Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Dec 15.
Published in final edited form as: Mol Cell Endocrinol. 2015 Feb 3;418(0 3):273–297. doi: 10.1016/j.mce.2015.01.035

miRNAs regulated by estrogens, tamoxifen, and endocrine disruptors and their downstream gene targets

Carolyn M Klinge 1
PMCID: PMC4523495  NIHMSID: NIHMS660834  PMID: 25659536

Abstract

MicroRNAs (miRNAs) are short (22 nucleotides), single-stranded, non-coding RNAs that form complimentary base-pairs with the 3’ untranslated region of target mRNAs within the RNA-induced silencing complex (RISC) and block translation and/or stimulate mRNA transcript degradation. The non-coding miRBase (release 21, June 2014) reports that human genome contains ~2,588 mature miRNAs which regulate ~ 60% of human protein-coding mRNAs. Dysregulation of miRNA expression has been implicated in estrogen-related diseases including breast and endometrial cancers. The mechanism for estrogen regulation of miRNA expression and the role of estrogen-regulated miRNAs in normal homeostasis, reproduction, lactation, and in cancer is an area of great research and clinical interest. Estrogens regulate miRNAs transcription through estrogen receptors α and β in a tissue-specific and cell-dependent manner. This review focuses primary on the regulation of miRNA expression by ligand-activated ERs and their bona fide gene targets and includes miRNAs regulation by tamoxifen and endocrine disrupting chemicals (EDCs) in breast cancer and cell lines.

Keywords: estrogen, estrogen receptor, miRNA, tamoxifen, transcription, mRNA stability, Dicer, Drosha, endocrine-resistance, endocrine disrupting chemical, epithelial-mesenchymal transformation (EMT)

1. Introduction

The three primary estrogenic steroid hormones: estradiol, estrone, and estriol regulate fertility, development, and homeostasis in various tissues including the brain, breast, cardiovascular system, colon, skin, brain, lung, and reproductive tract in both women and men. The word estrogen is often used in studies when referring to the use of estradiol (E2), the primary circulating estrogen in premenopausal women which is synthesized from cholesterol in the granulosa cells in the ovary in response to luteinizing hormone (LH). Estrone (E1) is the primary estrogen in postmenopausal women, synthesized primarily in adipose from adrenal androgens. E2 and E1 can also be formed locally, e.g., in breast (1) and lung (2).

Lifetime estrogen exposure is widely accepted as a major risk factor for the development of breast cancer (3). Because estrogens have a clear role in the majority of breast cancers and since estrogen receptor α (ERα)is the best prognostic indicator for breast cancer patients and is considered to be the most successful molecular target in the history of cancer drug discovery (4), much is known about the molecular mechanisms of estrogen regulation of transcription.

Data from ENCODE (Encyclopedia of DNA Elements, http://www.nature.com/encode/) revealed that ~ 75 % of the human genome is transcribed while only ~ 1% is protein-coding mRNA, suggesting that other RNA transcripts, including long non-coding RNAs (lncRNAs) and small RNAs (85% of which correspond to four major classes: small nuclear (sn)RNAs, small nucleolar (sno)RNAs, micro (mi)RNAs and transfer (t)RNAs), have regulatory functions (5). Next-generation sequencing (NGS) by RNA sequencing (RNA seq), also called ‘whole transcriptome shotgun sequencing’, is used to identify the transcriptome (6). The transcriptome includes all the RNAs in that source: mRNA, rRNA, and tRNA; and the non-coding RNAs (ncRNAs): miRNAs, enhancer RNAs (eRNAs), endogenous small-interfering RNAs (siRNAs), Piwi-interacting RNAs (piRNAs), and lncRNAs ranging from 1,000 to> 90,000 bases (7). Like miRNAs, siRNAs and piRNAs bind Argonaute family members and base pair with target RNA to cause RNA degradation and/or translation repression (8). LncRNAs are involved in assembly of active e.g., Neat1, or repressed, e.g., Xist, nuclear domains for transcription in a cell-dependent manner (9). This review focuses on estrogen regulation of miRNAs.

miRNAs, first described in 1993, are small (22 nucleotides), single-stranded non-coding, evolutionarily conserved RNA molecules that are related to, but distinct from, small interfering RNAs (siRNAs) which regulate mRNA translation or stability (1012). Comparative genomics analyses have revealed > 45,000 miRNA binding sites within human 3'UTRs that are conserved, indicating that > 60% of human protein-coding genes have been under selective pressure to maintain pairing to miRNAs (13). Compared to transcriptome or microarray analyses identifying miRNA expression patterns in different human cells, tissues, or with various treatments, there are far fewer published reports of estrogen or tamoxifen regulation of miRNAs expression in human cells or tissues (Figure 1). The pace of publication on miRNAs in humans has slowed since 2013 and publication rate on estrogen and human miRNA peaked in 2012 and is in decline. Given the role of estrogens in stimulating breast cancer, it is not surprising that most studies have examined changes in miRNA expression and their correlation with diagnostic markers used in breast cancer therapies, e.g., ERα and tumor grade (1424). Estrogens regulate miRNA expression by both genomic (transcriptional) and non-genomic/membrane-initiated mechanisms of action. Identification and characterization of estrogen-regulated miRNAs and their targets may provide new biomarkers and therapeutic targets in in diseases including breast cancer. There are many online resources about miRNA-mRNA targets recently compiled in http://multimir.ucdenver.edu/ and reviewed in (25).

Figure 1. History of PubMed citations on human miRNA, estrogen AND miRNA, and tamoxifen AND miRNA.

Figure 1

The search terms used were human AND miRNA (black closed circles) and human AND miRNA AND estrogen. Each point is the number of publications in the calendar year indicated. The number of citations was taken directly from an advanced search of PubMed and was not hand-curated to remove non-relevant citations.

2. Genomic ER activities

Transcription is initiated through a complex series of activities occurring through the cooperative interaction of multiple factors at the target gene promoter in association with interactions with other chromatin regions at great distances from the transcription start site- and even on different chromosomes (26). I will use the term ER to refer to either ERα or ERβ or to both subtypes. I will refer to each subtype individually when appropriate to differentiate their established differences. Estrogens bind the ligand binding domains (LBD) of ERα and ERβ which are members of the 48 member steroid/nuclear receptor (NR) superfamily of proteins (27). ERα and ERβ are highly conserved within the DNA binding domain (DBD, C domain), but differ in their N- and C- termini (28).

Crystal structure studies of the LBD of ERα, excluding the F domain, identified 12 alpha helices and found that E2 binding repositions helix 12 that acts as a “switch” controlling accessibility of coregulator interaction site: the ‘coactivator binding groove’ (29).

Chromatin forms a barrier for transcription factor binding. FoxA1, PBX, TLE1, AP2g, and GATA3 act as “pioneer factors” that remodel condensed chromatin to facilitate ERα binding (reviewed in (30)). ERα interacts directly with high affinity to a specific DNA sequence called the estrogen response element (ERE = 5’-AGGTCAnnnTGACCT-3’) (28). ER-ERE binding enhances the recruitment of coactivator/chromatin remodeling complexes resulting in histone modifications, nucleosomal repositioning, increased accessibility to the DNA template for RNA polymerase II interaction, and increased target gene transcription (reviewed in (31, 32)). Chromatin immunoprecipitation (ChIP) of ERα in cell lines, most notably MCF-7 human breast cancer cells, followed by sequencing of the bound DNA(ChIP seq) has established that EREs are located in gene promoters and at great distances from the transcription start site, including in the 3’ flanking regions of regulated genes (3340). Cell-specific ERα cistromes have been identified in ERα-transfected U2OS cells (41), MDA-MB-231 breast cancer cells(40)., and HeLa cells (42). In another example, ERα overexpression in ERα- HeLa cells identified only 9% of common promoter binding sites with MCF-7

In addition to direct ER-ERE binding, ER also activates transcription via a “tethering mechanism” whereby ER interacts directly with transcription factors, e.g. Sp1 (43) and AP-1 (44), bound to their response elements. ERβ binding sites appear enriched for AP-1 sites (45). ChIP-seq, ChIP-PET (ChIP for ERα followed by paired-end tag sequencing) and ChIP-chip experiments identified a number of transcription factor binding sites with which ERα interacts in MCF-7 cells including: AP-1, CEBP, FOXA1, PAX6, RORA, PITX2, and GATA2 (46).

3. Rapid, membrane-initiated, nongenomic estrogen action

In addition to its classical genomic/transcriptional effects mediated by ER-DNA interaction, described above, E2 has rapid “nongenomic, extra-nuclear, or membrane-initiated” effects that occur very rapidly, i.e., within seconds-minutes after E2 administration (reviewed in (47, 48)) These effects are independent and distinct from the genomic, i.e., ER-mediated transcription, activities reviewed in the preceding section. Rapid estrogen-stimulated intracellular activities are mediated by plasma membrane (PM)-associated ERα, ERβ, ERα splice variants: ERα46, ERα36, and/or by an ‘orphan’ G-protein coupled estrogen receptor GPR30/GPER (4960). Palmitoylation of ERα46 helps it to localize to the PM (6164). ERα36 is also recruited to the PM by palmitoylation (65). Evidence of the biological function of PM-associated ERs, including GPER, is supported by experiments in which cell-impermeable E2–bovine serum albumin (E2-BSA) or other E2-conjugates rapidly initiated intracellular kinase cascade activities including MAPK/ERK (p42/p44 MAPKs), endothelial nitric oxide synthase (eNOS), and PI3K/AKT (6675). Increased E2 during pregnancy activates GPER which, with activation of glucagon-like peptide 1 (GLP1) receptor, increases cAMP-PKA and decreases miR-338-3p resulting in increased expression of proliferation and/or anti-apoptotic genes and β-cell proliferation (76). Overexpression of ERα46 stimulates E2-induced endogenous miR-21 transcription and reduced miR-21 targets PTEN and PDCD4 in MCF-7 cells (77). ERα36 and miR-210 expression were correlated in TNBC tumors (78), but to my knowledge, no mechanistic studies have been performed on ERα36 regulation of miRNA transcription.

4. miRNA processing and general activity

The human genome contains ~ 2,588 mature miRNAs (June 2014, http://www.mirbase.org/) (79). The term miRNome is defined as the full spectrum of miRNAs for a specific genome (80). About half of miRNAs are expressed from introns of protein-coding transcripts and miRNAs have 5' and 3' sequence features that form boundaries including transcription start sites, CpG islands, and transcription factor binding recognition elements (81). miRNAs may be differentially processed from the sense and antisense strands of the same hairpin RNA or transcripts from the same locus (82). miRNAs are produced by canonical miRNA processing or noncanonical pathways (83).

The canonical and noncanonical pathways of miRNA biogenesis and the regulation of components of this pathway by miRNAs, phosphorylation, and protein: protein interactions and E2are depicted in Figure 2. miRNAs are transcribed as primary-micro-RNAs (pri-miRNAs) by RNA polymerase II either as independent transcription units or are cotranscribed within introns of pre-mRNAs (84). Pri-miRs are caped and polyadenylated (85). The self-base-pairing stem-loop structure of the pri-miR is cleaved by the microprocessor complex with catalytic Drosha (RNASEN), an RNAse III family endonuclease, and its cofactor DGCR8 (DiGeorge syndrome critical region 8 gene) into shorter (60 to 70 nt) imperfect hairpin-containing precursor-miRNAs (pre-miRNAs) (86). DGCR8 functions as an anchor by binding the pri-miRNA to direct cleavage by Drosha 11 bp from the dsRNA-ssRNA junction (84). The Drosha microprocessor also binds and regulates other cellular RNAs (84) and includes other proteins and hnRNPs shown in Figure 2: EWSR1, FUS< Nucleolin, p68, p72 which interacts with YAP2.

Figure 2. Model of canonical miRNA biogenesis and function.

Figure 2

Primary transcripts of microRNAs (pri-miRNAs) are transcribed by RNA polymerase II, processed by the RNAse III enzyme, Drosha and its cofactor DGCR8, to precursor microRNAs (pre-miRNAs) which are exported from the nucleus by Exportin/RAN-GTP (85). In the cytoplasm, pre-miRNAs are processed by the Microprocessor complex that includes Dicer, an RNAse III enzyme, to form mature ~22 nt transiently double-stranded miRNA duplexes that are transferred to Argonaute proteins (most notably AGO2 in the RNA-induced silencing complex (RISC), leading to unwinding of the duplexes to form single stranded miRNAs. The RISC complex binds either to the 3’ untranslated region (3’ UTR) or to the open reading frame (ORF) of its target mRNA. Binding of miRNA/RISC complex with the 3’UTR causes translational repression (18).

Exportin and Ran-GTP or CRM1 export pre-miRNAs from the nucleus. In the cytoplasm, pre-miRNAs are cleaved to the mature ~22 nt transiently double-stranded miRNA duplexes by the RNAse III enzyme Dicer. Dicer with its associated cofactors TRBP (TAR (transactivating response) RNA-binding protein) and PACT (protein activator of the interferon-induced protein kinase) transfers the miRNA to the RNA-induced silencing complex (RISC) containing the catalytic Argonaute proteins (AGO1, AGO2, AGO3, and AGO4 (87)) which unwind the duplexes to form single stranded miRNAs. One strand miRNA is preferentially selected to bind one of the AGO proteins and by base-pairing directs translational inhibition and/or mRNA degradation by binding either to the 3’ untranslated region (3’ UTR) or to the open reading frame (ORF) of its target mRNA (8891). AGO2 is the catalytic component of RISC. Dicer binds not only miRNAs, but also tRNAs, snoRNAs, mRNA and promoter RNAs (92). The widespread reduction of miRNAs in cancers is considered to be the result of defective miRNA processing as reflected in increased pri-miRNAs due to Hippo signaling regulation of p72 nuclear function by YAP sequestering p72 from the Microprocessor in a cell-density-dependent manner (93).

The non-canonical pathways of miRNA generation include the generation of mirtrons which are short hairpin pre-miRNAs directly produced by splicing, thus bypassing Drosha-mediated cleavage (94, 95). Some miRNAs function as bimodal miRNAs controlling different target gene sets depending on the region used for interaction. i.e., a canonical seed in positions 2–8 or positions nt 6–12, e.g., miR-4728-3p, encoded in intron 24 of HER2 gene (96) which downregulates ESR1 expression through an internal seed interaction (97).

Just like protein-coding genes, complexity of the miRNome has increased with further research. miRNAs are heterogeneous in length and sequence with isomiRs that are sequence variants of the canonical miRNA currently in the miRBase generated from a single miRNA locus by template and non-template variants (98). Templated isomiRs match the genomic sequence, but have different 5′-start and/or 3′-ends, resulting from imprecise Drosha or Dicer cleavage (99), whereas non-templated isomiRs diverge from the genomic sequence due to post-transcriptional enzymatic modification. The most common non-templated modification is adenylation, catalyzed by the adenosine deaminase (ADAR) family of enzymes (100). The expression isomiRNAs is dynamic, with differences between cell types and tissues. A tool called IsomiRage http://cru.genomics.iit.it/Isomirage/ is available for profiling the miRNAs/isomiRs and corresponding differential expression patterns using Illumina next-generation sequencing datasets of small RNA (99). When applied to primary breast normal and cancer cells the IsomiRage increased the number of detected miRNA species by ~40%, thus revealing additional information “hidden” in sequencing datasets (99). These isomiRNAs are effectively loaded on AGO/RISC complexes and thus are thought to function as canonical miRNAs, thus increasing the repertoire of mRNA targets.

Not only are miRNAs active in the cells in which they are transcribed, but miRNAs circulate in exosomes: 40–100 nM membrane-bound vesicles composed of different growth factors, cytokines, lipids, cytoplasmic proteins, and nucleic acids, including miRNAs, which circulate in the blood and lymph and deliver molecules between tissues (101). The exosomal content is tightly regulated by endosomal sorting complexes required for transport (ESCRT) (102). Specific cell surface markers allow cellular uptake of exosomes with high specificity. The physiological role of exosomes is controversial. Exosomes can facilitate tumor progression by supplying tumor niches with factors that favor proliferation, invasion, drug resistance, and metastasis (101). Circulating miRNAs embedded in exosomes reprogram cellular mechanisms in recipient cells (103, 104). Whether exosomal miRNAs will be makers in cancer is currently speculative. A recent study appears to be the first comparison between cell-free and exosomal miRNAs in breast cancer patients and healthy women (105). The authors reported higher exosomal miR-372 and cell free (not exosomal) miR-373 in triple negative breast cancer compared to luminal breast cancer patients and higher cell free miR-101 in both groups (102).

5. miRNA-mRNA interaction

The critical, perfectly complementary basepairing between 7 to 8 nucleotides at the 5’ end of the miRNA and its target mRNA is referred to as the ‘seed sequence’. Base pairing of the miRNA-RISC complex within the ORF requires almost perfect complimentarity and the mRNA is either degraded or translation is blocked (85). RNA binding proteins (RBP), e.g. HuR, hnRNP E1, and hnRNP L, and miRNAs compete and collaborate to regulate mRNA stability and RBPs can recruit miRNA-containing RICSs to target lncRNAs (106). There is evidence that miRNA-mRNA gene silencing occurs in the rough endoplasmic reticulum (RER) by interaction of components of Dicer, TRBP and PACT with the RER (107).

Most commonly, because of imperfect base pairing between the miRNA and the 3’UTR, the RISC complex causes translational repression by interaction with eIF6 which prevents 80S ribosomal assembly (108) or by inhibition of translation (18). The exact mechanisms of translational inhibition versus mRNA degradation have not yet been fully elucidated (109). miRNAs initiate target mRNA degradation by recruiting mRNA decay pathway effectors such as de-adenylation and de-capping enzymes (110). The miRNA-containing ribonucleoprotein particle (miRNP)-silenced mRNA is directed to the P-bodies and the mRNA is either released from its inhibition upon a cellular signal and/or actively degraded (111). Some miRNAs may also increase translation of select mRNAs in a cell cycle-dependent manner (112).

miRNAs are considered highly stable, although this is cell-type, cell cycle, and miRNA- specific; further target regulation can promote miRNA’s 3’-end uridylation and degradation (106). This means that an increase in target mRNA leads to a decrease in its target miRNAs. miRNAs are regulated by competing endogenous RNAs (ceRNAs) (113) which contain miRNA target sties and thus act as miRNA ‘sponges’ and sequester miRNAs from interaction with target mRNAs. Circular RNAs (circRNAs) are ceRNAs that contain miRNA binding sites and are resistant to miRNA-mediated destabilization (reviewed in (114)). Multiple non-coding RNA species, including sncRNAs, pseudogenes, lncRNAs and circRNAs appear to possess ceRNA activity (114).

miRNAs have important roles in regulating cellular processes including replication, differentiation, and apoptosis. In cancer, miRNAs can either act as ‘oncosuppressor miRNAs’ which are often downregulated in cancer, e.g., the miR29b-1/a in acute myeloid leukemia resulting in upregulation of oncoprotein BCL-2 (115),or, as ‘oncomiRs’, by decreasing the levels of tumor suppressor proteins, e.g., miR-21 decreasing PDCD4 (116). MiRNAs are expressed in a tissue-specific manner (117). Each miRNA targets ~ 200 transcripts directly or indirectly (118), but the bone fide physiological targets of the vast majority of miRNAs remain to be experimentally verified.

6. HITS-CLIP to identify miRNA-mRNA interaction by Ago2 immunoprecipitation

High-throughput RNA-seq isolated by crosslinking immunoprecipitation (HITS-CLIP) of Argonaute 2 (Ago 2, catalytic component of the RISC complex (119)) is used to identify putative miRNA-mRNA ternary complexes (120, 121). HITS-CLIP of E2-treated MCF-7 cells revealed Ago 2 footprints throughout ESR1 mRNA, including peaks in the 3’UTR and within the coding region, and follow-up experiments identified miR-9-5p binding the 3’ UTR, directly downregulating ERα protein levels (122).

7. Nomenclature of miRNA

miRNAs are preceded a three lettered prefix indicating the species of origin e.g., hsa for homo sapiens and mmu for mouse (123). miRNAs originating from different genomic loci are assigned a numerical suffix, i.e, hsa-miR-29b-1 and hsa-miR-29b-2. If transcripts are equally expressed they are referred to as miR-21-5p (from the 5’ arm) and miR-21-3p (from the 3’ arm) arise from the same hairpin precursor. Alternatively, miR-21* indicates the less predominant species in RISC (124). miRNAs differing by a few bases are given a lettered suffix, e.g.,miR-125a and miR-125b. miRNA families arise from a common ancestor and have similar sequences, e.g. miR-221 and miR-222 family. 61% of mammalian miRNAs are expressed from polycistronic clusters. reflecting shared biological functions for unrelated miRNAs in the same primary transcript (125). miRNA clusters arise due to gene duplication, e.g., the miR-200 cluster of miRNAs are located in two chromosomes, i.e., miR-200a, miR-200b, and miR-429 are located on chromosome 1 and miR-200c and miR-141 are located on chromosome 12 (126). Each cluster is transcribed into a common precursor RNA.

8. Regulation of miRNA expression

Levels of mature miRNA are regulated transcriptionally and by processing of pri-miRNAs and pre-miRNAs. In the microprocessor complex the ratio of Drosha and DGCR8 are tightly regulated (127). DGCR8 stabilizes Drosha and Drosha cleaves and inactivates DGCR8; providing a tight feedback loop (128). ERα interacts directly with helicases p68 and p72 (which are established ERα coregulators (129)). ERα-p68 interaction was reported to inhibit Drosha complex formation (130), and thus repress pri-miRNA processing. Importantly, this work was recently retracted (131). However, another group of investigators also reported that Drosha and p68/DDX5 could be co-purified with ERα in MCF-7 cells, but not with ERβ in ERβ-stably transfected MCF-7 cells (132). This report has not been confirmed.

Dicer processes pre-miRNA to mature miRNA. Dicer activity is enhanced by MAPK-phosphorylation of TRBP (Figure 2) which promotes miRNA processing (133). The RNA coactivator SRA (steroid receptor RNA activator) binds Dicer complex components PACT, TRBP, and PKR in various cell lines and also binds NRs, including ERα (134). Dicer acts as a NR coactivator in MCF-7 cells and is recruited to the PSA gene promoter in DHT-treated LNCaP prostate cancer cells with androgen receptor (AR) (134). These findings suggest that pre-miR processing may be coupled with ERα and AR regulation of gene transcription.

AGO2 is the catalytic component of the RISC complex and serves as a platform to recruit additional regulators of mRNA stability (125). AGO2 is regulated at the transcriptional and post-transcriptional level. For example, in MCF-7 breast cancer cells, E2 inhibits AGO2 expression by activating epidermal growth factor (EGF)-MAPK signaling (135). Direct interaction of EGF receptor (EGFR) with AGO2 in the cytoplasm phosphorylates AGO2 at Tyr 393 which reduced AGO2 association with Dicer (Figure 2) and TRBP suppresses maturation of specific tumor suppressor miRNAs under hypoxic conditions (136).

Nucleolin is a multifunctional protein concentrated in the nucleolus, but located throughout the cell, including the plasma membrane, and has roles in transcription, ribosome biogenesis, DNA replication, chromatin remodeling, apoptosis, and macropinocytosis (137, 138). There are several examples of nucleolin functioning as a transcription factor or as a coregulator through its interactions with other proteins (reviewed in (139)). Nucleolin was reported to promote the maturation of specific miRNAs implicated in carcinogenesis in MCF-7 and HeLa cells: miR-21, miR-103, miR-221, and miR-222 (140).

9. Estrogen regulation of miRNA expression overview

Regulation of miRNA expression by estrogens in animals, fish, and humans has been reviewed by us (141, 142) and others (143). Since my previous review, a non-inclusive list of new studies of E2 regulation of miRNA expression in animals includes: female Fischer 344 rat brain, specifically in the ventral and dorsal hippocampus, central amygdala, and paraventricular nucleus and as a function of aging (144); in female ACI rats in an E2-induced mammary carcinogenesis model (145); mouse aorta (146); mouse liver and primary murine hepatocytes (147); rat cardiac fibroblasts (148). I will not review these studies, but will focus on human cell lines and tissues.

10. ERα and ERβ regulate miRNA expression in a ligand-independent manner

ChIP studies have shown that ‘unliganded’ ERα (149)and ERβ (150) bind DNA in cells grown in serum-free or charcoal-stripped serum medium. Overexpression of ERα in MCF-7 cells upregulated mIR-17 (151). Overexpression of ERβ in non-hormone treated MCF-7 and ZR-75.1 human breast cancer cell lines was reported to regulate the expression of > 450 miRNAs in next-gen RNA sequencing experiments (152). Here I will focus on updating reports on ER ligand-responsive regulation of miRNA expression in human cell lines and tissues.

11. E2 and other ER ligands regulate miRNA expression in human cell lines and tissues

The hope of current studies of E2 regulation of miRNA expression in breast cancer cell lines is that identification of E2-regulated miRNAs and their gene targets may offer insight into mechanisms of estrogen in breast carcinogenesis and progression and identify targets for therapeutic interference. By far and large, E2 regulation of the transcriptome, including miRNAs is best characterized in breast cancer cell lines with MCF-7 studies predominant. This will be apparent in Tables 1 and 2 which summarize the regulation of miRs and their bona fide targets by ER ligands including E2, tamoxifen, 4-OHT, and endocrine disruptors in human cell lines and tissues. It is worth noting that there are conflicting results of E2 and other ER ligand regulation of miRNAs within cell lines, e.g., MCF-7 and T47D, between reports from different investigators and even within the same lab group in different publications. There are many likely explanations for these differences including cell lines and variations in cell treatment conditions, circadian regulation of ERα expression (153), normalization of data (154), and control genes used for qPCR (155).

Table 1. miRNAs upregulated by estradiol (E2), tamoxifen (TAM), 4-hydroxytamoxifen (4-OHT), Fulvestrant (ICI 182,780), or endocrine-disrupting chemicals (EDC) in animal studies and human cell lines.

The bona fide targets of the miRNAs are experimentally proven in the reference cited; however, this direct targeting is not necessary substantiated in E2 regulation in the cells indicated in column 3. DIANA-TarBase v7.0 (228) web site has a list of bona fide targets of miRNAs : http://diana.imis.athena-innovation.gr/DianaTools/

miRNA Ligand Human cell line/tissue Comments Bona fide targets
Let-7a,b,c,d,e,f, g, i E2 MCF-7 cells stably expressing a biscistronic vector control (157). MCF-7 cells (141, 229).

1 µM E2 in Ishikawa and ECC-1 ERα+ human endometrial cancer cells (158).

Let-7a and let-7f-1* were increased at 6,12, and 72 h but decreased at 24 h with 10 nM E2 in MCF-7 cells (159).

Let-7a* was increased in response to 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
Oncosuppressor miR- stimulate apoptosis (230) DICER1 (231); let-7g:COL1A2 (232)
miR-7 E2 10 nM E2 MCF-7 cells (141, 233) oncomiR XRCC2 (234)
KLF4 (235)
miR-10a
miR-10b
E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236). miR-10b is down-regulated in breast tumors and upregulated in sera (237). BUB1, PLK1, CCNA2 (238)
miR-15a E2 10 nM E2 MCF-7 cells (141). Upregulated by E2F1 (239). CCNE1 = CyclinE (239)
miR-16-1* E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-16-2* E2 10 nM E2 for 24 h in T47D cells (154).
miR-17* E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-17-3p E2 MCF-7 stably transfected to overexpress the aromatase gene (MCF-7aro) (240).
miR-17-92 E2 MCF-7 cells (233, 241, 242). miR-17-92 cluser encodes miR-17, 18, 19, 20, 19b-1, 92-1 miR-19a and miR-92a: PTEN (243)
miR-18a E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-18a* E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-18a is higher in ERα-breast tumors (244) ERα (241)
miR-18b E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159).

10 nM E2 for 6, 12 h in MCF-7 cells stably overexpressing inducible ERβ or ERα-downregulated at 24 and 72 h (132).
miR-18b* BPA 10 µM BPA for 18 h in MCF-7 cells (161)
miR-19a, 19b E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)

miR-19a and 19a* were increased by 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-19b-1 E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-19b E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-20a* E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-21 Fludioxonil fenhexamid

4-OHT
MCF-7 cells (176)

MCF-7 cells (156)
oncomiR

Fludioxonil and fenhexamid are endocrine disruptors
NFIB (245); PTEN, PDCD4 (156); RASA1 and RASA2 (148)
miR-22 E2

EDC
1 nM E2, 1 µM triclosan or 1 µM triclocarban for 18 h in MCF-7 cells (178). EDC
miR-23b* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ but not ERα (132).
miR-24 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-24-1* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ but not ERα (132).
miR-25 E2 MCF-7 cells (141, 233). miR-106b-25 cluster encodes miR-106b, miR-93, and miR-25in the 13th intron of the MCM7 gene (246) BIM (247); DR4 (248); MCU (249); Smad7 (250); LATS2 (251); RECK (252)
miR-25* E2 10 nM E2 12 and 24 h in MCF-7 and ZR-75-1 cells (159).
miR-26a E2 and fulvestrant Primary human myometrial smooth muscle cells (MSMC) (253) Oncosuppressor miR ESR1 (254)

CHD1, GREB1, and KPNA2 (255)
miR-27a E2 1 µM E2 in Ishikawa and ECC-1 ERα+ human endometrial cancer cells (158). OncomiR EGFR (256)
miR-27b E2 MCF-7 cells (233). Oncosuppressor miR Sp1 (257); LIMK1 (258); PPARγ (259)
miR-29a E2 MCF-7 cells (233). OncomiR: stimulates migration and invasion; Repressed by c-myc, YYI, NFκB, CEBPA and stimulated by p53 (260) BCL2, CDC42, CDK6, DNMT, MCL1, Osteonectin, TGFβ3m, TTP, TGF-β1, TGF-β2, TTP (260)
miR-29b-2* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ but inhibited by ERα (132).
miR-29c E2 10 nM E2 for 24 h in T47D cells (154).
miR-30b E2 MCF-7 cells (141) Oncosuppressor miR CCNE2(261); KRAS, PIK3CD and BCL2(262)
miR-30d E2 1 µM E2 in Ishikawa ERα+ human endometrial cancer cells (158).

10 µM BPA for 18 h in MCF-7 cells (161).
miR-32 E2 10 nM E2 72 h in MCF-7 cells stably overexpressing inducible ERβ (132).
miR-33a E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-92 E2 10 nM E2 24 and 72 h in MCF-7 cells (159)
miR-92a E2 1 µM E2 in ECC-1 ERα+ human endometrial cancer cells (158).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-92a-1* E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)
miR-92b E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-93 E2 10 nM E2 24 h in MCF-10A and T47D cells (263). 1 nM E2 for 18 h in MCF-7 cells (161).
miR-98 E2
BPA
MCF-7 cells (141).
10 µM BPA for 18 h in MCF-7 cells.
miR-99b E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-101 E2 10 nM E2 24 h in MCF-7 cells (264).
miR-101* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ (72 h) but not ERα (132).
miR-103 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-122 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-124 E2 MCF-7 cells (233). Oncosuppressor miR Ets1 (265)
miR-124-5p: LAMB1 (266)
ROCK1 (267)
FLOT1(268)
SphK1 (269)
CD151 (270)
iASPP (271)
Slug (272)
miR-130b E2 MCF-7 cells (242). TP53INP1 (273); DICER1 (274)
miR-135a E2 10 nM E2 6 h in MCF-7 cells (264)

10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)
miR-135b E2 10 nM E2 for 6 and 72 h in ZR-75-1 cells, but no change at 12 or 24 h (159).
miR-142-3p E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-148 E2 MCF-7 cells (233). miRNA-148/152 family include miR-148a, miR-148b, miR-152 (275) PXR, DNMT1, CAND1, BCL2, p27, ACVR1, PETN, WNT10B, MSK1, CDC25B, ROCK1, CCKBR, CCK2R, IGF-1R, IRS1 (275)
miR-149 E2 MCF-7 cells (233). GSK3α (276)
GIT1 (277)
AKT and E2F1 (278)
miR-151-5p E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-155 E2 100 nM E2 for 48 h in MCF-7 cells (279).

Higher levels circulating in the serum of breast cancer patients than healthy women (280).
oncomiR TRF1 (281).
TP53INP1(282)
miR-181a E2 1 µM E2 in Ishikawa ERα+ human endometrial cancer cells (158).
miR-181d E2 MCF-7 cells (233) CCND1 (245)
miR-186 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-190 E2 10 nM E2 for 6, 12, and 72 h in ZR-75-1 cells, but not 24 h (159).
miR-190a E2 100 nM E2 in MCF-7 cells increased ERα recruitment to the miR-190a promoter containing a half-site ERE (283). PAR-1 (283)
miR-190b E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-191 E2 10 nM E2 for 6 h in MCF-7 cells (284).

10 nM E2 (24 h) stimulation was inhibited by 100 nM tamoxifen and by siERα and siERβ in MCF-7 cells (285). ERα and ERβ ChIPped to the miR-191 promoter in MCF-7 cells (285). 1 nM E2 for 18 h in MCF-7 cells (161).
EGR1 (284)

CDK6, BDNF, and SATB1 (285)
miR-193a-5p E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-193b E2

EDC
MCF-7 cells (242)

1 nM E2, 1 µM triclosan or 1 µM triclocarban for 18 h in MCF-7 cells (178).
uPA (286); YWHAZ, SHMT2, AKR1C2 (287); miR-193-3p: MYB (288)
miR-194 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-195 E2 MCF-7 cells (141) CCND1 (245)
miR-195* E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells- highest at 6 h (159). ASF1B, BIM, BCL2L2, CCL5, CADM1, EZH2, FGF$1, HDGF, LTF, MAP2K3, NRAS, PTEN, TP53, TWIST1, XBP1 (and others) (289)
miR-196a2* E2 10 nM E2 6 h in MCF-7 cells (264) Mediated by ERα and the protein kinase ERK2 (264). By ChIP assay, both ERα and ERK2 were recruited to chromatin with 45 min 10 nM E2 alone with increased pSer5 RNA pol II recruitment (264). TP63 (264)
miR-198 E2 10 nM E2 for 24 h in T47D cells (154).
miR-199a/b-3p E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells, but not at 6h (159).
miR-199a-5p E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-200a E2 MCF-7 cells (141) BAP1, PTPRD, KLF11, SEPT7, HOX5B, ERBB2IP, RASSF2, ELMO2, SHC1, VAC14 (DIANA)
miR-200c none Endogenous ERα in MCF-10A cells ChIPed to the miR-200c promoter and Overexpression of ERα in MCF-10A cells increased miR-200c expression (290).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-203 E2 MCF-7 cells (141)
miR-205 E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).
miR-206 DPN

E2

EDC

ERβ-selective agonist in MCF-7 cells (181).

1 nM E2, 1 µM triclosan or 1 µM triclocarban for 18 h in MCF-7 cells (178)
Oncosuppressor miR
miR-210 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-216a E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-219-5p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-222 E2

BPA
1 nM E2 or 10 µM BPA for 18 h in MCF-7 cells (161). KIT (291); PPP2R2A (292); CDKN1C (293); CDK1B (294); DICER1 (229)
miR-223 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-301b E2 MCF-7 cells (242)
miR-320 E2 1 µM E2 in Ishikawa and ECC-1 ERα+ human endometrial cancer cells (158).
miR-320a E2 1 nM E2 or 10 µM BPA for 18 h in MCF-7 cells (161).
miR-320c E2

BPA
1 nM E2 or 10 µM BPA for 18 h in MCF-7 cells (161).
miR-330-5p E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ not ERα (132).
miR-335 E2 10 nM E2 for 6, 12 and 72 h in MCF-7 and ZR-75-1 cells, but not at 24 h (159).
miR-342 E2; Not blocked by 1 µM 4-OHT MCF-7-HER2 cells, MCF-7 cells stably overexpressing HER2, but still tamoxifen-sensitive (220)
miR-363 E2 10 nM E2 for 12 and 24 h in ZR-75-1 cells, but not 6 or 72 h (159).
miR-365 E2 MCF-7 cells (141)
miR-374a* E2 10 nM E2 for 6, 12 and 72 h in MCF-7 and ZR-75-1 cells, but repressed > 1.5 fold at 24 h (159).
miR-375 E2 10 nM E2 for 24 and 72 h in ZR-75-1 cells, but not 6 or 12 h (159).
miR-376b E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells- highest at 6 h (159).
miR-423-5p E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-424 E2 MCF-7 cells (165)

10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-424* E2 10 nM E2 for 6, 12 and 72 h in MCF-7 and ZR-75-1 cells, but not at 24 h (159).
miR-425 E2 1 µM E2 in Ishikawa and ECC-1 ERα+ human endometrial cancer cells (158).

10 nM E2 for 6 h in MCF-7 cells (284).
EGR1 (284)
miR-449a E2 10 nM E2 for 6, 12and 24 h in ZR-75-1 cells, but not 72 h (159)
miR-450b-3p,5p E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells- highest at 72 h (159)
miR-455-5p, 455-3p E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159)
miR-484 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-489 E2 10 nM E2 12, 24, and 72 h in MCF-7 and ZR-75-1 cells, but not at 6 h(159)
miR-491-3p E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-499-5p E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-515-5p tamoxifen 100 nM tamoxifen for 48 h ~ 25% decrease in MCF-7 cells (295). SK1 (295)
miR-520d E2 MCF-7 cells stably expressing a constitutively active AKT (157)
miR-542-5p E2 10 nM E2 for 72 h in MCF-7 cells (159)
miR-542-3p E2 10 nM E2 for 72 h in MCF-7 and ZR-75-1 cells (159)
miR-548d-3p E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ, but not ERα (132).
miR-548e E2 10 nM E2 for 6, 24, and 72 h in ZR-75-1 cells (159).
miR-550 E2 10 nM E2 for 72 h in MCF-7 cells (159).
miR-556-5p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells, but not at 24 h (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-560:9.1 E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-564 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-574-5p E2

PhIP
1 µM E2 in Ishikawa ERα+ human endometrial cancer cells (158).

10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-574-3p E2 or PhIP 10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-579 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159)
miR-590-3p E2 10 nM E2 highest stimulation at 6, 12 and 72 h in ZR-75-1 cells with no change detected at 24 h (159)
miR-594:9.1 E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 cells (159)
miR-615-3p E2 10 nM E2 6 h in MCF-7 cells (264)
miR-628-5p E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells (159).
miR-638 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-643 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-651 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-652 E2 10 nM E2 for 24 and 72 h in ZR-75-1 cells, but not at 6 or 12 h (159).
miR-653 E2 10 nM E2 for 72 h in MCF-7 and ZR-75-1 cells(159).
miR-653:9.1 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-660 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-663 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-663b E2 10 nM E2 for 6 and 24 h in ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-708 E2 10 nM E2 for 12, 24, and 72h in ZR-75-1 cells, but not at 6 h (159)
miR-720 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-760 E2 24 h and 3d in MCF-7 cells (165).

10 nM E2 for 24 and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-886-3p E2 10 nM E2 for 24 h in MCF-7 and ZR-75-1 cells, but not at 6, 12, or 72 h (159).
miR-938 E2 10 nM E2 for 6 h in MCF-7 cells (66).
miR-939 E2 10 nM E2 for 72 h in MCF-7 cells (159)
miR-940 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-942 E2 10 nM E2 for 72 h in MCF-7 and ZR-75-1 cells, but not 6, 12, or 24 h (159).
miR-944 E2 10 nM E2 for 6 h in MCF-7 cells (66)
miR-1206 E2 10 nM E2 for 72 h in MCF-7 cells (159)
miR-122 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-1248 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-1268 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159),
miR-1275 E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-1305 E2 10 nM E2 for 12 and 72 h in MCF-7 and ZR-75-1 cells (159)
miR-1323 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1826 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1915 E2
BPA
1 nM E2 for 10 µM BPA for 18 h in MCF-7 cells (161).

Table 2. Estradiol- and tamoxifen- inhibited miRNAs.

This table lists miRNAs whose expression is decreased by E2, tamoxifen, or 4-OHT. MCF-7, T47D, ZR-75-1, BT-474, and BG1 are ERα positive breast cancer cells.

miRNA Ligand Species/tissue/cell line Comments Bona fide
targets
Let-7g, -7f, -7a, -7c E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).10 nM E2 6 h in MCF-7 cells (141).

Let-7g in MCF-7 cells (297).

10 nM letrozole stimulated Let-7 expression in MCF-7 cells co-cultured with primary human stromal cells (167).

1 nM E2 for 18 h in MCF-7 cells (161).
Blocked by fulvestrant GAB2; FN1 (297)
Let-7b E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
Let-7f 4-OHT 1 µM 4-OHT for one month in MCF-7 cells (298)
Let-7i E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-7-1 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-9, miR-9-d E2 10 nM E2 for 24 h in ER β stably expressing SW480 colon cancer cells (236).
miR-15a* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-16 E2 10 nM E2 for 6, 24, and 48 h in MCF-7 cells; blocked by pretreatment with 1 uM ICI 182,780 (299).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-16-1* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-17 E2 10 nM E2 for 24 h ER β stably expressing SW480 colon cancer cells (236). Oncosuppressor miR206
miR-17* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ but increased by ERα (132).
miR-18a, miR-18b E2 10 nM E2 24 h ER β stably expressing SW480 colon cancer cells (236)
miR-19a, 19b E2 10 nM E2 24 h ER β stably expressing SW480 colon cancer cells (236).
miR-20a E2 24 h 10 nM E2 in isolated human endometrial glandular epithelial cell; blocked by ICI 182,780 (300).

10 nM E2 for 24 h ERβ stably expressing SW480 colon cancer cells (236).
miR-21 E2 24 h 10 nM E2 in isolated human endometrial glandular epithelial cells and in Primary human leiomyoma smooth muscle cells (LSMC) (253)

10 nM E2 for 48 h in MCF-7 cells (296) (181). 10 nM E2 6 h: ~ 60% reduction in miR-21 in MCF-7 cells (156)

10 nM E2 for 12 or 24 h in MCF-7 cells (264). 10 µM E2 for 24 h in MCF-7 cells, no effect in MDA-MB-231 cells (301). 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159). 10 nM E2 or 100 nM PhIP for 24 h in MCF-7 cells (160). 1 nM E2 for 18 h in MCF-7 cells (161).
blocked by ICI 182,780 isolated human endometrial glandular epithelial cells



ERα or ERK2 knock-down reduced E2-downregulation of miR-21 expression(264)
PTEN, PDCD4 (156)




JAG1 (301)
miR-22, 22* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-23a.
23b
E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).

miR-23a: 10 nM 3 h in MCF-7 cells (302) and 10 nM E2 for 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-24 E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296)
miR-25 E2 10 nM E2 for 24 h ERβ stably expressing SW480 colon cancer cells (236).
miR-26a E2 24 h 10 nM E2 LSMC(253).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-26a-2* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-26b E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).

10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-27a* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-27b E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).

10 nM E2 for 72 h in MCF-7 cells (159)
Oncosuppressor miR
miR-29a E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-29a* E2 10 nM E2 6, 12, 24, and 72 h in MCF- 7 cells (159)
miR-29b-1*, 29b-2* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)
miR-30a E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ (132). ERβ ChIPed to the promoter (132).
miR-30c-2* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-30d E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132). ERα was more inhibitory than ERβ.
miR-34a E2 10 nM E2 for 24 h MCF-7 cells (303)

10 nM E2 for 6 h in HUVEC, LNCaP, C38IM, and C27IM human prostate cancer cells (304).

Higher levels circulating in the serum of breast cancer patients than healthy women (280).

10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)
Oncosuppressor miR- stimulate apoptosis (230) LMTK3 (303)

SIRT1 (305)
miR-92a E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-99a E2 10 nM E2 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159)
miR-99b
E2
10 nM E2 for 6,12, 24 and 72 h in ZR-75-1 cells, most repressed at 72 h (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-105-2 4-OHT 1 µM 4-OHT for one month in MCF-7 cells (298)
miR-106 E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-106b E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-107 E2 10 nM E2, for 6, 12, 24 h and 3 d in MCF-7 cells (306).

10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-125a-3p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159). Oncosuppressor miR
miR-125a 4-OHT 1 µM 4-OHT for one month in MCF-7 cells (298)
miR-125b-2* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159). Oncosuppressor miR BAK1, BCL2, DICER1, ERBB2, ERBB3, ETS1, FGFR2, IL6R, JUN, LIN28A, LIN28B, MCL1, MUC1, NCOR2, SIRT7, STAT3, TNF, TP53 (and others)(289)
miR-128a:9.1 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159). oncomiR
miR-130b* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-132* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-135a E2 10 nM E2 for 24 h ERβ stably expressing SW480 colon cancer cells (236).

10 nM E2 24 h in MCF-7 cells (66).
miR-139-5p E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-140 E2 10 nM E2 for 24 h in ERα-stably transfected MCF-10A cells (307).

ERα binds the miR-140 promoter in E2 or BPA-treated MCF_7 cells.
SOX2 (307)
miR-140-5p E2 10 nM E2 for 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-141 E2 or PhIP 10 nM E2 or 100 nM PhIP for 24 h in MCF-7 cells (160).
miR-142-3p E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-143 E2 10 nM E2 for 6, 24, and 48 h in MCF-7 cells; blocked by pretreatment with 1 uM ICI 182,780 (299).
miR-148b* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-149 E2 10 nM E2 6 h in MCF-7 cells (141)
miR-142-3p E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-146b-5p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-181a, 181b, 181d E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).

miR-181a and 181b inhibited by 100 nM E2 in MCF-7 cells (163).
miR-181 4-OHT 100 nM 4-OHT for 6 h in MCF-7 cells (155).
miR-181a*, 181c* E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).

miR-181c* 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-181c E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-183 E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-185* E2 10 nM E2 for 12 and 72 h in ZR-75-1 cells, but not 6 or 24 h (159).
miR-186 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ, but increased by ERα (132).
miR-192 E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-193a E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).
miR-193a-3p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159)
miR-193b* E2 10 nM E2 for 72 h in ZR-75-1 cells (159).
miR-194 E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).
miR-194b* E2 10 nM E2 for 72 h in ZR-75-1 cells (159).
miR-196a E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).
miR-196b
E2
10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-199a/b-3p E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ but increased by ERα (132).
miR-199b-5p E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα-except that 24 h of E2 increased miR-199b-5p in ERα-MCF-7 cells (132).
miR-200a E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)

10 nM E2 6 h MCF-7, LCC1, and LCC2 breast cancer cells (214).
miR-200b E2

4-OHT
10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).

10 nM E2 6 h MCF-7, LCC1, LCC2, and LCC9 breast cancer cells (214).

500 nM 4-OHT for h in ECC-1 and Ishikawa endometrial cancer cells (308).
4-OHT induced c-Myc that inhibited miR-200a, miR-200b, and miR-429 transcription (308).

miR-200b promoter P2 is hypermethylated in primary breast tumors and ERα-negative cell lines (309).
ZEB2 (308)
miR-200c E2

4-OHT
10 nM E2 for 6 h in MCF-7 cells (141)

10 nM E2 for 6 h MCF-7, LCC1, LCC2, and LCC9 breast cancer cells (214). 500 nM 4-OHT for h in ECC-1 and Ishikawa endometrial cancer cells (308).
ZEB2 (308)
miR-203 E2 10 nM E2 for 6, 24, and 48 h in MCF-7 cells; blocked by pretreatment with 1 uM ICI 182,780 (299).

1 nM E2 for 18 h in MCF-7 cells (161).
miR-204 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-205 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159). Oncosuppressor miR
miR-206 1 nM E2 or 10 nM PPT (an ERα-selective agonist) MCF-7 cells (181). 80% reduction in expression with 24 h treatment
miR-218 E2 10 nM E2 for 24 and 72 h in MCF-7 cells (159).
miR-220c E2 10 nM E2 for 24 h in T47D cells (154).
miR-221 E2 10 nM E2 for 24 h ~ 80% reduction in MCF-7 and T47D cells (294).

Repressed by ERα knockdown

10 nM E2 48 h in MCF-7 cells (202).

10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236).

1 nM E2 for 18 h in MCF-7 cells (161).
pro-metastatic/pro-proliferative ESR1 = ERα (reviewed in (310))
miR-221* E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells (159).
miR-222 E2

BPA
10 nM E2 for 24 h ~ 80% reduction in MCF-7 and T47D cells (294).

Repressed by ERα knockdown

10 nM E2 for 48 h in MCF-7 cells (202).
miR-223 E2 10 nM E2 for 3 h in MCF-7 cells (302)
miR-301a E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-320b

miR-320d
E2 1 nM E2 for 18 h in MCF-7 cells (161).
miR-328 E2 10 nM E2 6 h in MCF-7 cells (141).

10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-330-5p E2



PhIP
10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).

10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-338-3p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159). 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-342 E2 10 nM E2 for 6 h in MCF-7 cells (141).
miR-345 E2 10 nM E2 for 72 h in ZR-75-1 cells (159).
miR-362-5p E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-365 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-374b* E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-375 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-376a E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-377 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-379 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-429 4-OHT 500 nM 4-OHT for h in ECC-1 and Ishikawa endometrial cancer cells (308).
miR-451 tamoxifen 1 µM tamoxifen repressed by 4 h and 90% at 24 h (311). Expression ~ 2-fold lower in tamoxifen-resistant MCF-7 cells (311)
miR-487b E2 10 nM E2 for 6,12, and 72 h in ZR-75-1 cells, but no significant expression at 24 h (159).
miR-499 E2 10 nM E2 for 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296).
miR-504 E2 10 nM E2 for72 h in MCF-7 and ZR-75-1 cells (159).
miR- E2 10 nM E2 for 24 h in MCF-7 cells (159)
miR-515-5p E2 10 nM E2 48 h in MCF-7 cells mediated by ERα binding (295). SK1 (295)
miR-518c* E2 or PhIP 10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-520d E2 10 nM E2 48 h in MCF-7 cells; Also repressed in T47D, ZR-75-1, BT-474, and BG1, but not SKBR3 breast cancer cells (296)
miR-548g E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159)
miR-570 E2 10 nM E2 for 6, 12, 24 h and 3 d in MCF-7 cells (306).

10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).
miR-574-3p 4-OHT 1 µM 4-OHT for one month in MCF-7 cells (298) Clathrin heavy chain (CLTC) (298)
miR-579 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-582-3p E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells (159).
miR-583-5p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-584 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-589 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-590-5p E2 10 nM E2 24 h ERβ stably expressing SW480 colon cancer cells (236)
miR-610 E2 10 nM E2 for 6,12, 24 and 72 h in ZR-75-1 cells, most repressed at 72 h (159).
miR-615-5p E2 or PhIP 10 nM E2 or 100 nM PhIP for 24 h in MCF-7 cells (160).
miR-616 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-618 E2 10 nM E2 for 6, 12, 24 h and 3 d in MCF-7 cells (306). 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-632 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-638 E2 or PhIP 10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-646 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159)
miR-650 E2 10 nM E2 for 24 h in T47D cells (154).
miR-663 E2 or PhIP 10 nM E2 or 100 nM PhIP for 4, 8, 12, or 24 h in MCF-7 cells (160).
miR-671:9-1, 671-3p E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-708* E2 10 nM E2 for 6, 24, and 72 h in ZR-75-1 cells, but not 12 h (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-874 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-877 4-OHT 1 µM 4-OHT for one month in MCF-7 cells (298)
miR-935 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).

10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-938 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159)
miR-1225 E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1228 E2 10 nM E2 for 24 h in T47D cells (154).
miR-1229 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1234 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159).
miR-1238 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 and ZR-75-1 cells (159) =.
miR-1257 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-1267 E2 10 nM E2 in MCF-7 cells stably overexpressing inducible ERβ or ERα (132).
miR-1301 E2 10 nM E2 for 6, 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1303 E2 10 nM E2 for 12, 24, and 72 h in ZR-75-1 cells (159).
miR-1468 E2 10 nM E2 for 6, 12, 24, and 72 h in MCF-7 cells (159).

Identification of E2- and 4-OHT- regulated miRNAs was originally performed by microarray by us (155, 156) and others (132, 157161). These reports are summarized in Tables 1 and 2. An Illumina human MicroRNA Expression Profiling Beadchip was used to identify E2-regulated miRNAs in MCF-7 and ZR-75.1 cells after 6, 12, 24, and 72 h of treatment following an initial 4 days of ‘hormone deprivation’ in medium containing 5% dextran-coated charcoal stripped FBS (159). The authors reported 230 significant miRNA changes (up- and down- regulation) that are summarized in Tables 1 and 2. The authors correlated miRNA expression with ERα in vivo binding in published data sets and found ERα binding within 10 kB of miR-125a-2, miR-181c, miR-23a, miR-27a, miR-24-2, and mIR-26 and ERα binding sites within 50kB of genes in which miRs are embedded: miR-25 in MCM2; miR-26a in CTDSP2, miR-424 in GBC16121, miR-618 in LIN7A, miR-760 in BCAR3, and miR-942 in TTF2(159). The authors noted that they found more of miR * strands regulated by E2 and suggested a possible role of ER in strand selection. Since the * strands are now known to be functional in Ago2-RISC complexes (162), these findings appear to reflect the wide range of miRNAs functionally regulating estrogen action in vivo.

GRO-seq (global nuclear run-on and sequencing) identified all RNA transcripts in E2-treated MCF-7 cells (163). The authors identified 119 miRNA transcripts as regulated by E2 at minimally one of the time points (10 and 40 min) examined with half of the miRNAs upregulated and half downregulated, the same as protein-coding transcripts. However, GRO-seq is unable to detect miRNAs that are co-transcribed as a part of their host gene within which they are embedded (164). Another genome wide analysis of E2-regulated miRNA expression was performed in MCF-7 and ZR-75-1 luminal-like breast cancer cells (165). In that study, E2 increased miR-760 and miR-424 and decreased miR-618, miR-570, and miR-107 expression. It will be of interest to correlate binding events, transcriptional regulation, and functional outcome in these large-scale studies.

Aromatase inhibitors are used to inhibit the endogenous synthesis of estrogens in postmenopausal breast cancer patients (166). The aromatase inhibitor letrozole (10 nM) stimulated the expression of let-7f, miR-146a, miR-150, miR-27a, miR-263, miR-19a, miR-372, miR-23b, miR-203, miR-10b, miR-128a, miR-9, and miR-126 and inhibited miR-134, miR-142-5p, miR-96, miR-148b, and miR-222 expression in MCF-7 cells co-cultured with primary human stromal cells (167). If these are E2-regulated miRNAs in MCF-7 cells, then we would expect E2 to increase miR-134, miR-142-5p, miR-96, miR-148b, and miR-222 and inhibit let-7f, miR-146a, miR-150, miR-27a, miR-263, miR-19a, miR-372, miR-23b, miR-203, miR-10b, miR-128a, miR-9, and miR126. We compared these expected results with published data summarized in Tables 1 and 2. E2 has not been reported to increase miR-134, miR-148b, or miR-96; however, in agreement with the expected results, E2 increased miR-142-3p and miR-222 in MCF-7 cells (Table 1). E2 has not been reported to inhibit miR-146a, miR-150, miR-263, miR-372, miR-10b, miR-9, or miR-126; however, E2 reduced let-7f, miR-27a, miR-19a, miR-23b, miR-203, miR-128a:9.1, in MCF-7 cells (Table 2).

12. Endocrine disrupting chemicals regulating miRNA expression

Endocrine disrupting chemicals (EDC) are environmental chemicals that mimic or block transcriptional activation elicited by naturally circulating steroid hormones by binding to steroid hormone receptors and either acting as agonists or antagonists of that receptor (168, 169). EDC may also affect the levels or activities enzymes involved in steroid hormone synthesis or metabolism, alter the expression or activities of transcriptional coregulators, and cause epigenetic changes(170) (168). The role of EDC in breast cancer is suspected, but not proven (171). Based on their widespread use, environmental persistence, the possible role of EDC in hormone-related cancers is of keen interest (168, 171, 172).

There are few reports examining how EDC affect miRNA expression in fish, animals or animal cell lines (173). Treatment of mouse TM4 Sertoli cells with 10 µ g/mL nonylphenol (NP) increased the expression of 47 miRNAs and down-regulated the expression of 100 miRNAs with 24 h of treatment (174). Only 10 miRNAs were increased > 1.5-fold with mmu-miR-135* being increased ~ 4-fold. The authors correlated the increase in miR-135* with decreased expression of 18 mRNAs in NP-treated cells, but did not confirm changes at the protein level or whether these are bona fide mRNA targets of mmu-miR-135a* (174). Neonatal exposure to the estrogenic analog estradiol benzoate (EB) from postnatal days (PND)1–5 with doses of 0, 0.75, 1.25, 2.5, or 25 µ g/d given sc, increased miR-29 (a,b, and c) in adult (PND90) rat testicular tissue with a concordant decrease in miR-29 target Mcl-1 protein (175).

To my knowledge, based on searching PubMed, there are only four studies of the effect of EDC on miRNA expression in human cell lines. One study showed that, like E2 (156), 10 µ M o,p-dichlorodiphenyltrichloroethane (DDT) and 10 µ M bisphenol A (BPA) activate ERα in MCF-7 cells and downregulated miR-21 (161). In addition, the authors reported that treatment of MCF-7 cells with 1 nM E2, 10 µ M BPA, or 10 µ M DDT reduced the expression of let-7a, b, c, d, e, and f, miR-15b, and miR-28b and upregulated miR-638, miR-663, and miR-1915. We reported that the anti-fungal agents fenhexamid and fludioxonil increased miR-21 expression in MCF-7, T47D, and MDA-MB-231 human breast cancer cells and reduced the expression of miR-125b and miR-181a (176). In MCF-7 cells, fenhexamid and fludioxonil induction of miR-21 was inhibited by fulvestrant; by AR antagonist, bicalutamide; by actinomycin D and cycloheximide, and by inhibitors of the mitogen-activated protein kinases (MAPK) and phosphoinositide 3-kinase (PI3K) pathways. Fenhexamid activation was inhibited by the arylhydrocarbon receptor antagonist α-napthoflavone.

The cooking of meat, particularly at high temperature with browning, e.g. grilling on a charcoal grill, results in the formation of heterocyclic amines (HCA), including the most abundant: 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) which is considered a mammary carcinogen (177). Treatment of MCF-7 cells with 100 nM PhIP decreased miR-21, miR-1, and miR-106b expression and increased miR-923, miR-574-3p, miR-574-5p, and miR-494 (160). Other miRNAs regulated by PhIP are listed in Tables 1 and 2.

The antimicrobial agents triclosan (TCS) and triclocarban (TCC) are widely used in many consumer products including soaps, skin creams, toothpastes and deodorants and are present in the aquatic and terrestrial environment (178). TCS and TCC are established EDS that compete with E2 for ERα and ERβ binding, albeit with lower affinity (179). TCS and TCC (each at 1 µ M) increased the expression of miR-22, miR-206, and miR-193b (2–3-fold) in MCF-7 cells, similar to the stimulation with 1 nM E2 (178).

13. miRNAs regulating ER expression

miRNAs can influence estrogen-regulated gene expression by directly reducing ERα mRNA stability or translation. Nine miRNAs have been reported to reduce ERα protein levels: miR-18a, miR 18b, miR-193b, miR-302c, miR-22 (180), miR-201, miR-221, and miR-222 (142), miR-206 (181), miR-222-3p (182), miR-4728-3p (97), miR-373 (105); miR-9-5p (122). let-7a, let-7b, and let-7i (183) (Figure 3). MiR-206 is inversely correlated with ERα expression, but not ERβ, in human breast tumors (184). miR-221/222 is higher in ERα negative than ERα positive breast cancer cell lines and human breast tumors (185, 186). Anti-miR-221 suppressed the growth of TAM-resistant breast cancer cells as xenografts in nude mice (187). Similarly, the expression of miR-22 was significantly lower in MCF-7, T-47D and BT474 ERα-positive versus ERα-negative MDA-MB-231 and SK-BR-3 breast cancer cells (188). A protein lysate microarray (LMA)-based strategy in which a library of pre-miRs was transiently transfected into MCF-7 and BT-474 cells in 384-well plates and ERα protein was subsequently analyzed in protein lysates that were printed on nitrocellulose-coated slides (189). miR-18a, miR-18b, miR -193, miR -206, and miR-302c reduced ERα by directly binding sites in the 3’UTR of ERα. Further, the authors reported an inverse correlation between the expression of miR-18a, -18b and ERα-negative breast tumor samples (189). ERα is upregulated during breast carcinogenesis and cancer stem cells (CSCs) isolated from MCF-7 and T47D cells had increased ERα and decreased let-7a, let-7b, let-7c, let-7d, let-7g levels (190). miR-873 was reported to inhibit E2-ERα-regulated gene transcription and cell proliferation by directly targeting CDK3, thus inhibiting ERα phosphorylation (Ser104, 106, and 118) and thus, ERα activity in MCF-7 cells (191). Stable overexpression of miR-873 in tamoxifen-resistant MCF-7 cells sensitized cells to tamoxifen (191).

Figure 3. Overview of miRNAs regulating ERα and ERβ expression and function.

Figure 3

MiRNAs that inhibit ERα, ERβ, and coregulators involved in gene transcription are indicated as discussed in the text.

14. miRNAs that regulate ER coregulators

miRNAs may also affect estrogen-regulated gene expression by reducing the expression of ER-interacting coactivators. miR-17-5p inhibited translation of coactivator SRC-3/AIB1/NCOA3 and reduced E2-ERα-ERE-luciferase activity in transfected cells (192). miR-195 inhibited SRC-3 expression in HepG2 cells by direct interaction with the 3’UTR region (193). There are 3 reports on miRNA regulation of corepressors that target ERα. miR-10a and -10b repress SMRT/NCOR2 (194). miR-184 (195) and miR-16 (196) represses SMRT/NCOR2 translation, but how they affect ER activity is unknown. MTA1 (metastatic tumor antigen 1) repressed miR-661, but the effect on ERα transcription was not evaluated (197). miR-615-3p repressed LCoR expression (198), but whether this affects ERα was not studied. Clearly, little is known about regulation of ER coactivators and corepressors by miRNAs.

15. E2 regulation of AGO2 in human breast cancer cell lines

The expression of Argonaut-2 (Ago2), the catalytic subunit of the RISC complex that mediates miRNA-dependent cleavage/degradation in mammals is higher in ERα-negative, HER2-positive than ERα-positive/HER2 negative (luminal) human breast cancer cell lines and tumors (16). However, E2 and the ERα-agonist PPT, but not the ERβ-agonist DPN, increased AGO2 protein expression in MCF-7 cells (16). Further studies showed that EGF acts through the MAPK pathway to increase Ago2 protein stability, but there were no studies examining the mechanism by which E2 and PPT, presumably through ERα, increase Ago2 protein levels. Surprisingly, Ago2 overexpression in MCF-7 cells increased ERα protein levels by 3-fold, despite also increasing miR-206 that reduces ERα (16). The authors concluded that this “discordant” finding indicates that there is a greater concentration of miRNAs than target proteins involved in ERα suppression than those that target ERα itself” (16). Microarray profiling shows that the expression of Ago1 and Ago2 proteins is higher while Dicer and TRBP1 is lower in ERα-negative versus ERα-positive breast cancer cells (199).

16. MicroRNA and endocrine-resistant breast cancer

Altered miRNA expression is likely to play a role in endocrine-resistance in breast cancer. A PubMed search for ‘MicroRNA and endocrine resistance in breast cancer’ generated nine new publications since my previous review (200). A recent review of mechanisms of endocrine resistance includes a paragraph on the upregulation of miR-221, miR-222, and miR-181b and downregulation of miR-21, miR-342, and miR-489 in tamoxifen-resistant breast cells (201). miR-221/222 promoted TAM-resistance by targeting ERα and the cell cycle regulator p27 (also known as Kip1) (185). Overexpression of miR-221/222 also associates with Fulvestrant-resistance (202). miR-221/222 is also increased in CD44+CD24−/low human breast cancer stem cells, indicating a role for these stem cells in endocrine resistance (203). miRNAs in CSCs and their role in chemoresistance has been recently reviewed (204).

My laboratory identified miRNAs that are differentially regulated by TAM in endocrine-sensitive MCF-7 and endocrine-resistant LY2 human breast cancer cells (155). LY2 cells were derived from MCF-7 by serial passage in the antiestrogen LY 117018, a precursor to Raloxifene (RAL) (205), and express wild-type ERα mRNA levels similar to MCF-7 cells (206), but are resistant to TAM, RAL, and Fulvestrant (ICI 182,780) (207). We identified 97 miRNAs regulated in the opposite direction in MCF-7 and LY2 cells. Quantitative real-time PCR (qPCR) selectively confirmed higher miR-200a, miR-200b, and miR-200c in MCF-7 than LY2 cells and higher miR-10a, miR-22, miR-29a, miR-125b, and miR-222 in LY2 than in MCF-7 cells (155). Some of the mRNA targets include PDCD4, BCL2, CYP1B1, and ERBB3.

Members of the miR-200 family and miR-221/222 are implicated in epithelial-mesenchymal transition (EMT) and metastasis (208). Many studies have identified an inverse relationship between the expression of the miR-200 family and its targets ZEB1/2 in cells (209213). ZEB1, a target of miR-200 family of miRNAs and a promoter of EMT, was found to be overexpressed in LY2 cells when compared to MCF-7 cells (155). We observed a progressive decrease in the expression of miR-200a, miR-200b, and miR-200c in an MCF-7-derived cell line model of TAM/endocrine resistance, i.e., decreasing from MCF-7, LCC1 (E2-independent, but TAM-sensitive; to the TAM-resistant LCC2, LCC9, and LY2 cell lines, respectively (214). Concurrently, we detected an increase in ZEB1 expression in LCC9 and LY2 cells. Overexpression of miR-200b and miR-200c enhanced the sensitivity of LY2 breast cancer cells to growth inhibition by antiestrogens 4-OHT and fulvestrant. These data are in agreement with other reports showing an inverse correlation between miR-200 family and ZEB1 expression in basal-like, triple negative breast cancer (TNBC) cells such as MDA-MB-231 and BT549 (210, 212, 213, 215). CpG island methylation of miR-200c/miR-141 promoter has been reported in breast and prostate cancer cells (216218). Treatment of MDA-MB-231 and BT549 breast and PC3 prostate cancer cells with 5-aza-2’-deoxycytidine (5-aza-dC), a demethylating agent, increased miR-200c and miR-141 expression (216). Our study agrees with these reports of epigenetic silencing of the miR-200 family, because we demonstrated that treatment of LY2 cells with 5-aza-dC + histone deacetylase inhibitor trichostatin A (TSA) increased miR-200b and miR-200c expression (214). There was a concomitant decrease in the expression of ZEB1 mRNA and protein and the LY2 cells appeared more epithelial in in morphology and were sensitized to TAM and fulvestrant inhibition. Likewise, knockdown of ZEB1 increased antiestrogen sensitivity of LY2 cells resulting in inhibition of cell proliferation (214).

Global miRNA analysis of 153 ERα+ primary breast tumors from women who subsequently took tamoxifen as an adjuvant mono-therapy revealed that no single miRNA profile was predictive of patient outcome (219). Decreased expression of miR-190b, miR-339-5p, miR-520c-3-, miR-520g, miR-520h, miR-139-3p, miR-204, miR-502-5p, miR-365, and miR-363 in the primary tumors was associated with recurrence in tamoxifen-treated patients (219).

miR-342 was downregulated in two TAM-resistant cell lines derived from MCF-7 cells called LCC2 and TAMR1 (220). Overexpression of miR-342 conferred TAM-sensitivity and increased apoptosis. miR-451, an oncosuppressor miRNA, was downregulated in TAM-resistant breast cancer cells (221). miR-451 targets 14-3-3ζ an anti-apoptotic gene that is overexpressed in TAM-resistant tumors and is associated with lower survival (221). Increased expression of ERα36, a truncated form of the full length ERα66, that blocks ERα66 genomic activity while activating MAPK signaling, has been reported in TAM-resistant breast tumors (222). Let-7a targets ERα36 and loss of Let-7 family members conferred TAM-resistance by activating non-genomic estrogen signaling mediated by ERα36 (223).

miRNA microarray profiling identified 10 miRNAs downregulated in a TAM-resistant MCF-7 cell line compared with wt MCF-7 cells: miR-125a, miR-489, miR-375, miR-653, miR-135b, miR-556-3p, miR-190b, miR-556-5p, miR-561, and miR-548h; while 12 miRs were upregulated: miR-551b, miR-519a, miR-376a*, miR-31, miR-224, miR-521, miR-31*, miR-655, miR-205, miR-518f, miR-520h, miR-455-3p (224). Transfection of TAM-resistant MCF-7 cells with pre-miR-375 re-sensitized the cells to ~ 15% growth inhibition by 5 µ M TAM, reduced mRNA expression of EMT markers: FN1, ZEB1, and SNAI2, and reverted EMT-like invasive appearance of the cells (224). MTDH was identified as a direct target of miR-375 and siMTDH in TAM-resistant MCF-7 cells partially sensitized the cells to tamoxifen and higher TDFH was correlated with reduced disease-free survival in tamoxifen-treated breast cancer patients (224).

The miRNA cluster C19MC, encoding 59 miRNAs spanning ~ 100 kB(225), is the largest known cluster of miRNAs in the human genome (226). Many miRNAs of C19MC are oncomiRs when re-expressed in tissues (225). miRNA microarray profiling revealed that 18 miRNAs in the C19MC cluster were upregulated in in a TAM-resistant MCF-7 cell line compared with wt MCF-7 cells including miR- 520c-3p, miR-519d, miR-518b, miR-520h, miR-521, miR-518f, miR-520b, miR-518c, miR-512-5p, miR-512-3p, miR-518e*, miR-515-5p, miR-517c, miR-522, and miR-519a (227). Overexpression of a miR-519a mimic in MCF-7 cells resulted in TAM-resistance and transfection of TAM-resistant MCF-7 cells with a miR-519a inhibitor restored TAM-growth inhibition on the cells (227). The authors verified CDKN1A, RB1, and PTEN as bona fide targets of miR-519a and correlated increased miR-519a expression with poorer disease-free survival in ERα+ breast cancer patients (227).

CONCLUSION

Estrogens, most commonly E2, and other ER ligands including tamoxifen and endocrine disruptors regulate diverse physiological effects through genomic and nongenomic/membrane-initiated mechanisms that alter cellular expression of miRNAs. miRNAs are post-transcriptional regulators of mRNA translation and stability. Although miRNA changes in fish, mice, rats, and human breast cancer cells in response to E2 and tamoxifen have been reported, there are relatively few studies examining the detailed mechanisms for these responses and their downstream bona fide targets. The effect of E2 varies between and within cell lines depending on the ratio of ERs, including GPER, expressed, coregulators, chromatin structure, cell cycle, circadian rhythms, and numerous other physiological parameters. Future HITS-CLIP and global high-throughput studies are needed to elucidate the general principles while detailed biochemical/molecular studies are required to dissect the specific mechanisms involved in ER/miRNA interactions and their roles in human health and disease.

Highlights.

  • Reviews miRNA biogenesis and regulation by estradiol

  • Two tables summarize miRNAs stimulated or repressed by estradiol and tamoxifen or other ER ligands in human tissues or cell lines

  • Reviews regulation of miRNAs by endocrine disrupting chemicals

Acknowledgement

This study was supported by NIH R01 CA138410 to C.M.K.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest: none

DISCLOSURE: This is an updated report of the earlier reviews: C.M. Klinge “Estrogen Regulation of MicroRNA Expression.” Curr Genomics 10:169–183, 2009 and C.M. Klinge “miRNAs and estrogen action. Trends in Endocrinology & Metabolism” 23:223–233, 2012.

References

  • 1.To SQ, Knower KC, Cheung V, Simpson ER, Clyne CD. Transcriptional control of local estrogen formation by aromatase in the breast. The Journal of Steroid Biochemistry and Molecular Biology. 2014 doi: 10.1016/j.jsbmb.2014.05.004. [DOI] [PubMed] [Google Scholar]
  • 2.James G. Gender is a risk factor for lung cancer. Med Hypotheses. 2011;76:328–331. doi: 10.1016/j.mehy.2010.10.030. [DOI] [PubMed] [Google Scholar]
  • 3.Henderson BE, Feigelson HS. Hormonal carcinogenesis. Carcinogenesis. 2000;21:427–433. doi: 10.1093/carcin/21.3.427. [DOI] [PubMed] [Google Scholar]
  • 4.Zhou W, Slingerland JM. Links between oestrogen receptor activation and proteolysis: relevance to hormone-regulated cancer therapy. Nat Rev Cancer. 2014;14:26–38. doi: 10.1038/nrc3622. [DOI] [PubMed] [Google Scholar]
  • 5.Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See L-H, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigo R, Gingeras TR. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Wolf JB. Principles of transcriptome analysis and gene expression quantification: an RNA-seq tutorial. Molecular ecology resources. 2013;13:559–572. doi: 10.1111/1755-0998.12109. [DOI] [PubMed] [Google Scholar]
  • 7.Marrone AK, Beland FA, Pogribny IP. Noncoding RNA response to xenobiotic exposure: an indicator of toxicity and carcinogenicity. Expert opinion on drug metabolism & toxicology. 2014:1–14. doi: 10.1517/17425255.2014.954312. [DOI] [PubMed] [Google Scholar]
  • 8.Watanabe T, Lin H. Posttranscriptional Regulation of Gene Expression by Piwi Proteins and piRNAs. Mol Cell. 2014;56:18–27. doi: 10.1016/j.molcel.2014.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rinn J, Guttman M. RNA and dynamic nuclear organization. Science. 2014;345:1240–1241. doi: 10.1126/science.1252966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zamore PD, Haley B. Ribo-gnome: The Big World of Small RNAs. Science. 2005;309:1519–1524. doi: 10.1126/science.1111444. [DOI] [PubMed] [Google Scholar]
  • 11.Zeng Y. Principles of micro-RNA production and maturation. Oncogene. 2006;25:6156–6162. doi: 10.1038/sj.onc.1209908. [DOI] [PubMed] [Google Scholar]
  • 12.Couzin J. Genetics. Erasing microRNAs reveals their powerful punch. Science. 2007;316:530. doi: 10.1126/science.316.5824.530. [DOI] [PubMed] [Google Scholar]
  • 13.Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009;19:92–105. doi: 10.1101/gr.082701.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Blenkiron C, Goldstein LD, Thorne NP, Spiteri I, Chin SF, Dunning MJ, Barbosa-Morais NL, Teschendorff AE, Green AR, Ellis IO, Tavare S, Caldas C, Miska EA. MicroRNA expression profiling of human breast cancer identifies new markers of tumor subtype. Genome Biol. 2007;8:R214. doi: 10.1186/gb-2007-8-10-r214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY. miR-21-mediated tumor growth. Oncogene. 2007;26:2799–2803. doi: 10.1038/sj.onc.1210083. [DOI] [PubMed] [Google Scholar]
  • 16.Adams BD, Claffey KP, White BA. Argonaute-2 Expression is Regulated by EGFR/MAPK Signaling and Correlates with a Transformed Phenotype in Breast Cancer Cells. Endocrinology:en.2008-0984. 2008 doi: 10.1210/en.2008-0984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Foekens JA, Sieuwerts AM, Smid M, Look MP, de Weerd V, Boersma AWM, Klijn JGM, Wiemer EAC, Martens JWM. Four miRNAs associated with aggressiveness of lymph node-negative, estrogen receptor-positive human breast cancer. Proceedings of the National Academy of Sciences. 2008;105:13021–13026-. doi: 10.1073/pnas.0803304105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lowery AJ, Miller N, McNeill RE, Kerin MJ. MicroRNAs as Prognostic Indicators and Therapeutic Targets: Potential Effect on Breast Cancer Management. Clin Cancer Res. 2008;14:360–365. doi: 10.1158/1078-0432.CCR-07-0992. [DOI] [PubMed] [Google Scholar]
  • 19.Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, Jacob S, Majumder S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27(Kip1) J Biol Chem. 2008;283:29897–29903. doi: 10.1074/jbc.M804612200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL, Massague J. Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 2008;451:147–152. doi: 10.1038/nature06487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yu Z, Wang C, Wang M, Li Z, Casimiro MC, Liu M, Wu K, Whittle J, Ju X, Hyslop T, McCue P, Pestell RG. A cyclin D1/microRNA 17/20 regulatory feedback loop in control of breast cancer cell proliferation. J Cell Biol. 2008;182:509–517. doi: 10.1083/jcb.200801079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Iorio MV, Ferracin M, Liu C-G, Veronese A, Spizzo R, Sabbioni S, Magri E, Pedriali M, Fabbri M, Campiglio M, Menard S, Palazzo JP, Rosenberg A, Musiani P, Volinia S, Nenci I, Calin GA, Querzoli P, Negrini M, Croce CM. MicroRNA Gene Expression Deregulation in Human Breast Cancer. Cancer Res. 2005;65:7065–7070. doi: 10.1158/0008-5472.CAN-05-1783. [DOI] [PubMed] [Google Scholar]
  • 23.Jiang J, Lee EJ, Gusev Y, Schmittgen TD. Real-time expression profiling of microRNA precursors in human cancer cell lines. Nucl Acids Res. 2005;33:5394–5403. doi: 10.1093/nar/gki863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mattie MD, Benz CC, Bowers J, Sensinger K, Wong L, Scott GK, Fedele V, Ginzinger D, Getts R, Haqq C. Optimized high-throughput microRNA expression profiling provides novel biomarker assessment of clinical prostate and breast cancer biopsies. Mol Cancer. 2006;5:24. doi: 10.1186/1476-4598-5-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ru Y, Kechris KJ, Tabakoff B, Hoffman P, Radcliffe RA, Bowler R, Mahaffey S, Rossi S, Calin GA, Bemis L, Theodorescu D. The multiMiR R package and database: integration of microRNA–target interactions along with their disease and drug associations. Nucleic Acids Res. 2015;42:e133. doi: 10.1093/nar/gku631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nunez E, Fu X-D, Rosenfeld MG. Nuclear organization in the 3D space of the nucleus -- cause or consequence? Curr Opin Genet Dev. 2009;19:424–436. doi: 10.1016/j.gde.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Maglich JM, Sluder A, Guan X, Shi Y, McKee DD, Carrick K, Kamdar K, Willson TM, Moore JT. Comparison of complete nuclear receptor sets from the human, Caenorhabditis elegans and Drosophila genomes. Genome Biol. 2001;2 doi: 10.1186/gb-2001-2-8-research0029. RESEARCH0029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Klinge CM. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res. 2001;29:2905–2919. doi: 10.1093/nar/29.14.2905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ruff M, Gangloff M, Wurtz JM, Moras D. Estrogen receptor transcription and transactivation: Structure-function relationship in DNA- and ligand-binding domains of estrogen receptors. Breast Cancer Res. 2000;2:353–359. doi: 10.1186/bcr80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Magnani L, Lupien M. Chromatin and epigenetic determinants of estrogen receptor alpha (ESR1) signaling. Mol Cell Endocrinol. 2014;382:633–641. doi: 10.1016/j.mce.2013.04.026. [DOI] [PubMed] [Google Scholar]
  • 31.Rosenfeld MG, Glass CK. Coregulator Codes of Transcriptional Regulation by Nuclear Receptors. J Biol Chem. 2001;276:36865–36868. doi: 10.1074/jbc.R100041200. [DOI] [PubMed] [Google Scholar]
  • 32.Dasgupta S, Lonard DM, O'Malley BW. Nuclear Receptor Coactivators: Master Regulators of Human Health and Disease. Annu Rev Med. 2014;65:279–292. doi: 10.1146/annurev-med-051812-145316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Carroll JS, Meyer CA, Song J, Li W, Geistlinger TR, Eeckhoute J, Brodsky AS, Keeton EK, Fertuck KC, Hall GF, Wang Q, Bekiranov S, Sementchenko V, Fox EA, Silver PA, Gingeras TR, Liu XS, Brown M. Genome-wide analysis of estrogen receptor binding sites. Nat Genet. 2006;38:1289–1297. doi: 10.1038/ng1901. [DOI] [PubMed] [Google Scholar]
  • 34.Carroll JS, Brown M. Estrogen Receptor Target Gene: An Evolving Concept. Mol Endocrinol. 2006;20:1707–1714. doi: 10.1210/me.2005-0334. [DOI] [PubMed] [Google Scholar]
  • 35.Liu Y, Gao H, Marstrand TT, Strom A, Valen E, Sandelin A, Gustafsson J-A, Dahlman-Wright K. The genome landscape of ER{alpha}- and ER{beta}-binding DNA regions. Proceedings of the National Academy of Sciences. 2008 doi: 10.1073/pnas.0712085105. 0712085105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lin CY, Vega VB, Thomsen JS, Zhang T, Kong SL, Xie M, Chiu KP, Lipovich L, Barnett DH, Stossi F, Yeo A, George J, Kuznetsov VA, Lee YK, Charn TH, Palanisamy N, Miller LD, Cheung E, Katzenellenbogen BS, Ruan Y, Bourque G, Wei CL, Liu ET. Whole-Genome Cartography of Estrogen Receptor alpha Binding Sites. PLoS Genet. 2007;3:e87. doi: 10.1371/journal.pgen.0030087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kwon Y-S, Garcia-Bassets I, Hutt KR, Cheng CS, Jin M, Liu D, Benner C, Wang D, Ye Z, Bibikova M, Fan J-B, Duan L, Glass CK, Rosenfeld MG, Fu X-D. Sensitive ChIP-DSL technology reveals an extensive estrogen receptor {alpha}-binding program on human gene promoters. PNAS. 2007;104:4852–4857. doi: 10.1073/pnas.0700715104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Welboren W-J, Sweep FCGJ, Span PN, Stunnenberg HG. Genomic actions of estrogen receptor {alpha}: what are the targets and how are they regulated? Endocr Relat Cancer. 2009;16:1073–1089. doi: 10.1677/ERC-09-0086. [DOI] [PubMed] [Google Scholar]
  • 39.Welboren W-J, van Driel MA, Janssen-Megens EM, van Heeringen SJ, Sweep FCGJ, Span PN, Stunnenberg HG. ChIP-Seq of ER[alpha] and RNA polymerase II defines genes differentially responding to ligands. EMBO J advanced online publication. 2009 doi: 10.1038/emboj.2009.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stender JD, Kim K, Charn TH, Komm B, Chang KCN, Kraus WL, Benner C, Glass CK, Katzenellenbogen BS. Genome-Wide Analysis of Estrogen Receptor {alpha} DNA Binding and Tethering Mechanisms Identifies Runx1 as a Novel Tethering Factor in Receptor-Mediated Transcriptional Activation. Mol Cell Biol. 2010;30:3943–3955. doi: 10.1128/MCB.00118-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Krum SA, Miranda-Carboni GA, Lupien M, Eeckhoute J, Carroll JS, Brown M. Unique ER{alpha} Cistromes Control Cell Type-Specific Gene Regulation. Mol Endocrinol. 2008;22:2393–2406. doi: 10.1210/me.2008-0100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Heldring N, Isaacs GD, Diehl AG, Sun M, Cheung E, Ranish JA, Kraus WL. Multiple Sequence-Specific DNA-Binding Proteins Mediate Estrogen Receptor Signaling through a Tethering Pathway. Mol Endocrinol. 2011;25:564–574. doi: 10.1210/me.2010-0425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Porter W, Wang F, Wang W, Duan R, Safe S. Role of estrogen receptor/Sp1 complexes in estrogen-induced heat shock protein 27 gene expression. Mol Endocrinol. 1996;10:1371–1378. doi: 10.1210/mend.10.11.8923463. [DOI] [PubMed] [Google Scholar]
  • 44.Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science. 1997;277:1508–1510. doi: 10.1126/science.277.5331.1508. [DOI] [PubMed] [Google Scholar]
  • 45.Zhao C, Gao H, Liu Y, Papoutsi Z, Jaffrey S, Gustafsson J-Å, Dahlman-Wright K. Genome-Wide Mapping of Estrogen Receptor-β–Binding Regions Reveals Extensive Cross-Talk with Transcription Factor Activator Protein-1. Cancer Res. 2010;70:5174–5183. doi: 10.1158/0008-5472.CAN-09-4407. [DOI] [PubMed] [Google Scholar]
  • 46.Gu F, Hsu H-K, Hsu P-Y, Wu J, Ma Y, Parvin J, Huang T, Jin V. Inference of hierarchical regulatory network of estrogen-dependent breast cancer through ChIP-based data. BMC Systems Biology. 2010;4:170. doi: 10.1186/1752-0509-4-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Watson CS, Jeng Y-J, Hu G, Wozniak A, Bulayeva N, Guptarak J. Estrogen- and xenoestrogen-induced ERK signaling in pituitary tumor cells involves estrogen receptor-α interactions with G protein-αi and caveolin I. Steroids. 2012;77:424–432. doi: 10.1016/j.steroids.2011.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Levin ER. Extranuclear estrogen receptor's roles in physiology: lessons from mouse models. American Journal of Physiology - Endocrinology and Metabolism. 2014;307:E133–E140. doi: 10.1152/ajpendo.00626.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Watson CS, Alyea RA, Jeng YJ, Kochukov MY. Nongenomic actions of low concentration estrogens and xenoestrogens on multiple tissues. Mol Cell Endocrinol. 2007;274:1–7. doi: 10.1016/j.mce.2007.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sandén C, Broselid S, Cornmark L, Andersson K, Daszkiewicz-Nilsson J, Mårtensson UEA, Olde B, Leeb-Lundberg LMF. G Protein-Coupled Estrogen Receptor 1/G Protein-Coupled Receptor 30 Localizes in the Plasma Membrane and Traffics Intracellularly on Cytokeratin Intermediate Filaments. Mol Pharmacol. 2011;79:400–410. doi: 10.1124/mol.110.069500. [DOI] [PubMed] [Google Scholar]
  • 51.Recchia AG, De Francesco EM, Vivacqua A, Sisci D, Panno ML, Andò S, Maggiolini M. The G Protein-coupled Receptor 30 Is Up-regulated by Hypoxia-inducible Factor-1α (HIF-1α) in Breast Cancer Cells and Cardiomyocytes. J Biol Chem. 2011;286:10773–10782. doi: 10.1074/jbc.M110.172247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Levin ER. Minireview: Extranuclear Steroid Receptors: Roles in Modulation of Cell Functions. Mol Endocrinol. 2011;25:377–384. doi: 10.1210/me.2010-0284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cheng S-B, Quinn JA, Graeber CT, Filardo EJ. Downmodulation of the G-protein-coupled estrogen receptor, GPER, from the cell surface occurs via a transgolgi-proteasome pathway. Journal of Biological Chemistry. 2011 doi: 10.1074/jbc.M111.224071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang D, Hu L, Zhang G, Zhang L, Chen C. G protein-coupled receptor 30 in tumor development. Endocrine. 2010;38:29–37. doi: 10.1007/s12020-010-9363-z. [DOI] [PubMed] [Google Scholar]
  • 55.Stratton RC, Squires PE, Green AK. 17β-Estradiol Elevates cGMP and, via Plasma Membrane Recruitment of Protein Kinase GIα, Stimulates Ca2+ Efflux from Rat Hepatocytes. J Biol Chem. 2010;285:27201–27212. doi: 10.1074/jbc.M110.103630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Madeo A, Maggiolini M. Nuclear Alternate Estrogen Receptor GPR30 Mediates 17β-Estradiol–Induced Gene Expression and Migration in Breast Cancer–Associated Fibroblasts. Cancer Res. 2010;70:6036–6046. doi: 10.1158/0008-5472.CAN-10-0408. [DOI] [PubMed] [Google Scholar]
  • 57.Kolkova Z, Noskova V, Ehinger A, Hansson S, Casslén B. G protein-coupled estrogen receptor 1 (GPER, GPR 30) in normal human endometrium and early pregnancy decidua. Mol Hum Reprod. 2010;16:743–751. doi: 10.1093/molehr/gaq043. [DOI] [PubMed] [Google Scholar]
  • 58.Ignatov A, Ignatov T, Roessner A, Costa S, Kalinski T. Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells. Breast Cancer Res Treat. 2010;123:87–96. doi: 10.1007/s10549-009-0624-6. [DOI] [PubMed] [Google Scholar]
  • 59.Prossnitz ER, Maggiolini M. Mechanisms of estrogen signaling and gene expression via GPR30. Mol Cell Endocrinol. 2009;308:32–38. doi: 10.1016/j.mce.2009.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Levin ER. G Protein-Coupled Receptor 30: Estrogen Receptor or Collaborator? Endocrinology. 2009;150:1563–1565. doi: 10.1210/en.2008-1759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Li L, Haynes MP, Bender JR. Plasma membrane localization and function of the estrogen receptor alpha variant (ER46) in human endothelial cells. Proc Natl Acad Sci U S A. 2003;100:4807–4812. doi: 10.1073/pnas.0831079100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M. S-palmitoylation modulates human estrogen receptor-alpha functions. Biochem Biophys Res Commun. 2004;316:878–883. doi: 10.1016/j.bbrc.2004.02.129. [DOI] [PubMed] [Google Scholar]
  • 63.Pedram A, Razandi M, Sainson RCA, Kim JK, Hughes CC, Levin ER. A Conserved Mechanism for Steroid Receptor Translocation to the Plasma Membrane. J Biol Chem. 2007;282:22278–22288. doi: 10.1074/jbc.M611877200. [DOI] [PubMed] [Google Scholar]
  • 64.Moriarty K, Kim KH, Bender JR. Estrogen Receptor-Mediated Rapid Signaling. Endocrinology. 2006;147:5557–5563. doi: 10.1210/en.2006-0729. [DOI] [PubMed] [Google Scholar]
  • 65.Chaudhri RA, Hadadi A, Lobachev KS, Schwartz Z, Boyan BD. Estrogen receptor-alpha 36 mediates the anti-apoptotic effect of estradiol in triple negative breast cancer cells via a membrane-associated mechanism. Biochim Biophys Acta. 2014;1843:2796–2806. doi: 10.1016/j.bbamcr.2014.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Razandi M, Pedram A, Levin ER. Plasma membrane estrogen receptors signal to antiapoptosis in breast cancer. Mol Endocrinol. 2000;14:1434–1447. doi: 10.1210/mend.14.9.0526. [DOI] [PubMed] [Google Scholar]
  • 67.Hisamoto K, Ohmichi M, Kurachi H, Hayakawa J, Kanda Y, Nishio Y, Adachi K, Tasaka K, Miyoshi E, Fujiwara N, Taniguchi N, Murata Y. Estrogen induces the Akt-dependent activation of endothelial nitric-oxide synthase in vascular endothelial cells. J Biol Chem. 2001;276:3459–3467. doi: 10.1074/jbc.M005036200. [DOI] [PubMed] [Google Scholar]
  • 68.Monje P, Boland R. Expression and cellular localization of naturally occurring beta estrogen receptors in uterine and mammary cell lines. J Cell Biochem. 2002;86:136–144. doi: 10.1002/jcb.10193. [DOI] [PubMed] [Google Scholar]
  • 69.Chen DB, Bird IM, Zheng J, Magness RR. Membrane estrogen receptor-dependent extracellular signal-regulated kinase pathway mediates acute activation of endothelial nitric oxide synthase by estrogen in uterine artery endothelial cells. Endocrinology. 2004;145:113–125. doi: 10.1210/en.2003-0547. [DOI] [PubMed] [Google Scholar]
  • 70.Belcher SM, Le HH, Spurling L, Wong JK. Rapid Estrogenic Regulation of Extracellular Signal- Regulated Kinase 1/2 Signaling in Cerebellar Granule Cells Involves a G Protein- and Protein Kinase A-Dependent Mechanism and Intracellular Activation of Protein Phosphatase 2A. Endocrinology. 2005;146:5397–5406. doi: 10.1210/en.2005-0564. [DOI] [PubMed] [Google Scholar]
  • 71.Mhyre AJ, Shapiro RA, Dorsa DM. Estradiol Reduces Nonclassical Transcription at Cyclic Adenosine 3',5'-Monophosphate Response Elements in Glioma Cells Expressing Estrogen Receptor Alpha. Endocrinology. 2006;147:1796–1804. doi: 10.1210/en.2005-1316. [DOI] [PubMed] [Google Scholar]
  • 72.Simoncini T, Scorticati C, Mannella P, Fadiel A, Giretti MS, Fu X-D, Baldacci C, Garibaldi S, Caruso A, Fornari L, Naftolin F, Genazzani AR. Estrogen Receptor {alpha} Interacts with G{alpha}13 to Drive Actin Remodeling and Endothelial Cell Migration via the RhoA/Rho Kinase/Moesin Pathway. Mol Endocrinol. 2006;20:1756–1771. doi: 10.1210/me.2005-0259. [DOI] [PubMed] [Google Scholar]
  • 73.Filardo E, Quinn J, Pang Y, Graeber C, Shaw S, Dong J, Thomas P. Activation of the Novel Estrogen Receptor G Protein-Coupled Receptor 30 (GPR30) at the Plasma Membrane. Endocrinology. 2007;148:3236–3245. doi: 10.1210/en.2006-1605. [DOI] [PubMed] [Google Scholar]
  • 74.Jaubert A-M, Mehebik-Mojaat N, Lacasa D, Sabourault D, Giudicelli Y, Ribiere C. Nongenomic Estrogen Effects on Nitric Oxide Synthase Activity in Rat Adipocytes. Endocrinology. 2007;148:2444–2452. doi: 10.1210/en.2006-1329. [DOI] [PubMed] [Google Scholar]
  • 75.Wang C, Prossnitz ER, Roy SK. G Protein-Coupled Receptor 30 Expression Is Required for Estrogen Stimulation of Primordial Follicle Formation in the Hamster Ovary. Endocrinology. 2008;149:4452–4461. doi: 10.1210/en.2008-0441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Jacovetti C, Regazzi R. Compensatory β-cell mass expansion: A big role for a tiny actor. Cell Cycle. 2013;12:197–198. doi: 10.4161/cc.23378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Klinge CM, Riggs KA, Wickramasinghe NS, Emberts CG, McConda DB, Barry PN, Magnusen JE. Estrogen receptor alpha 46 is reduced in tamoxifen resistant breast cancer cells and re-expression inhibits cell proliferation and estrogen receptor alpha 66-regulated target gene transcription. Mol Cell Endocrinol. 2010;323:268–276. doi: 10.1016/j.mce.2010.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pelekanou V, Notas G, Kampa M, Tsentelierou E, Radojicic J, Leclercq G, Castanas E, Stathopoulos EN. ERα36, a new variant of the ERα is expressed in triple negative breast carcinomas and has a specific transcriptomic signature in breast cancer cell lines. Steroids. 2012;77:928–934. doi: 10.1016/j.steroids.2011.12.016. [DOI] [PubMed] [Google Scholar]
  • 79.Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42:D68–D73. doi: 10.1093/nar/gkt1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Samantarrai D, Dash S, Chhetri B, Mallick B. Genomic and Epigenomic Cross-talks in the Regulatory Landscape of miRNAs in Breast Cancer. Mol Cancer Res. 2013;11:315–328. doi: 10.1158/1541-7786.MCR-12-0649. [DOI] [PubMed] [Google Scholar]
  • 81.Saini HK, Griffiths-Jones S, Enright AJ. Genomic analysis of human microRNA transcripts. Proceedings of the National Academy of Sciences. 2007;104:17719–17724. doi: 10.1073/pnas.0703890104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Amaral PP, Dinger ME, Mercer TR, Mattick JS. The Eukaryotic Genome as an RNA Machine. Science. 2008;319:1787–1789. doi: 10.1126/science.1155472. [DOI] [PubMed] [Google Scholar]
  • 83.Yang J-S, Lai Eric C. Alternative miRNA Biogenesis Pathways and the Interpretation of Core miRNA Pathway Mutants. Mol Cell. 2011;43:892–903. doi: 10.1016/j.molcel.2011.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Macias S, Cordiner RA, Caceres JF. Cellular functions of the microprocessor. Biochem Soc Trans. 2013;41:838–843. doi: 10.1042/BST20130011. [DOI] [PubMed] [Google Scholar]
  • 85.Verghese ET, Hanby AM, Speirs V, Hughes TA. Small is beautiful: microRNAs and breast cancer-where are we now? J Pathol. 2008;215:214–221. doi: 10.1002/path.2359. [DOI] [PubMed] [Google Scholar]
  • 86.Thomson JM, Newman M, Parker JS, Morin-Kensicki EM, Wright T, Hammond SM. Extensive post-transcriptional regulation of microRNAs and its implications for cancer. Genes Dev. 2006;20:2202–2207. doi: 10.1101/gad.1444406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hock J, Meister G. The Argonaute protein family. Genome Biol. 2008;9:210. doi: 10.1186/gb-2008-9-2-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pasquinelli AE, Hunter S, Bracht J. MicroRNAs: a developing story. Curr Opin Genet Dev. 2005;15:200–205. doi: 10.1016/j.gde.2005.01.002. [DOI] [PubMed] [Google Scholar]
  • 89.Sen GL, Blau HM. A brief history of RNAi: the silence of the genes. FASEB J. 2006;20:1293–1299. doi: 10.1096/fj.06-6014rev. [DOI] [PubMed] [Google Scholar]
  • 90.Berkhout B, Jeang K-T. RISCy Business: MicroRNAs, Pathogenesis, and Viruses. J Biol Chem. 2007;282:26641–26645. doi: 10.1074/jbc.R700023200. [DOI] [PubMed] [Google Scholar]
  • 91.Cuellar TL, McManus MT. MicroRNAs and endocrine biology. J Endocrinol. 2005;187:327–332. doi: 10.1677/joe.1.06426. [DOI] [PubMed] [Google Scholar]
  • 92.Rybak-Wolf A, Jens M, Murakawa Y, Herzog M, Landthaler M, Rajewsky N. A Variety of Dicer Substrates in Human and C. elegans. Cell. 2014;159:1153–1167. doi: 10.1016/j.cell.2014.10.040. [DOI] [PubMed] [Google Scholar]
  • 93.Mori M, Triboulet R, Mohseni M, Schlegelmilch K, Shrestha K, Camargo Fernando D, Gregory Richard I. Hippo Signaling Regulates Microprocessor and Links Cell-Density-Dependent miRNA Biogenesis to Cancer. Cell. 2014;156:893–906. doi: 10.1016/j.cell.2013.12.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Sibley CR, Seow Y, Saayman S, Dijkstra KK, El Andaloussi S, Weinberg MS, Wood MJA. The biogenesis and characterization of mammalian microRNAs of mirtron origin. Nucleic Acids Res. 2012;40:438–448. doi: 10.1093/nar/gkr722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ladewig E, Okamura K, Flynt AS, Westholm JO, Lai EC. Discovery of hundreds of mirtrons in mouse and human small RNA data. Genome Res. 2012;22:1634–1645. doi: 10.1101/gr.133553.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Persson H, Kvist A, Rego N, Staaf J, Vallon-Christersson J, Luts L, Loman N, Jonsson G, Naya H, Hoglund M, Borg A, Rovira C. Identification of New MicroRNAs in Paired Normal and Tumor Breast Tissue Suggests a Dual Role for the ERBB2/Her2 Gene. Cancer Res. 2011;71:78–86. doi: 10.1158/0008-5472.CAN-10-1869. [DOI] [PubMed] [Google Scholar]
  • 97.Newie I, Sokilde R, Persson H, Grabau D, Rego N, Kvist A, von Stedingk K, Axelson H, Borg A, Vallon-Christersson J, Rovira C. The HER2-encoded miR-4728-3p regulates ESR1 through a non-canonical internal seed interaction. PLoS One. 2014;9:e97200. doi: 10.1371/journal.pone.0097200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Neilsen CT, Goodall GJ, Bracken CP. IsomiRs – the overlooked repertoire in the dynamic microRNAome. Trends Genet. 2012;28:544–549. doi: 10.1016/j.tig.2012.07.005. [DOI] [PubMed] [Google Scholar]
  • 99.Muller H, Marzi MJ, Nicassio F. IsomiRage: From Functional Classification to Differential Expression of miRNA Isoforms. Frontiers in bioengineering and biotechnology. 2014;2:38. doi: 10.3389/fbioe.2014.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nishikura K. Functions and regulation of RNA editing by ADAR deaminases. Annu Rev Biochem. 2010;79:321–349. doi: 10.1146/annurev-biochem-060208-105251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Braicu C, Tomuleasa C, Monroig P, Cucuianu A, Berindan-Neagoe I, Calin GA. Exosomes as divine messengers: are they the Hermes of modern molecular oncology? Cell Death Differ. 2015;22:34–45. doi: 10.1038/cdd.2014.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–125. doi: 10.1016/j.ceb.2014.05.004. [DOI] [PubMed] [Google Scholar]
  • 103.Zomer A, Vendrig T, Hopmans ES, van Eijndhoven M, Middeldorp JM, Pegtel DM. Exosomes: Fit to deliver small RNA. Communicative & integrative biology. 2010;3:447–450. doi: 10.4161/cib.3.5.12339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Turchinovich A, Weiz L, Burwinkel B. Extracellular miRNAs: the mystery of their origin and function. Trends Biochem Sci. 2012;37:460–465. doi: 10.1016/j.tibs.2012.08.003. [DOI] [PubMed] [Google Scholar]
  • 105.Eichelser C, Stuckrath I, Muller V, Milde-Langosch K, Wikman H, Pantel K, Schwarzenbach H. Increased serum levels of circulating exosomal microRNA-373 in receptor-negative breast cancer patients. Oncotarget. 2014;5:9650–9663. doi: 10.18632/oncotarget.2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ho JJ, Marsden PA. Competition and collaboration between RNA-binding proteins and microRNAs. Wiley interdisciplinary reviews RNA. 2014;5:69–86. doi: 10.1002/wrna.1197. [DOI] [PubMed] [Google Scholar]
  • 107.Stalder L, Heusermann W, Sokol L, Trojer D, Wirz J, Hean J, Fritzsche A, Aeschimann F, Pfanzagl V, Basselet P, Weiler J, Hintersteiner M, Morrissey DV, Meisner-Kober NC. The rough endoplasmatic reticulum is a central nucleation site of siRNA-mediated RNA silencing. EMBO J. 2013;32:1115–1127. doi: 10.1038/emboj.2013.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chendrimada TP, Finn KJ, Ji X, Baillat D, Gregory RI, Liebhaber SA, Pasquinelli AE, Shiekhattar R. MicroRNA silencing through RISC recruitment of eIF6. Nature. 2007;447:823–828. doi: 10.1038/nature05841. [DOI] [PubMed] [Google Scholar]
  • 109.Leung AK, Sharp PA. Quantifying Argonaute proteins in and out of GW/P-bodies: implications in microRNA activities. Adv Exp Med Biol. 2013;768:165–182. doi: 10.1007/978-1-4614-5107-5_10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Behm-Ansmant I, Rehwinkel J, Izaurralde E. MicroRNAs silence gene expression by repressing protein expression and/or by promoting mRNA decay. Cold Spring Harb Symp Quant Biol. 2006;71:523–530. doi: 10.1101/sqb.2006.71.013. [DOI] [PubMed] [Google Scholar]
  • 111.Perron MP, Provost P. Protein interactions and complexes in human microRNA biogenesis and function. Front Biosci. 2008;13:2537–2547. doi: 10.2741/2865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Vasudevan S, Tong Y, Steitz JA. Switching from Repression to Activation: MicroRNAs Can Up-Regulate Translation. Science. 2007;318:1931–1934. doi: 10.1126/science.1149460. [DOI] [PubMed] [Google Scholar]
  • 113.Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi Pier P. A ceRNA Hypothesis: The Rosetta Stone of a Hidden RNA Language? Cell. 2011;146:353–358. doi: 10.1016/j.cell.2011.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505:344–352. doi: 10.1038/nature12986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kriegel AJ, Liu Y, Fang Y, Ding X, Liang M. The miR-29 family: genomics, cell biology, and relevance to renal and cardiovascular injury. Physiol Genomics. 2012;44:237–244. doi: 10.1152/physiolgenomics.00141.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Asangani IA, Rasheed SAK, Nikolova DA, Leupold JH, Colburn NH, Post S, Allgayer H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 2008;27:2128–2136. doi: 10.1038/sj.onc.1210856. [DOI] [PubMed] [Google Scholar]
  • 117.Volinia S, Calin GA, Liu C-G, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, Prueitt RL, Yanaihara N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris CC, Croce CM. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences. 2006;103:2257–2261. doi: 10.1073/pnas.0510565103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S, Naylor TL, Barchetti A, Ward MR, Yao G, Medina A, O'Brien-Jenkins A, Katsaros D, Hatzigeorgiou A, Gimotty PA, Weber BL, Coukos G. microRNAs exhibit high frequency genomic alterations in human cancer. PNAS. 2006;103:9136–9141. doi: 10.1073/pnas.0508889103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kawamata T, Tomari Y. Making RISC. Trends Biochem Sci. 2010;35:368–376. doi: 10.1016/j.tibs.2010.03.009. [DOI] [PubMed] [Google Scholar]
  • 120.Thomson DW, Bracken CP, Goodall GJ. Experimental strategies for microRNA target identification. Nucleic Acids Res. 2011;39:6845–6853. doi: 10.1093/nar/gkr330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Zhang C, Darnell RB. Mapping in vivo protein-RNA interactions at single-nucleotide resolution from HITS-CLIP data. Nat Biotech. 2011;29:607–614. doi: 10.1038/nbt.1873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pillai MM, Gillen AE, Yamamoto TM, Kline E, Brown J, Flory K, Hesselberth JR, Kabos P. HITS-CLIP reveals key regulators of nuclear receptor signaling in breast cancer. Breast Cancer Res Treat. 2014;146:85–97. doi: 10.1007/s10549-014-3004-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A, Enright AJ. miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 2006;34:D140–D144. doi: 10.1093/nar/gkj112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Okamura K, Phillips MD, Tyler DM, Duan H, Chou Y-t, Lai EC. The regulatory activity of microRNA[ast] species has substantial influence on microRNA and 3[prime] UTR evolution. Nat Struct Mol Biol. 2008;15:354–363. doi: 10.1038/nsmb.1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Gurtan AM, Sharp PA. The Role of miRNAs in Regulating Gene Expression Networks. J Mol Biol. 2013;425:3582–3600. doi: 10.1016/j.jmb.2013.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tanzer A, Stadler PF. Molecular evolution of a microRNA cluster. J Mol Biol. 2004;339:327–335. doi: 10.1016/j.jmb.2004.03.065. [DOI] [PubMed] [Google Scholar]
  • 127.Gregory RI, Yan K-p, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, Shiekhattar R. The Microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432:235–240. doi: 10.1038/nature03120. [DOI] [PubMed] [Google Scholar]
  • 128.Han J, Pedersen JS, Kwon SC, Belair CD, Kim Y-K, Yeom K-H, Yang W-Y, Haussler D, Blelloch R, Kim VN. Posttranscriptional Crossregulation between Drosha and DGCR8. Cell. 2009;136:75–84. doi: 10.1016/j.cell.2008.10.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wortham NC, Ahamed E, Nicol SM, Thomas RS, Periyasamy M, Jiang J, Ochocka AM, Shousha S, Huson L, Bray SE, Coombes RC, Ali S, Fuller-Pace FV. The DEAD-box protein p72 regulates ER[alpha]-/oestrogen-dependent transcription and cell growth, and is associated with improved survival in ER[alpha]-positive breast cancer. Oncogene. 2009;28:4053–4064. doi: 10.1038/onc.2009.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Yamagata K, Fujiyama S, Ito S, Ueda T, Murata T, Naitou M, Takeyama K-i, Minami Y, O'Malley BW, Kato S. Maturation of MicroRNA Is Hormonally Regulated by a Nuclear Receptor. Mol Cell. 2009;36:340–347. doi: 10.1016/j.molcel.2009.08.017. [DOI] [PubMed] [Google Scholar]
  • 131.Retraction notice to: Maturation of microRNA is hormonally regulated by a nuclear receptor. Mol Cell. 2014;54:536. doi: 10.1016/j.molcel.2014.04.018. [DOI] [PubMed] [Google Scholar]
  • 132.Paris O, Ferraro L, Grober OMV, Ravo M, De Filippo MR, Giurato G, Nassa G, Tarallo R, Cantarella C, Rizzo F, Di Benedetto A, Mottolese M, Benes V, Ambrosino C, Nola E, Weisz A. Direct regulation of microRNA biogenesis and expression by estrogen receptor beta in hormone-responsive breast cancer. Oncogene. 2012;31:4196–4206. doi: 10.1038/onc.2011.583. [DOI] [PubMed] [Google Scholar]
  • 133.Paroo Z, Ye X, Chen S, Liu Q. Phosphorylation of the Human MicroRNA-Generating Complex Mediates MAPK/Erk Signaling. Cell. 2009;139:112–122. doi: 10.1016/j.cell.2009.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Redfern AD, Colley SM, Beveridge DJ, Ikeda N, Epis MR, Li X, Foulds CE, Stuart LM, Barker A, Russell VJ, Ramsay K, Kobelke SJ, Li X, Hatchell EC, Payne C, Giles KM, Messineo A, Gatignol A, Lanz RB, O'Malley BW, Leedman PJ. RNA-induced silencing complex (RISC) Proteins PACT, TRBP, and Dicer are SRA binding nuclear receptor coregulators. Proceedings of the National Academy of Sciences. 2013;110:6536–6541. doi: 10.1073/pnas.1301620110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Adams BD, Claffey KP, White BA. Argonaute-2 expression is regulated by epidermal growth factor receptor and mitogen-activated protein kinase signaling and correlates with a transformed phenotype in breast cancer cells. Endocrinology. 2009;150:14–23. doi: 10.1210/en.2008-0984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Shen J, Xia W, Khotskaya YB, Huo L, Nakanishi K, Lim SO, Du Y, Wang Y, Chang WC, Chen CH, Hsu JL, Wu Y, Lam YC, James BP, Liu X, Liu CG, Patel DJ, Hung MC. EGFR modulates microRNA maturation in response to hypoxia through phosphorylation of AGO2. Nature. 2013;497:383–387. doi: 10.1038/nature12080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tuteja R, Tuteja N. Nucleolin: a multifunctional major nucleolar phosphoprotein. Crit Rev Biochem Mol Biol. 1998;33:407–436. doi: 10.1080/10409239891204260. [DOI] [PubMed] [Google Scholar]
  • 138.Bates PJ, Laber DA, Miller DM, Thomas SD, Trent JO. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Exp Mol Pathol. 2009;86:151–164. doi: 10.1016/j.yexmp.2009.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Litchfield LM, Riggs KA, Hockenberry AM, Oliver LD, Barnhart KG, Cai J, Pierce WM, Jr, Ivanova MM, Bates PJ, Appana SN, Datta S, Kulesza P, McBryan J, Young LS, Klinge CM. Identification and Characterization of Nucleolin as a COUP-TFII Coactivator of Retinoic Acid Receptor β Transcription in Breast Cancer Cells. PLoS ONE. 2012;7:e38278. doi: 10.1371/journal.pone.0038278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Pichiorri F, Palmieri D, De Luca L, Consiglio J, You J, Rocci A, Talabere T, Piovan C, Lagana A, Cascione L, Guan J, Gasparini P, Balatti V, Nuovo G, Coppola V, Hofmeister CC, Marcucci G, Byrd JC, Volinia S, Shapiro CL, Freitas MA, Croce CM. In vivo NCL targeting affects breast cancer aggressiveness through miRNA regulation. The Journal of Experimental Medicine. 2013 doi: 10.1084/jem.20120950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Klinge CM. Estrogen Regulation of MicroRNA Expression. Curr Genomics. 2009;10:169–183. doi: 10.2174/138920209788185289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Klinge CM. miRNAs and estrogen action. Trends Endocrinol Metab miRNAs and estrogen action. 2012 doi: 10.1016/j.tem.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Gupta A, Caffrey E, Callagy G, Gupta S. Oestrogen-dependent regulation of miRNA biogenesis: many ways to skin the cat. Biochem Soc Trans. 2012;40:752–758. doi: 10.1042/BST20110763. [DOI] [PubMed] [Google Scholar]
  • 144.Rao YS, Mott NN, Wang Y, Chung WC, Pak TR. MicroRNAs in the aging female brain: a putative mechanism for age-specific estrogen effects. Endocrinology. 2013;154:2795–2806. doi: 10.1210/en.2013-1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Munagala R, Aqil F, Vadhanam MV, Gupta RC. MicroRNA ‘signature’ during estrogen-mediated mammary carcinogenesis and its reversal by ellagic acid intervention. Cancer Lett. 2013;339:175–184. doi: 10.1016/j.canlet.2013.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Zhao J, Imbrie GA, Baur WE, Iyer LK, Aronovitz MJ, Kershaw TB, Haselmann GM, Lu Q, Karas RH. Estrogen receptor-mediated regulation of microRNA inhibits proliferation of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2013;33:257–265. doi: 10.1161/ATVBAHA.112.300200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Zhang Y, Wu L, Wang Y, Zhang M, Li L, Zhu D, Li X, Gu H, Zhang C-Y, Zen K. Protective Role of Estrogen-induced miRNA-29 Expression in Carbon Tetrachloride-induced Mouse Liver Injury. J Biol Chem. 2012;287:14851–14862. doi: 10.1074/jbc.M111.314922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Queiros AM, Eschen C, Fliegner D, Kararigas G, Dworatzek E, Westphal C, Sanchez Ruderisch H, Regitz-Zagrosek V. Sex- and estrogen-dependent regulation of a miRNA network in the healthy and hypertrophied heart. Int J Cardiol. 2013;169:331–338. doi: 10.1016/j.ijcard.2013.09.002. [DOI] [PubMed] [Google Scholar]
  • 149.Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M. Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell. 2000;103:843–852. doi: 10.1016/s0092-8674(00)00188-4. [DOI] [PubMed] [Google Scholar]
  • 150.Vivar OI, Zhao X, Saunier EF, Griffin C, Mayba OS, Tagliaferri M, Cohen I, Speed TP, Leitman DC. Estrogen receptor [beta] binds to and regulates three distinct classes of target genes. J Biol Chem. 2010;285:22059–22066. doi: 10.1074/jbc.M110.114116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Liao XH, Lu DL, Wang N, Liu LY, Wang Y, Li YQ, Yan TB, Sun XG, Hu P, Zhang TC. Estrogen receptor α mediates proliferation of breast cancer MCF–7 cells via a p21/PCNA/E2F1-dependent pathway. FEBS J. 2014;281:927–942. doi: 10.1111/febs.12658. [DOI] [PubMed] [Google Scholar]
  • 152.Nassa G, Tarallo R, Giurato G, De Filippo MR, Ravo M, Rizzo F, Stellato C, Ambrosino C, Baumann M, Lietzen N, Nyman TA, Weisz A. Post-transcriptional regulation of human breast cancer cell proteome by unliganded estrogen receptor beta via microRNAs. Mol Cell Proteomics. 2014;13:1076–1090. doi: 10.1074/mcp.M113.030403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vantaggiato C, Tocchetti M, Cappelletti V, Gurtner A, Villa A, Daidone MG, Piaggio G, Maggi A, Ciana P. Cell cycle dependent oscillatory expression of estrogen receptor-α links Pol II elongation to neoplastic transformation. Proceedings of the National Academy of Sciences. 2014;111:9561–9566. doi: 10.1073/pnas.1321750111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Katchy A, Edvardsson K, Aydogdu E, Williams C. Estradiol-activated estrogen receptor α does not regulate mature microRNAs in T47D breast cancer cells. J Steroid Biochem Mol Biol. 2012;128:145–153. doi: 10.1016/j.jsbmb.2011.10.008. [DOI] [PubMed] [Google Scholar]
  • 155.Manavalan TT, Teng Y, Appana SN, Datta S, Kalbfleisch TS, Li Y, Klinge CM. Differential expression of microRNA expression in tamoxifen-sensitive MCF-7 versus tamoxifen-resistant LY2 human breast cancer cells. Cancer Lett. 2011;313:26–43. doi: 10.1016/j.canlet.2011.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wickramasinghe N, Manavalan T, Dougherty S, Riggs K, Li Y, Klinge C. Estradiol downregulates miR-21 expression and increases miR-21 target gene expression in MCF-7 breast cancer cells. Nucleic Acids Res. 2009;37:2584–2595. doi: 10.1093/nar/gkp117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Bhat-Nakshatri P, Wang G, Collins NR, Thomson MJ, Geistlinger TR, Carroll JS, Brown M, Hammond S, Srour EF, Liu Y, Nakshatri H. Estradiol-regulated microRNAs control estradiol response in breast cancer cells. Nucl Acids Res. 2009;37:4850–4861. doi: 10.1093/nar/gkp500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhang R, He Y, Zhang X, Xing B, Sheng Y, Lu H, Wei Z. Estrogen receptor-regulated microRNAs contribute to the BCL2/BAX imbalance in endometrial adenocarcinoma and precancerous lesions. Cancer Lett. 2012;314:155–165. doi: 10.1016/j.canlet.2011.09.027. [DOI] [PubMed] [Google Scholar]
  • 159.Ferraro L, Ravo M, Nassa G, Tarallo R, De Filippo MR, Giurato G, Cirillo F, Stellato C, Silvestro S, Cantarella C, Rizzo F, Cimino D, Friard O, Biglia N, De Bortoli M, Cicatiello L, Nola E, Weisz A. Effects of oestrogen on microRNA expression in hormone-responsive breast cancer cells. Horm Cancer. 2012;3:65–78. doi: 10.1007/s12672-012-0102-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Papaioannou MD, Koufaris C, Gooderham NJ. The cooked meat-derived mammary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) elicits estrogenic-like microRNA responses in breast cancer cells. Toxicol Lett. 2014;229:9–16. doi: 10.1016/j.toxlet.2014.05.021. [DOI] [PubMed] [Google Scholar]
  • 161.Tilghman SL, Bratton MR, Segar HC, Martin EC, Rhodes LV, Li M, McLachlan JA, Wiese TE, Nephew KP, Burow ME. Endocrine Disruptor Regulation of MicroRNA Expression in Breast Carcinoma Cells. PLoS ONE. 2012;7:e32754. doi: 10.1371/journal.pone.0032754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Xue B, He L. An expanding universe of the non-coding genome in cancer biology. Carcinogenesis. 2014;35:1209–1216. doi: 10.1093/carcin/bgu099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Hah N, Danko Charles G, Core L, Waterfall Joshua J, Siepel A, Lis John T, Kraus WL. A Rapid, Extensive, and Transient Transcriptional Response to Estrogen Signaling in Breast Cancer Cells. Cell. 2011;145:622–634. doi: 10.1016/j.cell.2011.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hah N, Kraus WL. Hormone-regulated transcriptomes: Lessons learned from estrogen signaling pathways in breast cancer cells. Mol Cell Endocrinol. 2014;382:652–664. doi: 10.1016/j.mce.2013.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Cicatiello L, Mutarelli M, Grober OM, Paris O, Ferraro L, Ravo M, Tarallo R, Luo S, Schroth GP, Seifert M, Zinser C, Chiusano ML, Traini A, De Bortoli M, Weisz A. Estrogen receptor alpha controls a gene network in luminal-like breast cancer cells comprising multiple transcription factors and microRNAs. Am J Pathol. 2010;176:2113–2130. doi: 10.2353/ajpath.2010.090837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Santen RJ, Brodie H, Simpson ER, Siiteri PK, Brodie A. History of Aromatase: Saga of an Important Biological Mediator and Therapeutic Target. Endocr Rev. 2009;30:343–375. doi: 10.1210/er.2008-0016. [DOI] [PubMed] [Google Scholar]
  • 167.Shibahara Y, Miki Y, Onodera Y, Hata S, Chan MS, Yiu CC, Loo TY, Nakamura Y, Akahira J, Ishida T, Abe K, Hirakawa H, Chow LW, Suzuki T, Ouchi N, Sasano H. Aromatase inhibitor treatment of breast cancer cells increases the expression of let-7f, a microRNA targeting CYP19A1. J Pathol. 2012;227:357–366. doi: 10.1002/path.4019. [DOI] [PubMed] [Google Scholar]
  • 168.Diamanti-Kandarakis E, Bourguignon J-P, Giudice LC, Hauser R, Prins GS, Soto AM, Zoeller RT, Gore AC. Endocrine-Disrupting Chemicals: An Endocrine Society Scientific Statement. Endocr Rev. 2009;30:293–342. doi: 10.1210/er.2009-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Zoeller RT, Brown TR, Doan LL, Gore AC, Skakkebaek NE, Soto AM, Woodruff TJ, Vom Saal FS. Endocrine-Disrupting Chemicals and Public Health Protection: A Statement of Principles from The Endocrine Society. Endocrinology. 2012;153:4097–4110. doi: 10.1210/en.2012-1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Knower KC, To SQ, Leung Y-K, Ho S-M, Clyne CD. Endocrine disruption of the epigenome: a breast cancer link. Endocrine-Related Cancer. 2014;21:T33–T55. doi: 10.1530/ERC-13-0513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Weyandt J, Ellsworth RE, Hooke JA, Shriver CD, Ellsworth DL. Environmental chemicals and breast cancer risk--a structural chemistry perspective. Curr Med Chem. 2008;15:2680–2701. doi: 10.2174/092986708786242930. [DOI] [PubMed] [Google Scholar]
  • 172.Casals-Casas C, Desvergne B. Endocrine disruptors: from endocrine to metabolic disruption. Annu Rev Physiol. 2011;73:135–162. doi: 10.1146/annurev-physiol-012110-142200. [DOI] [PubMed] [Google Scholar]
  • 173.Collotta M, Bertazzi PA, Bollati V. Epigenetics and pesticides. Toxicology. 2013;307:35–41. doi: 10.1016/j.tox.2013.01.017. [DOI] [PubMed] [Google Scholar]
  • 174.Choi J-S, Oh J-H, Park H-J, Choi M-S, Park S-M, Kang S-J, Oh M-J, Kim SJ, Hwang SY, Yoon S. miRNA regulation of cytotoxic effects in mouse Sertoli cells exposed to nonylphenol. Reproductive Biology and Endocrinology. 2011;9:126. doi: 10.1186/1477-7827-9-126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Meunier L, Siddeek B, Vega A, Lakhdari N, Inoubli L, Bellon RP, Lemaire G, Mauduit C, Benahmed M. Perinatal Programming of Adult Rat Germ Cell Death After Exposure to Xenoestrogens: Role of microRNA miR-29 Family in the Down-Regulation of DNA Methyltransferases and Mcl-1. Endocrinology. 2012;153:1936–1947. doi: 10.1210/en.2011-1109. [DOI] [PubMed] [Google Scholar]
  • 176.Teng Y, Manavalan TT, Hu C, Medjakovic S, Jungbauer A, Klinge CM. Endocrine Disruptors Fludioxonil and Fenhexamid Stimulate miR-21 Expression in Breast Cancer Cells. Toxicol Sci. 2013;131:71–83. doi: 10.1093/toxsci/kfs290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Nowell SA, Ahn J, Ambrosone CB. Gene-nutrient interactions in cancer etiology. Nutr Rev. 2004;62:427–438. doi: 10.1111/j.1753-4887.2004.tb00014.x. [DOI] [PubMed] [Google Scholar]
  • 178.Huang H, Du G, Zhang W, Hu J, Wu D, Song L, Xia Y, Wang X. The in Vitro estrogenic activities of triclosan and triclocarban. J Appl Toxicol. 2014;34:1060–1067. doi: 10.1002/jat.3012. [DOI] [PubMed] [Google Scholar]
  • 179.Gee RH, Charles A, Taylor N, Darbre PD. Oestrogenic and androgenic activity of triclosan in breast cancer cells. J Appl Toxicol. 2008;28:78–91. doi: 10.1002/jat.1316. [DOI] [PubMed] [Google Scholar]
  • 180.Pandey DP, Picard D. miR-22 Inhibits Estrogen Signaling by Directly Targeting the Estrogen Receptor {alpha} mRNA. Mol Cell Biol. 2009;29:3783–3790. doi: 10.1128/MCB.01875-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Adams BD, Furneaux H, White BA. The Micro-Ribonucleic Acid (miRNA) miR-206 Targets the Human Estrogen Receptor-{alpha} (ER{alpha}) and Represses ER{alpha} Messenger RNA and Protein Expression in Breast Cancer Cell Lines. Mol Endocrinol. 2007;21:1132–1147. doi: 10.1210/me.2007-0022. [DOI] [PubMed] [Google Scholar]
  • 182.Liu B, Che Q, Qiu H, Bao W, Chen X, Lu W, Li B, Wan X. Elevated MiR-222-3p promotes proliferation and invasion of endometrial carcinoma via targeting ERalpha. PLoS One. 2014;9:e87563. doi: 10.1371/journal.pone.0087563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhao Y, Deng C, Wang J, Xiao J, Gatalica Z, Recker RR, Xiao GG. Let-7 family miRNAs regulate estrogen receptor alpha signaling in estrogen receptor positive breast cancer. Breast Cancer Res Treat. 2011;127:69–80. doi: 10.1007/s10549-010-0972-2. [DOI] [PubMed] [Google Scholar]
  • 184.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 Res. 2008;68:5004–5008. doi: 10.1158/0008-5472.CAN-08-0180. [DOI] [PubMed] [Google Scholar]
  • 185.Zhao J-J, Lin J, Yang H, Kong W, He L, Ma X, Coppola D, Cheng JQ. MicroRNA-221/222 negatively regulates ERalpha and associates with tamoxifen resistance in breast cancer. J Biol Chem. 2008;283:31079–31086. doi: 10.1074/jbc.M806041200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 186.Cochrane DR, Cittelly DM, Howe EN, Spoelstra NS, McKinsey EL, LaPara K, Elias A, Yee D, Richer JK. MicroRNAs link estrogen receptor alpha status and Dicer levels in breast cancer. Horm Cancer. 2010;1:306–319. doi: 10.1007/s12672-010-0043-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lu Y, Roy S, Nuovo G, Ramaswamy B, Miller T, Shapiro C, Jacob ST, Majumder S. Anti-miR-222 and -181B suppresses growth of tamoxifen resistant xenografts in mouse by targeting TIMP3 and modulating mitogenic signal. Journal of Biological Chemistry. 2011 doi: 10.1074/jbc.W118.002430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Xiong J, Yu D, Wei N, Fu H, Cai T, Huang Y, Wu C, Zheng X, Du Q, Lin D, Liang Z. An estrogen receptor alpha suppressor, microRNA-22, is downregulated in estrogen receptor alpha-positive human breast cancer cell lines and clinical samples. FEBS J. 2010 doi: 10.1111/j.1742-4658.2010.07594.x. [DOI] [PubMed] [Google Scholar]
  • 189.Leivonen SK, Makela R, Ostling P, Kohonen P, Haapa-Paananen S, Kleivi K, Enerly E, Aakula A, Hellstrom K, Sahlberg N, Kristensen VN, Borresen-Dale AL, Saviranta P, Perala M, Kallioniemi O. Protein lysate microarray analysis to identify microRNAs regulating estrogen receptor signaling in breast cancer cell lines. Oncogene. 2009;28:3926–3936. doi: 10.1038/onc.2009.241. [DOI] [PubMed] [Google Scholar]
  • 190.Sun X, Qin S, Fan C, Xu C, Du N, Ren H. Let-7: a regulator of the ERalpha signaling pathway in human breast tumors and breast cancer stem cells. Oncol Rep. 2013;29:2079–2087. doi: 10.3892/or.2013.2330. [DOI] [PubMed] [Google Scholar]
  • 191.Cui J, Bi M, Overstreet AM, Yang Y, Li H, Leng Y, Qian K, Huang Q, Zhang C, Lu Z, Chen J, Sun T, Wu R, Sun Y, Song H, Wei X, Jing P, Meredith A, Yang X, Zhang C. MiR-873 regulates ER[alpha] transcriptional activity and tamoxifen resistance via targeting CDK3 in breast cancer cells. Oncogene. 2014 doi: 10.1038/onc.2015.201. [DOI] [PubMed] [Google Scholar]
  • 192.Hossain A, Kuo MT, Saunders GF. Mir-17-5p Regulates Breast Cancer Cell Proliferation by Inhibiting Translation of AIB1 mRNA. Mol Cell Biol. 2006;26:8191–8201. doi: 10.1128/MCB.00242-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Jiang HL, Yu H, Ma X, Xu D, Lin GF, Ma DY, Jin JZ. MicroRNA-195 regulates steroid receptor coactivator-3 protein expression in hepatocellular carcinoma cells. Tumour Biol. 2014;35:6955–6960. doi: 10.1007/s13277-014-1933-x. [DOI] [PubMed] [Google Scholar]
  • 194.Foley NH, Bray I, Watters KM, Das S, Bryan K, Bernas T, Prehn JHM, Stallings RL. MicroRNAs 10a and 10b are potent inducers of neuroblastoma cell differentiation through targeting of nuclear receptor corepressor 2. Cell Death Differ. 2011;18:1089–1098. doi: 10.1038/cdd.2010.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Wu J, Bao J, Wang L, Hu Y, Xu C. MicroRNA-184 downregulates nuclear receptor corepressor 2 in mouse spermatogenesis. BMC Dev Biol. 2011;11:64. doi: 10.1186/1471-213X-11-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Zhou R, Li X, Hu G, Gong AY, Drescher KM, Chen XM. miR-16 targets transcriptional corepressor SMRT and modulates NF-kappaB-regulated transactivation of interleukin-8 gene. PLoS One. 2012;7:e30772. doi: 10.1371/journal.pone.0030772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Bui-Nguyen TM, Pakala SB, Sirigiri DR, Martin E, Murad F, Kumar R. Stimulation of Inducible Nitric Oxide by Hepatitis B Virus Transactivator Protein HBx Requires MTA1 Coregulator. J Biol Chem. 2010;285:6980–6986. doi: 10.1074/jbc.M109.065987. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 198.Jiang A, Zhang S, Li Z, Liang R, Ren S, Li J, Pu Y, Yang J. miR-615-3p promotes the phagocytic capacity of splenic macrophages by targeting ligand-dependent nuclear receptor corepressor in cirrhosis-related portal hypertension. Exp Biol Med. 2011;236:672–680. doi: 10.1258/ebm.2011.010349. [DOI] [PubMed] [Google Scholar]
  • 199.Cheng C, Fu X, Alves P, Gerstein M. mRNA expression profiles show differential regulatory effects of microRNAs between estrogen receptor-positive and estrogen receptor-negative breast cancer. Genome Biol. 2009;10:R90. doi: 10.1186/gb-2009-10-9-r90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Klinge CM. miRNAs and estrogen action. Trends in Endocrinology & Metabolism. 2012;23:223–233. doi: 10.1016/j.tem.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Zhao M, Ramaswamy B. Mechanisms and therapeutic advances in the management of endocrine-resistant breast cancer. World journal of clinical oncology. 2014;5:248–262. doi: 10.5306/wjco.v5.i3.248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Rao X, Di Leva G, Li M, Fang F, Devlin C, Hartman-Frey C, Burow ME, Ivan M, Croce CM, Nephew KP. MicroRNA-221/222 confers breast cancer fulvestrant resistance by regulating multiple signaling pathways. Oncogene. 2011;30:1082–1097. doi: 10.1038/onc.2010.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Shimono Y, Zabala M, Cho RW, Lobo N, Dalerba P, Qian D, Diehn M, Liu H, Panula SP, Chiao E, Dirbas FM, Somlo G, Pera RA, Lao K, Clarke MF. Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell. 2009;138:592–603. doi: 10.1016/j.cell.2009.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Sun X, Jiao X, Pestell TG, Fan C, Qin S, Mirabelli E, Ren H, Pestell RG. MicroRNAs and cancer stem cells: the sword and the shield. Oncogene. 2014;33:4967–4977. doi: 10.1038/onc.2013.492. [DOI] [PubMed] [Google Scholar]
  • 205.Bronzert DA, Greene GL, Lippman ME. Selection and characterization of a breast cancer cell line resistant to the antiestrogen LY 117018. Endocrinology. 1985;117:1409–1417. doi: 10.1210/endo-117-4-1409. [DOI] [PubMed] [Google Scholar]
  • 206.Mullick A, Chambon P. Characterization of the estrogen receptor in two antiestrogen-resistant cell lines, LY2 and T47D. Cancer Res. 1990;50:333–338. [PubMed] [Google Scholar]
  • 207.Crawford AC, Riggins RB, Shajahan AN, Zwart A, Clarke R. Co-Inhibition of BCL-W and BCL2 Restores Antiestrogen Sensitivity through BECN1 and Promotes an Autophagy-Associated Necrosis. PLoS ONE. 2010;5:e8604. doi: 10.1371/journal.pone.0008604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Sreekumar R, Sayan BS, Mirnezami AH, Sayan AE. MicroRNA Control of Invasion and Metastasis Pathways. Frontiers in genetics. 2011;2:58. doi: 10.3389/fgene.2011.00058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Bracken CP, Gregory PA, Kolesnikoff N, Bert AG, Wang J, Shannon MF, Goodall GJ. A Double-Negative Feedback Loop between ZEB1-SIP1 and the microRNA-200 Family Regulates Epithelial-Mesenchymal Transition. Cancer Res. 2008;68:7846–7854. doi: 10.1158/0008-5472.CAN-08-1942. [DOI] [PubMed] [Google Scholar]
  • 210.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 Dev. 2008;22:894–907. doi: 10.1101/gad.1640608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.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. J Biol Chem. 2008;283:14910–14914. doi: 10.1074/jbc.C800074200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T. A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008;9:582–589. doi: 10.1038/embor.2008.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Hurteau GJ, Carlson JA, Spivack SD, Brock GJ. Overexpression of the microRNA hsa-miR-200c leads to reduced expression of transcription factor 8 and increased expression of E-cadherin. Cancer Res. 2007;67:7972–7976. doi: 10.1158/0008-5472.CAN-07-1058. [DOI] [PubMed] [Google Scholar]
  • 214.Manavalan TT, Teng Y, Litchfield LM, Muluhngwi P, Al-Rayyan N, Klinge CM. Reduced Expression of miR-200 Family Members Contributes to Antiestrogen Resistance in LY2 Human Breast Cancer Cells. PLoS ONE. 2013;8:e62334. doi: 10.1371/journal.pone.0062334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008;10:593–601. doi: 10.1038/ncb1722. [DOI] [PubMed] [Google Scholar]
  • 216.Vrba L, Jensen TJ, Garbe JC, Heimark RL, Cress AE, Dickinson S, Stampfer MR, Futscher BW. Role for DNA methylation in the regulation of miR-200c and miR-141 expression in normal and cancer cells. PLoS One. 2010;5:e8697. doi: 10.1371/journal.pone.0008697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Neves R, Scheel C, Weinhold S, Honisch E, Iwaniuk KM, Trompeter HI, Niederacher D, Wernet P, Santourlidis S, Uhrberg M. Role of DNA methylation in miR-200c/141 cluster silencing in invasive breast cancer cells. BMC research notes. 2010;3:219. doi: 10.1186/1756-0500-3-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Davalos V, Moutinho C, Villanueva A, Boque R, Silva P, Carneiro F, Esteller M. Dynamic epigenetic regulation of the microRNA-200 family mediates epithelial and mesenchymal transitions in human tumorigenesis. Oncogene. 2012;31:2062–2074. doi: 10.1038/onc.2011.383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Lyng MB, Lænkholm A-V, Søkilde R, Gravgaard KH, Litman T, Ditzel HJ. Global MicroRNA Expression Profiling of High-Risk ER+ Breast Cancers from Patients Receiving Adjuvant Tamoxifen Mono-Therapy: A DBCG Study. PLoS ONE. 2012;7:e36170. doi: 10.1371/journal.pone.0036170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Cittelly DM, Das PM, Spoelstra NS, Edgerton SM, Richer JK, Thor AD, Jones FE. Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol Cancer. 2010;9:317. doi: 10.1186/1476-4598-9-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Bergamaschi A, Katzenellenbogen BS. Tamoxifen downregulation of miR-451 increases 14-3-3zeta and promotes breast cancer cell survival and endocrine resistance. Oncogene. 2011 doi: 10.1038/onc.2011.223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Fowler AM, Santen RJ, Allred DC. "Dwarf" Estrogen Receptor in Breast Cancer and Resistance to Tamoxifen. J Clin Oncol. 2009;27:3413–3415. doi: 10.1200/JCO.2009.21.8776. [DOI] [PubMed] [Google Scholar]
  • 223.Zhao Y, Deng C, Lu W, Xiao J, Ma D, Guo M, Recker RR, Gatalica Z, Wang Z, Xiao GG. let-7 microRNAs induce tamoxifen sensitivity by downregulation of estrogen receptor alpha signaling in breast cancer. Mol Med. 2011;17:1233–1241. doi: 10.2119/molmed.2010.00225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Ward A, Balwierz A, Zhang JD, Kublbeck M, Pawitan Y, Hielscher T, Wiemann S, Sahin O. Re-expression of microRNA-375 reverses both tamoxifen resistance and accompanying EMT-like properties in breast cancer. Oncogene. 2013;32:1173–1182. doi: 10.1038/onc.2012.128. [DOI] [PubMed] [Google Scholar]
  • 225.Flor I, Bullerdiek J. The dark side of a success story: microRNAs of the C19MC cluster in human tumours. J Pathol. 2012;227:270–274. doi: 10.1002/path.4014. [DOI] [PubMed] [Google Scholar]
  • 226.Bortolin-Cavaille ML, Dance M, Weber M, Cavaille J. C19MC microRNAs are processed from introns of large Pol-II, non-protein-coding transcripts. Nucleic Acids Res. 2009;37:3464–3473. doi: 10.1093/nar/gkp205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Ward A, Shukla K, Balwierz A, Soons Z, König R, Sahin Ö, Wiemann S. microRNA-519a is a novel oncomir conferring tamoxifen resistance by targeting a network of tumor-suppressor genes in ER+ breast cancer. The Journal of Pathology:n/a-n/a. 2014 doi: 10.1002/path.4363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Vlachos IS, Paraskevopoulou MD, Karagkouni D, Georgakilas G, Vergoulis T, Kanellos I, Anastasopoulos I-L, Maniou S, Karathanou K, Kalfakakou D, Fevgas A, Dalamagas T, Hatzigeorgiou AG. DIANA-TarBase v7.0: indexing more than half a million experimentally supported miRNA:mRNA interactions. Nucleic Acids Res. 2015;43:D153–D159. doi: 10.1093/nar/gku1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Cochrane D, Cittelly D, Howe E, Spoelstra N, McKinsey E, LaPara K, Elias A, Yee D, Richer J. MicroRNAs Link Estrogen Receptor Alpha Status and Dicer Levels in Breast Cancer. Hormones and Cancer. 2010;1:306–319. doi: 10.1007/s12672-010-0043-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.White NMA, Fatoohi E, Metias M, Jung K, Stephan C, Yousef GM. Metastamirs: a stepping stone towards improved cancer management. Nat Rev Clin Oncol. 2011;8:75–84. doi: 10.1038/nrclinonc.2010.173. [DOI] [PubMed] [Google Scholar]
  • 231.Forman JJ, Legesse-Miller A, Coller HA. A search for conserved sequences in coding regions reveals that the let-7 microRNA targets Dicer within its coding sequence. Proceedings of the National Academy of Sciences. 2008;105:14879–14884. doi: 10.1073/pnas.0803230105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Ji J, Zhao L, Budhu A, Forgues M, Jia H-L, Qin L-X, Ye Q-H, Yu J, Shi X, Tang Z-Y, Wang XW. Let-7g targets collagen type I α2 and inhibits cell migration in hepatocellular carcinoma. J Hepatol. 2010;52:690–697. doi: 10.1016/j.jhep.2009.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Masuda M, Miki Y, Hata S, Takagi K, Sakurai M, Ono K, Suzuki K, Yang Y, Abe E, Hirakawa H, Ishida T, Suzuki T, Ohuchi N, Sasano H. An induction of microRNA, miR-7 through estrogen treatment in breast carcinoma. J Transl Med. 2012;10(Suppl 1):S2. doi: 10.1186/1479-5876-10-S1-S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Xu K, Chen Z, Qin C, Song X. miR-7 inhibits colorectal cancer cell proliferation and induces apoptosis by targeting XRCC2. OncoTargets and therapy. 2014;7:325–332. doi: 10.2147/OTT.S59364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Meza-Sosa KF, Pérez-García EI, Camacho-Concha N, López-Gutiérrez O, Pedraza-Alva G, Pérez-Martínez L. MiR-7 Promotes Epithelial Cell Transformation by Targeting the Tumor Suppressor KLF4. PLoS ONE. 2014;9:e103987. doi: 10.1371/journal.pone.0103987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Edvardsson K, Nguyen-Vu T, M.Kalasekar S, Pontén F, Gustafsson J-Å, Williams C. Estrogen receptor β expression induces changes in the microRNA pool in human colon cancer cells. Carcinogenesis. 2013 doi: 10.1093/carcin/bgt067. [DOI] [PubMed] [Google Scholar]
  • 237.Chan M, Liaw CS, Ji SM, Tan HH, Wong CY, Thike AA, Tan PH, Ho GH, Lee AS-G. Identification of Circulating MicroRNA Signatures for Breast Cancer Detection. Clin Cancer Res. 2013;19:4477–4487. doi: 10.1158/1078-0432.CCR-12-3401. [DOI] [PubMed] [Google Scholar]
  • 238.Biagioni F, Bossel Ben-Moshe N, Fontemaggi G, Canu V, Mori F, Antoniani B, Di Benedetto A, Santoro R, Germoni S, De Angelis F, Cambria A, Avraham R, Grasso G, Strano S, Muti P, Mottolese M, Yarden Y, Domany E, Blandino G. miR-10b*, a master inhibitor of the cell cycle, is down-regulated in human breast tumours. EMBO Mol Med. 2012;4:1214–1229. doi: 10.1002/emmm.201201483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Ofir M, Hacohen D, Ginsberg D. miR-15 and miR-16 Are Direct Transcriptional Targets of E2F1 that Limit E2F-Induced Proliferation by Targeting Cyclin E. Mol Cancer Res. 2011;9:440–447. doi: 10.1158/1541-7786.MCR-10-0344. [DOI] [PubMed] [Google Scholar]
  • 240.Masri S, Liu Z, Phung S, Wang E, Yuan Y-C, Chen S. The role of microRNA-128a in regulating TGFbeta signaling in letrozole-resistant breast cancer cells. Breast Cancer Res Treat. 2010;124:89–99. doi: 10.1007/s10549-009-0716-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Castellano L, Giamas G, Jacob J, Coombes RC, Lucchesi W, Thiruchelvam P, Barton G, Jiao LR, Wait R, Waxman J, Hannon GJ, Stebbing J. The estrogen receptor-alpha induced microRNA signature regulates itself and its transcriptional response. Proc Natl Acad Sci USA. 2009;106:15732–15737. doi: 10.1073/pnas.0906947106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Wang G, Wang Y, Shen C, Huang Y-w, Huang K, Huang THM, Nephew KP, Li L, Liu Y. RNA Polymerase II Binding Patterns Reveal Genomic Regions Involved in MicroRNA Gene Regulation. PLoS ONE. 2010;5:e13798. doi: 10.1371/journal.pone.0013798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Zhu H, Han C, Lu D, Wu T. miR-17-92 Cluster Promotes Cholangiocarcinoma Growth: Evidence for PTEN as Downstream Target and IL-6/Stat3 as Upstream Activator. Am J Pathol. 2014;184:2828–2839. doi: 10.1016/j.ajpath.2014.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Yoshimoto N, Toyama T, Takahashi S, Sugiura H, Endo Y, Iwasa M, Fujii Y, Yamashita H. Distinct expressions of microRNAs that directly target estrogen receptor α in human breast cancer. Breast Cancer Res Treat. 2011;130:331–339. doi: 10.1007/s10549-011-1672-2. [DOI] [PubMed] [Google Scholar]
  • 245.Hannafon B, Sebastiani P, de las Morenas A, Lu J, Rosenberg C. Expression of microRNAs and their gene targets are dysregulated in pre-invasive breast cancer. Breast Cancer Research. 2011;13:R24. doi: 10.1186/bcr2839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Smith AL, Iwanaga R, Drasin DJ, Micalizzi DS, Vartuli RL, Tan AC, Ford HL. The miR-106b-25 cluster targets Smad7, activates TGF-[beta] signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer. Oncogene. 2012;31:5162–5171. doi: 10.1038/onc.2012.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Zhang H, Zuo Z, Lu X, Wang L, Wang H, Zhu Z. MiR-25 regulates apoptosis by targeting Bim in human ovarian cancer. Oncol Rep. 2012;27:594–598. doi: 10.3892/or.2011.1530. [DOI] [PubMed] [Google Scholar]
  • 248.Razumilava N, Bronk SF, Smoot RL, Fingas CD, Werneburg NW, Roberts LR, Mott JL. miR-25 targets TNF-related apoptosis inducing ligand (TRAIL) death receptor-4 and promotes apoptosis resistance in cholangiocarcinoma. Hepatology. 2012;55:465–475. doi: 10.1002/hep.24698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Marchi S, Lupini L, Patergnani S, Rimessi A, Missiroli S, Bonora M, Bononi A, Corra F, Giorgi C, De Marchi E, Poletti F, Gafa R, Lanza G, Negrini M, Rizzuto R, Pinton P. Downregulation of the mitochondrial calcium uniporter by cancer-related miR-25. Curr Biol. 2013;23:58–63. doi: 10.1016/j.cub.2012.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Li Q, Zou C, Zou C, Han Z, Xiao H, Wei H, Wang W, Zhang L, Zhang X, Tang Q, Zhang C, Tao J, Wang X, Gao X. MicroRNA-25 functions as a potential tumor suppressor in colon cancer by targeting Smad7. Cancer Lett. 2013;335:168–174. doi: 10.1016/j.canlet.2013.02.029. [DOI] [PubMed] [Google Scholar]
  • 251.Feng S, Pan W, Jin Y, Zheng J. MiR-25 promotes ovarian cancer proliferation and motility by targeting LATS2. Tumour Biol. 2014 doi: 10.1007/s13277-014-2546-0. [DOI] [PubMed] [Google Scholar]
  • 252.Zhao H, Wang Y, Yang L, Jiang R, Li W. MiR-25 promotes gastric cancer cells growth and motility by targeting RECK. Mol Cell Biochem. 2014;385:207–213. doi: 10.1007/s11010-013-1829-x. [DOI] [PubMed] [Google Scholar]
  • 253.Pan Q, Luo X, Chegini N. Differential expression of microRNAs in myometrium and leiomyomas and regulation by ovarian steroids. J Cell Mol Med. 2008;12:227–240. doi: 10.1111/j.1582-4934.2007.00207.x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 254.Chen L, Zheng J, Zhang Y, Yang L, Wang J, Ni J, Cui D, Yu C, Cai Z. Tumor-specific Expression of MicroRNA-26a Suppresses Human Hepatocellular Carcinoma Growth via Cyclin-dependent and -independent Pathways. Mol Ther. 2011;19:1521–1528. doi: 10.1038/mt.2011.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Tan S, Ding K, Li R, Zhang W, Li G, Kong X, Qian P, Lobie PE, Zhu T. Identification of miR-26 as a key mediator of estrogen stimulated cell proliferation by targeting CHD1, GREB1 and KPNA2. Breast Cancer Res. 2014;16:R40. doi: 10.1186/bcr3644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Zhang L, Chen X, Shi Y, Zhou B, Du C, Liu Y, Han S, Yin J, Peng B, He X, Liu W. miR-27a suppresses EV71 replication by directly targeting EGFR. Virus Genes. 2014 doi: 10.1007/s11262-014-1114-4. [DOI] [PubMed] [Google Scholar]
  • 257.Jiang J, Lv X, Fan L, Huang G, Zhan Y, Wang M, Lu H. MicroRNA-27b suppresses growth and invasion of NSCLC cells by targeting Sp1. Tumour Biol. 2014 doi: 10.1007/s13277-014-2294-1. [DOI] [PubMed] [Google Scholar]
  • 258.Wan L, Zhang L, Fan K, Wang J. MiR-27b targets LIMK1 to inhibit growth and invasion of NSCLC cells. Mol Cell Biochem. 2014;390:85–91. doi: 10.1007/s11010-013-1959-1. [DOI] [PubMed] [Google Scholar]
  • 259.Lee JJ, Drakaki A, Iliopoulos D, Struhl K. MiR-27b targets PPARgamma to inhibit growth, tumor progression and the inflammatory response in neuroblastoma cells. Oncogene. 2012;31:3818–3825. doi: 10.1038/onc.2011.543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Wang Y, Zhang X, Li H, Yu J, Ren X. The role of miRNA-29 family in cancer. Eur J Cell Biol. 2013;92:123–128. doi: 10.1016/j.ejcb.2012.11.004. [DOI] [PubMed] [Google Scholar]
  • 261.Ichikawa T, Sato F, Terasawa K, Tsuchiya S, Toi M, Tsujimoto G, Shimizu K. Trastuzumab produces therapeutic actions by upregulating miR-26a and miR-30b in breast cancer cells. PLoS One. 2012;7:e31422. doi: 10.1371/journal.pone.0031422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Liao WT, Ye YP, Zhang NJ, Li TT, Wang SY, Cui YM, Qi L, Wu P, Jiao HL, Xie YJ, Zhang C, Wang JX, Ding YQ. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2. J Pathol. 2014;232:415–427. doi: 10.1002/path.4309. [DOI] [PubMed] [Google Scholar]
  • 263.Singh B, Ronghe AM, Chatterjee A, Bhat NK, Bhat HK. MicroRNA-93 regulates NRF2 expression and is associated with breast carcinogenesis. Carcinogenesis. 2013;34:1165–1172. doi: 10.1093/carcin/bgt026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Kim K, Madak-Erdogan Z, Ventrella R, Katzenellenbogen BS. A MicroRNA196a2* and TP63 circuit regulated by estrogen receptor-alpha and ERK2 that controls breast cancer proliferation and invasiveness properties. Horm Cancer. 2013;4:78–91. doi: 10.1007/s12672-012-0129-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Li W, Zang W, Liu P, Wang Y, Du Y, Chen X, Deng M, Sun W, Wang L, Zhao G, Zhai B. MicroRNA-124 inhibits cellular proliferation and invasion by targeting Ets-1 in breast cancer. Tumour Biol. 2014 doi: 10.1007/s13277-014-2402-2. [DOI] [PubMed] [Google Scholar]
  • 266.Chen Q, Lu G, Cai Y, Li Y, Xu R, Ke Y, Zhang S. MiR-124-5p inhibits the growth of high-grade gliomas through posttranscriptional regulation of LAMB1. Neuro Oncol. 2014;16:637–651. doi: 10.1093/neuonc/not300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Gu X, Meng S, Liu S, Jia C, Fang Y, Li S, Fu C, Song Q, Lin L, Wang X. miR-124 represses ROCK1 expression to promote neurite elongation through activation of the PI3K/Akt signal pathway. J Mol Neurosci. 2014;52:156–165. doi: 10.1007/s12031-013-0190-6. [DOI] [PubMed] [Google Scholar]
  • 268.Li L, Luo J, Wang B, Wang D, Xie X, Yuan L, Guo J, Xi S, Gao J, Lin X, Kong Y, Xu X, Tang H, Xie X, Liu M. Microrna-124 targets flotillin-1 to regulate proliferation and migration in breast cancer. Mol Cancer. 2013;12:163. doi: 10.1186/1476-4598-12-163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Zhang H, Wang Q, Zhao Q, Di W. MiR-124 inhibits the migration and invasion of ovarian cancer cells by targeting SphK1. Journal of ovarian research. 2013;6:84. doi: 10.1186/1757-2215-6-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Han ZB, Yang Z, Chi Y, Zhang L, Wang Y, Ji Y, Wang J, Zhao H, Han ZC. MicroRNA-124 suppresses breast cancer cell growth and motility by targeting CD151. Cell Physiol Biochem. 2013;31:823–832. doi: 10.1159/000350100. [DOI] [PubMed] [Google Scholar]
  • 271.Liu K, Zhao H, Yao H, Lei S, Lei Z, Li T, Qi H. MicroRNA-124 regulates the proliferation of colorectal cancer cells by targeting iASPP. BioMed research international. 2013;2013:867537. doi: 10.1155/2013/867537. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 272.Liang YJ, Wang QY, Zhou CX, Yin QQ, He M, Yu XT, Cao DX, Chen GQ, He JR, Zhao Q. MiR-124 targets Slug to regulate epithelial-mesenchymal transition and metastasis of breast cancer. Carcinogenesis. 2013;34:713–722. doi: 10.1093/carcin/bgs383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Ma S, Tang KH, Chan YP, Lee TK, Kwan PS, Castilho A, Ng I, Man K, Wong N, To K-F, Zheng B-J, Lai PBS, Lo CM, Chan KW, Guan X-Y. miR-130b Promotes CD133+ Liver Tumor-Initiating Cell Growth and Self-Renewal via Tumor Protein 53-Induced Nuclear Protein 1. Cell Stem Cell. 2010;7:694–707. doi: 10.1016/j.stem.2010.11.010. [DOI] [PubMed] [Google Scholar]
  • 274.Li BL, Lu C, Lu W, Yang TT, Qu J, Hong X, Wan XP. miR-130b is an EMT-related microRNA that targets DICER1 for aggression in endometrial cancer. Medical oncology (Northwood, London, England) 2013;30:484. doi: 10.1007/s12032-013-0484-0. [DOI] [PubMed] [Google Scholar]
  • 275.Chen Y, Song YX, Wang ZN. The microRNA-148/152 family: multi-faceted players. Mol Cancer. 2013;12:43. doi: 10.1186/1476-4598-12-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Jin L, Hu WL, Jiang CC, Wang JX, Han CC, Chu P, Zhang LJ, Thorne RF, Wilmott J, Scolyer RA, Hersey P, Zhang XD, Wu M. MicroRNA-149*, a p53-responsive microRNA, functions as an oncogenic regulator in human melanoma. Proceedings of the National Academy of Sciences. 2011;108:15840–15845. doi: 10.1073/pnas.1019312108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Chan SH, Huang WC, Chang JW, Chang KJ, Kuo WH, Wang MY, Lin KY, Uen YH, Hou MF, Lin CM, Jang TH, Tu CW, Lee YR, Lee YH, Tien MT, Wang LH. MicroRNA-149 targets GIT1 to suppress integrin signaling and breast cancer metastasis. Oncogene. 2014;33:4496–4507. doi: 10.1038/onc.2014.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Lin RJ, Lin YC, Yu AL. miR-149* induces apoptosis by inhibiting Akt1 and E2F1 in human cancer cells. Mol Carcinog. 2010;49:719–727. doi: 10.1002/mc.20647. [DOI] [PubMed] [Google Scholar]
  • 279.Zhang C, Zhao J, Deng H. 17beta-estradiol up-regulates miR-155 expression and reduces TP53INP1 expression in MCF-7 breast cancer cells. Mol Cell Biochem. 2013;379:201–211. doi: 10.1007/s11010-013-1642-6. [DOI] [PubMed] [Google Scholar]
  • 280.Eichelser C, Flesch-Janys D, Chang-Claude J, Pantel K, Schwarzenbach H. Deregulated Serum Concentrations of Circulating Cell–Free MicroRNAs miR-17, miR-34a, miR-155, and miR-373 in Human Breast Cancer Development and Progression. Clin Chem. 2013;59:1489–1496. doi: 10.1373/clinchem.2013.205161. [DOI] [PubMed] [Google Scholar]
  • 281.Dinami R, Ercolani C, Petti E, Piazza S, Ciani Y, Sestito R, Sacconi A, Biagioni F, le Sage C, Agami R, Benetti R, Mottolese M, Schneider C, Blandino G, Schoeftner S. miR-155 drives telomere fragility in human breast cancer by targeting TRF1. Cancer Res. 2014;74:4145–4156. doi: 10.1158/0008-5472.CAN-13-2038. [DOI] [PubMed] [Google Scholar]
  • 282.Zhang CM, Zhao J, Deng HY. MiR-155 promotes proliferation of human breast cancer MCF-7 cells through targeting tumor protein 53-induced nuclear protein 1. J Biomed Sci. 2013;20:79. doi: 10.1186/1423-0127-20-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Chu HW, Cheng CW, Chou WC, Hu LY, Wang HW, Hsiung CN, Hsu HM, Wu PE, Hou MF, Shen CY, Yu JC. A novel estrogen receptor-microRNA 190a-PAR-1-pathway regulates breast cancer progression, a finding initially suggested by genome-wide analysis of loci associated with lymph-node metastasis. Hum Mol Genet. 2014;23:355–367. doi: 10.1093/hmg/ddt426. [DOI] [PubMed] [Google Scholar]
  • 284.Di Leva G, Piovan C, Gasparini P, Ngankeu A, Taccioli C, Briskin D, Cheung DG, Bolon B, Anderlucci L, Alder H, Nuovo G, Li M, Iorio MV, Galasso M, Ramasamy S, Marcucci G, Perrotti D, Powell KA, Bratasz A, Garofalo M, Nephew KP, Croce CM. Estrogen Mediated-Activation of miR-191/425 Cluster Modulates Tumorigenicity of Breast Cancer Cells Depending on Estrogen Receptor Status. PLoS Genet. 2013;9:e1003311. doi: 10.1371/journal.pgen.1003311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Nagpal N, Ahmad HM, Molparia B, Kulshreshtha R. MicroRNA-191, an estrogen-responsive microRNA, functions as an oncogenic regulator in human breast cancer. Carcinogenesis. 2013;34:1889–1899. doi: 10.1093/carcin/bgt107. [DOI] [PubMed] [Google Scholar]
  • 286.Li XF, Yan PJ, Shao ZM. Downregulation of miR-193b contributes to enhance urokinase-type plasminogen activator (uPA) expression and tumor progression and invasion in human breast cancer. Oncogene. 2009;28:3937–3948. doi: 10.1038/onc.2009.245. [DOI] [PubMed] [Google Scholar]
  • 287.Leivonen S-K, Rokka A, Östling P, Kohonen P, Corthals GL, Kallioniemi O, Perälä M. Identification of miR-193b targets in breast cancer cells and systems biological analysis of their functional impact. Molecular & Cellular Proteomics. 2011;10:M110 005322. doi: 10.1074/mcp.M110.005322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Mets E, Van der Meulen J, Van Peer G, Boice M, Mestdagh P, Van de Walle I, Lammens T, Goossens S, De Moerloose B, Benoit Y, Van Roy N, Clappier E, Poppe B, Vandesompele J, Wendel HG, Taghon T, Rondou P, Soulier J, Van Vlierberghe P, Speleman F. MicroRNA-193b-3p acts as a tumor suppressor by targeting the MYB oncogene in T-cell acute lymphoblastic leukemia. Leukemia. 2014 doi: 10.1038/leu.2014.276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Katoh M. Cardio-miRNAs and onco-miRNAs: circulating miRNA-based diagnostics for non-cancerous and cancerous diseases. Frontiers in cell and developmental biology. 2014;2:61. doi: 10.3389/fcell.2014.00061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Rokavec M, Wu W, Luo J-L. IL6-Mediated Suppression of miR-200c Directs Constitutive Activation of Inflammatory Signaling Circuit Driving Transformation and Tumorigenesis. Mol Cell. 2012;45:777–789. doi: 10.1016/j.molcel.2012.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Osada H, Takahashi T. MicroRNAs in biological processes and carcinogenesis. Carcinogenesis. 2007;28:2–12. doi: 10.1093/carcin/bgl185. [DOI] [PubMed] [Google Scholar]
  • 292.Wong QW-L, Ching AK-K, Chan AW-H, Choy K-W, To K-F, Lai PB-S, Wong N. MiR-222 Overexpression Confers Cell Migratory Advantages in Hepatocellular Carcinoma through Enhancing AKT Signaling. Clin Cancer Res. 2010;16:867–875. doi: 10.1158/1078-0432.CCR-09-1840. [DOI] [PubMed] [Google Scholar]
  • 293.Qian K, Hu L, Chen H, Li H, Liu N, Li Y, Ai J, Zhu G, Tang Z, Zhang H. Has-miR-222 Is Involved in Differentiation of Endometrial Stromal Cells in Vitro. Endocrinology. 2009;150:4734–4743. doi: 10.1210/en.2008-1629. [DOI] [PubMed] [Google Scholar]
  • 294.Di Leva G, Gasparini P, Piovan C, Ngankeu A, Garofalo M, Taccioli C, Iorio MV, Li M, Volinia S, Alder H, Nakamura T, Nuovo G, Liu Y, Nephew KP, Croce CM. MicroRNA Cluster 221–222 and Estrogen Receptor {alpha} Interactions in Breast Cancer. J Natl Cancer Inst. 2010;102:706–721. doi: 10.1093/jnci/djq102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Pinho FG, Frampton AE, Nunes J, Krell J, Alshaker H, Jacob J, Pellegrino L, Roca-Alonso L, de Giorgio A, Harding V, Waxman J, Stebbing J, Pchejetski D, Castellano L. Downregulation of microRNA-515-5p by the estrogen receptor modulates sphingosine kinase-1 and breast cancer cell proliferation. Cancer Res. 2013 doi: 10.1158/0008-5472.CAN-13-0158. [DOI] [PubMed] [Google Scholar]
  • 296.Maillot G, Lacroix-Triki M, Pierredon S, Gratadou L, Schmidt S, Benes V, Roche H, Dalenc F, Auboeuf D, Millevoi S, Vagner S. Widespread Estrogen-Dependent Repression of microRNAs Involved in Breast Tumor Cell Growth. Cancer Res. 2009;69:8332–8340. doi: 10.1158/0008-5472.CAN-09-2206. [DOI] [PubMed] [Google Scholar]
  • 297.Qian P, Zuo Z, Wu Z, Meng X, Li G, Wu Z, Zhang W, Tan S, Pandey V, Yao Y, Wang P, Zhao L, Wang J, Wu Q, Song E, Lobie PE, Yin Z, Zhu T. Pivotal Role of Reduced let-7g Expression in Breast Cancer Invasion and Metastasis. Cancer Res. 2011;71:6463–6474. doi: 10.1158/0008-5472.CAN-11-1322. [DOI] [PubMed] [Google Scholar]
  • 298.Ujihira T, Ikeda K, Suzuki T, Yamaga R, Sato W, Horie-Inoue K, Shigekawa T, Osaki A, Saeki T, Okamoto K, Takeda S, Inoue S. MicroRNA-574-3p, identified by microRNA library-based functional screening, modulates tamoxifen response in breast cancer. Sci Rep. 2015;5 doi: 10.1038/srep07641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Yu X, Zhang X, Dhakal IB, Beggs M, Kadlubar S, Luo D. Induction of cell proliferation and survival genes by estradiol-repressed microRNAs in breast cancer cells. BMC Cancer. 2012;12:29. doi: 10.1186/1471-2407-12-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Pan Q, Luo X, Toloubeydokhti T, Chegini N. The expression profile of micro-RNA in endometrium and endometriosis and the influence of ovarian steroids on their expression. Mol Hum Reprod. 2007;13:797–806. doi: 10.1093/molehr/gam063. [DOI] [PubMed] [Google Scholar]
  • 301.Selcuklu SD, Donoghue MTA, Kerin MJ, Spillane C. Regulatory interplay between miR-21, JAG1 and 17beta-estradiol (E2) in breast cancer cells. Biochem Biophys Res Commun. 2012;423:234–239. doi: 10.1016/j.bbrc.2012.05.074. [DOI] [PubMed] [Google Scholar]
  • 302.Saumet A, Vetter G, Bouttier M, Antoine E, Roubert C, Orsetti B, Theillet C, Lecellier CH. Estrogen and retinoic acid antagonistically regulate several microRNA genes to control aerobic glycolysis in breast cancer cells. Molecular bioSystems. 2012;8:3242–3253. doi: 10.1039/c2mb25298h. [DOI] [PubMed] [Google Scholar]
  • 303.Zhao G, Guo J, Li D, Jia C, Yin W, Sun R, Lv Z, Cong X. MicroRNA-34a suppresses cell proliferation by targeting LMTK3 in human breast cancer MCF-7 cell line. DNA Cell Biol. 2013;32:699–707. doi: 10.1089/dna.2013.2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Nanni S, Aiello A, Re A, Guffanti A, Benvenuti V, Colussi C, Castro-Vega LJ, Felsani A, Londono-Vallejo A, Capogrossi MC, Bacchetti S, Gaetano C, Pontecorvi A, Farsetti A. Estrogen-Dependent Dynamic Profile of eNOS-DNA Associations in Prostate Cancer. PLoS ONE. 2013;8:e62522. doi: 10.1371/journal.pone.0062522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Yamakuchi M, Ferlito M, Lowenstein CJ. miR-34a repression of SIRT1 regulates apoptosis. Proceedings of the National Academy of Sciences. 2008;105:13421–13426. doi: 10.1073/pnas.0801613105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Cicatiello L, Mutarelli M, Grober OMV, Paris O, Ferraro L, Ravo M, Tarallo R, Luo S, Schroth GP, Seifert M, Zinser C, Chiusano ML, Traini A, De Bortoli M, Weisz A. Estrogen Receptor {alpha} Controls a Gene Network in Luminal-Like Breast Cancer Cells Comprising Multiple Transcription Factors and MicroRNAs. Am J Pathol. 2010;176:2113–2130. doi: 10.2353/ajpath.2010.090837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Zhang Y, Eades G, Yao Y, Li Q, Zhou Q. Estrogen Receptor α Signaling Regulates Breast Tumor-initiating Cells by Down-regulating miR-140 Which Targets the Transcription Factor SOX2. J Biol Chem. 2012;287:41514–41522. doi: 10.1074/jbc.M112.404871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Bai J-X, Yan B, Zhao Z-N, Xiao X, Qin W-W, Zhang R, Jia L-T, Meng Y-L, Jin B-Q, Fan D-M, Wang T, Yang A-G. Tamoxifen Represses miR-200 MicroRNAs and Promotes Epithelial-to-Mesenchymal Transition by Up-Regulating c-Myc in Endometrial Carcinoma Cell Lines. Endocrinology. 2013;154:635–645. doi: 10.1210/en.2012-1607. [DOI] [PubMed] [Google Scholar]
  • 309.Wee EJH, Peters K, Nair SS, Hulf T, Stein S, Wagner S, Bailey P, Lee SY, Qu WJ, Brewster B, French JD, Dobrovic A, Francis GD, Clark SJ, Brown MA. Mapping the regulatory sequences controlling 93 breast cancer-associated miRNA genes leads to the identification of two functional promoters of the Hsa-mir-200b cluster, methylation of which is associated with metastasis or hormone receptor status in advanced breast cancer. Oncogene. 2012;31:4182–4195. doi: 10.1038/onc.2011.584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Guttilla IK, Adams BD, White BA. ERα, microRNAs, and the epithelial–mesenchymal transition in breast cancer. Trends in Endocrinology & Metabolism. 2012;23:73–82. doi: 10.1016/j.tem.2011.12.001. [DOI] [PubMed] [Google Scholar]
  • 311.Bergamaschi A, Katzenellenbogen BS. Tamoxifen downregulation of miR-451 increases 14-3-3[zeta] and promotes breast cancer cell survival and endocrine resistance. Oncogene. 2012;31:39–47. doi: 10.1038/onc.2011.223. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES