Cancer Letters

Cancer Letters

Volume 414, 1 February 2018, Pages 136-146
Cancer Letters

Original Article
MLKL-PITPα signaling-mediated necroptosis contributes to cisplatin-triggered cell death in lung cancer A549 cells

https://doi.org/10.1016/j.canlet.2017.10.047Get rights and content

Highlights

  • Autocrine TNF-α contributes to the initiation of cisplatin-triggered necroptosis of lung cancer cells.

  • PITPα interacts with MLKL.

  • PITPα is involved in the function of MLKL in necroptosis.

Abstract

Necroptosis has been reported to be involved in cisplatin-induced cell death, but the mechanisms underlying the occurrence of necroptosis are not fully elucidated. In this study, we show that apart from apoptosis, cisplatin induces necroptosis in A549 cells. The alleviation of cell death by two necroptosis inhibitors—necrostatin-1 (Nec-1) and necrosulfonamide (NSA), and the phosphorylation of mixed lineage kinase domain-like protein (MLKL) at serine 358, suggest the involvement of receptor-interacting protein kinase 1 (RIPK1)-RIPK3-MLKL signaling in cisplatin-treated A549 cells. Additionally, the initiation of cisplatin-induced necroptosis relies on autocrine tumor necrosis factor alpha (TNF-α). Furthermore, we present the first evidence that phosphatidylinositol transfer protein alpha (PITPα) is involved in MLKL-mediated necroptosis by interacting with the N terminal MLKL on its sixth helix and the preceding loop, which facilitates MLKL oligomerization and plasma membrane translocation in necroptosis. Silencing of PITPα expression interferes with MLKL function and reduces cell death. Our data elucidate that cisplatin-treated lung cancer cells undergo a new type of programmed cell death called necroptosis and shed new light on how MLKL translocates to the plasma membrane.

Introduction

The core of cancer therapy is to remove cancer cells from normal tissue. Apart from surgery, chemotherapy is the strategy that best manifests this idea. Cisplatin (cis-diamminedichloroplatinum(II), DDP), a chemotherapeutic drug targeting DNA, is still incorporated in a broad range of combined antineoplastic chemotherapy regimens for various solid tumors. Inasmuch as cisplatin remains a first-line agent in lung cancer chemotherapy, it is pivotal to fully understand its cytotoxic mechanisms in lung cancer cells. Generally, apoptosis is widely believed to be the fate of the cisplatin-treated cells. Recent researches have shown that cisplatin may cause a new type of programmed cell death known as necroptosis [1], [2], [3]. Necroptosis is substantially involved in pathological states such as organ inflammation [4], [5], arteriosclerosis [6], cerebral ischemia [7], and antiviral responses [8]. Mechanisms that initiate necroptosis differ. Chemical compounds (MNNG, shikonin), protein ligands (TNF-α, TRAIL), and analog of nucleic acids (poly (I:C)) are suggested to trigger necroptosis under certain circumstances [9], [10], [11], [12], [13]. Several antineoplastic drugs (5-fluorouracil, Obatoclax, and cisplatin) are demonstrated to kill tumor cells through necroptosis as well [14], [15], [16].

Interactions between RIPK1-RIPK3 or RIPK3-RIPK3 and the ensuing recruitment and phosphorylation of MLKL are widely acknowledged necroptosis pathways [17], [18], [19]. Phosphorylated MLKL then forms oligomers and translocates to the plasma membrane [20], [21]. It has been demonstrated that to kill cells, MLKL needs to bind on some species of phosphatidylinositol phosphates (PIPs) at the plasma membrane, and interfering with the synthesis of PIPs inhibits its killing ability [22], [23], [24]. However, the underlying mechanisms are not fully explored. PITPα belongs to the family of PITPs. It participates in the transfer of phosphatidylinositol (PI) between membranes, as well as in the regulation of PIPs synthesis [25]. Thus, we hypothesized that PITPα might assist in the function of MLKL.

In this study, we show that apart from apoptosis, cisplatin-treated A549 cells undergo necroptosis. We show that MLKL forms oligomers and shifts from the cytosol to the plasma membrane during necroptosis, and its oligomerization and membrane translocation partially depend on PITPα. Additionally, autocrine TNF-α signaling contributes to the initiation of cisplatin-induced necroptosis.

Section snippets

Reagents

Cisplatin was obtained from Qilu Pharmaceutical (Jinan, China). z-VAD(OME)-FMK was purchased from Santa Cruz Biotechnology (sc-311561; Dallas, TX, USA). Necrostatin-1 was obtained from Sigma-Aldrich (N9037; St. Louis, MO, USA). Necrosulfonamide was purchased from Abcam (ab143839; Cambridge, UK). Cholera Toxin Subunit B (Recombinant), Alexa Fluor® 594 Conjugate (CT-B) was obtained from Molecular Probes (C34777; Eugene, OR, USA). The human TNF-alpha ELISA Kit was obtained from Dakewe Biotech

Cisplatin induces both apoptosis and necroptotic-like cell death in lung cancer A549 cells

To test our hypothesis that necroptosis might contribute to cisplatin-triggered lung cancer cell death, we firstly pretreated A549 cells with zVAD-FMK, a pan-caspase inhibitor of apoptosis, before cisplatin treatment to see whether it could rescue all cells from cisplatin-induced cell death. Unexpectedly, we still noticed dead cells in A549 cells with zVAD-FMK pretreatment and the dead cells in supernatant were round and appeared to be identical, whereas, the morphology of the dead cells varied

Discussion

In many studies, the initiation of necroptosis can be ascribed to drug-induced up-regulation of RIPK3 or MLKL [1], [39]. However, in our study, the expression levels of RIPK1, RIPK3 and MLKL in A549 cells did not change after cisplatin treatment, which led us to reevaluate the mechanism and ultimately to ascribe it to autocrine TNF-α signaling. TNF-α is a classic necroptosis inducer and it triggers necroptosis especially when the capacity of caspases are inhibited [12]. Consistent with other

Funding

This work was supported by grants from the National Natural Science Foundation of China (31371405), and the National Basic Research Program of China (2015CB553701).

Acknowledgements

None.

References (51)

  • Y. Dondelinger et al.

    MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates

    Cell Rep.

    (2014)
  • G. Quarato et al.

    Sequential engagement of distinct MLKL phosphatidylinositol-binding sites executes necroptosis

    Mol. Cell

    (2016)
  • S. Cockcroft

    Phosphatidylinositol transfer proteins couple lipid transport to phosphoinositide synthesis

    Semin. Cell Dev. Biol.

    (2001)
  • S.J. Tilley et al.

    Structure-function analysis of human [corrected] phosphatidylinositol transfer protein alpha bound to phosphatidylinositol

    Structure

    (2004)
  • K. Moriwaki et al.

    Necrosis-dependent and independent signaling of the RIP kinases in inflammation

    Cytokine Growth Factor Rev.

    (2014)
  • J.M. Murphy et al.

    The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism

    Immunity

    (2013)
  • L. Duprez et al.

    RIP kinase-dependent necrosis drives lethal systemic inflammatory response syndrome

    Immunity

    (2011)
  • W. Sun et al.

    Cytosolic calcium mediates RIP1/RIP3 complex-dependent necroptosis through JNK activation and mitochondrial ROS production in human colon cancer cells

    Free Radic. Biol. Med.

    (2017)
  • M. Taipale et al.

    Quantitative analysis of HSP90-client interactions reveals principles of substrate recognition

    Cell

    (2012)
  • A.N. Hunt et al.

    Acyl chain-based molecular selectivity for HL60 cellular phosphatidylinositol and of phosphatidylcholine by phosphatidylinositol transfer protein alpha

    Biochim. Biophys. Acta

    (2004)
  • S. Hara et al.

    The first 5 amino acids of the carboxyl terminus of phosphatidylinositol transfer protein (PITP) alpha play a critical role in inositol lipid signaling. Transfer activity of PITP is essential but not sufficient for restoration of phospholipase C signaling

    J. Biol. Chem.

    (1997)
  • Y. Xu et al.

    A role for tubular necroptosis in cisplatin-induced AKI

    J. Am. Soc. Nephrol.

    (2015)
  • Y. Xu et al.

    Receptor interactive protein kinase 3 promotes cisplatin-triggered necrosis in apoptosis-resistant esophageal squamous cell carcinoma cells

    PLoS One

    (2014)
  • P.S. Welz et al.

    FADD prevents RIP3-mediated epithelial cell necrosis and chronic intestinal inflammation

    Nature

    (2011)
  • L. Cabon et al.

    BID regulates AIF-mediated caspase-independent necroptosis by promoting BAX activation

    Cell Death Differ.

    (2012)
  • Cited by (0)

    1

    These two authors contributed equally to this work.

    View full text