Knockdown of HMGA1 inhibits human breast cancer cell growth and metastasis in immunodeficient mice

https://doi.org/10.1016/j.bbrc.2013.03.064Get rights and content

Highlights

  • Overexpression of HMGA1 has been previously implicated in breast carcinogenesis.

  • An RNAi-based HMGA1-silencing construct with co-expression of EmGFP was generated.

  • HMGA1-silenced MDA-MB-231 human breast cancer cells were generated.

  • Knockdown of HMGA1 impairs anchorage-independent growth and tumorsphere formation.

  • Knockdown of HMGA1 impairs xenograft growth and metastasis in immunodeficient mice.

Abstract

The high mobility group A1 gene (HMGA1) has been previously implicated in breast carcinogenesis, and is considered an attractive target for therapeutic intervention because its expression is virtually absent in normal adult tissue. Other studies have shown that knockdown of HMGA1 reduces the tumorigenic potential of breast cancer cells in vitro. Therefore, we sought to determine if silencing HMGA1 can affect breast cancer development and metastatic progression in vivo. We silenced HMGA1 expression in the human breast cancer cell line MDA-MB-231 using an RNA interference vector, and observed a significant reduction in anchorage-independent growth and tumorsphere formation, which respectively indicate loss of tumorigenesis and self-renewal ability. Moreover, silencing HMGA1 significantly impaired xenograft growth in immunodeficient mice, and while control cells metastasized extensively to the lungs and lymph nodes, HMGA1-silenced cells generated only a few small metastases. Thus, our results show that interfering with HMGA1 expression reduces the tumorigenic and metastatic potential of breast cancer cells in vivo, and lend further support to investigations into targeting HMGA1 as a potential treatment for breast cancer.

Introduction

Breast cancer is the most frequent type of cancer and a leading cause of cancer death among women worldwide [1], [2], [3]. The proteins of the high mobility group A (HMGA) family have been previously implicated in breast carcinogenesis [4]. These are non-histone, DNA-binding proteins often referred to as architectural transcription factors. They contain basic A-T hook domains that mediate binding to the minor groove of AT-rich regions of chromosomal DNA. Upon binding to DNA, HMGA proteins regulate gene expression by organizing the transcriptional complex through protein–protein and protein–DNA interactions (reviewed in [5], [6], [7]). The HMGA family includes the products of the HMGA1 and HMGA2 genes. Of these, HMGA1 can generate three different protein isoforms through alternative splicing (HMGA1a, b and c). HMGA1a and HMGA1b are the most abundant isoforms, and differ by only 11 amino acids that are present in HMGA1a but not in HMGA1b [4]. HMGA1 is expressed almost exclusively during embryonic development [8], but has been found to be abnormally expressed in several types of cancer, including leukemia [9], pancreatic [10], [11], thyroid [12], colon [13], breast [14], [15], [16], lung [17], ovarian [18], endometrial [19], prostate [20], and head and neck cancer [21]. Several studies have also shown that overexpression of HMGA1 induces transformation both in vitro and in animal models (reviewed in [6], [7]).

The causal role of HMGA1 in breast cancer development and metastasis is supported by studies in cell lines [14], [22], [23] as well as by the analysis of clinical specimens [15], [16]. For example, elevated HMGA1 protein expression has been reported in breast carcinomas and hyperplastic lesions with cellular atypia, in contrast with normal breast tissue where HMGA1 was not detected [15], [16]. Similarly, HMGA1 overexpression has been observed in human breast cancer cell lines, with the highest levels in known metastatic lines, such as Hs578T and MDA-MB-231 [14], [22], [23]. Moreover, exogenous overexpression of HMGA1a was shown to induce transformation of the human non-tumorigenic mammary myoepithelial cell line Hs578Bst in vitro [14] and to increase the metastatic ability of MCF7 breast cancer cells in vivo [22]. Conversely, decreasing HMGA1 expression in Hs578T breast cancer cells was shown to cause a reduction in anchorage-independent growth, which is a typical feature of cancer cells [14].

HMGA1 is considered an attractive target for therapeutic intervention because its expression is virtually absent in normal adult tissue and knockdown of HMGA1 has been shown to interfere with the tumorigenic growth of multiple cancer cell lines [6], [7]. We therefore sought to determine if silencing HMGA1 can affect breast cancer development and metastatic progression in vivo using a human xenograft mouse model.

Section snippets

Cell lines, transfections and proliferation assays

The MDA-MB-231 breast cancer cell line was obtained from American Type Culture Collection, and cultured in DMEM (Cellgro 10-013), supplemented with 10% FBS and 5 μg/ml gentamicin. Cells were propagated for two weeks, aliquoted in media supplemented with 5% DMSO and stored in liquid nitrogen. Each aliquot was used for less than six months. MDA-MB-231 cells were transfected using Lipofectamine 2000 (Invitrogen). Stable transfectants were selected by adding 50 μg/ml of blasticidin to the media, and

Results and discussion

We generated an HMGA1-silencing construct that uses microRNA (miRNA) for RNA interference (RNAi), and co-expresses the fluorescent molecule EmGFP (co-cistronic) for easy detection of the transfected cells (pHMGA1-394-EmGFP-miR). We selected the breast cancer cell line MDA-MB-231 because it was previously shown to overexpress HMGA1 [14], and it is tumorigenic in immunodeficient mice. Stable, polyclonal MDA-MB-231 transfectants were generated, and sorted by flow cytometry. We isolated three

Acknowledgments

This study was supported by the Flight Attendant Medical Research Institute (062544_YCSA), and The National Institutes of Health (P30 CA006973). The funding sources had no role in the collection, analysis and interpretation of data, writing of the report, or in the decision to submit the article for publication.

References (28)

  • R. Reeves et al.

    HMGI/Y proteins: flexible regulators of transcription and chromatin structure

    Biochim. Biophys. Acta

    (2001)
  • M. Fedele et al.

    HMGA and cancer

    Biochim. Biophys. Acta

    (2010)
  • A.C. Hristov et al.

    HMGA1 correlates with advanced tumor grade and decreased survival in pancreatic ductal adenocarcinoma

    Mod. Pathol.

    (2010)
  • J. Ferlay, H.R. Shin, F. Bray, D. Forman, C. Mathers, D.M. Parkin, GLOBOCAN 2008 v1.2, Cancer Incidence and Mortality...
  • American Cancer Society, Cancer Facts & Figures 2013, American Cancer Society, Atlanta,...
  • J. Ferlay et al.

    Cancer incidence and mortality patterns in europe: estimates for 40 countries in 2012

    Eur. J. Cancer

    (2013)
  • S. Peluso et al.

    High-mobility group A (HMGA) proteins and breast cancer

    Breast Care (Basel)

    (2010)
  • L.M. Resar

    The high mobility group A1 gene: transforming inflammatory signals into cancer?

    Cancer Res.

    (2010)
  • A. Fusco et al.

    Roles of HMGA proteins in cancer

    Nat. Rev. Cancer

    (2007)
  • Y. Xu et al.

    The HMG-I oncogene causes highly penetrant, aggressive lymphoid malignancy in transgenic mice and is overexpressed in human leukemia

    Cancer Res.

    (2004)
  • N. Abe et al.

    Pancreatic duct cell carcinomas express high levels of high mobility group I(Y) proteins

    Cancer Res.

    (2000)
  • G. Chiappetta et al.

    Detection of high mobility group I HMGI(Y) protein in the diagnosis of thyroid tumors: HMGI(Y) expression represents a potential diagnostic indicator of carcinoma

    Cancer Res.

    (1998)
  • G. Chiappetta et al.

    High mobility group HMGI(Y) protein expression in human colorectal hyperplastic and neoplastic diseases

    Int. J. Cancer

    (2001)
  • C.E. Dolde et al.

    HMG-I/Y in human breast cancer cell lines

    Breast Cancer Res. Treat.

    (2002)
  • Cited by (30)

    • High Mobility Group A (HMGA) proteins: Molecular instigators of breast cancer onset and progression

      2018, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      In addition, loss of HMGA1 expression causes a strong reduction in mammosphere formation and dimension, decreases cell migration and invasion, impairs anchorage independent growth and the ability of cells to form metastasis in vivo. All these results have been confirmed in a series of back-to-back articles published in the same period [92,142]. Consistent with these results, several genes involved in stemness were found to be regulated upon HMGA1 silencing in MDA-MB-231 cells [73,92].

    • HMGA1 facilitates tumor progression through regulating Wnt/β-catenin pathway in endometrial cancer

      2016, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      HMGA1 was first discovered in highly proliferative and widely metastatic cervical cancer cells (HeLa) almost 30 years ago. Indeed, HMGA1 is over-expressed in diverse cancers, such as colon cancer, breast cancer, prostatic cancer, pancreatic cancer, and ovarian cancer [22,25–28]. In this study, an IHC analysis was conducted to show HMGA1 expression in EC samples and normal endometrial tissue samples.

    • MIR-625 suppresses cell proliferation and migration by targeting HMGA1 in breast cancer

      2016, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Overexpression of HMGA1 and its association with poor prognosis have been reported in bladder cancer [15], non-small cell lung cancer [16] and breast cancer [17]. The knockdown of HMGA1 in breast cancer led to a significant reduction in anchorage-independent growth and xenograft formation [18]. Mechanistically, HMGA1 was capable of modulating the activity of cyclin E2 to translocate YAP to nuclear, subsequently resulting in enhancement of cell motility and invasiveness [19].

    View all citing articles on Scopus
    View full text