Role of the autotaxin–lysophosphatidate axis in cancer resistance to chemotherapy and radiotherapy

https://doi.org/10.1016/j.bbalip.2012.08.015Get rights and content

Abstract

High expression of autotaxin in cancers is often associated with increased tumor progression, angiogenesis and metastasis. This is explained mainly since autotaxin produces the lipid growth factor, lysophosphatidate (LPA), which stimulates cell division, survival and migration. It has recently become evident that these signaling effects of LPA also produce resistance to chemotherapy and radiation-induced cell death. This results especially from the stimulation of LPA2 receptors, which depletes the cell of Siva-1, a pro-apoptotic signaling protein and stimulates prosurvival kinase pathways through a mechanism mediated via TRIP-6. LPA signaling also increases the formation of sphingosine 1-phosphate, a pro-survival lipid. At the same time, LPA decreases the accumulation of ceramides, which are used in radiation therapy and by many chemotherapeutic agents to stimulate apoptosis. The signaling actions of extracellular LPA are terminated by its dephosphorylation by a family of lipid phosphate phosphatases (LPP) that act as ecto-enzymes. In addition, lipid phosphate phoshatase-1 attenuates signaling downstream of the activation of both LPA receptors and receptor tyrosine kinases. This makes many cancer cells hypersensitive to the action of various growth factors since they often express low LPP1/3 activity. Increasing our understanding of the complicated signaling pathways that are used by LPA to stimulate cell survival should identify new therapeutic targets that can be exploited to increase the efficacy of chemo- and radio-therapy. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.

Highlights

► Autotaxin and LPA receptors are upregulated in metastatic cancers. ► LPA receptors, particularly the LPA2 subtype convey resistance against apoptosis. ► LPA is removed from the extracellular compartment by lipid phosphate phosphatases. ► Expression of lipid phosphate phosphatases is often low in cancers. ► The ATX–LPA receptor–LPP axis influences the chemo- and radiation-resistance of cancers.

Section snippets

Introduction and overview

The involvement of the lipid mediator lysophosphatidate (LPA) in regulating tumor progression, angiogenesis and metastasis has been increasingly recognized over the last decade. Much of the impetus for this started with the identification of autotaxin (ATX) as an extracellular lysophospholipase D, whose major signaling effect is to generate extracellular LPA [1], [2], [3]. ATX was originally identified in 1992 in the media derived from A2058 human melanoma cells. Stracke et al. showed that

Role of autotaxin and lysophosphatidate in tumor biology

Lysophosphatidylcholine (LPC) is the most abundant phospholipid in blood plasma where it reaches concentrations of about 200 μM in human beings [12]. The liver secretes unsaturated LPC [13] and presumably, this depends on the activity of a phospholipase A1. It is probable that other organs also have this ability to secrete unsaturated LPC. Saturated LPC in the circulation is produced mainly by lecithin:cholesterol acyltransferase acting on the phosphatidylcholine in high density lipoproteins.

Other routes of LPA formation

Apart from the actions of autotaxin, other pathways can produce extracellular LPA (Fig. 1). One such pathway involves secretory phospholipase A2 (sPLA2), which produces LPA from phosphatidate (PA). This latter lipid is present in micro-vesicles released during inflammation reactions [29]. Also, group VIA phospholipase A2 (Ca2 + independent phospholipase A2β, iPLA2β) produces extracellular LPA in human epithelial ovarian cancer cells [30], [31]. The importance of this reaction in ovarian cancer

Turnover of extracellular LPA through the ecto-activities of lipid phosphate phosphatases

Plasma LPA concentrations are normally < 1 μM, but they have been reported to reach > 10 μM in ovarian cancer and this depends mainly on LPA production by ATX [8]. LPA is also degraded rapidly as expected for an important bioactive compound (Fig. 1). This is achieved through the actions of a family of three enzymes that are called lipid phosphate phosphatases (LPPs) [39]. These enzymes dephosphorylate a large variety of bioactive lipid phosphates and pyrophosphates, including LPA [40]. The location

Role of lipid phosphate phosphatases in regulating cell signaling downstream of the activation of growth factor receptors

The LPPs are expressed on internal membranes including the endoplasmic reticulum [42], [54] and the Golgi network [55]. It is assumed that the catalytic sites face the lumenal side of these membranes. This internal LPP activity appears to regulate the turnover of lipid phosphates that are formed downstream of the activation of GPCR, including the LPA receptors, as well as receptor tyrosine kinases. This action controls the concentrations of the lipid phosphates relative to their

Effects of LPA signaling in controlling the proapoptotic effects of ceramides versus the survival signals from sphingosine 1-phosphate

Many chemotherapeutic agents (including Taxol, doxorubicin and vincristine) and radiation therapy increase ceramide formation as part of their therapeutic actions in stimulating cell death (Fig. 3) [89], [90], [91], [92], [93], [94], [95]. Ceramides are sphingolipids that release cytochrome C from mitochondria and activate caspases to initiate apoptosis [93], [96]. The increased balance of ceramide versus S1P signaling is considered to be an important “rheostat” that favors the death versus

Overexpression of LPA receptor subtypes in different types of cancers

LPA elicits many of its cellular actions via the stimulation of specific GPCRs. LPA activates two clusters of GPCR. To date, six GPCR have been validated as specific targets of LPA. These include three GPCR encoded by the Endothelial Differentiation Gene family and designated LPA1, LPA2, and LPA3. Three other GPCR that include LPA4(GPR23/P2Y9), LPA5(GPR92), and LPA6(P2Y5) are related to the purinergic family of GPCR. There are three other GPCR including GPR87 [103], [104], P2Y10 [105], and

Role of LPA receptors in mediating resistance to genotoxic agents (chemo and radiation therapy)

There is evidence for the oncogenic transforming action of LPA GPCR. Taghavi et al. [10] have evaluated the role of LPA1/2/3/4 in malignant transformation and found that the rank order of transforming activity was LPA2 > LPA1 > LPA4. LPA3 did not promote the growth in soft-agar of mouse embryonic fibroblasts immortalized with the Tbx2 transcriptional suppressor and c-Myc. When these fibroblasts were injected into nude mice the rank order of survival was LPA1 < LPA2 < LPA4 whereas, LPA3 cells did not

Unique aspects of LPA2 receptor signaling underlying radiation and chemoresistance

Tumor cells upregulate lipid synthesis as a requirement for increased cell proliferation. Lipogenesis is controlled by the sterol regulatory element binding proteins (SREBP). Mukherjee and colleagues recently showed in carcinoma cells, but not in nontransformed cells, that LPA upregulates the transcriptional activity of SREBP [146], which is the rate-limiting step in lipogenesis. The 5′ adenosine monophosphate-activated protein kinase (AMPK) plays a role in cellular energy homeostasis. The net

Concluding remarks

The treatment of various cancers often fails due to the development of resistance to the actions of various chemotherapies and radiotherapy and patients die mainly because of the spread of metastases. This resistance depends largely on the effects of various growth factors including estrogens, EGF, PDGF, VEGF etc. that enable cancer cells to avoid cell death. We propose that another growth factor called LPA contributes to these problems of chemo-resistance. LPA is generated outside the cells

Acknowledgements

The work was supported by grants to DNB from the Canadian Institutes of Health Research, the Alberta Cancer Foundation and the Canadian Breast Cancer Foundation, by grants to GT from the National Cancer Institute, CA92160, AI80405 from the National Institute of Allergy Medical Countermeasures Radiological and Nuclear Threats Program, and by the Harriet Van Vleet Endowment for Basic Oncology Research.

References (164)

  • O. Fourcade et al.

    Secretory phospholipase A2 generates the novel lipid mediator lysophosphatidic acid in membrane microvesicles shed from activated cells

    Cell

    (1995)
  • X. Zhao et al.

    Caspase-3-dependent activation of calcium-independent phospholipase A2 enhances cell migration in non-apoptotic ovarian cancer cells

    J. Biol. Chem.

    (2006)
  • N.A. Moughal et al.

    Nerve growth factor signaling involves interaction between the Trk A receptor and lysophosphatidate receptor 1 systems: nuclear translocation of the lysophosphatidate receptor 1 and Trk A receptors in pheochromocytoma 12 cells

    Cell. Signal.

    (2004)
  • F. Gobeil et al.

    Modulation of pro-inflammatory gene expression by nuclear lysophosphatidic acid receptor type-1

    J. Biol. Chem.

    (2003)
  • G.D. Girnun et al.

    Synergy between PPARgamma ligands and platinum-based drugs in cancer

    Cancer Cell

    (2007)
  • J. Yue et al.

    Mice with transgenic overexpression of lipid phosphate phosphatase-1 display multiple organotypic deficits without alteration in circulating lysophosphatidate level

    Cell. Signal.

    (2004)
  • D. Brindley et al.

    Phosphatidate degradation: phosphatidate phosphatases (lipins) and lipid phosphate phosphatases

    Biochim. Biophys. Acta

    (2009)
  • S. Milstien et al.

    Targeting sphingosine-1-phosphate: a novel avenue for cancer therapeutics

    Cancer Cell

    (2006)
  • Y. Zhao et al.

    Intracellular generation of sphingosine 1-phosphate in human lung endothelial cells: role of lipid phosphate phosphatase-1 and sphingosine kinase 1

    J. Biol. Chem.

    (2007)
  • A. Martin et al.

    Decreased activities of phosphatidate phosphohydrolase and phospholipase D in Ras and tyrosine kinase (fps) transformed fibroblasts

    J. Biol. Chem.

    (1993)
  • D. Barila et al.

    The Dri 42 gene, whose expression is up-regulated during epithelial differentiation, encodes a novel endoplasmic reticulum resident transmembrane protein

    J. Biol. Chem.

    (1996)
  • M. Kai et al.

    Cloning and characterization of two human isozymes of Mg2 +‐independent phosphatidic acid phosphatase

    J. Biol. Chem.

    (1997)
  • S. Pyne et al.

    Lysophosphatidic acid and sphingosine 1-phosphate biology: the role of lipid phosphate phosphatases

    Semin. Cell Dev. Biol.

    (2004)
  • F. Alderton et al.

    G-protein-coupled receptor stimulation of the p42/p44 mitogen-activated protein kinase pathway is attenuated by lipid phosphate phosphatases 1, 1a, and 2 in human embryonic kidney 293 cells

    J. Biol. Chem.

    (2001)
  • C. Pilquil et al.

    Lipid phosphate phosphatase-1 regulates lysophosphatidate-induced fibroblast migration by controlling phospholipase D2-dependent phosphatidate generation

    J. Biol. Chem.

    (2006)
  • I. Garcia-Murillas et al.

    Lazaro encodes a lipid phosphate phosphohydrolase that regulates phosphatidylinositol turnover during Drosophila phototransduction

    Neuron

    (2006)
  • S. Ghosh et al.

    Raf-1 kinase possesses distinct binding domains for phosphatidylserine and phosphatidic acid. Phosphatidic acid regulates the translocation of Raf-1 in 12-O-tetradecanoylphorbol-13-acetate-stimulated Madin–Darby canine kidney cells

    J. Biol. Chem.

    (1996)
  • M.A. Rizzo et al.

    Phospholipase D and its product, phosphatidic acid, mediate agonist-dependent raf-1 translocation to the plasma membrane and the activation of the mitogen-activated protein kinase pathway

    J. Biol. Chem.

    (1999)
  • C. Delon et al.

    Sphingosine kinase 1 is an intracellular effector of phosphatidic acid

    J. Biol. Chem.

    (2004)
  • D.Y. Noh et al.

    Overexpression of phospholipase D1 in human breast cancer tissues

    Cancer Lett.

    (2000)
  • S.M. Mandala

    Sphingosine-1-phosphate phosphatases

    Prostaglandins Other Lipid Mediat.

    (2001)
  • K.E. Morris et al.

    Lipid phosphate phosphatase-2 activity regulates S-phase entry of the cell cycle in Rat2 fibroblasts

    J. Biol. Chem.

    (2006)
  • M. Serrano et al.

    Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a

    Cell

    (1997)
  • R. Martinez et al.

    Doxorubicin induces ceramide and diacylglycerol accumulation in rat hepatocytes through independent routes

    Toxicol. Lett.

    (2009)
  • A. Gomez-Munoz et al.

    Cell-permeable ceramides inhibit the stimulation of DNA synthesis and phospholipase D activity by phosphatidate and lysophosphatidate in rat fibroblasts

    J. Biol. Chem.

    (1994)
  • M. Umezu-Goto et al.

    Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production

    J. Cell Biol.

    (2002)
  • S.W. Nam et al.

    Autotaxin (NPP-2), a metastasis-enhancing motogen, is an angiogenic factor

    Cancer Res.

    (2001)
  • M. Umezu-Goto et al.

    Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production

    J. Cell Biol.

    (2002)
  • S.Y. Yang et al.

    Expression of autotaxin (NPP-2) is closely linked to invasiveness of breast cancer cells

    Clin. Exp. Metastasis

    (2002)
  • R. Bekele et al.

    Role of autotaxin and lysophosphatidate in cancer progression and resistance to chemotherapy and radiotherapy

    Clin. Lipidol.

    (2012)
  • P. Taghavi et al.

    In vitro genetic screen identifies a cooperative role for LPA signaling and c-Myc in cell transformation

    Oncogene

    (2008)
  • G. Tigyi

    Aiming drug discovery at lysophosphatidic acid targets

    Br. J. Pharmacol.

    (2010)
  • W.H. Moolenaar et al.

    The ins and outs of lysophosphatidic acid signaling

    Bioessays

    (2004)
  • C.G. Gaetano et al.

    Inhibition of autotaxin production or activity blocks lysophosphatidylcholine-induced migration of human breast cancer and melanoma cells

    Mol. Carcinog.

    (2009)
  • N. Samadi et al.

    Autotaxin protects MCF-7 breast cancer and MDA-MB-435 melanoma cells against Taxol-induced apoptosis

    Oncogene

    (2009)
  • H.M. Albers et al.

    Boronic acid-based inhibitor of autotaxin reveals rapid turnover of LPA in the circulation

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

    (2010)
  • G. Ferry et al.

    S32826, a nanomolar inhibitor of autotaxin: discovery, synthesis and applications as a pharmacological tool

    J. Pharmacol. Exp. Ther.

    (2008)
  • J. Gierse et al.

    A novel autotaxin inhibitor reduces lysophosphatidic acid levels in plasma and the site of inflammation

    J. Pharmacol. Exp. Ther.

    (2010)
  • A.L. Parrill et al.

    Autotaxin inhibition: challenges and progress toward novel anti-cancer agents

    Anticancer Agents Med Chem.

    (2008)
  • J. Hausmann et al.

    Structural basis of substrate discrimination and integrin binding by autotaxin

    Nat. Struct. Mol. Biol.

    (2011)
  • Cited by (106)

    • Role of adipose-derived stem cells in breast cancer

      2023, Chinese Journal of Plastic and Reconstructive Surgery
    • Molecular modelling guided design, synthesis and QSAR analysis of new small molecule non-lipid autotaxin inhibitors

      2020, Bioorganic Chemistry
      Citation Excerpt :

      In recent years there has been extensive research on autotaxin (ATX), a member of the ectonucleotide pyrophosphatase phosphodiesterase (ENPP) enzyme family (a.k.a. ENPP2), which is responsible for catalyzing the hydrolysis of lysophosphatidyl choline (LPC) and similar lysophospholipids to a growth-factor like bioactive phospholipid, lysophosphatidic acid (LPA) [1–13]. On the cell surface, LPA acts via six transmembrane G-protein coupled receptors, LPAR1-6 (LPAR), to promote a variety of physiological responses, including cell proliferation, migration, motility, survival, and platelet aggregation [6,7,13–15]. Thus, the ATX-LPA signaling pathway has been considered to be at the core of a number of diseases, including fibrotic diseases, cardiovascular diseases, inflammation, and central pain [6,7,10,13,16,17].

    • Role of the autotaxin–lysophosphatidate axis in the development of resistance to cancer therapy

      2020, Biochimica et Biophysica Acta - Molecular and Cell Biology of Lipids
    View all citing articles on Scopus

    This article is part of a Special Issue entitled Advances in Lysophospholipid Research.

    View full text