Abstract

Abstract BACKGROUND: A GWAS for the MA.27 aromatase inhibitors (AIs) adjuvant trial (4,406 controls and 252 cases) identified variant (V) SNPs in a long noncoding (lnc) RNA, MIR2052HG, that were associated with longer breast cancer free interval (HR= 0.37, P= 2.15E-07). V SNPs (MAF= 0.32 to 0.42) were associated with lower MIR2052HG and ERα expression in the presence of AIs. MIR2052HG maintained ERα both by promoting AKT/FOXO3-mediated ESR1 transcription and by limiting ubiquitin-mediated ERα degradation. (Cancer Res 76:7012-23, 2016). Our goal was to further elucidate MIR2052HG's mechanism of action. METHODS: RNA-Binding Protein Immunoprecipitation (RBPI) assays were performed to demonstrate that the transcription factor, early growth response protein 1 (EGR1), worked together with MIR2052HG to regulate lemur tyrosine kinase-3 (LMTK3) transcription in MCF7/AC1 and CAMA-1 cells. The location of EGR1 on the LMTK3 gene locus was mapped using chromatin immunoprecipitation (ChIP) assays. The co-localization of MIR2052HG RNA and the LMTK3 gene locus was determined using RNA-DNA dual fluorescent in situ hybridization (FISH). SNP effects were evaluated using a panel of human lymphoblastoid cell lines. RESULTS: TCGA analysis revealed LMTK3 and MIR2052HG expression were highly correlated in ERα-positive breast cancer patients. We found that the MIR2052HG transcript was located in the LMTK3 gene locus by RNA-DNA FISH. Among all of the 12 potential LMTK3 transcription factors identified in the Encode database that were examined by RBPI, only EGR1 showed an interaction with MIR2052HG. CHIP assays confirmed EGR1 binding to the two putative EGR1 binding sites in LMTK3 gene.Depletion of MIR2052HG reduced the binding of EGR1 to the LMTK3 promoter and decreased LMTK3 expression, suggesting that it might function as a scaffold. Mechanistically, decreased LMTK3 levels further increased protein kinase C (PKC) activity and downstream AKT activity, leading to reduced ESR1 mRNA levels via increased pFOXO3. At the protein level, in MIR2052HG depleted cells, increased PKC activity increased the phosphorylation of MEK, ERK, and RSK1, leading to increased ERα phosphorylation at Ser167 and increased ERα degradation. Conversely, overexpression of LMTK3 in MIR2052HG depleted cells reversed these phenotypes. MIR2052HG regulated LMTK3 and ERα expression in a SNP- dependent fashion: the MIR2052HG V SNP, relative to wild-type (WT) genotype, increased LMTK3/ERα expression in response to androstenedione due to increased binding between EGR1 and the LMTK3 promoter in LCLs. However, AI treatment reduced this binding in MIR2052HG variant cells but increased binding in WT cells, resulting in decreased LMTK3/ERα in V cells and increased expression in WT cells. CONCLUSIONS: Our findings support a model in which the protective MIR2052HG variant genotype regulates LMTK3 via MIR2052HG/EGR1, and LMTK3 regulates ERα stability via the PKC/MEK/ERK/RSK1 axis. This regulation may explain the effect of the MIR2052HG variant genotype on cell proliferation and response to AIs in MA.27. These findings provide new insight into the mechanism of action of MIR2052HG and suggest that LMTK3 may be a new therapeutic target in ERα-positive breast cancer patients treated with AIs. Citation Format: Cairns J, Ingle JN, Shepherd LE, Kubo M, Goetz MP, Weinshilboum RM, Kalari KR, Wang L. LncRNA MIR2052HG regulates ERα level and endocrine resistance through LMTK3 by recruiting early growth response protein 1 [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P5-07-01.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call