Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jul 1.
Published in final edited form as: Pharmacol Ther. 2015 Apr 4;151:141–151. doi: 10.1016/j.pharmthera.2015.04.002

Targeting Cdc20 as a novel cancer therapeutic strategy

Lixia Wang a,#, Jinfang Zhang b,#, Lixin Wan b,#, Xiuxia Zhou a, Zhiwei Wang a,*, Wenyi Wei b,*
PMCID: PMC4457591  NIHMSID: NIHMS678202  PMID: 25850036

Abstract

The Anaphase Promoting Complex (APC, also called APC/C) regulates cell cycle progression by forming two closely related, but functionally distinct E3 ubiquitin ligase sub-complexes, APCCdc20 and APCCdh1, respectively. Emerging evidence has begun to reveal that Cdc20 and Cdh1 have opposing functions in tumorigenesis. Specifically, Cdh1 functions largely as a tumor suppressor, whereas Cdc20 exhibits an oncogenic function, suggesting that Cdc20 could be a promising therapeutic target for combating human cancer. However, the exact underlying molecular mechanisms accounting for their differences in tumorigenesis remain largely unknown. Therefore, in this review, we summarize the downstream substrates of Cdc20 and the critical functions of Cdc20 in cell cycle progression, apoptosis, ciliary disassembly and brain development. Moreover, we briefly describe the upstream regulators of Cdc20 and the oncogenic role of Cdc20 in a variety of human malignancies. Furthermore, we summarize multiple pharmacological Cdc20 inhibitors including TAME and Apcin, and their potential clinical benefits. Taken together, development of specific Cdc20 inhibitors could be a novel strategy for the treatment of human cancers with elevated Cdc20 expression.

Keywords: Cancer, Cdc20, SCF, E3 ligase, Drug, Target

1. Introduction

Ubiquitination has been characterized to play a critical role in regulating diverse cellular processes including cell cycle progression, cell proliferation, apoptosis, DNA damage, migration and invasion (Hoeller et al., 2006; Nakayama & Nakayama, 2006). It has been well accepted that ubiquitination by the ubiquitin proteasome system (UPS) is a post-translational modification that controls protein degradation thereby the abundance of essential proteins involved in a plethora of cellular processes (Bassermann et al., 2014; Lipkowitz & Weissman, 2011; Wang, Z. et al., 2014). A wealth of evidence has emerged that two related, multi-subunit E3 ubiquitin ligase enzymes, the Anaphase Promoting Complex (APC) and the Skp1-Cullin1-F-box complex (SCF) have been considered as the major driving forces governing cell cycle progression (Lau et al., 2012; Wang et al., 2012; Wang, Z. et al., 2014; Zhang, J. et al., 2014). Notably, APC is the most complex E3 ubiquitin ligase that consists of at least 14 subunits (namely, APC1/TSG24, APC2, APC3/Cdc27, APC4, APC5, APC6/Cdc6, APC7, APC8/Cdc23, APC10/Doc1, APC11, APC13/SWM1, APC15/Mnd2, APC16, and Cdc26) and either one of two co-activators, Cdh1 or Cdc20 (Chang & Barford, 2014; Foe & Toczyski, 2011; Schreiber et al., 2011). Due to its large size and complex nature, the structure of the full APC holoenzyme remained poorly understood until recently, when its structure was elucidated by the Cryo-electron microscopy technology (Chang & Barford, 2014; Chang et al., 2014; Kulkarni et al., 2013). These structural insights support the model that the APC consists of a scaffolding subunit (including APC1, APC4, APC5), a catalytic and substrate recognition subunit (APC2, APC11, APC10), a tetratricopeptide repeat arm (APC3, APC6, APC8), and an accessory subunit (APC13, Cdc26, APC16) (Figure 1) (McLean et al., 2011; Vodermaier et al., 2003). Without a doubt, it is necessary to further determine the architectural details of the APC to aid in further understanding its biological functions.

Figure 1. A schematic illustration of the APC ubiquitin E3 ligase complex.

Figure 1

The APC core complex includes a scaffolding subunit (APC1, APC4, APC5), a catalytic and substrate recognition subunit (APC2, APC11, APC10), a tetratricopeptide repeat arm (APC3, APC6, APC8), and an accessory subunit (APC13, Cdc26, APC16).

To exert its biological functions, the APC core is associated with two activators, Cdc20 (cell division cycle 20 homologue, also called Fizzy) and Cdh1 (Cdc20 homologue 1, also known as Fizzy-related protein 1, FZR1), respectively, leading to two distinct E3 ubiquitin ligase complexes, APCCdc20 and APCCdh1 (Penas et al., 2011; Wang, Z. et al., 2013). Cdc20 contains seven WD40 repeats that are necessary for mediating protein-protein interactions (Hartwell et al., 1973). Emerging evidence has also revealed that Cdc20 and Cdh1 control the substrate specificity of the APC core-complex to bind and ubiquitinate target proteins for subsequent degradation. Notably, it has been demonstrated that Cdc20 and Cdh1 recruit their substrates via different motifs. For example, APCCdc20 typically targets its substrates which contain a Destruction-box (D-box) (Clute & Pines, 1999; Michaelis et al., 1997; Nasmyth, 2001), TEK (Jin et al., 2008) or the newly identified ABBA (Di Fiore et al., 2015) motifs (Table 1). On the other hand, APCCdh1 largely recruits substrates with either KEN-box (McGarry & Kirschner, 1998; Petersen et al., 2000), D-box (Bashir et al., 2004; den Elzen & Pines, 2001; Geley et al., 2001; Lindon & Pines, 2004; McGarry & Kirschner, 1998; Petersen et al., 2000; Wei et al., 2004), A-box (Littlepage & Ruderman, 2002), O-box (Araki et al., 2003), CRY box (Reis et al., 2006), LLK (Gao et al., 2009) or GxEN box (Castro et al., 2003) motifs (Table 1). It is still not fully understood how APCCdc20 and APCCdh1 mechanistically recruit their substrates with different motifs, but it provides a possible molecular explanation for their distinct roles in tumorigenesis that might stem from their abilities in targeting a different spectrum of substrates for destruction.

Table 1. The different types degrons that are recognized by Cdh1 and Cdc20.

APCCdh1 APCCdc20

Degron Consensus Representative Substrates References Degron Consensus Representative Substrates References
D-box RxxLx(2-5)N/D/E Cyclin A2, Plk1, Skp2, Geminin, Cdc6 (Bashir et al., 2004; den Elzen & Pines, 2001; Geley et al., 2001; Lindon & Pines, 2004; McGarry & Kirschner, 1998; Petersen et al., 2000; Wei et al., 2004) D-box RxxLx(2-5)N/D/E Cyclin B1, Securin, etc. (Clute & Pines, 1999; Michaelis et al., 1997; Nasmyth, 2001)
KEN box KENxxD/Q/E/N Geminin, Cdc6 (McGarry & Kirschner, 1998; Petersen et al., 2000) TEK TEK Securin (Jin et al., 2008)
A box QRVL Aurora A, Aurora B (Littlepage & Ruderman, 2002) ABBA Motif KxxFxxYxDxxE Cyclin A1, Cyclin A2 (Di Fiore et al., 2015)
O box paspLtekNak ORC1 (Araki et al., 2003) LXPKXLF Motif LXPKXLF PHF8 (Lim et al., 2013)
GxEN GxEN X-kid (Castro et al., 2003) LR Motif LR Kif18A (Sedgwick et al., 2013)
LLK LLK Claspin (Gao et al., 2009) MR Motif MR Nek2A (Sedgwick et al., 2013)
CRY CRY Cdc20 (Reis et al., 2006)

Consistent with this notion, although both Cdc20 and Cdh1 can activate the APC E3 ligase, they have distinct biological functions (Clijsters et al., 2013; Yu, 2002). For example, APCCdc20 exerts its function during the metaphase to anaphase transition through destruction of critical cell cycle regulators (Kim & Yu, 2011; Yu, 2007), whereasAPCCdh1 plays a key role in the late M and G1 phases (Hu et al., 2011; Qiao et al., 2010). Moreover, Cdh1 is considered as a tumor suppressor, while Cdc20 exhibits its oncogenic function (Penas et al., 2011; Wang, Z. et al., 2013). It is known that Cdc20 is an essential developmental gene, whose disruption in mice caused embryonic lethality and displayed condensed chromosomes, in part due to aberrant mitotic arrest (Li et al., 2007). Consistently, ablation of endogenous Cdc20 blocks in vivo tumorigenesis in a skin-tumor mouse model induced by a two-stage carcinogenesis protocol, largely due to elevated cellular apoptosis (Manchado et al., 2010). Furthermore, depleting endogenous Cdc20 in various cancer cell lines also led to a mitotic arrest followed by cell death. Together, these studies suggest that inhibition of APCCdc20 enzymatic activity might lead to an elevated cellular apoptosis. Although the exact molecular mechanism underlying Cdc20 loss-induced apoptosis remains unknown, these studies strongly argue for Cdc20 as a novel anti-cancer therapeutic drug target. Indeed, inactivating APC by an IR-mimetic inhibitor, pro-TAME, which targets both APCCdc20 and APCCdh1, also induced cell death in multiple cancer cell lines (Zeng et al., 2010). Therefore, in this article, we summarize the oncogenic role of Cdc20 in a variety of human cancers including pancreatic cancer, breast cancer, prostate cancer, colorectal cancer, lung cancer, glioblastomas, bladder, hepatocellular carcinoma and other cancers. Moreover, we discuss how aberrant overexpression of Cdc20 in various types of human cancers could be used to guide the development and use of Cdc20 inhibitors for treating human cancers. Finally, we describe several Cdc20 inhibitors and their potential clinical benefits.

2. Cdc20 exerts its biological functions largely by targeting its downstream substrates for ubiquitination and subsequent degradation

In recent years, many downstream targets of Cdc20 have been identified by various groups (Table 2). The initial role of Cdc20 was elucidated primarily in regulating cell cycle progression after it was discovered nearly half a century ago (Hartwell et al., 1970). Cells with Cdc20 mutants blocked cell division and stopped cell cycle progression toward anaphase and chromosome segregation (Hartwell et al., 1970). Mechanistically, many identified substrates of Cdc20 are involved in mitotic procession including Securin (Zur & Brandeis, 2001), Cyclin B1 (Lim et al., 1998; Shirayama et al., 1999), Cyclin A (Geley et al., 2001; Ohtoshi et al., 2000), Nek2A (Hames et al., 2001), Cenp-F (Gurden et al., 2010) and p21 (Amador et al., 2007). Further studies implicated Cdc20 in governing cellular apoptosis through regulating the stability of Mcl-1 (Harley et al., 2010) and Bim (Wan et al., 2014). Interestingly, Cdc20 has also been reported to play a key role in ciliary disassembly (Wang, W. et al., 2014) and brain development (Yang et al., 2007; Yang et al., 2009). In the following sections, we will summarize the different biological functions of Cdc20 in cell cycle progression, apoptosis, ciliary disassembly and brain development.

Table 2. Summary of the identified ubiquitination substrates for APCCdc20.

Substrates Functions/signaling pathways of substrates Coactivators References
Bard1 Controls spindle pole formation Cdc20/Cdh1 (Song & Rape, 2010)
Bim Plays key roles in regulating apoptosis Cdc20 (Wan et al., 2014)
Cenp-F Functions in kinetochore and chromosome segregation in mitosis Cdc20 (Gurden et al., 2010)
Conductin Inhibits the Wnt signaling pathway Cdc20 (Hadjihannas et al., 2012)
Cyclin A Controls S phase and G2/M transition Cdc20/Cdh1 (den Elzen & Pines, 2001; Geley et al., 2001)
Cyclin B Activates Cdk1 and controls the G2/M transition Cdc20/Cdh1 (Clute & Pines, 1999)
E2F1 Governs G1/S transition and apoptosis Cdc20/Cdh1 (Budhavarapu et al., 2012)
Hmmr Regulates the localization of Tpx2 at the spindle pole Cdc20/Cdh1 (Song & Rape, 2010)
HURP Nucleates and crosslinks microtubules in the vicinity of chromatin Cdc20/Cdh1 (Song & Rape, 2010)
Id1 Inhibits dendrite growth Cdc20 (Kim, A. H. et al., 2009)
Kif18A Plays a role in chromosome congression Cdc20 (Sedgwick et al., 2013)
Mcl-1 Anti-apoptotic protein Cdc20 (Harley et al., 2010)
Mps1 Regulates the spindle assembly checkpoint and chromosome-microtubule attachments Cdc20/Cdh1 (Cui et al., 2010)
Nek1 Functions in primary cilium formation Cdc20 (Shalom et al., 2008)
Nek2A Regulates centrosome separation and spindle formation Cdc20 (Hames et al., 2001; Hayes et al., 2006)
NeuroD2 Inhibits presynaptic differentiation Cdc20 (Yang et al., 2009)
Nlp Functions in centrosome maturation Cdc20/Cdh1 (Wang & Zhan, 2007)
NuSAP Nucleates and crosslinks microtubules in the vicinity of chromatin Cdc20/Cdh1 (Song & Rape, 2010)
PHF8 Activates gene transcription by demethylating histon H3 and H4 Cdc20 (Lim et al., 2013)
p21Cip1 Inhibits cyclin-dependent kinase activity Cdc20 (Amador et al., 2007)
RAP80 Recruits BRCA1 to DNA damage sites Cdc20/Cdh1 (Cho et al., 2012)
REV1 Functions on replicating across DNA lesions at the stalled replication fork Cdc20/Cdh1 (Chun et al., 2013)
Securin Inhibits separase activity Cdc20/Cdh1 (Michaelis et al., 1997; Nasmyth, 2001)
Sp100 Participates in viral resistance, transcriptional regulation and apoptosis Cdc20 (Wang et al., 2011)
TRRAP Histone acetyltransferase complex component Cdc20/Cdh1 (Ichim et al., 2013)

2.1. Regulation of cell cycle

Different from APCCdh1 with major functions in late M and G1 phases, APCCdc20 plays an indispensable role during the metaphase to anaphase transition by targeting critical cell cycle regulators including Securin (Michaelis et al., 1997; Nasmyth, 2001) and Cyclin B (Clute & Pines, 1999) for ubiquitination-mediated destruction. It has been also identified that Cdc20 binds p21 in a D-box motif-dependent manner to promote the timely degradation of p21 in prometaphase, whereas Skp2 degrades p21 during the G1/S transition (Amador et al., 2007). Another study also proposed that Cdc20-mediated degradation of conductin governs Wnt/β-catenin signaling and controls the cell cycle (Hadjihannas et al., 2012). In line with this finding, Cdc20-resistant conductin blocked Wnt signaling and inhibited colony formation of colorectal cancer cells (Hadjihannas et al., 2012). More recently, one elegant study revealed that APCCdc20 controls cell cycle through temporal degradation of Nek2A and Kif18A (Sedgwick et al., 2013). Notably, Cdc20-mediated Kif18A degradation depends on a C-terminal LR motif, whereas degradation of Nek2A by APCCdc20 depends on an MR motif (Sedgwick et al., 2013). Additionally, Lim et al. found that the ubiquitination and degradation of the histone-demethylase PHF8 is also regulated by APCCdc20, whereas depletion of endogenous PHF8 led to prolonged G2 phase and defective mitosis. Interestingly, PHF8 contains a unique LXPKXLF motif that is required for binding to Cdc20 (Lim et al., 2013), but further studies are required to demonstrate whether other Cdc20 substrates also possess this degron. Moreover, Song et al. found that the APCCdc20 complex promoted the degradation of four proteins that are required for spindle assembly including Bard1, HURP, NuSAP, and Hmmr (Song & Rape, 2010), further expanding the role of Cdc20 in spindle checkpoint assembly. On the other hand, Cho et al. discovered that APCCdc20 controls mitotic progression via targeting RAP80 (receptor-associated protein 80) (Cho et al., 2012). Given a critical role of RAP80 in DNA damage repair pathway, it suggests a possible role of Cdc20 in DNA damage repair pathway by linking mitotic regulation with chromosome stability control. To this end, it has also been recently demonstrated that degradation of TRRAP (TRansformation/TRanscription domain-Associated Protein) by APCCdc20 is required for a proper condensation of chromatin and chromosome segregation to govern the faithful segregation of duplicated DNA strands (Ichim et al., 2014). Taken together, identifying additional ubiquitin substrates will help us to better appreciate the molecular basis of the essential role of APCCdc20 in timely regulation of cell cycle progression.

2.2. Regulation of apoptosis

In addition to regulating mitotic progression, Cdc20 has been implicated in the regulation of other cellular processes such as apoptosis through targeting Mcl-1 (Harley et al., 2010) and Bim (Wan et al., 2014). It is well known that the Bcl-2 protein family plays a critical role in the apoptotic signaling pathway (Cory & Adams, 2002). The Bcl-2 family of proteins are classified into anti-apoptotic group and pro-apoptotic members (Cragg et al., 2009). The anti-apoptotic members contain Bcl-2, Bcl-xL, Bcl-w and Mcl-1, while the pro-apoptotic Bcl-2 members include Bax, Bak and BH3-only proteins such as Bim (Cory & Adams, 2002). Initiating the spindle assembly checkpoint by Taxol or Nocodazole, largely through Mad2-dependent suppression of APCCdc20 has been used as anti-cancer treatments (Janssen & Medema, 2011). Consistently, depleting Cdc20 or pharmacological inhibition of APC leads to elevated cellular apoptosis (Manchado et al., 2010), but the underlying molecular mechanism remains unclear. To this end, we recently reported that Cdc20 governs apoptosis largely through controlling the ubiquitination and stability of the pro-apoptotic protein, Bim (Wan et al., 2014). Notably, the pro-apoptotic protein Bim has attracted increasing attention as a pivotal regulator of apoptosis. The Bim expression level is controlled at both transcriptional and post-transcriptional levels in a cell- and tissuespecific manner (Akiyama et al., 2009). Moreover, Bim knockout mice exhibit a systematic lupus erythematosus-like autoimmune disease along with an abnormal accumulation of hematopoietic cells, suggesting that Bim is involved in the regulation of hematopoietic cells and the immune system (Akiyama & Tanaka, 2011). Further studies have also revealed critical roles for Bim in bone homeostasis and tumorigenesis, in part due to its critical contribution to trigger apoptosis (Akiyama & Tanaka, 2011).

Several E3 ligases have been reported to negatively regulate Bim stability. For example, c-Cbl and the ElonginB/C-Cullin2-CIS E3 ligase have been found to promote Bim degradation (Thien et al., 2010; Zhang et al., 2008). However, controversial results have been reported by different groups, thereby refuting a possible physiological role of either c-Cbl or CIS in Bim ubiquitination (Akiyama et al., 2003; Wiggins et al., 2007). Recently, it has been reported that Bim is degraded by β-TRCP (Moustafa-Kamal et al., 2013). However, as β-TRCP-mediated destruction of Bim required prior-phosphorylation, which is restricted to late G1 phase and is not relevant to the chemo-resistance associated with Taxol treatment (Dehan et al., 2009). On the other hand, we reported that APCCdc20 is a physiological E3 ligase that governs the ubiquitination and destruction of Bim (Wan et al., 2014). In support of this concept, we provided evidence to validate Bim as an APCCdc20 ubiquitin substrate. First, Bim abundance displayed a dramatic reduction during mitosis when APCCdc20 is most active. Second, depletion of Cdc20 in various cell lines led to a significant upregulation of Bim abundance. Third, Bim was stabilized in Cdc20-depleted M phase cells. Fourth, Cdc20 specifically interacts with Bim through its C-terminal WD40 repeats motifs. Fifth, Bim contains two evolutionarily conserved D boxes. As such, Cdc20 promotes Bim ubiquitination and subsequent degradation in a D-box-dependent manner. More importantly, we pinpointed the physiological function of Cdc20-mediated Bim degradation. Specifically, we found that hyperactive Cdc20 contributes to chemoresistance by promoting Bim destruction in HTLV-1 positive adult T-cell leukemia cells. Notably, depletion of Cdc20 induced chemoradiation sensitization in head and neck cancer cells (Wan et al., 2014). These studies therefore reveal that Cdc20 plays a crucial role in promoting the survival of cancer cells through inhibiting apoptosis and that Cdc20 allows the acquisition of chemo- or radioresistance partly by enhancing Bim destruction to evade apoptosis triggered by chemotherapeutic agents and/or irradiation. It has been suggested that anti-mitotic reagents that were used as anti-cancer agents activate the Spindle Assembly Checkpoint (SAC) primarily by suppressing APCCdc20, and induce apoptosis after prolonged-treatment (Huang et al., 2009; Wan et al., 2014). Interestingly, one excellent study recently demonstrated that the mitotic checkpoint complex binds a second Cdc20 to inhibit active APC and this is essential for the SAC (Izawa & Pines, 2015). It is important to note that Cdc20 could induce apoptosis through promoting the degradation of anti-apoptotic protein Mcl-1 (Harley et al., 2010). Another study suggests that Cdc20 governs apoptosis via mediating the degradation of PC-PLC (phosphatidylcholine specific phospholipase C) in hepatocellular carcinoma cells (Chen et al., 2010). Altogether, it is eager to further mechanistically define the role of Cdc20 involved in the regulation of apoptosis in human cancer cells.

2.3 Brain development and other functions

Yang et al. identified that APCCdc20 triggered presynaptic differentiation through degradation of NeuroD2, suggesting a potential role for Cdc20 in neuronal connectivity and plasticity in the brain (Yang et al., 2007; Yang et al., 2009). Consistently, Kim et al. found that Cdc20 has an essential function in dendrite morphogenesis in postmitotic neurons (Kim, A. H. et al., 2009). Remarkably, HDAC6 (histone deacetylase 6) promoted the polyubiquitination of Cdc20, activated Cdc20 activity, and enhanced the differentiation of dendrites, suggesting that Cdc20 is involved in neuronal connectivity and plasticity (Kim, A. H. et al., 2009). In addition to its role in neuronal differentiation, Wang et al. discovered a novel role for APCCdc20 beyond cell cycle control and apoptosis, and implicated its function in ciliary disassembly in part through regulating Nek1 stability (Wang, W. et al., 2014). Furthermore, Chun et al. discovered that overexpression of Cdc20 enhanced polyubiquitination and proteosomal degradation of REV1, which is a specialized DNA polymerase for DNA repair (Chun et al., 2013). Notably, Sp100, which participates in viral resistance, transcriptional regulation, and apoptosis, was degraded by APCCdc20 (Wang et al., 2011). These recent studies thus support the notion that beyond its canonical role in cell cycle regulation, Cdc20 may participate in many other important cellular processes including neuronal differentiation, ciliary formation or DNA damage repair. However, further in-depth studies are required to understand whether these new functions depend on its E3 ligase role or not (Wan et al., 2011), and whether they are intrinsically linked to its pivotal cell cycle role.

3. Cdc20 is regulated by multiple upstream factors

In addition to the extensive research efforts in determining the downstream substrates of Cdc20, recent studies have begun to define the upstream regulators of Cdc20 (Fang et al., 1998; Kidokoro et al., 2008; Reimann, Freed, et al., 2001). Here, we summarize the upstream regulators of Cdc20, allowing the readers to fully appreciate the complicated Cdc20 regulatory network system. Notably, it has been reported that p53 negatively regulates Cdc20 expression, which is supported by the demonstration that overexpression of p53 inhibited Cdc20, whereas depletion of p53 induced Cdc20 expression (Kidokoro et al., 2008). Specifically, Cdc20 expression was inhibited by genotoxic stresses in a p53-dependent manner largely through CDE-CHR elements present in the Cdc20 promoter. Moreover, this study also indicates that p53 exerted its anti-tumor activity via the indirect regulation of Cdc20 (Kidokoro et al., 2008). Strikingly, DNA damage-induced p53 downregulated Cdc20 through direct binding to its promoter, leading to chromatin remodeling (Banerjee et al., 2009). Furthermore, the function of p53 in HSF1 (heat shock factor 1)-mediated mitotic regulation and genomic instability could also be through regulation of the interaction between Cdc20 and HSF1 (Kim, H. S. et al., 2009). Additionally, the spindle checkpoint protein Mad2 was found to inhibit APCCdc20 activity through forming a Mad2-Cdc20-APC complex to control anaphase initiation (Fang et al., 1998). Moreover, the F-box protein Emi1 (early mitotic inhibitor 1) controls mitosis via binding to APCCdc20 as a pseudo-Cdc20-substrate and inhibiting its E3 ligase activity (Reimann, Freed, et al., 2001; Reimann, Gardner, et al., 2001). Another study showed that Bub1 and Bub3 blocked mitosis through suppression of APCCdc20-mediated degradation of Pds1 and Cyclin B (Fraschini et al., 1999). Interestingly, USP44 (ubiquitin-specific protease 44) deubiquitinated Cdc20 and blocked premature activation of APC via stabilization of the APC-inhibitory Mad2-Cdc20 complex (Stegmeier et al., 2007). Notably, tumor suppressor RASSF1A was reported to also inhibit APCCdc20 and block degradation of Cyclins A and B at the spindle poles (Song et al., 2004). In addition to these regulatory mechanisms mediated by protein components, miR-449 and miR-494 have been discovered to regulate Cdc20 mRNA levels (Lize et al., 2010; Yamanaka et al., 2012). Hence, we are of the opinion that further in-depth exploration is required to determine more Cdc20 upstream regulators for understanding how Cdc20 function is misregulated in tumorigenesis.

4. Role of Cdc20 in human malignancies

Mounting evidence has revealed that Cdc20 plays an oncogenic role in human tumorigenesis. Overexpression of Cdc20 was observed in a variety of human tumors. Moreover, higher expression of Cdc20 is associated with clinicopathological parameters in various types of human cancers. Therefore, in the following sections, we will summarize the critical role of Cdc20 in a wide range of human cancers.

4.1. Pancreatic cancer

Pancreatic cancer, one of the most common malignancies, is the fourth leading cause of cancer-related death in the United States (Siegel et al., 2015). It is estimated that 48,960 Americans are expected to be diagnosed with pancreatic cancer and 40,560 people will die from this disease in the US in 2015 (Siegel et al., 2015). Currently, the 5-year relative survival rate for pancreatic cancer is only 7%. Pancreatic cancer has been shown to display Cdc20 overexpression at high frequencies. For example, over-expression of Cdc20 was detected in pancreatic tumor tissues compared with normal adjacent tissues from pancreatic cancer patients (Li et al., 2003). Interestingly, STK15, identified as a Cdc20-associated protein, was also overexpressed in 58% of pancreatic tumor tissues. However, STK15 level was not correlated with tumor size, differentiation, and metastasis (Li et al., 2003). In line with this finding, Chang et al. performed an excellent retrospective study and identified Cdc20 expression as a useful biomarker in pancreatic cancer prognosis (Chang et al., 2012). Specifically, Cdc20 expression is significantly higher in pancreatic tumor tissues than in chronic pancreatitis tissue and normal pancreatic tissues (Chang et al., 2012). Notably, high expression of Cdc20 was associated with poor differentiation and a lower 5-year recurrence-free survival rate (Chang et al., 2012). Consistently, depletion of Cdc20 suppressed cell growth in human pancreatic cancer cells and induced G2/M cell cycle arrest (Taniguchi et al., 2008). More importantly, depletion of Cdc20 led to enhanced cytotoxicity upon paclitaxel treatment, and increased effects of gamma-irradiation against pancreatic cancer cells (Taniguchi et al., 2008). Taken together, Cdc20 could be a useful marker of pancreatic cancer progression and a novel therapeutic target for the treatment of this deadly disease.

4.2. Breast cancer

Breast cancer is the most commonly diagnosed cancer and the second most lethal malignancy in women in the US. About 60,290 cases of female breast carcinoma are expected to be diagnosed in 2015 (Siegel et al., 2015). This disease is the leading cause of cancer death in women aged 20-59 years. These data suggest that early diagnosis and prevention are required to reduce mortality associated with this disease. To this end, some studies have demonstrated that Cdc20 plays an essential role in breast cancer progression. Specifically, Yuan et al. reported that the mRNA and protein levels of Cdc20 were significantly higher in breast cancer cells and high-grade primary breast cancer tissues (Yuan et al., 2006). In support of this notion, another independent study screened Cdc20 expression in 445 breast cancer patients with up to 20 years of follow-up and validated that Cdc20 is highly expressed in breast cancer patients (Karra et al., 2014). Importantly, overexpression of Cdc20 was associated with an aggressive course of breast cancer. Consistently, high expression of Cdc20 and securin are correlated to extremely poor outcome of breast cancer patients (Karra et al., 2014). However, it is important to recognize that further study is necessary to determine the oncogenic role of Cdc20 in breast tumorigenesis.

4.3. Prostate Cancer

Prostate cancer is the most frequently diagnosed tumors in men and the leading cause of cancer-related death followed by cancers of lung and bronchus in the US (Siegel et al., 2015). The treatment of prostate cancer has been improved due to the use of widespread prostate-specific antigen (PSA) testing for early detection of asymptomatic prostate cancer (Siegel et al., 2015), but there is an urgent need to develop new treatments for patients with castration resistance in late stage prostate cancers. To this end, several studies have identified that Cdc20 is involved in prostate tumorigenesis. One study has shown that Daxx, an APC inhibitor, interacts with Cdc20 and inhibits the degradation of APCCdc20 substrates, leading to a transient delay in mitotic progression and chromosome instability (Kwan et al., 2013). Consistently, Daxx is overexpressed in prostate cancer tissues and positively correlated with the Gleason score and metastasis (Kwan et al., 2013). Moreover, LATs (L-type amino acid transporters) protein was expressed at all stages of prostate cancer. Inhibition of LAT suppressed tumor growth, cell cycle progression, and metastasis partly through downregulation of M-phase cell cycle genes including Cdc20 and mTORC1 in prostate cancer (Wang, Q. et al., 2013). Another study has suggested that knockdown of COX-2 by shRNA or using pharmacological COX inhibitors inhibited prostate cancer cell proliferation and arrested cell cycle progression via suppressing several key proteins in the kinetochore/centromere assembly including Cdc20 (Bieniek et al., 2014). Surprisingly, there is no direct evidence to show the physiological role of Cdc20 in prostate cancer development and progression, which awaits further studies.

4.4. Lung cancer

Lung cancer is the most common cause of cancer death, which kills more than 158,000 people in the US this year (Siegel et al., 2015). The 5-year relative survival rate is currently 18% for lung cancer patients. Using spiral computed tomography for screening for early signs of lung cancer, the survival rate of lung cancer has been improved (Siegel et al., 2015). It has been documented that lung cancer can be categorized into two types including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). Multiple studies have indicated that Cdc20 is highly expressed, and could be a potential prognostic marker in human NSCLC (Kato et al., 2012). Notably, relatively higher expression of Cdc20 was associated with pleural invasion, and shorter 5-year overall survival in NSCLC in a gender-specific manner (male specificity) (Kato et al., 2012). In line with this notion, deregulation of Cdc20 was observed in lung adenocarcinoma tissue samples (Zhang, W. et al., 2014). Notably, knockdown of Cdc20 inhibited cell growth, induced G2/M cell cycle arrest, and retarded colony formation of lung cancer cells (Kidokoro et al., 2008). However, the exact molecular mechanism of Cdc20-mediated lung tumorigenesis is still elusive and needs to be further explored.

4.5. Colorectal cancer

Colorectal cancer is the second leading cause of cancer death in the United States (Siegel et al., 2015). Nearly 49,700 Americans are anticipated to die from this deadly disease in 2015 (Siegel et al., 2015). Colorectal cancer has been known to contain numerous genetic and physiological alterations, leading to enhanced cell growth and survival (Kuipers et al., 2013). Emerging evidence has revealed that Cdc20 is overexpressed in colorectal cancer cell lines and primary cancer tissues compared with normal colon epithelial cells and adjacent noncancerous tissue samples, respectively (Wu et al., 2013). Remarkably, Cdc20 expression was associated with clinical stage, metastasis, and shorter overall survival, suggesting Cdc20 could serve as an independent prognostic biomarker for human colorectal cancer (Wu et al., 2013). In support of this finding, another independent study also identified that expression of Cdc20 was increased by more than five-fold in 77% of colorectal cancer tissues (Kidokoro et al., 2008). In line with this finding, depletion of Cdc20 inhibited Wnt signaling via upregulation of conductin. Moreover, Cdc20-resistant conductin suppressed Wnt signaling and subsequently attenuated colony formation of colorectal cancer cells (Hadjihannas et al., 2012). Interestingly, in a separate study, it has also been observed that expression of Cdc20 was decreased in the human colorectal cancer tissues compared with the proximal tissues (Storcelova et al., 2013), suggesting that further in-depth investigation is needed to define the exact role of Cdc20 in colorectal cancer.

4.6. Hepatocellular carcinoma

Hepatocellular carcinoma is a common malignant cancer worldwide, especially in Asian countries including China. Cdc20 has been validated to be critically involved in hepatocellular carcinoma. Li et al. reported that overexpression of Cdc20 was observed in 68% hepatocellular carcinoma tissues compared to adjacent non-tumor liver tissues. Strikingly, high levels of Cdc20 were positively correlated with gender, tumor differentiation, and TNM stage (Li et al., 2014). In further support of this concept, depletion of endogenous Cdc20 decreased cell proliferation and induced G2/M cell cycle phase in hepatocellular carcinoma cells (Li et al., 2014). Moreover, one recent study demonstrated that SIRT2 regulated APC E3 ligase activity partly through deacetylating Cdc20, and that Sirt2-deficient male mice developed hepatocellular carcinoma (Kim et al., 2011). Therefore, Cdc20 could be a promising therapeutic target for human hepatocellular carcinoma, while additional studies are warranted to validate the clinical benefits of Cdc20 inhibition in treating liver cancers.

4.7. Glioblastoma

Glioblastoma is the most common type of malignant brain tumor. The expression of Cdc20 was up-regulated in glioblastoma and down-regulated in low-grade gliomas (Marucci et al., 2008). Overexpression of Cdc20 is highly related to glioblastoma (Marucci et al., 2008). Notably, Dai et al. further found that Cdc20 plays a critical role in FoxM1-triggered cell survival in human glioblastoma (Dai et al., 2013). Furthermore, MIIP (migration and invasion inhibitor protein) interacts with Cdc20 and inhibits APC-mediated degradation of Cyclin B1, thereby inhibiting of glioma development and progression (Ji et al., 2010), while the physiological and pathological contribution of Cdc20 to glioblastoma needs further validation and investigation.

4.8. Gastric cancer

Gastric carcinoma is a common and lethal malignancy in the world. Importantly, Cdc20 was found to have a pivotal role in governing the progression of gastric cancer. Specifically, the expression of Cdc20 was upregulated in various gastric cell lines and tumor tissue samples (Kim et al., 2005). Furthermore, another independent study confirmed that Cdc20 expression was significantly higher in gastric cancer tissues than in corresponding noncancerous tissues (Ding et al., 2014). Notably, overexpression of Cdc20 was positively associated with tumor size, TNM stage, histological grade, and lymph node metastasis. More importantly, upregulation of Cdc20 was closely correlated with poor overall survival (Ding et al., 2014). Taken together, Cdc20 may be an independent marker for predicting clinical outcomes of gastric cancer and therefore, inhibiting Cdc20 might be a promising anti-gastric cancer approach at least for patients with elevated Cdc20 expression levels.

4.9. Other types of human cancers

Additional studies have validated the oncogenic function of Cdc20 in other cancers such as bladder cancer, oral cancer and cervical cancer. For instance, high expression of Cdc20 was observed in urothelial carcinoma of the human bladder (Choi et al., 2013; Kidokoro et al., 2008). This finding was confirmed by microarrays in urinanry bladder cancer tissues (Zaravinos et al., 2011). Notably, upregulation of Cdc20 was correlated with advanced age and stage, high grade, distant metastasis, shorter recurrence-free survival and poorer overall survival in bladder cancer patients (Choi et al., 2013). Furthermore, Mondal et al. observed up-regulation of Cdc20 in several oral squamous cell carcinoma cell lines and primary head and neck tumors (Mondal et al., 2007; Thirthagiri et al., 2007). Moreover, overexpression of Cdc20 in OSCC cell lines led to aneuploidy due to deregulating the activity of APC in promoting premature anaphase (Mondal et al., 2007). Furthermore, high levels of Cdc20 protein expression were observed in 37% of oral squamous cell carcinoma tissues. Strikingly, higher expression of Cdc20 was associated with shorter cancer-specific survival rate in oral cancer. These results thus indicate that Cdc20 may serve as an independent prognostic factor and a therapeutic target for oral cancer (Moura et al., 2014). In addition, Cdc20 was overexpressed in the high-grade squamous intraepithelial lesions and invasive squamous cell carcinoma in cervical cancer (Kim et al., 2014). Consistently, another independent study also defined that Cdc20 was up-regulated in invasive cervical cancers (Rajkumar et al., 2011). Recently, inhibition of Cdc20 was also reported to lead to G2/M cell cycle arrest and tumor growth inhibition in melanoma (Majumder et al., 2014). These results suggest that Cdc20 could function as a common oncoprotein in a majority of human cancers, therefore advocating for additional scientific investigation as well as translational studies to validate the clinical benefits of Cdc20 pharmacological inhibitors in treating various types of human cancers that are driven by Cdc20 overexpression.

5. Targeting Cdc20 for cancer therapies

Given the important oncogenic role of Cdc20 in tumorigenesis, its inhibitors could provide a therapeutic window in a range of human malignancies. It has been known that proteasome inhibitors can block ubiquitination-dependent proteolysis. Thus, many scientists have developed multiple proteasome inhibitors for treating human cancers (Adams, 2004; Allegra et al., 2014; Skaar et al., 2014). Remarkably, proteasome inhibitor bortezomib (Velcade™ Millennium Pharmaceuticals, Inc) and Carfizomib have been approved for treating multiple myeloma (Andreu-Vieyra & Berenson, 2014; Caravita et al., 2006; Mahindra et al., 2012). Moreover, several other proteasome inhibitors including Oprozomib, Delanzomib, and Marizomib have been used in clinical trials (Dou & Zonder, 2014). Due to their inhibitory effects on degradation of many key protein, these proteasome inhibitors have been shown to be toxic to normal cells, leading to numerous side effects including fever, anemia, diarrhea and nausea in patients (Dou & Zonder, 2014). These undesirable effects could be overcome by targeting specific components of the UPS system. To this end, discovering and developing small molecule inhibitors specifically targeting the Cdc20 oncoprotein could possibly be a novel strategy for the treatment of many types of human cancers.

5.1. TAME and pro-TAME

Studies from the King group have elegantly revealed that a small molecule, named as TAME (tosyl-L-arginine methyl ester), could bind APC and suppress its activation by Cdc20 and Cdh1 (Figure 2). A previous study from this group showed that TAME is an inhibitor of cyclin proteolysis in mitotic Xenopus egg extract (Verma et al., 2004). Moreover, they found that TAME reduced Cdc20 association with the APC and subsequent inhibited APC E3 ligase activity (Zeng et al., 2010). It is to be noted that TAME also inhibited the binding of Cdh1 to APC and reduced APC activation (Zeng et al., 2010). Given that TAME is not cell permeable, a TAME prodrug (pro-TAME), which can be processed by intracellular esterases to yield the active form of TAME, was synthesized. Further evidence revealed that pro-TAME disrupted the APC-Cdc20/Cdh1 interaction to reduce APC activation. Moreover, pro-TAME was found to induce mitotic arrest in the absence of spindle damage (Zeng et al., 2010). Intriguingly, pro-TAME in combination with microtubule inhibitors could have greater effects on enhancing cell death (Zeng et al., 2010). Strikingly, this group identified the precise mechanism by which TAME suppressed APC activation. TAME inhibits the binding of free Cdc20 to the APC. On the other hand, in the absence of APC substrates, TAME can promote Cdc20 dissociation from the APC by induction of Cdc20 auto-ubiquitination in its N-terminal region (Zeng & King, 2012). This process is suppressed by the binding of APC substrates such as cyclin B1, indicating that TAME stabilizes cyclin B1 by prematurely terminating its ubiquitination (Zeng & King, 2012). Further investigation is required to determine whether TAME and pro-TAME are clinically useful to retard tumorigenesis in vivo.

Figure 2. A schematic illustration of how pro-TAME and Apcin inhibit the APC E3 ligase activity.

Figure 2

pro-TAME, an IR-mimetic, directly blocks the recruitment of APC coactivators including Cdc20 and Cdh1, to the APC core complex. On the other hand, Apcin occupies the D-box-binding pocket on the side of the WD40 domain of Cdc20 and therefore blocking substrate-induced Cdc20 loading onto the APC core complex.

5.2. Apcin

Recently, another elegant study from the King laboratory validated that another small molecule, apcin (APC inhibitor), binds Cdc20 and prevents substrate recognition, thereby leading to competitively inhibition of the ubiquitination of Cdc20 substrates (Sackton et al., 2014). This group further explored the crystal structure of the apcin-Cdc20 complex and validated that apcin occupies the D-box-binding pocket within the WD40 domain (Sackton et al., 2014). It has been known that substrates can promote cooperative Cdc20 binding to the APC via a co-receptor interaction. To this end, further analysis has validated that apcin also blocks substrate-induced Cdc20 loading onto the APC core complex (Sackton et al., 2014). Interestingly, apcin is highly specific for stabilization of substrates that interact with APC through a D-box motif including cyclin B1 and securin, while TAME inhibited the degradation of all APC substrates due to its ability in directly blocking the recruitment of Cdc20 to the APC core complex (Sackton et al., 2014). Since apcin and TAME have distinct mechanisms to inhibit APC activation, the combination of apcin and TAME caused a synergistic stabilization of APC substrates such as cyclin B1, securin, and cyclin A2. Notably, apcin and pro-TAME synergized to increase the mitotic fraction in human cancer cell lines (Sackton et al., 2014). Taken together, the function of apcin can be dramatically enhanced by the combination of TAME, arguing that simultaneous inhibition of multiple protein-protein interactions by multiple compounds could represent a novel approach for the therapeutic targeting of protein complexes.

5.3. Withaferin A

Withaferin A, a bioactive component from Withania somnifera, has been confirmed to exhibit its anti-tumor activity against various types of human cancers including leukemia, pancreatic cancer, breast cancer and colorectal cancer. One recent study showed that withaferin A decreased STAT3 and induced cell death in neuroblastoma and multiple myeloma (Yco et al., 2014). Li et al. reported that withaferin A enhanced oxaliplatin-induced growth suppression and apoptosis via reactive oxygen species-mediated inactivation of the PI3K/Akt pathway in pancreatic cancer cells (Li et al., 2015). Moreover, withaferin A was found to inhibit canonical and constitutive NF-κB activities, resulting in induction of cellular apoptosis in lymphoma lines (Jackson et al., 2014). Strikingly, withaferin A eliminates cancer stem cells in ovarian cancer cells, leading to reduction in tumor growth and inhibition of metastasis in an ovarian orthotopic mouse model (Kakar et al., 2014). Notably, one elegant study demonstrated that withaferin A exerts its anti-tumor effects on breast cancer through regulation of ERK (extracellular signal-regulated kinase)/RSK (ribosomal S6 kinase) and DR5 (death receptor 5) as well as Elk1 (ETS-like transcription factor 1) and CHOP (C-EBP homologous protein) (Nagalingam et al., 2014). Furthermore, withaferin A treatment led to G2/M phase arrest and apoptosis in colorectal cancer cell lines (Das et al., 2014). Importantly, withaferin A exerts its anti-cancer activity through enhanced degradation of Cdc20 and Mad2, and also blocks SAC function, leading to mitotic delay, indicating that suppressing Cdc20 activity could be one molecular mechanism underlying the anti-cancer nature of Withaferin A (Das et al., 2014).

5.4. NAHA

It has been reported that NAHA, a N-alkylated amino acid-derived sulfonamide hydroxamate, has been shown to inhibit the expression of Cdc20 in breast cancer cells, but the underlying molecular mechanisms remains largely unclear (Jiang et al., 2012). Specifically, NAHA was found to enhance cell proliferation inhibition potency (Stanger et al., 2006). Further study revealed that NAHA inhibited proliferation and colony formation together with decreased Cdc20 levels. Moreover, NAHA retarded cell adhesion, invasion, and migration partly through inhibiting secretion of uPA(urokinase-type plasminogen activator) (Jiang et al., 2012). Notably, NAHA also inhibited breast cancer cell-mediated angiogenesis in part via down-regulation of VEGF (vascular endothelial growth factor). Consistent with this finding, NAHA decreased tumor volume and tumor weight as well as angiogenesis in mouse xenograft model of breast cancer (Jiang et al., 2012). However, further in-depth study is required to determine mechanistically how NAHA regulates the expression of Cdc20 in human cancer cells.

5.5. Ganodermanontriol

Ganodermanontriol (GDNT), a ganoderma alcohol from medicinal mushroom, has been discovered to inhibit cell proliferation via targeting Cdc20 in breast cancer cells (Jiang et al., 2011). Mechanistically, GDNT treatment led to the inhibition of cell growth, colony formation and invasion partly through down-regulation of Cdc20, uPA and uPAR (urokinase-type plasminogen activator receptor) expression (Jiang et al., 2011). Moreover, MycoPhyto® Complex (MC), a novel medicinal mushroom blend, was also identified to inhibit the expression of multiple cell cycle regulatory genes including Cdc20, leading to inhibition of cell proliferation and induction of cell cycle arrest as well as suppressing cell invasiveness in breast cancer cells (Jiang & Sliva, 2010). Altogether, GDNT and MC could be non-specific Cdc20 inhibitors.

5.6. Genistein

Genistein, a phytoestrogenic isoflavonid, has been believed to have pleiotropic biological effects in human malignancies, with relatively low toxicity to normal cells (Banerjee et al., 2008). A growing body of data implicates that genistein as a protein tyrosine kinase inhibitor that inhibits cell growth, migration, invasion, angiogenesis, and metastasis, and induces apoptosis and cell cycle arrest through regulation of multiple cellular signaling pathways (Sarkar et al., 2010). Emerging evidence also demonstrated that genistein deregulates Akt, NF-κB, Wnt and Hedgehog signaling pathways in human cancers (Sarkar et al., 2006). Further study has demonstrated that genistein exhibits its anti-carcinogenic properties through down-regulation of core regulatory genes including Cdc20 in primary glioblastoma, rhabdomyosarcoma, hepatocellular carcinoma and human embryonic carcinoma cells (Regenbrecht et al., 2008). In addition, recent study has also indicated that genistein governed the expression of Cdc20, leading to control of cell cycle in breast cancer cells (Zhang et al., 2015). Interestingly, emerging evidence has shown that genistein upregulated many genes involved in cell cycle such as Cdc20 in breast cancer patients (Shike et al., 2014). Therefore, it is necessary to further determine the role of genistein in breast cancer and whether inhibition of Cdc20 is the major signaling pathway through which genistein exerts its anti-cancer effects.

5.7. CFM-4 and BCHHD

CARP-1 is a peri-nuclear phosphoprotein that regulates cell growth and apoptosis. Studies have revealed that CARP-1 is a part of the NF-κB proteome and β-catenin signaling pathways (Jamal et al., 2014). Moreover, CARP-1 is also an co-activator of steroid/thyroid nuclear receptors. Furthermore, CARP-1 was found to bind APC2 and Cdc20 as well as Cdh1 (Puliyappadamba et al., 2011). CFM-4 (CARP-1 functional mimetic 4) was discovered to disrupt the CARP-1/APC-2 binding. CFM-4 prevents the binding between CARP-1 and APC2, leading to cell cycle arrest and apoptosis. CFM-4-induced apoptosis is involved in down-regulation of Cdc20 in breast cancer cells (Puliyappadamba et al., 2011). Additionally, CFM-4 inhibited cell growth and invasion in malignant pleural mesothelioma (Jamal et al., 2014). Moreover, the 6-brominated coumarin hydrazide-hydrazone derivative (BCHHD) 7c was recently found to inhibit Cdc20 expression in drug-resistant pancreatic cancer cells, suggesting that BCHHD 7c could be a potent anti-tumor drug to overcome drug resistance in pancreatic cancer (Nasr et al., 2014), but the contribution of the Cdc20 pathway in this process needs additional in-depth investigation.

6. Conclusions and future perspectives

In conclusion, as Cdc20 is critically involved in human tumorigenesis, development of specific Cdc20 inhibitors could be a strategy for improving the treatment of human cancers. It is noteworthy that Apcin is a specific inhibitor with a direct action against the APCCdc20 complex, while other inhibitors are not specific to target Cdc20. Therefore, more efforts are needed to discover other more specific Cdc20 inhibitors. We hope this article could stimulate more research efforts to develop specific Cdc20 inhibitors as anti-cancer agents. This is an important research direction given that increasing evidence suggest Cdh1 as a tumor suppressor while Cdc20 as an oncogene, therefore excluding the usage of pan-APC inhibitors, but advocating for specific Cdc20 inhibitors as a novel anti-cancer approach. One alternative approach may be to regulate Cdc20 upstream regulators including p53, Emi1, and USP44. On the other hand, it is also feasible to inhibit the E3 ubiquitin ligase activity of the APCCdc20 complex to block Cdc20 oncogenic function for cancer therapy. Due to their non-toxic nature, inhibiting Cdc20 by natural agents such as withaferin, GDNT, genistein could be a safer approach for better treatment of human cancers. We believe that deeper investigation is required to explore the mechanisms of Cdc20-mediated tumorigenesis, which will provide the rationale for developing specific Cdc20 inhibitors as effective anti-cancer agents in the near future. For future studies to determine the role of Cdc20, identification of its additional ubiquitin substrates by novel methods is critical (Figure 3). It is also important to explore the novel role of Cdc20 outside cell cycle progression control and apoptosis. To fully understand the function of Cdc20 in tumorigenesis, generation of Cdc20 conditional knockout (KO) or knockin mouse models will be necessary to better appreciate the physiological role of Cdc20 in various human cancer settings (Figure 3). To achieve better treatment outcome, Cdc20 inhibitors with greater specificity and efficacy should be developed and validated by both in vitro cell culture based studies and in vivo mouse modeling studies.

Figure 3. Future Perspectives.

Figure 3

A schematic illustration of the future studies to determine the oncogenic role of Cdc20 in various cancer settings, as well as to further validate whether pharmacological inhibition of Cdc20 could achieve clinical benefits for cancer patients, at least those with elevated Cdc20 expression levels.

Table 3. The list of compounds targeting the Cdc20 activity.

Compound Target and function Reference
TAME Reduces Cdc20 association with the APC and subsequently inhibits APC activity. (Zeng et al., 2010)
Pro-TAME A TAME pro-drug, disrupts the APC-Cdc20 interaction and then reduces APC activation. (Zeng et al., 2010)
Apcin Occupies the D-box-binding pocket on the side of the WD40 domain, blocks substrate-induced Cdc20 loading onto the APC. (Sackton et al., 2014)
Withaferin A Enhances degradation of Cdc20, blocks SAC function, leading to mitotic delay. (Das et al., 2014)
NAHA Inhibits the expression of Cdc20 in breast cancer cells, retards cell proliferation and colony formation. (Jiang et al., 2012)
Ganodermanontriol, Mycophyto complex Down-regulates Cdc20 expression, inhibits cell proliferation and invasion in breast cancer cells. (Jiang et al., 2011; Jiang & Sliva, 2010)
Genistein Regulation of Cdc20 in various human cancers to exert its anti-tumor activity. (Regenbrecht et al., 2008; Shike et al., 2014; Zhang et al., 2015)
CFM-4 Down-regulates Cdc20 in breast cancer cells, induces apoptosis. (Puliyappadamba et al., 2011)
BCHHD 7c Inhibits Cdc20 expression in drug resistant pancreatic cancer cells. (Nasr et al., 2014)

Acknowledgments

This work was supported in part by the NIH grants to W.W. (GM094777 and CA177910). W.W. is an ACS research scholar and a LLS research scholar. This work was also supported by grant from NSFC (81172087) and a project funded by the priority academic program development of Jiangsu higher education institutions.

Footnotes

Conflict of Interest: The authors declare no conflict of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Adams J. The proteasome: a suitable antineoplastic target. Nature reviews Cancer. 2004;4:349–360. doi: 10.1038/nrc1361. [DOI] [PubMed] [Google Scholar]
  2. Akiyama T, Bouillet P, Miyazaki T, Kadono Y, Chikuda H, Chung UI, Fukuda A, Hikita A, Seto H, Okada T, Inaba T, Sanjay A, Baron R, Kawaguchi H, Oda H, Nakamura K, Strasser A, Tanaka S. Regulation of osteoclast apoptosis by ubiquitylation of proapoptotic BH3-only Bcl-2 family member Bim. EMBO J. 2003;22:6653–6664. doi: 10.1093/emboj/cdg635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Akiyama T, Dass CR, Choong PF. Bim-targeted cancer therapy: a link between drug action and underlying molecular changes. Mol Cancer Ther. 2009;8:3173–3180. doi: 10.1158/1535-7163.MCT-09-0685. [DOI] [PubMed] [Google Scholar]
  4. Akiyama T, Tanaka S. Bim: guardian of tissue homeostasis and critical regulator of the immune system, tumorigenesis and bone biology. Archivum immunologiae et therapiae experimentalis. 2011;59:277–287. doi: 10.1007/s00005-011-0126-1. [DOI] [PubMed] [Google Scholar]
  5. Allegra A, Alonci A, Gerace D, Russo S, Innao V, Calabro L, Musolino C. New orally active proteasome inhibitors in multiple myeloma. Leukemia research. 2014;38:1–9. doi: 10.1016/j.leukres.2013.10.018. [DOI] [PubMed] [Google Scholar]
  6. Amador V, Ge S, Santamaria PG, Guardavaccaro D, Pagano M. APC/C(Cdc20) controls the ubiquitin-mediated degradation of p21 in prometaphase. Mol Cell. 2007;27:462–473. doi: 10.1016/j.molcel.2007.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Andreu-Vieyra C, Berenson JR. Carfilzomib in multiple myeloma. Expert opinion on biological therapy. 2014;14:1685–1699. doi: 10.1517/14712598.2014.953050. [DOI] [PubMed] [Google Scholar]
  8. Araki M, Wharton RP, Tang Z, Yu H, Asano M. Degradation of origin recognition complex large subunit by the anaphase-promoting complex in Drosophila. EMBO J. 2003;22:6115–6126. doi: 10.1093/emboj/cdg573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Banerjee S, Li Y, Wang Z, Sarkar FH. Multi-targeted therapy of cancer by genistein. Cancer letters. 2008;269:226–242. doi: 10.1016/j.canlet.2008.03.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Banerjee T, Nath S, Roychoudhury S. DNA damage induced p53 downregulates Cdc20 by direct binding to its promoter causing chromatin remodeling. Nucleic acids research. 2009;37:2688–2698. doi: 10.1093/nar/gkp110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bashir T, Dorrello NV, Amador V, Guardavaccaro D, Pagano M. Control of the SCF(Skp2-Cks1) ubiquitin ligase by the APC/C(Cdh1) ubiquitin ligase. Nature. 2004;428:190–193. doi: 10.1038/nature02330. [DOI] [PubMed] [Google Scholar]
  12. Bassermann F, Eichner R, Pagano M. The ubiquitin proteasome system - implications for cell cycle control and the targeted treatment of cancer. Biochimica et biophysica acta. 2014;1843:150–162. doi: 10.1016/j.bbamcr.2013.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bieniek J, Childress C, Swatski MD, Yang W. COX-2 inhibitors arrest prostate cancer cell cycle progression by down-regulation of kinetochore/centromere proteins. The Prostate. 2014;74:999–1011. doi: 10.1002/pros.22815. [DOI] [PubMed] [Google Scholar]
  14. Budhavarapu VN, White ED, Mahanic CS, Chen L, Lin FT, Lin WC. Regulation of E2F1 by APC/C Cdh1 via K11 linkage-specific ubiquitin chain formation. Cell Cycle. 2012;11:2030–2038. doi: 10.4161/cc.20643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Caravita T, de Fabritiis P, Palumbo A, Amadori S, Boccadoro M. Bortezomib: efficacy comparisons in solid tumors and hematologic malignancies. Nature clinical practice Oncology. 2006;3:374–387. doi: 10.1038/ncponc0555. [DOI] [PubMed] [Google Scholar]
  16. Castro A, Vigneron S, Bernis C, Labbe JC, Lorca T. Xkid is degraded in a D-box, KEN-box, and A-box-independent pathway. Mol Cell Biol. 2003;23:4126–4138. doi: 10.1128/MCB.23.12.4126-4138.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chang DZ, Ma Y, Ji B, Liu Y, Hwu P, Abbruzzese JL, Logsdon C, Wang H. Increased CDC20 expression is associated with pancreatic ductal adenocarcinoma differentiation and progression. Journal of hematology & oncology. 2012;5:15. doi: 10.1186/1756-8722-5-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chang L, Barford D. Insights into the anaphase-promoting complex: a molecular machine that regulates mitosis. Current opinion in structural biology. 2014;29:1–9. doi: 10.1016/j.sbi.2014.08.003. [DOI] [PubMed] [Google Scholar]
  19. Chang L, Zhang Z, Yang J, McLaughlin SH, Barford D. Molecular architecture and mechanism of the anaphase-promoting complex. Nature. 2014;513:388–393. doi: 10.1038/nature13543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen Z, Yu Y, Fu D, Li Z, Niu X, Liao M, Lu S. Functional roles of PC-PLC and Cdc20 in the cell cycle, proliferation, and apoptosis. Cell Biochem Funct. 2010;28:249–257. doi: 10.1002/cbf.1634. [DOI] [PubMed] [Google Scholar]
  21. Cho HJ, Lee EH, Han SH, Chung HJ, Jeong JH, Kwon J, Kim H. Degradation of Human RAP80 is Cell Cycle Regulated by Cdc20 and Cdh1 Ubiquitin Ligases. Mol Cancer Res. 2012;10:615–625. doi: 10.1158/1541-7786.MCR-11-0481. [DOI] [PubMed] [Google Scholar]
  22. Choi JW, Kim Y, Lee JH, Kim YS. High expression of spindle assembly checkpoint proteins CDC20 and MAD2 is associated with poor prognosis in urothelial bladder cancer. Virchows Archiv: an international journal of pathology. 2013;463:681–687. doi: 10.1007/s00428-013-1473-6. [DOI] [PubMed] [Google Scholar]
  23. Chun AC, Kok KH, Jin DY. REV7 is required for anaphase-promoting complexdependent ubiquitination and degradation of translesion DNA polymerase REV1. Cell Cycle. 2013;12:365–378. doi: 10.4161/cc.23214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Clijsters L, Ogink J, Wolthuis R. The spindle checkpoint, APC/C(Cdc20), and APC/C(Cdh1) play distinct roles in connecting mitosis to S phase. The Journal of cell biology. 2013;201:1013–1026. doi: 10.1083/jcb.201211019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Clute P, Pines J. Temporal and spatial control of cyclin B1 destruction in metaphase. Nat Cell Biol. 1999;1:82–87. doi: 10.1038/10049. [DOI] [PubMed] [Google Scholar]
  26. Cory S, Adams JM. The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer. 2002;2:647–656. doi: 10.1038/nrc883. [DOI] [PubMed] [Google Scholar]
  27. Cragg MS, Harris C, Strasser A, Scott CL. Unleashing the power of inhibitors of oncogenic kinases through BH3 mimetics. Nat Rev Cancer. 2009;9:321–326. doi: 10.1038/nrc2615. [DOI] [PubMed] [Google Scholar]
  28. Cui Y, Cheng X, Zhang C, Zhang Y, Li S, Wang C, Guadagno TM. Degradation of the human mitotic checkpoint kinase Mps1 is cell cycle-regulated by APC-cCdc20 and APCcCdh1 ubiquitin ligases. J Biol Chem. 2010;285:32988–32998. doi: 10.1074/jbc.M110.140905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dai B, Gong A, Jing Z, Aldape KD, Kang SH, Sawaya R, Huang S. Forkhead box M1 is regulated by heat shock factor 1 and promotes glioma cells survival under heat shock stress. The Journal of biological chemistry. 2013;288:1634–1642. doi: 10.1074/jbc.M112.379362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle assembly checkpoint by degradation of Mad2-Cdc20 complex in colorectal cancer cell lines. Biochemical pharmacology. 2014;91:31–39. doi: 10.1016/j.bcp.2014.06.022. [DOI] [PubMed] [Google Scholar]
  31. Dehan E, Bassermann F, Guardavaccaro D, Vasiliver-Shamis G, Cohen M, Lowes KN, Dustin M, Huang DC, Taunton J, Pagano M. betaTrCP- 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]
  32. den Elzen N, Pines J. Cyclin A is destroyed in prometaphase and can delay chromosome alignment and anaphase. J Cell Biol. 2001;153:121–136. doi: 10.1083/jcb.153.1.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Di Fiore B, Davey NE, Hagting A, Izawa D, Mansfeld J, Gibson TJ, Pines J. The ABBA Motif Binds APC/C Activators and Is Shared by APC/C Substrates and Regulators. Dev Cell. 2015;32:358–372. doi: 10.1016/j.devcel.2015.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Ding ZY, Wu HR, Zhang JM, Huang GR, Ji DD. Expression characteristics of CDC20 in gastric cancer and its correlation with poor prognosis. International journal of clinical and experimental pathology. 2014;7:722–727. [PMC free article] [PubMed] [Google Scholar]
  35. Dou QP, Zonder JA. Overview of proteasome inhibitor-based anti-cancer therapies: perspective on bortezomib and second generation proteasome inhibitors versus future generation inhibitors of ubiquitin-proteasome system. Current cancer drug targets. 2014;14:517–536. doi: 10.2174/1568009614666140804154511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Fang G, Yu H, Kirschner MW. The checkpoint protein MAD2 and the mitotic regulator CDC20 form a ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes Dev. 1998;12:1871–1883. doi: 10.1101/gad.12.12.1871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Foe I, Toczyski D. Structural biology: a new look for the APC. Nature. 2011;470:182–183. doi: 10.1038/470182a. [DOI] [PubMed] [Google Scholar]
  38. Fraschini R, Formenti E, Lucchini G, Piatti S. Budding yeast Bub2 is localized at spindle pole bodies and activates the mitotic checkpoint via a different pathway from Mad2. J Cell Biol. 1999;145:979–991. doi: 10.1083/jcb.145.5.979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gao D, Inuzuka H, Korenjak M, Tseng A, Wu T, Wan L, Kirschner M, Dyson N, Wei W. Cdh1 regulates cell cycle through modulating the claspin/Chk1 and the Rb/E2F1 pathways. Mol Biol Cell. 2009;20:3305–3316. doi: 10.1091/mbc.E09-01-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Geley S, Kramer E, Gieffers C, Gannon J, Peters JM, Hunt T. Anaphase-promoting complex/cyclosome-dependent proteolysis of human cyclin A starts at the beginning of mitosis and is not subject to the spindle assembly checkpoint. J Cell Biol. 2001;153:137–148. doi: 10.1083/jcb.153.1.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gurden MD, Holland AJ, van Zon W, Tighe A, Vergnolle MA, Andres DA, Spielmann HP, Malumbres M, Wolthuis RM, Cleveland DW, Taylor SS. Cdc20 is required for the post-anaphase, KEN-dependent degradation of centromere protein F. J Cell Sci. 2010;123:321–330. doi: 10.1242/jcs.062075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Hadjihannas MV, Bernkopf DB, Bruckner M, Behrens J. Cell cycle control of Wnt/beta-catenin signalling by conductin/axin2 through CDC20. EMBO Rep. 2012;13:347–354. doi: 10.1038/embor.2012.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Hames RS, Wattam SL, Yamano H, Bacchieri R, Fry AM. APC/C-mediated destruction of the centrosomal kinase Nek2A occurs in early mitosis and depends upon a cyclin A-type D-box. EMBO J. 2001;20:7117–7127. doi: 10.1093/emboj/20.24.7117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Harley ME, Allan LA, Sanderson HS, Clarke PR. Phosphorylation of Mcl-1 by CDK1-cyclin B1 initiates its Cdc20-dependent destruction during mitotic arrest. EMBO J. 2010;29:2407–2420. doi: 10.1038/emboj.2010.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Hartwell LH, Culotti J, Reid B. Genetic control of the cell-division cycle in yeast. I. Detection of mutants. Proc Natl Acad Sci U S A. 1970;66:352–359. doi: 10.1073/pnas.66.2.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Hartwell LH, Mortimer RK, Culotti J, Culotti M. Genetic Control of the Cell Division Cycle in Yeast: V. Genetic Analysis of cdc Mutants. Genetics. 1973;74:267–286. doi: 10.1093/genetics/74.2.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hayes MJ, Kimata Y, Wattam SL, Lindon C, Mao G, Yamano H, Fry AM. Early mitotic degradation of Nek2A depends on Cdc20-independent interaction with the APC/C. Nat Cell Biol. 2006;8:607–614. doi: 10.1038/ncb1410. [DOI] [PubMed] [Google Scholar]
  48. Hoeller D, Hecker CM, Dikic I. Ubiquitin and ubiquitin-like proteins in cancer pathogenesis. Nat Rev Cancer. 2006;6:776–788. doi: 10.1038/nrc1994. [DOI] [PubMed] [Google Scholar]
  49. Hu D, Qiao X, Wu G, Wan Y. The emerging role of APC/CCdh1 in development. Seminars in cell & developmental biology. 2011;22:579–585. doi: 10.1016/j.semcdb.2011.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Huang HC, Shi J, Orth JD, Mitchison TJ. Evidence that mitotic exit is a better cancer therapeutic target than spindle assembly. Cancer Cell. 2009;16:347–358. doi: 10.1016/j.ccr.2009.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Ichim G, Mola M, Finkbeiner MG, Cros MP, Herceg Z, Hernandez-Vargas H. The histone acetyltransferase component TRRAP is targeted for destruction during the cell cycle. Oncogene. 2013 doi: 10.1038/onc.2012.570. [DOI] [PubMed] [Google Scholar]
  52. Ichim G, Mola M, Finkbeiner MG, Cros MP, Herceg Z, Hernandez-Vargas H. The histone acetyltransferase component TRRAP is targeted for destruction during the cell cycle. Oncogene. 2014;33:181–192. doi: 10.1038/onc.2012.570. [DOI] [PubMed] [Google Scholar]
  53. Izawa D, Pines J. The mitotic checkpoint complex binds a second CDC20 to inhibit active APC/C. Nature. 2015;517:631–634. doi: 10.1038/nature13911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Jackson SS, Oberley C, Hooper CP, Grindle K, Wuerzberger-Davis S, Wolff J, McCool K, Rui L, Miyamoto S. Withaferin A disrupts ubiquitin-based NEMO reorganization induced by canonical NF-kappaB signaling. Experimental cell research. 2014 doi: 10.1016/j.yexcr.2014.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Jamal S, Cheriyan VT, Muthu M, Munie S, Levi E, Ashour AE, Pass HI, Wali A, Singh M, Rishi AK. CARP-1 functional mimetics are a novel class of small molecule inhibitors of malignant pleural mesothelioma cells. PLoS One. 2014;9:e89146. doi: 10.1371/journal.pone.0089146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Janssen A, Medema RH. Mitosis as an anti-cancer target. Oncogene. 2011;30:2799–2809. doi: 10.1038/onc.2011.30. [DOI] [PubMed] [Google Scholar]
  57. Ji P, Smith SM, Wang Y, Jiang R, Song SW, Li B, Sawaya R, Bruner JM, Kuang J, Yu H, Fuller GN, Zhang W. Inhibition of gliomagenesis and attenuation of mitotic transition by MIIP. Oncogene. 2010;29:3501–3508. doi: 10.1038/onc.2010.114. [DOI] [PubMed] [Google Scholar]
  58. Jiang J, Jedinak A, Sliva D. Ganodermanontriol (GDNT) exerts its effect on growth and invasiveness of breast cancer cells through the down-regulation of CDC20 and uPA. Biochem Biophys Res Commun. 2011;415:325–329. doi: 10.1016/j.bbrc.2011.10.055. [DOI] [PubMed] [Google Scholar]
  59. Jiang J, Sliva D. Novel medicinal mushroom blend suppresses growth and invasiveness of human breast cancer cells. Int J Oncol. 2010;37:1529–1536. doi: 10.3892/ijo_00000806. [DOI] [PubMed] [Google Scholar]
  60. Jiang J, Thyagarajan-Sahu A, Krchnak V, Jedinak A, Sandusky GE, Sliva D. NAHA, a novel hydroxamic acid-derivative, inhibits growth and angiogenesis of breast cancer in vitro and in vivo. PLoS One. 2012;7:e34283. doi: 10.1371/journal.pone.0034283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Jin L, Williamson A, Banerjee S, Philipp I, Rape M. Mechanism of ubiquitin-chain formation by the human anaphase-promoting complex. Cell. 2008;133:653–665. doi: 10.1016/j.cell.2008.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kakar SS, Ratajczak MZ, Powell KS, Moghadamfalahi M, Miller DM, Batra SK, Singh SK. Withaferin a alone and in combination with Cisplatin suppresses growth and metastasis of ovarian cancer by targeting putative cancer stem cells. PLoS One. 2014;9:e107596. doi: 10.1371/journal.pone.0107596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Karra H, Repo H, Ahonen I, Loyttyniemi E, Pitkanen R, Lintunen M, Kuopio T, Soderstrom M, Kronqvist P. Cdc20 and securin overexpression predict short-term breast cancer survival. British journal of cancer. 2014;110:2905–2913. doi: 10.1038/bjc.2014.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kato T, Daigo Y, Aragaki M, Ishikawa K, Sato M, Kaji M. Overexpression of CDC20 predicts poor prognosis in primary non-small cell lung cancer patients. J Surg Oncol. 2012 doi: 10.1002/jso.23109. [DOI] [PubMed] [Google Scholar]
  65. Kidokoro T, Tanikawa C, Furukawa Y, Katagiri T, Nakamura Y, Matsuda K. CDC20, a potential cancer therapeutic target, is negatively regulated by p53. Oncogene. 2008;27:1562–1571. doi: 10.1038/sj.onc.1210799. [DOI] [PubMed] [Google Scholar]
  66. Kim AH, Puram SV, Bilimoria PM, Ikeuchi Y, Keough S, Wong M, Rowitch D, Bonni A. A centrosomal Cdc20-APC pathway controls dendrite morphogenesis in postmitotic neurons. Cell. 2009;136:322–336. doi: 10.1016/j.cell.2008.11.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kim HS, Jeon YK, Ha GH, Park HY, Kim YJ, Shin HJ, Lee CG, Chung DH, Lee CW. Functional interaction between BubR1 and securin in an anaphase-promoting complex/cyclosomeCdc20-independent manner. Cancer Res. 2009;69:27–36. doi: 10.1158/0008-5472.CAN-08-0820. [DOI] [PubMed] [Google Scholar]
  68. Kim HS, Vassilopoulos A, Wang RH, Lahusen T, Xiao Z, Xu X, Li C, Veenstra TD, Li B, Yu H, Ji J, Wang XW, Park SH, Cha YI, Gius D, Deng CX. SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity. Cancer Cell. 2011;20:487–499. doi: 10.1016/j.ccr.2011.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kim JM, Sohn HY, Yoon SY, Oh JH, Yang JO, Kim JH, Song KS, Rho SM, Yoo HS, Kim YS, Kim JG, Kim NS. Identification of gastric cancer-related genes using a cDNA microarray containing novel expressed sequence tags expressed in gastric cancer cells. Clin Cancer Res. 2005;11:473–482. [PubMed] [Google Scholar]
  70. Kim S, Yu H. Mutual regulation between the spindle checkpoint and APC/C. Semin Cell Dev Biol. 2011;22:551–558. doi: 10.1016/j.semcdb.2011.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kim Y, Choi JW, Lee JH, Kim YS. MAD2 and CDC20 are upregulated in high-grade squamous intraepithelial lesions and squamous cell carcinomas of the uterine cervix. International journal of gynecological pathology. 2014;33:517–523. doi: 10.1097/PGP.0000000000000082. [DOI] [PubMed] [Google Scholar]
  72. Kuipers EJ, Rosch T, Bretthauer M. Colorectal cancer screening--optimizing current strategies and new directions. Nature reviews Clinical oncology. 2013;10:130–142. doi: 10.1038/nrclinonc.2013.12. [DOI] [PubMed] [Google Scholar]
  73. Kulkarni K, Zhang Z, Chang L, Yang J, da Fonseca PC, Barford D. Building a pseudo-atomic model of the anaphase-promoting complex. Acta crystallographica Section D, Biological crystallography. 2013;69:2236–2243. doi: 10.1107/S0907444913018593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kwan PS, Lau CC, Chiu YT, Man C, Liu J, Tang KD, Wong YC, Ling MT. Daxx regulates mitotic progression and prostate cancer predisposition. Carcinogenesis. 2013;34:750–759. doi: 10.1093/carcin/bgs391. [DOI] [PubMed] [Google Scholar]
  75. Lau AW, Fukushima H, Wei W. The Fbw7 and betaTRCP E3 ubiquitin ligases and their roles in tumorigenesis. Frontiers in bioscience. 2012;17:2197–2212. doi: 10.2741/4045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Li D, Zhu J, Firozi PF, Abbruzzese JL, Evans DB, Cleary K, Friess H, Sen S. Overexpression of oncogenic STK15/BTAK/Aurora A kinase in human pancreatic cancer. Clin Cancer Res. 2003;9:991–997. [PubMed] [Google Scholar]
  77. Li J, Gao JZ, Du JL, Huang ZX, Wei LX. Increased CDC20 expression is associated with development and progression of hepatocellular carcinoma. International journal of oncology. 2014;45:1547–1555. doi: 10.3892/ijo.2014.2559. [DOI] [PubMed] [Google Scholar]
  78. Li M, York JP, Zhang P. Loss of Cdc20 causes a securin-dependent metaphase arrest in two-cell mouse embryos. Mol Cell Biol. 2007;27:3481–3488. doi: 10.1128/MCB.02088-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Li X, Zhu F, Jiang J, Sun C, Wang X, Shen M, Tian R, Shi C, Xu M, Peng F, Guo X, Wang M, Qin R. Synergistic antitumor activity of withaferin A combined with oxaliplatin triggers reactive oxygen species-mediated inactivation of the PI3K/AKT pathway in human pancreatic cancer cells. Cancer letters. 2015;357:219–230. doi: 10.1016/j.canlet.2014.11.026. [DOI] [PubMed] [Google Scholar]
  80. Lim HH, Goh PY, Surana U. Cdc20 is essential for the cyclosome-mediated proteolysis of both Pds1 and Clb2 during M phase in budding yeast. Curr Biol. 1998;8:231–234. doi: 10.1016/s0960-9822(98)70088-0. [DOI] [PubMed] [Google Scholar]
  81. Lim HJ, Dimova NV, Tan MK, Sigoillot FD, King RW, Shi Y. The G2/M regulator histone demethylase PHF8 is targeted for degradation by the anaphase-promoting complex containing CDC20. Molecular and cellular biology. 2013;33:4166–4180. doi: 10.1128/MCB.00689-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Lindon C, Pines J. Ordered proteolysis in anaphase inactivates Plk1 to contribute to proper mitotic exit in human cells. J Cell Biol. 2004;164:233–241. doi: 10.1083/jcb.200309035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Lipkowitz S, Weissman AM. RINGs of good and evil: RING finger ubiquitin ligases at the crossroads of tumour suppression and oncogenesis. Nature reviews Cancer. 2011;11:629–643. doi: 10.1038/nrc3120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Littlepage LE, Ruderman JV. Identification of a new APC/C recognition domain, the A box, which is required for the Cdh1-dependent destruction of the kinase Aurora-A during mitotic exit. Genes Dev. 2002;16:2274–2285. doi: 10.1101/gad.1007302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lize M, Herr C, Klimke A, Bals R, Dobbelstein M. MicroRNA-449a levels increase by several orders of magnitude during mucociliary differentiation of airway epithelia. Cell Cycle. 2010;9:4579–4583. doi: 10.4161/cc.9.22.13870. [DOI] [PubMed] [Google Scholar]
  86. Mahindra A, Laubach J, Raje N, Munshi N, Richardson PG, Anderson K. Latest advances and current challenges in the treatment of multiple myeloma. Nature reviews Clinical oncology. 2012;9:135–143. doi: 10.1038/nrclinonc.2012.15. [DOI] [PubMed] [Google Scholar]
  87. Majumder P, Bhunia S, Bhattacharyya J, Chaudhuri A. Inhibiting tumor growth by targeting liposomally encapsulated CDC20siRNA to tumor vasculature: therapeutic RNA interference. Journal of controlled release : official journal of the Controlled Release Society. 2014;180:100–108. doi: 10.1016/j.jconrel.2014.02.012. [DOI] [PubMed] [Google Scholar]
  88. Manchado E, Guillamot M, de Carcer G, Eguren M, Trickey M, Garcia-Higuera I, Moreno S, Yamano H, Canamero M, Malumbres M. Targeting mitotic exit leads to tumor regression in vivo: Modulation by Cdk1, Mastl, and the PP2A/B55alpha,delta phosphatase. Cancer Cell. 2010;18:641–654. doi: 10.1016/j.ccr.2010.10.028. [DOI] [PubMed] [Google Scholar]
  89. Marucci G, Morandi L, Magrini E, Farnedi A, Franceschi E, Miglio R, Calo D, Pession A, Foschini MP, Eusebi V. Gene expression profiling in glioblastoma and immunohistochemical evaluation of IGFBP-2 and CDC20. Virchows Arch. 2008;453:599–609. doi: 10.1007/s00428-008-0685-7. [DOI] [PubMed] [Google Scholar]
  90. McGarry TJ, Kirschner MW. Geminin, an inhibitor of DNA replication, is degraded during mitosis. Cell. 1998;93:1043–1053. doi: 10.1016/s0092-8674(00)81209-x. [DOI] [PubMed] [Google Scholar]
  91. McLean JR, Chaix D, Ohi MD, Gould KL. State of the APC/C: organization, function, and structure. Crit Rev Biochem Mol Biol. 2011;46:118–136. doi: 10.3109/10409238.2010.541420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Michaelis C, Ciosk R, Nasmyth K. Cohesins: chromosomal proteins that prevent premature separation of sister chromatids. Cell. 1997;91:35–45. doi: 10.1016/s0092-8674(01)80007-6. [DOI] [PubMed] [Google Scholar]
  93. Mondal G, Sengupta S, Panda CK, Gollin SM, Saunders WS, Roychoudhury S. Overexpression of Cdc20 leads to impairment of the spindle assembly checkpoint and aneuploidization in oral cancer. Carcinogenesis. 2007;28:81–92. doi: 10.1093/carcin/bgl100. [DOI] [PubMed] [Google Scholar]
  94. Moura IM, Delgado ML, Silva PM, Lopes CA, do Amaral JB, Monteiro LS, Bousbaa H. High CDC20 expression is associated with poor prognosis in oral squamous cell carcinoma. Journal of oral pathology & medicine. 2014;43:225–231. doi: 10.1111/jop.12115. [DOI] [PubMed] [Google Scholar]
  95. Moustafa-Kamal M, Gamache I, Lu Y, Li S, Teodoro JG. BimEL is phosphorylated at mitosis by Aurora A and targeted for degradation by betaTrCP1. Cell death and differentiation. 2013;20:1393–1403. doi: 10.1038/cdd.2013.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Nagalingam A, Kuppusamy P, Singh SV, Sharma D, Saxena NK. Mechanistic elucidation of the antitumor properties of withaferin a in breast cancer. Cancer research. 2014;74:2617–2629. doi: 10.1158/0008-5472.CAN-13-2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. 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]
  98. Nasmyth K. Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu Rev Genet. 2001;35:673–745. doi: 10.1146/annurev.genet.35.102401.091334. [DOI] [PubMed] [Google Scholar]
  99. Nasr T, Bondock S, Youns M. Anticancer activity of new coumarin substituted hydrazide-hydrazone derivatives. European journal of medicinal chemistry. 2014;76:539–548. doi: 10.1016/j.ejmech.2014.02.026. [DOI] [PubMed] [Google Scholar]
  100. Ohtoshi A, Maeda T, Higashi H, Ashizawa S, Hatakeyama M. Human p55(CDC)/Cdc20 associates with cyclin A and is phosphorylated by the cyclin A-Cdk2 complex. Biochem Biophys Res Commun. 2000;268:530–534. doi: 10.1006/bbrc.2000.2167. [DOI] [PubMed] [Google Scholar]
  101. Penas C, Ramachandran V, Ayad NG. The APC/C Ubiquitin Ligase: From Cell Biology to Tumorigenesis. Frontiers in oncology. 2011;1:60. doi: 10.3389/fonc.2011.00060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Petersen BO, Wagener C, Marinoni F, Kramer ER, Melixetian M, Lazzerini Denchi E, Gieffers C, Matteucci C, Peters JM, Helin K. Cell cycle- and cell growth-regulated proteolysis of mammalian CDC6 is dependent on APC-CDH1. Genes Dev. 2000;14:2330–2343. doi: 10.1101/gad.832500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Puliyappadamba VT, Wu W, Bevis D, Zhang L, Polin L, Kilkuskie R, Finley RL, Jr, Larsen SD, Levi E, Miller FR, Wali A, Rishi AK. Antagonists of anaphase-promoting complex (APC)-2-cell cycle and apoptosis regulatory protein (CARP)-1 interaction are novel regulators of cell growth and apoptosis. J Biol Chem. 2011;286:38000–38017. doi: 10.1074/jbc.M111.222398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Qiao X, Zhang L, Gamper AM, Fujita T, Wan Y. APC/C-Cdh1: from cell cycle to cellular differentiation and genomic integrity. Cell Cycle. 2010;9:3904–3912. doi: 10.4161/cc.9.19.13585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Rajkumar T, Sabitha K, Vijayalakshmi N, Shirley S, Bose MV, Gopal G, Selvaluxmy G. Identification and validation of genes involved in cervical tumourigenesis. BMC Cancer. 2011;11:80. doi: 10.1186/1471-2407-11-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Regenbrecht CR, Jung M, Lehrach H, Adjaye J. The molecular basis of genistein-induced mitotic arrest and exit of self-renewal in embryonal carcinoma and primary cancer cell lines. BMC Med Genomics. 2008;1:49. doi: 10.1186/1755-8794-1-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Reimann JD, Freed E, Hsu JY, Kramer ER, Peters JM, Jackson PK. Emi1 is a mitotic regulator that interacts with Cdc20 and inhibits the anaphase promoting complex. Cell. 2001;105:645–655. doi: 10.1016/s0092-8674(01)00361-0. [DOI] [PubMed] [Google Scholar]
  108. Reimann JD, Gardner BE, Margottin-Goguet F, Jackson PK. Emi1 regulates the anaphase-promoting complex by a different mechanism than Mad2 proteins. Genes Dev. 2001;15:3278–3285. doi: 10.1101/gad.945701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Reis A, Levasseur M, Chang HY, Elliott DJ, Jones KT. The CRY box: a second APCcdh1-dependent degron in mammalian cdc20. EMBO Rep. 2006;7:1040–1045. doi: 10.1038/sj.embor.7400772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Sackton KL, Dimova N, Zeng X, Tian W, Zhang M, Sackton TB, Meaders J, Pfaff KL, Sigoillot F, Yu H, Luo X, King RW. Synergistic blockade of mitotic exit by two chemical inhibitors of the APC/C. Nature. 2014;514:646–649. doi: 10.1038/nature13660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Sarkar FH, Adsule S, Padhye S, Kulkarni S, Li Y. The role of genistein and synthetic derivatives of isoflavone in cancer prevention and therapy. Mini reviews in medicinal chemistry. 2006;6:401–407. doi: 10.2174/138955706776361439. [DOI] [PubMed] [Google Scholar]
  112. Sarkar FH, Li Y, Wang Z, Padhye S. Lesson learned from nature for the development of novel anti-cancer agents: implication of isoflavone, curcumin, and their synthetic analogs. Current pharmaceutical design. 2010;16:1801–1812. doi: 10.2174/138161210791208956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Schreiber A, Stengel F, Zhang Z, Enchev RI, Kong EH, Morris EP, Robinson CV, da Fonseca PC, Barford D. Structural basis for the subunit assembly of the anaphase-promoting complex. Nature. 2011;470:227–232. doi: 10.1038/nature09756. [DOI] [PubMed] [Google Scholar]
  114. Sedgwick GG, Hayward DG, Di Fiore B, Pardo M, Yu L, Pines J, Nilsson J. Mechanisms controlling the temporal degradation of Nek2A and Kif18A by the APC/C-Cdc20 complex. The EMBO journal. 2013;32:303–314. doi: 10.1038/emboj.2012.335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Shalom O, Shalva N, Altschuler Y, Motro B. The mammalian Nek1 kinase is involved in primary cilium formation. FEBS letters. 2008;582:1465–1470. doi: 10.1016/j.febslet.2008.03.036. [DOI] [PubMed] [Google Scholar]
  116. Shike M, Doane AS, Russo L, Cabal R, Reis-Filo J, Gerald W, Cody H, Khanin R, Bromberg J, Norton L. The effects of soy supplementation on gene expression in breast cancer: a randomized placebo-controlled study. Journal of the National Cancer Institute. 2014;106 doi: 10.1093/jnci/dju189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Shirayama M, Toth A, Galova M, Nasmyth K. APC(Cdc20) promotes exit from mitosis by destroying the anaphase inhibitor Pds1 and cyclin Clb5. Nature. 1999;402:203–207. doi: 10.1038/46080. [DOI] [PubMed] [Google Scholar]
  118. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA: a cancer journal for clinicians. 2015;65:5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
  119. Skaar JR, Pagan JK, Pagano M. SCF ubiquitin ligase-targeted therapies. Nature reviews Drug discovery. 2014;13:889–903. doi: 10.1038/nrd4432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Song L, Rape M. Regulated degradation of spindle assembly factors by the anaphase-promoting complex. Molecular cell. 2010;38:369–382. doi: 10.1016/j.molcel.2010.02.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Song MS, Song SJ, Ayad NG, Chang JS, Lee JH, Hong HK, Lee H, Choi N, Kim J, Kim H, Kim JW, Choi EJ, Kirschner MW, Lim DS. The tumour suppressor RASSF1A regulates mitosis by inhibiting the APC-Cdc20 complex. Nat Cell Biol. 2004;6:129–137. doi: 10.1038/ncb1091. [DOI] [PubMed] [Google Scholar]
  122. Stanger KJ, Sliva D, Jiang J, Krchnak V. Synthesis and screening of N-alkyl hydroxamates for inhibition of cancer cell proliferation. Combinatorial chemistry & high throughput screening. 2006;9:651–661. doi: 10.2174/138620706778700161. [DOI] [PubMed] [Google Scholar]
  123. Stegmeier F, Rape M, Draviam VM, Nalepa G, Sowa ME, Ang XL, McDonald ER, 3rd, Li MZ, Hannon GJ, Sorger PK, Kirschner MW, Harper JW, Elledge SJ. Anaphase initiation is regulated by antagonistic ubiquitination and deubiquitination activities. Nature. 2007;446:876–881. doi: 10.1038/nature05694. [DOI] [PubMed] [Google Scholar]
  124. Storcelova M, Vician M, Reis R, Zeman M, Herichova I. Expression of cell cycle regulatory factors hus1, gadd45a, rb1, cdkn2a and mre11a correlates with expression of clock gene per2 in human colorectal carcinoma tissue. Molecular biology reports. 2013;40:6351–6361. doi: 10.1007/s11033-013-2749-2. [DOI] [PubMed] [Google Scholar]
  125. Taniguchi K, Momiyama N, Ueda M, Matsuyama R, Mori R, Fujii Y, Ichikawa Y, Endo I, Togo S, Shimada H. Targeting of CDC20 via small interfering RNA causes enhancement of the cytotoxicity of chemoradiation. Anticancer Res. 2008;28:1559–1563. [PubMed] [Google Scholar]
  126. Thien CB, Dagger SA, Steer JH, Koentgen F, Jansen ES, Scott CL, Langdon WY. c-Cbl promotes T cell receptor-induced thymocyte apoptosis by activating the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. 2010;285:10969–10981. doi: 10.1074/jbc.M109.094920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Thirthagiri E, Robinson CM, Huntley S, Davies M, Yap LF, Prime SS, Paterson IC. Spindle assembly checkpoint and centrosome abnormalities in oral cancer. Cancer Lett. 2007;258:276–285. doi: 10.1016/j.canlet.2007.09.008. [DOI] [PubMed] [Google Scholar]
  128. Verma R, Peters NR, D'Onofrio M, Tochtrop GP, Sakamoto KM, Varadan R, Zhang M, Coffino P, Fushman D, Deshaies RJ, King RW. Ubistatins inhibit proteasome-dependent degradation by binding the ubiquitin chain. Science. 2004;306:117–120. doi: 10.1126/science.1100946. [DOI] [PubMed] [Google Scholar]
  129. Vodermaier HC, Gieffers C, Maurer-Stroh S, Eisenhaber F, Peters JM. TPR subunits of the anaphase-promoting complex mediate binding to the activator protein CDH1. Curr Biol. 2003;13:1459–1468. doi: 10.1016/s0960-9822(03)00581-5. [DOI] [PubMed] [Google Scholar]
  130. Wan L, Tan M, Yang J, Inuzuka H, Dai X, Wu T, Liu J, Shaik S, Chen G, Deng J, Malumbres M, Letai A, Kirschner MW, Sun Y, Wei W. APC(Cdc20) suppresses apoptosis through targeting Bim for ubiquitination and destruction. Developmental cell. 2014;29:377–391. doi: 10.1016/j.devcel.2014.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Wan L, Zou W, Gao D, Inuzuka H, Fukushima H, Berg AH, Drapp R, Shaik S, Hu D, Lester C, Eguren M, Malumbres M, Glimcher LH, Wei W. Cdh1 regulates osteoblast function through an APC/C-independent modulation of Smurf1. Mol Cell. 2011;44:721–733. doi: 10.1016/j.molcel.2011.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Wang Q, Tiffen J, Bailey CG, Lehman ML, Ritchie W, Fazli L, Metierre C, Feng YJ, Li E, Gleave M, Buchanan G, Nelson CC, Rasko JE, Holst J. Targeting amino acid transport in metastatic castration-resistant prostate cancer: effects on cell cycle, cell growth, and tumor development. Journal of the National Cancer Institute. 2013;105:1463–1473. doi: 10.1093/jnci/djt241. [DOI] [PubMed] [Google Scholar]
  133. Wang R, Li KM, Zhou CH, Xue JL, Ji CN, Chen JZ. Cdc20 mediates D-box-dependent degradation of Sp100. Biochem Biophys Res Commun. 2011;415:702–706. doi: 10.1016/j.bbrc.2011.10.146. [DOI] [PubMed] [Google Scholar]
  134. Wang W, Wu T, Kirschner MW. The master cell cycle regulator APC-Cdc20 regulates ciliary length and disassembly of the primary cilium. eLife. 2014;3:e03083. doi: 10.7554/eLife.03083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Wang Y, Zhan Q. Cell cycle-dependent expression of centrosomal ninein-like protein in human cells is regulated by the anaphase-promoting complex. J Biol Chem. 2007;282:17712–17719. doi: 10.1074/jbc.M701350200. [DOI] [PubMed] [Google Scholar]
  136. Wang Z, Gao D, Fukushima H, Inuzuka H, Liu P, Wan L, Sarkar FH, Wei W. Skp2: a novel potential therapeutic target for prostate cancer. Biochimica et biophysica acta. 2012;1825:11–17. doi: 10.1016/j.bbcan.2011.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wang Z, Liu P, Inuzuka H, Wei W. Roles of F-box proteins in cancer. Nature reviews Cancer. 2014;14:233–247. doi: 10.1038/nrc3700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Wang Z, Wan L, Zhong J, Inuzuka H, Liu P, Sarkar FH, Wei W. Cdc20: a potential novel therapeutic target for cancer treatment. Current pharmaceutical design. 2013;19:3210–3214. doi: 10.2174/1381612811319180005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Wei W, Ayad NG, Wan Y, Zhang GJ, Kirschner MW, Kaelin WG., Jr Degradation of the SCF component Skp2 in cell-cycle phase G1 by the anaphase-promoting complex. Nature. 2004;428:194–198. doi: 10.1038/nature02381. [DOI] [PubMed] [Google Scholar]
  140. Wiggins CM, Band H, Cook SJ. c-Cbl is not required for ERK1/2-dependent degradation of BimEL. Cellular signalling. 2007;19:2605–2611. doi: 10.1016/j.cellsig.2007.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Wu WJ, Hu KS, Wang DS, Zeng ZL, Zhang DS, Chen DL, Bai L, Xu RH. CDC20 overexpression predicts a poor prognosis for patients with colorectal cancer. Journal of translational medicine. 2013;11:142. doi: 10.1186/1479-5876-11-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Yamanaka S, Campbell NR, An F, Kuo SC, Potter JJ, Mezey E, Maitra A, Selaru FM. Coordinated effects of microRNA-494 induce G 2/M arrest in human cholangiocarcinoma. Cell Cycle. 2012;11 doi: 10.4161/cc.21105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Yang M, Li B, Tomchick DR, Machius M, Rizo J, Yu H, Luo X. p31comet blocks Mad2 activation through structural mimicry. Cell. 2007;131:744–755. doi: 10.1016/j.cell.2007.08.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Yang Y, Kim AH, Yamada T, Wu B, Bilimoria PM, Ikeuchi Y, de la Iglesia N, Shen J, Bonni A. A Cdc20-APC ubiquitin signaling pathway regulates presynaptic differentiation. Science. 2009;326:575–578. doi: 10.1126/science.1177087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Yco LP, Mocz G, Opoku-Ansah J, Bachmann AS. Withaferin A Inhibits STAT3 and Induces Tumor Cell Death in Neuroblastoma and Multiple Myeloma. Biochemistry insights. 2014;7:1–13. doi: 10.4137/BCI.S18863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Yu H. Regulation of APC-Cdc20 by the spindle checkpoint. Curr Opin Cell Biol. 2002;14:706–714. doi: 10.1016/s0955-0674(02)00382-4. [DOI] [PubMed] [Google Scholar]
  147. Yu H. Cdc20: a WD40 activator for a cell cycle degradation machine. Mol Cell. 2007;27:3–16. doi: 10.1016/j.molcel.2007.06.009. [DOI] [PubMed] [Google Scholar]
  148. Yuan B, Xu Y, Woo JH, Wang Y, Bae YK, Yoon DS, Wersto RP, Tully E, Wilsbach K, Gabrielson E. Increased expression of mitotic checkpoint genes in breast cancer cells with chromosomal instability. Clin Cancer Res. 2006;12:405–410. doi: 10.1158/1078-0432.CCR-05-0903. [DOI] [PubMed] [Google Scholar]
  149. Zaravinos A, Lambrou GI, Boulalas I, Delakas D, Spandidos DA. Identification of common differentially expressed genes in urinary bladder cancer. PLoS One. 2011;6:e18135. doi: 10.1371/journal.pone.0018135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Zeng X, King RW. An APC/C inhibitor stabilizes cyclin B1 by prematurely terminating ubiquitination. Nat Chem Biol. 2012;8:383–392. doi: 10.1038/nchembio.801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Zeng X, Sigoillot F, Gaur S, Choi S, Pfaff KL, Oh DC, Hathaway N, Dimova N, Cuny GD, King RW. Pharmacologic inhibition of the anaphase-promoting complex induces a spindle checkpoint-dependent mitotic arrest in the absence of spindle damage. Cancer Cell. 2010;18:382–395. doi: 10.1016/j.ccr.2010.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Zhang J, Wan L, Dai X, Sun Y, Wei W. Functional characterization of Anaphase Promoting Complex/Cyclosome (APC/C) E3 ubiquitin ligases in tumorigenesis. Biochimica et biophysica acta. 2014;1845:277–293. doi: 10.1016/j.bbcan.2014.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Zhang L, Yang B, Zhou K, Li H, Li D, Gao H, Zhang T, Wei D, Li Z, Diao Y. Potential therapeutic mechanism of genistein in breast cancer involves inhibition of cell cycle regulation. Molecular medicine reports. 2015;11:1820–1826. doi: 10.3892/mmr.2014.2907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Zhang W, Cheng GZ, Gong J, Hermanto U, Zong CS, Chan J, Cheng JQ, Wang LH. RACK1 and CIS mediate the degradation of BimEL in cancer cells. J Biol Chem. 2008;283:16416–16426. doi: 10.1074/jbc.M802360200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Zhang W, Gong W, Ai H, Tang J, Shen C. Gene expression analysis of lung adenocarcinoma and matched adjacent non-tumor lung tissue. Tumori. 2014;100:338–345. doi: 10.1700/1578.17222. [DOI] [PubMed] [Google Scholar]
  156. Zur A, Brandeis M. Securin degradation is mediated by fzy and fzr, and is required for complete chromatid separation but not for cytokinesis. EMBO J. 2001;20:792–801. doi: 10.1093/emboj/20.4.792. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES