Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Semin Cancer Biol. 2020 Mar 9;67(Pt 2):80–91. doi: 10.1016/j.semcancer.2020.03.001

APC/C ubiquitin ligase: functions and mechanisms in tumorigenesis

Morgan S Schrock 1,2, Benjamin R Stromberg 1,2, Luke Scarberry 1,2, Matthew K Summers 1,ǂ
PMCID: PMC7483777  NIHMSID: NIHMS1582920  PMID: 32165320

Abstract

The anaphase promoting complex/ cyclosome (APC/C), is an evolutionarily conserved protein complex essential for cellular division due to its role in regulating the mitotic transition from metaphase to anaphase. In this review, we highlight recent work that has shed light on our understanding of the role of APC/C coactivators, Cdh1 and Cdc20, in cancer initiation and development. We summarize the current state of knowledge regarding APC/C structure and function, as well as the distinct ways Cdh1 and Cdc20 are dysregulated in human cancer. We also discuss APC/C inhibitors, novel approaches for targeting the APC/C as a cancer therapy, and areas for future work.

Keywords: Anaphase promoting complex/ cyclosome, Cdc20, Cdh1, Genome instability, Cancer

I. Introduction: APC/C is an essential E3 ubiquitin ligase

The anaphase promoting complex/ cyclosome (APC/C), is an evolutionarily conserved multi-subunit E3 ubiquitin ligase that is primarily known for its ability to trigger the metaphase to anaphase transition during mitosis. Although other functions of the APC/C have been described, its pivotal role in cellular division make it essential to viability and development, and also implicate its dysregulation in tumorigenesis. In this review, we highlight the involvement of APC/C in tumorigenesis and discuss current strategies for targeting APC/C as a cancer therapy. First, we will briefly summarize the current state of knowledge regarding APC/C structure and function. We will then discuss the distinct ways APC/C is dysregulated in human cancer in the context of its two main regulators: Cdc20 (cell division cycle 20 homologue, also called Fizzy) and Cdh1 (Cdc20 homologue 1, also known as Fizzy-related protein 1, FZR1)[1, 2]. Lastly, we will identify current approaches to targeting APC/C for therapeutic benefit and discuss areas that need to be addressed in order to fully exploit APC/C dysregulation as a cancer therapy.

APC/C Structure and Function

Human APC/C is composed of 14 proteins for a total of 19 subunits, which can be categorized into three subcomplexes: the catalytic core (APC2, APC11, and APC10), the platform (APC1, APC4, APC5, and APC15), and the tetratricopeptide repeat (TPR) lobe (APC3, APC6, APC7, APC8, APC12, APC13, and APC16) [38] (Figure 1). APC1 is the largest subunit within the complex and acts to bridge the TPR lobe, platform and catalytic core. The catalytic core is responsible for APC/C E3 ubiquitin ligase activity, which allows it to catalyze the generation of poly-ubiquitin chains upon its substrates, triggering their degradation by the 26S proteasome. The attachment of ubiquitin is a multi-step process involving a ubiquitin-activating enzyme (E1), a ubiquitin-conjugating enzyme (E2), and a ubiquitin ligase (E3). There are two E1 enzymes in the human genome, approximately 40 E2 enzymes, and over 500 E3 ligases. This review focuses on the E3 ligase APC/C, which interacts with two predominant E2s: UbcH10 and Ube2s and to a lesser extent UbcH5 [916]. To begin the ubiquitination process, ubiquitin is activated through an ATP-dependent interaction with E1. The activated ubiquitin is then transferred to E2, which is recruited by E3, in this case, APC/C. The APC/C subunit Apc11 RING H2 domain interacts with the E2, which catalyzes the transfer of ubiquitin from the E2 active site to a lysine on the target protein [4, 1720]. Repeating this process generates a chain of ubiquitins on the substrate, labeling it for degradation by the 26S proteasome.

Figure 1.

Figure 1.

Structure of the APC/C including Cdh1 and substrate, Emi1. Image created in PyMOL using structure PDB 4UI9.

The ubiquitination of substrates by the APC/C requires the formation of an APC/C-activator-substrate complex. The APC/C is controlled by two main activators, Cdc20 and Cdh1, each targeting APC/C to specific substrates at precise times during the cell cycle. Binding of Cdc20 or Cdh1 to the APC/C holoenzyme induces a conformational change to the active forms, APC/CCdc20 and APC/CCdh1, respectively. The Cdc20 and Cdh1 WD40 domains located at their C termini provide a binding platform to recruit APC/C substrates, which in concert with APC10 functions as the substrate receptor of the APC/C [19, 2124]. APC/CCdc20 is present at the onset of mitosis until the beginning of anaphase (Figure 2A). In early mitosis, Cdc20 becomes active and associates with phosphorylated APC/C leading to the degradation of prometaphase substrates, such as cyclin A and Nek2 [2529]. Once all chromosomes achieve proper microtubule biorientation in metaphase, APCCdc20 degrades securin and cyclin B to promote cellular transition to anaphase. Degradation of securin releases separase, an enzyme that is responsible for cleaving Scc1, a component of the cohesion complex that holds sister chromatids together, and allows physical separation of sister chromatids. Degradation of cyclin B abolishes cyclin-Cdk activity at anaphase and allows the activation of APC/CCdh1, which destroys the remaining mitotic regulators, including Cdc20 [26, 30]. APC/CCdh1 is present at the onset of anaphase through G1 and promotes efficient mitotic exit and maintenance of the G1 state (Figure 2A).

Figure 2.

Figure 2.

APC/C activity fluctuates throughout the cell cycle and regulates cell cycle progression. A) APC/CCdc20 increases during mitosis and regulates the cell cycle through cellular division. During late mitosis APC/CCdh1 accumulates and ubiquitinate Cdc20 and regulates exit from mitosis. Once in G1 APC/CCdh1 acts as a tumor suppressor and halts cells in a quiescent state. Once APC/CCdh1 is degraded cells progress into S-phase. B) Substrate recognition and regulation of APC/CCdh1 is dependent on the cell cycle. During G1, APC/CCdh1 is active and suppresses the activity of Emi1 and Cyclin F (amongst others). However, as cells transition into S-phase Cdk2 phosphorylates, therefore deactivating, Cdh1 which allows Emi1, Cyclin F, and SCFβTRCP to further inhibit APC/CCdh1. Although Cdh1 levels are low during S-phase and G2 it can still function by ubiquitinating the tumor suppressor FBXO31. However, this interaction is disrupted if FBXO31 is phosphorylated by the DNA damage response kinase ATM.

Substrates targeted for degradation by the APC/C are as varied as the processes they regulate and include cell cycle regulators such as the mitotic cyclins, Securin, and Geminin, as well as the transcription and differentiation inhibitor Id2, the TGF-β regulator SnoN, the replication checkpoint regulator Claspin, and the adaptive glycolytic enzyme Pfkfb3 [3138]. E3s recognize substrates that contain cis-acting sequence motifs, also known as degrons. The degrons recognized by the APC/C are the destruction-box (D-box) (RXXLXXI/VXN) and/or KEN-box (KENXXXN/D). However, other APC/C-targeting/interacting sequences have been identified such as Cry-box, C-box, and O-box and are thought to interact with the adaptors in a similar fashion to the KEN and D-box. [39, 40]. APC/C substrate degrons directly interact with Cdc20 and Cdh1 via their WD40 domains, which leads to their ubiquitination by E2s and degradation by the 26S proteasome [4, 22, 23, 40, 41].

Because APC/C contributes to many different cellular pathways depending on which activator is bound, a more in depth examination of APC/C activity in respect to Cdc20 or Cdh1 is necessary to review its diverse pathway contributions to cancer development. The key cancer-related interactions of Cdc20 and Cdh1 detailed in the following sections are summarized in Table 1.

Table 1.

APC/C Interactors and Implications in Cancer

Activator Interactor Effect on Interactor Role in Cancer APC/C- Dependent Reference
Cdc20 Axin Degradation Axin degradation leads to increased β-Catenin signaling. Yes 7172
SOX2 Stabilization Promotes self-renewal of CSCs Yes 74
p21CIP1/WAF1 Degradation Promotes CSCs tumorigenicity Yes 68
Bim Degradation Attenuates induction of apoptosis by anti-mitotic agents. Yes 80
Mcl-1 Degradation? Cdc20-Mcl-1 interaction may alter the timing of mitotic slippage in drug- induced arrest Yes 8183
SMAR1 Degradation Reduces tumor suppressor function of SMAR1 Yes 70
SPOP Degradation of Cdc20 SPOP mutations in prostate cancer lead to increased Cdc20 levels. Yes 90
Cdh1 FBXO31 Degradation Akt-induced degradation of FBXO31 by APC/CCdh1 attenuates its tumor suppressor function. Yes 122
SKP2 Degradation Inhibits cell growth promoting functions of Skp2 such as triggering destruction of p27 Yes 123, 124
STIL Degradation Low levels of STIL lead to elevated centriole numbers and promote genome instability Yes 136139
CENP-A Degradation Elevated CENP-A promotes chromosomal instability Yes 140
SPOP Degradation Degradation of SPOP alleviates its inhibition of PD-L1 protein levels. Yes 146
BRAF Degradation Limits BRAF activity, patients with mutation in the targeting degron may lead to a higher higher incidence of skin cancer. Yes 126, 148
PAX3 Degradation Increased PAX3 levels drive melanoma proliferation and chemoresistance. Yes 149
WWP2 Cdh1 binding holds WWP2 in an inactive conformation. Cdh1-mediated inhibition of WWP2 stabilizes WWP2 substrates such as the tumor suppressor PTEN No 151
PTEN None PTEN promotes APC/C-Cdh1 complex formation and activity Yes 153
Src Cdh1 binding holds Src in an inactive conformation. Src phosphorylates Cdh1 preventing binding to the APC/C Cdh1-binding limits the activity of the oncogene Src, which promotes proliferation. Src activity reduces tumor suppressor activity of APC/CCdh1 No 127

II. APC/CCdc20 Regulation and Alteration in Cancer

APC/CCdc20 is inactive throughout most of the cell cycle due to competition with Cdh1. During a majority of the cell cycle, Cdc20 levels are low and the APC/C lacks sufficient phosphorylation for the formation of the activated APC/CCdc20 complex, allowing Cdh1 to outcompete it in binding to the APC/C [28, 42, 43]. APCCdc20 is inhibited to allow the G2/M transition through three mechanisms [37]. The first mechanism is via inhibition by Emi1. Although primarily thought of as a regulator of APC/CCdh1 at the G1/S transition, Emi1 is a potent inhibitor of APC/CCdc20 as well and may contribute to APC/CCdc20 regulation during the G2/M transition and early mitosis [44, 45]. Indeed, although the majority of Emi1 is degraded in G2 to allow mitotic progression the remaining pool is important for assembly of the mitotic spindle [4649]. The second method of inhibition utilizes Mad1 and Mad2, where a complex of Mad1-Mad2 converts cytosolic Mad2 from its open conformation (O-Mad2) to its closed conformation (C-Mad2). C-Mad2 then binds Cdc20, inhibiting it from binding to the APC/C [37, 5057]. During mitosis, the spindle assembly checkpoint (SAC) is activated by unattached kinetochores (Figure 3A). The SAC induces generation of the mitotic checkpoint complex (MCC), consisting of C-Mad2, Cdc20, BubR1, and Bub3, which functions to inactivate APC/C Cdc20 [5861]. This is accomplished through the generation of C-Mad2 at the kinetochore. C-Mad2 is generated in the same fashion that allows for the G2/M transition. Upon C-Mad2 binding to Cdc20, the MCC assembles. Once all kinetochores have properly attached to microtubules, the MCC disassembles allowing for the activation of APC/CCdc20 (Figure 3B). APC/CCdc20 leads to the subsequent degradation of Cyclin B and Securin to allow for mitotic progression (Figure 3C). The third method of regulating APC/CCdc20 is accomplished through the phosphorylation of Cdc20, reducing its binding affinity for APC/C. Cdk1 is able to phosphorylate Cdc20, which reduces its affinity for the APC/C and allows for the accumulation of Cyclin B, allowing for the G2/M transition [37]. Similarly, Bub1 and Plk1 phosphorylate and inhibit APC/CCdc20 independent of the MCC [62]. In mitosis Cdc20 must then be dephosphorylated to become activated, which is mediated by the PP2AB56 phosphatase, a known tumor suppressor [63]. This phosphatase is essential for the activation of APC/CCdc20. Upon depletion of functional PP2AB56 cells had a reduction in their ability to exit mitosis. This dysfunction resulted from an inability of Cdc20 to associate and activate the APC/C [64]. Proper regulation of Cdc20 contributes to mitotic maintenance and genome stability.

Figure 3.

Figure 3.

Schematic of APC/C silencing and activation associated with the Spindle Assembly Checkpoint. A) Unattached kinetochores at the metaphase plate activate the spindle assembly checkpoint, inducing formation of C-Mad2 at the kinetochore. C-Mad2 binds to Cdc20, stimulating the formation of the mitotic checkpoint complex, consisting of Bub3, BubR1, Mad2, Cdc20, and APC/C. Upon assembly of the MCC, the APC/C is inactivated, inhibiting the progression from metaphase to anaphase. B) Once all kinetochores are correctly attached, it signals for the disassembly of the MCC, allowing for the activation of APC/CCdc20. C) Activated APC/CCdc20 ubiquinates Cyclin B and Securin, leading to their subsequent degradation by the 26S proteasome and the transition from Metaphase to Anaphase.

The role of Cdc20 in tumorigenesis

In recent years, overexpression of Cdc20 has been associated with a multitude of different cancers such as prostate, glioblastoma, bladder, breast, oral, and diffuse large B cell and mantle cell lymphomas [6570]. Overexpression of Cdc20 allows a cell to bypass the SAC and exit mitosis prematurely, thus leading to genomic instability. This overexpression commonly results in a poor prognosis for patients due to Cdc20’s effects on a variety of cellular processes. Cdc20 appears to play a strong role in the maintenance of tumors through its expression in cancer stem-like cells (CSCs)/tumor initiating cells (TICs), as discussed below.

Overexpression of Cdc20 has also been correlated with activation of the WNT/ β-catenin pathway, which is strongly associated with the progression of cancer [7173]. Canonically, upon activation of WNT, β-catenin is stabilized and translocated into the nucleus. Upon translocation, β -catenin interacts with T-cell factor/lymphoid enhancing factors (TCF/LEF) family of DNA-binding factors and activates genes such as stromelysin, fibroblast growth factor (FGF), epidermal growth factor (EGF), cyclin D1, c-Myc, CD44 and ALDH [67]. Inhibition of Cdc20 in cutaneous squamous cell carcinoma through RNA interference resulted in a reduction in β-catenin. While not showing a direct interaction between Cdc20 and β -catenin, it is apparent that Cdc20 plays a role in regulating β-catenin [72]. Another recent study revealed a distinct mechanism through which β-catenin is stabilized in prostate CSCs. Cdc20 was shown to induce the degradation of Axin1, a key regulator of β-catenin, through its proteasome activity [71]. This provides a direct relationship for Cdc20 contributing to the upregulation of β-catenin and leading to cancer progression.

Another mechanism whereby CSCs/TICs can be maintained by APC/CCdc20 is accomplished through the SOX2 signaling pathway. SOX2 is a transcription factor associated with pluripotency [74]. APC/CCdc20 was shown to form a complex with nuclear SOX2 and to maintain its stability, contributing to neoplastic cells’ ability for self-renewal in glioblastoma. It remains unclear on how this stability is achieved, however the authors speculate it is possible that APC/CCdc20 alters post-translational modifications on SOX2, thus providing stabilization. TICs can also be maintained by Cdc20 through the degradation of p21CIP1/WAF1, which plays a pivotal role in suppressing TICs. p21CIP1/WAF1 functions as a CDK inhibitor and therefore plays an important role in cell cycle control [75, 76]. Importantly, upon overexpression of Cdc20, TICs increased their ability to generate tumors in vivo, highlighting the oncogenic potential of Cdc20. [68]

In recent years Cdc20 has appeared to play a role both in the positive and negative regulation of apoptosis. This was thought to be accomplished through APC/CCdc20 targeting both Bim and Mcl-1 for destruction. As death elicited by anti-mitotic agents (e.g., taxanes) is modulated by both the cells’ ability to induce apoptosis and the duration of the arrest, these observations suggested that APC/CCdc20 plays central roles in the cellular response to anti-mitotics by coordinating both fate-determining pathways [7779]. Bim was shown to specifically bind to Cdc20’s WD-40 motif, thus allowing for the polyubiquitination of Bim and leading to its subsequent degradation and promoting resistance to apoptosis induction by anti-mitotic agents [80]. In contrast, APC/CCdc20 was also implicated to play a role in the degradation of Mcl-1, a pro-survival protein, due to evidence of Mcl-1 interacting with APC/CCdc20 [81, 82]. However, further study failed to elucidate a clear relationship of Mcl-1 and Cdc20-mediated degradation. Rather, it appears that Mcl-1 may not be dependent on ubiquitin ligases for degradation and that Mcl-1 may function to regulate Cdc20 activity toward substrates such as Cyclin B, thus altering the rate of mitotic slippage and affecting induction of death by prolonged mitotic arrest [83]. Cdc20’s role in regulating apoptosis has clear implications for therapeutic response and it will be important to understand further whether cell or tissue-specific differences may influence its ability to coordinate the mechanisms regulating mitotic slippage and cellular survival.

Cdc20 also plays a role in the regulation of SMAR1, a tumor suppressor that functions to regulate the genome, the cell cycle, and apoptosis [8487]. SMAR1 works by activating p53 to arrest cells at the G1 and G2/M transition. It is also involved in the DNA damage repair pathway, as well as ensuring the proper splice variant of CD44, a putative cancer stem cell marker and modulator of metastasis, is produced [70, 88, 89]. In higher grades of cancer, SMAR1 levels are often reduced, in contrast to high levels of Cdc20. Further research, using breast cancer, ovarian, melanoma, cervical, and colon cancer cell lines, elucidated that Cdc20 regulates the degradation of SMAR1 through ubiquitination. It is important to note that under stress, Cdc20 did not interact with SMAR1, possibly due to SMAR1 playing an active role in the DNA damage repair response [70].

Given the numerous cancers with reported overexpression of Cdc20, it is important to know how this upregulation occurs. While upregulation of Cdc20 transcription by deregulation of the Rb-E2F pathway or amplification have been described, post translational control of protein stability or function may also contribute to enhanced APC/CCdc20 activity in cancer. One potential route for post-translational upregulation is through Speckle-type POZ protein (SPOP). SPOP functions as a tumor suppressor through its function as a substrate-interacting adaptor protein of CRL3, an E3 ubiquitin ligase, specifically by targeting known oncogenic proteins such as ci/Gli, androgen receptor, SRC-3, DEK, TRIM24, MacroH2A, SENP7, and ERG for degradation. SPOP has been shown to often be mutated in cases of prostate cancer and prostate cancer-associated mutations in the MATH domain of SPOP were shown to abolish the interaction with Cdc20. The lack in interaction resulted in a reduction in the polyubiquitination of Cdc20, stabilizing it and inhibiting its degradation through the proteasome pathway [90]. With the prevalence of Cdc20 overexpression in cancers, it is probable that there are a multitude of mechanisms in which Cdc20 is overexpressed, however these mechanism continue to be elucidated.

Overall, Cdc20 allows for the progression of cancer through several pathways, allowing for increased tumor proliferation and metastasis. Importantly, Cdc20’s role in cancer development and progression is not limited to a specific type of cancer. The diverse roles Cdc20 plays in tumorigenesis makes it a potent oncogenic factor and a potential target for therapeutic intervention.

III. APC/CCdh1 Regulation and Alteration in Cancer

Cdh1 was the second APC/C coactivator discovered, hence the name Cdc20 homolog1 [1]. It is also referred to as FZR1 (based on the drosophila homolog fizzy-related 1) but it should not be confused with the gene CDH1 (e-cadherin). The general function of this coactivator is similar to Cdc20, however it is primarily active during late mitosis and G1 (Figure 2A). The ability of APC/CCdh1 to regulate the cell cycle is the most well understood function of this E3 ubiquitin ligase. During late mitosis APC/CCdh1 aids in the transition into G1 by initiating the degradation of mitotic proteins such as Cdc20 [91], Plk1 [92], and Aurora kinases A [93] and B [94, 95]. However, it should be noted that Cdh1 is not essential for mitotic exit [93, 96]. Once in G1, APC/CCdh1 continues to destabilize mitotic proteins to maintain a G1 state. Additionally, Cdh1 targets proteins such as Cdc25A [97], Skp2 [98], and USP37 [99] to delay the transition from G1 to S. In order to inactivate APC/CCdh1, and progress into S-phase, a number of factors act on APC/CCdh1 to diminish its activity and allow the accumulation of S-phase proteins. Some of these factors include Emi1, which binds to and inactivates APC/CCdh1 [4, 44, 100102], Skp1-Cul1-F-box (SCF)BTRCP and SCFCyclin F, which ubiquitinate Cdh1 [103, 104], and Cyclin/Cdk complexes, which phosphorylate Cdh1 to reduce binding of Cdh1 to APC/C [4, 105108] (Figure 2B). The ability of APC/CCdh1 to regulate the cell cycle is highly regulated and has clear connections to cancer, however most of these topics have been thoroughly covered in previous reviews [109]. Reviews have also covered important topics such as structural insights into APC/C [110, 111], the function of APC/CCdh1 in the nervous system [112], as well as how APC/C uses degrons to recognize substrates [41]. The following sections will discuss more recent publications and highlight current research that has improved our understanding of Cdh1 and cancer. Specifically, as it relates to cell cycle regulation, genome stability, and other cancer related pathways. We will also highlight some of the APC/C independent functions of Cdh1 that have been discovered in the last few years.

APC/CCdh1 and Cell Cycle Regulation

Although there have been copious amounts of research focused on how APC/CCdh1 regulates the cell cycle, new insights continue to be uncovered. One of the most significant recent findings demonstrated how Cdh1 inactivation is a critical step during S-phase entry [113]. Normal human cells will spend a majority of their time in a quiescent state. Therefore, advancement into S-phase is a significant progression that commits the cell to division and proliferation. Earlier understanding of S-phase entry focused on the importance of the tumor suppressor retinoblastoma (Rb) and the transcription factor E2F. During G1, Rb represses E2F by binding to the transcription factor [114]. As cells move into S-phase cyclin E accumulates and activates Cdk2 which phosphorylates Rb and releases E2F [115]. Once E2F is released it localizes to the nucleus and increases the expression of S-phase proteins. It was previously understood that the phosphorylation of Rb was the point at which cells were committed to S-phase, however recent work on APC/CCdh1 has challenged this theory. It was observed that Cdk2 temporally phosphorylates Rb followed by Cdh1, which inactivates APC/CCdh1 by displacing the coactivator [113]. Interestingly, human cells were able to return to a quiescent state when Rb was phosphorylated but not when APC/CCdh1 was inactivated. This suggests that APC/CCdh1 inactivation is the point at which cells are committed to the cell cycle. Furthermore, it has been shown that once APC/CCdh1 is inactivated by Cdk2, Emi1 transitions from an APC/CCdh1 substrate to an APC/CCdh1 inhibitor [116] (Figure 2B). These findings that human cells commit to S-phase entry following the inactivation of APC/CCdh1 by Cdk2 phosphorylation and an increase in Emi1 levels further support APC/CCdh1 as an important tumor suppressor, which like Rb and p53 , plays a key role in preventing inappropriate entry into the cell cycle.

Although APC/CCdh1 activity is greatly reduced beyond the G1/S transition there have been some studies that indicate Cdh1 does contribute to cell cycle control during G2. Several recent studies extend these findings to demonstrate additional interplay between the APC/C and SCF ligases. FBXO31 is a substrate adaptor for the E3 ligase SCF, and it is known as a tumor suppressor that is commonly mutated in breast, ovarian, and prostate cancers [117121]. Recently, it was identified as an APC/CCdh1 substrate, however the ability of APC/CCdh1 to recognize FBXO31 is dependent on two different kinases, AKT and ATM [122]. It was observed that in unstressed HEK293T cells, the pro-survival kinase and oncogene AKT would phosphorylate FBXO31 and target it for ubiquitination by APC/CCdh1 (Figure 4). Conversely, when HEK293T cells were stressed with ionizing radiation, ATM would phosphorylate FBXO31 and rescue it from APC/CCdh1 targeting [122] (Figure 2B). These results suggest Cdh1 plays an important role during ATM-mediated cell cycle arrest and DNA damage repair. AKT activity also regulates the ability of APC/CCdh1 to recognize other SCF components, Skp2 [123] and cyclin F [103] (Figure 4). Skp2 recognizes and ubiquitinates the cyclin dependent kinase inhibitor p27 [124]. AKT stabilizes Skp2 by phosphorylating it and disrupting binding between Skp2 and APC/CCdh1 [123]. Therefore, once AKT is active Skp2 degrades p27 and the cell cycle progresses. Interestingly, SCFCyclin F ubiquitinates Cdh1, and conversely APC/CCdh1 ubiquitinates Cyclin F. This regulatory loop is broken when AKT phosphorylates and stabilizes Cyclin F [125]. Once Cyclin F is stable, it then begins to ubiquitinate and destabilize Cdh1 leading to cell cycle progression. All together these experiments detail a novel mechanism in which AKT activity modulates Cdh1 activity and consequently regulates the cell cycle (Figure 4).

Figure 4.

Figure 4.

APC/CCdh1 substrate recognition is regulated by AKT activity. During a quiescent state APC/CCdh1 will degrade substrates such as Skp2 and Cyclin F. Downregulation of these proteins allow cells to remain in G1/G0. However, once AKT becomes active it phosphorylates Skp2, Cyclin F, and FBXO31. This perturbs the interaction between Cdh1 and Skp/Cyclin F. Conversely, phosphorylation of FBXO31 recruits it to APC/CCdh1. Ultimately, AKT mediated regulation of these substrates contributes to cellular survival and proliferation.

APC/CCdh1 Regulates Genome Stability

Cdh1 has been characterized as a tumor suppressor mainly because mice with heterozygous Cdh1 knock-out develop tumors [96]. Similarly, cultured melanocytes are transformed when Cdh1 and PTEN are knocked down [126], immortalized breast epithelial cells and colonic fibroblasts are transformed by loss of Cdh1, and loss of Cdh1 increases the proliferation of breast cancer cell lines [127]. Recent work has tried to elucidate why these Cdh1 deficient mice develop tumors and has identified that Cdh1 is important in regulating genome stability and the DNA damage response [93, 128135]. Specifically, APC/CCdh1 has an important role regulating chromosomal instability and DNA replication. Mechanistic studies have shown that APC/CCdh1 is capable of regulating levels of the centrosome factor STIL in human cells [136]. The dysregulation of STIL is known to cause chromosomal rearrangement and acute lymphoblastic leukemia [137]. Therefore the mechanisms that control the levels of STIL are highly important. Interestingly, it has been observed that disrupting APC/CCdh1 control of STIL, by deleting the KEN box degron, caused an increase in centriole numbers [138]. This observation has since been reproduced by studying the interaction between Cdh1 and the STIL homolog, SAS-5, in C. elegans [139]. Other studies have identified the histone H3 variant, CENP-A, as a novel substrate of APC/CCdh1 in Drosophila which further connects APC/CCdh1 to chromosomal instability [140]. CENP-A localizes to and identifies centromeres. It has been observed that overexpression, and consequently mislocalization, of CENP-A contributes to aberrant kinetochore formation and chromosomal missegregation [141]. Therefore, dysregulation of APC/CCdh1 activity in cancer could be leading to more CENP-A expression and increased genome instability.

Cdh1 also protects genome integrity by regulating replication stress. The loss of Cdh1 is associated with replication errors leading to DNA damage and genomic instability [93, 142]. More specifically, there were slower replication forks, and consequently DNA breakage, when Cdh1 was knocked out [143]. While the shortened G1 phase of Cdh1-deficient cells may contribute to replication stress by multiple mechanisms, recent research in mouse embryonic fibroblasts suggests that the most likely cause of replication stress in the absence of Cdh1 is a decrease in the abundance of dNTPs [143].There are several reasons why loss of Cdh1 causes reduced dNTP levels. First, Cdh1 knock-down cells have a shorter G1, enter S-phase prematurely, [144] and may simply not have enough time to accumulate the appropriate amount of dNTPs as suggested by the ability of prolonging G1 in these cells to improve genomic integrity [143]. Second, it is known that a decrease in Cdh1 can lead to an increase of Cyclin F (which is an APC/CCdh1 substrate) [103]. As mentioned previously, Cyclin F is an E3 ligase substrate adaptor and it is known to negatively regulate critical subunits of ribonucleotide reductase (which is an essential enzyme in dNTP synthesis) [145]. Therefore, loss of Cdh1 could indirectly reduce the ability of cells to synthesize dNTPs by destabilizing subunits of ribonucleotide reductase. It will be interesting in future work to understand how the replication stress caused by loss of Cdh1 contributes to that induced by oncogenes to generate genomic instability.

The Role of Cdh1 in other Cancer Related Pathways

APC/CCdh1 has clear connections to cancer by regulating cell cycle control and genome stability, however Cdh1 has also been connected to other cancer related pathways. For instance, APC/CCdh1 can increase the ability of cancer cells to temper the immune response by indirectly regulating the levels of programmed death-ligand 1 (PD-L1) [146] (Figure 5). Cancer cells with increased PD-L1 suppress the adaptive immune response by destroying active T-cells and B-cells. A recent study identified that PD-L1 is ubiquitinated by the E3 ligase Cullin3SPOP, and that SPOP is ubiquitinated by APC/CCdh1 [146]. Therefore, when APC/CCdh1 is active PD-L1 levels increase which allows neoplastic cells to subdue the immune response. This is a unique function of Cdh1 because in most instances Cdh1 functions as a tumor suppressor.

Figure 5. (Boxes).

Figure 5.

Cdh1 regulates immune response and cell signaling in an APC/C dependent and independent manner.

For instance, APC/CCdh1 is able to regulate the MEK/ERK oncogenic signaling cascade (Figure 5). In normal cells the MEK/ERK pathway is stimulated by an extracellular signal that activates EGFR and stimulates a kinase cascade that leads to cell growth and proliferation. BRAF is an important oncogenic kinase in the MEK/ERK pathway and is constitutively active in many cancer types such as skin, lung, colorectal, and brain [147]. EGFR and BRAF inhibitors can suppress MEK/ERK signaling and are currently being used to treat patients with cancer. Interestingly, APC/CCdh1 is also able to suppress MEK/ERK signaling by directly ubiquitinating and destabilizing BRAF in human cells [126]. Further investigation showed that melanocytes have decreased Cdh1 expression, and increased BRAF expression, when exposed to UV radiation [126, 148, 149]. Researchers were also able to identify that Cdh1 interacts with BRAF through the degron D-box 4. According to the Catalog of Somatic Mutations in Cancer, mutations in D-box 4 (R671Q) have been found in patients with skin cancer (mutation Id COSM159405, cancer.sanger.ac.uk). Researchers were able to show that BRAF with a R671Q mutation showed a decrease in Cdh1 binding and increased BRAF stability [126]. This data suggests that when melanocytes are subjected to UV light there is a decrease in APC/CCdh1 function and an increase in MEK/ERK signaling because of the increase in BRAF expression. Additionally, patients with BRAF mutations in D-box 4 could have a high risk of developing cancers because APC/CCdh1 is unable to regulate BRAF expression. In addition to BRAF, loss of Cdh1 function in melanocytes leads to increased stability of PAX3 an upstream transcriptional regulator of the oncogene MITF that is sufficient to promote melanoma cell proliferation and resistance to chemotherapies [149].

Functions of Cdh1 Independent of APC/C

Most of the research that focuses on the role and regulation of Cdh1 in cancer has concentrated on its APC/C-dependent functions. However, there have been some recent studies that have demonstrated the ability of Cdh1 to regulate cancer associated functions independent of the APC/C (Figure 5). One of these pathways is the PTEN regulation of PI3K/AKT. As previously mentioned AKT is a prosurvival kinase that phosphorylates a number of substrates and can regulate processes such as metabolism, proliferation and cell migration. PI3K is another kinase that is upstream of AKT and initiates its activation. PTEN is a phosphatase and one of the major antagonists of this pathway, and importantly loss of PTEN is common in cancers and leads to increased PI3K/AKT signaling [150]. Novel research has shown that decreasing the amount of Cdh1 in cancer cells caused a decrease in WWP2 substrates such as PTEN [151]. WWP2 is an E3 ligase that ubiquitinates PTEN [152]. Interestingly, the ability of Cdh1 to inhibit WWP2 did not depend on the ability of Cdh1 to interact with APC/C. Conversely, Cdh1 directly binds to WWP2 and locks the E3 ligase in an inactive conformation [151]. Interestingly, another study showed that nuclear PTEN is capable of recruiting Cdh1 to APC/C [153]. This means that there is a positive feedback loop between APC/CCdh1 and PTEN. Both are capable of positively regulating each other and this strengthens tumor suppressor capabilities.

Cdh1 has also been show to regulate the non-receptor tyrosine kinase Src [127]. This kinase was one of the first oncogenes identified and is responsible to transducing extracellular signaling. Src targets many substrates and therefore has been connected to numerous pathways and cellular functions. Most commonly, Src has been shown to regulate proliferation, migration, and apoptosis [154]. Given the oncogenic potential of Src it is important to understand how its expression and activity is regulated in human cells. In breast cancer cells lines it was observed that Src activity increased when Cdh1 was knocked down, but there was no change in the abundance of Src [127]. Typical APC/CCdh1 substrates have an increase in expression when Cdh1 is knocked down. Through immunoprecipitation experiments it was determined that Cdh1 was binding to the N and C terminus ends of Src and locking it in it inactive conformation [127]. This is a similar mechanism to how Cdh1 inactivates WWP2. Ultimately these studies suggest Cdh1 should not be exclusively characterized as a coactivator for APC/C. It is becoming clear Cdh1 has important functions that are ubiquitin independent (Figure 5).

The diverse nature of the processes that Cdh1 participates in provide a likely explanation of its role as a haploinsufficient tumor suppressor. First, due to the many events that it regulates in the cell cycle alone, it is advantageous for cancer cells to maintain some level of Cdh1 activity. Second, while some of the oncogenic targets of Cdh1 (both APC/C-dependent and -independent) can have enhanced activity by amplification or other Cdh1-independent mechanisms, which may contribute to tumorigenesis, in general it seems that the combined deregulation of multiple pathways by partial reduction in Cdh1 function would be sufficient to create a permissive environment for the evolution of neoplastic cells.

IV. APC/C Subunits in Cancer

Heterozygous mutations of APC/C subunits were identified nearly two decades ago in colorectal cancer cell lines as well as a primary colorectal tumor [155]. Four of five mutations identified in that study impacted proteins of the TPR lobe with two mutations causing truncation of APC6 or APC8 and it was suggested that these mutations impaired mitotic progression. A third subunit of the TPR lobe, CDC27, is down-regulated in breast cancer and mutated in colorectal and testicular germ cell tumors leading to its characterization as a tumor suppressor [156159]. Although not necessarily prevalent, examination of the cancer genome atlas (TCGA) datasets revealed that mutation of at least one APC/C subunit can be found in up to 24% of cases, depending on the cancer [160]. Interestingly, a relatively high frequency of mutations are truncating and many others are predicted to have impacts on APC/C function based on the analysis of the APC/C structure. CRISPR/Cas9-mediated correction of the previously discovered APC8 truncation mutant in HT29 cells accelerated mitotic progression in these cells whereas heterozygous introduction of patient-derived CDC27, CDC16, APC1, or APC4 truncation mutants into multiple cell lines lengthened mitosis. While the idea that cancer cells select for a delayed mitosis may be unexpected, analysis of chromosome segregation in these cellular models demonstrated that weakened APC/C activity and prolonged mitosis allow cells to limit chromosomal instability (CIN) either in cells with high rates of CIN or after chemical induction of CIN via MPS1 inhibition resulting in resistance to these compounds. Similar attenuation of CIN was observed upon modest inhibition of APC/C by proTAME or perturbation of APC/C function via deletion of APC7, APC16 (which has a high proportion of truncation mutants), or the E2 enzymes UBE2C and UBE2S [160]. Consistent with these reports, down-regulation of multiple APC/C subunits has also been found to confer resistance to MPS1 inhibitors and expression of APC/C subunits was found to correlate strongly with cellular response to MPS1 inhibition in panels of breast cancer or lung cancer cell lines [161]. Examination of TCGA breast cancer data in this study identified decreased expression of at least one APC/C subunit in 46% of cases. Together these results suggest that cancer cells select for events (e.g., mutations) that weaken APC/C activity as a means to limit chromosomal instability and could also be a mechanism of resistance to therapies that target mitosis. In contrast, overexpression of UbcH10 or expression of a Cdc20 mutant that is refractory to SAC, enhance APC/C activity and are associated with increased CIN [162, 163]. Consistent with this idea, increased translation of APC3 and APC11 overexpression is also associated with CIN [164, 165]. As numerous reports suggest that overexpression of APC/C subunits is associated with poor prognosis, it is tempting to speculate increased CIN, which is also associated with poor prognosis, underlies this association. However, the impact of increased expression of APC/C subunits on mitotic timing and CIN remains to be determined.

As mentioned above, many of the mutations in APC/C subunits, particularly the truncation and frameshift mutants studied thus far are predicted to perturb the APC/C in ways that are not expected to be specific for APC/CCdc20. For example, truncation of APC8, APC3 or APC1 are expected to weaken interactions between APC10 and/or the co-activators within the remainder of the enzyme. Thus, it is tempting to speculate that, in addition to attenuating APC/CCdc20 activity in mitosis to limit CIN, these mutations serve to perturb tumor suppression by APC/CCdh1 as well. Additional research will be required to test this notion as well as gain an understanding of whether the majority of mutations in the APC/C subunits are deleterious as suggested by analyses so far.

V. Therapeutic Approaches for APC/C Inhibition in Cancer

APC/C is essential for normal development and is dysregulated in cancer

Mouse models involving Cdc20 and Cdh1 confirm that APC/C activity is essential for normal development and viability. Mouse embryos with homozygous deletions of Cdc20 are unable to undergo embryongenesis and arrest at the two-cell stage in metaphase [166]. Likewise, loss of Cdh1 leads to embryonic lethality with death occurring by embryonic day 10, due to defects in trophoblast endoreduplication [167] [168]. Lethality of the Cdh1-deficient embryos can be rescued with Cdh1 expression in the placenta, however the mice develop structural and numeric aberrations and proliferate inefficiently. Mouse models also confirm the oncogenic function of Cdc20 and the tumor suppressor function of Cdh1. Cdh1 heterozygous mice have an increased incidence of spontaneous tumor development, confirming the role of Cdh1 as a haploinsufficient tumor suppressor [96, 169]. Cdc20 hypomorphic mice develop considerable aneuploidy [170] and Cre-induced ablation of Cdc20 in mice carrying one null Cdc20 allele and one floxed Cdc20 knockout allele, ablated in vivo tumorigenesis in a skin-tumor mouse model [171]. Importantly, loss of Cdc20 elicited dramatic regression in skin tumors due to apoptotic death, whereas anti-mitotics exhibited more limited efficacy, confirming the rationale for inhibition of Cdc20 as a cancer therapy.

TAME and apcin directly inhibit APC/C and can be used in combination with microtubule-inhibiting drugs

Because APC/C is essential for mitotic exit during cellular division, its inhibition remains an attractive approach for cancer therapies. Indeed SAC-mediated inhibition of APC/CCdc20 is an important component of the cellular response to anti-mitotic agents such as taxanes and vinca alkaloids. Tosyl-L-Arginine Methyl Ester (TAME) is an APC/C inhibitor that was first identified as an inhibitor of cyclin proteolysis in a phenotypic Xenopous egg extract screen [172]. Because TAME is not cell permeable, the pro-drug form, pro-TAME, was developed in 2010 by the same lab and was found to inhibit Cdc20 and Cdh1 binding to APC/C, thereby preventing APC/C activation [173]. Later work revealed that TAME preferentially suppresses APC/CCdc20 rather than APC/CCdh1 [28]. Cells exposed to proTAME exhibit a prolonged mitotic arrest, which causes SAC reactivation and cohesion fatigue, resulting in cell death [174]. A second inhibitor, apcin, was described in 2014 [175]. Apcin binds to Cdc20 and competitively inhibits the ubiquitylation of D-box-containing substrates through occupation of the D-box-binding pocket within the Cdc20 WD40 domain [175]. Treatment of cells with apcin alone does not significantly affect cell survival because it is an inefficient inhibitor of APC/C. However, apcin is synthetically lethal in cohesion-defective cells and cancers [176].

Because apcin and TAME inhibit APC/C via different protein-protein interactions, a combination treatment synergizes to prolong mitotic duration in human cancer cell lines. For instance, the combination treatment apcin and proTAME has been shown to decrease tumor cell growth in multiple myeloma and osteosarcoma cancer cell lines, as well as in GBM cancer stem cells [177179]. APC/C inhibitors are also being tested in combination with microtubule-inhibiting drugs to further sustain mitotic arrest and enhance cell death. Crawford et al. show that proTAME in combination with the topoisomerase II inhibitors, etoposide and doxorubicin, and vincristine, a microtubule destabilizing agent, were effective at decreasing cell survival in multiple myeloma cells [180]. In addition, siRNA knockdown of Cdc20 in combination with docetaxel, a microtubule stabilizing agent, decreased castration-resistant prostate cancer cell growth [181]. In a recent study, Raab et al (2019) employed a combination treatment of paclitaxel and Plk1 inhibitor (BI6727) followed by proTAME in a ‘two-punch’ strategy to cause strong mitotic arrest followed by blocking mitotic exit to induce apoptosis in ovarian cancer primary cells and cell lines [182]. In addition, the Malumbres lab found that chemical inhibition of APC/C with proTAME caused increased sensitivity to Eg5 inhibitors and dramatic sensitivity to topoisomerase II inhibitors [183].

Other drugs are being explored as APC/C inhibitors

Numerous studies have implicated APC/C involvement in the response to newly emerging drugs. For example, Liu et al. show that treatment of colorectal cancer cell lines with Indoleamine 2, 3-dioxygenase (IDO), an enzyme that is the rate-limiting step in tryptophan metabolism, coincides with decreased Cdc20 expression and G2/M arrest [184]. Zhang et al. report that curcumin, a substance in turmeric, causes decreased pancreatic cell proliferation presumably due to downregulation of Cdc20 [185]. Rottlerin, polyphenol natural product isolated from the Asian tree Mallotus philippensis, was also shown to decrease glioma cell growth via many pathways [186]. Although the exact mechanism is not known, the authors show decreased Cdc20 expression is glioma cells treated with Rottlerein as well as treatment rescue with Cdc20 overexpression. Another study demonstrated that triterpenes from Poria cocos inhibit the migration of pancreatic cancer cells in correlation with decreased CDC20 [187]. These studies underscore the potential importance of Cdc20 inhibition as a therapeutic strategy in cancer.

VI. Conclusion

APC/CCdc20 inhibition is an attractive cancer therapy because of the essential role APC/C plays in cell cycle regulation and its contributions to other cancer-promoting pathways. However, current APC/C inhibitors, proTAME and apcin, are still in pre-clinical development and in vivo experiments have not been published. One obstacle for transitioning these inhibitors into experiments with mice is likely drug solubility. ProTAME has an IC50 in the μM range for HeLa cells [173] and multiple myeloma cells [179] (~3 μM to 20 μM). Apcin is synergistic with proTAME in the 200 μM range [175]. Another challenge associated with the use of these compounds is toxicity. Because APC/C is essential, a therapeutic strategy needs to be developed that restricts APC/C inhibition to tumor cells to avoid toxic effects from inhibiting cellular division in normal tissues. This is supported by Parmar et al. who used survivin and APC/C polyplexes to show therapeutic strategy proof-of-principle in breast cancer cell lines, but also saw the effect in normal cells [188]. One approach to circumventing APC/C inhibition in normal tissues is to envelop drug or siRNAs in nanoparticles for specific delivery only to tumor tissue. However, this technology is still developing [189, 190]. As an alternative to developing a small molecule inhibitor, the Wan lab used a protein-degradation strategy, Proteolysis Targeting Chimeras (PROTACs) to create CP5V, a potent Cdc20 degrader. CP5V works as a bridge to form a ternary complex between Cdc20 and the E3 ligase VHL to facilitate Cdc20 ubiquitination and subsequent proteasome-dependent degradation. CP5V elicits significant therapeutic efficacy at 2 μM in a subcutaneous xenograft breast cancer mouse mode with no apparent toxicity to normal tissue, showing therapeutic promise for this strategy [191].

Cdh1 is more difficult to approach as a clinical target. Although Cdh1 is generally accepted as a tumor suppressor, there have been studies that indicate Cdh1 could be overexpressed in some cancers at the RNA and/or protein level [192]. Similar to diminished activity, elevated Cdh1 activity is associated with DNA damage and rereplication, which could contribute to tumorigenesis. However, given that there are multiple mechanisms that destabilize Cdh1 protein, it is possible that elevated RNA is not indicative of elevated protein levels. Along these lines, whereas RNA expression profiles of the TCGA breast cancer cohort indicates increased expression of Cdh1, examination of Cdh1 protein levels as determined by mass spectrometry in these samples showed an overall reduction in Cdh1 (Figure 6) [16, 173, 193]. In addition, Cdh1 protein did not exhibit a negative correlation with APC/C substrates in these samples suggesting that the presence of Cdh1 protein many not equate to functional APC/CCdh1 in the tumors. Indeed, a multi-cancer analysis of tissue microarrays (TMA), which showed elevated Cdh1 in tumors, also found that Cdh1 levels were positively correlated with protein levels of APC/CCdh1 substrates [192]. Multiple mechanisms may contribute to the discrepancy between protein levels and apparent APC/CCdh1 activity in tumors such as phosphorylation by Cyclin-Cdk or Src leading to attenuated APC/C interaction with Cdh1, elevated expression of Emi1, or potential impacts of mutations in the APC/C subunits discussed above. It should also be noted that, in the context of the cell cycle, Cdh1 expression is not necessarily indicative of activity. Cdh1 expression peaks in G2 and M, but is held inactive until late mitosis. As the expression of many APC/C substrates also peak during G2 and M, it is possible that the positive correlation of Cdh1 protein and APC/C substrates observed in the TMA analysis is reflective of the cell cycle state of proliferating tumor cells.

Figure 6.

Figure 6.

Expression of Cdh1 (FZR1) in breast cancer. Data and analysis of the TCGA breast cancer data sets was obtained from the UALCAN server for (A) RNA; n=114, normal and n=1097, tumor and (B) protein; n=18, normal and n=125, tumor.

As covered earlier in this review, there is now substantial evidence to indicate that low Cdh1 levels and/or activity correlate with genome instability and replication errors as well as the regulation of numerous cancer-associated pathways. However, the discussion above highlights the deficit in our understanding of the mechanisms that lead to reduced APC/CCdh1 activity in cancers. Classic tumor suppressors accumulate point mutations rendering both alleles inactive, or in cases of haploinsufficiency, one null allele is sufficient to provide a pathogenic phenotype. Examination of Cdh1 sequences in human tumors and cancer cell lines reveals mutations in approximately 1–5% mutations in Cdh1, with the majority being variants of unknown significance (TCGA, cBioPortal). Altogether, it seems likely that the haploinsufficiency of Cdh1 activity is achieved in cancer by the potentially combintatorial activity of a variety of mechanisms, predominantly post-translational, that complicate the development of strategies to leverage Cdh1 activity as a cancer treatment. However, growing evidence suggests that enhanced APC/CCdh1 activity may contribute to the activity of current therapeutic strategies including inhibition of Akt, Src, and potentially Cdks. Cdc20 remains the paramount APC/C clinical target as it is the co-activator essential for exit from mitosis. In summary, mouse models and preclinical experiments underscore that Cdc20 inhibition is an effective cancer therapy, developing a drug which is soluble for in vivo studies that also exhibits tumor specificity remains a challenge.

Acknowledgements

This work was funded by an American Brain Tumor Association Basic Research Fellowship sponsored by an Anonymous Corporate Donor (MSS), NIH grants GM108743 (MKS) and GM112895 (MKS) and The Ohio State University Comprehensive Cancer Center/Department of Radiation Oncology start-up funds (MKS). The content of this work is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies. We apologize to our colleagues whose work we were unable to discuss within the confines of this review.

Abbreviations

APC/C

Anaphase Promoting Complex/Cyclosome

TPR

Tetratricopeptide Repeat

D-Box

Destruction Box, degron

KEN Box

Lysine-Glutamic Acid- containing degron

C-Mad2

Closed Mad2 Conformation

O-Mad2

Open Mad2 Conformation

SAC

Spindle Assembly Checkpoint

MCC

Mitotic Checkpoint Complex

CSCs

Cancer Stem-like Cells

TICs

Tumor Initiating Cells

SPOP

Speckle-type POZ protein, Cullin 3 substrate adaptor

SCF

Skp1-Cul1-F-box, Ubiquitin Ligase

PD-L1

Programmed-Death Ligand 1

CIN

Chromosomal Instability

TAME

Tosyl-L-Arginine Methyl Ester (APC/C inhibitor)

IDO

Indoleamine 2,3-dioxegnase

PROTACS

Proteolysis Targeting Chimeras

TCGA

The Cancer Genome Atlas

TMA

Tissue Microarray

Footnotes

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 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.

Conflict of Interest

The authors declare no competing interests.

References

  • 1.Visintin R, Prinz S, and Amon A, CDC20 and CDH1: a family of substrate-specific activators of APC-dependent proteolysis. Science, 1997. 278(5337): p. 460–3. [DOI] [PubMed] [Google Scholar]
  • 2.Fang G, Yu H, and Kirschner MW, Direct binding of CDC20 protein family members activates the anaphase-promoting complex in mitosis and G1. Mol Cell, 1998. 2(2): p. 163–71. [DOI] [PubMed] [Google Scholar]
  • 3.Yamaguchi M, et al. , Structure of an APC3–APC16 Complex: Insights into Assembly of the Anaphase-Promoting Complex/Cyclosome. Journal of Molecular Biology, 2015. 427(8): p. 1748–1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chang L, et al. , Atomic structure of the APC/C and its mechanism of protein ubiquitination. Nature, 2015. 522(7557): p. 450–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Uzunova K, et al. , APC15 mediates CDC20 autoubiquitylation by APC/CMCC and disassembly of the mitotic checkpoint complex. Nature Structural & Molecular Biology, 2012. 19(11): p. 1116–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Schreiber A, et al. , Structural basis for the subunit assembly of the anaphase-promoting complex. Nature, 2011. 470(7333): p. 227–232. [DOI] [PubMed] [Google Scholar]
  • 7.Ohi MD, et al. , Structural organization of the anaphase-promoting complex bound to the mitotic activator Slp1. Mol Cell, 2007. 28(5): p. 871–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang J, et al. , Insights into anaphase promoting complex TPR subdomain assembly from a CDC26-APC6 structure. Nat Struct Mol Biol, 2009. 16(9): p. 987–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stewart MD, et al. , E2 enzymes: more than just middle men. Cell Research, 2016. 26(4): p. 423–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rape M and Kirschner MW, Autonomous regulation of the anaphase-promoting complex couples mitosis to S-phase entry. Nature, 2004. 432(7017): p. 588–95. [DOI] [PubMed] [Google Scholar]
  • 11.Williamson A, et al. , Identification of a physiological E2 module for the human anaphase-promoting complex. Proc Natl Acad Sci U S A, 2009. 106(43): p. 18213–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Summers MK, et al. , The unique N terminus of the UbcH10 E2 enzyme controls the threshold for APC activation and enhances checkpoint regulation of the APC. Mol Cell, 2008. 31(4): p. 544–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Yu H, et al. , Identification of a novel ubiquitin-conjugating enzyme involved in mitotic cyclin degradation. Curr Biol, 1996. 6(4): p. 455–66. [DOI] [PubMed] [Google Scholar]
  • 14.Wu T, et al. , UBE2S drives elongation of K11-linked ubiquitin chains by the anaphase-promoting complex. Proc Natl Acad Sci U S A, 2010. 107(4): p. 1355–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Garnett MJ, et al. , UBE2S elongates ubiquitin chains on APC/C substrates to promote mitotic exit. Nat Cell Biol, 2009. 11(11): p. 1363–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wild T, et al. , The Spindle Assembly Checkpoint Is Not Essential for Viability of Human Cells with Genetically Lowered APC/C Activity. Cell Rep, 2016. 14(8): p. 1829–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Varshavsky A, The early history of the ubiquitin field. Protein Science, 2006. 15(3): p. 647–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Tang Z, et al. , APC2 Cullin protein and APC11 RING protein comprise the minimal ubiquitin ligase module of the anaphase-promoting complex. Mol Biol Cell, 2001. 12(12): p. 3839–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chang LF, et al. , Molecular architecture and mechanism of the anaphase-promoting complex. Nature,2014. 513(7518): p. 388–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Brown NG, et al. , Dual RING E3 Architectures Regulate Multiubiquitination and Ubiquitin Chain Elongation by APC/C. Cell, 2016. 165(6): p. 1440–1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Passmore LA, et al. , Doc1 mediates the activity of the anaphase-promoting complex by contributing to substrate recognition. Embo J, 2003. 22(4): p. 786–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.da Fonseca PC, et al. , Structures of APC/C(Cdh1) with substrates identify Cdh1 and Apc10 as the D-box co-receptor. Nature, 2011. 470(7333): p. 274–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kraft C, et al. , The WD40 propeller domain of Cdh1 functions as a destruction box receptor for APC/C substrates. Mol Cell, 2005. 18(5): p. 543–53. [DOI] [PubMed] [Google Scholar]
  • 24.Tian W, et al. , Structural analysis of human Cdc20 supports multisite degron recognition by APC/C. Proc Natl Acad Sci U S A, 2012. 109(45): p. 18419–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kraft C, et al. , Mitotic regulation of the human anaphase-promoting complex by phosphorylation. Embo J, 2003. 22(24): p. 6598–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kramer ER, et al. , Mitotic regulation of the APC activator proteins CDC20 and CDH1. Mol Biol Cell, 2000. 11(5): p. 1555–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Qiao R, et al. , Mechanism of APC/CCDC20 activation by mitotic phosphorylation. Proc Natl Acad Sci U S A, 2016. 113(19): p. E2570–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang S, et al. , Molecular mechanism of APC/C activation by mitotic phosphorylation. Nature, 2016. 533(7602): p. 260–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fujimitsu K, Grimaldi M, and Yamano H, Cyclin-dependent kinase 1–dependent activation of APC/C ubiquitin ligase. Science, 2016. 352(6289): p. 1121–1124. [DOI] [PubMed] [Google Scholar]
  • 30.Listovsky T, et al. , Mammalian Cdh1/Fzr mediates its own degradation. EMBO J, 2004. 23(7): p. 1619–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Glotzer M, Murray AW, and Kirschner MW, Cyclin is degraded by the ubiquitin pathway. Nature, 1991. 349(6305): p. 132–8. [DOI] [PubMed] [Google Scholar]
  • 32.Cohen-Fix O, et al. , Anaphase initiation in Saccharomyces cerevisiae is controlled by the APC-dependent degradation of the anaphase inhibitor Pds1p. Genes Dev, 1996. 10(24): p. 3081–93. [DOI] [PubMed] [Google Scholar]
  • 33.McGarry TJ and Kirschner MW, Geminin, an inhibitor of DNA replication, is degraded during mitosis. Cell, 1998. 93(6): p. 1043–53. [DOI] [PubMed] [Google Scholar]
  • 34.Wan Y, Liu X, and Kirschner MW, The anaphase-promoting complex mediates TGF-beta signaling by targeting SnoN for destruction. Mol Cell, 2001. 8(5): p. 1027–39. [DOI] [PubMed] [Google Scholar]
  • 35.Lasorella A, et al. , Degradation of Id2 by the anaphase-promoting complex couples cell cycle exit and axonal growth. Nature, 2006. 442(7101): p. 471–474. [DOI] [PubMed] [Google Scholar]
  • 36.Almeida A, Bolanos JP, and Moncada S, E3 ubiquitin ligase APC/C-Cdh1 accounts for the Warburg effect by linking glycolysis to cell proliferation. Proc Natl Acad Sci U S A, 2010. 107(2): p. 738–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lara-Gonzalez P, et al. , The G2-to-M Transition Is Ensured by a Dual Mechanism that Protects Cyclin B from Degradation by Cdc20–Activated APC/C. Developmental Cell, 2019. 51(3): p. 313–325.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhang S, Tischer T, and Barford D, Cyclin A2 degradation during the spindle assembly checkpoint requires multiple binding modes to the APC/C. Nature Communications, 2019. 10(1): p. 3863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Primorac I and Musacchio A, Panta rhei: The APC/C at steady state. The Journal of Cell Biology, 2013. 201(2): p. 177–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.He J, et al. , Insights into degron recognition by APC/C coactivators from the structure of an Acm1-Cdh1 complex. Mol Cell, 2013. 50(5): p. 649–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Davey NE and Morgan DO, Building a Regulatory Network with Short Linear Sequence Motifs: Lessons from the Degrons of the Anaphase-Promoting Complex. Mol Cell, 2016. 64(1): p. 12–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fujimitsu K, Grimaldi M, and Yamano H, Cyclin-dependent kinase 1-dependent activation of APC/C ubiquitin ligase. Science, 2016. 352(6289): p. 1121–4. [DOI] [PubMed] [Google Scholar]
  • 43.Qiao R, et al. , Mechanism of APC/CCDC20 activation by mitotic phosphorylation. Proceedings of the National Academy of Sciences, 2016. 113(19): p. E2570–E2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Reimann JD, et al. , Emi1 is a mitotic regulator that interacts with Cdc20 and inhibits the anaphase promoting complex. Cell, 2001. 105(5): p. 645–55. [DOI] [PubMed] [Google Scholar]
  • 45.Reimann JD, et al. , Emi1 regulates the anaphase-promoting complex by a different mechanism than Mad2 proteins. Genes Dev, 2001. 15(24): p. 3278–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Margottin-Goguet F, et al. , Prophase destruction of Emi1 by the SCF(betaTrCP/Slimb) ubiquitin ligase activates the anaphase promoting complex to allow progression beyond prometaphase. Dev Cell, 2003. 4(6): p. 813–26. [DOI] [PubMed] [Google Scholar]
  • 47.Guardavaccaro D, et al. , Control of meiotic and mitotic progression by the F box protein beta-Trcp1 in vivo. Dev Cell, 2003. 4(6): p. 799–812. [DOI] [PubMed] [Google Scholar]
  • 48.Moshe Y, et al. , Role of Polo-like kinase in the degradation of early mitotic inhibitor 1, a regulator of the anaphase promoting complex/cyclosome. Proc Natl Acad Sci U S A, 2004. 101(21): p. 7937–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ban KH, et al. , The END network couples spindle pole assembly to inhibition of the anaphase-promoting complex/cyclosome in early mitosis. Dev Cell, 2007. 13(1): p. 29–42. [DOI] [PubMed] [Google Scholar]
  • 50.Luo X, et al. , Structure of the Mad2 spindle assembly checkpoint protein and its interaction with Cdc20. Nat Struct Biol, 2000. 7(3): p. 224–9. [DOI] [PubMed] [Google Scholar]
  • 51.Luo X, et al. , The Mad2 spindle checkpoint protein undergoes similar major conformational changes upon binding to either Mad1 or Cdc20. Mol Cell, 2002. 9(1): p. 59–71. [DOI] [PubMed] [Google Scholar]
  • 52.Luo X, et al. , The Mad2 spindle checkpoint protein has two distinct natively folded states. Nat Struct Mol Biol, 2004. 11(4): p. 338–45. [DOI] [PubMed] [Google Scholar]
  • 53.Xia G, et al. , Conformation-specific binding of p31(comet) antagonizes the function of Mad2 in the spindle checkpoint. Embo J, 2004. 23(15): p. 3133–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Simonetta M, et al. , The influence of catalysis on mad2 activation dynamics. PLoS Biol, 2009. 7(1): p. e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.DeAntoni A, Sala V, and Musacchio A, Explaining the oligomerization properties of the spindle assembly checkpoint protein Mad2. Philos Trans R Soc Lond B Biol Sci, 2005. 360(1455): p. 637–47, discussion 447–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nezi L, et al. , Accumulation of Mad2-Cdc20 complex during spindle checkpoint activation requires binding of open and closed conformers of Mad2 in Saccharomyces cerevisiae. J Cell Biol, 2006. 174(1): p. 39–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sironi L, et al. , Mad2 binding to Mad1 and Cdc20, rather than oligomerization, is required for the spindle checkpoint. EMBO J, 2001. 20(22): p. 6371–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Musacchio A and Salmon ED, The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol, 2007. 8(5): p. 379–93. [DOI] [PubMed] [Google Scholar]
  • 59.Diaz-Martinez LA and Yu H, Running on a treadmill: dynamic inhibition of APC/C by the spindle checkpoint. Cell Div, 2007. 2: p. 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sudakin V, Chan GK, and Yen TJ, Checkpoint inhibition of the APC/C in HeLa cells is mediated by a complex of BUBR1, BUB3, CDC20, and MAD2. J Cell Biol, 2001. 154(5): p. 925–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Alfieri C, et al. , Molecular basis of APC/C regulation by the spindle assembly checkpoint. Nature, 2016. 536(7617): p. 431–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jia L, Li B, and Yu H, The Bub1-Plk1 kinase complex promotes spindle checkpoint signalling through Cdc20 phosphorylation. Nat Commun, 2016. 7: p. 10818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sangodkar J, et al. , All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase. FEBS J, 2016. 283(6): p. 1004–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lee SJ, et al. , The PP2A(B56) phosphatase promotes the association of Cdc20 with APC/C in mitosis. Journal of cell science, 2017. 130(10): p. 1760–1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gujar AD, Yano H, and Kim AH, The CDC20-APC/SOX2 signaling axis: An achilles’ heel for glioblastoma. Molecular & cellular oncology, 2015. 3(3): p. e1075644–e1075644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wu F, et al. , Prostate cancer-associated mutation in SPOP impairs its ability to target Cdc20 for poly-ubiquitination and degradation. Cancer Lett, 2017. 385: p. 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Garg M and Maurya N, WNT/β-catenin signaling in urothelial carcinoma of bladder. World journal of nephrology, 2019. 8(5): p. 83–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mao DD, et al. , A CDC20-APC/SOX2 Signaling Axis Regulates Human Glioblastoma Stem-like Cells. Cell reports, 2015. 11(11): p. 1809–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Maes A, et al. , The anaphase-promoting complex/cyclosome: a new promising target in diffuse large B-cell lymphoma and mantle cell lymphoma. British Journal of Cancer, 2019. 120(12): p. 1137–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Paul D, et al. , Cdc20 directs proteasome-mediated degradation of the tumor suppressor SMAR1 in higher grades of cancer through the anaphase promoting complex. Cell death & disease, 2017. 8(6): p. e2882–e2882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zhang Q, et al. , Cell division cycle 20 (CDC20) drives prostate cancer progression via stabilization of β-catenin in cancer stem-like cells. EBioMedicine, 2019. 42: p. 397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chu Z, et al. , CDC20 contributes to the development of human cutaneous squamous cell carcinoma through the Wnt/β‑ catenin signaling pathway. International journal of oncology, 2019. 54(5): p. 1534–1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Anastas JN and Moon RT, WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer, 2013. 13(1): p. 11–26. [DOI] [PubMed] [Google Scholar]
  • 74.Avilion AA, et al. , Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev, 2003. 17(1): p. 126–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.el-Deiry WS, et al. , WAF1, a potential mediator of p53 tumor suppression. Cell, 1993. 75(4): p. 817–25. [DOI] [PubMed] [Google Scholar]
  • 76.Harper JW, et al. , The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell, 1993. 75(4): p. 805–16. [DOI] [PubMed] [Google Scholar]
  • 77.Gascoigne KE and Taylor SS, Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell, 2008. 14(2): p. 111–22. [DOI] [PubMed] [Google Scholar]
  • 78.Brito DA and Rieder CL, Mitotic checkpoint slippage in humans occurs via cyclin B destruction in the presence of an active checkpoint. Curr Biol, 2006. 16(12): p. 1194–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Brito DA and Rieder CL, The ability to survive mitosis in the presence of microtubule poisons differs significantly between human nontransformed (RPE-1) and cancer (U2OS, HeLa) cells. Cell Motil Cytoskeleton, 2009. 66(8): p. 437–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wan L, et al. , APC(Cdc20) suppresses apoptosis through targeting Bim for ubiquitination and destruction. Developmental cell, 2014. 29(4): p. 377–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Allan LA, et al. , Atypical APC/C-dependent degradation of Mcl-1 provides an apoptotic timer during mitotic arrest. EMBO J, 2018. 37(17). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Harley ME, et al. , Phosphorylation of Mcl-1 by CDK1-cyclin B1 initiates its Cdc20-dependent destruction during mitotic arrest. EMBO J, 2010. 29(14): p. 2407–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sloss O, et al. , Mcl-1 dynamics influence mitotic slippage and death in mitosis. Oncotarget, 2016. 7(5): p. 5176–5192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chaudhary N, et al. , SMAR1 coordinates HDAC6-induced deacetylation of Ku70 and dictates cell fate upon irradiation. Cell Death Dis, 2014. 5: p. e1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Singh K, et al. , p53 target gene SMAR1 is dysregulated in breast cancer: its role in cancer cell migration and invasion. PLoS One, 2007. 2(7): p. e660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Sinha S, et al. , Coordinated regulation of p53 apoptotic targets BAX and PUMA by SMAR1 through an identical MAR element. EMBO J, 2010. 29(4): p. 830–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Taye N, et al. , SMAR1 inhibits Wnt/beta-catenin signaling and prevents colorectal cancer progression. Oncotarget, 2018. 9(30): p. 21322–21336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hofmann M, et al. , CD44 splice variants confer metastatic behavior in rats: homologous sequences are expressed in human tumor cell lines. Cancer Res, 1991. 51(19): p. 5292–7. [PubMed] [Google Scholar]
  • 89.Yan Y, Zuo X, and Wei D, Concise Review: Emerging Role of CD44 in Cancer Stem Cells: A Promising Biomarker and Therapeutic Target. Stem Cells Transl Med, 2015. 4(9): p. 1033–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wu F, et al. , Prostate cancer-associated mutation in SPOP impairs its ability to target Cdc20 for poly-ubiquitination and degradation. Cancer letters, 2017. 385: p. 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Huang JN, et al. , Activity of the APC(Cdh1) form of the anaphase-promoting complex persists until S phase and prevents the premature expression of Cdc20p. J Cell Biol, 2001. 154(1): p. 85–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lindon C and Pines J, Ordered proteolysis in anaphase inactivates Plk1 to contribute to proper mitotic exit in human cells. 2004. 164(2): p. 233–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sigl R, et al. , Loss of the mammalian APC/C activator FZR1 shortens G1 and lengthens S phase but has little effect on exit from mitosis. J Cell Sci, 2009. 122(Pt 22): p. 4208–17. [DOI] [PubMed] [Google Scholar]
  • 94.Littlepage LE and 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(17): p. 2274–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Stewart S and Fang G, Destruction Box–Dependent Degradation of Aurora B Is Mediated by the Anaphase-Promoting Complex/Cyclosome and Cdh1. 2005. 65(19): p. 8730–8735. [DOI] [PubMed] [Google Scholar]
  • 96.García-Higuera I, et al. , Genomic stability and tumour suppression by the APC/C cofactor Cdh1. Nature Cell Biology, 2008. 10 (7): p. 802–811. [DOI] [PubMed] [Google Scholar]
  • 97.Donzelli M, et al. , Dual mode of degradation of Cdc25 A phosphatase. EMBO J, 2002. 21(18): p. 4875–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Bashir T, et al. , Control of the SCF(Skp2-Cks1) ubiquitin ligase by the APC/C(Cdh1) ubiquitin ligase. Nature, 2004. 428(6979): p. 190–3. [DOI] [PubMed] [Google Scholar]
  • 99.Huang X, et al. , Deubiquitinase USP37 is activated by CDK2 to antagonize APC(CDH1) and promote S phase entry. Mol Cell, 2011. 42(4): p. 511–23. [DOI] [PubMed] [Google Scholar]
  • 100.Frye JJ, et al. , Electron microscopy structure of human APC/C(CDH1)-EMI1 reveals multimodal mechanism of E3 ligase shutdown. Nat Struct Mol Biol, 2013. 20(7): p. 827–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Miller JJ, et al. , Emi1 stably binds and inhibits the anaphase-promoting complex/cyclosome as a pseudosubstrate inhibitor. Genes Dev, 2006. 20(17): p. 2410–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wang W and Kirschner MW, Emi1 preferentially inhibits ubiquitin chain elongation by the anaphase-promoting complex. Nat Cell Biol, 2013. 15(7): p. 797–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Choudhury R, et al. , APC/C and SCF(cyclin F) Constitute a Reciprocal Feedback Circuit Controlling S-Phase Entry. Cell Rep, 2016. 16(12): p. 3359–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Fukushima H, et al. , SCF-mediated Cdh1 degradation defines a negative feedback system that coordinates cell-cycle progression. Cell Rep, 2013. 4(4): p. 803–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lukas C, et al. , Accumulation of cyclin B1 requires E2F and cyclin-A-dependent rearrangement of the anaphase-promoting complex. Nature, 1999. 401: p. 815. [DOI] [PubMed] [Google Scholar]
  • 106.Keck JM, et al. , Cyclin E overexpression impairs progression through mitosis by inhibiting APC(Cdh1). J Cell Biol, 2007. 178(3): p. 371–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lau AW, et al. , Regulation of APC(Cdh1) E3 ligase activity by the Fbw7/cyclin E signaling axis contributes to the tumor suppressor function of Fbw7. Cell Res, 2013. 23(7): p. 947–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zachariae W, et al. , Control of cyclin ubiquitination by CDK-regulated binding of Hct1 to the anaphase promoting complex. Science, 1998. 282(5394): p. 1721–4. [DOI] [PubMed] [Google Scholar]
  • 109.Zhou Z, et al. , Insights into APC/C: from cellular function to diseases and therapeutics. 2016. 11(1): p. 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Yamano H, APC/C: current understanding and future perspectives. F1000Res, 2019. 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Watson ER, et al. , Posing the APC/C E3 Ubiquitin Ligase to Orchestrate Cell Division. Trends Cell Biol, 2019. 29(2): p. 117–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fuchsberger T, Lloret A, and Viña J, New Functions of APC/C Ubiquitin Ligase in the Nervous System and Its Role in Alzheimer’s Disease. Int J Mol Sci, 2017. 18(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cappell SD, et al. , EMI1 switches from being a substrate to an inhibitor of APC/C(CDH1) to start the cell cycle. Nature, 2018. 558(7709): p. 313–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Yao G, et al. , A bistable Rb–E2F switch underlies the restriction point. Nature Cell Biology, 2008. 10(4): p. 476–482. [DOI] [PubMed] [Google Scholar]
  • 115.Narasimha AM, et al. , Cyclin D activates the Rb tumor suppressor by mono-phosphorylation. Elife, 2014. 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Cappell SD, et al. , Irreversible APC(Cdh1) Inactivation Underlies the Point of No Return for Cell-Cycle Entry. Cell, 2016. 166(1): p. 167–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jeffery JM, et al. , FBXO31 protects against genomic instability by capping FOXM1 levels at the G2/M transition. Oncogene, 2017. 36(7): p. 1012–1022. [DOI] [PubMed] [Google Scholar]
  • 118.Jia L and Sun Y, F-box proteins FBXO31 and FBX4 in regulation of cyclin D1 degradation upon DNA damage. Pigment Cell Melanoma Res, 2009. 22(5): p. 518–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Kumar R, et al. , FBXO31 is the chromosome 16q24.3 senescence gene, a candidate breast tumor suppressor, and a component of an SCF complex. Cancer Res, 2005. 65(24): p. 11304–13. [DOI] [PubMed] [Google Scholar]
  • 120.Malonia SK, et al. , F-box protein FBXO31 directs degradation of MDM2 to facilitate p53-mediated growth arrest following genotoxic stress. Proc Natl Acad Sci U S A, 2015. 112(28): p. 8632–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Santra MK, Wajapeyee N, and Green MR, F-box protein FBXO31 mediates cyclin D1 degradation to induce G1 arrest after DNA damage. Nature, 2009. 459(7247): p. 722–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Choppara S, et al. , Degradation of FBXO31 by APC/C is regulated by AKT- and ATM-mediated phosphorylation. Proceedings of the National Academy of Sciences, 2018. 115(5): p. 998–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wei W, et al. , Degradation of the SCF component Skp2 in cell-cycle phase G1 by the anaphase-promoting complex. Nature, 2004. 428(6979): p. 194–8. [DOI] [PubMed] [Google Scholar]
  • 124.Carrano AC, et al. , SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nature Cell Biology, 1999. 1(4): p. 193–199. [DOI] [PubMed] [Google Scholar]
  • 125.Choudhury R, et al. , The E3 Ubiquitin Ligase SCF(Cyclin F) Transmits AKT Signaling to the Cell-Cycle Machinery. Cell Rep, 2017. 20(13): p. 3212–3222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Wan L, et al. , The APC/C E3 Ligase Complex Activator FZR1 Restricts BRAF Oncogenic Function. Cancer Discov, 2017. 7(4): p. 424–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Han T, et al. , Interplay between c-Src and the APC/C co-activator Cdh1 regulates mammary tumorigenesis. Nat Commun, 2019. 10(1): p. 3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Engelbert D, et al. , The ubiquitin ligase APC(Cdh1) is required to maintain genome integrity in primary human cells. Oncogene, 2008. 27(7): p. 907–17. [DOI] [PubMed] [Google Scholar]
  • 129.Bassermann F, et al. , The Cdc14B-Cdh1-Plk1 axis controls the G2 DNA-damage-response checkpoint. Cell, 2008. 134(2): p. 256–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lafranchi L, et al. , APC/C(Cdh1) controls CtIP stability during the cell cycle and in response to DNA damage. EMBO J, 2014. 33(23): p. 2860–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sudo T, et al. , Activation of Cdh1-dependent APC is required for G1 cell cycle arrest and DNA damage-induced G2 checkpoint in vertebrate cells. EMBO J, 2001. 20(22): p. 6499–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Zhang T, et al. , DNA damage checkpoint maintains CDH1 in an active state to inhibit anaphase progression. Dev Cell, 2009. 17(4): p. 541–51. [DOI] [PubMed] [Google Scholar]
  • 133.Ha K, et al. , The anaphase promoting complex impacts repair choice by protecting ubiquitin signalling at DNA damage sites. Nature Communications, 2017. 8(1): p. 15751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lin H, et al. , Cdc14A and Cdc14B Redundantly Regulate DNA Double-Strand Break Repair. Mol Cell Biol, 2015. 35(21): p. 3657–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Greil C, et al. , The role of APC/C(Cdh1) in replication stress and origin of genomic instability. Oncogene, 2016. 35(23): p. 3062–70. [DOI] [PubMed] [Google Scholar]
  • 136.Arquint C, et al. , Cell-cycle-regulated expression of STIL controls centriole number in human cells. 2012. 125(5): p. 1342–1352. [DOI] [PubMed] [Google Scholar]
  • 137.Aplan PD, Lombardi DP, and Kirsch IR, Structural characterization of SIL, a gene requently disrupted in T-cell acute lymphoblastic leukemia. Mol Cell Biol, 1991. 11(11): p. 5462–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Arquint C and Erich A Nigg, STIL Microcephaly Mutations Interfere with APC/C-Mediated Degradation and Cause Centriole Amplification. Current Biology, 2014. 24(4): p. 351–360. [DOI] [PubMed] [Google Scholar]
  • 139.Medley JC, et al. , APC/C(FZR-1) Controls SAS-5 Levels To Regulate Centrosome Duplication in Caenorhabditis elegans. G3 (Bethesda), 2017. 7(12): p. 3937–3946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Moreno-Moreno O, Torras-Llort M, and Azorin F, The E3-ligases SCFPpa and APC/CCdh1 co-operate to regulate CENP-ACID expression across the cell cycle. Nucleic Acids Res, 2019. 47(7): p. 3395–3406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Amato A, et al. , CENPA overexpression promotes genome instability in pRb-depleted human cells. Mol Cancer, 2009. 8: p. 119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sorensen CS, et al. , Nonperiodic activity of the human anaphase-promoting complex-Cdh1 ubiquitin ligase results in continuous DNA synthesis uncoupled from mitosis. Mol Cell Biol, 2000. 20(20): p. 7613–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Garzón J, et al. , Shortage of dNTPs underlies altered replication dynamics and DNA breakage in the absence of the APC/C cofactor Cdh1. Oncogene, 2017. 36(42): p. 5808–5818. [DOI] [PubMed] [Google Scholar]
  • 144.Yuan X, et al. , Uncovering the role of APC-Cdh1 in generating the dynamics of S-phase onset. Mol Biol Cell, 2014. 25(4): p. 441–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.D’Angiolella V, et al. , Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell, 2012. 149(5): p. 1023–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Zhang J, et al. , Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature, 2018. 553(7686): p. 91–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Dankert JF, et al. , Cyclin F-Mediated Degradation of SLBP Limits H2A.X Accumulation and Apoptosis upon Genotoxic Stress in G2. Mol Cell, 2016. 64(3): p. 507–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Liu W, et al. , Proteolysis of CDH1 enhances susceptibility to UV radiation-induced apoptosis. Carcinogenesis, 2008. 29(2): p. 263–272. [DOI] [PubMed] [Google Scholar]
  • 149.Cao J, et al. , The E3 ligase APC/C(Cdh1) promotes ubiquitylation-mediated proteolysis of PAX3 to suppress melanocyte proliferation and melanoma growth. Sci Signal, 2015. 8(392): p. ra87. [DOI] [PubMed] [Google Scholar]
  • 150.Carnero A and Paramio JM, The PTEN/PI3K/AKT Pathway in vivo, Cancer Mouse Models. 2014. 4(252). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Liu J, et al. , Cdh1 inhibits WWP2-mediated ubiquitination of PTEN to suppress tumorigenesis in an APC-independent manner. Cell Discov, 2016. 2: p. 15044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Maddika S, et al. , WWP2 is an E3 ubiquitin ligase for PTEN. Nat Cell Biol, 2011. 13(6): p. 728–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Song MS, et al. , Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner. Cell, 2011. 144(2): p. 187–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Sen B and Johnson FM, Regulation of SRC family kinases in human cancers. J Signal Transduct, 2011. 2011: p. 865819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Wang Q, et al. , Alterations of anaphase-promoting complex genes in human colon cancer cells. Oncogene, 2003. 22(10): p. 1486–90. [DOI] [PubMed] [Google Scholar]
  • 156.Cancer Genome Atlas N, Comprehensive molecular characterization of human colon and rectal cancer. Nature, 2012. 487(7407): p. 330–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Lawrence MS, et al. , Discovery and saturation analysis of cancer genes across 21 tumour types. Nature, 2014. 505(7484): p. 495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Litchfield K, et al. , Whole-exome sequencing reveals the mutational spectrum of testicular germ cell tumours. Nat Commun, 2015. 6: p. 5973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Pawar SA, et al. , C/EBP{delta} targets cyclin D1 for proteasome-mediated degradation via induction of CDC27/APC3 expression. Proc Natl Acad Sci U S A, 2010. 107(20): p. 9210–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Sansregret L, et al. , APC/C Dysfunction Limits Excessive Cancer Chromosomal Instability. Cancer Discov, 2017. 7(2): p. 218–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Thu KL, et al. , Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer. Proc Natl Acad Sci U S A, 2018. 115(7): p. E1570–E1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Li M, et al. , Loss of spindle assembly checkpoint-mediated inhibition of Cdc20 promotes tumorigenesis in mice. J Cell Biol, 2009. 185(6): p. 983–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.van Ree JH, et al. , Overexpression of the E2 ubiquitin-conjugating enzyme UbcH10 causes chromosome missegregation and tumor formation. J Cell Biol, 2010. 188(1): p. 83–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Link LA, et al. , PCBP1/HNRNP E1 Protects Chromosomal Integrity by Translational Regulation of CDC27. Mol Cancer Res, 2016. 14(7): p. 634–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Drouet Y, et al. , Integrated analysis highlights APC11 protein expression as a likely new independent predictive marker for colorectal cancer. Sci Rep, 2018. 8(1): p. 7386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Li M, York JP, and Zhang P, Loss of Cdc20 Causes a Securin-Dependent Metaphase Arrest in Two-Cell Mouse Embryos. Molecular and Cellular Biology, 2007. 27(9): p. 3481–3488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Li M, et al. , The adaptor protein of the anaphase promoting complex Cdh1 is essential in maintaining replicative lifespan and in learning and memory. Nature Cell Biology, 2008. 10(9): p. 1083–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Garcia-Higuera I, et al. , Genomic stability and tumour suppression by the APC/C cofactor Cdh1. Nat Cell Biol, 2008. 10(7): p. 802–11. [DOI] [PubMed] [Google Scholar]
  • 169.Wasch R, Robbins JA, and Cross FR, The emerging role of APC/CCdh1 in controlling differentiation, genomic stability and tumor suppression. Oncogene, 2010. 29(1): p. 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Malureanu L, et al. , Cdc20 hypomorphic mice fail to counteract de novo synthesis of cyclin B1 in mitosis. The Journal of cell biology, 2010. 191(2): p. 313–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Manchado E, et al. , Targeting mitotic exit leads to tumor regression in vivo: Modulation by Cdk1, Mastl, and the PP2A/B55alpha,delta phosphatase. Cancer Cell, 2010. 18(6): p. 641–54. [DOI] [PubMed] [Google Scholar]
  • 172.Verma R, et al. , Ubistatins Inhibit Proteasome-Dependent Degradation by Binding the Ubiquitin Chain. Science, 2004. 306(5693): p. 117–120. [DOI] [PubMed] [Google Scholar]
  • 173.Zeng X, et al. , Pharmacologic inhibition of the anaphase-promoting complex induces a spindle checkpoint-dependent mitotic arrest in the absence of spindle damage. Cancer Cell, 2010. 18(4): p. 382–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Lara-Gonzalez P and Taylor SS, Cohesion fatigue explains why pharmacological inhibition of the APC/C induces a spindle checkpoint-dependent mitotic arrest. PloS one, 2012. 7(11): p. e49041–e49041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Sackton KL, et al. , Synergistic blockade of mitotic exit by two chemical inhibitors of the APC/C. Nature, 2014. 514(7524): p. 646–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.de Lange J, et al. , Defective sister chromatid cohesion is synthetically lethal with impaired APC/C function. Nature Communications, 2015. 6(1): p. 8399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.De K, et al. , Hyperphosphorylation of CDH1 in Glioblastoma Cancer Stem Cells Attenuates APC/CCDH1 Activity and Pharmacologic Inhibition of APC/CCDH1/CDC20 Compromises Viability. Molecular Cancer Research, 2019. 17(7): p. 1519–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Gao Y, et al. , Cdc20 inhibitor apcin inhibits the growth and invasion of osteosarcoma cells. Oncol Rep, 2018. 40(2): p. 841–848. [DOI] [PubMed] [Google Scholar]
  • 179.Lub S, et al. , Inhibiting the anaphase promoting complex/cyclosome induces a metaphase arrest and cell death in multiple myeloma cells. Oncotarget, 2016. 7(4): p. 4062–4076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Crawford LJ, et al. , Identification of the APC/C co-factor FZR1 as a novel therapeutic target for multiple myeloma. Oncotarget, 2016. 7(43): p. 70481–70493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Li K, et al. , Silencing of CDC20 suppresses metastatic castration-resistant prostate cancer growth and enhances chemosensitivity to docetaxel. Int J Oncol, 2016. 49(4): p. 1679–85. [DOI] [PubMed] [Google Scholar]
  • 182.Raab M, et al. , Blocking Mitotic Exit of Ovarian Cancer Cells by Pharmaceutical Inhibition of the Anaphase-Promoting Complex Reduces Chromosomal Instability. Neoplasia, 2019. 21(4): p. 363–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Eguren M, et al. , A Synthetic Lethal Interaction between APC/C and Topoisomerase Poisons Uncovered by Proteomic Screens. Cell Reports, 2014. 6(4): p. 670–683. [DOI] [PubMed] [Google Scholar]
  • 184.Liu X, et al. , 1-L-MT, an IDO inhibitor, prevented colitis-associated cancer by inducing CDC20 inhibition-mediated mitotic death of colon cancer cells. International Journal of Cancer, 2018. 143(6): p. 1516–1529. [DOI] [PubMed] [Google Scholar]
  • 185.Zhang Y, et al. , Inhibition of Cell Survival by Curcumin Is Associated with Downregulation of Cell Division Cycle 20 (Cdc20) in Pancreatic Cancer Cells. Nutrients, 2017. 9(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Wang L, et al. , Rottlerin inhibits cell growth and invasion via down-regulation of Cdc20 in glioma cells. Oncotarget, 2016. 7(43). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Cheng S, Castillo V, and Sliva D, CDC20 associated with cancer metastasis and novel mushroomderived CDC20 inhibitors with antimetastatic activity. Int J Oncol, 2019. 54(6): p. 2250–2256. [DOI] [PubMed] [Google Scholar]
  • 188.Parmar MB, et al. , Additive Polyplexes to Undertake siRNA Therapy against CDC20 and Survivin in Breast Cancer Cells. Biomacromolecules, 2018. 19(11): p. 4193–4206. [DOI] [PubMed] [Google Scholar]
  • 189.Mukherjee A, et al. , Lipid-polymer hybrid nanoparticles as a next-generation drug delivery platform: state of the art, emerging technologies, and perspectives. Int J Nanomedicine, 2019. 14: p. 1937–1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Parmar MB, et al. , Targeting Cell Cycle Proteins in Breast Cancer Cells with siRNA by Using Lipid-Substituted Polyethylenimines. Frontiers in bioengineering and biotechnology, 2015. 3: p. 14–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Chi JJ, et al. , A novel strategy to block mitotic progression for targeted therapy. EBioMedicine, 2019. 49: p. 40–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Lehman NL, et al. , Oncogenic regulators and substrates of the anaphase promoting complex/cyclosome are frequently overexpressed in malignant tumors. Am J Pathol, 2007. 170(5): p. 1793–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wild T, et al. , Deletion of APC7 or APC16 Allows Proliferation of Human Cells without the Spindle Assembly Checkpoint. Cell Rep, 2018. 25(9): p. 2317–2328 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES