Regulation mechanism of Fbxw7-related signaling pathways (Review)

  • Authors:
    • Zhenyu Zhou
    • Chuanchao He
    • Jie Wang
  • View Affiliations

  • Published online on: August 26, 2015     https://doi.org/10.3892/or.2015.4227
  • Pages: 2215-2224
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

F-box and WD repeat domain-containing 7 (Fbxw7), the substrate-recognition component of SCFFbxw7 complex, is thought to be a tumor suppressor involved in cell growth, proliferation, differentiation and survival. Although an increasing number of ubiquitin substrates of Fbxw7 have been identified, the best characterized substrates are cyclin E and c-Myc. Fbxw7/cyclin E and Fbxw7/c-Myc pathways are tightly regulated by multiple regulators. Fbxw7 has been identified as a tumor suppressor in hepatocellular carcinoma. This review focused on the regulation of Fbxw7/cyclin E and Fbxw7/c-Myc pathways and discussed findings to gain a better understanding of the role of Fbxw7 in hepatocellular carcinoma.

1. Introduction

Cancer is considered a terrible disease that leads to a global health threat. Although significant improvements have been made in the management of cancer as well as the comprehension of the molecular mechanisms of neoplasm pathogenesis and progression, cancer remains a common disorder worldwide, accounting for 12.7 million new cancer cases and 7.6 million cancer deaths in 2008, worldwide (1,2). Previous findings have shown that tumorigenesis in humans is a multi-step process foundational with mutations which can activate oncogenes and inactivate tumor-suppressor genes. The genetic alterations can result in changes of the proteome, and these changes eventually drive the malignant biological behavior through complex signaling pathways (39).

The ubiquitin-proteasome system (UPS), which is responsible for the degradation of >80% of cell proteins, is the main proteolytic mechanisms involved in eukaryocytes (3,4). Most of the proteins involved in cell cycle progression, proliferation and apoptosis are regulated by the UPS (5). Dysregulation of the UPS may contribute to tumor progression, drug resistance and altered immune surveillance (5). Previous studies have focused on the relationship between epithelial-mesenchymal transition (EMT) signal transduction pathways and UPS, and found that many signal transducers and transcription factors involved in EMT are regulated by ubiquitination and the UPS (6). It is well-known that the UPS consists of three classes of enzymes: the ubiquitin-activating enzyme (E1), the ubiquitin-conjugating enzyme (E2) and ubiquitin ligases (E3). In most cases, the three enzymes together with 26S proteasome drive target substrate degradation through a series of catalytic processes (5,7). In the human genome there are two E1s, ~30–40 E2s and >600 E3 ligases (8). It should be emphasized that during the ubiquitination, E3 mediates the transfer of ubiquitin from E2 to the substrate protein. Thus, it has a central role in the substrate decision (9).

The SCF complexes belong to the cullin-RING ligase family and are the largest family of E3 ligases comprising four subunits: S-phase kinase-associated protein 1 (SKP1), which is responsible for recruiting the variable F-box protein; cullin 1 (CUL1), which provides a rigid scaffold connecting SKP1 and RBX1 on opposite ends; RING box 1 (RBX1; also known as ROC1 or HRT1), which serves as an interface for E2 ubiquitin-conjugating enzymes; and a member of the F-box protein family, which functions as a substrate-recognition component (10,11). F-box and WD repeat domain-containing 7 (Fbxw7; also known as Fbw7, hCdc4, hAGO and Sel10) is an evolutionarily conserved protein belonging to the F-box family. It was first identified as Cdc4 in budding yeast in 1973 (12). Sel10 was subsequently identified in Caenorhabditis elegans as a negative regulator of Lin-12 (homology of Notch) (13). In parallel, archipelago, a gene in Drosophila melanogaster encoding a protein containing an F-box domain and seven tandem WD repeats (AGO) domain, was identified (14). From those studies, the human homologue was ascertained and was designated as Fbxw7 or Fbw7.

Fbxw7 is thought to be a tumor suppressor involved in cell growth, proliferation, differentiation and survival (15). Fbxw7 has been found to be inactivated by mutation in various human cancer types (1618). Fbxw7 has an unusual mutation spectrum whereby biallelic, simple loss-of-function mutations are rare. Instead, most mutations are monoallelic missense changes involving specific arginine residues at β-sheet propellor tips that allow the Fbxw7 protein to recognize its substrates (19). Inactivation of the Fbxw7 protein is associated with the deregulation of several well-known oncoproteins with significant capabilities in pathways that manage cell division and growth, including cyclin E (20), c-Myc (21), c-Jun (22), Notch (23), Mcl-1 (24) and mammalian target of rapamycin (mTOR) (25) (Table I).

Table I

Substrates of Fbxw7.

Table I

Substrates of Fbxw7.

Protein nameFull nameRef.
Cyclin E20
c-Myc21
c-Jun22
Notch23
Mcl-1Myeloid cell leukemia 124
mTORMammalian target of rapamycin25
PS1Presenilin 126
N-Myc27
p6328
c-Myb29
HIF-1αHypoxia-inducible transcription factor-1α30
AIB1 (SRC-3)Amplified in breast cancer 131
SREBP1Sterol regulatory element binding protein-132
JunB33
NF-κB2 (p100)Nuclear factor of κ light polypeptide gene enhancer in B cells 234
Aurora AAurora kinase B35
Aurora BAurora kinase A36
CCDC6Coiled-coil domain containing 637
MED13LMediator complex subunit 13-like38
MED13Mediator complex subunit 1338
NF1Neurofibromatosis type 139
KLF2Krüppel-like factor 240
KLF5Krüppel-like factor 541
C/EBPαCCAAT/enhancer binding protein α42
C/EBPδCCAAT/enhancer binding protein δ43
Nrf1Nuclear factor E2-related factor 144
G-CSFRGranulocyte colony stimulating factor receptor45
CREB3L1 (OASIS)cAMP responsive element binding protein 3-like 146
CREB3L2 (BBF2H7)cAMP responsive element binding protein 3-like 246
TopoIIαTopoisomerase IIα47
TGIF1TGFβ-induced factor 148
Eya1Eyes absent 149
GATA3GATA binding protein 350
DAB2IPDisabled homolog 2 interacting protein51
PGC-1αPeroxisome proliferator-activated receptor-γ coactivator 1α52
YAPYes-associated protein53

Therefore, the dysregulation of Fbxw7-mediated proteasome degradation is likely involved in many signaling pathways which play important roles in human cancers. Among these oncogenic Fbxw7 substrates, cyclin E and c-Myc may be the most clearly investigated oncoproteins that have certain contributions to Fbxw7-associated cancers. In this review, we discuss the two major signaling pathways impacted by Fbxw7 to show how Fbxw7-related signaling pathways are regulated in cancer. In addition, since few studies have focused on the relationship between hepatocellular carcinoma (HCC) and Fbxw7, we assess molecular mechanisms by which Fbxw7 exerts antitumor activity in HCC.

2. Fbxw7 and the mechanism of degradation of its substrates

Fbxw7, which shares the WD40 repeats structure (a substrate interaction domain used to classify F-box proteins), is a well-studied member of the F-box family (54). The human FBXW7 gene consists of 4 introns and 13 exons and its gene locus maps to chromosome region 4q32, which is commonly deleted in many types of human malignancies (17,55). The FBXW7 gene encodes the Fbxw7α, Fbxw7β and Fbxw7γ protein isoforms, which have distinct subcellular localizations, with Fbxw7α mainly localizing to nucleoplasm, Fbxw7β to cytoplasm and Fbxw7γ to nucleolus (56). The three isoforms also seem to have tissue specificity with the α-form being found to be extensively expressed in human tissues, while the β-form and/or γ-form are present at lower levels, except in skeletal muscle, brain and to a lesser degree, heart (17).

In addition to the N-terminal region which contains dominant signals for the subcellular localization, each isoform of Fbxw7 shares conserved interaction domains in the C-terminal region: the eight WD40 repeats that determine target specificity; the F-box that recruits the SKP1 of the SCF complex; and the D domain that dimerize the SCFFbxw7, which allows it to target substrates with low-affinity Cdc4 phosphodegron (CPDs) (15). All these domains are essential for the degradation of its substrates.

Although the three Fbxw7 isoforms share many identical functional domains, recent studies have identified that each isoform has its special function. The α-form is most abundant and accounts for degradation of most tested substrates. Fbxw7β is proved to reside in the endoplasmic reticulum membrane and protects cells from oxidative stress (57). Moreover, Fbxw7α and Fbxw7β are found to play an opposite role in their substrates. For example, PGC-1α, a transcriptional coactivator with broad effects on cellular energy metabolism, is found to have a different fate under different Fbxw7 isoforms (52). Fbw7β reduces cellular PGC-1α via ubiquitin-mediated degradation, whereas Fbw7α increases cellular PGC-1α via ubiquitin-mediated stabilization (52).

Evidence suggests that substrates of Fbxw7 are polyubiquitinated in a GSK3-dependent manner by SCFFbxw7 (22,33,58). Glycogen synthase kinase 3 (GSK3), firstly identified in 1980, is a constitutively active and ubiquitously expressed serine/threonine kinase (59,60). Human cells contain two GSK3 isoforms, known as GSK3α and GSK3β, which are highly similar with respect to sequence (share 97% amino acid sequence within their catalytic domains) and function (61). The activation of GSK3 is dominated by phosphorylation on Ser-21 of GSK3α and Ser-9 of GSK3β (62). It is well established that these phosphorylations are regulated via PI3K/Akt pathway (63). In most cases, inactivation of the PI3K/Akt pathway leads to dephosphorylation of GSK3, which results in the activation of GSK3. The activated GSK3 subsequently phosphorylates the CPDs of the substrates that are primed by phosphorylation at position +4 of the CPD by a yet to-be-identified kinase (in the cyclin E case, which is Cdk2) (33,64). The CPDs of substrates are recognized and interacted with the eight WD40 repeats of Fbxw7 for ubiquitin-mediated proteolytic degradation (65).

CPD is a phosphodegron motif existing in the substrates of Fbxw7. The phosphorylation of CPD by GSK3 is crucial for the interaction between Fbxw7 and its substrate. Mutation of critical residues within the CPD lead to stabilization of substrates, such as c-Myc and has been observed in many human cancers (66). Data gathering from Fbxw7 substrates have ascertained the conserved CDP sequence as ΦXΦΦΦ-T/S-PPX-S/T/E, with Φ standing for a hydrophobic residue and X for any amino acid (65,67). The T/S residue can be phosphorylated by GSK3 and the phosphorylation of S/T/E residue serves as a priming signal for GSK3 phosphorylation. Moreover, studies also found that some substrates have more than one CPD. For example, studies have revealed that cyclin E has two CPDs, with one located in the T380 and the other one being centered at ~T62 (68,69). The two CPDs of cyclin E were essential for efficiency of Fbxw7 binding, in response to different signaling pathways (70).

3. Regulation of Fbxw7-cyclin E/c-Myc signaling pathways

Over the past decade, a number of studies have contributed to the understanding of Fbxw7 molecular mechanisms in human cancers. Several molecules that play an essential role in tumor pathogenesis and progression have been identified to be regulated by Fbxw7, and are now recognized as potential therapeutic targets. In the present review, we focus on the mechanism by which cyclin E and c-Myc are regulated by Fbxw7.

Fbxw7/cyclin E pathway

By binding to its kinase partner Cdk2, cyclin E regulates the cell cycle by promoting the G1-S transition (71). Deregulation of cyclin E causes genomic instability and is thought to directly contribute to cell transformation and tumorigenesis (72). Cyclin E, containing two CPDs that are phosphorylated by GSK3 and autophosphorylated by Cdk2, respectively, is the most well-studied substrate of Fbxw7 (69). Cyclin E has gained much attention as a key mediator for the tumor-suppressor ability of Fbxw7. Specifically, Fbxw7 tightly regulates the abundance of cyclin E through many molecular mechanisms.

p53 has a complicated and incompletely understood interplay between Fbxw7 and cyclin E. Previous observations revealed that the p53-p21 pathway, which is induced by excess cyclin E, suppresses cyclin E kinase activity and this pathway cooperates with SCFFbxw7 to suppress cyclin E-induced genome instability (73,74). Moreover, when using a cDNA-microarray system, p53 protein is found to be a transcriptional activator of FBXW7b (75). p53 arrests cell cycle progression at G0-G1 by inducing the Fbxw7β-mediated downregulation of cyclin E expression (75). Similarly, another group provided results showing that, p53 regulates the levels of cyclin E protein to impose a G1-S block through the activation of ago in Drosophila (76). However, previous findings have also shown that, Fbxw7β is not necessary for the degradation of cyclin E in mammals as mentioned below. Fbxw7 was eventually proven to act as an upstream of p53 by inducing Aurora A degradation (77). Aurora A phosphorylates p53 at S315/215 and thus reduces p53 levels and transcriptional activity (77) (Fig. 1). However, more studies are needed to clarify these paradoxical and complicated relationships.

Some molecules influence the process whereby Fbxw7 modulates cyclin E protein degradation, for instance rictor (78), Artemis (79) and SV40 large T (LT) (80) (Fig. 1). Although rictor is usually identified as a binding partner of mTOR, numerous complexes containing rictor have been recognized and shown to be mTOR-independent (81). Fbxw7α was recently shown to form a complex with rictor. The rictor/Fbxw7 complex functions as an E3 ligase complex, and promotes the degradation of cyclin E in an mTOR-independent manner (78). Artemis, a member of the SNM1 gene family, is a known phosphorylation target of ATM, ATR and DNA-PKcs in response to various types of genotoxic stress (82). In response to UV irradiation, Artemis is phosphorylated at S516 and S645 by the ATR kinase. The phosphorylated Artemis then interacts with Fbxw7α or Fbxw7γ and induces strong ubiquitylation of cyclin E (79). Similarly, the knockdown of rictor by shRNA and inactivation of Artemis by the mutation of S516/645 to alanine can lead to a decreasing ubiquitination of cyclin E (78,79). Contrary to rictor and Artemis, LT negatively regulates the degradation of cyclin E by Fbxw7. LT is a viral oncoprotein producing in cells infected with simian virus 40 (SV40) (83). SV40 LT protein binds to a number of host cell proteins and disrupts their normal functions. Welcker and and Clurman (80) found that LT also has a consensus CPD in its C terminus that can be phosphorylated at T701, through which LT functions as a competitive inhibitor of Fbxw7 and then deregulates ubiquitination of cyclin E. The Ras/MAPK pathway is also involved in the induction of cyclin E stability by altering the physical interaction between Fbxw7 and cyclin E, but not by altering cyclin E phosphorylation on any of its known regulatory sites (84,85) (Fig. 1).

In addition to interacting with Fbxw7, some factors may target cyclin E and disturb the interaction between Fbxw7 and cyclin E. For example, the protein phosphatase PP2A-B55β targets the N- and C-terminal phosphodegrons of cyclin E1 for dephosphorylation, thereby protecting it from degradation mediated by the SCFFbxw7 ubiquitin ligase (86) (Fig. 1). These data suggest the interaction between Fbxw7 and cyclin E is tightly regulated by different regulators. However, as yet unknown regulators remain to be identified in the future.

It has been demonstrated that ubiquitin-mediated degradation of cyclin E in mammals can be divided into two ways: one requires only Fbxw7α and the other requires Fbxw7α and Fbxw7γ, although not Fbxw7β (56,87,88) (Fig. 2). When the expression of cyclin E is low, presumably normal, the inactivation of cyclin E requires Fbxw7α and Fbxw7γ in a two-step manner. Firstly, cyclin E, which is phosphorylated at S384 by Cdk2, interacts with prolyl cis-trans isomerase Pin1 in conjunction with SCFFbxw7α, carries out a noncanonical isomerization of a proline-proline bond in the cyclin E phosphodegron (P382) (56). Then, being mediated by nucleolar protein nucleophosmin (NPM), the complex binds to SCFFbxw7γ and translocates from the nucleoplasm into the nucleolus where cyclin E is multiubiquitylated, but does not execute proteasomal degradation (87). Notably, Cdk2 kinase was recently demonstrated to interact with Fbxw7γ, result in Fbxw7γ degradation (89). This system serves a novel mechanism for rapid inactivation of cyclin E, through separation of cyclin E from its targets.

Some studies have focused on the upstream of the Fbxw7/cyclin E pathway. Their findings have identified many factors that increase or decrease Fbxw7 expression, thus regulating the Fbxw7/cyclin E pathway (Fig. 1). The PI3K/Akt pathway, which was previously thought to be pivotal for the regulation of the GSK phosphorylation, was recently reported to mediate the phosphorylation of Fbxw7α at S227 (90). This modification stabilizes Fbxw7 and promotes ubiquitylation of the two substrates, cyclin E and c-Myc (90). In addition, Sim et al reported a novel mechanism whereby TRIP-Br proteins links to E2F to act as upstream regulators of the Fbxw7/cyclin E pathway in the maintenance of genomic stability (91). This mechanism reveals a function distinct from the conventional function of E2F, which increases cyclin E expression at the transcriptional level. TRIP-Br proteins interact with PHD zinc finger and/or bromodomain proteins such as KRIP-1 and p300/CBP, upregulate the FBXW7 gene product via the activation of E2F transcriptional activity, and subsequently lead to downregulation of the cyclin E protein (91). Polo-like kinase 2 (PIK2) is also found to directly phosphorylate Fbxw7 at S25, S176 and S349. However, these phosphorylations reduce the stability of Fbxw7, thus stabilizing cyclin E, and contributing to the duplication of centrioles and aneuploidy (92). TAL1, a class II basic helix-loop-helix (bHLH) transcription factor, promotes the malignant phenotype in T-ALL through the repression of Fbxw7 in a miR223-dependent manner. This effect leads to a marked increase of the expression of cyclin E and c-Myc (93).

The most distinctive function of cyclin E is to bind to and activate Cdk2. Activation of the cyclin E/Cdk2 complex subsequently leads to the phosphorylation of its substrates, such as retinoblastoma (81), cdc6, NPM, p21 and p27 (72). Of note, cyclin E/Cdk2 was reported to directly phosphorylate Cdh1, thus inactivating APCCdh1, an E3 ligase important for genomic stability (94). Furthermore, Lau et al showed that the Fbxw7/cyclin E pathway regulates the activation of APCCdh1 through direct phosphorylation of Cdh1 (95). This subsequently leads to the inactivation of APCCdh1 E3 ligase and upregulation of APCCdh1-specific substrates, which are well-characterized oncoproteins (Fig. 1). These results suggested that Fbxw7 regulates cell cycle through, not only cyclin E itself, but also the downstream substrates of cyclin E/Cdk2 complex.

Taken together, these findings suggest that Fbxw7/cyclin E is regulated by multiple regulators, which partly explains why there are no FBXW7 gene mutations in some tumors. Moreover, these findings provide insight into the tumor-suppressive function of Fbxw7.

Fbxw7/c-Myc pathway

The c-Myc protein, a basic helix-loop-helix zipper (bHLH/Zip)-type transcription factor, generally combines with its cofactor MAX and activates their target genes transcription by binding E-box motifs (CACGTG), and thus playing a predominant role in cell proliferation and tumorigenesis (96). Accumulated evidence has indicated that c-Myc protein turnover is tightly regulated at the post-translational level through ubiquitin-proteasome pathway controlled by the SCFFbxw7 complex (97,98). The oncoprotein c-Myc has only one CPD, containing the sequence PTPPLSP (residues 57–63 in human c-Myc), within which T58 and S62 are the phosphorylation sites (98). However, these two phosphorylation sites exert opposite functions on c-Myc degradation, as the phosphorylation of S62 results in c-Myc stabilization, whereas the T58 phosphorylation by GSK3β contributes to the interaction between Fbxw7 and c-Myc (98,99). The phosphorylation of T58 depends on the prime phosphorylation of Ser-62. This phosphorylation-dependent proteolysis controlled by SCFFbxw7 is involved with complicated feedback mechanisms.

The activation of Ras-dependent phosphorylation pathways is considered to be important for c-Myc stability mainly through two effective pathways: the Raf/MEK/ERK pathway stabilizes c-Myc by enhancing S62 phosphorylation, and the PI3K/Akt pathway which disrupts the interaction between Fbxw7 and c-Myc by phosphorylating GSK3β at S9, which in turn decreases the phosphorylation of T58 within c-Myc (99). Thus in quiescent cells, where the growth stimulus disappears, Ras activity declines, and the activity of the PI3K/Akt pathway is also downregulated, resulting in the enhancement of T58 phosphorylation and the degradation of c-Myc. It has been shown that the dephosphorylation of S62 is also crucial for the ubiquitin-mediated degradation of c-Myc. T58 phosphorylation is found to facilitate the interaction between c-Myc and Pin1 which catalyzes the isomerization of proline residues in c-Myc to promote S62 dephosphorylation by protein phosphatase 2A (PP2A) (100). Further study showed that Axin1, a multi-domain scaffold protein, regulates this process by facilitating the association of GSK3β, B56α (one of the regulatory subunits of PP2A), and Pin1 with c-Myc, forming an Axin-Pin1-GSK3β-PP2A/B56α complex to promote the ubiquitin-mediated degradation of c-Myc (101). c-Myc has been reported to bind to and transcriptionally activate the PPP2R5D gene that encodes B56δ (another regulatory subunit of PP2A) (102). PP2A-B56δ, not only S dephosphorylates S62 within c-Myc in the same manner as PP2A-B56α, but also reverses the GSK3β inhibitory phosphorylation at S9 by PI3K/Akt (102). These studies provide a mechanism that links c-Myc protein degradation controlled by Fbxw7 to a complicated feedback pathway. In addition, Cdk5, Cdk1 and CIP2A increase S62 of c-Myc phosphorylation in a direct or indirect manner, resulting in c-Myc stabilization (103105) (Fig. 3).

Similar to cyclin E, the Fbxw7/c-Myc pathway appears to be linked to many signal molecules. Previous findings have shown that rictor binds to Fbxw7 and facilitates ubiquitination of c-Myc (78). In addition to rictor, other interaction partners such as stomatin-like protein 1 (SLP-1), NPM, and Bloom (BLM) forms complexes with Fbxw7; in particular, Fbxw7γ, which is specific for the ubiquitination of c-Myc (89,106108). By performing a two-hybrid screen, Zhang et al identified that SLP-1, as a novel interaction partner, can bind to the N-terminus of Fbxw7γ and stabilize Fbxw7γ, leading to an even greater reduction in c-Myc abundance (89). NPM is required by Fbxw7γ for the proper folding, nucleolar localization and stabilization. Mutation of NPM induces delocalization and destabilization of Fbxw7γ and stabilization of c-Myc (106). Recently, BLM, which is a helicase mutated in Bloom syndrome and is conclusively regarded as a sensor of DNA lesion, was reported to have contact with c-Myc and Fbxw7, leading to the degradation of c-Myc and subsequent delay of colorectal tumorigenesis (108,109).

By contrast, many factors serve as binding partners of c-Myc, and block the degradation of c-Myc in a Fbxw7-dependent manner. For example, NF-κB essential modulator (NEMO) induces the upregulation of c-Myc protein through direct interaction with c-Myc protein and inhibits ubiquitination activity of Fbxw7 without interfering with the interaction of Fbxw7 with T58-phoshorylated c-Myc (110).

Although some studies focus on the interaction mechanism between Fbxw7 and c-Myc, other studies provide mechanistic insights for the regulation of the Fbxw7/c-Myc pathway through a reduction of Fbxw7 expression. For example, NF-κB1 (p50), a ubiquitously expressed subunit of NF-κB, is thought to suppress FBXW7 gene transcription and upregulate c-Myc protein expression (111). Huang et al revealed that Fbxw7 was profoundly upregulated in p50-deficient cells in comparison to that in p50 intact cells, whereas knockdown of Fbxw7 in p50−/− cells restored arsenite-induced c-Myc protein accumulation (111). COP9 signalosome (CSN) has been found to facilitate the autoubiquitination/degradation of Fbxw7, thereby stabilizing c-Myc (112). Similar to protein factors, some miRNAs participate in the regulation of c-Myc by interfering with the expression of Fbxw7. For example, miR-92 mediates the proteolytic degradation of c-Myc by direct repression of Fbxw7 in a Eµ-myc Burkitt's lymphoma model (113). Thus, miR-92 overexpression leads to increase of aberrant c-Myc.

Taken together, the abovementioned studies showed that the Fbxw7/c-Myc pathway is regulated by accurate mechanisms. These mechanisms ensure the cell cycle progression in normal cells and regulate cell proliferation and tumorigenesis in malignant tumors. Moreover, these regulation mechanisms (regulation of the Fbxw7/cylin E and Fbxw7/c-Myc pathways) may also exist in other Fbxw7-specific substrates. Furthermore, the fact that some regulators are involved in the Fbxw7/cyclin E and Fbxw7/c-Myc pathways suggests that the two pathways are important for tumor cell proliferation, and the abnormality of these pathways may be partially regulated by the same mechanisms.

4. Role of Fbxw7 in hepatocellular carcinoma

As a tumor suppressor, mRNA and protein expression levels of Fbxw7 have been shown to be downregulated in various types of human cancer. Fbxw7 has been found to be inactivated by mutation in several malignancies with an overall mutation frequency of ~6% (18). However, no studies have yet reported Fbxw7 mutations in HCC, one of the leading causes of cancer-related mortality worldwide. Notably, Fbxw7 expression is reported to be important in the regulation of lipogenesis and cell proliferation and differentiation in the liver (114). Using liver-specific Fbxw7 null mice, Onoyama et al found that the hepatic ablation of Fbxw7 resulted in hepatomegaly and steatohepatitis, and long-term Fbxw7 deficiency resulted in marked proliferation of the biliary system and development of hamartomas (114). Moreover, double heterozygous p53−/− Fbxw7−/− mice have been proven to develop hepatocarcinomas (35). These results suggest that Fbxw7 is critical for the liver development and tumorigenesis.

Previous studies from different groups have identified the role of Fbxw7 as a tumor suppressor in HCC. Chen et al were the first group to investigate Fbxw7 in HCC (47). Their findings revealed that Fbxw7 promoted ubiquitin-dependent degradation of TopoIIα in a HADC-dependent manner (47). Liu et al found that Fbxw7 was involved in the ubiquitin-dependent degradation of AIB1 in Hbx-related HCC (31). Notably, Tu et al were the first group to report that the Fbxw7 mRNA and protein expression in HCC tissues was significantly lower than that in normal tumor-adjacent tissues (115). Subsequently, they also identified that c-Myc, cyclin E, and YAP proteins abundance in HCC was regulated by Fbxw7 (53,116). Consistent with Tu et al, Imura et al reported a lower Fbxw7 expression in HCC tissues compared with non-tumor liver tissues (117). Coincidentally, all these substrates of Fbxw7 in HCC are closely associated with promoting cancer cell proliferation and tumorigenesis (53,71,96,118,119). Given that c-Myc, YAP and AIB1 have transcriptional activity and promote the transcription of many oncogenes with different functions, Fbxw7 may be associated with controlling cell growth and regulating other malignant behavior, such as invasion and metastasis (96,120,121) (Fig. 4). Taken together, these results show that Fbxw7 plays a critical role in HCC. The inactivation of Fbxw7 in HCC is involved in tumorigenesis. However, in-depth investigation is required to determine how Fbxw7 is inactivated in HCC and whether other signal pathways, through which Fbxw7 plays as a tumor suppressor in HCC, exist.

5. Conclusions

Most cell proteins involved in cell cycle progression, proliferation and apoptosis are regulated by the UPS, which consist of three classes of enzymes (E1, E2 and E3). The SCFFbxw7 complex is one of the most well-known E3 ligases. Fbxw7, as the substrates-recognition component of the SCFFbxw7 complex, regulates cell proliferation, genetic stability, and tumorigenesis in humans by coordinating the ubiquitin-dependent proteolysis of several key oncoproteins. Several studies (refs?) have identified a growing list of specific substrates of Fbxw7, such as YAP and Eya1. However, cyclin E and c-Myc are the best characterized oncoproteins among the substrates of Fbxw7. Multiple factors tightly regulate the Fbxw7/cyclin E and Fbxw7/c-Myc pathways in different mechanisms. These regulation mechanisms may also exist in other Fbxw7-specific substrates. These mechanisms therefore may be useful in understanding the functions of Fbxw7-related signaling pathways in the regulation of cell proliferation and tumorigenesis in cancer. Furthermore, this understanding reveals Fbxw7-related signaling pathways have potential in developing new targets in cancer therapy. In addition, since many studies have reported that Fbxw7 functions as a tumor suppressor and plays a critical role in regulating several key oncoproteins in HCC, Fbxw7 is a potential therapeutic target in this cancer. However, future studies should be conducted to invesigate the mechanism of Fbxw7 and how it may serve as a tumor suppressor in HCC.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81301768 and 81372565). The authors would like to thank Professor Qingguang Liu and Dr Kangsheng Tu (Xi'an Jiaotong University, Xi'an, China) for their technical assistance.

Abbreviations:

Fbxw7

F-box and WD repeat domain-containing 7

HCC

hepatocellular carcinoma

UPS

ubiquitin-proteasome system

EMT

epithelial-mesenchymal transition

E1

ubiquitin-activating enzyme

E2

ubiquitin-conjugating enzyme

E3

ubiquitin ligases

SKP1

S-phase kinase-associated protein 1

CUL1

cullin 1

RBX1

RING box 1

mTOR

mammalian target of rapamycin

CPD

Cdc4 phosphodegron

GSK3

glycogen synthase kinase 3

NPM

nucleolar protein nucleophosmin

PIK2

Polo-like kinase 2

bHLH

basic helix-loop-helix

bHLH/Zip

basic helix-loop-helix zipper

PP2A

protein phosphatase 2A

BLM

bloom

NEMO

NF-κB essential modulator

CSN

COP9 signalosome

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar

2 

Allemani C, Weir HK, Carreira H, Harewood R, Spika D, Wang XS, Bannon F, Ahn JV, Johnson CJ, Bonaventure A, et al CONCORD Working Group: Global surveillance of cancer survival 1995–2009: Analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (CONCORD-2). Lancet. 385:977–1010. 2015. View Article : Google Scholar

3 

Adams J: Development of the proteasome inhibitor PS-341. Oncologist. 7:9–16. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Tu Y, Chen C, Pan J, Xu J, Zhou ZG and Wang CY: The Ubiquitin Proteasome Pathway (UPP) in the regulation of cell cycle control and DNA damage repair and its implication in tumorigenesis. Int J Clin Exp Pathol. 5:726–738. 2012.PubMed/NCBI

5 

Bedford L, Lowe J, Dick LR, Mayer RJ and Brownell JE: Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov. 10:29–46. 2011. View Article : Google Scholar

6 

Voutsadakis IA: The ubiquitin-proteasome system and signal transduction pathways regulating Epithelial Mesenchymal transition of cancer. J Biomed Sci. 19:672012. View Article : Google Scholar : PubMed/NCBI

7 

Devoy A, Soane T, Welchman R and Mayer RJ: The ubiquitin-proteasome system and cancer. Essays Biochem. 41:187–203. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Komander D: The emerging complexity of protein ubiquitination. Biochem Soc Trans. 37:937–953. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Pickart CM: Mechanisms underlying ubiquitination. Annu Rev Biochem. 70:503–533. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Zheng N, Schulman BA, Song L, Miller JJ, Jeffrey PD, Wang P, Chu C, Koepp DM, Elledge SJ, Pagano M, et al: Structure of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature. 416:703–709. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Schulman BA, Carrano AC, Jeffrey PD, Bowen Z, Kinnucan ER, Finnin MS, Elledge SJ, Harper JW, Pagano M and Pavletich NP: Insights into SCF ubiquitin ligases from the structure of the Skp1-Skp2 complex. Nature. 408:381–386. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Hartwell LH, Mortimer RK, Culotti J and Culotti M: Genetic control of the cell division cycle in yeast: V. genetic analysis of cdc mutants. Genetics. 74:267–286. 1973.PubMed/NCBI

13 

Hubbard EJ, Wu G, Kitajewski J and Greenwald I: sel-10, a negative regulator of lin-12 activity in Caenorhabditis elegans, encodes a member of the CDC4 family of proteins. Genes Dev. 11:3182–3193. 1997. View Article : Google Scholar

14 

Moberg KH, Bell DW, Wahrer DC, Haber DA and Hariharan IK: Archipelago regulates Cyclin E levels in Drosophila and is mutated in human cancer cell lines. Nature. 413:311–316. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Welcker M and Clurman BE: FBW7 ubiquitin ligase: A tumour suppressor at the crossroads of cell division, growth and differentiation. Nat Rev Cancer. 8:83–93. 2008. View Article : Google Scholar

16 

Rajagopalan H, Jallepalli PV, Rago C, Velculescu VE, Kinzler KW, Vogelstein B and Lengauer C: Inactivation of hCDC4 can cause chromosomal instability. Nature. 428:77–81. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Spruck CH, Strohmaier H, Sangfelt O, Müller HM, Hubalek M, Müller-Holzner E, Marth C, Widschwendter M and Reed SI: hCDC4 gene mutations in endometrial cancer. Cancer Res. 62:4535–4539. 2002.PubMed/NCBI

18 

Akhoondi S, Sun D, von der Lehr N, Apostolidou S, Klotz K, Maljukova A, Cepeda D, Fiegl H, Dafou D, Marth C, et al: FBXW7/hCDC4 is a general tumor suppressor in human cancer. Cancer Res. 67:9006–9012. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Davis H, Lewis A, Behrens A and Tomlinson I: Investigation of the atypical FBXW7 mutation spectrum in human tumours by conditional expression of a heterozygous propellor tip missense allele in the mouse intestines. Gut. 63:792–799. 2014. View Article : Google Scholar :

20 

Koepp DM, Schaefer LK, Ye X, Keyomarsi K, Chu C, Harper JW and Elledge SJ: Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase. Science. 294:173–177. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Yada M, Hatakeyama S, Kamura T, Nishiyama M, Tsunematsu R, Imaki H, Ishida N, Okumura F, Nakayama K and Nakayama KI: Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7. EMBO J. 23:2116–2125. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Wei W, Jin J, Schlisio S, Harper JW and Kaelin WG Jr: The v-Jun point mutation allows c-Jun to escape GSK3-dependent recognition and destruction by the Fbw7 ubiquitin ligase. Cancer Cell. 8:25–33. 2005. View Article : Google Scholar : PubMed/NCBI

23 

O'Neil J, Grim J, Strack P, Rao S, Tibbitts D, Winter C, Hardwick J, Welcker M, Meijerink JP, Pieters R, et al: FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to gamma-secretase inhibitors. J Exp Med. 204:1813–1824. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Inuzuka H, Shaik S, Onoyama I, Gao D, Tseng A, Maser RS, Zhai B, Wan L, Gutierrez A, Lau AW, et al: SCFFBW7 regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction. Nature. 471:104–109. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Mao JH, Kim IJ, Wu D, Climent J, Kang HC, DelRosario R and Balmain A: FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression. Science. 321:1499–1502. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Li J, Pauley AM, Myers RL, Shuang R, Brashler JR, Yan R, Buhl AE, Ruble C and Gurney ME: SEL-10 interacts with presenilin 1, facilitates its ubiquitination, and alters A-beta peptide production. J Neurochem. 82:1540–1548. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Brockmann M, Poon E, Berry T, Carstensen A, Deubzer HE, Rycak L, Jamin Y, Thway K, Robinson SP, Roels F, et al: Small molecule inhibitors of aurora-a induce proteasomal degradation of N-myc in childhood neuroblastoma. Cancer Cell. 24:75–89. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Galli F, Rossi M, D'Alessandra Y, De Simone M, Lopardo T, Haupt Y, Alsheich-Bartok O, Anzi S, Shaulian E, Calabrò V, et al: MDM2 and Fbw7 cooperate to induce p63 protein degradation following DNA damage and cell differentiation. J Cell Sci. 123:2423–2433. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Kitagawa K, Hiramatsu Y, Uchida C, Isobe T, Hattori T, Oda T, Shibata K, Nakamura S, Kikuchi A and Kitagawa M: Fbw7 promotes ubiquitin-dependent degradation of c-Myb: Involvement of GSK3-mediated phosphorylation of Thr-572 in mouse c-Myb. Oncogene. 28:2393–2405. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Cassavaugh JM, Hale SA, Wellman TL, Howe AK, Wong C and Lounsbury KM: Negative regulation of HIF-1α by an FBW7-mediated degradation pathway during hypoxia. J Cell Biochem. 112:3882–3890. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Liu Y, Tong Z, Li T, Chen Q, Zhuo L, Li W, Wu RC and Yu C: Hepatitis B virus X protein stabilizes amplified in breast cancer 1 protein and cooperates with it to promote human hepatocellular carcinoma cell invasiveness. Hepatology. 56:1015–1024. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Tu K, Zheng X, Yin G, Zan X, Yao Y and Liu Q: Evaluation of Fbxw7 expression and its correlation with expression of SREBP-1 in a mouse model of NAFLD. Mol Med Rep. 6:525–530. 2012.PubMed/NCBI

33 

Pérez-Benavente B, García JL, Rodríguez MS, Pineda-Lucena A, Piechaczyk M, Font de Mora J and Farràs R: GSK3-SCFFBXW7 targets JunB for degradation in G2 to preserve chromatid cohesion before anaphase. Oncogene. 32:2189–2199. 2013. View Article : Google Scholar

34 

Arabi A, Ullah K, Branca RM, Johansson J, Bandarra D, Haneklaus M, Fu J, Ariës I, Nilsson P, Den Boer ML, et al: Proteomic screen reveals Fbw7 as a modulator of the NF-κB pathway. Nat Commun. 3:9762012. View Article : Google Scholar

35 

Mao JH, Perez-Losada J, Wu D, Delrosario R, Tsunematsu R, Nakayama KI, Brown K, Bryson S and Balmain A: Fbxw7/Cdc4 is a p53-dependent, haploinsufficient tumour suppressor gene. Nature. 432:775–779. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Teng CL, Hsieh YC, Phan L, Shin J, Gully C, Velazquez-Torres G, Skerl S, Yeung SC, Hsu SL and Lee MH: FBXW7 is involved in Aurora B degradation. Cell Cycle. 11:4059–4068. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Zhao J, Tang J, Men W and Ren K: FBXW7-mediated degradation of CCDC6 is impaired by ATM during DNA damage response in lung cancer cells. FEBS Lett. 586:4257–4263. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Davis MA, Larimore EA, Fissel BM, Swanger J, Taatjes DJ and Clurman BE: The SCF-Fbw7 ubiquitin ligase degrades MED13 and MED13L and regulates CDK8 module association with Mediator. Genes Dev. 27:151–156. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Tan M, Zhao Y, Kim SJ, Liu M, Jia L, Saunders TL, Zhu Y and Sun Y: SAG/RBX2/ROC2 E3 ubiquitin ligase is essential for vascular and neural development by targeting NF1 for degradation. Dev Cell. 21:1062–1076. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Wang R, Wang Y, Liu N, Ren C, Jiang C, Zhang K, Yu S, Chen Y, Tang H, Deng Q, et al: FBW7 regulates endothelial functions by targeting KLF2 for ubiquitination and degradation. Cell Res. 23:803–819. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Bialkowska AB, Liu Y, Nandan MO and Yang VW: A colon cancer-derived mutant of Krüppel-like factor 5 (KLF5) is resistant to degradation by glycogen synthase kinase 3β (GSK3β) and the E3 ubiquitin ligase F-box and WD repeat domain-containing 7α (FBW7α). J Biol Chem. 289:5997–6005. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Bengoechea-Alonso MT and Ericsson J: The ubiquitin ligase Fbxw7 controls adipocyte differentiation by targeting C/EBPalpha for degradation. Proc Natl Acad Sci USA. 107:11817–11822. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Balamurugan K, Sharan S, Klarmann KD, Zhang Y, Coppola V, Summers GH, Roger T, Morrison DK, Keller JR and Sterneck E: FBXW7α attenuates inflammatory signalling by downregulating C/EBPδ and its target gene Tlr4. Nat Commun. 4:16622013. View Article : Google Scholar

44 

Biswas M, Phan D, Watanabe M and Chan JY: The Fbw7 tumor suppressor regulates nuclear factor E2-related factor 1 transcription factor turnover through proteasome-mediated proteolysis. J Biol Chem. 286:39282–39289. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Lochab S, Pal P, Kapoor I, Kanaujiya JK, Sanyal S, Behre G and Trivedi AK: E3 ubiquitin ligase Fbw7 negatively regulates granulocytic differentiation by targeting G-CSFR for degradation. Biochim Biophys Acta. 1833:2639–2652. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Yumimoto K, Matsumoto M, Onoyama I, Imaizumi K and Nakayama KI: F-box and WD repeat domain-containing-7 (Fbxw7) protein targets endoplasmic reticulum-anchored osteogenic and chondrogenic transcriptional factors for degradation. J Biol Chem. 288:28488–28502. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Chen MC, Chen CH, Chuang HC, Kulp SK, Teng CM and Chen CS: Novel mechanism by which histone deacetylase inhibitors facilitate topoisomerase IIα degradation in hepatocellular carcinoma cells. Hepatology. 53:148–159. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Bengoechea-Alonso MT and Ericsson J: Tumor suppressor Fbxw7 regulates TGFβ signaling by targeting TGIF1 for degradation. Oncogene. 29:5322–5328. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Sun Y and Li X: The canonical wnt signal restricts the glycogen synthase kinase 3/fbw7-dependent ubiquitination and degradation of eya1 phosphatase. Mol Cell Biol. 34:2409–2417. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Kitagawa K, Shibata K, Matsumoto A, Matsumoto M, Ohhata T, Nakayama KI, Niida H and Kitagawa M: Fbw7 targets GATA3 through cyclin-dependent kinase 2-dependent proteolysis and contributes to regulation of T-cell development. Mol Cell Biol. 34:2732–2744. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Dai X, North BJ and Inuzuka H: Negative regulation of DAB2IP by Akt and SCFFbw7 pathways. Oncotarget. 5:3307–3315. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Trausch-Azar JS, Abed M, Orian A and Schwartz AL: Isoform-specific SCFFbw7 ubiquitination mediates differential regulation of PGC-1α. J Cell Physiol. 230:842–852. 2015. View Article : Google Scholar

53 

Tu K, Yang W, Li C, Zheng X, Lu Z, Guo C, Yao Y and Q: Fbxw7 is an independent prognostic marker and induces apoptosis and growth arrest by regulating YAP abundance in hepatocellular carcinoma. Mol Cancer. 13:1102014. View Article : Google Scholar : PubMed/NCBI

54 

Jin J, Cardozo T, Lovering RC, Elledge SJ, Pagano M and Harper JW: Systematic analysis and nomenclature of mammalian F-box proteins. Genes Dev. 18:2573–2580. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Sterian A, Kan T, Berki AT, Mori Y, Olaru A, Schulmann K, Sato F, Wang S, Paun B, Cai K, et al: Mutational and LOH analyses of the chromosome 4q region in esophageal adenocarcinoma. Oncology. 70:168–172. 2006. View Article : Google Scholar : PubMed/NCBI

56 

van Drogen F, Sangfelt O, Malyukova A, Matskova L, Yeh E, Means AR and Reed SI: Ubiquitylation of cyclin E requires the sequential function of SCF complexes containing distinct hCdc4 isoforms. Mol Cell. 23:37–48. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Matsumoto A, Tateishi Y, Onoyama I, Okita Y, Nakayama K and Nakayama KI: Fbxw7β resides in the endoplasmic reticulum membrane and protects cells from oxidative stress. Cancer Sci. 102:749–755. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Ren H, Zhao L, Li Y, Yue P, Deng X, Owonikoko TK, Chen M, Khuri FR and Sun SY: The PI3 kinase inhibitor NVP-BKM120 induces GSK3/FBXW7-dependent Mcl-1 degradation, contributing to induction of apoptosis and enhancement of TRAIL-induced apoptosis. Cancer Lett. 338:229–238. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Embi N, Rylatt DB and Cohen P: Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase. Eur J Biochem. 107:519–527. 1980. View Article : Google Scholar : PubMed/NCBI

60 

Woodgett JR: Molecular cloning and expression of glycogen synthase kinase-3/factor A. EMBO J. 9:2431–2438. 1990.PubMed/NCBI

61 

Wu D and Pan W: GSK3: A multifaceted kinase in Wnt signaling. Trends Biochem Sci. 35:161–168. 2010. View Article : Google Scholar :

62 

Kim L and Kimmel AR: GSK3, a master switch regulating cell-fate specification and tumorigenesis. Curr Opin Genet Dev. 10:508–514. 2000. View Article : Google Scholar : PubMed/NCBI

63 

Buttrick GJ and Wakefield JG: PI3-K and GSK-3: Akt-ing together with microtubules. Cell Cycle. 7:2621–2625. 2008. View Article : Google Scholar : PubMed/NCBI

64 

Welcker M, Singer J, Loeb KR, Grim J, Bloecher A, Gurien-West M, Clurman BE and Roberts JM: Multisite phosphorylation by Cdk2 and GSK3 controls cyclin E degradation. Mol Cell. 12:381–392. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Hao B, Oehlmann S, Sowa ME, Harper JW and Pavletich NP: Structure of a Fbw7-Skp1-cyclin E complex: Multisite-phosphorylated substrate recognition by SCF ubiquitin ligases. Mol Cell. 26:131–143. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Bahram F, von der Lehr N, Cetinkaya C and Larsson LG: c-Myc hot spot mutations in lymphomas result in inefficient ubiquitination and decreased proteasome-mediated turnover. Blood. 95:2104–2110. 2000.PubMed/NCBI

67 

Tan Y, Sangfelt O and Spruck C: The Fbxw7/hCdc4 tumor suppressor in human cancer. Cancer Lett. 271:1–12. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Strohmaier H, Spruck CH, Kaiser P, Won KA, Sangfelt O and Reed SI: Human F-box protein hCdc4 targets cyclin E for proteolysis and is mutated in a breast cancer cell line. Nature. 413:316–322. 2001. View Article : Google Scholar : PubMed/NCBI

69 

Ye X, Nalepa G, Welcker M, Kessler BM, Spooner E, Qin J, Elledge SJ, Clurman BE and Harper JW: Recognition of phosphodegron motifs in human cyclin E by the SCFFbw7 ubiquitin ligase. J Biol Chem. 279:50110–50119. 2004. View Article : Google Scholar : PubMed/NCBI

70 

Welcker M and Clurman BE: Fbw7/hCDC4 dimerization regulates its substrate interactions. Cell Div. 2:72007. View Article : Google Scholar : PubMed/NCBI

71 

Geng Y, Lee YM, Welcker M, Swanger J, Zagozdzon A, Winer JD, Roberts JM, Kaldis P, Clurman BE and Sicinski P: Kinase-independent function of cyclin E. Mol Cell. 25:127–139. 2007. View Article : Google Scholar : PubMed/NCBI

72 

Siu KT, Rosner MR and Minella AC: An integrated view of cyclin E function and regulation. Cell Cycle. 11:57–64. 2012. View Article : Google Scholar :

73 

Minella AC, Grim JE, Welcker M and Clurman BE: p53 and SCFFbw7 cooperatively restrain cyclin E-associated genome instability. Oncogene. 26:6948–6953. 2007. View Article : Google Scholar : PubMed/NCBI

74 

Minella AC, Swanger J, Bryant E, Welcker M, Hwang H and Clurman BE: p53 and p21 form an inducible barrier that protects cells against cyclin E-cdk2 deregulation. Curr Biol. 12:1817–1827. 2002. View Article : Google Scholar : PubMed/NCBI

75 

Kimura T, Gotoh M, Nakamura Y and Arakawa H: hCDC4b, a regulator of cyclin E, as a direct transcriptional target of p53. Cancer Sci. 94:431–436. 2003. View Article : Google Scholar : PubMed/NCBI

76 

Mandal S, Freije WA, Guptan P and Banerjee U: Metabolic control of G1-S transition: Cyclin E degradation by p53-induced activation of the ubiquitin-proteasome system. J Cell Biol. 188:473–479. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Finkin S, Aylon Y, Anzi S, Oren M and Shaulian E: Fbw7 regulates the activity of endoreduplication mediators and the p53 pathway to prevent drug-induced polyploidy. Oncogene. 27:4411–4421. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Guo Z, Zhou Y, Evers BM and Wang Q: Rictor regulates FBXW7-dependent c-Myc and cyclin E degradation in colorectal cancer cells. Biochem Biophys Res Commun. 418:426–432. 2012. View Article : Google Scholar : PubMed/NCBI

79 

Wang H, Zhang X, Geng L, Teng L and Legerski RJ: Artemis regulates cell cycle recovery from the S phase checkpoint by promoting degradation of cyclin E. J Biol Chem. 284:18236–18243. 2009. View Article : Google Scholar : PubMed/NCBI

80 

Welcker M and Clurman BE: The SV40 large T antigen contains a decoy phosphodegron that mediates its interactions with Fbw7/hCdc4. J Biol Chem. 280:7654–7658. 2005. View Article : Google Scholar

81 

Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, Erdjument-Bromage H, Tempst P and Sabatini DM: Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol. 14:1296–1302. 2004. View Article : Google Scholar : PubMed/NCBI

82 

Poinsignon C, de Chasseval R, Soubeyrand S, Moshous D, Fischer A, Haché RJ and de Villartay JP: Phosphorylation of Artemis following irradiation-induced DNA damage. Eur J Immunol. 34:3146–3155. 2004. View Article : Google Scholar : PubMed/NCBI

83 

Ahuja D, Sáenz-Robles MT and Pipas JM: SV40 large T antigen targets multiple cellular pathways to elicit cellular transformation. Oncogene. 24:7729–7745. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Minella AC, Welcker M and Clurman BE: Ras activity regulates cyclin E degradation by the Fbw7 pathway. Proc Natl Acad Sci USA. 102:9649–9654. 2005. View Article : Google Scholar : PubMed/NCBI

85 

Hynes NE and Lane HA: ERBB receptors and cancer: The complexity of targeted inhibitors. Nat Rev Cancer. 5:341–354. 2005. View Article : Google Scholar : PubMed/NCBI

86 

Tan Y, Sun D, Jiang W, Klotz-Noack K, Vashisht AA, Wohlschlegel J, Widschwendter M and Spruck C: PP2A-B55β antagonizes cyclin E1 proteolysis and promotes its dysregulation in cancer. Cancer Res. 74:2006–2014. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Bhaskaran N, van Drogen F, Ng HF, Kumar R, Ekholm-Reed S, Peter M, Sangfelt O and Reed SI: Fbw7α and Fbw7γ collaborate to shuttle cyclin E1 into the nucleolus for multiubiquitylation. Mol Cell Biol. 33:85–97. 2013. View Article : Google Scholar :

88 

Reed SI: Cooperation between different Cdc4/Fbw7 isoforms may be associated with 2-step inactivation of SCFCdc4 targets. Cell Cycle. 5:1923–1924. 2006. View Article : Google Scholar : PubMed/NCBI

89 

Zhang W, MacDonald EM and Koepp DM: The stomatin-like protein SLP-1 and Cdk2 interact with the F-Box protein Fbw7-γ. PLoS One. 7:e477362012. View Article : Google Scholar

90 

Schülein C, Eilers M and Popov N: PI3K-dependent phosphorylation of Fbw7 modulates substrate degradation and activity. FEBS Lett. 585:2151–2157. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Sim KG, Zang Z, Yang CM, Bonventre JV and Hsu SI: TRIP-Br links E2F to novel functions in the regulation of cyclin E expression during cell cycle progression and in the maintenance of genomic stability. Cell Cycle. 3:1296–1304. 2004. View Article : Google Scholar : PubMed/NCBI

92 

Cizmecioglu O, Krause A, Bahtz R, Ehret L, Malek N and Hoffmann I: Plk2 regulates centriole duplication through phosphorylation-mediated degradation of Fbxw7 (human Cdc4). J Cell Sci. 125:981–992. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Mansour MR, Sanda T, Lawton LN, Li X, Kreslavsky T, Novina CD, Brand M, Gutierrez A, Kelliher MA, Jamieson CH, et al: The TAL1 complex targets the FBXW7 tumor suppressor by activating miR-223 in human T cell acute lymphoblastic leukemia. J Exp Med. 210:1545–1557. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Keck JM, Summers MK, Tedesco D, Ekholm-Reed S, Chuang LC, Jackson PK and Reed SI: Cyclin E overexpression impairs progression through mitosis by inhibiting APCCdh1. J Cell Biol. 178:371–385. 2007. View Article : Google Scholar : PubMed/NCBI

95 

Lau AW, Inuzuka H, Fukushima H, Wan L, Liu P, Gao D, Sun Y and Wei W: Regulation of APCCdh1 E3 ligase activity by the Fbw7/cyclin E signaling axis contributes to the tumor suppressor function of Fbw7. Cell Res. 23:947–961. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Meyer N and Penn LZ: Reflecting on 25 years with MyC. Nat Rev Cancer. 8:976–990. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Onoyama I, Tsunematsu R, Matsumoto A, Kimura T, de Alborán IM, Nakayama K and Nakayama KI: Conditional inactivation of Fbxw7 impairs cell-cycle exit during T cell differentiation and results in lymphomatogenesis. J Exp Med. 204:2875–2888. 2007. View Article : Google Scholar : PubMed/NCBI

98 

Welcker M, Orian A, Jin J, Grim JE, Harper JW, Eisenman RN and Clurman BE: The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation. Proc Natl Acad Sci USA. 101:9085–9090. 2004. View Article : Google Scholar : PubMed/NCBI

99 

Sears R, Nuckolls F, Haura E, Taya Y, Tamai K and Nevins JR: Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 14:2501–2514. 2000. View Article : Google Scholar : PubMed/NCBI

100 

Yeh E, Cunningham M, Arnold H, Chasse D, Monteith T, Ivaldi G, Hahn WC, Stukenberg PT, Shenolikar S, Uchida T, et al: A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol. 6:308–318. 2004. View Article : Google Scholar : PubMed/NCBI

101 

Arnold HK, Zhang X, Daniel CJ, Tibbitts D, Escamilla-Powers J, Farrell A, Tokarz S, Morgan C and Sears RC: The Axin1 scaffold protein promotes formation of a degradation complex for c-Myc. EMBO J. 28:500–512. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Liu L and Eisenman RN: Regulation of c-Myc protein abundance by a protein phosphatase 2A-glycogen synthase kinase 3β-negative feedback pathway. Genes Cancer. 3:23–36. 2012. View Article : Google Scholar : PubMed/NCBI

103 

Seo HR, Kim J, Bae S, Soh JW and Lee YS: Cdk5-mediated phosphorylation of c-Myc on Ser-62 is essential in transcriptional activation of cyclin B1 by cyclin G1. J Biol Chem. 283:15601–15610. 2008. View Article : Google Scholar : PubMed/NCBI

104 

Sjostrom SK, Finn G, Hahn WC, Rowitch DH and Kenney AM: The Cdk1 complex plays a prime role in regulating N-myc phosphorylation and turnover in neural precursors. Dev Cell. 9:327–338. 2005. View Article : Google Scholar : PubMed/NCBI

105 

Junttila MR, Puustinen P, Niemelä M, Ahola R, Arnold H, Böttzauw T, Ala-aho R, Nielsen C, Ivaska J, Taya Y, et al: CIP2A inhibits PP2A in human malignancies. Cell. 130:51–62. 2007. View Article : Google Scholar : PubMed/NCBI

106 

Bonetti P, Davoli T, Sironi C, Amati B, Pelicci PG and Colombo E: Nucleophosmin and its AML-associated mutant regulate c-Myc turnover through Fbw7 gamma. J Cell Biol. 182:19–26. 2008. View Article : Google Scholar : PubMed/NCBI

107 

Welcker M, Orian A, Grim JE, Eisenman RN and Clurman BE: A nucleolar isoform of the Fbw7 ubiquitin ligase regulates c-Myc and cell size. Curr Biol. 14:1852–1857. 2004. View Article : Google Scholar : PubMed/NCBI

108 

Chandra S, Priyadarshini R, Madhavan V, Tikoo S, Hussain M, Mudgal R, Modi P, Srivastava V and Sengupta S: Enhancement of c-Myc degradation by BLM helicase leads to delayed tumor initiation. J Cell Sci. 126:3782–3795. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Tikoo S and Sengupta S: Time to bloom. Genome Integr. 1:142010. View Article : Google Scholar : PubMed/NCBI

110 

Kim BY, Yang JS, Kwak SY, Zhang XK and Han YH: NEMO stabilizes c-Myc through direct interaction in the nucleus. FEBS Lett. 584:4524–4530. 2010. View Article : Google Scholar : PubMed/NCBI

111 

Huang H, Ma L, Li J, Yu Y, Zhang D, Wei J, Jin H, Xu D, Gao J and Huang C: NF-κB1 inhibits c-Myc protein degradation through suppression of FBW7 expression. Oncotarget. 5:493–505. 2014. View Article : Google Scholar : PubMed/NCBI

112 

Chen J, Shin JH, Zhao R, Phan L, Wang H, Xue Y, Post SM, Ho Choi H, Chen JS, Wang E, et al: CSN6 drives carcinogenesis by positively regulating Myc stability. Nat Commun. 5:53842014. View Article : Google Scholar : PubMed/NCBI

113 

Olive V, Sabio E, Bennett MJ, De Jong CS, Biton A, McGann JC, Greaney SK, Sodir NM, Zhou AY, Balakrishnan A, et al: A component of the mir-17-92 polycistronic oncomir promotes oncogene-dependent apoptosis. eLife. 2:e008222013. View Article : Google Scholar : PubMed/NCBI

114 

Onoyama I, Suzuki A, Matsumoto A, Tomita K, Katagiri H, Oike Y, Nakayama K and Nakayama KI: Fbxw7 regulates lipid metabolism and cell fate decisions in the mouse liver. J Clin Invest. 121:342–354. 2011. View Article : Google Scholar :

115 

Tu K, Zheng X, Zan X, Han S, Yao Y and Liu Q: Evaluation of Fbxw7 expression and its correlation with the expression of c-Myc, cyclin E and p53 in human hepatocellular carcinoma. Hepatol Res. 42:904–910. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Tu K, Zheng X, Zhou Z, Li C, Zhang J, Gao J, Yao Y and Liu Q: Recombinant human adenovirus-p53 injection induced apoptosis in hepatocellular carcinoma cell lines mediated by p53-Fbxw7 pathway, which controls c-Myc and cyclin E. PLoS One. 8:e685742013. View Article : Google Scholar : PubMed/NCBI

117 

Imura S, Tovuu LO, Utsunomiya T, Morine Y, Ikemoto T, Arakawa Y, Kanamoto M, Iwahashi S, Saito Y, Takasu C, et al: The role of Fbxw7 expression in hepatocellular carcinoma and adjacent non-tumor liver tissue. J Gastroenterol Hepatol. 29:1822–1829. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Tien JC and Xu J: Steroid receptor coactivator-3 as a potential molecular target for cancer therapy. Expert Opin Ther Targets. 16:1085–1096. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Chen T, Sun Y, Ji P, Kopetz S and Zhang W: Topoisomerase IIα in chromosome instability and personalized cancer therapy. Oncogene. Oct 20–2014.Epub ahead of print. View Article : Google Scholar

120 

Piccolo S, Dupont S and Cordenonsi M: The biology of YAP/TAZ: Hippo signaling and beyond. Physiol Rev. 94:1287–1312. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Lahusen T, Henke RT, Kagan BL, Wellstein A and Riegel AT: The role and regulation of the nuclear receptor co-activator AIB1 in breast cancer. Breast Cancer Res Treat. 116:225–237. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 34 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou Z, He C and Wang J: Regulation mechanism of Fbxw7-related signaling pathways (Review). Oncol Rep 34: 2215-2224, 2015
APA
Zhou, Z., He, C., & Wang, J. (2015). Regulation mechanism of Fbxw7-related signaling pathways (Review). Oncology Reports, 34, 2215-2224. https://doi.org/10.3892/or.2015.4227
MLA
Zhou, Z., He, C., Wang, J."Regulation mechanism of Fbxw7-related signaling pathways (Review)". Oncology Reports 34.5 (2015): 2215-2224.
Chicago
Zhou, Z., He, C., Wang, J."Regulation mechanism of Fbxw7-related signaling pathways (Review)". Oncology Reports 34, no. 5 (2015): 2215-2224. https://doi.org/10.3892/or.2015.4227