Elsevier

Biochimie

Volume 173, June 2020, Pages 114-122
Biochimie

Research paper
Effects of folic acid withdrawal on transcriptomic profiles in murine triple-negative breast cancer cell lines

https://doi.org/10.1016/j.biochi.2020.04.005Get rights and content

Highlights

  • Folic acid withdrawal alters gene expression in mouse mammary cancer cell lines.

  • Changes are most pronounced in non-metastatic mesenchymal cells.

  • Interferon signaling pathway genes are upregulated by folic acid withdrawal.

  • Reactivation of the type I interferon pathway may have clinical utility.

Abstract

We have previously shown that withdrawal of folic acid led to metabolic reprogramming and a less aggressive phenotype in a mouse cell model of triple-negative breast cancer (TNBC). Herein, we evaluate the effects of folic acid withdrawal on transcriptomic profiles in these cells. Murine cell lines were originally derived from a pool of spontaneous mammary tumors grown in MMTV-Wnt1 transgenic mice. Based on their differential molecular characteristics and metastatic potential, these cell lines were previously characterized as non-metastatic epithelial (E-Wnt), non-metastatic mesenchymal (M-Wnt) and metastatic mesenchymal (metM-Wntliver) cells. Using custom two-color 180K Agilent microarrays, we have determined gene expression profiles for three biological replicates of each subtype kept on standard medium (2.2 μM folic acid) or folic acid-free medium for 72 h. The analyses revealed that more genes were differentially expressed upon folic acid withdrawal in M-Wnt cells (1884 genes; Benjamini-Hochberg-adjusted P-value <0.05) compared to E-Wnt and metM-Wntliver cells (108 and 222 genes, respectively). Pathway analysis has identified that type I interferon signaling was strongly affected by folic acid withdrawal, with interferon-responsive genes consistently being upregulated upon folic acid withdrawal in M-Wnt cells. Of note, repressed interferon signaling has been established as one of the characteristics of aggressive human TNBC, and hence reactivation of this pathway may be a promising therapeutic approach. Overall, while our study indicates that the response to folic acid withdrawal varies by molecular subtype and cellular phenotype, it also underscores the necessity to further investigate one-carbon metabolism as a potential therapeutic means in the treatment of advanced TNBC.

Introduction

Cancer metastasis is the most common cause of cancer-related death [[1], [2], [3]]. Metastasis is a complex multi-step process, with cells undergoing metabolic reprograming to support their energetic, biosynthetic, and redox needs at various stages of metastasis [[4], [5], [6]]. During invasion, metabolic adaptation also enables cancer cells to survive in nutrient-limited environments [5,6]. Epithelial-to-mesenchymal transition (EMT), a process where epithelial cancer cells acquire mesenchymal features, plays an important role in cancer cell invasion and metastasis [6,7]. EMT is regulated by several transcription factors, which control, among others, numerous metabolic genes [5,6].

One of the key metabolic branches in the cell is the network of folate-dependent biochemical pathways. Folate (vitamin B9) functions as a coenzyme in numerous reactions of one-carbon transfer, which are involved in nucleotide biosynthesis, amino acid metabolism, redox homeostasis, and epigenetic regulation [[8], [9], [10]]. Since humans cannot synthesize folate, folate metabolism is regulated to a significant extent by dietary intake of the vitamin. Low folate intake or status has been associated with an increased risk of certain diseases, most notably neural tube defects [[10], [11], [12]], but also particular forms of cancer [13,14]. In 1996, the U.S. Food and Drug Administration (FDA) approved mandatory fortification of several types of grain foods in the US with folic acid, a synthetic form of the vitamin, to prevent neural tube defects [15].

Replenishing the intracellular folate pool by the intake of exogenous folates, such as folic acid, is especially important for rapidly dividing cells, including cancer cells. Ample preclinical evidence links cellular replication to folate availability [[16], [17], [18], [19], [20], [21], [22], [23]]. Such a link raised the concern that excessive consumption of dietary folate or folic acid may promote the progression of neoplastic lesions and increase the risk of malignancies and cancer-related death [11,[24], [25], [26]]. In support of such concern, animal studies have demonstrated that high intake of folic acid promotes the progression of established tumors [[27], [28], [29], [30], [31]]. A similar conclusion has been drawn based on the observed association between serum folate levels and proliferation of cancer cells in men with prostate cancer [32]. Of note, while additional studies support this conclusion [33], other studies have shown inverse associations between dietary folate and tumor development [34]. Nevertheless, the possibility that excessive intake of folates might promote development of malignancies should not be ignored [13,35,36].

The role of folate in cancer metastasis is even less clear and the literature on this subject is limited and somewhat controversial. Low extracellular folate is associated with altered expression of genes involved in cell adhesion, migration and invasion [[37], [38], [39]] indicating that the vitamin might play a role in the mechanism of metastasis. Of note, the positive association between folic acid supplementation and the migratory ability of cancer cells, as well as the inhibitory effects of antifolates on cell migration, have been demonstrated [[40], [41], [42]]. In further support of the role of folate metabolism in metastasis, metastasizing melanomas have been shown to have increased dependence on NADPH-generating folate pathways [43]. Also, antifolate pemetrexed has been approved by the FDA for the treatment of advanced or metastatic non-small cell lung cancer [[44], [45], [46]]. Several studies, however, suggested that folate supplementation would inhibit rather than promote metastasis. For example, a recent report has demonstrated that in cultured cells folate deficiency causes the elevation of reactive oxygen species, which leads to the suppression of proliferation, but at the same time enhances metastatic potential [47]. In agreement with these findings, folate deficiency promoted metastasis in a xenograft mouse model [47]. In another study, folate deprivation enhanced invasiveness of colon cancer cells through the activation of sonic hedgehog and NF-κB signaling [48]. Inhibitory effects of folic acid on metastasis and proliferation have been also linked to the activation of the folate receptor-ERK1/2-TSLC1 signaling pathway [49]. Contrary to these studies, we have previously reported that folate deprivation inhibits migration and invasion of cultured cancer cells and decreases tumor growth and metastasis in a mouse model [28,42,50].

In a recent study, we have shown that folic acid restriction led to metabolic reprogramming and a less aggressive phenotype in cultured murine triple-negative breast cancer (TNBC) cells [50]. Specifically, cells kept on folic acid-free medium displayed reduced migration and had weaker invasion potential [50]. Interestingly, effects of folic acid withdrawal were more profound in more tumorigenic mesenchymal cells than in epithelial cells [50]. To obtain insight into potential mechanisms related to folate restriction, in the present study we have evaluated transcriptomic profiles in three mouse TNBC cell lines grown in medium with or without folic acid.

Section snippets

Cell lines

Epithelial (E-Wnt) and mesenchymal (M-Wnt) cells were each derived from the same pool of mammary tumors from mouse mammary tumor virus (MMTV)-Wnt-1 transgenic mice [50,51]. Spontaneous mammary tumors were excised, dissected and mechanically dissociated, and viable cells were plated [51]. Based on morphology, specific clones with epithelial and mesenchymal properties were selected, further characterized and eventually denoted as E-Wnt and M-Wnt cells, respectively [51]. Metastatic metM-Wntliver

Effects of folic acid withdrawal on transcriptomic profiles in mouse TNBC cell lines

We have previously demonstrated that folic acid withdrawal suppresses proliferation and motility, while promoting metabolic reprogramming, in E-Wnt, M-Wnt and metM-Wntliver cells [50]. Herein, we have determined the transcriptional response of these cells following withdrawal of folic acid. Based on the top 500 probes with most variable gene expression, the epithelial cells (E-Wnt) showed expression profiles distinct from the mesenchymal cells (M-Wnt and metM-Wntliver). Additionally, the M-Wnt

Discussion and conclusions

Folate is known to play an important role in human health and disease due to its participation in diverse metabolic pathways as well as in the regulation of methylation reactions. Pleiotropic effects of folate in cancer cells have been linked to migratory capacity and invasiveness; though effects may differ by cancer type, stage and molecular subtype [50,61,62]. We have previously shown that folic acid withdrawal resulted in metabolic and bioenergetic changes indicative of a less proliferative

CRediT authorship contribution statement

Dieuwertje E. Kok: Formal analysis, Writing - original draft, Writing - review & editing. Ciara H. O’Flanagan: Data curation, Writing - review & editing. Michael F. Coleman: Data curation, Writing - review & editing. Zahra Ashkavand: Data curation, Writing - review & editing. Stephen D. Hursting: Conceptualization, Supervision, Writing - review & editing. Sergey A. Krupenko: Conceptualization, Supervision, Writing - original draft, Writing - review & editing.

Declaration of competing interest

The authors declare no conflict of interest.

Acknowledgments

This study was supported by a grant from the National Cancer Institute (R35 CA197627) to SDH. SAK is supported by the National Institutes of Health grant DK117854. DEK is supported by a Veni grant (016.Veni.188.082) of the Dutch Research Council. Funding agencies had no involvement in study design, collection, analysis or interpretation of data, writing of the report or decision to submit the manuscript. The authors thank Dr. David Horita for carefully reading the manuscript.

References (82)

  • A.M. Troen et al.

    Unmetabolized folic acid in plasma is associated with reduced natural killer cell cytotoxicity among postmenopausal women

    J. Nutr.

    (2006)
  • H. Sawaengsri et al.

    High folic acid intake reduces natural killer cell cytotoxicity in aged mice

    J. Nutr. Biochem.

    (2016)
  • X. Yang et al.

    Targeting the tumor microenvironment with interferon-β bridges innate and adaptive immune responses

    Canc. Cell

    (2014)
  • O. Gluz et al.

    Triple-negative high-risk breast cancer derives particular benefit from dose intensification of adjuvant chemotherapy: results of WSG AM-01 trial

    Ann. Oncol.

    (2008)
  • L.Y. Zhou et al.

    Potential role of pemetrexed in metastatic breast cancer patients pre-treated with anthracycline or taxane

    Chronic Dis Transl Med

    (2015)
  • A.F. Chambers et al.

    Dissemination and growth of cancer cells in metastatic sites

    Nat. Rev. Canc.

    (2002)
  • N. Sethi et al.

    Unravelling the complexity of metastasis - molecular understanding and targeted therapies

    Nat. Rev. Canc.

    (2011)
  • E. Sahai

    Illuminating the metastatic process

    Nat. Rev. Canc.

    (2007)
  • T.M. Thomson et al.

    Metabolic plasticity and epithelial-mesenchymal transition

    J. Clin. Med.

    (2019)
  • C. Lehuede et al.

    Metabolic plasticity as a determinant of tumor growth and metastasis

    Canc. Res.

    (2016)
  • M. Sciacovelli et al.

    Metabolic reprogramming and epithelial-to-mesenchymal transition in cancer

    FEBS J.

    (2017)
  • B. De Craene et al.

    Regulatory networks defining EMT during cancer initiation and progression

    Nat. Rev. Canc.

    (2013)
  • J. Fan et al.

    Quantitative flux analysis reveals folate-dependent NADPH production

    Nature

    (2014)
  • A.S. Tibbetts et al.

    Compartmentalization of Mammalian folate-mediated one-carbon metabolism

    Annu. Rev. Nutr.

    (2010)
  • K.C. Strickland et al.

    Molecular mechanisms underlying the potentially adverse effects of folate

    Clin. Chem. Lab. Med.

    (2013)
  • P.J. Stover

    Physiology of folate and vitamin B12 in health and disease

    Nutr. Rev.

    (2004)
  • E.A. Williams

    Folate, colorectal cancer and the involvement of DNA methylation

    Proc. Nutr. Soc.

    (2012)
  • M.A. Honein et al.

    Impact of folic acid fortification of the US food supply on the occurrence of neural tube defects

    J. Am. Med. Assoc.

    (2001)
  • M.J. Koury et al.

    Apoptosis mediates and thymidine prevents erythroblast destruction in folate deficiency anemia

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

    (1994)
  • C. Courtemanche et al.

    Folate deficiency inhibits the proliferation of primary human CD8+ T lymphocytes in vitro

    J. Immunol.

    (2004)
  • Y. Chen et al.

    Folic acid deficiency inhibits neural rosette formation and neuronal differentiation from rhesus monkey embryonic stem cells

    J. Neurosci. Res.

    (2012)
  • C. Moussa et al.

    Altered folate metabolism modifies cell proliferation and progesterone secretion in human placental choriocarcinoma JEG-3 cells

    Br. J. Nutr.

    (2015)
  • N. Lamm et al.

    Folate levels modulate oncogene-induced replication stress and tumorigenicity

    EMBO Mol. Med.

    (2015)
  • S.Y. Hwang et al.

    Folic acid is necessary for proliferation and differentiation of C2C12 myoblasts

    J. Cell. Physiol.

    (2018)
  • M. Ebbing et al.

    Cancer incidence and mortality after treatment with folic acid and vitamin B12

    J. Am. Med. Assoc.

    (2009)
  • K.S. Crider et al.

    Folic acid food fortification-its history, effect, concerns, and future directions

    Nutrients

    (2011)
  • J.B. Mason

    Folate, cancer risk, and the Greek god, Proteus: a tale of two chameleons

    Nutr. Rev.

    (2009)
  • M.F. Hansen et al.

    High folic acid diet enhances tumour growth in PyMT-induced breast cancer

    Br. J. Canc.

    (2017)
  • G. Bistulfi et al.

    Dietary folate deficiency blocks prostate cancer progression in the TRAMP model

    Canc. Prev. Res.

    (2011)
  • S. Deghan Manshadi et al.

    Folic acid supplementation promotes mammary tumor progression in a rat model

    PloS One

    (2014)
  • G.M. Lindzon et al.

    Effect of folic acid supplementation on the progression of colorectal aberrant crypt foci

    Carcinogenesis

    (2009)
  • Cited by (7)

    • Effect of folic acid supplementation on lactation performance of Holstein dairy cows: A meta-analysis

      2023, Animal Feed Science and Technology
      Citation Excerpt :

      Menzies et al. (2009) utilized comparative functional genomics to lactation adapted cows and found a 12.7-fold increase of folate receptor α gene expression at periods of high milk protein production. Folic acid, as a coenzyme in one-carbon transfer reactions (Kok et al., 2020), is involved in the biosynthesis of amino acids and further affect milk protein production. La et al. (2019) proved that FA heightened gene expression of protein synthesis in liver of dairy cows.

    • Suboptimal folic acid exposure rewires oncogenic metabolism and proteomics signatures to mediate human breast cancer malignancy

      2022, Journal of Nutritional Biochemistry
      Citation Excerpt :

      By using metabolomics to quantify metabolite changes in mouse mammary tumor–derived BC subtypes, Krupenko and colleagues [20] reported that invasive and metastatic BC subtypes grown in folate-free microenvironment exhibited less aggressive phenotypes in terms of inhibited glycolysis; decreased lactate; and restricted BC proliferation, migration, and invasion. In another study, the authors demonstrated that transcriptomic profiling of these cells in FA-free media strongly affects interferon-response genes, and the variation in gene expression is based on cell phenotype and BC subtypes [21]. Microarray transcriptome-wide analysis revealed that exposure of nontransformed breast and three BC subtypes to 100 nM FA (i.e., low FA levels) led to the upregulation of pathways associated with cell migration, proliferation, and vascularization [22].

    • Food and omics: unraveling the role of food in breast cancer development

      2021, Current Opinion in Food Science
      Citation Excerpt :

      The same technology has confirmed an important modification of gene expression upon docosahexaenoic acid (DHA) incorporation in the same triple-negative cell line (MDA-MB-231) [58]. Similarly, gene expression microarrays have been used to investigate the potential of folic acid for metabolic reprogramming of triple-negative breast cancer in mouse cell models [59]. In a study performed on animal models, Li et al. [16] combined tumor observation and histology with epigenetics and transcriptomics.

    View all citing articles on Scopus
    1

    These authors contributed equally.

    View full text