Genome-wide study of ER-regulated lncRNAs shows AP000439.3 may function as a key regulator of cell cycle in breast cancer

  • Authors:
    • Yin Zhang
    • Dan-Lan Wang
    • Hai-Yan Yan
    • Jian-You Liao
    • Jie-Hua He
    • Kai-Shun Hu
    • Wei-Xi Deng
    • Yan-Jie Wang
    • Hong-Tao Xing
    • H. Phillip Koeffler
    • Dong Yin
  • View Affiliations

  • Published online on: September 20, 2017     https://doi.org/10.3892/or.2017.5975
  • Pages: 3227-3237
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Estrogen receptor (ER) plays important roles in cell growth, development and tumorigenesis. Although ER-regulated genes have been extensively investigated, little is known about roles of ER-regulated lncRNAs in breast cancer. Here, we conducted genome-wide study of ER-regulated lncRNAs by using RNA-seq, ChIP-seq and TCGA data. A total of identified 114 ER-regulated lncRNAs were identified, many of them were overexpressed in ER+ breast cancer and co-expressed with some key regulators. Silencing one of most prominent lncRNA, AP000439.3, resulted in inhibition of cell cycle progression and proliferation. Further study revealed AP000439.3 can regulate expression of CCND1 through enhancing estrogen receptor induction of CCND1. This finding revealed lncRNAs may serve as important effectors of ER in regulation of gene expression and cell phenotype in breast cancer.

Introduction

Estrogen plays critical roles in normal mammary gland development, physiological processes as well as tumorigenesis. Estrogen receptor (ER) is the major mediator of the effect of estrogen. After binding to estrogen, ER is activated and translocates into the cell nucleus (1). In the nucleus, ER can transcriptionally regulate target genes either by directly binding to estrogen response elements (EREs) or through protein-protein interactions with other transcriptional factors such as AP1, SP1 and NF-κB (2). Moreover, ~70% of breast cancers express ER. ER is essential for the initiation and development of breast cancers (1,3,4). The pro-oncogenic effect of ER is mediated primarily by transcriptionally regulating its target genes. Various genes that play crucial roles in cell proliferation and apoptosis such as c-Myc, CCND1 and BCL2 have been identified as ER-regulated genes. Hundreds of protein-coding genes have been identified as ER-regulated genes by using microarray and RNA-seq as well as ChIP-seq (59).

Long non-coding RNAs (lncRNAs) refer to non-coding RNAs consisting of >200 nucleotides. Recently, the important roles of lncRNAs in breast cancer cell proliferation, apoptosis, metastasis and endocrine resistance have been reported (1012). Because of their crucial roles in breast cancer, we explored ER-regulated lncRNAs. Although a few studies have been carried out to identify ER-regulated lncRNAs very recently (1315), their roles in regulation of gene expression and cell phenotype remains largely unknown. Here, we report a genome-wide study of ER-regulated lncRNAs by conducting an integrate analysis of ChIP-seq, strand-specific RNA-seq and TCGA clinical data. We observed that many of these ER-regulated lncRNAs were overexpressed in ER+ breast cancer and exhibited co-expression with several key regulator proteins. Moreover, we found one of most prominent lncRNAs, AP000439.3, can promote cell cycle progression through enhancing CCDN1 expression induced by estrogen. These findings reveal ER can regulate many lncRNAs that exhibit important functions in regulation of gene expression and cell phenotype in breast cancer.

Materials and methods

Datasets and computational analysis

The 17β-estradiol (E2) and vehicle treated MCF7 RNA-seq data (pair-end, strand-specific) was previously reported by Dago et al (16) and obtained from GEO dataset (GSE64590). For RNA-seq analysis, the sequenced reads were aligned to human reference genome (Hg38) and transcriptome (GENCODE.v23) using STAR (17) and then processed by RSEM (18). Genes with FDR<0.05 generated by both DESeq (19) and edgeR (20) were considered as differentially expressed genes.

ERα ChIP-seq of estrogen treated and untreated MCF7 were reported by Franco et al (21) and obtained from GEO dataset (GSE59530). Reads of ChIP-seq were aligned to human reference genome (Hg38) using BOWTIE (22). The estrogen treated peaks were generated by MACS (23) using untreated as control. The ER binding sites within ±100 kb region of transcriptional start sites (TSS) of protein-coding genes and lncRNAs (GENCODE.v23) were considered as potential ER regulatory sites.

The TCGA lncRNA data were downloaded from TANRIC database (24), mRNA and RPPAs data were downloaded from Broad Dashboard-Stddata (https://confluence.broadinstitute.org/display/GDAC/Dashboard-Stddata).

Cell culture and treatments

MCF7, ZR-75-1 and T47D cells were obtained from ATCC. MCF7 cells were cultured in Eagle's minimum essential medium supplemented with 10% FBS; ZR-75-1 and T47D were cultured in RPMI-1640 media supplemented with 10% FBS. Before estrogen treatments, the cells were grown for 72 h in phenol red-free MEM Eagle medium supplemented with 10% charcoal-dextran-treated FBS. The cells were then treated with ethanol (vehicle) or 10 nM E2 for 24 h. siRNAs were synthesized in GenePharma Co. (Shanghai, China). The sequences of siRNA are listed in Table II. siRNA was transfected at a final concentration of 50 nM using Lipofectamine 2000 reagent (Invitrogen).

Table II.

Primers and siRNAs used in this study.

Table II.

Primers and siRNAs used in this study.

qPCR primersForwardReverse
AP000439.3 CCCCAGGCTAGGAAGATGT GAGCCAAGAGGTCCTCACAG
RP11-321G12.1 GGTTTGGTTCCCAATTGTTG TTGGAAGACCCCATCTTCAC
RP11-150O12.3 ACCATTTCCAAACTGCCAAG GCTCCATGCACACTCAAGAA
LINC01016 TACAGCATGGTTCCCAAATG GGGCCATGGTCACTCATATT
SIAH2-AS1 CTCCTCAATCCCCACACAGT TGCAGACGTGTATTCGGGTA
RP11-95G17.2 TTGCTGTAGTGCGGCTTAAA TAACCCCTTGCAATCAGCTC

ChIP PCR primersForwardReverse

ER binding site 1 AGGGAGAGTTCCCAGGAGTC CAGCCCTGTCTGAGCAATTT
ER binding site 2 CTCCACCGAGCACTCCATAC TGCCTCTTGTTTCCCCTAAA

siRNAssenseantisene

AP000439.3 siRNA 1# GCUAGGAAGAUGUGCACCU AGGUGCACAUCUUCCUAGC
AP000439.3 siRNA 2# GCAAAGCUCACAGGAAAUA UAUUUCCUGUGAGCUUUGC
ERα siRNA CGAGUAUGAUCCUACCAGA UCUGGUAGGAUCAUACUCG
Real-time quantitative PCR and western blotting

Cells were grown and treated as described above and then RNA was collected using TRIzol reagent (Invitrogen), RNA was reverse-transcribed using PrimeScript RT reagent kit (Takara RR047A) and analyzed by LightCycler® 96 real-time PCR thermocycler (Roche), primers are listed in Table II. The lncRNA expression was normalized to β-actin transcript as an internal standard. The CCND1 and α-tubulin protein were detected by western blotting using cyclin D1 mouse mAb (BD Pharmigen #556470) and α-tubulin antibody (Santa Cruz, SC-5286).

Cell cycle analysis and colony formation assay

Seventy-two hours after siRNA transfection, cells were digested and washed with PBS and then fixed in 70% ethanol overnight at 4°C, then incubated with RNase and propidium iodide (PI) for 10 min and analyzed by flow cytometry.

For colony forming analysis, 1,000 cells were plated in 6-well plates and grown for 2 weeks, colonies were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet. The number of colonies were counted and analyzed by ImageJ software.

Chromatin immunoprecipitation (ChIP) assay

The ChIP assay were performed as previously described (25) with MCF7 cells using 3 µg normal IgG (CST, #2729) and ER (Santa Cruz sc-543) antibody. The ChIP PCR primers were listed in Table II.

Statistical analysis

Wilcoxon rank-sum test was used for comparing fold change between lncRNA and protein. Spearman's rank moment correlation coefficient was calculated for analysis of the co-expression of lncRNAs and RPPAs, Pearson's product moment correlation for lncRNA-mRNA co-expression. For qRT-PCR and colony formation assay, Student's t-test was used to test for statistical significance of the differences between the different group parameters. p-values of <0.05 was considered statistically significant.

Results

Global identification and characterization of ER-regulated lncRNAs

To identify ER-regulated lncRNAs in a genome-wide manner, we exploited published RNA-seq of E2- and vehicle-treated cells that were originally used for analyzing alternative mRNA splicing (16) (GSE64590). We chose this dataset because: i) this RNA-seq was strand-specific, which was superb for analyzing antisense transcripts that overlapped with host genes. ii) Three repetitions with high reproducibility occurred in this dataset. We conducted an analysis of differential expression (DE) transcripts using edgeR (20) and DEseq (19) processed by RSEM (18). We observed 2,869 DE transcripts, which included 1,784 upregulated and 1,085 downregulated transcripts. The dominant types of DE transcripts were protein-coding genes (92.5%); 147 lncRNAs were found to express differentially upon E2 treatment (Fig. 1A). Of these lncRNAs, 45.6% were long intergenic RNA (lincRNAs), 19.7% were antisense and 16.3% were processed transcripts (Fig. 1B). DE lncRNAs showed significantly lower expression than protein-coding genes (Fig. 1C), which is in agreement with previous reports (26). Despite low expression of DE lncRNAs, we found DE lncRNAs exhibited a higher fold-change than DE protein-coding genes in the presence of estrogen (Fig. 1D). LncRNAs tend to be more specifically expressed than protein (27,28); moreover, lncRNAs differ not just between tissues, but also between closely related cell types, which indicates lncRNAs are likely under stricter regulations (29). In agreement with this notion, our analysis revealed lncRNAs were under more rigorous regulation of activated ER. To investigate whether these DE transcripts were transcriptionally regulated by ER, we conducted an analysis of ER binding sites using published ChIP-seq data, and 114 of 147 (77.6%) DE lncRNAs had at least one ER binding site within their genomic domain (within ± 100 kb from TSS). Half of these lncRNAs were upregulated by E2 and these lncRNAs were referred to as putative ER-upregulated lncRNAs (Table I). To explore the potential roles of these lncRNAs, we conducted co-expression analysis between lncRNAs and reverse-phase protein array (RPPAs) proteomics data (30), which includes extensively validated antibodies to nearly 200 proteins and phosphoproteins of TCGA clinical samples. We found most of these lncRNAs co-expressed with some key regulators of the cell cycle (cyclin family) and apoptosis (Bcl-2 family) as well as key components of important signaling pathway such as IGF1R, MAPK (pT202/Y204) and JNK2. These data suggest that ER can transcriptionally regulate hundreds of lncRNAs that may participate in some important cellular processes.

Table I.

List of ER-upregluated lncRNAs.

Table I.

List of ER-upregluated lncRNAs.

Ensembl IDLncRNA nameFold change (log2)FDR
ENSG00000273565.1CTD-3075F15.15.130.000258667
ENSG00000249346.6LINC010164.301.89E-107
ENSG00000266036.1RP11-452I5.24.043.29E-06
ENSG00000254290.1RP11-150O12.32.746.46E-08
ENSG00000258354.1MIR3180–12.696.93E-06
ENSG00000272472.1RP11-95G17.22.515.95E-15
ENSG00000244265.1SIAH2-AS12.480.000112224
ENSG00000227036.6LINC005111.975.34E-30
ENSG00000259459.5RP11-321G12.11.860.000182177
ENSG00000229525.1AC053503.41.833.17E-06
ENSG00000233885.7YEATS2-AS11.482.03E-14
ENSG00000259080.1RP11-158I13.21.461.04E-05
ENSG00000261578.1RP11-21L23.21.391.73E-26
ENSG00000280186.1RP11-483I13.61.359.59E-53
ENSG00000204792.2LINC012911.321.38E-21
ENSG00000253125.1RP11-459E5.11.246.02E-05
ENSG00000260401.1RP11-800A3.41.232.02E-94
ENSG00000261051.1RP11-274H2.51.221.07E-16
ENSG00000266709.1RP11-214O1.21.191.26E-05
ENSG00000223749.7MIR503HG1.182.99E-42
ENSG00000280924.1LINC006281.162.97E-05
ENSG00000226471.6CTA-292E10.61.169.45E-19
ENSG00000277159.1RP11-88E10.41.122.90E-09
ENSG00000281207.1SLFNL1-AS11.036.93E-06
ENSG00000280073.1CTD-2525P14.50.951.47E-05
ENSG00000232956.8SNHG150.931.38E-37
ENSG00000230836.1LINC012930.845.77E-15
ENSG00000246174.7KCTD21-AS10.793.09E-08
ENSG00000236144.6TMEM147-AS10.428.75E-07
ENSG00000213062.4RP1-206D15.60.751.69E-05
ENSG00000270344.2RP11-734K2.40.739.31E-17
ENSG00000260257.2RP5-1085F17.30.720.000169607
ENSG00000261226.1RP11-830F9.70.637.69E-06
ENSG00000249859.7PVT10.625.88E-46
ENSG00000271020.1RP11-10C24.10.616.26E-05
ENSG00000247092.6SNHG100.572.45E-12
ENSG00000232445.1RP11-132A1.40.572.35E-06
ENSG00000271643.1RP11-10C24.30.569.12E-08
ENSG00000262202.4RP11-160E2.60.565.30E-09
ENSG00000259977.1AL121578.20.537.72E-06
ENSG00000255774.1AP000439.30.400.000181605
ENSG00000252690.3SCARNA150.391.54E-08
ENSG00000255717.6SNHG10.346.40E-61
ENSG00000236824.1BCYRN10.324.03E-08
ENSG00000236830.6CBR3-AS10.316.86E-12
ENSG00000226950.6DANCR0.307.63E-15
ENSG00000234912.9SNHG200.253.78E-05
ENSG00000227051.5C14orf1320.233.24E-16
ENSG00000235123.5DSCAM-AS10.222.09E-29
ENSG00000163597.14SNHG160.203.35E-46
ENSG00000259623.1RP11-156E6.10.191.91E-06
ENSG00000260032.1LINC006570.163.44E-37
ENSG00000224032.6EPB41L4A-AS10.165.66E-06
ENSG00000242125.3SNHG30.141.29E-05
ENSG00000244879.4GABPB1-AS10.111.09E-05
ENSG00000234741.7GAS50.066.73E-08
ENSG00000175061.17LRRC75A-AS10.053.01E-05
Detection of ER-regulated lncRNAs

To verify the DE lncRNAs analyzed by RNA-seq, we selected six lncRNAs for further analysis. We first performed qPCR experiments after E2 treatment. Consistent with RNA-seq results. In MCF7, the most significantly changed lncRNA is AP000439.3, which increased 6 times. The expressions of this lncRNA in T47D was too low to be effectively detected. We also observed significant increase of AP000439.3 after E2 treatment in ZR-75-1 cells (data not shown). The other lncRNAs also increased remarkably after E2 treatment of both in MCF7 and T47D (Fig. 2A and C). To confirm that these DE lncRNAs are regulated by ER, we silenced ERα in MCF7 and T47D by siRNA. In MCF7 cells, all of these lncRNAs dramatically decreased when silencing ERα, some of the lncRNAs such as RP11-150O12.3, AP000439.3 and RP11-95G17.2 decreased by >60% (Fig. 2B and D). Similar results were observed when ER was knocked down in T47D. Although LINC01016 is a significantly upregulated lncRNA in T47D, silencing ER did not result in its reduction. LINC01016 has been reported as an ER-target lncRNA in a previous study (13,14). Failure to observe a decrease of LINC01016 when silencing ERα may be due to its low expression level in T47D, hence hard to downregulate further, but easy to upregulate.

ER-regulated lncRNA AP000439.3 promotes cell cycle progression

To further characterize these ER-regulated lncRNAs, we analyzed the expression of lncRNAs in breast tissues using The Cancer Genome Atlas (TCGA) data (31). Most of these selected lncRNAs were dramatically overexpressed in ER+ breast cancer compared to ER breast cancer and normal breast tissues (p<0.001, Fig. 3).

To explore the roles of ER target lncRNAs in breast cancer, we selected one of the most significantly upregulated lncRNA (AP000439.3) for further analysis. Using ChIPBase, an integrated database for decoding transcriptional regulators of genes and lncRNAs (32,33), we found ERα is the top ranked regulator. To verify that AP000439.3 is regulated by ER, ChIP PCR assay was performed on potential ER binding sites identified by ChIP-seq data and ChIPBase. As shown in Fig. 4A, ER can directly bind upstream of AP000439.3.

Since ER can regulate dozens of genes that play crucial roles in breast cancer, we speculate these lncRNAs may also have an important impact on breast cancer cell function. AP000439.3 was silenced using two individual siRNAs by >50% (Fig. 4B). The most dominant role of ER in breast cancer is promotion of cell proliferation and cell cycle progression (1). Hence the cell cycle progression was detected using flow cytometry assay. Silencing AP000439.3 resulted in a dramatic inhibition of the G1-S transition in MCF7 cells (Fig. 4C) and in ZR-75-1 cells (Fig. 4D). Likewise, silencing AP000439.3 significantly suppressed clonogenic proliferation (Fig. 4E and F). Taken together, lncRNA AP000439.3 is stimulated by activated ER, it is overexpressed in ER+ breast cancer and promotes cell cycle progression and proliferation.

AP000439.3 facilitates estrogen induced CCDN1 expression

To understand how AP000439.3 regulates the cell cycle, we first investigated its location and gene structure. AP000439.3 is a long intergenic non-coding RNA (lincRNA) located at chromosome 11q13. AP000439.3 is 160 kb upstream of the CCDN1 transcriptional start site (TSS) (Fig. 5A), AP000439.3 and CCDN1 are transcribed divergently (head to head). CCND1 encodes the cyclin D1 protein that serves as a regulator of cyclin-dependent kinase as a crucial regulator of the cell cycle. Amplification of CCND1 has been reported in many kinds of tumors including breast cancer, its amplification is associated with a poor prognosis (34). Analysis of COSMIC CNV data (35) revealed that CCND1 amplification occurred in 68/919 (7.4%) breast cancer samples, 58 of 68 (85%) of these also have an amplification of AP000439.3 (Fig. 5B). Expression analysis of CCND1 and AP000439.3 showed a high correlation between them (R=0.61, p<10−15, Fig. 5C). CCND1 is overexpressed in ~50% breast cancer, while amplification of the CCND1 gene is present only in a minority of CCND1-overexpressed breast cancers (36), directly regulated by ER is another important reason for overexpression of CCND1 (37).

LncRNAs are likely to regulate the expression of an adjacent gene (3840). Hence we speculated that AP000439.3 may influence the expression of CCND1. We silenced AP000439.3 in MCF7 and then treated these cells with either E2 or vehicle. Both qPCR and western blotting showed that knockdown of AP000439.3 could impair CCND1 expression induced by E2 (Fig. 5D). Taken together, these data suggest ER-regulated lncRNA AP000439.3 can facilitate ER regulation of CCND1 and thus promote cell cycle progression (Fig. 6).

Discussion

In this study, we described a genome-wide identification and characterization of ER-regulated lncRNAs in breast cancer cells. We found many of these lncRNAs were overexpressed in ER+ breast cancer and co-expressed with some key regulators. Moreover, we found one of the most prominent lncRNA, AP000439.3, can promote cell cycle progression through enhancing CCDN1 expression induced by estrogen.

The mechanisms of how AP000439.3 facilitate CCND1 expression remains to be further investigated. Considering AP000439.3 can influence transcription of CCND1, it may involve changes in chromatin organization. This mechanism have been reported by lncRNA CCAT1-L regulation on its adjacent gene C-MYC in colorectal cancer (41).

The roles of ER is mediated primarily by its downstream effectors, the ER-regulated protein-coding genes and their roles have been well studied. However, some important issues such as the mechanisms by which ER-mediated drug resistance remains largely unknown (42). Study of ER-regulated lncRNAs may provide a unique prospective to answer these questions.

Our findings may provide another layer of estrogenic control of gene expression: ER can promote expression of lncRNAs that are adjacent to protein-coding genes; these lncRNAs can serve as positive regulators that further facilitate transcriptional regulation by ER. It remains to be further investigated how many lncRNAs function in this way and whether these lncRNAs specifically regulate their neighboring adjacent genes or have a more extensive impact on ER regulation. Since these ER-regulated lncRNAs showed highly cell-type specificity, some oncogenic lncRNAs can be potential biomarkers and therapeutic targets.

Acknowledgements

We would like to thank Ms. Qing Wei and Ms. Fang Su for flow cytometry analysis. We thank The Cancer Genome Atlas (TCGA) project and their contributors for providing this valuable public data set. This study was supported by grants from the Natural Science Foundation of China (81572484, 81420108026 and 81621004 to D.Y.); Guangdong Science and Technology Department (2014A050503026 to D.Y., 2015B050501004 and S2012030006287); Guangzhou Bureau of Science and Information Technology (201704030036 to D.Y. This study was also supported by the RNA Biology Center at the Cancer Science Institute of Singapore, NUS, as part of funding under the Singapore Ministry of Education's Tier 3 grants, grant no. MOE2014-T3-1-006.

Glossary

Abbreviations

Abbreviations:

ER

estrogen receptor

lncRNA

long non-coding RNA

EREs

estrogen response elements

lincRNA

long intergenic non-coding RNA

DE

differential expression

qPCR

quantitative polymerase chain reaction

TCGA

The Cancer Genome Atlas

RPM

reads per million

FPKM

reads per kilobase of exon model per million mapped reads

ChIP

chromatin immunoprecipitation

References

1 

Liang J and Shang Y: Estrogen and cancer. Annu Rev Physiol. 75:225–240. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Marino M, Galluzzo P and Ascenzi P: Estrogen signaling multiple pathways to impact gene transcription. Curr Genomics. 7:497–508. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Yager JD and Davidson NE: Estrogen carcinogenesis in breast cancer. N Engl J Med. 354:270–282. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Shao W and Brown M: Advances in estrogen receptor biology: Prospects for improvements in targeted breast cancer therapy. Breast Cancer Res. 6:39–52. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Frasor J, Danes JM, Komm B, Chang KC, Lyttle CR and Katzenellenbogen BS: Profiling of estrogen up- and down-regulated gene expression in human breast cancer cells: Insights into gene networks and pathways underlying estrogenic control of proliferation and cell phenotype. Endocrinology. 144:4562–4574. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Coser KR, Chesnes J, Hur J, Ray S, Isselbacher KJ and Shioda T: Global analysis of ligand sensitivity of estrogen inducible and suppressible genes in MCF7/BUS breast cancer cells by DNA microarray. Proc Natl Acad Sci USA. 100:pp. 13994–13999. 2003; View Article : Google Scholar : PubMed/NCBI

7 

Inoue A, Yoshida N, Omoto Y, Oguchi S, Yamori T, Kiyama R and Hayashi S: Development of cDNA microarray for expression profiling of estrogen-responsive genes. J Mol Endocrinol. 29:175–192. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Soulez M and Parker MG: Identification of novel oestrogen receptor target genes in human ZR75-1 breast cancer cells by expression profiling. J Mol Endocrinol. 27:259–274. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Lin CY, Ström A, Vega VB, Kong SL, Yeo AL, Thomsen JS, Chan WC, Doray B, Bangarusamy DK, Ramasamy A, et al: Discovery of estrogen receptor alpha target genes and response elements in breast tumor cells. Genome Biol. 5:R662004. View Article : Google Scholar : PubMed/NCBI

10 

Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, Ronning P, Reuland B, Guenther K, Beadnell TC, et al: PVT1 dependence in cancer with MYC copy-number increase. Nature. 512:82–86. 2014.PubMed/NCBI

11 

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

12 

Xue X, Yang YA, Zhang A, Fong KW, Kim J, Song B, Li S, Zhao JC and Yu J: LncRNA HOTAIR enhances ER signaling and confers tamoxifen resistance in breast cancer. Oncogene. 35:2746–2755. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Miano V, Ferrero G, Reineri S, Caizzi L, Annaratone L, Ricci L, Cutrupi S, Castellano I, Cordero F and De Bortoli M: Luminal long non-coding RNAs regulated by estrogen receptor alpha in a ligand-independent manner show functional roles in breast cancer. Oncotarget. 7:3201–3216. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Jonsson P, Coarfa C, Mesmar F, Raz T, Rajapakshe K, Thompson JF, Gunaratne PH and Williams C: Single-molecule sequencing reveals estrogen-regulated clinically relevant lncRNAs in breast cancer. Mol Endocrinol. 29:1634–1645. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Qiu JJ, Ye LC, Ding JX, Feng WW, Jin HY, Zhang Y, Li Q and Hua KQ: Expression and clinical significance of estrogen-regulated long non-coding RNAs in estrogen receptor α-positive ovarian cancer progression. Oncol Rep. 31:1613–1622. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Dago DN, Scafoglio C, Rinaldi A, Memoli D, Giurato G, Nassa G, Ravo M, Rizzo F, Tarallo R and Weisz A: Estrogen receptor beta impacts hormone-induced alternative mRNA splicing in breast cancer cells. BMC Genomics. 16:3672015. View Article : Google Scholar : PubMed/NCBI

17 

Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M and Gingeras TR: STAR: Ultrafast universal RNA-seq aligner. Bioinformatics. 29:15–21. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Li B and Dewey CN: RSEM: Accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 12:3232011. View Article : Google Scholar : PubMed/NCBI

19 

Anders S and Huber W: Differential expression analysis for sequence count data. Genome Biol. 11:R1062010. View Article : Google Scholar : PubMed/NCBI

20 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Franco HL, Nagari A and Kraus WL: TNFα signaling exposes latent estrogen receptor binding sites to alter the breast cancer cell transcriptome. Mol Cell. 58:21–34. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Langmead B, Trapnell C, Pop M and Salzberg SL: Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10:R252009. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W, et al: Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9:R1372008. View Article : Google Scholar : PubMed/NCBI

24 

Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, Weinstein JN and Liang H: TANRIC: An interactive open platform to explore the function of lncRNAs in cancer. Cancer Res. 75:3728–3737. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Dong S, Han J, Chen H, Liu T, Huen MS, Yang Y, Guo C and Huang J: The human SRCAP chromatin remodeling complex promotes DNA-end resection. Curr Biol. 24:2097–2110. 2014. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Ulitsky I and Bartel DP: lincRNAs: Genomics, evolution, and mechanisms. Cell. 154:26–46. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A and Rinn JL: Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25:1915–1927. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Amin V, Harris RA, Onuchic V, Jackson AR, Charnecki T, Paithankar S, Subramanian S Lakshmi, Riehle K, Coarfa C and Milosavljevic A: Epigenomic footprints across 111 reference epigenomes reveal tissue-specific epigenetic regulation of lincRNAs. Nat Commun. 6:63702015. View Article : Google Scholar : PubMed/NCBI

30 

Li J, Lu Y, Akbani R, Ju Z, Roebuck PL, Liu W, Yang JY, Broom BM, Verhaak RG, Kane DW, et al: TCPA: A resource for cancer functional proteomics data. Nat Methods. 10:1046–1047. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C and Stuart JM: Cancer Genome Atlas Research Network: The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet. 45:1113–1120. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Yang JH, Li JH, Jiang S, Zhou H and Qu LH: ChIPBase: A database for decoding the transcriptional regulation of long non-coding RNA and microRNA genes from ChIP-Seq data. Nucleic Acids Res. 41D:D177–D187. 2013. View Article : Google Scholar

33 

Zhou KR, Liu S, Sun WJ, Zheng LL, Zhou H, Yang JH and Qu LH: ChIPBase v2.0: Decoding transcriptional regulatory networks of non-coding RNAs and protein-coding genes from ChIP-seq data. Nucleic Acids Res. 45D:D43–D50. 2017. View Article : Google Scholar

34 

Musgrove EA, Caldon CE, Barraclough J, Stone A and Sutherland RL: Cyclin D as a therapeutic target in cancer. Nat Rev Cancer. 11:558–572. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, Jia M, Shepherd R, Leung K, Menzies A, et al: COSMIC: Mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39(Database): D945–D950. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Arnold A and Papanikolaou A: Cyclin D1 in breast cancer pathogenesis. J Clin Oncol. 23:4215–4224. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Eeckhoute J, Carroll JS, Geistlinger TR, Torres-Arzayus MI and Brown M: A cell-type-specific transcriptional network required for estrogen regulation of cyclin D1 and cell cycle progression in breast cancer. Genes Dev. 20:2513–2526. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Berghoff EG, Clark MF, Chen S, Cajigas I, Leib DE and Kohtz JD: Evf2 (Dlx6as) lncRNA regulates ultraconserved enhancer methylation and the differential transcriptional control of adjacent genes. Development. 140:4407–4416. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Spurlock CF III, Tossberg JT, Guo Y, Collier SP, Crooke PS III and Aune TM: Expression and functions of long noncoding RNAs during human T helper cell differentiation. Nat Commun. 6:69322015. View Article : Google Scholar : PubMed/NCBI

40 

Luo S, Lu JY, Liu L, Yin Y, Chen C, Han X, Wu B, Xu R, Liu W, Yan P, et al: Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell. 18:637–652. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Xiang JF, Yin QF, Chen T, Zhang Y, Zhang XO, Wu Z, Zhang S, Wang HB, Ge J, Lu X, et al: Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 24:513–531. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Xu CY, Jiang ZN, Zhou Y, Li JJ and Huang LM: Estrogen receptor α roles in breast cancer chemoresistance. Asian Pac J Cancer Prev. 14:4049–4052. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 38 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Wang D, Yan H, Liao J, He J, Hu K, Deng W, Wang Y, Xing H, Koeffler HP, Koeffler HP, et al: Genome-wide study of ER-regulated lncRNAs shows AP000439.3 may function as a key regulator of cell cycle in breast cancer. Oncol Rep 38: 3227-3237, 2017
APA
Zhang, Y., Wang, D., Yan, H., Liao, J., He, J., Hu, K. ... Yin, D. (2017). Genome-wide study of ER-regulated lncRNAs shows AP000439.3 may function as a key regulator of cell cycle in breast cancer. Oncology Reports, 38, 3227-3237. https://doi.org/10.3892/or.2017.5975
MLA
Zhang, Y., Wang, D., Yan, H., Liao, J., He, J., Hu, K., Deng, W., Wang, Y., Xing, H., Koeffler, H. P., Yin, D."Genome-wide study of ER-regulated lncRNAs shows AP000439.3 may function as a key regulator of cell cycle in breast cancer". Oncology Reports 38.5 (2017): 3227-3237.
Chicago
Zhang, Y., Wang, D., Yan, H., Liao, J., He, J., Hu, K., Deng, W., Wang, Y., Xing, H., Koeffler, H. P., Yin, D."Genome-wide study of ER-regulated lncRNAs shows AP000439.3 may function as a key regulator of cell cycle in breast cancer". Oncology Reports 38, no. 5 (2017): 3227-3237. https://doi.org/10.3892/or.2017.5975