: 10.1158/0008-5472.CAN-09-2111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kalluri R, Weinberg RA. The basics of epithelial–mesenchymal transition. The Journal of Clinical Investigation. 2009;119(6):1420–1428. doi: 10.1172/JCI39104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nature Reviews Cancer. 2002;2(6):442–454. doi: 10.1038/nrc822. [DOI] [PubMed] [Google Scholar]
  • 14.Yang J, Weinberg RA. Epithelial–mesenchymal transition: at the crossroads of development and tumor metastasis. Developmental Cell. 2008;14(6):818–829. doi: 10.1016/j.devcel.2008.05.009. [DOI] [PubMed] [Google Scholar]
  • 15.Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial–mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gupta PB, Chaffer CL, Weinberg RA. Cancer stem cells: mirage or reality? Nature Medicine. 2009;15(9):1010–1012. doi: 10.1038/nm0909-1010. [DOI] [PubMed] [Google Scholar]
  • 17.Cano A, Perez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del Barrio MG, et al. The transcription factor snail controls epithelial–mesenchymal transitions by repressing E-cadherin expression. Nature Cell Biology. 2000;2(2):76–83. doi: 10.1038/35000025. [DOI] [PubMed] [Google Scholar]
  • 18.Hajra KM, Chen DY, Fearon ER. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Research. 2002;62(6):1613–1618. [PubMed] [Google Scholar]
  • 19.Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117(7):927–939. doi: 10.1016/j.cell.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 20.Eger A, Aigner K, Sonderegger S, Dampier B, Oehler S, Schreiber M, et al. DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene. 2005;24(14):2375–2385. doi: 10.1038/sj.onc.1208429. [DOI] [PubMed] [Google Scholar]
  • 21.Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, et al. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Molecular Cell. 2001;7(6):1267–1278. doi: 10.1016/s1097-2765(01)00260-x. [DOI] [PubMed] [Google Scholar]
  • 22.Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nature Cell Biology. 2008;10(5):593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 23.Park SM, Gaur AB, Lengyel E, Peter ME. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes & Development. 2008;22(7):894–907. doi: 10.1101/gad.1640608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Shimono Y, Zabala M, Cho RW, Lobo N, Dalerba P, Qian D, et al. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell. 2009;138(3):592–603. doi: 10.1016/j.cell.2009.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gregory PA, Bracken CP, Smith E, Bert AG, Wright JA, Roslan S, et al. An autocrine TGF-beta/ZEB/miR-200 signaling network regulates establishment and maintenance of epithelial–mesenchymal transition. Molecular Biology of the Cell. 2011;22(10):1686–1698. doi: 10.1091/mbc.E11-02-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, et al. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Reports. 2008;9(6):582–589. doi: 10.1038/embor.2008.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dykxhoorn DM, Wu Y, Xie H, Yu F, Lal A, Petrocca F, et al. miR-200 enhances mouse breast cancer cell colonization to form distant metastases. PLoS One. 2009;4(9):e7181. doi: 10.1371/journal.pone.0007181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Korpal M, Ell BJ, Buffa FM, Ibrahim T, Blanco MA, Celia-Terrassa T, et al. Direct targeting of Sec23a by miR-200s influences cancer cell secretome and promotes metastatic colonization. Nature Medicine. 2011;17(9):1101–1108. doi: 10.1038/nm.2401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, et al. miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nature Cell Biology. 2010;12(3):247–256. doi: 10.1038/ncb2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Martello G, Rosato A, Ferrari F, Manfrin A, Cordenonsi M, Dupont S, et al. A MicroRNA targeting dicer for metastasis control. Cell. 2010;141(7):1195–1207. doi: 10.1016/j.cell.2010.05.017. [DOI] [PubMed] [Google Scholar]
  • 31.Di Leva G, Gasparini P, Piovan C, Ngankeu A, Garofalo M, Taccioli C, et al. MicroRNA cluster 221–222 and estrogen receptor alpha interactions in breast cancer. Journal of the National Cancer Institute. 2010;102(10):706–721. doi: 10.1093/jnci/djq102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cochrane DR, Cittelly DM, Howe EN, Spoelstra NS, McKinsey EL, LaPara K, et al. MicroRNAs link estrogen receptor alpha status and Dicer levels in breast cancer. Hormones and Cancer. 2011;1(6):306–319. doi: 10.1007/s12672-010-0043-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stinson S, Lackner MR, Adai AT, Yu N, Kim HJ, O’Brien C, et al. miR-221/222 targeting of trichorhinophalangeal 1 (TRPS1) promotes epithelial-to-mesenchymal transition in breast cancer. Science Signaling. 2011;4(186):pt5. doi: 10.1126/scisignal.2002258. [DOI] [PubMed] [Google Scholar]
  • 34.Kong W, Yang H, He L, Zhao JJ, Coppola D, Dalton WS, et al. MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Molecular and Cellular Biology. 2008;28(22):6773–6784. doi: 10.1128/MCB.00941-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kumarswamy R, Mudduluru G, Ceppi P, Muppala S, Kozlowski M, Niklinski J, et al. MicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer. International Journal of Cancer. 2012;130(9):2044–2053. doi: 10.1002/ijc.26218. [DOI] [PubMed] [Google Scholar]
  • 36.Wang FE, Zhang C, Maminishkis A, Dong L, Zhi C, Li R, et al. MicroRNA-204/211 alters epithelial physiology. The FASEB Journal. 2010;24(5):1552–1571. doi: 10.1096/fj.08-125856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ma L, Teruya-Feldstein J, Weinberg RA. Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007;449(7163):682–688. doi: 10.1038/nature06174. [DOI] [PubMed] [Google Scholar]
  • 38.Carrio M, Arderiu G, Myers C, Boudreau NJ. Homeobox D10 induces phenotypic reversion of breast tumor cells in a three-dimensional culture model. Cancer Research. 2005;65(16):7177–7185. doi: 10.1158/0008-5472.CAN-04-1717. [DOI] [PubMed] [Google Scholar]
  • 39.Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464(7291):1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sasayama T, Nishihara M, Kondoh T, Hosoda K, Kohmura E. MicroRNA-10b is overexpressed in malignant glioma and associated with tumor invasive factors, uPAR and RhoC. International Journal of Cancer. 2009;125(6):1407–1413. doi: 10.1002/ijc.24522. [DOI] [PubMed] [Google Scholar]
  • 41.Sun L, Yan W, Wang Y, Sun G, Luo H, Zhang J, et al. MicroRNA-10b induces glioma cell invasion by modulating MMP-14 and uPAR expression via HOXD10. Brain Research. 2011;1389:9–18. doi: 10.1016/j.brainres.2011.03.013. [DOI] [PubMed] [Google Scholar]
  • 42.Tian Y, Luo A, Cai Y, Su Q, Ding F, Chen H, et al. MicroRNA-10b promotes migration and invasion through KLF4 in human esophageal cancer cell lines. Journal of Biological Chemistry. 2010;285(11):7986–7994. doi: 10.1074/jbc.M109.062877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gabriely G, Yi M, Narayan RS, Niers JM, Wurdinger T, Imitola J, et al. Human glioma growth is controlled by microRNA-10b. Cancer Research. 2011;71(10):3563–3572. doi: 10.1158/0008-5472.CAN-10-3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Pan X, Wang ZX, Wang R. MicroRNA-21: a novel therapeutic target in human cancer. Cancer Biology & Therapy. 2011;10(12):1224–1232. doi: 10.4161/cbt.10.12.14252. [DOI] [PubMed] [Google Scholar]
  • 45.Medina PP, Nolde M, Slack FJ. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature. 2010;467(7311):86–90. doi: 10.1038/nature09284. [DOI] [PubMed] [Google Scholar]
  • 46.Wu WY, Xue XY, Chen ZJ, Han SL, Huang YP, Zhang LF, et al. Potentially predictive microRNAs of gastric cancer with metastasis to lymph node. World Journal of Gastroenterology. 2011;17(31):3645–3651. doi: 10.3748/wjg.v17.i31.3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S, et al. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27(15):2128–2136. doi: 10.1038/sj.onc.1210856. [DOI] [PubMed] [Google Scholar]
  • 48.Yan LX, Huang XF, Shao Q, Huang MY, Deng L, Wu QL, et al. MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA. 2008;14(11):2348–2360. doi: 10.1261/rna.1034808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo YY. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Research. 2008;18(3):350–359. doi: 10.1038/cr.2008.24. [DOI] [PubMed] [Google Scholar]
  • 50.Huang TH, Wu F, Loeb GB, Hsu R, Heidersbach A, Brincat A, et al. Up-regulation of miR-21 by HER2/neu signaling promotes cell invasion. Journal of Biological Chemistry. 2009;284(27):18515–18524. doi: 10.1074/jbc.M109.006676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Li T, Li D, Sha J, Sun P, Huang Y. MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochemical and Biophysical Research Communications. 2009;383(3):280–285. doi: 10.1016/j.bbrc.2009.03.077. [DOI] [PubMed] [Google Scholar]
  • 52.Wang P, Zou F, Zhang X, Li H, Dulak A, Tomko RJ, Jr, et al. microRNA-21 negatively regulates Cdc25A and cell cycle progression in colon cancer cells. Cancer Research. 2009;69(20):8157–8165. doi: 10.1158/0008-5472.CAN-09-1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Connolly EC, Van Doorslaer K, Rogler LE, Rogler CE. Overexpression of miR-21 promotes an in vitro metastatic phenotype by targeting the tumor suppressor RHOB. Molecular Cancer Research. 2010;8(5):691–700. doi: 10.1158/1541-7786.MCR-09-0465. [DOI] [PubMed] [Google Scholar]
  • 54.Cottonham CL, Kaneko S, Xu L. miR-21 and miR-31 converge on TIAM1 to regulate migration and invasion of colon carcinoma cells. Journal of Biological Chemistry. 2010;285(46):35293–35302. doi: 10.1074/jbc.M110.160069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lou Y, Yang X, Wang F, Cui Z, Huang Y. MicroRNA-21 promotes the cell proliferation, invasion and migration abilities in ovarian epithelial carcinomas through inhibiting the expression of PTEN protein. International Journal of Molecular Medicine. 2010;26(6):819–827. doi: 10.3892/ijmm_00000530. [DOI] [PubMed] [Google Scholar]
  • 56.Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, Nagel R, et al. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell. 2006;124(6):1169–1181. doi: 10.1016/j.cell.2006.02.037. [DOI] [PubMed] [Google Scholar]
  • 57.Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, et al. The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nature Cell Biology. 2008;10(2):202–210. doi: 10.1038/ncb1681. [DOI] [PubMed] [Google Scholar]
  • 58.Preis M, Gardner TB, Gordon SR, Pipas JM, Mackenzie TA, Klein EE, et al. MicroRNA-10b expression correlates with response to neoadjuvant therapy and survival in pancreatic ductal adenocarcinoma. Clinical Cancer Research. 2011;17(17):5812–5821. doi: 10.1158/1078-0432.CCR-11-0695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Nakata K, Ohuchida K, Mizumoto K, Kayashima T, Ikenaga N, Sakai H, et al. MicroRNA-10b is overex-pressed in pancreatic cancer, promotes its invasiveness, and correlates with a poor prognosis. Surgery. 2011;150(5):916–922. doi: 10.1016/j.surg.2011.06.017. [DOI] [PubMed] [Google Scholar]
  • 60.Valastyan S, Reinhardt F, Benaich N, Calogrias D, Szasz AM, Wang ZC, et al. A pleiotropically acting microRNA, miR-31, inhibits breast cancer metastasis. Cell. 2009;137(6):1032–1046. doi: 10.1016/j.cell.2009.03.047. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 61.Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, et al. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451(7175):147–152. doi: 10.1038/nature06487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Song G, Zhang Y, Wang L. MicroRNA-206 targets notch3, activates apoptosis, and inhibits tumor cell migration and focus formation. Journal of Biological Chemistry. 2009;284(46):31921–31927. doi: 10.1074/jbc.M109.046862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu B, Peng XC, Zheng XL, Wang J, Qin YW. MiR-126 restoration down-regulate VEGF and inhibit the growth of lung cancer cell lines in vitro and in vivo. Lung Cancer. 2009;66(2):169–175. doi: 10.1016/j.lungcan.2009.01.010. [DOI] [PubMed] [Google Scholar]
  • 64.Crawford M, Brawner E, Batte K, Yu L, Hunter MG, Otterson GA, et al. MicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines. Biochemical and Biophysical Research Communications. 2008;373(4):607–612. doi: 10.1016/j.bbrc.2008.06.090. [DOI] [PubMed] [Google Scholar]
  • 65.Png KJ, Halberg N, Yoshida M, Tavazoie SF. A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells. Nature. 2011;481(7380):190–194. doi: 10.1038/nature10661. [DOI] [PubMed] [Google Scholar]
  • 66.Valastyan S, Chang A, Benaich N, Reinhardt F, Weinberg RA. Concurrent suppression of integrin alpha5, radixin, and RhoA phenocopies the effects of miR-31 on metastasis. Cancer Research. 2010;70(12):5147–5154. doi: 10.1158/0008-5472.CAN-10-0410. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 67.Valastyan S, Chang A, Benaich N, Reinhardt F, Weinberg RA. Activation of miR-31 function in already-established metastases elicits metastatic regression. Genes & Development. 2011;25(6):646–659. doi: 10.1101/gad.2004211. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 68.Valastyan S, Benaich N, Chang A, Reinhardt F, Weinberg RA. Concomitant suppression of three target genes can explain the impact of a microRNA on metastasis. Genes & Development. 2009;23(22):2592–2597. doi: 10.1101/gad.1832709. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 69.Bussing I, Slack FJ, Grosshans H. let-7 microRNAs in development, stem cells and cancer. Trends in Molecular Medicine. 2008;14(9):400–409. doi: 10.1016/j.molmed.2008.07.001. [DOI] [PubMed] [Google Scholar]
  • 70.Boyerinas B, Park SM, Hau A, Murmann AE, Peter ME. The role of let-7 in cell differentiation and cancer. Endocrine-Related Cancer. 2010;17(1):F19–F36. doi: 10.1677/ERC-09-0184. [DOI] [PubMed] [Google Scholar]
  • 71.Mayr C, Hemann MT, Bartel DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science. 2007;315(5818):1576–1579. doi: 10.1126/science.1137999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, et al. RAS is regulated by the let-7 microRNA family. Cell. 2005;120(5):635–647. doi: 10.1016/j.cell.2005.01.014. [DOI] [PubMed] [Google Scholar]
  • 73.Yu F, Yao H, Zhu P, Zhang X, Pan Q, Gong C, et al. let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007;131(6):1109–1123. doi: 10.1016/j.cell.2007.10.054. [DOI] [PubMed] [Google Scholar]
  • 74.Liu H, Patel MR, Prescher JA, Patsialou A, Qian D, Lin J, et al. Cancer stem cells from human breast tumors are involved in spontaneous metastases in orthotopic mouse models. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(42):18115–18120. doi: 10.1073/pnas.1006732107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Malanchi I, Santamaria-Martinez A, Susanto E, Peng H, Lehr HA, Delaloye JF, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2011;481(7379):85–89. doi: 10.1038/nature10694. [DOI] [PubMed] [Google Scholar]
  • 76.Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature. 2005;435(7043):834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
  • 77.Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(30):10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of bio-markers for diagnosis of cancer and other diseases. Cell Research. 2008;18(10):997–1006. doi: 10.1038/cr.2008.282. [DOI] [PubMed] [Google Scholar]
  • 79.Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, et al. Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential marker for colorectal cancer screening. Gut. 2009;58(10):1375–1381. doi: 10.1136/gut.2008.167817. [DOI] [PubMed] [Google Scholar]
  • 80.Huang Z, Huang D, Ni S, Peng Z, Sheng W, Du X. Plasma microRNAs are promising novel biomarkers for early detection of colorectal cancer. International Journal of Cancer. 2009;127(1):118–126. doi: 10.1002/ijc.25007. [DOI] [PubMed] [Google Scholar]
  • 81.Camps C, Buffa FM, Colella S, Moore J, Sotiriou C, Sheldon H, et al. hsa-miR-210 Is induced by hypoxia and is an independent prognostic factor in breast cancer. Clinical Cancer Research. 2008;14(5):1340–1348. doi: 10.1158/1078-0432.CCR-07-1755. [DOI] [PubMed] [Google Scholar]
  • 82.Volinia S, Galasso M, Sana ME, Wise TF, Palatini J, Huebner K, et al. Breast cancer signatures for invasiveness and prognosis defined by deep sequencing of microRNA. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(8):3024–3029. doi: 10.1073/pnas.1200010109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Li J, Huang H, Sun L, Yang M, Pan C, Chen W, et al. MiR-21 indicates poor prognosis in tongue squamous cell carcinomas as an apoptosis inhibitor. Clinical Cancer Research. 2009;15(12):3998–4008. doi: 10.1158/1078-0432.CCR-08-3053. [DOI] [PubMed] [Google Scholar]
  • 84.Zhi F, Chen X, Wang S, Xia X, Shi Y, Guan W, et al. The use of hsa-miR-21, hsa-miR-181b and hsa-miR-106a as prognostic indicators of astrocytoma. European Journal of Cancer. 2010;46(9):1640–1649. doi: 10.1016/j.ejca.2010.02.003. [DOI] [PubMed] [Google Scholar]
  • 85.Jiang J, Zheng X, Xu X, Zhou Q, Yan H, Zhang X, et al. Prognostic significance of miR-181b and miR-21 in gastric cancer patients treated with S-1/Oxaliplatin or Doxifluridine/Oxaliplatin. PLoS One. 2011;6(8):e23271. doi: 10.1371/journal.pone.0023271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pramanik D, Campbell NR, Karikari C, Chivukula R, Kent OA, Mendell JT, et al. Restitution of tumor suppressor microRNAs using a systemic nanovector inhibits pancreatic cancer growth in mice. Molecular Cancer Therapeutics. 2011;10(8):1470– 1480. doi: 10.1158/1535-7163.MCT-11-0152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ma L, Reinhardt F, Pan E, Soutschek J, Bhat B, Marcusson EG, et al. Therapeutic silencing of miR- 10b inhibits metastasis in a mouse mammary tumor model. Nature Biotechnology. 2010;28(4):341–347. doi: 10.1038/nbt.1618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Makeyev EV, Maniatis T. Multilevel regulation of gene expression by microRNAs. Science. 2008;319(5871):1789–1790. doi: 10.1126/science.1152326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mei M, Ren Y, Zhou X, Yuan XB, Han L, Wang GX, et al. Downregulation of miR-21 enhances chemotherapeutic effect of taxol in breast carcinoma cells. Technology in Cancer Research & Treatment. 2010;9(1):77–86. doi: 10.1177/153303461000900109. [DOI] [PubMed] [Google Scholar]
  • 90.Iorio MV, Casalini P, Piovan C, Di Leva G, Merlo A, Triulzi T, et al. microRNA-205 regulates HER3 in human breast cancer. Cancer Research. 2009;69(6):2195–2200. doi: 10.1158/0008-5472.CAN-08-2920. [DOI] [PubMed] [Google Scholar]
  • 91.Lujambio A, Calin GA, Villanueva A, Ropero S, Sanchez-Cespedes M, Blanco D, et al. A microRNA DNA methylation signature for human cancer metastasis. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(36):13556–13561. doi: 10.1073/pnas.0803055105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Sun L, Yao Y, Liu B, Lin Z, Lin L, Yang M, et al. MiR-200b and miR-15b regulate chemotherapy-induced epithelial–mesenchymal transition in human tongue cancer cells by targeting BMI1. Oncogene. 2011;31(4):432–445. doi: 10.1038/onc.2011.263. [DOI] [PubMed] [Google Scholar]
  • 93.Zhang Z, Liu S, Shi R, Zhao G. miR-27 promotes human gastric cancer cell metastasis by inducing epithelial-to-mesenchymal transition. Cancer Genetics. 2011;204(9):486–491. doi: 10.1016/j.cancergen.2011.07.004. [DOI] [PubMed] [Google Scholar]
  • 94.Gebeshuber CA, Zatloukal K, Martinez J. miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Reports. 2009;10(4):400–405. doi: 10.1038/embor.2009.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Dong P, Kaneuchi M, Watari H, Hamada J, Sudo S, Ju J, et al. MicroRNA-194 inhibits epithelial to mesenchymal transition of endometrial cancer cells by targeting oncogene BMI-1. Molecular Cancer. 2011;10:99. doi: 10.1186/1476-4598-10-99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial–mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. Journal of Biological Chemistry. 2008;283(22):14910–14914. doi: 10.1074/jbc.C800074200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kim T, Veronese A, Pichiorri F, Lee TJ, Jeon YJ, Volinia S, et al. p53 regulates epithelial–mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. The Journal of Experimental Medicine. 2011;208(5):875–883. doi: 10.1084/jem.20110235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Vetter G, Saumet A, Moes M, Vallar L, Le Bechec A, Laurini C, et al. miR-661 expression in SNAI1-induced epithelial to mesenchymal transition contributes to breast cancer cell invasion by targeting Nectin-1 and StarD10 messengers. Oncogene. 2010;29(31):4436–4448. doi: 10.1038/onc.2010.181. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 99.Han HB, Gu J, Zuo HJ, Chen ZG, Zhao W, Li M, et al. Let-7c functions as a metastasis suppressor by targeting MMP11 and PBX3 in colorectal cancer. The Journal of Pathology. 2012;226(3):544–555. doi: 10.1002/path.3014. [DOI] [PubMed] [Google Scholar]
  • 100.Ji J, Zhao L, Budhu A, Forgues M, Jia HL, Qin LX, et al. Let-7g targets collagen type I alpha2 and inhibits cell migration in hepatocellular carcinoma. Journal of Hepatology. 2010;52(5):690–697. doi: 10.1016/j.jhep.2009.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Qian P, Zuo Z, Wu Z, Meng X, Li G, Zhang W, et al. Pivotal role of reduced let-7g expression in breast cancer invasion and metastasis. Cancer Research. 2011;71(20):6463–6474. doi: 10.1158/0008-5472.CAN-11-1322. [DOI] [PubMed] [Google Scholar]
  • 102.Yang Q, Jie Z, Cao H, Greenlee AR, Yang C, Zou F, et al. Low-level expression of let-7a in gastric cancer and its involvement in tumorigenesis by targeting RAB40C. Carcinogenesis. 2011;32(5):713–722. doi: 10.1093/carcin/bgr035. [DOI] [PubMed] [Google Scholar]
  • 103.Chang TC, Zeitels LR, Hwang HW, Chivukula RR, Wentzel EA, Dews M, et al. Lin-28B transactivation is necessary for Myc-mediated let-7 repression and proliferation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(9):3384–3389. doi: 10.1073/pnas.0808300106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Dangi-Garimella S, Yun J, Eves EM, Newman M, Erkeland SJ, Hammond SM, et al. Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7. EMBO Journal. 2009;28(4):347–358. doi: 10.1038/emboj.2008.294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kefas B, Godlewski J, Comeau L, Li Y, Abounader R, Hawkinson M, et al. microRNA-7 inhibits the epidermal growth factor receptor and the Akt pathway and is down-regulated in glioblastoma. Cancer Research. 2008;68(10):3566–3572. doi: 10.1158/0008-5472.CAN-07-6639. [DOI] [PubMed] [Google Scholar]
  • 106.Reddy SD, Ohshiro K, Rayala SK, Kumar R. MicroRNA-7, a homeobox D10 target, inhibits p21-activated kinase 1 and regulates its functions. Cancer Research. 2008;68(20):8195–8200. doi: 10.1158/0008-5472.CAN-08-2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Weiss FU, Marques IJ, Woltering JM, Vlecken DH, Aghdassi A, Partecke LI, et al. Retinoic acid receptor antagonists inhibit miR-10a expression and block metastatic behavior of pancreatic cancer. Gastroenterology. 2009;137(6):2136–2145. e2131–2137. doi: 10.1053/j.gastro.2009.08.065. [DOI] [PubMed] [Google Scholar]
  • 108.Li G, Wu Z, Peng Y, Liu X, Lu J, Wang L, et al. MicroRNA-10b induced by Epstein–Barr virus-encoded latent membrane protein-1 promotes the metastasis of human nasopharyngeal carcinoma cells. Cancer Letters. 2010;299(1):29–36. doi: 10.1016/j.canlet.2010.07.021. [DOI] [PubMed] [Google Scholar]
  • 109.Takeshita F, Patrawala L, Osaki M, Takahashi RU, Yamamoto Y, Kosaka N, et al. Systemic delivery of synthetic microRNA-16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell-cycle genes. Molecular Therapy. 2010;18(1):181–187. doi: 10.1038/mt.2009.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dews M, Homayouni A, Yu D, Murphy D, Sevignani C, Wentzel E, et al. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nature Genetics. 2006;38(9):1060–1065. doi: 10.1038/ng1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liu S, Goldstein RH, Scepansky EM, Rosenblatt M. Inhibition of rho-associated kinase signaling prevents breast cancer metastasis to human bone. Cancer Research. 2009;69(22):8742–8751. doi: 10.1158/0008-5472.CAN-09-1541. [DOI] [PubMed] [Google Scholar]
  • 112.Xu D, Takeshita F, Hino Y, Fukunaga S, Kudo Y, Tamaki A, et al. miR-22 represses cancer progression by inducing cellular senescence. The Journal of Cell Biology. 2011;193(2):409–424. doi: 10.1083/jcb.201010100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Coulouarn C, Factor VM, Andersen JB, Durkin ME, Thorgeirsson SS. Loss of miR-122 expression in liver cancer correlates with suppression of the hepatic phenotype and gain of metastatic properties. Oncogene. 2009;28(40):3526–3536. doi: 10.1038/onc.2009.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Tsai WC, Hsu PW, Lai TC, Chau GY, Lin CW, Chen CM, et al. MicroRNA-122, a tumor suppressor microRNA that regulates intrahepatic metastasis of hepatocellular carcinoma. Hepatology. 2009;49(5):1571–1582. doi: 10.1002/hep.22806. [DOI] [PubMed] [Google Scholar]
  • 115.Li Y, Vandenboom TG, 2nd, Wang Z, Kong D, Ali S, Philip PA, et al. miR-146a suppresses invasion of pancreatic cancer cells. Cancer Research. 2010;70(4):1486–1495. doi: 10.1158/0008-5472.CAN-09-2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lin SL, Chiang A, Chang D, Ying SY. Loss of mir-146a function in hormone-refractory prostate cancer. RNA. 2008;14(3):417–424. doi: 10.1261/rna.874808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Xia H, Qi Y, Ng SS, Chen X, Li D, Chen S, et al. microRNA-146b inhibits glioma cell migration and invasion by targeting MMPs. Brain Research. 2009;1269:158–165. doi: 10.1016/j.brainres.2009.02.037. [DOI] [PubMed] [Google Scholar]
  • 118.Kogo R, Mimori K, Tanaka F, Komune S, Mori M. Clinical significance of miR-146a in gastric cancer cases. Clinical Cancer Research. 2011;17(13):4277–4284. doi: 10.1158/1078-0432.CCR-10-2866. [DOI] [PubMed] [Google Scholar]
  • 119.Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, Benz CC. Expression of microRNA-146 suppresses NF-kappaB activity with reduction of metastatic potential in breast cancer cells. Oncogene. 2008;27(42):5643–5647. doi: 10.1038/onc.2008.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Edmonds MD, Hurst DR, Vaidya KS, Stafford LJ, Chen D, Welch DR. Breast cancer metastasis suppressor 1 coordinately regulates metastasis-associated microRNA expression. International Journal of Cancer. 2009;125(8):1778–1785. doi: 10.1002/ijc.24616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Meng Z, Fu X, Chen X, Zeng S, Tian Y, Jove R, et al. miR-194 is a marker of hepatic epithelial cells and suppresses metastasis of liver cancer cells in mice. Hepatology. 2010;52(6):2148–2157. doi: 10.1002/hep.23915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kondo N, Toyama T, Sugiura H, Fujii Y, Yamashita H. miR-206 Expression is down-regulated in estrogen receptor alpha-positive human breast cancer. Cancer Research. 2008;68(13):5004–5008. doi: 10.1158/0008-5472.CAN-08-0180. [DOI] [PubMed] [Google Scholar]
  • 123.Yan D, da Dong XE, Chen X, Wang L, Lu C, Wang J, et al. MicroRNA-1/206 targets c-Met and inhibits rhabdomyosarcoma development. Journal of Biological Chemistry. 2009;284(43):29596–29604. doi: 10.1074/jbc.M109.020511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Penna E, Orso F, Cimino D, Tenaglia E, Lembo A, Quaglino E, et al. microRNA-214 contributes to melanoma tumour progression through suppression of TFAP2C. EMBO Journal. 2011;30(10):1990–2007. doi: 10.1038/emboj.2011.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Xu Y, Zhao F, Wang Z, Song Y, Luo Y, Zhang X, et al. MicroRNA-335 acts as a metastasis suppressor in gastric cancer by targeting Bcl-w and specificity protein 1. Oncogene. 2012;31(11):1398–1407. doi: 10.1038/onc.2011.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Png KJ, Yoshida M, Zhang XH, Shu W, Lee H, Rimner A, et al. MicroRNA-335 inhibits tumor reinitiation and is silenced through genetic and epigenetic mechanisms in human breast cancer. Genes & Development. 2011;25(3):226–231. doi: 10.1101/gad.1974211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lee DY, Deng Z, Wang CH, Yang BB. MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(51):20350–20355. doi: 10.1073/pnas.0706901104. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES