The inhibition of MAPK potentiates the anti-angiogenic efficacy of mTOR inhibitors

https://doi.org/10.1016/j.bbrc.2011.03.086Get rights and content

Abstract

The mammalian target of rapamycin (mTOR) which is part of two functionally distinct complexes, mTORC1 and mTORC2, plays an important role in vascular endothelial cells. Indeed, the inhibition of mTOR with an allosteric inhibitor such as rapamycin reduces the growth of endothelial cell in vitro and inhibits angiogenesis in vivo. Recent studies have shown that blocking mTOR results in the activation of other prosurvival signals such as Akt or MAPK which counteract the growth inhibitory properties of mTOR inhibitors. However, little is known about the interactions between mTOR and MAPK in endothelial cells and their relevance to angiogenesis. Here we found that blocking mTOR with ATP-competitive inhibitors of mTOR or with rapamycin induced the activation of the mitogen-activated protein kinase (MAPK) in endothelial cells. Downregulation of mTORC1 but not mTORC2 had similar effects showing that the inhibition of mTORC1 is responsible for the activation of MAPK. Treatment of endothelial cells with mTOR inhibitors in combination with MAPK inhibitors reduced endothelial cell survival, proliferation, migration and tube formation more significantly than either inhibition alone. Similarly, in a tumor xenograft model, the anti-angiogenic efficacy of mTOR inhibitors was enhanced by the pharmacological blockade of MAPK. Taken together these results show that blocking mTORC1 in endothelial cells activates MAPK and that a combined inhibition of MAPK and mTOR has additive anti-angiogenic effects. They also provide a rationale to target both mTOR and MAPK simultaneously in anti-angiogenic treatment.

Highlights

► Targeting mTOR in endothelial cell activates MAPK. ► Blocking MAPK enhances the anti-angiogenic effects of mTOR inhibitors. ► The anti-angiogenic efficacy of ATP-competitive inhibitors of mTOR is superior to that of rapamycin.

Introduction

Angiogenesis, the formation of new blood vessels from pre-existing ones, is a critical event in many physiological and pathological processes such as tumor growth and metastasis [1]. In fact, in order to grow, a tumor needs to develop new blood vessels once it reaches the dimension of 2 mm3 [2]. Angiogenesis is a multi-step process which involves endothelial cell proliferation, survival, and migration. Several molecules implicated in angiogenesis have been identified including growth factors (e.g. vascular endothelial growth factor, fibroblast growth factors and angiopoietins), integrins, notch receptors and their ligands, as well as molecules involved in mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathways [3], [4], [5]. Since angiogenesis plays a key role in tumor growth, targeting tumor angiogenesis represents a promising approach in cancer therapy. Accordingly, targeting the vascular endothelial growth factor (VEGF) has shown clinical efficacy and has been approved for the treatment of various cancers [6]. However, the benefits of anti-angiogenic therapies are transitory and most of the tumors eventually progress under therapy. Therefore, a strong need exists to design new therapeutic strategies that confer enduring anti-angiogenic effects.

mTOR is a key regulator of cell growth, proliferation, and survival as being part of two distinct complexes, mTORC1 and mTORC2. While mTORC1 is composed of five different components: mTOR, raptor, mLST8, PRAS40 and deptor, mTORC2 consists of mTOR, rictor, mSin1, mLST8, deptor and protor-1 [7]. mTORC1 phosphorylates, among others, S6K1 and 4E-BP1 resulting in the regulation of translation initiation and protein synthesis. mTORC2 phosphorylates Akt, SGK and PKC and is involved in cell proliferation, survival and cytoskeletal organization [8]. Numerous studies demonstrated that blocking mTOR with rapamycin reduces tumor angiogenesis [9]. Indeed, rapamycin inhibits the functions of endothelial cells relevant to angiogenesis in vitro and reduces angiogenesis in several models in vivo [10], [11], [12]. However, emerging evidence has shown that targeting mTOR also stops a negative feedback loop which results in the activation of proliferative signals such as Akt or MAPK that reduce the growth inhibitory properties of mTOR inhibitors [13]. The effect of mTOR inhibition on MAPK activity in endothelial cells as well as its relevance to angiogenesis has however not been determined. In this study, we evaluated the consequences of mTOR inhibition either by ATP-competitive inhibitors of mTOR or by rapamycin on MAPK activity in endothelial cells. We also explored the anti-angiogenic effects of mTOR inhibition in combination with MAPK inhibition both in vitro and in vivo.

Section snippets

Antibodies and chemicals

NVP-BEZ235, PP242, WYE-354, Ku-0063794 were from Chemdea. Rapamycin and UO126 were from LC laboratories. Antibodies directed against phospho-MAPK (Thr202, Tyr204), MAPK, phospho-Akt (S473), Akt, phospho-S6 ribosomal protein (Ser235/236), S6 ribosomal protein, raptor and rictor were from Cell Signaling.

Cell culture

Human umbilical vein endothelial cells (HUVEC) were purchased from Millipore and cultured in EndoGRO-VEGF complete medium (Millipore). HUVEC were used for the experiments between passages 2 and 5.

Cell transfection

ATP-competitive inhibitors of mTOR activate MAPK in endothelial cells

To assess the effects of mTOR inhibition on MAPK activity in endothelial cells, HUVEC were exposed to increasing concentrations of different ATP-competitive inhibitors of mTOR (Ku-0063794, WYE-354, PP242 or NVP-BEZ235) for 4 h and Western Blot analysis was performed on cell lysates. While Ku-0063794 [17], WYE-354 [18] and PP242 [19] specifically block mTOR activity, NVP-BEZ235 [20] also inhibits PI3K in addition to mTOR. We found that Ku-0063794, WYE-354 or PP242, blocked mTORC1 activity at 10 nM

Discussion

Despite encouraging experimental studies showing the anti-tumoral efficacy of rapamycin and rapamycin like drugs (rapalogs), the clinical benefit has been less successful than expected. Part of it might be explained by the identification of multiple crosstalks between mTOR signaling pathway and other pathways implicated in cell growth. It has been well described that blocking mTORC1 by rapamycin stops a negative feedback loop resulting in the activation of proliferative and pro-survival signals

Acknowledgments

This work was supported by a Research Grant of the Swiss National Science Foundation (SCORE 32323B-123821 to O.D.).

References (26)

  • R.H. Adams et al.

    Molecular regulation of angiogenesis and lymphangiogenesis

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • A.S. Chung et al.

    Targeting the tumor vasculature: insights from physiological angiogenesis

    Nat. Rev. Cancer

    (2010)
  • R. Zoncu et al.

    mTOR: from growth signal integration to cancer diabetes and ageing

    Nat. Rev. Mol. Cell Biol.

    (2011)
  • Cited by (25)

    • MTOR kinase inhibitors as potential cancer therapeutic drugs

      2013, Cancer Letters
      Citation Excerpt :

      Accordingly treatment of endothelial cells with Ku-0063794 in combination with U0126 reduced endothelial cell survival, proliferation, migration and tube formation more significantly than either inhibitor alone. Similarly, in a tumor xenograft model, the anti-angiogenic efficacy of TORKinibs was enhanced by the pharmacological blockade of MAPK [69]. The identification of TORKinibs not only provides us with valuable research tools for fully understanding the biological functions of mTORCs essential for regulation of cell proliferation and survival, but also promises the development of new and efficacious anticancer drugs.

    • Potent dual inhibitors of TORC1 and TORC2 complexes (KU-0063794 and KU-0068650) demonstrate in vitro and ex vivo anti-keloid scar activity

      2013, Journal of Investigative Dermatology
      Citation Excerpt :

      We hypothesized that dual mTORC1 and mTORC2 inhibition provides superior inhibition of Akt signaling and anti-angiogenic activity. Unlike Rapamycin, which inhibits mTORC1 alone (Benjamin et al., 2011), here we demonstrate that both KU-0063794 and KU-0068650 (referred to collectively as AstraZeneca (AZ) compounds) are highly selective adenosine triphosphate-competitive inhibitors (Garcia-Martinez et al., 2009) of mTOR kinase activity, with no toxicity in vivo (Malagu et al., 2009; Janes et al., 2010; Dormond-Meuwly et al., 2011), similar in mechanism of action to AZD8055 (Chresta et al., 2010; Sini et al., 2010). Therefore, we investigated (i) the baseline cellular levels of mTOR, p70S6K, and their activated forms between KD and extra-lesional tissue (ELT) obtained from the same patient, (ii) the effect of both AZ compounds on KD growth and ECM deposition in vitro and ex vivo, and (iii) differences between KU-0063794 and KU-0068650 to a well-recognized mTOR inhibitor Rapamycin.

    • DEPTOR regulates vascular endothelial cell activation & proinflammatory & angiogenic responses

      2013, Blood
      Citation Excerpt :

      Similar to other cell types,21,23-25 we find that DEPTOR expression in EC regulates mTORC1-dependent activation responses, but in these studies, we also identify an important regulatory role for DEPTOR in ERK1/2 activity. Although there is a well-established intracellular cross-talk among the mTOR signaling pathway (and intermediaries) and ERK1/2 signaling,41-43 we find that the treatment of ECs with pharmacological inhibitors of mTOR activity (either rapamycin or Torin1) fails to alter the effect of DEPTOR knockdown on the augmentation of ERK1/2 activity. Furthermore, we find that the treatment of ECs with a pharmacological inhibitor of MEK-ERK1/2 activity (U0126) fails to alter the effect of DEPTOR knockdown on mTORC1-induced signals.

    • Targeting the intragraft microenvironment and the development of chronic allograft rejection

      2012, Human Immunology
      Citation Excerpt :

      Compared to rapalogs, ATP-competitive inhibitors of mTOR target both mTORC1 and mTORC2. Initial studies have demonstrated that their ability to inhibit EC proliferation and survival are greater than raplogs [131], and they also inhibit VEGF production more effectively. These differences indicate that ATP-competitive inhibitors of mTOR have promise as novel therapeutics in the future.

    View all citing articles on Scopus
    View full text