Elsevier

Cell Calcium

Volume 79, May 2019, Pages 80-88
Cell Calcium

The lysosomal TRPML1 channel regulates triple negative breast cancer development by promoting mTORC1 and purinergic signaling pathways

https://doi.org/10.1016/j.ceca.2019.02.010Get rights and content

Highlights

  • TRPML1 is specifically upregulated in triple-negative breast cancers (TNBCs).

  • Downregulation and pharmacological inhibition suppress the growth of TNBCs.

  • TRPML1 regulates TNBC development through controlling mTORC1 activity and lysosomal ATP release.

Abstract

The triple-negative breast cancer (TNBC) that comprises approximately 10%–20% of breast cancers is an aggressive subtype lacking effective therapeutics. Among various signaling pathways, mTORC1 and purinergic signals have emerged as potentially fruitful targets for clinical therapy of TNBC. Unfortunately, drugs targeting these signaling pathways do not successfully inhibit the progression of TNBC, partially due to the fact that these signaling pathways are essential for the function of all types of cells. In this study, we report that TRPML1 is specifically upregulated in TNBCs and that its genetic downregulation and pharmacological inhibition suppress the growth of TNBC. Mechanistically, we demonstrate that TRPML1 regulates TNBC development, at least partially, through controlling mTORC1 activity and the release of lysosomal ATP. Because TRPML1 is specifically activated by cellular stresses found in tumor microenvironments, antagonists of TRPML1 could represent anticancer drugs with enhanced specificity and potency. Our findings are expected to have a major impact on drug targeting of TNBCs.

Introduction

Breast cancer is a very common and fatal cancer in women. It is a heterogeneous disease classified into three major subtypes. Two of these are defined by the expression of steroid hormone receptors (estrogen receptor [ER] and progesterone receptor [PR]) and overexpression and/or amplification of the receptor tyrosine kinase HER2 (also known as ERBB2). Agents targeting these proteins have led to a significant increase in patient survival [[1], [2], [3], [4], [5]]. In contrast, the most aggressive and metastatic form of breast cancer is the triple-negative breast cancer (TNBC) subtype, which is defined by the lack of expression of ER, PR and HER2 and its resistance to common therapies targeting these molecules [[1], [2], [3], [4], [5]]. TNBC represents approximately 10%–20% of breast cancers [6,7]. The major cause of TNBC mortality is the propensity of TNBC cells to metastasize to nearby normal tissues and organs and even spread to distant parts of the body through the blood or lymph system. Therefore, understanding the basic cellular and molecular mechanisms of TNBC development is crucial for devising new therapies for this disease.

The mammalian target of rapamycin complex 1 (mTORC1) is crucial for autophagy induction in response to nutrient shortage and cellular stress. Continuous loss of mTORC1 activity during nutrient and energy deprivation leads to cell death [8,9] because mTORC1 is also required for protein synthesis, metabolism, cell growth and proliferation and cellular homeostasis [[10], [11], [12]]. Mounting evidence suggests that mTORC1 represents a major signaling pathway responsible for cancer cell growth and survival [13,14]. Hyperactivation of mTORC1 pathway is common in human cancer including breast cancer [12,14,15]. In particular, activated mTORC1 is more frequently observed in TNBC (36%) than in other subtypes of breast cancer, rendering mTORC1 a potential target for TNBC therapy [[16], [17], [18]]. As such, several molecules that inhibit mTORC1 are currently in clinical trials for the treatment of multiple cancer types, including breast cancer [17,[19], [20], [21], [22], [23]].

Adenosine triphosphate (ATP) is actively released to the extracellular environment in response to tissue damage and cellular stress [24]. Increased levels of extracellular ATP at tumor sites have been suggested to play a key role in host-tumor interaction [[24], [25], [26]], likely acting on their specific cell surface receptors to increase motility, invasion and metastasis of many cancers [[27], [28], [29], [30]], including breast cancer [31,32]. Interestingly, the lysosome plays an important role in mTORC1 signaling and is a major source of ATP. Specifically, mTORC1 is recruited to the surface of the lysosome to be activated by nutrients within the lysosome [33]. Lysosomal ATP (˜ 1 mM) [34,35] can be released into extracellular space via lysosomal exocytosis [[35], [36], [37]], thereby regulating cancer migration and invasion [36,38]. Thus, lysosomes may regulate cancer development by controlling both mTORC1 and ATP signaling pathways.

Recently, we [39], and others [40,41], demonstrated that Transient Receptor Potential Mucolipin 1 (TRPML1 or ML1), the lysosomal Ca2+ release channel, is required for sustained activity of mTORC1 under nutrient stress but not under normal conditions. This regulation of mTORC1 by ML1 is essential for cells to survive extreme conditions [39]. Mounting evidence also suggests that ML1 is a key regulator of lysosomal exocytosis [[42], [43], [44]]. Here we show that ML1 supports tumor development by promoting both mTORC1 activity and lysosomal ATP release. We show that ML1 is markedly upregulated in highly metastatic TNBC when compared with non-metastatic ER+/PR+ MCF-7 breast cancer cell, as well as the non-tumorigenic MCF-10 A epithelial cell line. The mRNA of ML1 from TNBC patients is also significantly higher than that from ER+/PR+ breast cancer patients. Molecular abrogation or pharmacological inhibition of ML1 suppresses TNBC cell proliferation and invasion in vitro, and tumor growth and progression in vivo. In addition, we demonstrate that ML1 regulates TNBC growth by promoting mTORC1 activity and facilitating TNBC invasion through increasing lysosomal ATP release. Our work suggests that inhibiting ML1 could be a potential therapeutic strategy for TNBC.

Section snippets

Cell culture

MCF10 A breast normal cells were grown in DMEM/F12 (1:1). Human breast cancer cell lines MDA-MB-231, MCF7, and Hs 578 T were grown in DMEM, while SUM159PT were cultured using F12 medium (Life Technologies, Carlsbad, CA) supplemented with 10% FBS (Life Technologies). All cultures were maintained at 37 °C in a 5% CO2 incubator.

Antibodies, plasmids and reagents

The following primary antibodies were used for western blotting and immunofluorescence staining: anti-p-p70S6K (T389) (9206, Cell Signaling Technology), anti-p70S6K (Cell

ML1 is highly expressed in metastatic breast cancer cells and breast tumors

We first investigated the expression profile of ML1 in non-tumorigenic MCF-10 A cells and several breast cancer cell lines, including the non-metastatic ER+/PR+ breast cancer cell line MCF-7 and three metastatic TNBC cell lines MDA-MB-231, Hs 578 T and SUM159PT [50,51]. ML1 expression levels were gauged by quantitative PCR with reverse transcription (RT-PCR). ML1 mRNA was found to be specifically upregulated in aggressive, metastatic TNBC cell lines including MDA-MB-231, Hs 578 T and SUM159PT

Discussion

Although TNBC is the most aggresive malignant cancercausing high mortality rates among women, therapeutic options for this disease remain limited. This is partially due to the lack of our understanding of key pathways associated with TNBCs. In this study, we report that ML1 is specifically elevated in TNBC cell lines and patients and is associated with increased mTORC1 activity and lysosomal ATP release into the extracellular space to promote TNBC growth and invasion.

In agreement with previous

Conflicts of interest statement

The authors declare that there is no conflict of interest.

Acknowledgements

This work was supported by DMRF Equipment Grant, CIHR grant (PJT-156102), and CFI Leaders Opportunity Fund-Funding for research infrastructure (29291). We thank Mitsunori Fukuda for the Syt-VII-DN plasmid and Haoxing Xu for his constant support. We appreciate the encouragement and helpful comments from other members of the Dong laboratory and El Hiani laboratories.

References (91)

  • D. Schumacher et al.

    Platelet-derived nucleotides promote tumor-cell transendothelial migration and metastasis via P2Y2 receptor

    Cancer Cell

    (2013)
  • Q. Cao et al.

    Dong XP: SLC17A9 protein functions as a lysosomal ATP transporter and regulates cell viability

    J. Biol. Chem.

    (2014)
  • C.O. Wong et al.

    Drosophila TRPML is required for TORC1 activation

    Curr. Biol.

    (2012)
  • Q. Cao et al.

    BK channels alleviate lysosomal storage diseases by providing positive feedback regulation of lysosomal Ca2+ release

    Dev. Cell

    (2015)
  • M. Samie et al.

    A TRP channel in the lysosome regulates large particle phagocytosis via focal exocytosis

    Dev. Cell

    (2013)
  • M.A. Samie et al.

    Lysosomal exocytosis and lipid storage disorders

    J. Lipid Res.

    (2014)
  • Y. Sancak et al.

    Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids

    Cell

    (2010)
  • M. Vilella-Bach et al.

    The FKBP12-rapamycin-binding domain is required for FKBP12-rapamycin-associated protein kinase activity and G1 progression

    J. Biol. Chem.

    (1999)
  • Y. Miao et al.

    A TRP channel senses lysosome neutralization by pathogens to trigger their expulsion

    Cell

    (2015)
  • D. Hoshino et al.

    Exosome secretion is enhanced by invadopodia and drives invasive behavior

    Cell Rep.

    (2013)
  • P. Marcato et al.

    Aldehyde dehydrogenase 1A3 influences breast cancer progression via differential retinoic acid signaling

    Mol. Oncol.

    (2015)
  • G.P. Gupta et al.

    Cancer metastasis: building a framework

    Cell

    (2006)
  • M.N. Sharifi et al.

    Autophagy promotes focal adhesion disassembly and cell motility of metastatic tumor cells through the direct interaction of Paxillin with LC3

    Cell Rep.

    (2016)
  • H. Maes et al.

    Tumor vessel normalization by chloroquine independent of autophagy

    Cancer Cell

    (2014)
  • A.K. Murugan et al.

    Mutations in critical domains confer the human mTOR gene strong tumorigenicity

    J. Biol. Chem.

    (2013)
  • B. Levine et al.

    Autophagy in the pathogenesis of disease

    Cell

    (2008)
  • C. Cang et al.

    Ren D: mTOR regulates lysosomal ATP-sensitive two-pore Na(+) channels to adapt to metabolic state

    Cell

    (2013)
  • Y. Zhang et al.

    BRMS1 sensitizes breast Cancer cells to ATP-Induced growth suppression

    Biores. Open Access

    (2013)
  • C. Arsenis et al.

    Degradation of nucleic acids by lysosomal extracts of rat liver and Ehrlich ascites tumor cells

    J. Biol. Chem.

    (1970)
  • L. Ma et al.

    Tumour invasion and metastasis initiated by microRNA-10b in breast cancer

    Nature

    (2007)
  • L. Lin et al.

    CCL18 from tumor-associated macrophages promotes angiogenesis in breast cancer

    Oncotarget

    (2015)
  • C. Yam et al.

    Targeting the molecular subtypes of triple negative breast cancer: understanding the diversity to progress the field

    Oncologist

    (2017)
  • L.A. Carey et al.

    The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes

    Clin. Cancer Res.

    (2007)
  • M.C. Maiuri et al.

    Self-eating and self-killing: crosstalk between autophagy and apoptosis

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • Y. Liu et al.

    Autosis and autophagic cell death: the dark side of autophagy

    Cell Death Differ.

    (2015)
  • A.C. Hsieh et al.

    The translational landscape of mTOR signalling steers cancer initiation and metastasis

    Nature

    (2012)
  • S.A. Wander et al.

    PI3K/mTOR inhibition can impair tumor invasion and metastasis in vivo despite a lack of antiproliferative action in vitro: implications for targeted therapy

    Breast Cancer Res. Treat.

    (2013)
  • M. Cargnello et al.

    The expanding role of mTOR in cancer cell growth and proliferation

    Mutagenesis

    (2015)
  • S.H. Ueng et al.

    Phosphorylated mTOR expression correlates with poor outcome in early-stage triple negative breast carcinomas

    Int. J. Clin. Exp. Pathol.

    (2012)
  • R. Hatem et al.

    Targeting mTOR pathway inhibits tumor growth in different molecular subtypes of triple-negative breast cancers

    Oncotarget

    (2016)
  • E. Paplomata et al.

    The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers

    Ther. Adv. Med. Oncol.

    (2014)
  • S.A. Wander et al.

    Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy

    J. Clin. Invest.

    (2011)
  • B. Marty et al.

    Frequent PTEN genomic alterations and activated phosphatidylinositol 3-kinase pathway in basal-like breast cancer cells

    Breast Cancer Res.

    (2008)
  • R.J. Shaw et al.

    Ras, PI(3)K and mTOR signalling controls tumour cell growth

    Nature

    (2006)
  • F. Di Virgilio et al.

    Extracellular purines, purinergic receptors and tumor growth

    Oncogene

    (2016)
  • Cited by (39)

    • p53 mitigates the effects of oncogenic HRAS in urothelial cells via the repression of MCOLN1

      2021, iScience
      Citation Excerpt :

      As a result, TRPML1 inhibition or MCOLN1 knockdown disrupts ERK activation and cell proliferation in HRAS-driven cancer cells. TRPML1-dependent vesicle exocytosis is also necessary for the release of lysosomal ATP, which in the case of triple-negative breast cancers, stimulates cell migration and metastases (Xu et al., 2019). In other cancers, TRPML1 promotes untrammeled proliferation and tumor growth via mTORC1 (Hu et al., 2019; Xu et al., 2019).

    • Two-pore and TRPML cation channels: Regulators of phagocytosis, autophagy and lysosomal exocytosis

      2021, Pharmacology and Therapeutics
      Citation Excerpt :

      Increased levels of ATP secretion, which occurs via lysosomal exocytosis, result in increased motility, invasion and metastasis of many cancers (W. H. Li et al., 2013; Schumacher, Strilic, Sivaraj, Wettschureck, & Offermanns, 2013; Shi, Queiroz, Stap, Richel, & Spek, 2013; Y. Zhang et al., 2010). Xu and collaborators showed that, in TNBC, TRPML1 supports tumor growth by promoting mTORC1 activity and lysosomal ATP release by enhancing lysosomal exocytosis (M. Xu et al., 2019). Thus, molecular abrogation or pharmacological inhibition of TRPML1 was shown to suppress TNBC proliferation and invasion in vitro, and tumor growth and progression in vivo (M. Xu et al., 2019).

    View all citing articles on Scopus
    1

    Equal contribution.

    View full text