Long non-coding RNAs as novel biomarkers for breast cancer invasion and metastasis (Review)

  • Authors:
    • Kaijiong Zhang
    • Zhenglian Luo
    • Yi Zhang
    • Xiaoyu Song
    • Li Zhang
    • Lichun Wu
    • Jinbo Liu
  • View Affiliations

  • Published online on: June 22, 2017     https://doi.org/10.3892/ol.2017.6462
  • Pages: 1895-1904
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer (BC) is now the most common malignancy worldwide, with high prevalence and lethality among women. Invasion and metastasis are the major reasons for breast cancer‑associated mortality. However, the underlying mechanism of invasion and metastasis has not been entirely elucidated. Long non‑coding RNAs (lncRNAs) are a large class of non‑coding transcripts that are >200 bases in length and cannot encode proteins. Evidence has indicated that lncRNAs regulate gene expression at the levels of epigenetic modification, transcription and post‑transcription. In addition, they are involved in diverse tumor biological processes, including cell proliferation, apoptosis, invasion, metastasis and angiogenesis. The present review focuses on the recent progress of lncRNAs in breast cancer invasion and metastasis, aiming to provide novel strategies for the clinical prevention, diagnosis and treatment of breast cancer.

Introduction

Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer-associated mortality among women worldwide, with an estimated 1.7 million cases and 521,900 mortalities in 2012, which accounts for 25% of all cancer cases and 15% of all cancer-associated mortalities among females (1). With the development of society and the economy, the incidence of breast cancer is increasing rapidly, particularly in the less developed countries of the world (1). Although advances have been made in the treatment of breast cancer over the previous decades, the prognosis for patients remains poor due to the high rate of metastasis (1,2). Metastasis is the primary cause of relapse and mortality of patients with breast cancer (35). Once metastasis occurs, the 5-year survival rate is <25% (6). Therefore, there is an urgent requirement to identify novel molecular biomarkers of metastasis, which may be useful in the early diagnosis, prognosis assessment and prediction of response to therapy for breast cancer.

Metastasis is a multifactorial and multistep process, which is regulated by the abnormal expression of various genes, including snail family zinc finger (Snail1 and Snail2), zinc finger E-box binding homeobox (ZEB1 and ZEB2), twist basic helix-loop-helix transcription factor 1 (Twist1) and matrix metalloproteinases (MMPs) (7). In previous years, although numerous studies (79) have focused on the identification and characterization of the genes and gene products that drive a metastatic process, the precise molecular mechanism that governs the process has been largely unknown. The identification of non-coding RNAs (ncRNAs) has opened a novel avenue for studies on cancer metastasis. It has been reported that only ~2% of the human genome encodes protein-coding genes, and at least 70% of the human genome is actively transcribed, generating a vast range of ncRNAs (10,11). Although these ncRNAs were previously regarded as transcriptional ‘noise’, it has become clear that ncRNAs perform a crucial role in cellular development and tumor progression, including metastasis (12,13). Based on transcript size, ncRNAs are classified into two major groups. Transcripts shorter than 200 nucleotides (nts) are referred to as small ncRNAs, such as microRNAs (miRNAs/miRs) (14), which have been well established to extensively affect cancer invasion and metastasis (15,16). The other group is composed of long non-coding RNAs (lncRNAs), which are pervasively transcribed in the genome and exhibit emerging roles in tumorigenesis (1720).

lncRNAs are a group of RNA molecules, which are >200 nts in length, lack functional open reading frames (ORFs) and cannot encode proteins; they regulate gene expression at the epigenetic, transcriptional and post-transcriptional levels (21,22). The majority of lncRNAs are located in the cytoplasm (2325), and are transcribed by RNA polymerase II, spliced and polyadenylated (20,26). lncRNAs can be classified into five general categories according to their location relative to nearby protein-coding genes: Sense lncRNA, antisense lncRNA, bidirectional lncRNA, intronic lncRNA and intergenic lncRNA (27,28). Although thousands of lncRNAs have been identified to be involved in numerous physiological and pathological processes, including cellular metabolism, differentiation, proliferation and apoptosis (2931), the precise mechanisms by which lncRNAs function remain poorly understood. An ever-growing amount of lncRNAs identified have essential roles in regulating oncogenic processes, such as metastasis, and their mechanisms require additional investigation to be fully understood.

Mechanisms of invasion and metastasis in breast cancer

Metastatic breast cancer frequently represents an incurable impasse and a poor prognosis for patients. Subsequent to receiving diagnosis, the mechanisms involved in metastasis are largely unknown (32). Metastasis is a complex succession of cell-biological events, collectively termed the invasion-metastasis cascade, which involves multiple factors and steps: Cancer cells break away from primary tumors, invade locally through surrounding extracellular matrix and stromal cell layers, intravasate into the circulation of blood or lymphatic vessels, survive within the vasculature, extravasate into distant tissues, and reinitiate proliferation and growth programs of tumors at metastatic sites (7). These complex cell-biological processes are coordinated by the expression of various genes and the regulation of signaling cascades, which confer the ability of incipient metastatic tumor cells to successfully generate high-grade, life-threatening malignancies (7,33).

Epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition (MET), are essential for cancer metastasis and distal colonization (34,35). EMT is typically believed to be the first step of the invasion-metastasis cascade, and the significance of EMT for breast cancer invasion and metastasis has been shown in a large number of in vitro and in vivo studies (3638). The most critical and well-studied EMT-associated molecule is the epithelial adhesion molecule, E-cadherin (39,40). In EMT, the expression of E-cadherin is decreased, while the expression of mesenchymal markers such as N-cadherin, vimentin or fibronectin is increased, and this switch promotes the capacity of cancer cell invasion and mobility (41,42). A decrease in E-cadherin expression is considered to be a fundamental event of EMT, and is prominently associated with breast cancer invasion and metastasis (4345). The decrease in E-cadherin leads to inactivation of glycogen synthase kinase 3β through phosphorylation, thus stabilizing the released β-catenin, which results in the induction of wide-ranging transcriptional and functional changes, thereby promoting breast cancer cell metastatic dissemination (46). Notably, the expression of E-cadherin will be re-established in cells that come from metastatic tumor colonies (47). Once cancer cells disseminate to a secondary organ, the process of MET may occur, and the levels of E-cadherin will be increased in these cells compared with the cells from the primary tumor (48). Furthermore, Snail1, Snail2, ZEB1, ZEB2 and Twist1 have all been shown to be important master factors of EMT (49,50). A typical example illustrated that Snail1 represses expression of epithelial genes by binding to E-box DNA sequences in the proximal promoter region of E-cadherin, which recruits the polycomb repressive complex 2 (PRC2), resulting in methylation and acetylation at histone H3 lysine (H3K)4, H3K9 and H3K27, and thereby silencing the expression of E-cadherin and initiating EMT-relevant processes to promote cancer metastasis (5155).

Several signaling pathways have also been reported to be involved in EMT and metastasis, including the transforming growth factor (TGF)-β, Wnt/β-catenin, Hedgehog, phosphoinositide 3-kinase (PI3K)/AKT and nuclear factor (NF)-κB pathways (5660). These pathways contribute to the activation of downstream transcriptional targets and ultimately initiate the program of cell adhesion, migration and metastasis. In addition to transcription factors of EMT, the invasion-metastasis cascade is also regulated by post-transcriptional regulatory networks, including lncRNAs and miRNAs (61,62). A previous study demonstrated that miRNAs exert their effects at multiple steps in the metastatic cascade by effecting cancer cell adherence, migration, invasion, motility and angiogenesis (63). However, the molecular mechanisms driving breast cancer invasion and metastasis have been scant, as lncRNAs have only recently been identified. The extent to which lncRNAs specifically facilitate metastasis remains unknown. This review focuses on individual lncRNAs involved in breast cancer metastasis, and investigates the mechanisms and functions of several known lncRNAs that have only recently been identified. This may provide valuable targets for potential clinical applications of breast cancer in early diagnosis and management.

Roles of invasion and metastasis-associated lncRNAs in breast cancer

HOX antisense intergenic RNA (HOTAIR)

HOTAIR, a 2158-nt ncRNA located on chromosome 12q13.13, was identified in 2007 (64). HOTAIR expression is increased in primary and metastatic breast cancer, and its expression level in primary tumors is a powerful predictor of eventual metastasis and mortality (65). Overexpression of HOTAIR in breast cancer cells induced genome-wide retargeting of PRC2, leading to trimethylation of H3K27 (H3K27me3), gene expression, and increased cancer invasiveness and metastasis (65). Enhancer of zeste homolog 2 (EZH2), a subunit of PRC2, is highly expressed in breast cancer, and the levels of EZH2 have been found to be strongly associated with breast cancer invasion and metastasis (66). HOTAIR and EZH2 expression levels are highly associated; co-expression of HOTAIR and EZH2 are associated with a worse clinical outcome (67).

HOTAIR serves as a key regulator in controlling the multiple signaling mechanisms involved in EMT and stemness establishment; it promotes the repression of E-cadherin by PRC2, and silencing the expression of HOTAIR can prevent EMT induction and cancer stem cells arising (68). A subsequent study revealed that HOTAIR functions as a competing endogenous RNA to regulate EMT and the invasive program by competing for miR-7; the decrease in miR-7 promotes EMT of breast cancer stem cells by activating the signal transducer and activator of transcription 3 (STAT3) pathway, and that inhibits the expression of HOTAIR, which may serve as a good strategy for treating highly invasive breast cancer (69). In metastatic breast cancer, HOTAIR also inhibits miR-568 to maintain the expression of nuclear factor of activated T cells 5, which promotes EMT and metastasis of breast cancer cells by transcriptionally activating calcium-binding protein S100A4 and vascular endothelial growth factor C (70).

HOTAIR is also significantly associated with estrogen receptor (ER) (71,72). The promoter of HOTAIR contains multiple functional estrogen response elements, and is transcriptionally induced by estradiol (E2) (73). ER and various ER coregulators, including histone methylases such as mixed-lineage leukemia (MLL)1 and MLL3, and CREB-binding protein/p300, bind to the promoter of HOTAIR in an E2-dependent manner (73,74). Knockdown of ER and MLLs can decrease E2-induced HOTAIR expression (73). HOTAIR overexpression may serve as an independent biomarker for the prediction of the risk of metastasis in patients with ER-positive breast cancer (72). Recently, Tao et al (75) demonstrated that estrogen promoted HOTAIR through G protein-coupled estrogen receptor 1-mediated miR-148a inhibition, and that estrogen-induced breast cancer cell migration was reversed by silencing HOTAIR in triple-negative breast cancer.

H19

H19 was the first lncRNA identified in 1991 (76); it is derived from a large imprinted locus on human chromosome 11p15.5, and performs a crucial role in genomic imprinting during growth and development (77). H19 is transcribed by RNA polymerase II, spliced and polyadenylated in the cytosol (78), and is largely or exclusively expressed from the maternal allele (79). Although H19 has been demonstrated to be involved in diverse biological processes of multiple numbers of tumors, there remains considerable debate about its roles. It has been reported that H19 serves as a tumor suppressor in Wilm's tumors, embryonic rhabdomyosarcoma and Beckwith-Wiedemann syndrome (80), while it functions as an oncogene in pancreatic (81), bladder (82), colorectal (83) and gastric (84) cancer. In breast cancer, H19 is proposed to function as an oncogene to potentiate the properties of tumorigenesis (85). H19 promotes breast cancer cell proliferation and invasion by hepatocyte growth factor and E2F transcription factor 1 (86,87). Matouk et al (88) indicated that H19 was highly expressed in all common metastatic sites regardless of tumor primary origin, and that its expression level was significantly associated with the metastatic potential of breast cancer cells. The overexpression of H19 abolished E-cadherin expression and increased N-cadherin and Slug expression through a mechanism that involves miR-675. Slug could also serve as a positive feedback loop to upregulate H19 expression and activate its promoter (88). In addition, TGF-β-induced EMT and H19/miR-675 expression level is associated with the PI3K/AKT pathway (89). Recently, Vennin et al (90) demonstrated that H19 could decrease ubiquitin ligase E3 family (c-Cbl and Cbl-b) expression through its miR-675 directly binding c-Cbl and Cbl-b, which increases the stability and the activation of epidermal growth factor receptor and c-Met, leading to sustained activation of their downstream AKT and ERK signaling pathways, thereby enhancing breast cancer cell proliferation, migration, tumor growth and metastasis.

Metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1)

MALAT-1, also termed nuclear-enriched abundant transcript 2, is a highly conserved lncRNA that is ~8,000 nts in length, which was identified in non-small cell lung cancer as a prognostic marker of distant metastasis (91). A number of studies have found that MALAT-1 is extensively dysregulated in several types of human malignant tumors, including lung cancer (91), pancreatic cancer (92), hepatocellular carcinoma (93), cervical cancer (94), colorectal cancer (95), bladder cancer (96) and breast cancer (97). MALAT-1 enhances the potential transcription of EMT and regulates tumor cell proliferation, migration, invasion and metastasis (98,99). MALAT-1 was markedly increased in breast cancer cells compared with in non-cancerous cell lines (97). High concentration E2 treatment causes transcriptional inhibition of MALAT-1 RNA, thus inhibiting the proliferation, migration and invasion of breast cancer cells (97). However, by constrast, Xu et al (100) revealed that MALAT-1 expression was decreased in breast cancer cell lines and tumor tissue. Lower expression of MALAT-1 was associated with axillary lymph node metastasis and clinical disease progression. knockdown of MALAT1 in breast cancer cell lines induced an EMT program by activating the PI3K/AKT pathway, thereby promoting breast cancer cell migration and invasion (100).

Long stress-induced non-coding transcript 5 (LSINCT5)

LSINCT5 is a 2.6-kb transcript that is polyadenylated and transcribed on the negative strand. LSINCT5 is localized in the nucleus and potentially transcribed by RNA polymerase III (101). Studies by Silva et al (101,102) demonstrated that LSINCT5 is greatly overexpressed in breast cancer cell lines and tumor tissues, and its expression is associated with cellular proliferation. In addition, LSINCT5 is also involved in breast cancer invasion and metastasis. Knocking down the expression of LSINCT5 leads to decreased expression of CXC chemokine receptor 4 (CXCR4), a crucial breast cancer marker associated with invasion and metastasis (103). The decrease in CXCR4 will cause a decrease in MMP-9 expression and reversion of EMT, thereby enhancing apoptosis and reducing the incidence of tumor invasion and metastasis (103105).

Long intergenic non-protein coding RNA-regulator of reprogramming (lincRNA-ROR)

lincRNA-ROR is a large intergenic non-coding RNA ~2,600 nts in length that modulates reprogramming (106). lincRNA-ROR was first discovered in induced pluripotent stem cells (iPSCs), and it was reported to serve a vital role in promoting survival in iPSCs and embryonic stem cells, possibly by preventing the activation of cellular stress pathways, including the p53 response (106). lincRNA-ROR suppresses p53 translation through direct interaction with the heterogeneous nuclear ribonucleoprotein (hnRNP) I, and inhibits p53-mediated cell cycle arrest and apoptosis (107). lincRNA-ROR expression is controlled by a key cytoprotective transcription factor, nuclear factor-erythroid 2-related factor (NRF2), in mammary stem cells. NRF2 directly binds the ROR promoter at NRF2 response elements and subsequently inhibits lincRNA-ROR transcriptional activity (108). In addition to transcriptional silencing, knockdown of NRF2 leads to a significant decrease in EZH2 binding and in the repressive marker H3K27me3, which is associated with increased lincRNA-ROR expression, resulting in mammary stem cell self-renewal (108).

lincRNA-ROR is also associated with miRNAs and may function as a competing endogenous RNA for specific miRNAs to interfere in their function. Wang et al (109) demonstrated that lincRNA-ROR is enriched in the cytoplasm and functions as a molecular sponge for miRNA-145 to upregulate its target genes, including octamer-binding transcription factor 4, sex-determining region Y-box (Sox)2, Kruppel-like factor 4 and Nanog expression. These genes are crucial for stem cell characteristics and the EMT program (110112). Eades et al (113) identified that lincRNA-ROR is markedly upregulated in triple-negative breast cancer [ER(−), human epidermal growth factor receptor 2(−) and progesterone receptor(−)] and in metastatic disease, and competitive inhibition of miR-145 by lincRNA-ROR results in expression of small GTPase ADP-ribosylation factor 6 (ARF6). ARF6 has an essential role in breast cancer cell invasion (114). The overexpression of ARF6 regulates E-cadherin localization and affects cell-cell adhesion, and then performs an important role in the metastatic process (114116). Additionally, lincRNA-ROR also competitively binds miRNA-205 to prevent the degradation of miRNA-205 target genes, such as ZEB1 and ZEB2 (116). lincRNA-ROR, as a critical positive regulator of EMT, potentiates breast cancer cell migration and invasion, and promotes the generation of stem cell properties (117). Silencing of lincRNA-ROR will repress breast cancer tumorigenesis and metastasis (116,117).

Translational regulatory lncRNA (treRNA)

treRNA (originally termed ncRNA-a7) was identified through genome-wide computational analysis in 2010 and was revealed to function as an enhancer and regulate Snail transcription (118). treRNA consists of two exons and contains three putative ORFs, and its transcript is spliced and polyadenylated. treRNA employs different molecular mechanisms of gene regulation depending on its cellular location. In the nucleus, treRNA acts as an enhancer for neighboring genes such as Snail (118). In the cytoplasm (~75%), treRNA functions to downregulate the expression of E-cadherin by suppressing the translation of its mRNA. treRNA reduces the level of E-cadherin protein without affecting the level of its mRNA, but alters the polysome distribution of E-cadherin mRNA. In addition, treRNA promotes the formation of a novel ribonucleoprotein complex with RNA-binding proteins (such as hnRNP K, FMR autosomal homolog (FXR)1, FXR2, poly(U) binding splicing factor 60 and splicing factor 3b subunit 3). This treRNP complex interacts directly or indirectly with the 3′UTR of E-cadherin mRNA, and reduces the translation efficiency (119,120).

In breast cancer samples, treRNA is significantly upregulated in paired primary and lymph-node metastasis compared with healthy controls. Enforced expression of treRNA significantly promotes breast cancer cell migration and invasion in vitro and tumor lung metastases in vivo, whereas knockdown of treRNA markedly suppresses tumor cellular migration, invasion and metastasis (119). Furthermore, enhanced expression of treRNA could decrease the expression of E-cadherin, zonulaoccludens-1 and β-catenin, and increase the expression of fibronectin and vimentin, thus affecting the process of EMT, invasion and metastasis (119). Investigation of the functions of treRNA and RNA-binding proteins may serve as potential diagnostic and prognostic markers, as well as providing therapeutic targets for breast cancer (119).

Brain cytoplasmic RNA 200 (BC200)

BC200 is a 200-nt cytoplasmic non-coding RNA that is selectively expressed in the primate nervous system, where it is transcribed by RNA polymerase III (121123). BC200 is not generally expressed in non-neural organs such as the colon, heart, kidney, liver, spleen or skeletal muscle (122). However, the neuron-specific expression is lost in various human tumors, including breast cancer (123). A previous study revealed that this non-coding RNA is more highly expressed in invasive compared with normal breast tissues or in benign tumors of breast cancer, and its expression in breast cancer cells may serve as a molecular indicator of invasive or potentially invasive malignancy (124). Furthermore, BC200 expression is strongly associated with tumor grade, and it may be used as a prognostic indicator to assess tumor progression (124,125). Therefore, the expression of BC200 may serve as a molecular marker in the diagnosis or prognosis of breast cancer (124,125).

lncRNA-activated by TGF-β (lncRNA-ATB)

lncRNA-ATB was first identificated in human hepatocellular carcinoma (HCC) cells following TGF-β treatment in 2014 (126). The lncRNA is poly(A)-negative, mainly localized in the cytoplasm and has three highly homologous loci on chromosomes 13, 14 and 22 (126,127). High expression of lncRNA-ATB is associated with tumor metastases and a poor prognosis (128,129). Evidence has indicated that lncRNA-ATB promotes HCC cell invasion by competitively binding the miR-200 family, upregulating ZEB1 and ZEB2 mRNA and protein levels, and then inducing EMT and invasion (126). By contrast, depletion of lncRNA-ATB is sufficient to abolish TGF-β-induced EMT, even though TGF-β is known to strongly induce a number of other EMT drivers, including Snail, Slug and Twist (127). In addition, lncRNA-ATB promotes organ colonization of disseminated tumor cells by binding interleukin-11 (IL-11) mRNA, increasing IL-11 mRNA stability, causing autocrine induction of IL-11 and triggering STAT3 signaling (126). IL-11 is a TGF-β target gene, the expression of which has been shown to promote bone metastasis of breast cancer (130). lncRNA-ATB has been found to upregulate and induce EMT not only in HCC, but also in colorectal (128) and breast (131) cancer. Shi et al (131) revealed that lncRNA-ATB is markedly upregulated in breast cancer cells and tissues. lncRNA-ATB may promote trastuzumab resistance and the invasion-metastasis cascade by competitively binding miR-200c and upregulating ZEB1 and zinc finger protein 217, thereby inducing EMT (131). Furthermore, the high level of lncRNA-ATB is associated with the trastuzumab resistance of patients with breast cancer (131). Therefore, lncRNA-ATB may serve as a promising biomarker and therapeutic target for breast cancer invasion and metastasis.

Breast cancer anti-estrogen resistance 4 (BCAR4)

BCAR4 was identified by functional genetic screening in anti-estrogen resistance human breast cancer cells (132). It was reported to be associated with tumor aggressiveness and tamoxifen resistance (132,133). High BCAR4 mRNA levels may serve as an independent predictive factor for poor survival following tamoxifen therapy in breast cancer (133). In addition, elevated BCAR4 expression is associated with ER, PR and ERBB2/3 signaling. Knockdown of ERBB2 or ERBB3 could inhibit cell proliferation (133,134). Recently, novel evidence demonstrated that BCAR4 presents close relevance with breast cancer metastasis and regulates a non-canonical Hedgehog/GLI2 transcriptional program (135,136). BCAR4 provides a driving force in the process of breast cancer progression and metastasis, which is markedly upregulated in breast cancer tissues and cells, and elevated expression of BCAR4 is associated with advanced lymph node metastasis stage and poor survival for patients with breast cancer (135). BCAR4 binds to the domain of unknown function of smad nuclear interacting protein 1 (SNIP1) and releases the inhibitory effect of SNIP1 on the histone acetyltransferase activity of p300, leading to the activation of p300 (135). The activated p300 enhances the acetylation of H3K18 on the promoters of GLI2 target transcription units, which releases the inhibitory roles of serine/threonine-protein phosphatase 1 regulatory subunit 10 (PPP1R10, also known as PNUTS) on protein phosphatase 1 enzymatic activity, which consequently modulates RNA Pol II Ser5 phosphorylation levels at GLI2 target gene promoter regions, thus activating the GLI-mediated transcriptional program and promoting cell migration and metastasis (135,137).

HOXA transcript induced by TGF-β (HIT)

lncRNA-HIT is an uncharacterized lncRNA located in the HOXA gene cluster, which functions in the nucleus and is significantly induced by TGF-β (138). Richards et al (138) found that lncRNA-HIT performs an important role in TGF-β-induced EMT, migration and invasion. Depletion of lncRNA-HIT significantly increases E-cadherin and reduces vimentin in mouse mammary gland epithelial cells, and inhibits TGF-β induced migration, invasion and EMT. lncRNA-HIT is also significantly elevated in the highly metastatic mouse mammary tumor cell line (4T1) (138). Knockdown of lncRNA-HIT in this cell line results in significant reduction of cell migration, invasion, tumor growth and lung metastasis (138). Furthermore, lncRNA-HIT is conserved in humans and its expression is associated with more invasive breast cancer progression (138). The expression of lncRNA-HIT could be a potential therapeutic target of breast cancer.

NF-κB-interacting lncRNA (NKILA)

NKILA, an NF-κB-induced lncRNA, is a 2570-nt intronless transcript that is encoded at chromosome 20q13. NKILA exerts its biological function in the cytoplasm, serves as a negative feedback regulator of NF-κB in resting and activated cells, and performs a pivotal inhibitory role in breast cancer progression and metastasis (139). NKILA binds to the NF-κB/inhibitor of NF-κB (IκB) complex, and inhibits IκB kinase-induced IκB phosphorylation, the NF-κB pathway and cancer-associated inflammation (139). In addition, NKILA is significantly reduced by miR-103/107-mediated degradation in high-metastatic breast cancer cells, and the decrease of NKILA results in abnormal NF-κB activation and thus contributes to cancer metastasis and poor patient outcome (139).

Neuroblastoma-associated transcript 1 (NBAT1)

NBAT1 was identified in neuroblastoma as a tumor suppressing lncRNA, and it controls tumor progression by epigenetic silencing of target genes, including Sox9, oncostatin M receptor and versican (140). NBAT1 loss affects neuronal differentiation through activation of the neuronal-specific transcription factor neuron restrictive silencer factor/RE1-silencing transcription factor. Furthermore, loss of NBAT1 is associated with cellular proliferation and invasion (140). NBAT1 regulates tumor biological processes by interacting with PRC2 member EZH2, which suppresses its target gene that affects cell proliferation and migration (140,141). In breast cancer, loss of NBAT1 is also associated with tumor metastasis and poor clinical prognosis (142). Overexpression of NBAT1 could inhibit the migration and invasion of breast cancer cells through EZH2 (142). In addition, NBAT1 could suppress breast cancer metastasis by regulating dickkopf Wnt signaling pathway inhibitor 1 (DKK1) in a PRC2-dependent manner. DKK1 is an inhibitor of the Wnt signaling pathway, which inhibits the migration and invasion of breast cancer (143145). Mechanically, overexpression of NBAT1 decreases the H3K27me3 level of the DKK1 promoter and activates the expression of DKK1, inhibiting breast cancer cell migration and invasion (146). Therefore, NBAT1 may serve as a breast cancer prognostic biomarker and as a potential therapeutic target to inhibit breast cancer metastasis.

Other lncRNAs

Other lncRNAs have also been found to be involved in breast cancer invasion and metastasis, including steroid receptor RNA activator (SRA), linc00617, LOC554202 and AK058003. SRA was reported to be overexpressed in breast cancer cells; knockdown of SRA in MDA-MB-231 cells reduced the invasiveness and expression of certain critical genes, including thrombospondin 1, caveolin 1, transmembrane protease serine (TMPRSS)2, TMPRSS3, MMP-1 and MMP-9 (147). linc00617 promotes breast cancer cell motility and invasion in vitro and potentiates breast cancer cell metastasis in vivo (148). Furthermore, linc00617 induces the generation of stem cells; it activates the transcription of stemness factor Sox2, which stimulates EMT transition and promotes breast cancer progression and metastasis (148). In addition, LOC554202 (149) and AK058003 (150) were also revealed to be upregulated in breast cancer cells and tissues, and the expression levels of these two lncRNAs were strongly associated with tumor size and advanced clinical stage. Knockdown of Loc554202 and AK058003 significantly inhibits breast cancer cell proliferation, invasion and metastasis (149,150).

Conclusions

Metastasis is the major cause of breast cancer-associated mortlaity, and the invasion-metastasis cascade of breast cancer is formed from a diverse range of biological processes. Therefore, it is essential to identify how to diagnose breast cancer at the early stage and develop anti-metastatic therapeutics. Emerging evidence is beginning to reveal the potential role of lncRNA in breast cancer pathogenesis, and it is now considered to perform an essential role in pro- and anti-metastatic processes (Table I). These novel findings provide fresh molecular insights into the invasion and metastasis of breast cancer and may suggest a novel strategy for breast cancer therapy. However, studies of lncRNAs remain in the early stages, and the underlying mechanism of lncRNAs in regulating the invasion and metastasis of breast cancer is not clear. Furthermore, the expression of lncRNAs in breast cancer lacks specificity and the biological functions of certain lncRNAs remain under considerable debate. Therefore, identification of highly specific and sensitive lncRNAs for the early monitoring of the invasion and metastasis of breast cancer remains an arduous task. An improved understanding of the network of genes and cellular pathways regulated by lncRNAs will aid in improving the understanding of breast cancer pathogenesis, and thus will open novel avenues for breast cancer diagnosis, prognosis and therapy.

Table I.

Functions of lncRNAs in breast cancer invasion and metastasis.

Table I.

Functions of lncRNAs in breast cancer invasion and metastasis.

lncRNAExpressionRegulatory mechanisms of invasion and metastasis
HOTAIRUpregulatedTrimethylation of histone H3K27 by recruiting PRC2
Repression of E-cadherin by PRC2
Competitively binds miR-7 and miR-568
Transcriptionally induced by estradiol
Induces EMT program and CSCs arising
H19UpregulatedInduced by HGF and E2F1
Increases EGFR and c-Met
Decreases E-cadherin, increases N-cadherin and Slug
Activates Akt and ERK signaling pathways
MALAT-1UpregulatedActives transcription
Regulates expression of cell cycle genes
Regulates EMT program
LSINCT5UpregulatedRegulates CXCR4 expression
lincRNA-RORUpregulatedSuppresses p53 translation
Transcriptionally inhibited by NRF2
Competitively binds miRNA-145 and miRNA-205
Promotes survival of iPSCs and ESCs
treRNAUpregulatedPromotes formation of a ribonucleoprotein complex with
hnRNP K, FXR1, FXR2, PUF60 and SF3B3
Enhances Snail transcription
Decreases expression of E-cadherin, zonulaoccludens-1 and β-catenin
Increases fibronectin and vimentin expression
BC200UpregulatedSequesters PABP, preventing protein synthesis
lncRNA-ATBUpregulatedRegulates trastuzumab resistance
Competitively binds miR-200c, upregulating ZEB1 and ZNF-217
Induces IL-11 expression
BCAR4UpregulatedInduced by tamoxifen resistance
Associated with ER, PR and ERBB2/3 signaling
Activation of Hedgehog/GLI2
Activation of p300
lncRNA-HITUpregulatedInduced by TGF-β
Decreases E-cadherin and increases vimentin
Regulator of TGF-β induced EMT
NKILADownregulatedBinds to NF-κB/IκB and represses NF-κB activation
Regulated by miR-103/107
Inhibits the NF-κB signaling pathway
Inhibits cancer-associated inflammation
NBAT1DownregulatedInteraction with PRC2 member EZH2
Activation of DKK1 expression
Inhibits the Wnt signaling pathway
SRAUpregulatedActivation of GR, ERα, PR and SRAP
Coactivates THBS1, CAV1, TMPRSS2, TMPRSS3, MMP-1 and MMP-9
linc00617UpregulatedActivates the transcription of stemness factor Sox2
Induces generation of stem cells
AK058003UpregulatedRegulates γ-synuclein gene

[i] lncRNA, long non-coding RNA; HOTAIR, HOX antisense intergenic RNA; H3K27, histone H3 lysine 27; miR, microRNA; PRC2, polycomb repressive complex 2; HGF, hepatocyte growth factor; E2F1, E2F transcription factor 1; EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; MALAT-1, metastasis-associated lung adenocarcinoma transcript-1; EMT, epithelial-mesenchymal transition; CXCR4, chemokine (C-X-C motif) receptor 4; lincRNA-ROR, long intergenic non-protein coding RNA-regulator of reprogramming; NRF2, nuclear factor-erythroid 2-related factor; iPSC, induced pluripotent stem cell; ESC, endothelial progenitor cell; treRNA, translational regulatory lncRNA; hnRNP K, heterogeneous nuclear ribonucleoprotein K; FXR, FMR autosomal homolog; PUF60, poly(U) binding splicing factor 60; SF3B3, splicing factor 3b subunit 3; PABP, polyadenylate-binding protein; TGF-β, transforming growth factor-β; lncRNA-ATB, lncRNA-activated by TGF-β; ZEB1, zinc finger E-box binding homeobox 1; ZNF-217, zinc finger protein 217; IL-11, interleukin-11; ER, estrogen receptor; PR, progesterone receptor; ERBB2/3, erb-b2 receptor tyrosine kinase 2/3; BCAR4, breast cancer anti-estrogen resistance 4; HIT, HOXA transcript induced by TGF-β; NF-κB, nuclear factor-κB; NKILA, NF-κB interacting lncRNA; NBAT1, neuroblastoma-associated transcript 1; EZH2, enhancer of zeste homolog 2; DKK1, dickkopf Wnt signaling pathway inhibitor 1; GR, glucocorticoid receptor; SRAP, steroid receptor activator protein; THBS1, thrombospondin 1; CAV1, caveolin 1; TMPRSS2/3, transmembrane protease, serine 2/3; MMP-1/9, matrix metalloproteinase-1/9; Sox2, sex-determining region Y-box 2.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

DeSantis C, Ma J, Bryan L and Jemal A: Breast cancer statistics, 2013. CA Cancer J Clin. 64:52–62. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Huober J and Thürlimann B: The role of combination chemotherapy in the treatment of patients with metastatic breast cancer. Breast Care (Basel). 4:367–372. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Cardoso F, Harbeck N, Fallowfield L, Kyriakides S and Senkus E; ESMO Guidelines Working Group, : Locally recurrent or metastatic breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 23 Suppl 7:vii11–vii19. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Chaffer CL and Weinberg RA: A perspective on cancer cell metastasis. Science. 331:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Zhang J, Tian XJ, Zhang H, Teng Y, Li R, Bai F, Elankumaran S and Xing J: TGF-β-induced epithelial-to-mesenchymal transition proceeds through stepwise activation of multiple feedback loops. Sci Signal. 7:ra912014. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

van Bakel H and Hughes TR: Establishing legitimacy and function in the new transcriptome. Brief Funct Genomic Proteomic. 8:424–436. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Perkel JM: Visiting ‘noncodarnia’. Biotechniques. 54:301–304. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Fabian MR and Sonenberg N: The mechanics of miRNA-mediated gene silencing: A look under the hood of miRISC. Nat Struct Mol Biol. 19:586–593. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Kong YW, Ferland-McCollough D, Jackson TJ and Bushell M: microRNAs in cancer management. Lancet Oncol. 13:e249–e258. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Huarte M and Rinn JL: Large non-coding RNAs: Missing links in cancer? Hum Mol Genet. 19:R152–R161. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Gibb EA, Brown CJ and Lam WL: The functional role of long non-coding RNA in human carcinomas. Mol Cancer. 10:382011. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Hauptman N and Glavač D: Long non-coding RNA in cancer. Int J Mol Sci. 14:4655–4669. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Ernst C and Morton CC: Identification and function of long non-coding RNA. Front Cell Neurosci. 7:1682013. View Article : Google Scholar : PubMed/NCBI

23 

Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, et al: The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Res. 22:1775–1789. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Nie L, Wu HJ, Hsu JM, Chang SS, Labaff AM, Li CW, Wang Y, Hsu JL and Hung MC: Long non-coding RNAs: Versatile master regulators of gene expression and crucial players in cancer. Am J Transl Res. 4:127–150. 2012.PubMed/NCBI

25 

van Heesch S, van Iterson M, Jacobi J, Boymans S, Essers PB, de Bruijn E, Hao W, MacInnes AW, Cuppen E and Simonis M: Extensive localization of long noncoding RNAs to the cytosol and mono- and polyribosomal complexes. Genome Biol. 15:R62014. View Article : Google Scholar : PubMed/NCBI

26 

Wang KC and Chang HY: Molecular mechanisms of long noncoding RNAs. Mol Cell. 43:904–914. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Guttman M and Rinn JL: Modular regulatory principles of large non-coding RNAs. Nature. 482:339–346. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Mattick JS: The genetic signatures of noncoding RNAs. PLoS Genet. 5:e10004592009. View Article : Google Scholar : PubMed/NCBI

31 

Bergmann JH and Spector DL: Long non-coding RNAs: Modulators of nuclear structure and function. Curr Opin Cell Biol. 26:10–18. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Foroni C, Broggini M, Generali D and Damia G: Epithelial-mesenchymal transition and breast cancer: Role, molecular mechanisms and clinical impact. Cancer Treat Rev. 38:689–697. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Vanharanta S and Massagué J: Origins of metastatic traits. Cancer Cell. 24:410–421. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Lamouille S, Subramanyam D, Blelloch R and Derynck R: Regulation of epithelial-mesenchymal and mesenchymal-epithelial transitions by microRNAs. Curr Opin Cell Biol. 25:200–207. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Brabletz T: To differentiate or not-routes towards metastasis. Nat Rev Cancer. 12:425–436. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Chaw SY, Majeed AA, Dalley AJ, Chan A, Stein S and Farah CS: Epithelial to mesenchymal transition (EMT) biomarkers-E-cadherin, beta-catenin, APC and Vimentin-in oral squamous cell carcinogenesis and transformation. Oral Oncol. 48:997–1006. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Marsan M, Van den Eynden G, Limame R, Neven P, Hauspy J, Van Dam PA, Vergote I, Dirix LY, Vermeulen PB and Van Laere SJ: A core invasiveness gene signature reflects epithelial-to-mesenchymal transition but not metastatic potential in breast cancer cell lines and tissue samples. PLoS One. 9:e892622014. View Article : Google Scholar : PubMed/NCBI

38 

Jin L, Chen J, Li L, Li C, Chen C and Li S: CRH suppressed TGFβ1-induced Epithelial-Mesenchymal Transition via induction of E-cadherin in breast cancer cells. Cell Signal. 26:757–765. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Gumbiner BM: Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol. 6:622–634. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Gheldof A and Berx G: Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl Sci. 116:317–336. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Acloque H, Adams MS, Fishwick K, Bronner-Fraser M and Nieto MA: Epithelial-mesenchymal transitions: The importance of changing cell state in development and disease. J Clin Invest. 119:1438–1449. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Oka H, Shiozaki H, Kobayashi K, Inoue M, Tahara H, Kobayashi T, Takatsuka Y, Matsuyoshi N, Hirano S, Takeichi M, et al: Expression of E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. Cancer Res. 53:1696–1701. 1993.PubMed/NCBI

44 

Schipper JH, Frixen UH, Behrens J, Unger A, Jahnke K and Birchmeier W: E-cadherin expression in squamous cell carcinomas of head and neck: Inverse correlation with tumor dedifferentiation and lymph node metastasis. Cancer Res. 51:6328–6337. 1991.PubMed/NCBI

45 

Derksen PW, Liu X, Saridin F, van der Gulden H, Zevenhoven J, Evers B, van Beijnum JR, Griffioen AW, Vink J, Krimpenfort P, et al: Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell. 10:437–449. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Onder TT, Gupta PB, Mani SA, Yang J, Lander ES and Weinberg RA: Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68:3645–3654. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Mallini P, Lennard T, Kirby J and Meeson A: Epithelial-to-mesenchymal transition: What is the impact on breast cancer stem cells and drug resistance. Cancer Treat Rev. 40:341–348. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Chao Y, Wu Q, Acquafondata M, Dhir R and Wells A: Partial mesenchymal to epithelial reverting transition in breast and prostate cancer metastases. Cancer Microenviron. 5:19–28. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Moody SE, Perez D, Pan TC, Sarkisian CJ, Portocarrero CP, Sterner CJ, Notorfrancesco KL, Cardiff RD and Chodosh LA: The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell. 8:197–209. 2005. View Article : Google Scholar : PubMed/NCBI

50 

De Craene B and Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 13:97–110. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Lin T, Ponn A, Hu X, Law BK and Lu J: Requirement of the histone demethylase LSD1 in Snai1-mediated transcriptional repression during epithelial-mesenchymal transition. Oncogene. 29:4896–4904. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Tong ZT, Cai MF, Wang XG, Kong LL, Mai SJ, Liu YH, Zhang HB, Liao YJ, Zheng F, Zhu W, et al: EZH2 supports nasopharyngeal carcinoma cell aggressiveness by forming a co-repressor complex with HDAC1/HDAC2 and Snail to inhibit E-cadherin. Oncogene. 31:583–594. 2012.PubMed/NCBI

53 

Herranz N, Pasini D, Díaz VM, Francí C, Gutierrez A, Dave N, Escrivà M, Hernandez-Muñoz I, Di Croce L, Helin K, et al: Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor. Mol Cell Biol. 28:4772–4781. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Dong C, Wu Y, Wang Y, Wang C, Kang T, Rychahou PG, Chi YI, Evers BM and Zhou BP: Interaction with Suv39H1 is critical for Snail-mediated E-cadherin repression in breast cancer. Oncogene. 32:1351–1362. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Dong C, Wu Y, Yao J, Wang Y, Yu Y, Rychahou PG, Evers BM and Zhou BP: G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. J Clin Invest. 122:1469–1486. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Polyak K and Weinberg RA: Transitions between epithelial and mesenchymal states: Acquisition of malignant and stem cell traits. Nat Rev Cancer. 9:265–273. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Yang J and Weinberg RA: Epithelial-mesenchymal transition: At the crossroads of development and tumor metastasis. Dev Cell. 14:818–829. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Dubrovska A, Kim S, Salamone RJ, Walker JR, Maira SM, García-Echeverría C, Schultz PG and Reddy VA: The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci USA. 106:pp. 268–273. 2009; View Article : Google Scholar : PubMed/NCBI

59 

Padua D and Massagué J: Roles of TGFbeta in metastasis. Cell Res. 19:89–102. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Zhou BP and Hung MC: Wnt, hedgehog and snail: Sister pathways that control by GSK-3beta and beta-Trcp in the regulation of metastasis. Cell Cycle. 4:772–776. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Guo F, Kerrigan BC Parker, Yang D, Hu L, Shmulevich I, Sood AK, Xue F and Zhang W: Post-transcriptional regulatory network of epithelial-to-mesenchymal and mesenchymal-to-epithelial transitions. J Hematol Oncol. 7:192014. View Article : Google Scholar : PubMed/NCBI

62 

Ceppi P and Peter ME: MicroRNAs regulate both epithelial-to-mesenchymal transition and cancer stem cells. Oncogene. 33:269–278. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Dumont N and Tlsty TD: Reflections on miRing effects in metastasis. Cancer Cell. 16:3–4. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, et al: EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA. 100:pp. 11606–11611. 2003; View Article : Google Scholar : PubMed/NCBI

67 

Chisholm KM, Wan Y, Li R, Montgomery KD, Chang HY and West RB: Detection of long non-coding RNA in archival tissue: Correlation with polycomb protein expression in primary and metastatic breast carcinoma. PLoS One. 7:e479982012. View Article : Google Scholar : PubMed/NCBI

68 

Alves C Padua, Fonseca AS, Muys BR, de Barros E Lima Bueno R, Bürger MC, de Souza JE, Valente V, Zago MA and Silva WA Jr: Brief report: The lincRNA Hotair is required for epithelial-to-mesenchymal transition and stemness maintenance of cancer cell lines. Stem Cells. 31:2827–2832. 2013. View Article : Google Scholar : PubMed/NCBI

69 

Zhang H, Cai K, Wang J, Wang X, Cheng K, Shi F, Jiang L, Zhang Y and Dou J: MiR-7, inhibited indirectly by lincRNA HOTAIR, directly inhibits SETDB1 and reverses the EMT of breast cancer stem cells by downregulating the STAT3 pathway. Stem Cells. 32:2858–2868. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Li JT, Wang LF, Zhao YL, Yang T, Li W, Zhao J, Yu F, Wang L, Meng YL, Liu NN, et al: Nuclear factor of activated T cells 5 maintained by Hotair suppression of miR-568 upregulates S100 calcium binding protein A4 to promote breast cancer metastasis. Breast Cancer Res. 16:4542014. View Article : Google Scholar : PubMed/NCBI

71 

Gokmen-Polar Y, Vladislav IT, Neelamraju Y, Janga SC and Badve S: Prognostic impact of HOTAIR expression is restricted to ER-negative breast cancers. Sci Rep. 5:87652015. View Article : Google Scholar : PubMed/NCBI

72 

Sorensen KP, Thomassen M, Tan Q, Bak M, Cold S, Burton M, Larsen MJ and Kruse TA: Long non-coding RNA HOTAIR is an independent prognostic marker of metastasis in estrogen receptor-positive primary breast cancer. Breast Cancer Res Treat. 142:529–536. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Bhan A, Hussain I, Ansari KI, Kasiri S, Bashyal A and Mandal SS: Antisense transcript long noncoding RNA (lncRNA) HOTAIR is transcriptionally induced by estradiol. J Mol Biol. 425:3707–3722. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Bhan A, Hussain I, Ansari KI, Bobzean SA, Perrotti LI and Mandal SS: Bisphenol-A and diethylstilbestrol exposure induces the expression of breast cancer associated long noncoding RNA HOTAIR in vitro and in vivo. J Steroid Biochem Mol Biol. 141:160–170. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Tao S, He H and Chen Q: Estradiol induces HOTAIR levels via GPER-mediated miR-148a inhibition in breast cancer. J Transl Med. 13:1312015. View Article : Google Scholar : PubMed/NCBI

76 

Bartolomei MS, Zemel S and Tilghman SM: Parental imprinting of the mouse H19 gene. Nature. 351:153–155. 1991. View Article : Google Scholar : PubMed/NCBI

77 

Gabory A, Jammes H and Dandolo L: The H19 locus: Role of an imprinted non-coding RNA in growth and development. Bioessays. 32:473–480. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Brannan CI, Dees EC, Ingram RS and Tilghman SM: The product of the H19 gene may function as an RNA. Mol Cell Biol. 10:28–36. 1990. View Article : Google Scholar : PubMed/NCBI

79 

Giannoukakis N, Deal C, Paquette J, Goodyer CG and Polychronakos C: Parental genomic imprinting of the human IGF2 gene. Nat Genet. 4:98–101. 1993. View Article : Google Scholar : PubMed/NCBI

80 

Soejima H and Higashimoto K: Epigenetic and genetic alterations of the imprinting disorder Beckwith-Wiedemann syndrome and related disorders. J Hum Genet. 58:402–409. 2013. View Article : Google Scholar : PubMed/NCBI

81 

Ma C, Nong K, Zhu H, Wang W, Huang X, Yuan Z and Ai K: H19 promotes pancreatic cancer metastasis by derepressing let-7's suppression on its target HMGA2-mediated EMT. Tumour Biol. 35:9163–9169. 2014. View Article : Google Scholar : PubMed/NCBI

82 

Luo M, Li Z, Wang W, Zeng Y, Liu Z and Qiu J: Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 333:213–221. 2013. View Article : Google Scholar : PubMed/NCBI

83 

Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ and Kwok TT: Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 31:350–358. 2010. View Article : Google Scholar : PubMed/NCBI

84 

Li H, Yu B, Li J, Su L, Yan M, Zhu Z and Liu B: Overexpression of lncRNA H19 enhances carcinogenesis and metastasis of gastric cancer. Oncotarget. 5:2318–2329. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Lottin S, Adriaenssens E, Dupressoir T, Berteaux N, Montpellier C, Coll J, Dugimont T and Curgy JJ: Overexpression of an ectopic H19 gene enhances the tumorigenic properties of breast cancer cells. Carcinogenesis. 23:1885–1895. 2002. View Article : Google Scholar : PubMed/NCBI

86 

Adriaenssens E, Lottin S, Berteaux N, Hornez L, Fauquette W, Fafeur V, Peyrat JP, Le Bourhis X, Hondermarck H, Coll J, et al: Cross-talk between mesenchyme and epithelium increases H19 gene expression during scattering and morphogenesis of epithelial cells. Exp Cell Res. 275:215–229. 2002. View Article : Google Scholar : PubMed/NCBI

87 

Berteaux N, Lottin S, Monte D, Pinte S, Quatannens B, Coll J, Hondermarck H, Curgy JJ, Dugimont T and Adriaenssens E: H19 mRNA-like noncoding RNA promotes breast cancer cell proliferation through positive control by E2F1. J Biol Chem. 280:29625–29636. 2005. View Article : Google Scholar : PubMed/NCBI

88 

Matouk IJ, Raveh E, Abu-lail R, Mezan S, Gilo M, Gershtain E, Birman T, Gallula J, Schneider T, Barkali M, et al: Oncofetal H19 RNA promotes tumor metastasis. Biochim Biophys Acta. 1843:1414–1426. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Xue G, Restuccia DF, Lan Q, Hynx D, Dirnhofer S, Hess D, Rüegg C and Hemmings BA: Akt/PKB-mediated phosphorylation of Twist1 promotes tumor metastasis via mediating cross-talk between PI3K/Akt and TGF-beta signaling axes. Cancer discov. 2:248–259. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Vennin C, Spruyt N, Dahmani F, Julien S, Bertucci F, Finetti P, Chassat T, Bourette RP, Le Bourhis X and Adriaenssens E: H19 non coding RNA-derived miR-675 enhances tumorigenesis and metastasis of breast cancer cells by downregulating c-Cbl and Cbl-b. Oncotarget. 6:29209–29223. 2015.PubMed/NCBI

91 

Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al: MALAT-1, a novel noncoding RNA and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 22:8031–8041. 2003. View Article : Google Scholar : PubMed/NCBI

92 

Jiao F, Hu H, Han T, Yuan C and Wang L, Jin Z, Guo Z and Wang L: Long noncoding RNA MALAT-1 enhances stem cell-like phenotypes in pancreatic cancer cells. Int J Mol Sci. 16:6677–6693. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Lai MC, Yang Z, Zhou L, Zhu QQ, Xie HY, Zhang F, Wu LM, Chen LM and Zheng SS: Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 29:1810–1816. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Zhang ZJ, Tong YQ, Wang JJ, Yang C, Zhou GH, Li YH, Xie PL, Hu JY and Li GC: Spaceflight alters the gene expression profile of cervical cancer cells. Chin J Cancer. 30:842–852. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Xu C, Yang M, Tian J, Wang X and Li Z: MALAT-1: A long non-coding RNA and its important 3′ end functional motif in colorectal cancer metastasis. Int J Oncol. 39:169–175. 2011.PubMed/NCBI

96 

Ying L, Chen Q, Wang Y, Zhou Z, Huang Y and Qiu F: Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 8:2289–2294. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Zhao Z, Chen C, Liu Y and Wu C: 17β-Estradiol treatment inhibits breast cell proliferation, migration and invasion by decreasing MALAT-1 RNA level. Biochem Biophys Res Commun. 445:388–393. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Tano K and Akimitsu N: Long non-coding RNAs in cancer progression. Front Genet. 3:2192012. View Article : Google Scholar : PubMed/NCBI

100 

Xu S, Sui S, Zhang J, Bai N, Shi Q, Zhang G, Gao S, You Z, Zhan C, Liu F and Pang D: Downregulation of long noncoding RNA MALAT1 induces epithelial-to-mesenchymal transition via the PI3K-AKT pathway in breast cancer. Int J Clin Exp Pathol. 8:4881–4891. 2015.PubMed/NCBI

101 

Silva JM, Boczek N, Berres MW, Ma X and Smith DI: LSINCT5 is over expressed in breast and ovarian cancer and affects cellular proliferation. RNA Biol. 8:496–505. 2011. View Article : Google Scholar : PubMed/NCBI

102 

Silva JM, Perez DS, Pritchett JR, Halling ML, Tang H and Smith DI: Identification of long stress-induced non-coding transcripts that have altered expression in cancer. Genomics. 95:355–362. 2010. View Article : Google Scholar : PubMed/NCBI

103 

Hassan S, Buchanan M, Jahan K, Aguilar-Mahecha A, Gaboury L, Muller WJ, Alsawafi Y, Mourskaia AA, Siegel PM, Salvucci O and Basik M: CXCR4 peptide antagonist inhibits primary breast tumor growth, metastasis and enhances the efficacy of anti-VEGF treatment or docetaxel in a transgenic mouse model. Int J Cancer. 129:225–232. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Sobolik T, Su YJ, Wells S, Ayers GD, Cook RS and Richmond A: CXCR4 drives the metastatic phenotype in breast cancer through induction of CXCR2 and activation of MEK and PI3K pathways. Mol Biol Cell. 25:566–582. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Yang P, Liang SX, Huang WH, Zhang HW, Li XL, Xie LH, Du CW and Zhang GJ: Aberrant expression of CXCR4 significantly contributes to metastasis and predicts poor clinical outcome in breast cancer. Curr Mol Med. 14:174–184. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Loewer S, Cabili MN, Guttman M, Loh YH, Thomas K, Park IH, Garber M, Curran M, Onder T, Agarwal S, et al: Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet. 42:1113–1117. 2010. View Article : Google Scholar : PubMed/NCBI

107 

Zhang A, Zhou N, Huang J, Liu Q, Fukuda K, Ma D, Lu Z, Bai C, Watabe K and Mo YY: The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res. 23:340–350. 2013. View Article : Google Scholar : PubMed/NCBI

108 

Zhang Y, Xia J, Li Q, Yao Y, Eades G, Gernapudi R, Duru N, Kensler TW and Zhou Q: NRF2/long noncoding RNA ROR signaling regulates mammary stem cell expansion and protects against estrogen genotoxicity. J Biol Chem. 289:31310–31318. 2014. View Article : Google Scholar : PubMed/NCBI

109 

Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, Wu M, Xiong J, Guo X and Liu H: Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell. 25:69–80. 2013. View Article : Google Scholar : PubMed/NCBI

110 

Yin X, Zhang BH, Zheng SS, Gao DM, Qiu SJ, Wu WZ and Ren ZG: Coexpression of gene Oct4 and Nanog initiates stem cell characteristics in hepatocellular carcinoma and promotes epithelial-mesenchymal transition through activation of Stat3/Snail signaling. J Hematol Oncol. 8:232015. View Article : Google Scholar : PubMed/NCBI

111 

Liu L, Zhang J, Fang C, Zhang Z, Feng Y and Xi X: OCT4 mediates FSH-induced epithelial-mesenchymal transition and invasion through the ERK1/2 signaling pathway in epithelial ovarian cancer. Biochem Biophys Res Commun. 461:525–532. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Hu J, Guo H, Li H, Liu Y, Liu J, Chen L, Zhang J and Zhang N: MiR-145 regulates epithelial to mesenchymal transition of breast cancer cells by targeting Oct4. PLoS One. 7:e459652012. View Article : Google Scholar : PubMed/NCBI

113 

Eades G, Wolfson B, Zhang Y, Li Q, Yao Y and Zhou Q: lincRNA-RoR and miR-145 regulate invasion in triple-negative breast cancer via targeting ARF6. Mol Cancer Res. 13:330–338. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Hashimoto S, Onodera Y, Hashimoto A, Tanaka M, Hamaguchi M, Yamada A and Sabe H: Requirement for Arf6 in breast cancer invasive activities. Proc Natl Acad Sci USA. 101:pp. 6647–6652. 2004; View Article : Google Scholar : PubMed/NCBI

115 

Sabe H, Hashimoto S, Morishige M, Ogawa E, Hashimoto A, Nam JM, Miura K, Yano H and Onodera Y: The EGFR-GEP100-Arf6-AMAP1 signaling pathway specific to breast cancer invasion and metastasis. Traffic. 10:982–993. 2009. View Article : Google Scholar : PubMed/NCBI

116 

Hou P, Zhao Y, Li Z, Yao R, Ma M, Gao Y, Zhao L, Zhang Y, Huang B and Lu J: LincRNA-ROR induces epithelial-to-mesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis. 5:e12872014. View Article : Google Scholar : PubMed/NCBI

117 

Chen YM, Liu Y, Wei HY, Lv KZ and Fu P: Linc-ROR induces epithelial-mesenchymal transition and contributes to drug resistance and invasion of breast cancer cells. Tumour Biol. 37:10861–10870. 2016. View Article : Google Scholar : PubMed/NCBI

118 

Orom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, et al: Long noncoding RNAs with enhancer-like function in human cells. Cell. 143:46–58. 2010. View Article : Google Scholar : PubMed/NCBI

119 

Gumireddy K, Li A, Yan J, Setoyama T, Johannes GJ, Orom UA, Tchou J, Liu Q, Zhang L, Speicher DW, et al: Identification of a long non-coding RNA-associated RNP complex regulating metastasis at the translational step. EMBO J. 32:2672–2684. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Hansji H, Leung EY, Baguley BC, Finlay GJ and Askarian-Amiri ME: Keeping abreast with long non-coding RNAs in mammary gland development and breast cancer. Front Genet. 5:3792014. View Article : Google Scholar : PubMed/NCBI

121 

Watson JB and Sutcliffe JG: Primate brain-specific cytoplasmic transcript of the Alu repeat family. Mol Cell Biol. 7:3324–3327. 1987. View Article : Google Scholar : PubMed/NCBI

122 

Tiedge H, Chen W and Brosius J: Primary structure, neural-specific expression, and dendritic location of human BC200 RNA. J Neurosci. 13:2382–2390. 1993.PubMed/NCBI

123 

Chen W, Böcker W, Brosius J and Tiedge H: Expression of neural BC200 RNA in human tumours. J Pathol. 183:345–351. 1997. View Article : Google Scholar : PubMed/NCBI

124 

Iacoangeli A, Lin Y, Morley EJ, Muslimov IA, Bianchi R, Reilly J, Weedon J, Diallo R, Böcker W and Tiedge H: BC200 RNA in invasive and preinvasive breast cancer. Carcinogenesis. 25:2125–2133. 2004. View Article : Google Scholar : PubMed/NCBI

125 

De Leeneer K and Claes K: Non Coding RNA Molecules as Potential Biomarkers in Breast Cancer. Adv Exp Med Biol. 867:263–275. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014. View Article : Google Scholar : PubMed/NCBI

127 

Li W and Kang Y: A new Lnc in metastasis: Long noncoding RNA mediates the prometastatic functions of TGF-β. Cancer Cell. 25:557–559. 2014. View Article : Google Scholar : PubMed/NCBI

128 

Iguchi T, Uchi R, Nambara S, Saito T, Komatsu H, Hirata H, Ueda M, Sakimura S, Takano Y, Kurashige J, et al: A long noncoding RNA, lncRNA-ATB, is involved in the progression and prognosis of colorectal cancer. Anticancer Res. 35:1385–1388. 2015.PubMed/NCBI

129 

Saito T, Kurashige J, Nambara S, Komatsu H, Hirata H, Ueda M, Sakimura S, Uchi R, Takano Y, Shinden Y, et al: A long non-coding RNA activated by transforming growth factor-beta is an independent prognostic marker of gastric cancer. Ann Surg Oncol. 22 Suppl 3:915–922. 2015. View Article : Google Scholar

130 

Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA and Massagué J: A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 3:537–549. 2003. View Article : Google Scholar : PubMed/NCBI

131 

Shi SJ, Wang LJ, Yu B, Li YH, Jin Y and Bai XZ: LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget. 6:11652–11663. 2015. View Article : Google Scholar : PubMed/NCBI

132 

Meijer D, van Agthoven T, Bosma PT, Nooter K and Dorssers LC: Functional screen for genes responsible for tamoxifen resistance in human breast cancer cells. Mol Cancer Res. 4:379–386. 2006. View Article : Google Scholar : PubMed/NCBI

133 

Godinho MF, Sieuwerts AM, Look MP, Meijer D, Foekens JA, Dorssers LC and van Agthoven T: Relevance of BCAR4 in tamoxifen resistance and tumour aggressiveness of human breast cancer. Br J Cancer. 103:1284–1291. 2010. View Article : Google Scholar : PubMed/NCBI

134 

Godinho M, Meijer D, Setyono-Han B, Dorssers LC and van Agthoven T: Characterization of BCAR4, a novel oncogene causing endocrine resistance in human breast cancer cells. J Cell Physiol. 226:1741–1749. 2011. View Article : Google Scholar : PubMed/NCBI

135 

Xing Z, Lin A, Li C, Liang K, Wang S, Liu Y, Park PK, Qin L, Wei Y, Hawke DH, et al: lncRNA directs cooperative epigenetic regulation downstream of chemokine signals. Cell. 159:1110–1125. 2014. View Article : Google Scholar : PubMed/NCBI

136 

Xing Z, Lin C and Yang L: Unraveling the therapeutic potential of the LncRNA-dependent noncanonical Hedgehog pathway in cancer. Mol Cell Oncol. 2:e9989002015. View Article : Google Scholar : PubMed/NCBI

137 

Xing Z, Park PK, Lin C and Yang L: LncRNA BCAR4 wires up signaling transduction in breast cancer. RNA Biol. 12:681–689. 2015. View Article : Google Scholar : PubMed/NCBI

138 

Richards EJ, Zhang G, Li ZP, Permuth-Wey J, Challa S, Li Y, Kong W, Dan S, Bui MM, Coppola D, et al: Long non-coding RNAs (LncRNA) regulated by transforming growth factor (TGF) β: LncRNA-hit-mediated TGFβ-induced epithelial to mesenchymal transition in mammary epithelia. J Biol Chem. 290:6857–6867. 2015. View Article : Google Scholar : PubMed/NCBI

139 

Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, Lin L, Yao H, Su F, Li D, et al: A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell. 27:370–381. 2015. View Article : Google Scholar : PubMed/NCBI

140 

Pandey GK, Mitra S, Subhash S, Hertwig F, Kanduri M, Mishra K, Fransson S, Ganeshram A, Mondal T, Bandaru S, et al: The risk-associated long noncoding RNA NBAT-1 controls neuroblastoma progression by regulating cell proliferation and neuronal differentiation. Cancer Cell. 26:722–737. 2014. View Article : Google Scholar : PubMed/NCBI

141 

Pandey GK and Kanduri C: Fighting Neuroblastomas with NBAT1. Oncoscience. 2:79–80. 2015. View Article : Google Scholar : PubMed/NCBI

142 

Hu P, Chu J, Wu Y, Sun L, Lv X, Zhu Y, Li J, Guo Q, Gong C, Liu B and Su S: NBAT1 suppresses breast cancer metastasis by regulating DKK1 via PRC2. Oncotarget. 6:32410–32425. 2015.PubMed/NCBI

143 

Cowling VH, D'Cruz CM, Chodosh LA and Cole MD: c-Myc transforms human mammary epithelial cells through repression of the Wnt inhibitors DKK1 and SFRP1. Mol Cell Biol. 27:5135–5146. 2007. View Article : Google Scholar : PubMed/NCBI

144 

Sato N, Yamabuki T, Takano A, Koinuma J, Aragaki M, Masuda K, Ishikawa N, Kohno N, Ito H, Miyamoto M, et al: Wnt inhibitor Dickkopf-1 as a target for passive cancer immunotherapy. Cancer Res. 70:5326–5336. 2010. View Article : Google Scholar : PubMed/NCBI

145 

Xu WH, Liu ZB, Yang C, Qin W and Shao ZM: Expression of dickkopf-1 and beta-catenin related to the prognosis of breast cancer patients with triple negative phenotype. PLoS One. 7:e376242012. View Article : Google Scholar : PubMed/NCBI

146 

Hu P, Chu J, Wu Y, Sun L, Lv X, Zhu Y, Li J, Guo Q, Gong C, Liu B and Su S: NBAT1 suppresses breast cancer metastasis by regulating DKK1 via PRC2. Oncotarget. 6:32410–32425. 2015.PubMed/NCBI

147 

Foulds CE, Tsimelzon A, Long W, Le A, Tsai SY, Tsai MJ and O'Malley BW: Research resource: Expression profiling reveals unexpected targets and functions of the human steroid receptor RNA activator (SRA) gene. Mol Endocrinol. 24:1090–1105. 2010. View Article : Google Scholar : PubMed/NCBI

148 

Li H, Zhu L, Xu L, Qin K, Liu C, Yu Y, Su D, Wu K and Sheng Y: Long noncoding RNA linc00617 exhibits oncogenic activity in breast cancer. Mol Carcinog. 56:3–17. 2017. View Article : Google Scholar : PubMed/NCBI

149 

Shi Y, Lu J, Zhou J, Tan X, He Y, Ding J, Tian Y, Wang L and Wang K: Long non-coding RNA Loc554202 regulates proliferation and migration in breast cancer cells. Biochem Biophys Res Commun. 446:448–453. 2014. View Article : Google Scholar : PubMed/NCBI

150 

He K and Wang P: Unregulated long non-coding RNA-AK058003 promotes the proliferation, invasion and metastasis of breast cancer by regulating the expression levels of the γ-synuclein gene. Exp Ther Med. 9:1727–1732. 2015.PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang K, Luo Z, Zhang Y, Song X, Zhang L, Wu L and Liu J: Long non-coding RNAs as novel biomarkers for breast cancer invasion and metastasis (Review). Oncol Lett 14: 1895-1904, 2017
APA
Zhang, K., Luo, Z., Zhang, Y., Song, X., Zhang, L., Wu, L., & Liu, J. (2017). Long non-coding RNAs as novel biomarkers for breast cancer invasion and metastasis (Review). Oncology Letters, 14, 1895-1904. https://doi.org/10.3892/ol.2017.6462
MLA
Zhang, K., Luo, Z., Zhang, Y., Song, X., Zhang, L., Wu, L., Liu, J."Long non-coding RNAs as novel biomarkers for breast cancer invasion and metastasis (Review)". Oncology Letters 14.2 (2017): 1895-1904.
Chicago
Zhang, K., Luo, Z., Zhang, Y., Song, X., Zhang, L., Wu, L., Liu, J."Long non-coding RNAs as novel biomarkers for breast cancer invasion and metastasis (Review)". Oncology Letters 14, no. 2 (2017): 1895-1904. https://doi.org/10.3892/ol.2017.6462