Nuclear accumulation of MKL1 in luminal breast cancer cells impairs genomic activity of ERα and is associated with endocrine resistance

https://doi.org/10.1016/j.bbagrm.2020.194507Get rights and content

Highlights

  • MKL1 is a master regulator of actin dynamic and cellular motile functions.

  • Nuclear translocation of MKL1 is associated with endocrine resistance.

  • Nuclear translocation of MKL1 induces a mixed luminal/basal phenotype.

  • Nuclear translocation of MKL1 suppresses estrogen-mediated control of gene expression.

  • Nuclear translocation of MKL1 induces a profound reprogramming in ERα cistrome associated with a massive loss of ERα binding sites (ERBSs).

Abstract

Estrogen receptor (ERα) is central in driving the development of hormone-dependent breast cancers. A major challenge in treating these cancers is to understand and overcome endocrine resistance. The Megakaryoblastic Leukemia 1 (MKL1, MRTFA) protein is a master regulator of actin dynamic and cellular motile functions, whose nuclear translocation favors epithelial-mesenchymal transition. We previously demonstrated that nuclear accumulation of MKL1 in estrogen-responsive breast cancer cell lines promotes hormonal escape. In the present study, we confirm through tissue microarray analysis that nuclear immunostaining of MKL1 is associated with endocrine resistance in a cohort of breast cancers and we decipher the underlining mechanisms using cell line models. We show through gene expression microarray analysis that the nuclear accumulation of MKL1 induces dedifferentiation leading to a mixed luminal/basal phenotype and suppresses estrogen-mediated control of gene expression. Chromatin immunoprecipitation of DNA coupled to high-throughput sequencing (ChIP-Seq) shows a profound reprogramming in ERα cistrome associated with a massive loss of ERα binding sites (ERBSs) generally associated with lower ERα-binding levels. Novel ERBSs appear to be associated with EGF and RAS signaling pathways. Collectively, these results highlight a major role of MKL1 in the loss of ERα transcriptional activity observed in certain cases of endocrine resistances, thereby contributing to breast tumor cells malignancy.

Introduction

Breast cancers exhibit strong heterogeneity from genetic and phenotypic features to clinical behavior [1]. Based on global gene expression profiles, at least four major molecular subtypes have been identified, including luminal A, luminal B, HER2-enriched and basal-like tumor [2,3]. Estrogen receptor-alpha (ERα) expression defines the luminal subtypes. More than two-thirds of breast cancers overexpress ERα allowing most of them to depend on estrogen to proliferate [4,5]. This specificity makes ERα an ideal target for endocrine therapies that use selective estrogen receptor modulators (SERM) such as tamoxifen and/or aromatase inhibitors to block estrogen-dependent cell proliferation [6,7]. Unfortunately, a significant number of ERα positive breast tumors fail to initially respond to endocrine therapy or stop benefiting from such treatments and acquire resistance [8,9]. Endocrine resistance often relies on changes in the functional properties of ERα, whose activity may shift from ligand-dependent to ligand-independent activity [9]. Actually, luminal A breast cancers generally express higher level of ERα, exhibit better response to endocrine therapy and have better prognosis than luminal B subtype [10,11]. These last years, estrogen signaling in luminal breast cancer cells was deeply explored through gene expression and genome-wide chromatin binding profiling using both cell lines and human tumors [[12], [13], [14]]. Data highlighted that ERα binds to several thousand of sites across the genome and that most of these sites are located far away from the promoter regions. The genomic regions bound by ERα are enriched with cis-regulatory elements bound by pioneer factors, notably FOXA1 and GATA3 whose expression is correlated with luminal subtype [15]. ERα binding to chromatin still occurs in breast cancer cell lines and tumors that are resistant to antiestrogen therapies [14]. However, these cells do present a modified cistrome of ERα, having lost a number of ERα binding sites (ERBSs) but also harboring specific ones. Interestingly, these novel ERBSs found in antiestrogen-resistant cells, are still bound by FOXA1 but are depleted in GATA3 motifs. In addition, the ERα cistrome in drug resistant cell lines and tumors is characterized by an increased average ERα binding signal intensity likely resulting from a constitutively active estrogen receptor. The transcriptomic signature of these cells remains associated with ERα-positive luminal subtype [14]. More aggressive in nature, with higher proliferation and metastasis potential than luminal subtypes, basal-like tumors often have a triple ERα/progesterone receptor (PR)/HER2 negative phenotype [2,16]. Consequently, these tumors are not amenable to conventional targeted therapies. Recent studies suggest that basal-like breast cancers originate from luminal cells rather than a mammary stem cell (MaSC) [1,17,18]. Up to 30% of initially ERα-positive tumors that have developed resistance to antiestrogen therapies lose part of ERα expression [19]. Therefore, endocrine resistance may also rely on a dedifferentiation process leading luminal cancers to switch to an ERα-negative phenotype.

MKL1, also called MRTFA, MAL or BSAC, is a member of the myocardin-related transcription factor (MRTF) family, whose members are coactivators of the serum response factor (SRF) [20,21]. Its main role is to sense the degree of actin polymerization controlled by Rho GTPases and to subsequently integrate this information at the gene expression level through a nuclear translocation [20]. As a master regulator of cellular motile and contractile functions, MKL1 exerts important roles in vascular smooth muscle cell and cardiac myocyte differentiation, and neuronal migration [21]. In mammary gland, MKL1 is essential for the basal/myoepithelial cell differentiation and function [22,23]. During tumorigenesis, MKL1 is required for tumor cell invasion and metastasis mediating the adaptive changes in cell shape, adhesion, and migration linked to the actin cytoskeleton [24]. Recent genome-wide association studies have identified MKL1 locus as a susceptible risk factor for breast cancer and especially triple-negative breast cancer (TNBC) [[25], [26], [27]]. We previously showed that the activation of MKL1 in estrogen sensitive breast cancer cell lines leads to hormonal resistance and reduced expression of ERα, PR and HER2, recalling the triple negative phenotype [28]. In the present study, we show that nuclear accumulation of MKL1 is associated with endocrine resistance in a cohort of breast cancers. Using genome wide analysis of gene expression and ERα chromatin binding, we demonstrate that, upon the expression of a constitutively active form of MKL1, initially ERα-positive breast cancer cells initiate a dedifferentiation process associated with a profound reprogramming in ERα cistrome leading to hormonal escape.

Section snippets

Cell culture and transfection

Stably transfected MCF7 T-Rex sub-clones (T-Rex system, Invitrogen), MCF7-control (with empty pcDNA4/TO expression vector) and MCF7-MKL1ΔN200 (with MKL1ΔN200 pcDNA4/TO expression vector), were previously described [28,29]. MCF7 T-Rex sub-clones as well as MCF10A, T47D, ZR-75-1, MCF7 AKT+ [express myr-akt1 (activated) plasmid (Upstate cell signaling solutions)], MDA-MB-468, MDA-MB-231 and SUM159PT cell lines were routinely maintained in DMEM (Invitrogen) supplemented with 10% fetal calf serum

Nuclear accumulation of MKL1 favors invasiveness and impairs transactivation efficiency of ERα

We previously showed that breast cancer cell lines exhibit different MKL1 activity [28]. As shown in Fig. 1A and Fig. S1A, MKL1 is almost cytoplasmic and inactive in luminal ERα-positive breast cancer cell lines such as MCF7, T47D or ZR-75-1, which exhibit a well-differentiated epithelial phenotype. In the non-tumorigenic epithelial cell line MCF10A, the vast majority of cells exhibit a cytoplasmic localization of MKL1. Only cells on islet periphery present a nuclear translocation of MKL1.

Discussion

Thirty to 50% of recurrent tumors that arise from ERα-positive primary breast cancer patients fail to respond to endocrine therapy [8,9]. While the majority of these breast tumors retain ERα expression, high reduction or loss of ERα expression represents a non-negligible step in the progression from endocrine sensitive to resistance [19]. Notably, luminal B breast cancers, which generally express lower level of ERα, exhibit worse response to endocrine therapy. Understanding the underlying

Author contributions

G.F. conceived the project. C.J., T.F.-C., F.P., E.J., P.G. and G.F. conducted the experiments. T.F.-C., D.H., S.A. and R.M. analyzed the microarray and ChIP-Seq data. C.J., M.M.C., and S.J. performed tissue microarrays. M.M. and D.M. contributed to the data analysis. T.F.-C., C.J., R.M. and G.F. wrote the manuscript with input from all authors.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

We thank the biosit Health genomics and H2P2-Histo pathology High precision core facilities of biogenouest and the IGBMC Micrroarray and Sequencing platform at Illkrich. This work was supported by the University of Rennes 1, Inserm, CNRS, and the Ligue Contre le Cancer.

References (59)

  • A. Skibinski et al.

    The origin of breast tumor heterogeneity

    Oncogene

    (2015)
  • C.M. Perou et al.

    Molecular portraits of human breast tumours

    Nature

    (2000)
  • T. Sørlie et al.

    Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • K. Dahlman-Wright et al.

    International Union of Pharmacology. LXIV. Estrogen receptors

    Pharmacol. Rev.

    (2006)
  • C.K. Osborne

    Steroid hormone receptors in breast cancer management

    Breast Cancer Res. Treat.

    (1998)
  • D.L. Wickerham et al.

    The use of tamoxifen and raloxifene for the prevention of breast cancer

    Recent Results Cancer Res. Fortschritte Krebsforsch. Progres Dans Rech. Sur Cancer.

    (2009)
  • V.C. Jordan et al.

    Selective estrogen-receptor modulators and antihormonal resistance in breast cancer

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (2007)
  • E.A. Musgrove et al.

    Biological determinants of endocrine resistance in breast cancer

    Nat. Rev. Cancer

    (2009)
  • V.N.R. Gajulapalli et al.

    Oestrogen receptor negativity in breast cancer: a cause or consequence?

    Biosci. Rep.

    (2016)
  • J.S. Carroll et al.

    Genome-wide analysis of estrogen receptor binding sites

    Nat. Genet.

    (2006)
  • W.-J. Welboren et al.

    ChIP-Seq of ERalpha and RNA polymerase II defines genes differentially responding to ligands

    EMBO J.

    (2009)
  • C.S. Ross-Innes et al.

    Differential oestrogen receptor binding is associated with clinical outcome in breast cancer

    Nature

    (2012)
  • K.M. Jozwik et al.

    Pioneer factors in hormone-dependent cancers

    Nat. Rev. Cancer

    (2012)
  • E. Lim et al.

    Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers

    Nat. Med.

    (2009)
  • T. Kuukasjärvi et al.

    Loss of estrogen receptor in recurrent breast cancer is associated with poor response to endocrine therapy

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (1996)
  • G.C.T. Pipes et al.

    The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis

    Genes Dev.

    (2006)
  • S. Li et al.

    Requirement of a myocardin-related transcription factor for development of mammary myoepithelial cells

    Mol. Cell. Biol.

    (2006)
  • Y. Sun et al.

    Acute myeloid leukemia-associated Mkl1 (Mrtf-a) is a key regulator of mammary gland function

    Mol. Cell. Biol.

    (2006)
  • S. Medjkane et al.

    Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis

    Nat. Cell Biol.

    (2009)
  • Cited by (8)

    View all citing articles on Scopus
    1

    Contributed equally to the work.

    View full text