Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2012 May;14(5):360–367. doi: 10.1593/neo.12532

Targeting the mTOR-DEPTOR Pathway by CRL E3 Ubiquitin Ligases: Therapeutic Application1

Yongchao Zhao 1, Yi Sun 1
PMCID: PMC3384423  PMID: 22745582

Abstract

The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine/threonine protein kinase, integrates both intracellular and extracellular signals and serves as a central regulator of cell metabolism, growth, proliferation, survival, and autophagy. The mTOR pathway is frequently activated in many human cancers, mainly resulting from alterations in the upstream regulators, such as phosphoinositide 3-kinase (PI3K)/AKT activation, PTEN loss or dysregulation of mTOR-negative regulators (e.g., TSC1/2), leading to uncontrolled proliferation. Thus, inhibiting the PI3K/AKT/mTOR pathways is widely considered as an effective approach for targeted cancer therapy. Recently, we and others found that DEPTOR, a naturally occurring inhibitor of both mTORC1 and mTORC2, was degraded by SCF (Skp1-Cullin-F box proteins) E3 ubiquitin ligase, the founding member of cullin-RING-ligases (CRLs), resulting in mTOR activation and cell proliferation. In addition to DEPTOR, previous studies have demonstrated that several other negative regulators of mTOR pathway are also substrates of CRL/SCF E3s. Thus, targeting CRL/SCF E3s is expected to cause the accumulation of these mTOR signal inhibitors to effectively block the mTOR pathway. In this review, we will discuss mTOR signaling pathway, how DEPTOR regulates mTOR/AKT axis, thus acting as a tumor suppressor or oncogene in some cases, how DEPTOR is ubiquitinated and degraded by SCFβ-TrCP E3, and how MLN4924, a small-molecule indirect inhibitor of CRL/SCF E3 ligases through blocking cullin neddylation, might be useful as a novel approach of mTOR pathway targeting for cancer therapy.

The mTOR and mTOR Signaling Pathway: An Introduction

The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine/threonine protein kinase, integrates both intracellular and extracellular signals and serves as a central regulator of cell metabolism, growth, proliferation, survival, and autophagy [1,2]. In mammalian cells, mTOR forms two structurally and functionally distinct complexes, namely mTORC1 and mTORC2 [3,4]. Whereas mTORC1 consists of mTOR, Raptor, mLST8/GβL, PRAS40, and DEPTOR, mTORC2 is composed of mTOR, Rictor, GβL, Protor, Sin1, and DEPTOR [1,2]. It is well known that mTORC1 mainly promotes protein translation and cell growth by phosphorylating S6K1 and 4E-BP1, whereas mTORC2 regulates cytoskeletal organization [5] as well as cell survival through directly phosphorylating and activating AKT [3,4]. In addition, as a negative feedback loop, mTORC1 through S6K inhibits AKT by downregulating the expression of IRS-1/2 to block phosphoinositide 3-kinase (PI3K) activation [3,4]. Furthermore, mTORs are well-established negative regulators of autophagy [6,7]; whereas mTORC1 inhibits autophagosome formation by phosphorylating Ulk1/2 and Atg13 [8,9], mTORC2 represses the expression of some autophagy-related genes and other autophagy regulators [10–13]. Likewise, rapamycin, a potent inhibitor of mTOR, is commonly used as an autophagy inducer [6,14].

In response to multiple signals, including growth factors, nutrients, energy, and stress, mTOR is activated or inactivated, resulting in altered cellular processes [1,5,15]. On exposure to growth factors, mTOR is activated through 1) the PI3K-AKT pathway, leading to inactivation of TSC2, a negative regulator of mTOR [16], which complexes with TSC1 to inactivate Rheb GTPase, an mTORC1 activator [16–18]; and 2) the RAS-ERK-RSK pathway, which also phosphorylates and inhibits TSC2 [19,20]. We recently showed that RSK1 could directly target the mTOR complex by phosphorylating DEPTOR, an mTOR inhibitor [21], thereby promoting its degradation [22] (see below). In response to hypoxia, HIF-1 is induced to block mTORC1 through the HIF-1-REDD1 axis, which activates the TSC1–TSC2 complex through less well-defined mechanisms or a mechanism involving 14-3-3 [23,24]. Furthermore, TSC2 is activated by the cellular energy sensor AMP-activated protein kinase (AMPK). When the ratio of AMP/ATP is increased, AMPK is activated to phosphorylate and activate TSC2, leading to mTORC1 inactivation [25]. AMPK also inhibits mTOR by directly phosphorylating and inactivating Raptor in a TSC2-independent manner [26]. By phosphorylating and activating AMPK, the tumor suppressor LKB1 also significantly abrogates mTOR signaling [27]. Finally, mTORC1 can be activated by amino acids through Rag GTPases [28,29]. However, the upstream activators of mTORC2 are ill defined, but it is generally thought that mTORC2 is activated directly or indirectly only by growth factors [2]. Figure 1 briefly summarizes the signaling pathways that respond to various stimuli, leading to mTOR activation and subsequent cell growth and proliferation, survival, and autophagy inhibition.

Figure 1.

Figure 1

The mTOR signaling pathway. Arrows stand for activation; bars represent inhibition. graphic file with name neo1405_0360_fig005.jpg represents phosphorylation. For details, see text.

DEPTOR, an mTOR Inhibitor: A Tumor Suppressor or an Oncogene?

Recently, Sabatini's group discovered a novel mTOR binding partner, designated as DEPTOR in reference to the presence of two tandem N-terminal DEP (disheveled, egl-10, pleckstrin) domains with unknown function [30] and its specific interaction with mTOR [21]. DEPTOR also contains a C-terminal PDZ (postsynaptic density 95, discslarge, zonula occludens-1) domain [31], which is often involved in protein-protein interaction. Indeed, DEPTOR binds to FAT domain on mTOR through its PDZ domain [21]. Significantly, DEPTOR binds to both mTORC1 and mTORC2 and inhibits their activities, as measured by in vitro kinase assay as well as in cell-based assays. Nevertheless, the messenger RNA (mRNA) and protein levels of DEPTOR are negatively regulated by the activities of both mTORC1 and mTORC2, establishing a regulatory feedback loop [21].

By blocking the activity of mTOR, DEPTOR acts, in general, as a tumor suppressor through inhibiting protein synthesis, cell proliferation, and survival. Indeed, small interfering RNA knockdown of DEPTOR increased the activities of both mTORC1 and mTORC2, as evidenced by increased S6K1/AKT phosphorylation, and thereby promoting cell proliferation and survival [1,21] (Figure 2A). However, under certain circumstances, DEPTOR could act as an oncogene [21,32]. High levels of DEPTOR inhibited mTORC1 and activated AKT by relieving the feedback inhibition from S6K1 to PI3K, as demonstrated by reduced S6K1 phosphorylation and increased AKT phosphorylation, thus promoting the survival of cancer cells [21,22] (Figure 2B). It is not surprising that the levels of DEPTOR are downregulated inmost cancers because the mTOR pathway negatively regulates DEPTOR mRNA expression and protein stability [21]. In a subset (∼28%) of human multiple myelomas, however, DEPTOR was overexpressed. In these cells, high levels of DEPTOR were required for the maintenance of PI3K/AKT activation and small interfering RNA knockdown of DEPTOR induced apoptotic cell death [21]. Consistently, increased DEPTOR rendered cancer cells more resistant to chemotherapeutic drugs [22,33,34]. Finally, DEPTOR overexpression was found in hepatocellular carcinoma and differentiated thyroid carcinoma, which correlated with the poor prognosis of patients [35,36]. These conflicting data suggest a complex role of DEPTOR in the regulation of mTOR signaling pathways as well as other yet-to-be-identified pathways in a tissue-specific and cell context-dependent manner. The in vivo physiological role of DEPTOR as a tumor suppressor or oncogene will only be clearly demonstrated in mouse knockout studies by determining the effect on tumorigenesis on Deptor deletion (total or tissue specific) alone or in combination with, for example, Ras activation or Pten loss.

Figure 2.

Figure 2

DEPTOR has tumor suppressive and oncogenic properties. (A) DEPTOR acts as a tumor suppressor. (B) DEPTOR acts as an oncogene. For details, see text.

DEPTOR Ubiquitination and Degradation by SCFβ-TrCP E3 Ubiquitin Ligase

Although it was established that mTOR promotes DEPTOR degradation on mitogen stimulation [21], the E3 ubiquitin ligase, responsible for DEPTOR degradation, was unknown until most recently. Three independent groups, including ours, identified SCFβ-TrCP E3 ubiquitin ligase is the E3 that promotes the ubiquitination and degradation of DEPTOR in response to serum stimulation [22,33,37].

Cullin-RING ligases (CRLs) with the founding member, SCF E3, are the largest E3 ubiquitin ligase family. CRLs seem to account for ∼20% of total protein degradation by the ubiquitin-proteasome system [38]. By promoting ubiquitination and degradation of a variety of key substrates, CLRs control a number of important biologic processes including cell cycle progression, signal transduction, gene transcription, embryonic development, genomic integrity, and tumor suppression [39–41]. In mammalian cells, there are a total of eight members of the cullin family of proteins (CUL1, 2, 3, 4A, 4B, 5, 7, and PARC) [42], which individually binds to an adaptor protein (such as SKP1 and DDB1) and a substrate receptor protein (such as F-box protein) at the N-terminus, and a RING protein, RBX1 or RBX2 (also known as ROC2 or SAG) at the C-terminus [43]. It is well established that the substrate specificity of CRLs/SCF E3s is determined by substrate receptor proteins, such as F-box proteins [44], whereas the core ligase activity is possessed by the cullins-RBX1/2 complex [45].

Now how does SCFβ-TrCP promote DEPTOR ubiquitination and degradation? It is well established that in order for F-box proteins to bind to its substrates for subsequent ubiquitination and degradation, the substrates have to be phosphorylated first at the serine or threonine residue(s) on a consensus-binding motif of a given F-box protein [39]. In the case of DEPTOR, we did identify an evolutionarily conserved consensus β-TrCP binding motif (DSGXXS) on residues 286 to 291 (SSGYFS). The only mismatch here is the replacement of the conserved aspartic acid (D) by a serine (S) (see Figure S1A in Zhao et al. [22]), which, nevertheless, behaves as aspartic acid on phosphorylation. The exact same sequence of the binding motif was identified on BimEL, a previously characterized substrate of SCFβ-TrCP [46]. We further identified that kinases S6K1, a downstream target of mTORC1, and RSK1 are responsible for DEPTOR phosphorylation at the S286-S287-S291 residues, with S6K1 acting as a prime kinase, although which kinase actually phosphorylates which serine residue remains to be defined [22,47]. Conversely, two other groups reported that mTOR and CK1α are the responsible kinases with mTOR acting as a prime kinase to facilitate DEPTOR phosphorylation on three serine residues at the binding motif (286SSGYFS291) by CK1α [33,37]. To further clarify this discrepancy, we performed an in vitro kinase assay for DEPTOR phosphorylation on these three serine residues (Ser286,287,291), using a specific phosphor antibody raised against a peptide with all three serine residues phosphorylated [22]. As shown in Figure 3A, immune-purified FLAG-tagged DEPTOR was phosphorylated by mTOR (slightly) and CK1α alone or in combination or by the combination of S6K1 and RSK1. Thus, the combination of either mTOR/CK1α or S6K1/RSK1 was able to phosphorylate this binding motif in vitro. The in vivo DEPTOR phosphorylation for its targeted degradation, however, will likely be triggered by a different set of kinases activated in response to a given stimulus. Taken together, these combined results indicate that mTOR positively regulates its own activity by inducing the ubiquitination and degradation of DEPTOR, one of its own endogenous inhibitors, either directly or through its downstream target S6K1 [22,33,37,47]. On phosphorylation on β-TrCP binding motif, DEPTOR is bound to β-TrCP, which recruits E2-loaded RBX1-Cullin 1 complex, catalyzing ubiquitin transfer from E2 to DEPTOR. Multiple runs of this E1/E2/E3-mediated chain reaction cause DEPTOR polyubiquitination, which is then recognized by the 26S proteasome for subsequent degradation (Figure 3B) [22,33,37].

Figure 3.

Figure 3

DEPTOR is ubiquitinated by SCFβ-TrCP E3 ligase on phosphorylation. (A) In vitro phosphorylation of DEPTOR on the consensus-binding motif. FLAG-tagged DEPTOR was transfected into 293 cells, purified by IP using bead-conjugated anti-FLAG Ab, and incubated with active CK1 or mTOR alone or in combination, as well as the combination of active S6K1 and RSK1 in a kinase reaction mixture. DEPTOR phosphorylation on Ser286,287,291 was detected by immunoblotting using a DEPTOR phosphor-specific Ab [22]. (B) Schematic summary of DEPTOR ubiquitination by SCFβ-TrCP E3 ubiquitin ligase and degradation by 26S proteasome. In response to mitogen, RSK1 and S6K1 are activated to phosphorylate DEPTOR at β-TrCP degron site [22]. Conversely, DEPTOR is phosphorylated by mTOR at non-degron site, which acts as primer for subsequent phosphorylation at degron site by CK1α [33,37]. Phosphorylated DEPTOR is then recognized by β-TrCP for subsequent ubiquitination by SCF E3 and degradation by proteasome.

CRLs/SCF E3 Ligases Degrade Multiple Components of mTOR Pathway

In addition to DEPTOR, a variety of components, both upstream and downstream, of mTOR signaling pathway were found to be the substrates of CRLs/SCF E3 ligases (Figure 4A). First of all, mTOR itself was reported to undergo ubiquitin-mediated degradation by Cul1-Skp1-Fbw7 E3 ligase, although the kinase(s) responsible for prerequisite phosphorylation was not defined [48]. Second, TSC2 was identified as the substrate of Cul4-DDB1-Fbw5 E3 ligase that mediated TSC2 protein stability and TSC complex turnover [49]. Third, REDD1 was rapidly degraded by Cul4A-DDB1-β-TrCP E3 ligase with GSK-3β as a corresponding kinase for prerequisite phosphorylation [50]. Fourth, HIF-1α was a well-characterized substrate of Cul2-Elongin BC-VHL E3 ligase [51,52]. Fifth, IRS1 can be ubiquitinated and degraded either by Cul7-Skp1-Fbw8 in a manner dependent of mTOR and S6K [53] or by Cul1-Skp1-Fbxo40 E3 ligase with ligase activity enhanced by increased tyrosine phosphorylation of IRS1 [54]. Sixth, PHLPP1, a protein phosphatase that dephosphorylates activated AKT, was found to be substrate of SCFβ-TrCP on phosphorylation by CK1 and GSK-3β [55]. Finally, PDCD4, a downstream target of mTOR/S6K1, was identified as a substrate of Cul1-Skp1-β-TrCP E3 ligase on prerequisite phosphorylation at Ser67 by S6K1 [56]. Thus, given the fact that all these CRLs E3 substrates, except mTOR itself and IRS1, are the negative regulators of mTOR pathway, the small-molecule inhibitor of CRLs/SCF E3 ligases would likely cause the accumulation of these negative regulators to shut down mTOR pathways, leading to suppression of cell proliferation and survival.

Figure 4.

Figure 4

CRLs/SCF E3 ubiquitin ligases regulate mTOR pathway. (A) Schematic summary of CRLs/SCF E3 substrates in mTOR signaling pathway. (B) Schematic summary of cullin neddylation. (C) Schematic summary of ubiquitination reaction catalyzed by SCF E3 ubiquitin ligases and potential targeting points for inhibition. For details, see text.

Targeting mTOR Pathway by Inactivating CRL/SCF E3s: Therapeutic Application

The mTOR pathway is frequently and constitutively activated in many types of human cancer mainly through activation of PI3K/AKT and inactivation of PTEN in the upstream signaling pathways [3,5,57,58]. The PI3K/AKT signaling is activated through a variety of mechanisms, including mutations or amplifications of cell surface receptor tyrosine kinases (RTKs), such as EGFR [59–61] and IGFR [62,63], the gain-of-function mutations in the PIK3CA gene itself, which encodes the p110a catalytic subunit of PI3K [64–66], and loss of expression of the tumor suppressor PTEN, which encodes a lipid and protein phosphatase [67]. PTEN inactivation is found in many cancers, such as carcinomas of breast, endometrium, thyroid, and prostate as well as melanoma, leukemia, lymphoma, and glioblastoma, through several mechanisms, including mutations, deletions, loss of heterozygosity, methylation, and microRNA silencing [67]. Furthermore, mTOR activation was also found in several benign tumor syndromes, including tuberous sclerosis (TSC1/2 mutations) [25], Peutz-Jeghers syndrome (LKB1 mutations) [27], Cowden syndrome (PTEN mutations) [68], and neurofibromatosis (NF1 mutations) [69]. Thus, PI3K/AKT/mTOR pathways are considered as attractive targets for effective cancer treatment [64,70–74].

Given the association of mTOR with a wide variety of diseases including cancer, metabolic disorders, neurologic diseases, and autoimmune diseases [2], mTOR inhibitors may therefore have a broad application in the treatment of these diseases. Rapamycin (sirolimus) and its analogs (rapalogs), which include temsirolimus (CCI-779) [75], everoliums (RAD001) [76], and deforolimus (AP23573) [77], are the first generation of mTOR inhibitors [73], which form a complex with the immunophilin FKBP12 and inhibit mTORC1 activity through binding to FRB (FKBP12-rapamycin-binding) domain on mTOR [78]. Numerous clinical trials are ongoing to test their efficacy as anticancer agents, but with limited success [73,79,80]. The cancer resistance to rapamycin type of drugs could be attributable to the existence of the negative feedback mechanism by which inactivation of mTORC1/S6K1 releases S6K1 inhibition on IRS-PI3K, leading to activation of AKT for cancer cell survival [73]. Thus, inhibition of mTORC1 reduces cell growth but could improve the survival [81]. Recently, ATP-competitive inhibitors, such as Torin1 [82], PP242 and PP30 [83], which directly target the catalytic site of mTOR kinase and inhibit both mTORC1 and mTORC2 kinase activity, were developed as the second generation of mTOR inhibitors, currently in early stage of clinical trials with a better anticancer efficacy than rapalogs in preclinical settings [80,84].

An alternative approach of blocking mTOR or mTOR pathway, which has never been tested before, would be through modulating the stability of mTOR-negative regulators. Given the fact that most mTOR signal blockers, such as DEPTOR, HIF-1α, REDD1, TSC2, and PDCD4, are substrates of CRLs/SCF E3 ligases, small-molecule inhibitors of the CRL/SCF E3s are expected to cause their accumulation, thus abrogating mTOR pathway, particularly in human cancers with constitutive mTOR activation. Unfortunately, specific small-molecule inhibitor against the multicomponent CRL/SCF E3 ligases has not been discovered to test this hypothesis. However, MLN4924, a newly discovered small-molecule inhibitor of NEDD8-activating Enzyme (NAE) [38], could act as an indirect inhibitor of CRL/SCF E3s, thus to be used as an alternative option.

Like ubiquitination, neddylation requires E1 NEDD8-activating enzyme, E2 NEDD8-conjugating enzyme (UBC12), and E3 NEDD8 ligase to catalyze the transfer of NEDD8 to a target molecule [85,86] (Figure 4B). Inhibition of either enzyme in the reaction would abrogate substrate neddylation. MLN4924 is a newly discovered small-molecule inhibitor of NAE [38]. MLN4924 binds to NAE to create a covalent NEDD8-MLN4924 adduct, which cannot be further used in subsequent intraenzyme reactions, thus blocking NAE enzymatic activity [87] (Figure 4B). Because the activity of CRL/SCF E3s requires cullins neddylation, which disrupts inhibitory binding by CAND1 and retains the CRLs/SCFs in an active conformation [88–91], MLN4924 became the first-in-class indirect inhibitor of CRL/SCF E3s [38]. By inactivating CRL/SCF E3s, MLN4924 causes the accumulation of a number of SCF E3 substrates to inducing apoptosis [38,92–95] and senescence [96–98], thus inhibiting tumor growth both in vitro and in vivo. We used MLN4924 as an alternative mTOR pathway inhibitor and found that it effectively blocked the activity of mTORC1 in both time- and dose-dependent manners in almost all tested human cancer cell lines derived from carcinomas of breast, prostate, colon, brain, and cervix (unpublished observations). Thus, antitumor activity of MLN4924 could be attributable to its inhibition of the mTOR pathway. It is worth noting that among these reported substrates of CRL/SCF E3s in the mTOR pathway, HIF-1α, DEPTOR, and PDCD4 were most frequently accumulated, whereas mTOR itself, TSC2, and REDD1 were never accumulated on MLN4924 treatment in all tested cell lines under our assay conditions (unpublished observations). The differential MLN4924 responses, in substrate accumulation among tested cancer lines, are likely attributable to cell line- and cell context-dependent availability of F-box/adaptor proteins as well as of kinases prerequisite for substrate phosphorylation.

It should be clearly noted that, although MLN4924 demonstrated a universal inhibitory activity against mTORC1, it suffers from a major specificity issue as an mTOR pathway blocker. First of all, there are thousands of CRL/SCF substrates, given the fact that CRLs can be assembled into more than 200 different E3 enzymes by different combinations of its multiple components [39,99]. Apparently, some of the substrates are oncogenes, whereas others are tumor suppressors [40,100]. The net biologic outcome of MLN4924 action (such as induction of apoptosis, senescence, or even autophagy; unpublished observation) would likely depend on the interaction of these substrates in a cell context-, temporal-, and spatial-dependent manner. Second, MLN4924 is an NAE inhibitor and would likely inhibit other cellular neddylation reactions (much to be learned) [85,86], although cullins are the only physiological substrates [85,101]. Third, it has been recently reported that cancer cells could develop the resistance to MLN4924 by selection of rare clones with heterozygous mutations in targeting enzyme NAEβ [102,103]. Nevertheless, MLN4924 is the first-in-class and only indirect inhibitor of SCF E3 ligases, which demonstrated potent anticancer activity with limited toxicity in mice in preclinical settings [38,104] and has been advanced to several phase 1 clinical trials [105,106].

Future Perspectives on DEPTOR Biology and CLR/SCF E3 Inhibitors

Although activation of PI3K-AKT-mTOR pathway, resulting from PI3K/AKT mutation and/or PTEN loss, plays a key role in promoting cell proliferation/survival and tumorigenesis in many human cancers [64,67,70,107], it is totally unknown how this oncogenic pathway is regulated by DEPTOR under in vivo physiological settings, particularly given the fact that DEPTOR could act as a tumor suppressor or oncogene in cell culture settings [1,21]. Several DEPTOR biologic questions remain to be addressed: 1) Is Deptor a tumor suppressor or an oncogene in vivo when acting alone? 2) Can Deptor affect tumorigenesis triggered by activation of PI3K-AKT-mTOR axis as a result of Ras activation or Pten loss? 3) Can targeting DEPTOR degradation suppress tumorigenesis in vivo? The answers to these fundamental questions by the use of genetically modified mouse models and human tumor xenograft models would certainly further our current understanding of DEPTOR function and its regulation of PI3K-AKT-mTOR axis, and provide molecular basis for rational drug design in targeting this oncogenic axis.

With regard to specific inhibitors of CRL/SCF E3s, which have been validated as the attractive anticancer targets [108,109], encouraging progresses are being made. Examples include the discovery of Cpd A that disrupted Skp2-p27 binding to abrogate p27 degradation [110] and discovery of few SCF-Cdc4 inhibitors [111,112] and a Cdc34 inhibitor [113]. Other options include 1) disruption of E2 binding with RING proteins, RBX1/RBX2; 2) disruption of receptor binding with tumor suppressive substrates; and 3) inhibition of kinases that phosphorylate tumor suppressor substrates for targeted degradation (Figure 4C). It is hoped that specific inhibitors targeting CRL/SCF E3s will be eventually discovered and further developed as novel anticancer agents for the treatment of human cancers with activated CRL/SCF E3s.

Footnotes

1

This work was supported by the National Cancer Institute grants (CA118762 and CA156744) to Y.S.

References

  • 1.Efeyan A, Sabatini DM. mTOR and cancer: many loops in one pathway. Curr Opin Cell Biol. 2010;22:169–176. doi: 10.1016/j.ceb.2009.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22. doi: 10.1016/j.ccr.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 4.Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–734. doi: 10.1038/nrc1974. [DOI] [PubMed] [Google Scholar]
  • 5.Corradetti MN, Guan KL. Upstream of the mammalian target of rapamycin: do all roads pass through mTOR? Oncogene. 2006;25:6347–6360. doi: 10.1038/sj.onc.1209885. [DOI] [PubMed] [Google Scholar]
  • 6.Jung CH, Ro SH, Cao J, Otto NM, Kim DH. mTOR regulation of autophagy. FEBS Lett. 2010;584:1287–1295. doi: 10.1016/j.febslet.2010.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science. 2004;306:990–995. doi: 10.1126/science.1099993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Alers S, Loffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol. 2012;32:2–11. doi: 10.1128/MCB.06159-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yang ZF, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22:124–131. doi: 10.1016/j.ceb.2009.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Cardenas ME, Cutler NS, Lorenz MC, Di Como CJ, Heitman J. The TOR signaling cascade regulates gene expression in response to nutrients. Genes Dev. 1999;13:3271–3279. doi: 10.1101/gad.13.24.3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, Moffat J, Brown M, Fitzgerald KJ, Sabatini DM. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCα, but not S6K1. Dev Cell. 2006;11:859–871. doi: 10.1016/j.devcel.2006.10.007. [DOI] [PubMed] [Google Scholar]
  • 12.Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF, Tavare S, Arakawa S, Shimizu S, Watt FM. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009;23:798–803. doi: 10.1101/gad.519709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhao J, Brault JJ, Schild A, Goldberg AL. Coordinate activation of autophagy and the proteasome pathway by FoxO transcription factor. Autophagy. 2008;4:378–380. doi: 10.4161/auto.5633. [DOI] [PubMed] [Google Scholar]
  • 14.Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726–734. doi: 10.1038/nrc1692. [DOI] [PubMed] [Google Scholar]
  • 15.Sarbassov DD, Ali SM, Sabatini DM. Growing roles for the mTOR pathway. Curr Opin Cell Biol. 2005;17:596–603. doi: 10.1016/j.ceb.2005.09.009. [DOI] [PubMed] [Google Scholar]
  • 16.Inoki K, Li Y, Zhu TQ, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002;4:648–657. doi: 10.1038/ncb839. [DOI] [PubMed] [Google Scholar]
  • 17.Potter CJ, Pedraza LG, Xu T. Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol. 2002;4:658–665. doi: 10.1038/ncb840. [DOI] [PubMed] [Google Scholar]
  • 18.Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/Akt pathway. Mol Cell. 2002;10:151–162. doi: 10.1016/s1097-2765(02)00568-3. [DOI] [PubMed] [Google Scholar]
  • 19.Roux PP, Ballif BA, Anjum R, Gygi SP, Blenis J. Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci USA. 2004;101:13489–13494. doi: 10.1073/pnas.0405659101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ma L, Chen ZB, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosis and cancer pathogenesis. Cell. 2005;121:179–193. doi: 10.1016/j.cell.2005.02.031. [DOI] [PubMed] [Google Scholar]
  • 21.Peterson TR, Laplante M, Thoreen CC, Sancak Y, Kang SA, Kuehl WM, Gray NS, Sabatini DM. DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell. 2009;137:873–886. doi: 10.1016/j.cell.2009.03.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhao Y, Xiong X, Sun Y. DEPTOR, an mTOR inhibitor, is a physiological substrate of SCFβTrCP E3 ubiquitin ligase and regulates survival and autophagy. Mol Cell. 2011;44:304–316. doi: 10.1016/j.molcel.2011.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sofer A, Lei K, Johannessen CM, Ellisen LW. Regulation of mTOR and cell growth in response to energy stress by REDD1. Mol Cell Biol. 2005;25:5834–5845. doi: 10.1128/MCB.25.14.5834-5845.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.DeYoung MP, Horak P, Sofer A, Sgroi D, Ellisen LW. Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling. Genes Dev. 2008;22:239–251. doi: 10.1101/gad.1617608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Inoki K, Zhu TQ, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003;115:577–590. doi: 10.1016/s0092-8674(03)00929-2. [DOI] [PubMed] [Google Scholar]
  • 26.Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008;30:214–226. doi: 10.1016/j.molcel.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shaw RJ, Bardeesy N, Manning BD, Lopez L, Kasmatka M, DePinho RA, Cantley LC. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell. 2004;6:91–99. doi: 10.1016/j.ccr.2004.06.007. [DOI] [PubMed] [Google Scholar]
  • 28.Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science. 2008;320:1496–1501. doi: 10.1126/science.1157535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan K-L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat Cell Biol. 2008;10:935–945. doi: 10.1038/ncb1753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen S, Hamm HE. DEP domains: more than just membrane anchors. Dev Cell. 2006;11:436–438. doi: 10.1016/j.devcel.2006.09.011. [DOI] [PubMed] [Google Scholar]
  • 31.Jemth P, Gianni S. PDZ domains: folding and binding. Biochemistry. 2007;46:8701–8708. doi: 10.1021/bi7008618. [DOI] [PubMed] [Google Scholar]
  • 32.Proud CG. Dynamic balancing: DEPTOR tips the scales. J Mol Cell Biol. 2009;1:61–63. doi: 10.1093/jmcb/mjp012. [DOI] [PubMed] [Google Scholar]
  • 33.Gao D, Inuzuka H, Tan M-KM, Fukushima H, Locasale JW, Liu P, Wan L, Zhai B, Chin YR, Shaik S, et al. mTOR drives its own activation via SCFβTrCP-dependent degradation of the mTOR inhibitor DEPTOR. Mol Cell. 2011;44:290–303. doi: 10.1016/j.molcel.2011.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Foster H, Coley HM, Goumenou A, Pados G, Harvey A, Karteris E. Differential expression of mTOR signalling components in drug resistance in ovarian cancer. Anticancer Res. 2010;30:3529–3534. [PubMed] [Google Scholar]
  • 35.Pei L, Xie PF, Zhou EX, Yang Q, Luo Y, Tang ZH. Overexpression of DEP domain containing mTOR-interacting protein correlates with poor prognosis in differentiated thyroid carcinoma. Mol Med Report. 2011;4:817–823. doi: 10.3892/mmr.2011.503. [DOI] [PubMed] [Google Scholar]
  • 36.Yen CH, Lu YC, Li CH, Lee CM, Chen CY, Cheng MY, Huang SF, Chen KF, Cheng AL, Liao LY, et al. Functional characterization of glycine N-methyltransferase and its interactive protein-DEPDC6/DEPTOR in hepatocellular carcinoma. Mol Med. 2012;18:286–296. doi: 10.2119/molmed.2011.00331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Duan S, Skaar JR, Kuchay S, Toschi A, Kanarek N, Ben-Neriah Y, Pagano M. mTOR generates an auto-amplification loop by triggering the βTrCP- and CK1α-dependent degradation of DEPTOR. Mol Cell. 2011;44:317–324. doi: 10.1016/j.molcel.2011.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Soucy TA, Smith PG, Milhollen MA, Berger AJ, Gavin JM, Adhikari S, Brownell JE, Burke KE, Cardin DP, Critchley S, et al. An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer. Nature. 2009;458:732–736. doi: 10.1038/nature07884. [DOI] [PubMed] [Google Scholar]
  • 39.Deshaies RJ, Joazeiro CAP. RING domain E3 ubiquitin ligases. Annu Rev Biochem. 2009;78:399–434. doi: 10.1146/annurev.biochem.78.101807.093809. [DOI] [PubMed] [Google Scholar]
  • 40.Nakayama KI, Nakayama K. Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer. 2006;6:369–381. doi: 10.1038/nrc1881. [DOI] [PubMed] [Google Scholar]
  • 41.Petroski MD, Deshaies RJ. Function and regulation of cullin-RING ubiquitin ligases. Nat Rev Mol Cell Biol. 2005;6:9–20. doi: 10.1038/nrm1547. [DOI] [PubMed] [Google Scholar]
  • 42.Sarikas A, Hartmann T, Pan ZQ. The cullin protein family. Genome Biol. 2011;12:220. doi: 10.1186/gb-2011-12-4-220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zheng N, Schulman BA, Song LZ, Miller JJ, Jeffrey PD, Wang P, Chu C, Koepp DM, Elledge SJ, Pagano M, et al. Structure of the Cul1-Rbx1-Skp1-F box(Skp2) SCF ubiquitin ligase complex. Nature. 2002;416:703–709. doi: 10.1038/416703a. [DOI] [PubMed] [Google Scholar]
  • 44.Jin JP, Cardozo T, Lovering RC, Elledge SJ, Pagano M, Harper JW. Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 2004;18:2573–2580. doi: 10.1101/gad.1255304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wu K, Fuchs SY, Chen G, Tan P, Gomez C, Ronai Z, Pan Z-Q. The SCFHOS/β-TRCP-ROC1 E3 ubiquitin ligase utilizes two distinct domains within CUL1 for substrate targeting and ubiquitin ligation. Mol Cell Biol. 2000;20:1382–1393. doi: 10.1128/mcb.20.4.1382-1393.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dehan E, Bassermann F, Guardavaccaro D, Vasiliver-Shamis G, Cohen M, Lowes KN, Dustin M, Huang DC, Taunton J, Pagano M. βTrCP- and Rsk1/2-mediated degradation of BimEL inhibits apoptosis. Mol Cell. 2009;33:109–116. doi: 10.1016/j.molcel.2008.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wrighton KH. Cell signalling: mTOR targets its own inhibitor. Nat Rev Mol Cell Biol. 2011;12:769. doi: 10.1038/nrm3229. [DOI] [PubMed] [Google Scholar]
  • 48.Mao J-H, Kim I-J, Wu D, Climent J, Kang HC, DelRosario R, Balmain A. FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression. Science. 2008;321:1499–1502. doi: 10.1126/science.1162981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hu J, Zacharek S, He YJ, Lee H, Shumway S, Duronio RJ, Xiong Y. WD40 protein FBW5 promotes ubiquitination of tumor suppressor TSC2 by DDB1-CUL4-ROC1 ligase. Genes Dev. 2008;22:866–871. doi: 10.1101/gad.1624008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Katiyar S, Liu E, Knutzen CA, Lang ES, Lombardo CR, Sankar S, Toth JI, Petroski MD, Ronai Z, Chiang GG. REDD1, an inhibitor of mTOR signalling, is regulated by the CUL4A-DDB1 ubiquitin ligase. EMBO Rep. 2009;10:866–872. doi: 10.1038/embor.2009.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, von Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, et al. Targeting of HIF-α to the von Hippel-Lindau ubiquitylation complex by O-2-regulated prolyl hydroxylation. Science. 2001;292:468–472. doi: 10.1126/science.1059796. [DOI] [PubMed] [Google Scholar]
  • 52.Ivan M, Kondo K, Yang HF, Kim W, Valiando J, Ohh M, Salic A, Asara JM, Lane WS, Kaelin WG. HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O-2 sensing. Science. 2001;292:464–468. doi: 10.1126/science.1059817. [DOI] [PubMed] [Google Scholar]
  • 53.Xu X, Sarikas A, Dias-Santagata DC, Dolios G, Lafontant PJ, Tsai SC, Zhu W, Nakajima H, Nakajima HO, Field LJ, et al. The CUL7 E3 ubiquitin ligase targets insulin receptor substrate 1 for ubiquitin-dependent degradation. Mol Cell. 2008;30:403–414. doi: 10.1016/j.molcel.2008.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shi J, Luo L, Eash J, Ibebunjo C, Glass DJ. The SCF-Fbxo40 complex induces IRS1 ubiquitination in skeletal muscle, limiting IGF1 signaling. Dev Cell. 2011;21:835–847. doi: 10.1016/j.devcel.2011.09.011. [DOI] [PubMed] [Google Scholar]
  • 55.Li X, Liu J, Gao T. β-TrCP-mediated ubiquitination and degradation of PHLPP1 are negatively regulated by Akt. Mol Cell Biol. 2009;29:6192–6205. doi: 10.1128/MCB.00681-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dorrello NV, Peschiaroli A, Guardavaccaro D, Colburn NH, Sherman NE, Pagano M. S6K1- and βTRCP-mediated degradation of PDCD4 promotes protein translation and cell growth. Science. 2006;314:467–471. doi: 10.1126/science.1130276. [DOI] [PubMed] [Google Scholar]
  • 57.Guertin DA, Sabatini DM. An expanding role for mTOR in cancer. Trends Mol Med. 2005;18:286–296. doi: 10.1016/j.molmed.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 58.Yang Q, Guan KL. Expanding mTOR signaling. Cell Res. 2007;17:666–681. doi: 10.1038/cr.2007.64. [DOI] [PubMed] [Google Scholar]
  • 59.Arteaga CL. EGF receptor mutations in lung cancer: from humans to mice and maybe back to humans. Cancer Cell. 2006;9:421–423. doi: 10.1016/j.ccr.2006.05.014. [DOI] [PubMed] [Google Scholar]
  • 60.Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5:341–354. doi: 10.1038/nrc1609. [DOI] [PubMed] [Google Scholar]
  • 61.Hynes NE, MacDonald G. ErbB receptors and signaling pathways in cancer. Curr Opin Cell Biol. 2009;21:177–184. doi: 10.1016/j.ceb.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 62.Wang Y, Sun Y. Insulin-like growth factor receptor-1 as an anticancer target: blocking transformation and inducing apoptosis. Curr Cancer Drug Targets. 2002;2:191–207. doi: 10.2174/1568009023333863. [DOI] [PubMed] [Google Scholar]
  • 63.Werner H, LeRoith D. The role of the insulin-like growth factor system in human cancer. Adv Cancer Res. 1996;68:183–223. doi: 10.1016/s0065-230x(08)60354-1. [DOI] [PubMed] [Google Scholar]
  • 64.Cantley LC. The phosphoinositide 3-kinase pathway. Science. 2002;296:1655–1657. doi: 10.1126/science.296.5573.1655. [DOI] [PubMed] [Google Scholar]
  • 65.Yuan TL, Cantley LC. PI3K pathway alterations in cancer: variations on a theme. Oncogene. 2008;27:5497–5510. doi: 10.1038/onc.2008.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, Gazdar A, Powell SM, Riggins GJ, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. doi: 10.1126/science.1096502. [DOI] [PubMed] [Google Scholar]
  • 67.Hollander MC, Blumenthal GM, Dennis PA. PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat Rev Cancer. 2011;11:289–301. doi: 10.1038/nrc3037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liaw D, Marsh DJ, Li J, Dahia PLM, Wang SI, Zheng ZM, Bose S, Call KM, Tsou HC, Peacocke M, et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet. 1997;16:64–67. doi: 10.1038/ng0597-64. [DOI] [PubMed] [Google Scholar]
  • 69.Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and mTOR. Proc Natl Acad Sci USA. 2005;102:8573–8578. doi: 10.1073/pnas.0503224102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Luo J, Manning BD, Cantley LC. Targeting the PI3K-Akt pathway in human cancer: rationale and promise. Cancer Cell. 2003;4:257–262. doi: 10.1016/s1535-6108(03)00248-4. [DOI] [PubMed] [Google Scholar]
  • 71.Meric-Bernstam F, Gonzalez-Angulo AM. Targeting the mTOR signaling network for cancer therapy. J Clin Oncol. 2009;27:2278–2287. doi: 10.1200/JCO.2008.20.0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Baselga J. Targeting the phosphoinositide-3 (PI3) kinase pathway in breast cancer. Oncologist. 2011;16:12–19. doi: 10.1634/theoncologist.2011-S1-12. [DOI] [PubMed] [Google Scholar]
  • 73.Guertin DA, Sabatini DM. The pharmacology of mTOR inhibition. Sci Signal. 2009;2:pe24. doi: 10.1126/scisignal.267pe24. [DOI] [PubMed] [Google Scholar]
  • 74.Vanhaesebroeck B, Stephens L, Hawkins P. PI3K signalling: the path to discovery and understanding. Nat Rev Mol Cell Biol. 2012;13:195–203. doi: 10.1038/nrm3290. [DOI] [PubMed] [Google Scholar]
  • 75.Rini BI. Temsirolimus, an inhibitor of mammalian target of rapamycin. Clin Cancer Res. 2008;14:1286–1290. doi: 10.1158/1078-0432.CCR-07-4719. [DOI] [PubMed] [Google Scholar]
  • 76.Gabardi S, Baroletti SA. Everolimus: a proliferation signal inhibitor with clinical applications in organ transplantation, oncology, and cardiology. Pharmacotherapy. 2010;30:1044–1056. doi: 10.1592/phco.30.10.1044. [DOI] [PubMed] [Google Scholar]
  • 77.Mita M, Sankhala K, Abdel-Karim I, Mita A, Giles F. Deforolimus (AP23573) a novel mTOR inhibitor in clinical development. Expert Opin Investig Drugs. 2008;17:1947–1954. doi: 10.1517/13543780802556485. [DOI] [PubMed] [Google Scholar]
  • 78.Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane WS, Schreiber SL. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369:756–758. doi: 10.1038/369756a0. [DOI] [PubMed] [Google Scholar]
  • 79.Dazert E, Hall MN. mTOR signaling in disease. Curr Opin Cell Biol. 2011;23:744–755. doi: 10.1016/j.ceb.2011.09.003. [DOI] [PubMed] [Google Scholar]
  • 80.Benjamin D, Colombi M, Moroni C, Hall MN. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov. 2011;10:868–880. doi: 10.1038/nrd3531. [DOI] [PubMed] [Google Scholar]
  • 81.Menon S, Manning BD. Common corruption of the mTOR signaling network in human tumors. Oncogene. 2008;27(suppl 2):S43–S51. doi: 10.1038/onc.2009.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y, Reichling LJ, Sim T, Sabatini DM, Gray NS. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem. 2009;284:8023–8032. doi: 10.1074/jbc.M900301200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Feldman ME, Apsel B, Uotila A, Loewith R, Knight ZA, Ruggero D, Shokat KM. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 2009;7:371–383. doi: 10.1371/journal.pbio.1000038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hoang B, Frost P, Shi YJ, Belanger E, Benavides A, Pezeshkpour G, Cappia S, Guglielmelli T, Gera J, Lichtenstein A. Targeting TORC2 in multiple myeloma with a new mTOR kinase inhibitor. Blood. 2010;116:4560–4568. doi: 10.1182/blood-2010-05-285726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Deshaies RJ, Emberley ED, Saha A. Conjugation and Deconjugation of Ubiquitin Family Modifiers: Subcellular Biochemistry. 2010. Control of cullin-ring ubiquitin ligase activity by Nedd8; pp. 41–56. [DOI] [PubMed] [Google Scholar]
  • 86.Xirodimas DP. Novel substrates and functions for the ubiquitin-like molecule NEDD8. Biochem Soc Trans. 2008;36:802–806. doi: 10.1042/BST0360802. [DOI] [PubMed] [Google Scholar]
  • 87.Brownell JE, Sintchak MD, Gavin JM, Liao H, Bruzzese FJ, Bump NJ, Soucy TA, Milhollen MA, Yang X, Burkhardt AL, et al. Substrate-assisted inhibition of ubiquitin-like protein-activating enzymes: the NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ. Mol Cell. 2010;37:102–111. doi: 10.1016/j.molcel.2009.12.024. [DOI] [PubMed] [Google Scholar]
  • 88.Kamura T, Conrad MN, Yan Q, Conaway RC, Conaway JW. The Rbx1 subunit of SCF and VHL E3 ubiquitin ligase activates Rub1 modification of cullins Cdc53 and Cul2. Genes Dev. 1999;13:2928–2933. doi: 10.1101/gad.13.22.2928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Goldenberg SJ, Cascio TC, Shumway SD, Garbutt KC, Liu JD, Xiong Y, Zheng N. Structure of the Cand1-Cul1-Roc1 complex reveals regulatory mechanisms for the assembly of the multisubunit cullin-dependent ubiquitin ligases. Cell. 2004;119:517–528. doi: 10.1016/j.cell.2004.10.019. [DOI] [PubMed] [Google Scholar]
  • 90.Duda DM, Borg LA, Scott DC, Hunt HW, Hammel M, Schulman BA. Structural insights into NEDD8 activation of cullin-RING ligases: conformational control of conjugation. Cell. 2008;134:995–1006. doi: 10.1016/j.cell.2008.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yamoah K, Oashi T, Sarikas A, Gazdoiu S, Osman R, Pan Z-Q. Autoinhibitory regulation of SCF-mediated ubiquitination by human cullin 1's C-terminal tail. Proc Natl Acad Sci USA. 2008;105:12230–12235. doi: 10.1073/pnas.0806155105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Milhollen MA, Traore T, Adams-Duffy J, Thomas MP, Berger AJ, Dang L, Dick LR, Garnsey JJ, Koenig E, Langston SP, et al. MLN4924, a NEDD8-activating enzyme inhibitor, is active in diffuse large B-cell lymphoma models: rationale for treatment of NF-κB-dependent lymphoma. Blood. 2010;116:1515–1523. doi: 10.1182/blood-2010-03-272567. [DOI] [PubMed] [Google Scholar]
  • 93.Swords RT, Kelly KR, Smith PG, Garnsey JJ, Mahalingam D, Medina E, Oberheu K, Padmanabhan S, O'Dwyer M, Nawrocki ST, et al. Inhibition of NEDD8-activating enzyme: a novel approach for the treatment of acute myeloid leukemia. Blood. 2010;115:3796–3800. doi: 10.1182/blood-2009-11-254862. [DOI] [PubMed] [Google Scholar]
  • 94.Tan M, Li Y, Yang R, Xi N, Sun Y. Inactivation of SAG E3 ubiquitin ligase blocks embryonic stem cell differentiation and sensitizes leukemia cells to retinoid acid. PLoS One. 2011;6:e27726. doi: 10.1371/journal.pone.0027726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Milhollen MA, Narayanan U, Soucy TA, Veiby PO, Smith PG, Amidon B. Inhibition of NEDD8-activating enzyme induces rereplication and apoptosis in human tumor cells consistent with deregulating CDT1 turnover. Cancer Res. 2011;71:3042–3051. doi: 10.1158/0008-5472.CAN-10-2122. [DOI] [PubMed] [Google Scholar]
  • 96.Jia L, Li H, Sun Y. Induction of p21-dependent senescence by an NAE inhibitor, MLN4924, as a mechanism of growth suppression. Neoplasia. 2011;13:561–569. doi: 10.1593/neo.11420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Lin HK, Chen Z, Wang G, Nardella C, Lee SW, Chan CH, Yang WL, Wang J, Egia A, Nakayama KI, et al. Skp2 targeting suppresses tumorigenesis by Arf-p53-independent cellular senescence. Nature. 2010;464:374–379. doi: 10.1038/nature08815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Lin JJ, Milhollen MA, Smith PG, Narayanan U, Dutta A. NEDD8-targeting drug MLN4924 elicits DNA rereplication by stabilizing Cdt1 in S phase, triggering checkpoint activation, apoptosis, and senescence in cancer cells. Cancer Res. 2010;70:10310–10320. doi: 10.1158/0008-5472.CAN-10-2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Willems AR, Schwab M, Tyers M. A hitchhiker's guide to the cullin ubiquitin ligases: SCF and its kin. Biochim Biophys Acta. 2004;1695:133–170. doi: 10.1016/j.bbamcr.2004.09.027. [DOI] [PubMed] [Google Scholar]
  • 100.Skaar JR, D'Angiolella V, Pagan JK, Pagano M. SnapShot: F Box Proteins II. Cell. 2009;137:1358. doi: 10.1016/j.cell.2009.05.040. 1358.e1. [DOI] [PubMed] [Google Scholar]
  • 101.Rabut G, Peter M. Function and regulation of protein neddylation. ‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep9. 2008:969–976. doi: 10.1038/embor.2008.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Milhollen MA, Thomas MP, Narayanan U, Traore T, Riceberg J, Amidon BS, Bence NF, Bolen JB, Brownell J, Dick LR, et al. Treatment-emergent mutations in NAEβ confer resistance to the NEDD8-activating enzyme inhibitor MLN4924. Cancer Cell. 2012;21:388–401. doi: 10.1016/j.ccr.2012.02.009. [DOI] [PubMed] [Google Scholar]
  • 103.Toth JI, Yang L, Dahl R, Petroski MD. A gatekeeper residue for NEDD8-activating enzyme inhibition of MLN4924. Cell Reports. 2012;1:309–316. doi: 10.1016/j.celrep.2012.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wei D, Li H, Yu J, Sebolt JT, Zhao L, Lawrence TS, Smith PG, Morgan MA, Sun Y. Radiosensitization of human pancreatic cancer cells by MLN4924, an investigational NEDD8-activating enzyme inhibitor. Cancer Res. 2012;72:282–293. doi: 10.1158/0008-5472.CAN-11-2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Soucy TA, Dick LR, Smith PG, Milhollen MA, Brownell JE. The NEDD8 conjugation pathway and its relevance in cancer biology and therapy. Genes Cancer. 2010;1:708–716. doi: 10.1177/1947601910382898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Soucy TA, Smith PG, Rolfe M. Targeting NEDD8-activated cullin-RING ligases for the treatment of cancer. Clin Cancer Res. 2009;15:3912–3916. doi: 10.1158/1078-0432.CCR-09-0343. [DOI] [PubMed] [Google Scholar]
  • 107.Lindsley CW. The Akt/PKB family of protein kinases: a review of small molecule inhibitors and progress towards target validation: a 2009 update. Curr Top Med Chem. 2010;10:458–477. doi: 10.2174/156802610790980602. [DOI] [PubMed] [Google Scholar]
  • 108.Jia L, Sun Y. SCF E3 ubiquitin ligases as anticancer targets. Curr Cancer Drug Targets. 2011;11:347–356. doi: 10.2174/156800911794519734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Nalepa G, Rolfe M, Harper JW. Drug discovery in the ubiquitin-proteasome system. Nat Rev Drug Discov. 2006;5:596–613. doi: 10.1038/nrd2056. [DOI] [PubMed] [Google Scholar]
  • 110.Chen Q, Xie W, Kuhn DJ, Voorhees PM, Lopez-Girona A, Mendy D, Corral LG, Krenitsky VP, Xu W, Moutouh-de Parseval L, et al. Targeting the p27 E3 ligase SCF(Skp2) results in p27- and Skp2-mediated cell-cycle arrest and activation of autophagy. Blood. 2008;111:4690–4699. doi: 10.1182/blood-2007-09-112904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Aghajan M, Jonai N, Flick K, Fu F, Luo M, Cai X, Ouni I, Pierce N, Tang X, Lomenick B, et al. Chemical genetics screen for enhancers of rapamycin identifies a specific inhibitor of an SCF family E3 ubiquitin ligase. Nat Biotechnol. 2010;28:738–742. doi: 10.1038/nbt.1645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Orlicky S, Tang X, Neduva V, Elowe N, Brown ED, Sicheri F, Tyers M. An allosteric inhibitor of substrate recognition by the SCF(Cdc4) ubiquitin ligase. Nat Biotechnol. 2010;28:733–737. doi: 10.1038/nbt.1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ceccarelli DF, Tang X, Pelletier B, Orlicky S, Xie W, Plantevin V, Neculai D, Chou YC, Ogunjimi A, Al-Hakim A, et al. An allosteric inhibitor of the human Cdc34 ubiquitin-conjugating enzyme. Cell. 2011;145:1075–1087. doi: 10.1016/j.cell.2011.05.039. [DOI] [PubMed] [Google Scholar]

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES