Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship

https://doi.org/10.1016/j.jbior.2019.03.003Get rights and content

Abstract

Activation of Mechanistic target of rapamycin (mTOR) signaling plays a crucial role in tumorigenesis of numerous malignancies including glioblastoma (GB). The Canonical PI3K/Akt/mTOR signaling cascade is commonly upregulated due to loss of the tumor suppressorm PTEN, a phosphatase that acts antagonistically to the kinase (PI3K) in conversion of PIP2 to PIP3. mTOR forms two multiprotein complexes, mTORC1 and mTORC2 which are composed of discrete protein binding partners to regulate cell growth, motility, and metabolism. These complexes are sensitive to distinct stimuli, as mTORC1 is sensitive to nutrients while mTORC2 is regulated via PI3K and growth factor signaling. The main function of mTORC1 is to regulate protein synthesis and cell growth through downstream molecules: 4E-BP1 (also called EIF4E-BP1) and S6K. On the other hand, mTORC2 is responsive to growth factor signaling by phosphorylating the C-terminal hydrophobic motif of some AGC kinases like Akt and SGK and it also plays a crucial role in maintenance of normal and cancer cells through its association with ribosomes, and is involved in cellular metabolic regulation. mTORC1 and mTORC2 regulate each other, as shown by the fact that Akt regulates PRAS40 phosphorylation, which disinhibits mTORC1 activity, while S6K regulates Sin1 to modulate mTORC2 activity.

Allosteric inhibitors of mTOR, rapamycin and rapalogs, remained ineffective in clinical trials of Glioblastoma (GB) patients, in part due to their incomplete inhibition of mTORC1 as well as unexpected activation of mTOR via the loss of negative feedback loops. In recent years, novel ATP binding inhibitors of mTORC1 and mTORC2 suppress mTORC1 activity completely by total dephosphorylation of its downstream substrate pS6KSer235/236, while effectively suppressing mTORC2 activity, as demonstrated by complete dephosphorylation of pAKTSer473. Furthermore by these novel combined mTORC1/mTORC2 inhibitors reduced the proliferation and self-renewal of GB cancer stem cells. However, a search of more effective way to target mTOR has generated a third generation inhibitor of mTOR, “Rapalink”, that bivalently combines rapamycin with an ATP-binding inhibitor, which effectively abolishes the mTORC1 activity. All in all, the effectiveness of inhibitors of mTOR complexes can be judged by their ability to suppress both mTORC1/mTORC2 and their ability to impede both cell proliferation and migration along with aberrant metabolic pathways.

Introduction

Mechanistic target of Rapamycin (mTOR; AKA Mammalian target of Rapamycin) is a large protein product of 289kDA and a product of a single gene localized to the chromosome 1p36.2. The Rapamycin was first discovered in the soil bacterium Streptomyces hygroscopicus present in soil samples from the Easter Island (also locally termed Rapa Nui) in the 1970s, and found to have potent antifungal activity. The target of rapamycin (TOR) was identified in the budding yeast Saccharomyces Cerevisiae in 1991 (Heitman et al., 1991). mTOR, a serine-threonine protein kinase, belongs to phosphoinositide 3-kinase-related kinase (PI3K) family with homologs in all eukaryotes (Laplante and Sabatini, 2012; Russell et al., 2011). Structurally, the N-terminus of mTOR contains several huntingtin elongation factor 3 protein phosphatase 2A TOR1 (HEAT) repeats to mediate the majority of interactions with other proteins. The C- terminus contains FKB-binding domain and a kinase domain that places it in the phosphatidylinositol-3-kinase (PI3K) family. The PI3K/Akt/mTOR signaling axis regulates various physiological functions such as cell cycle progression, transcription, mRNA translation, differentiation, apoptosis, autophagy, motility, and metabolism (Guertin and Sabatini, 2009; Jacinto et al., 2004; Laplante and Sabatini, 2012; Saxton and Sabatini, 2017). Hyperactivation of mTOR activity results in an increase in cell growth, and can cause some cell types to enter the cell cycle (Laplante and Sabatini, 2012). Constitutive activation of mTOR via single point mutations has been shown in many cancers including adenocarcinoma and renal cell carcinoma (Sato et al., 2010).

Aberrant PI3K/Akt signaling has been reported in many human cancers, which is a major cause of hyperactivation of the mTOR pathway contributing to both cancer pathogenesis as well as therapy resistance. Loss-of-function due to mutations in tumor suppressors, such as phosphatase and tensin homolog (PTEN), tuberous sclerosis 1/2 (TSC1/2), neurofibromin 1/2 (NF1/2), or oncogenic mutations in KRAS, PIK3CA, or AKT are the most common causes of mTOR signaling hyperactivity. Moreover, constitutive activation of the PI3K/Akt/mTOR signaling network is seen in patients with acute myeloid leukemia (AML) (Martelli et al., 2007). Additionally, studies suggest that tumor suppressors including PTEN and p53 may regulate stem cell populations by controlling self-renewal of cancer stem cells (Korkaya and Wicha, 2007).

It has been demonstrated that mTOR signaling is aberrantly regulated in Glioblastoma (GB), leading to abnormalities in protein synthesis, metabolism and motility, thereby resulting in uncontrolled growth and dissemination (Jhanwar-Uniyal et al., 2011, 2015a, 2015b). GB is WHO defined Grade IV astrocytoma, is the most common and aggressive CNS malignancy. Despite current treatment modalities survival rate remains dismal. As such, GB tumors are mostly PTEN and/or p53 deregulated (Ohgaki and Kleihues, 2009). Mutations of PTEN are found in approximately 70–90% of GB. Accordingly, there is up-regulation of the PI3K/Akt pathway in GB (Hay and Sonenberg, 2004; Phillips et al., 2006). The role of Akt in formation of gliomagenesis has been demonstrated in animal models and along with its downstream target mTOR, controls distinct cellular functions (Guertin et al., 2009; Holland, 2001). Because of overactivation of mTOR pathways, Rapamycin and its analogue, have been considered for the treatment of many cancers (under the trade name Sirolimus), as an FDA-approved immunosuppressant and chemotherapeutic agent (see Fig. 1).

Section snippets

Two complexes of mTOR: mTORC1 and mTORC2

mTOR forms two functionally distinct complexes in mammalian cells, namely mTOR complex 1 (mTORC1), which contains mTOR, Rapamycin-sensitive adapter protein of mTOR (Raptor), and LST8; and mTOR complex 2 (mTORC2), which is comprised of Rapamycin-insensitive companion of mTOR (Rictor), LST8, and Sin1 along with other proteins(Loewith et al., 2002). Activated mTORC1 regulates protein translation through activation of p70 S6 Kinase (p70 S6K), and inhibition of eukaryotic initiation factor 4E

Cellular localization of mTOR and its components

mTOR may function via nucleo-cytoplasmic signaling (Bachmann et al., 2006). We have previously revealed that PDGF stimulates the cellular localization of mTOR (Jhanwar-Uniyal et al., 2013). Many components of the mTOR pathway are expressed in both nuclear as well as cytoplasmic compartments. Many of the mTOR pathway proteins are localized to the nucleus because the major role of mTORC1 is in ribosome biogenesis or transcription. PI3K has been shown to be nuclear, while PDK1, Akt and PTEN

mTOR pathways in cancer

mTOR is often activated by mutations in its upstream regulators such as gain-of-function mutation of PI3K and loss-of-function mutation of the tumor suppressor gene PTEN (Alessi et al., 1997; Long et al., 2005; Thorpe et al., 2015; Yang et al., 2017; Zhang et al., 2017). Inhibition of mTORC1 signaling has been considered as an anticancer strategy. Rapamycin and its analogues, termed rapalogues, remained partial inhibitors of downstream effectors of the mTORC1, mainly, 4EBP and lead to

Summary

Since the discovery of mTOR, some four decades ago, our understanding the cellular and molecular mechanism of mTOR complexes have made unprecedented progress. Nevertheless, many aspects of their molecular and cellular regulation remains to be determined, for example not much is known about the regulation of mTORC1 via non-canonical regulation. We are just beginning to recognize the molecular mechanism of mTORC2 activation and its regulation besides PI3K. mTOR integrates extracellular growth

Conflict of interest

None.

Acknowledgment

Supported by funds from Advanced Research Foundation.

References (93)

  • M. Jhanwar-Uniyal et al.

    Deciphering the signaling pathways of cancer stem cells of glioblastoma multiforme: role of Akt/mTOR and MAPK pathways

    Adv. Enzym. Regul.

    (2011)
  • M. Jhanwar-Uniyal et al.

    Distinct signaling mechanisms of mTORC1 and mTORC2 in glioblastoma multiforme: a tale of two complexes

    Adv. Biol. Regulat.

    (2015)
  • M. Jhanwar-Uniyal et al.

    Deconstructing mTOR complexes in regulation of Glioblastoma Multiforme and its stem cells

    Adv. Biol. Regulat.

    (2013)
  • D.H. Kim et al.

    mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery

    Cell

    (2002)
  • J. Kim et al.

    mTOR as a central hub of nutrient signalling and cell growth

    Nat. Cell Biol.

    (2019)
  • N. Kubica et al.

    Activation of the mammalian target of rapamycin complex 1 is both necessary and sufficient to stimulate eukaryotic initiation factor 2Bvarepsilon mRNA translation and protein synthesis

    Int. J. Biochem. Cell Biol.

    (2008)
  • M. Laplante et al.

    mTOR signaling in growth control and disease

    Cell

    (2012)
  • R. Loewith et al.

    Two TOR complexes, only one of which is rapamycin sensitive, have distinct roles in cell growth control

    Mol. Cell

    (2002)
  • X. Long et al.

    Rheb binds and regulates the mTOR kinase

    Curr. Biol. : CB

    (2005)
  • H. Nojima et al.

    The mammalian target of rapamycin (mTOR) partner, raptor, binds the mTOR substrates p70 S6 kinase and 4E-BP1 through their TOR signaling (TOS) motif

    J. Biol. Chem.

    (2003)
  • H.S. Phillips et al.

    Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis

    Cancer Cell

    (2006)
  • A.R. Ramos et al.

    Phosphoinositide 5-phosphatase activities control cell motility in glioblastoma: two phosphoinositides PI(4,5)P2 and PI(3,4)P2 are involved

    Adv. Biol. Regulat.

    (2018)
  • P. Salomoni et al.

    The role of PML in tumor suppression

    Cell

    (2002)
  • J.N. Sarkaria et al.

    North Central Cancer Treatment Group Phase I trial N057K of everolimus (RAD001) and temozolomide in combination with radiation therapy in patients with newly diagnosed glioblastoma multiforme

    Int. J. Radiat. Oncol. Biol. Phys.

    (2011)
  • R.A. Saxton et al.

    mTOR signaling in growth, metabolism, and disease

    Cell

    (2017)
  • J. Tamburini et al.

    Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways

    Blood

    (2008)
  • L. Wang et al.

    PRAS40 regulates mTORC1 kinase activity by functioning as a direct inhibitor of substrate binding

    J. Biol. Chem.

    (2007)
  • J. Zhang et al.

    Inhibition of Rb phosphorylation leads to mTORC2-mediated activation of Akt

    Mol. Cell

    (2016)
  • L. Albert et al.

    Inhibition of mTOR activates the MAPK pathway in glioblastoma multiforme

    CANCER GENOMICS PROTEOMICS

    (2009)
  • S.M. Chang et al.

    Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme

    Investig. New Drugs

    (2005)
  • W.H. Chappell et al.

    Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health

    Oncotarget

    (2011)
  • M.G. Chheda et al.

    Vandetanib plus sirolimus in adults with recurrent glioblastoma: results of a phase I and dose expansion cohort study

    J. Neuro-Oncol.

    (2015)
  • C.M. Chresta et al.

    AZD8055 is a potent, selective, and orally bioavailable ATP- competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity

    Cancer Res.

    (2010)
  • T.F. Cloughesy et al.

    Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN- deficient glioblastoma

    PLoS Med.

    (2008)
  • F.A. Dick et al.

    Molecular mechanisms underlying RB protein function

    Nat. Rev. Mol. Cell Biol.

    (2013)
  • L. Doherty et al.

    Pilot study of the combination of EGFR and mTOR inhibitors in recurrent malignant gliomas

    Neurology

    (2006)
  • N.V. Dorrello et al.

    S6K1- and betaTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth

    Science (New York, N.Y.)

    (2006)
  • M. Ebner et al.

    Localization of mTORC2 activity inside cells

    J. Cell Biol.

    (2017)
  • V. Facchinetti et al.

    The mammalian target of rapamycin complex 2 controls folding and stability of Akt and protein kinase C

    EMBO J.

    (2008)
  • E. Galanis et al.

    Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a North central cancer treatment group study

    J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol.

    (2005)
  • J.M. Garcia-Martinez et al.

    mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1)

    Biochem. J.

    (2008)
  • A.P. Ghosh et al.

    Point mutations of the mTOR-RHEB pathway in renal cell carcinoma

    Oncotarget

    (2015)
  • B.C. Grabiner et al.

    A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity

    Cancer Discov.

    (2014)
  • D.A. Guertin et al.

    The pharmacology of mTOR inhibition

    Sci. Signal.

    (2009)
  • N. Gulati et al.

    Involvement of mTORC1 and mTORC2 in regulation of glioblastoma multiforme growth and motility

    Int. J. Oncol.

    (2009)
  • J.D. Hainsworth et al.

    Phase II study of concurrent radiation therapy, temozolomide, and bevacizumab followed by bevacizumab/everolimus as first-line treatment for patients with glioblastoma

    Clin. Adv. Hematol. Oncol. : HO (Hum. Organ.)

    (2012)
  • Cited by (180)

    View all citing articles on Scopus
    View full text