Elsevier

Pharmacology & Therapeutics

Volume 151, July 2015, Pages 141-151
Pharmacology & Therapeutics

Associate editor: B. Teicher
Targeting Cdc20 as a novel cancer therapeutic strategy

https://doi.org/10.1016/j.pharmthera.2015.04.002Get rights and content

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.

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 and 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 (Lipkowitz and Weissman, 2011, Bassermann et al., 2014, Wang et al., 2014a). 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, Zhang et al., 2014b, Wang et al., 2014a). 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 (Foe and Toczyski, 2011, Schreiber et al., 2011, Chang and Barford, 2014). 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 (Kulkarni et al., 2013, Chang and Barford, 2014, Chang et al., 2014). 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) (Fig. 1) (Vodermaier et al., 2003, McLean et al., 2011). Without a doubt, it is necessary to further determine the architectural details of the APC to aid in further understanding its biological functions.

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 et al., 2013b). 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) (Michaelis et al., 1997, Clute and Pines, 1999, 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 and Kirschner, 1998, Petersen et al., 2000), D-box (McGarry and Kirschner, 1998, Petersen et al., 2000, den Elzen and Pines, 2001, Geley et al., 2001, Bashir et al., 2004, Lindon and Pines, 2004, 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.

Consistent with this notion, although both Cdc20 and Cdh1 can activate the APC E3 ligase, they have distinct biological functions (Yu, 2002, Clijsters et al., 2013). For example, APCCdc20 exerts its function during the metaphase to anaphase transition through destruction of critical cell cycle regulators (Yu, 2007, Kim and Yu, 2011), whereas APCCdh1 plays a key role in the late M and G1 phases (Qiao et al., 2010, Hu et al., 2011). Moreover, Cdh1 is considered as a tumor suppressor, while Cdc20 exhibits its oncogenic function (Penas et al., 2011, Wang et al., 2013b). 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.

Section snippets

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

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, Reimann et al., 2001a, Kidokoro et al., 2008). 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

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.

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 TM Millennium Pharmaceuticals, Inc) and Carfizomib have

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 (Table 3). Therefore, more efforts are needed to discover other more specific Cdc20 inhibitors. We hope this article could stimulate more research efforts to

Conflict of interest

The authors declare no conflict of interest.

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.

References (155)

  • D. Hu et al.

    The emerging role of APC/CCdh1 in development

    Semin Cell Dev Biol

    (2011)
  • H.C. Huang et al.

    Evidence that mitotic exit is a better cancer therapeutic target than spindle assembly

    Cancer Cell

    (2009)
  • S.S. Jackson et al.

    Withaferin A disrupts ubiquitin-based NEMO reorganization induced by canonical NF-kappaB signaling

    Exp Cell Res

    (2015)
  • J. Jiang et al.

    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)
  • L. Jin et al.

    Mechanism of ubiquitin-chain formation by the human anaphase-promoting complex

    Cell

    (2008)
  • A.H. Kim et al.

    A centrosomal Cdc20-APC pathway controls dendrite morphogenesis in postmitotic neurons

    Cell

    (2009)
  • H.S. Kim et al.

    SIRT2 maintains genome integrity and suppresses tumorigenesis through regulating APC/C activity

    Cancer Cell

    (2011)
  • S. Kim et al.

    Mutual regulation between the spindle checkpoint and APC/C

    Semin Cell Dev Biol

    (2011)
  • X. Li et al.

    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 Lett

    (2015)
  • P. Majumder et al.

    Inhibiting tumor growth by targeting liposomally encapsulated CDC20siRNA to tumor vasculature: therapeutic RNA interference

    J Control Release

    (2014)
  • E. Manchado et al.

    Targeting mitotic exit leads to tumor regression in vivo: modulation by Cdk1, Mastl, and the PP2A/B55alpha, delta phosphatase

    Cancer Cell

    (2010)
  • T.J. McGarry et al.

    Geminin, an inhibitor of DNA replication, is degraded during mitosis

    Cell

    (1998)
  • C. Michaelis et al.

    Cohesins: chromosomal proteins that prevent premature separation of sister chromatids

    Cell

    (1997)
  • T. Nasr et al.

    Anticancer activity of new coumarin substituted hydrazide–hydrazone derivatives

    Eur J Med Chem

    (2014)
  • A. Ohtoshi et al.

    Human p55(CDC)/Cdc20 associates with cyclin A and is phosphorylated by the cyclin A-Cdk2 complex

    Biochem Biophys Res Commun

    (2000)
  • J. Adams

    The proteasome: a suitable antineoplastic target

    Nat Rev Cancer

    (2004)
  • T. Akiyama et al.

    Regulation of osteoclast apoptosis by ubiquitylation of proapoptotic BH3-only Bcl-2 family member Bim

    EMBO J

    (2003)
  • T. Akiyama et al.

    Bim-targeted cancer therapy: a link between drug action and underlying molecular changes

    Mol Cancer Ther

    (2009)
  • T. Akiyama et al.

    Bim: guardian of tissue homeostasis and critical regulator of the immune system, tumorigenesis and bone biology

    Arch Immunol Ther Exp (Warsz)

    (2011)
  • C. Andreu-Vieyra et al.

    Carfilzomib in multiple myeloma

    Expert Opin Biol Ther

    (2014)
  • M. Araki et al.

    Degradation of origin recognition complex large subunit by the anaphase-promoting complex in Drosophila

    EMBO J

    (2003)
  • T. Banerjee et al.

    DNA damage induced p53 downregulates Cdc20 by direct binding to its promoter causing chromatin remodeling

    Nucleic Acids Res

    (2009)
  • T. Bashir et al.

    Control of the SCF(Skp2-Cks1) ubiquitin ligase by the APC/C(Cdh1) ubiquitin ligase

    Nature

    (2004)
  • J. Bieniek et al.

    COX-2 inhibitors arrest prostate cancer cell cycle progression by down-regulation of kinetochore/centromere proteins

    Prostate

    (2014)
  • V.N. Budhavarapu et al.

    Regulation of E2F1 by APC/C Cdh1 via K11 linkage-specific ubiquitin chain formation

    Cell Cycle

    (2012)
  • T. Caravita et al.

    Bortezomib: efficacy comparisons in solid tumors and hematologic malignancies

    Nat Clin Pract Oncol

    (2006)
  • A. Castro et al.

    Xkid is degraded in a D-box, KEN-box, and A-box-independent pathway

    Mol Cell Biol

    (2003)
  • D.Z. Chang et al.

    Increased CDC20 expression is associated with pancreatic ductal adenocarcinoma differentiation and progression

    J Hematol Oncol

    (2012)
  • L. Chang et al.

    Molecular architecture and mechanism of the anaphase-promoting complex

    Nature

    (2014)
  • Z. Chen et al.

    Functional roles of PC-PLC and Cdc20 in the cell cycle, proliferation, and apoptosis

    Cell Biochem Funct

    (2010)
  • H.J. Cho et al.

    Degradation of human RAP80 is cell cycle regulated by Cdc20 and Cdh1 ubiquitin ligases

    Mol Cancer Res

    (2012)
  • J.W. Choi et al.

    High expression of spindle assembly checkpoint proteins CDC20 and MAD2 is associated with poor prognosis in urothelial bladder cancer

    Virchows Arch

    (2013)
  • A.C. Chun et al.

    REV7 is required for anaphase-promoting complex-dependent ubiquitination and degradation of translesion DNA polymerase REV1

    Cell Cycle

    (2013)
  • L. Clijsters et al.

    The spindle checkpoint, APC/C(Cdc20), and APC/C(Cdh1) play distinct roles in connecting mitosis to S phase

    J Cell Biol

    (2013)
  • P. Clute et al.

    Temporal and spatial control of cyclin B1 destruction in metaphase

    Nat Cell Biol

    (1999)
  • S. Cory et al.

    The Bcl2 family: regulators of the cellular life-or-death switch

    Nat Rev Cancer

    (2002)
  • M.S. Cragg et al.

    Unleashing the power of inhibitors of oncogenic kinases through BH3 mimetics

    Nat Rev Cancer

    (2009)
  • N. den Elzen et al.

    Cyclin A is destroyed in prometaphase and can delay chromosome alignment and anaphase

    J Cell Biol

    (2001)
  • Z.Y. Ding et al.

    Expression characteristics of CDC20 in gastric cancer and its correlation with poor prognosis

    Int J Clin Exp Pathol

    (2014)
  • Q.P. Dou et al.

    Overview of proteasome inhibitor-based anti-cancer therapies: perspective on bortezomib and second generation proteasome inhibitors versus future generation inhibitors of ubiquitin-proteasome system

    Curr Cancer Drug Targets

    (2014)
  • Cited by (0)

    1

    These authors contributed equally to this article.

    View full text