Cancer Letters

Cancer Letters

Volume 333, Issue 2, 10 June 2013, Pages 239-243
Cancer Letters

Inhibition of S6 kinase suppresses the apoptotic effect of eIF4E ablation by inducing TGF-β-dependent G1 cell cycle arrest

https://doi.org/10.1016/j.canlet.2013.01.041Get rights and content

Abstract

The mammalian target of rapamycin complex 1 (mTORC1) is a critical regulator of cap-dependent translation through its direct activation of ribosomal protein p70 S6 kinase (S6 kinase) and indirect activation of eukaryotic initiation factor 4E (eIF4E). We recently reported that inhibition of eIF4E expression caused apoptosis in cancer cells in the absence of serum. This was indicated by treatment with the mTORC1 inhibitor rapamycin, which suppressed both S6 kinase and 4E-BP1 phosphorylation (dephosphorylated 4E-BP1 binds and inactivates eIF4E), or by knockdown of eIF4E. We report here that knockdown of eIF4E also causes apoptosis in the presence of serum. This was unexpected because rapamycin induces G1 cell cycle arrest in the presence of serum. Upon investigation, we have found that inactivated S6 kinase prevents the apoptotic effect observed by singular knockdown of eIF4E and results in G1 cell cycle arrest. This effect is dependent on TGF-β (transforming growth factor-β) signaling which contributes to G1 cell cycle arrest. Suppression of S6 kinase phosphorylation alone is insufficient to mediate cell cycle arrest, indicating that complete G1 cell cycle arrest is due to suppression of both S6 kinase and eIF4E. These data indicate that the cytostatic effect of rapamycin is suppression of both S6 kinase and eIF4E, while the cytotoxic effects are due suppression of eIF4E in the absence of S6 kinase-dependent activation of TGF-β signals. Our findings place an importance on the evaluating the activity/expression level of S6 kinase and eIF4E as readouts for rapamycin/rapalog efficacy.

Highlights

Rapamycin induces apoptosis in cancer cells by indirect inhibition of eIF4E through suppression of 4E-BP1 phosphorylation. ► The apoptotic effect of rapamycin occurs in the absence of serum – in the presence of serum, rapamycin induces G1 arrest. ► In contrast with rapamycin, suppression of eIF4E expression causes apoptosis in both the absence and presence of serum. ► In addition to suppressing 4E-BP1 phosphorylation, rapamycin suppresses phosphorylation of the mTORC1 substrate S6 kinase. ► Suppression of both S6 kinase and 4E-BP1 phosphorylation leads to G1 arrest in the presence of serum and prevents apoptosis.

Introduction

It is widely accepted that rapamycin suppresses phosphorylation of mTOR complex 1 (mTORC1) substrate S6 kinase at low nano-molar doses. Through an allosteric mechanism, rapamycin preferentially inhibits mammalian target of rapamcyin complex 1 (mTORC1) but also inhibits mTORC2 under certain conditions [1], [2]. We recently reported that high (micro-molar) doses of rapamycin also suppressed phosphorylation of another mTORC1 substrate, eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1). The high dose induced apoptosis in cancer cells in the absence of serum. We attributed cell death to indirect inhibition of the cap-dependent translation initiator, eIF4E, because de-phosphorylated 4E-BP1 binds eIF4E [3], [4]. Supportively, eIF4E ablation also caused apoptosis. This finding correlated with previous reports that the key mTORC1 substrate for cell proliferation and survival is eIF4E [5], [6]. In contrast, rapamycin induced G1 cell cycle arrest in the presence of serum that protected cells from progressing into S-phase [7], wherein the lack of eIF4E activity would otherwise cause apoptosis [7], [8]. G1 cell cycle arrest was dependent on TGF-β signaling [7] which is suppressed by mTOR [9], [10], Low-dose rapamycin concentrations sufficient to suppress phosphorylation of S6 kinase (but not 4E-BP1) induced TGF-β signaling [10]. While low-dose rapamycin treatment impeded G1 cell cycle progression, high-dose treatment was required to completely block progression from mitosis to S-phase [8]. In sum, rapamycin induces G1 cell cycle arrest at doses that effectively suppress the phosphorylation of both S6 kinase and 4E-BP1.

In this report, we investigated the role of eIF4E ablation on cell cycle arrest in the presence of serum. As aforementioned, inhibiting S6 kinase (low-dose rapamycin) was insufficient to mediate complete G1 arrest. Here, our results indicate similarly, eIF4E ablation is insufficient to induce cell cycle arrest and instead results in apoptotic cell death. Surprisingly, suppression of S6 kinase effectively prevented the apoptotic effect of eIF4E ablation. Dual inhibition of both proteins causes a TGF-β-dependent G1 cell cycle arrest. The findings reveal complex effects of rapamycin on mTORC1 substrates with clinical implications for using translational activators S6 kinase and eIF4E as readouts for clinical efficacy.

Section snippets

Cells, cell culture conditions and cell viability

The human cancer cell lines MDA-MB-231 and BT-549 cells were obtained from the American Tissue Type Culture Collection (ATCC) and cultured in Dulbecco’s Modified Eagle Medium (DMEM) (Sigma) supplemented with 10% Fetal Bovine Serum (Sigma). Cell viability was determined 24 h after treatment as described previously [8].

Antibodies and reagents

The following antibodies: PARP, cleaved PARP, P-S6 kinase T389, S6 kinase, and eIF4E were from Cell Signaling; α-actin was from Sigma. Negative control siRNA (Dharmacon), siRNAs

Ablation of eIF4E expression induces apoptosis in the presence of serum in MDA-MB-231 cells

Previously, we reported that high (20 μM) rapamycin doses induced apoptosis in MDA-MB-231 as well as in other cancer cell lines in the absence of serum [7], [8], [11], [12]. The apoptotic effect was due to complete dephosphorylation of 4E-BP1, which subsequently inactivated eIF4E [3], [4]. The indirect effect of rapamycin on eIF4E correlated with knockdown of eIF4E as this also induced apoptosis. Consistent with the effect of high dose rapamycin being due to suppression of eIF4E, knockdown of

Discussion

Previously, we reported that rapamycin caused G1 cell cycle arrest in the presence of serum [8]. We associated the effect of high-dose rapamycin on inhibition of 4E-BP1 phosphorylation with an indirect inhibition of eIF4E [8]. Here, we investigated the role of eIF4E in cell cycle arrest. Surprisingly, we observed cell death upon eIF4E knockdown in MDA-MB-231 breast cancer cells in both the presence and absence of serum. Since high-dose rapamycin treatment continues to suppress phosphorylation

Acknowledgements

This work was supported by a grant from the National Cancer Institute (CA46677). Research Centers in Minority Institutions (RCMI) award RR-03037 from the National Center for Research Resources of the National Institutes of Health, which supports infrastructure and instrumentation, is also acknowledged. PY was supported by a Gene Center Fellowship from the RCMI.

References (33)

  • A. Toschi et al.

    Regulation of mTORC1 and mTORC2 complex assembly by phosphatidic acid: competition with rapamycin

    Mol. Cell. Biol.

    (2009)
  • J.D. Richter et al.

    Regulation of cap-dependent translation by eIF4E inhibitory proteins

    Nature

    (2005)
  • R.J. Dowling et al.

    MTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs

    Science

    (2010)
  • N. Gadir et al.

    Defective TGF-β signaling sensitizes human cancer cells to rapamycin

    Oncogene

    (2008)
  • P. Yellen et al.

    High-dose rapamycin induces apoptosis in human cancer cells by dissociating mTOR complex1 and suppressing phosphorylation of 4E-BP1

    Cell Cycle

    (2011)
  • K. Song et al.

    Novel roles of Akt and mTOR in suppressing TGF-β/ALK5-mediated Smad3 activation

    EMBO J.

    (2006)
  • Cited by (16)

    • Rapamycin-induced G1 cell cycle arrest employs both TGF-β and Rb pathways

      2015, Cancer Letters
      Citation Excerpt :

      The phosphorylation of 4E-BP1 by mTORC1 results in the liberation of eIF4E, which can then facilitate cap-dependent translation of RNAs that encode proteins critical for cell cycle progression [17]. We demonstrated previously that suppressing expression of both S6K and eIF4E resulted in G1 cell cycle arrest [11]. To determine whether the phosphorylation of Rb stimulated by high dose rapamycin was dependent on the inhibition of 4E-BP1 phosphorylation, we investigated whether suppressing eIF4E expression would suppress Rb phosphorylation.

    • Reciprocal regulation of AMP-activated protein kinase and phospholipase D

      2015, Journal of Biological Chemistry
      Citation Excerpt :

      It is not clear how PLD and mTOR suppress AMPK; however, it was reported previously that rapamycin can stimulate the phosphorylation of AMPK at the LKB1 site at Thr-172 and of ACC at the AMPK site at Ser-79 (30). This can be achieved at low nanomolar concentrations of rapamycin that suppress S6 kinase phosphorylation but that have no effect on 4E-BP1 phosphorylation (37, 38), suggesting a role for S6 kinase. In contrast with cancer and other proliferating cells, AMPK stimulates glucose uptake when ATP levels are reduced (39).

    • Erythroid induction of K562 cells treated with mithramycin is associated with inhibition of raptor gene transcription and mammalian target of rapamycin complex 1 (mTORC1) functions

      2015, Pharmacological Research
      Citation Excerpt :

      The results are shown in Fig. 7A, and clearly sustain the concept that MTH inhibits the phosphorylation of p70S6 kinase. Fig. 7B shows the involvement of p70S6 kinase on the cell cycle [43,45], and demonstrates that MTH treatment of K562 cells leads to an increase of G1-phase cells, in association with a sharp decrease of the proportion of S-phase cells. This study investigated the possible effects of mithramycin, known to be a strong inducer of fetal hemoglobin in erythroid cells from normal and β-thalassmic patients, on the mTOR pathway.

    • MAP kinase-interacting kinases - Emerging targets against cancer

      2014, Chemistry and Biology
      Citation Excerpt :

      A similar effect was observed in UMSCC22B cells, where cell cycle progression was disturbed at a G1-S checkpoint when using eIF4E small interfering RNA (Oridate et al., 2005). In addition, knockdown of eIF4E has been shown to induce apoptotic cell death in MDA-MB-231 cells accompanied by cleavage of the caspase 3 substrate poly (adenosine diphosphate-ribose) polymerase (Yellen et al., 2013). Silencing eIF4E in NIH 3T3 cells significantly reduced the levels of antiapoptotic proteins, including BI-1, dad1, and survivin, which explained the proapoptotic effect of eIF4E depletion (Mamane et al., 2007).

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    2

    Present address: Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States.

    View full text