Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2019 Oct 1;244(16):1419–1429. doi: 10.1177/1535370219881360

Highlight article: Role of E2Fs and mitotic regulators controlled by E2Fs in the epithelial to mesenchymal transition

Shirley Jusino 1, Harold I Saavedra 1,
PMCID: PMC6900705  PMID: 31575294

Short abstract

The epithelial-to-mesenchymal transition (EMT) is a complex cellular process in which epithelial cells acquire mesenchymal properties. EMT occurs in three biological settings: development, wound healing and fibrosis, and tumor progression. Despite occurring in three independent biological settings, EMT signaling shares some molecular mechanisms that allow epithelial cells to de-differentiate and acquire mesenchymal characteristics that confer cells invasive and migratory capacity to distant sites. Here we summarize the molecular mechanism that delineates EMT and we will focus on the role of E2 promoter binding factors (E2Fs) in EMT during tumor progression. Since the E2Fs are presently undruggable due to their control in numerous pivotal cellular functions and due to the lack of selectivity against individual E2Fs, we will also discuss the role of three mitotic regulators and/or mitotic kinases controlled by the E2Fs (NEK2, Mps1/TTK, and SGO1) in EMT that can be useful as drug targets.

Impact statement

The study of the epithelial to mesenchymal transition (EMT) is an active area of research since it is one of the early intermediates to invasion and metastasis—a state of the cancer cells that ultimately kills many cancer patients. We will present in this review that besides their canonical roles as regulators of proliferation, unregulated expression of the E2F transcription factors may contribute to cancer initiation and progression to metastasis by signaling centrosome amplification, chromosome instability, and EMT. Since our discovery that the E2F activators control centrosome amplification and mitosis in cancer cells, we have identified centrosome and mitotic regulators that may represent actionable targets against EMT and metastasis in cancer cells. This is impactful to all of the cancer patients in which the Cdk/Rb/E2F pathway is deregulated, which has been estimated to be most cancer patients with solid tumors.

Keywords: Centrosome amplification, E2Fs, cell cycle, cancer, the epithelial to mesenchymal transition, metastasis

Introduction

The concept of epithelial to mesenchymal transformation, now referred to as the epithelial-to-mesenchymal transition (EMT), was first coined by Elizabeth Hay1 when she observed that epithelial cells undergo complex cellular processes to acquire mesenchymal characteristics in order to invade and migrate during primitive streak development in chick embryos. EMT initiates by the loss of epithelial junctions and apical-basal polarity.24 Loss of epithelial junctions and cell polarity results in cytoskeleton reorganization and morphological changes.

These molecular changes are accompanied by changes in protein expression, such as the downregulation of proteins that compose the junction complexes and epithelial markers, as well as the upregulation of mesenchymal markers.5,6 The loss of E-cadherin is considered a hallmark of EMT.7 Typically, the loss of E-cadherin results in expression shift with another cadherin isoform, a process known as “cadherin switching.” As discussed in Wheelock et al.,7 this cadherin switch is not restricted to E-cadherin to N-cadherin and is context-dependent. Other molecular changes include changes in integrin expression, which correlates with increased expression of matrix metalloproteinases (MMPs) that contribute to the loss of epithelial junctions, enhance ECM degradation, and enable cell invasion.8,9 EMT can also be regulated at the RNA level either by alternative splicing that generates alternative isoforms of pivotal EMT-related proteins or non-coding miRNA that bind to mRNAs coding for epithelial markers and inhibit their translation or promote their degradation. Most molecular changes are mainly orchestrated by EMT transcription factor families such as SNAIL,10 bHLH (which includes TWIST1),6,11 ZEB,6,12 and many others. Several signaling pathways are involved in EMT regulation and can be further reviewed in Lamouille et al.4 Ultimately, all the processes described above result in the acquisition of mesenchymal features that allow the cells to invade and migrate through the extracellular matrix (ECM).

EMT can occur in three distinct biological settings with clearly delineated functional consequences: Type 1 (associated with embryogenesis and development), Type 2 (associated with wound healing and fibrosis), and Type 3 (associated with cancer progression).13 The latter is our main interest since EMT is a precursor of cancer metastasis, in which prevention and treatment remain a key clinical challenge. However, EMT does not only promote metastasis, but also promote tumor initiation, tumor stemness, and resistance to therapy.11,14,15

EMT has been described as a binary process with two distinct populations: epithelial and mesenchymal. However, recent studies have demonstrated the presence of intermediate states referred to as partial, incomplete, or hybrid EMT states. Pastushenko and Blanpain16 classified the states as epithelial, early hybrid EMT, late hybrid EMT, and full EMT or mesenchymal. Each transition state is grouped according to their distinct characteristics including cell shape, cell adhesion, expression of surface markers, transcription factors, and EMT-associated genes. They also classified these transition states accordingly to proliferative, invasiveness, plasticity, stemness, and metastatic properties.

Since EMT is a key precursor of cancer metastasis, we will describe the role of E2 promoter binding factors (E2Fs) and a subset of mitotic kinases controlled by E2Fs in EMT and cancer.

E2Fs in cancer, EMT, and metastasis

The E2F family of transcription factors is divided into three major groups: activators (E2F1, E2F2, and E2F3a), canonical repressors (E2f3b-E2F6), and atypical repressors (E2F7 and E2F8) of gene transcription.17 The E2F activators are silenced throughout G1 phase by physical association with the Rb tumor suppressor and are activated by the sequential phosphorylation of Rb first by the G1/S phase CDKs (initially by cyclin D/Cdk4/Cdk6 and then by cyclin E/A/Cdk2); this hyper-phosphorylation results in the occupancy of a plethora of promoters by the E2F activators.1824 The E2Fs have many functions such as the regulation of the cell cycle, centriole duplication, cell differentiation, tissue development, DNA repair, genomic integrity, apoptosis, metabolism, and angiogenesis.17,2527 Early reports focused on the ability of unregulated E2F activators to trigger hyper-proliferation and apoptosis.2833

Different subsets of E2Fs influence the survival outcomes of several cancers; for example, Huang et al.34 showed that all eight E2Fs are significantly upregulated in hepatocellular carcinoma patients and that overexpression of E2F3, E2F5, and E2F6 correlated with worse overall- and disease-free survival. Studies from our laboratory showed that overexpression of E2F1 and E2F3 strongly correlate with poor overall- and relapse-free overall survival of breast cancer patients, while E2F2 did not.35 Similarly, Liu et al.36 showed that all E2Fs are overexpressed in breast cancer with the exception of E2F4 and that E2F1, E2F3, E2F7, and E2F8 associated with poor relapse-free survival in breast cancer patients, and similar results were obtained by Li et al.37

The E2Fs can act as tumor suppressors or oncogenes depending on the tissue context. For example, loss of E2F3 in Rb+/− mice suppressed pituitary tumors but accelerated tumor formation and metastasis of thyroid tumors38; in contrast, the ablation of E2F1 in Rb+/− mice suppresses both of these tumor types.39 Likewise, increased expression of E2F1 driven by the K5 promoter in older transgenic mice resulted in a variety of spontaneous tumors including the skin, but E2F1 expression in the same mouse model blocked skin tumor development in the two-stage induction model.40 Also, the ablation of E2F1 and E2F3 in an MMTV-neu-driven mouse model of mammary cancer could severely restrict tumor growth.41 A similar MMTV-neu model showed loss of E2F1 accelerated tumorigenesis and E2F2 could suppress it, but that ablation of E2F1 or E2F2 suppressed metastasis.42 Also, E2F2 behaves differently than E2F1 or E2F3 in tumors initiated by MMTV-c-Myc, since genetic ablation of E2F2 accelerated metastasis to the lungs of mice, and increased migration and lung-colonization ability when knocked down in triple-negative MDA-MB-231 cells.43

While mechanistically the E2Fs could act as tumor suppressors or oncogenes due to their ability to suppress or induce proliferation and apoptosis in different tissue contexts, Saavedra et al.44 demonstrated that genetic ablation of E2F3a and E2F3b in primary mouse fibroblasts resulted in centrosome amplification—defined as the acquisition of three or more centrosomes that result in pseudo bipolar and multipolar mitotic spindles and aneuploidy; neither ablation of E2F1, E2F2, E2F4 or E2F5 could cause these phenotypes. These experiments suggested that loss of repression likely due to the loss of E2F3b was central in suppressing centrosome amplification and chromosome instability. Our group first demonstrated that the overexpression of E2F activators (E2F1, E2F2, and E2F3a) in MCF10A mammary epithelial cells increases centrosome amplification, chromosome instability, and deregulate timing of mitosis/cytokinesis.45 Moreover, depletion of E2F3a and E2F3b in Her2+ breast cancer cells reduced tumor growth and burden by suppressing centrosome amplification and unregulated mitosis.46 This is an important finding since centrosome amplification and chromosome instability can lead to tumor initiation,47,48 maintenance,4953 and progression46,5456. Centrosome amplification correlates with poor prognostic factors,56 causes chromosomal instability49 tumor heterogeneity,57 and promotes invasion45 through changes in cell polarity.58,59 These results suggest that activation or repression by E2Fs plays a major role in maintaining centrosome homeostasis and genomic integrity and may signal invasion and EMT through centrosome amplification and loss of cell polarity.

It is widely recognized that the E2Fs signal EMT, invasion, and metastasis.28,60 For example, overexpression of E2F1 in squamous carcinoma cells enhances their ability to invade.61 Elegant studies from the Chellappan laboratory showed that several MMPs genes are regulated by the E2Fs through an Rb-Raf-1 interaction.62 They predicted E2F binding sites on all 23 human MMPs gene promoters and showed that MMP9, MMP14, and MMP15 were E2F1-responsive. Furthermore, they demonstrated that the depletion of E2Fs reduced invasion and migration in vitro, and reduced metastasis in tail vein lung metastatic models in mice. Further, several receptor proteins have been identified and associated with E2F1 expression and EMT in breast, bladder, and lung cancers.65,66 Wang et al.67 showed that E2F1 promotes EMT by the direct regulation of ZEB2 in small cell lung carcinoma. Previous studies from these authors also showed that E2F1 regulates MMP-9 and MMP-16.68 The E2F1/SP3/STAT6 axis has been shown to increase the expression of ZEB1 and ZEB2, and signal IL-4 induced EMT in colorectal cancer.69 Also, depletion of E2F1 resulted in decreased expression of DDR1, which in turn decreased migration and invasion of osteosarcoma cells.70 Further analysis demonstrated that STAT3 signaling was impaired upon E2F1 depletion, resulting in decreased vimentin, MMP2, and MMP9, and increased E-cadherin. Liang et al.71 demonstrated that E2F1 depletion inhibited proliferation, invasion, and migration in prostate cancer cells. Meanwhile, E2F1 depletion in tumor xenograft suppressed tumor growth, invasion, and EMT by increasing E-cadherin and decreasing the expression of vimentin; in this study, CD147 was identified as an interaction partner for E2F1. Further, Knoll et al.72 showed that the E2F1-miR-224/452-TXNIP axis promotes EMT, migration, and invasion in melanoma, and can serve as a predictor of patient survival.

Fewer studies have been devoted to study the role of E2F2 and E2F3 in EMT. One of the few studies done with E2F2 shows that it correlates with elevated levels of vimentin and inversely correlates with miR-99 in lung cancer, that E2F2 inhibition allows miR-99 to repress stemness in lung cancer and that overexpression of E2F2 rescues back the migratory potential of cells overexpressing miR-99.73 Studies focused on E2F3 also center in non-coding micro RNAs. Studies from Ye et al.74 showed that forced overexpression of miR-363 decreased proliferation, migration, invasion of hepatocellular carcinoma cells, and reversed EMT in vitro and in vivo. Further analysis revealed that miR-363 targets the 3ʹ untranslated region of E2F3 and that they are inversely correlated, and that overexpression of E2F3 in cells expressing miR-363 restored colony formation, migration, and invasion capacity. The control of E2F3 by miR-363 has implications for several cancers since the aberrant expression of miR-363 and its role in tumor pathogenesis and progression have already been reported in gallbladder, colorectal, gastric, head and neck, and breast cancers. This is not exclusive to miR-363 since E2F3 has been reported to be regulated in HCC cells by several miRNAs, such as miR-141, miR-144, miR-503, miR-214 miR-424, and miR-217. In another study, it was shown that miR-200c, which is down-regulated in bladder cancer, regulates BMI-1 and E2F3.75 Mutations in two putative miR-200c-binding sites resulted in reduced cell invasion, migration, proliferation, and increased E-cadherin levels. Other studies show that the E2F activators can contribute to metastasis by influencing the tumor microenvironment. For example, a study from the Leone laboratory evidenced the role of E2F3 in metastasis, although not by inducing EMT. In their study, it was shown that the ablation of E2F3 in tumor-associated macrophages led to decreased expression of extracellular matrix modifiers that resulted in decreased pulmonary metastasis of breast cancer cells.76 Another study showed in an MMTV-polyomavirus-induced model of mammary tumor metastasis that the ablation of E2F1 and E2F2 reduced metastatic capacity by reducing the expression of several genes involved in angiogenesis, remodeling of the extracellular matrix, tumor cell survival, and tumor-cell interactions with vascular endothelial cells that promote lung metastases.77

Some studies have been devoted to study the role of the remaining E2Fs (E2F4-E2F8). To our best knowledge, the only two studies that show any effect of E2F4 in EMT were done by the Chellappan laboratory (briefly discussed above)62 and one by Baiz et al.78 In this last study bortezomib, an anti-proliferative drug that inhibits the 26S proteasome subunit promotes cell cycle arrest and apoptosis was tested in hepatocellular carcinoma cells and it promoted downregulation of E2F2 and upregulation of E2F4, E2F6, and E2F8. These changes correlated with the expression of some EMT-related genes in a microarray; however, no functional nor other expression analysis was conducted to evaluate how these changes modulate EMT. Another study showed that the treatment of NMuMG cells (mouse mammary cells) with TGFβ1 (a known inducer of EMT) resulted in E2F5 downregulation.79 Further, a gene expression analysis identified E2F5 as a possible target of LOXL2 (lysyl oxidase-like 2), which stabilizes SNAIL1 and is upregulated in pancreatic cancer.76 In addition to the study of Baiz et al., a study done in endometrial carcinoma cells showed that miR-424 (which associates with poor predictor factors, cell invasion, and migration) negatively regulates E2F663; this was evidenced by dual-luciferase reporter system and expression assays. Inhibition of miR-424 with an antisense oligonucleotide sequence increased the expression levels of E2F6. Lastly, it was shown that miR-30a-5p decreased EMT and metastasis in gallbladder carcinoma cells by targeting E2F7.64 Inhibition of miR-30a-5p resulted in increased cell proliferation, migration, invasion, and nuclear translocation of β-catenin. Meanwhile, NOZ (a gallbladder carcinoma cell line) transfected with miR-30a-5p and transplanted in nude mice resulted in slower tumor growth and reduced hepatic metastasis. E2F7 and E2F8 have been shown to also induce angiogenesis, which facilitates metastasis, by interacting with HIF1and regulating VEGFA.80

In summary, these studies together with the scarce research focus on E2F2-8 and EMT accentuate the need to redirect our attention to the study of E2F2-E2F8 because even though they have redundant functions, they have unique functions, expression, and prognostic values.

Current therapies against E2Fs, mitotic regulators, and EMT

Despite their significant contribution to tumorigenesis through cell-cycle deregulation, centrosome amplification, and chromosome instability, and their apparent role in EMT, invasion, and metastasis, E2F activators are virtually undruggable.81,82 Current therapies directed against E2Fs in cancer include inhibition of upstream regulators using CDK4/6 inhibitors (which prevent the initial phosphorylation of Rb and keep E2Fs in a repressed state), use of truncated E2F1 protein, use of small molecules or peptides to inhibit E2F activity, and the use of E2F-driven oncolytic viruses. The Leone83 and Knudsen84 groups reviewed in detail these therapies and their advantages and disadvantages. Out of these four possible therapies directed to E2Fs, CDK4/6 inhibitors and E2F-driven oncolytic viruses are in clinical trials.85,86 The major disadvantages of the CDK4/6 inhibitors are that they do not work if RB is not intact or if there is E2F amplification.84,87 On the other hand, E2F-driven oncolytic virus treatment has shown promising results. One example is ICOVIR-7, which features an RGD-4C modification of the fiber HI-loop of serotype 5 adenoviruses for enhanced entry into tumor cells and has been tested clinically.86 Treatment with ICOVIR-7 showed anti-tumoral efficiency in 9 out of 17 patients, while 5 out of 12 exhibited stabilization or reduced tumor size. The remaining two target therapies mentioned above are not yet in clinical trials and major complications include the delivery method, specificity against individual E2Fs, and potential side effects such as mounting a general autoimmune response against viruses.

Despite E2Fs not currently being ideal targets for cancer therapy because they are transcription factors that regulate numerous essential normal cell processes, and due to the lack of selectivity against specific E2Fs, E2F-regulated mitotic kinases and regulators, including NEK2, Mps1/TTK, BUB1, BUBR1, and SGO1 can serve as potential drug targets. We recently demonstrated that individual expression of E2F1 or E2F3 in mammary epithelial cells elevates the levels of multiple regulators of mitosis and/or the centrosome, including NEK2, Mps1/TTK, SGO1, HEC1, BUBR1, and PP2AC.35 Some of these centrosome/mitotic regulators have effects on EMT and this notion was first explored by investigating the effect of another gene, Aurora-A. Early studies from Wan et al.88 showed that Aurora-A, a mitotic kinase involved in centrosome segregation and spindle assembly, mediates cell invasion and EMT (denoted by changes in E-cadherin and β-catenin) in nasopharyngeal carcinoma through the MAPK pathway. This was the first report that linked a mitotic regulator with EMT induction. Here, we will discuss briefly the role of these E2F targets in EMT. However, bear in mind that E2F regulates many targets that can promote EMT through pathways not yet consider here—many of which have been discussed in the previous section.

NEK2

NEK2 (never-in-mitosis-a-related kinase 2) is a serine/threonine that localizes in the centrosome and has multiple roles in the cell cycle. NEK2 belongs to the NEK family (which has 11 other members). However, NEK2 has received special attention in cancer due to its numerous functions in normal and oncogenic states. The NEK2 gene has three splice variants (NEK2A, NEK2B, NEK2C), being NEK2A the full-length protein.8991 The N-terminal of NEK2 is the catalytic kinase domain, while the C-terminal is the regulatory domain. NEK2 expression peaks in S and G2 phase.90 It has been shown that E2F4 binds to the NEK2 promoter during the G1 phase serving as a transcriptional repressor.92 Our lab also showed that NEK2 is under the control of the Cdk4 pathway and the E2Fs and that NEK2 can rescue back centrosome amplification in Her2+ breast cancer cells silenced for Cdk4 or E2F3.35,9395 NEK2 is also repressed by p53 and miRNA-128.96,97 On the other hand, NEK2 is positively regulated by FoxM1.98,99 NEK2 is degraded by the ubiquitin-proteosome complex and activated by auto-phosphorylation at several residues—reviewed in Fang and Zhang.100 NEK2 participates in centrosome duplication and centrosome separation by phosphorylating c-Nap1, Rootletin, and Cep68.101103 Moreover, NEK2 stabilizes microtubules through phosphorylation of Nlp and centrobin during interphase.104 Also, NEK2 phosphorylates Hec1 to control kinetochore attachment and spindle assembly, specifically chromosome alignment by direct interaction with MAD1 or phosphorylation of Hec1 and SGO1.105107 NEK2 also mediates chromosome separation.100,108 Apart from these roles in the cell cycle, NEK2 is a novel regulator of B cell development and immune response.109

NEK2 is clinically relevant for cancer treatment since it has been shown to be upregulated in diverse cancer cells and cancer types.100,110 We demonstrated that Nek2 may represent an important target against mammary cancers since it can significantly suppress centrosome amplification and chromosome instability triggered by Her2, H-RasG12V, or H-RasG12V and c-Myc.35,9395 Xenograft studies have demonstrated that NEK2 depletion results in reduced tumor growth.111 Furthermore, studies have shown that uncontrolled NEK2 activity can lead to chromosomal instability,112 which can have an effect on tumorigenesis and tumor progression.57 Our studies showed that NEK2 overexpression in Her2+ breast cancer cells downregulated for E2F3 generates pre-invasive protrusions in 3D cell culture models (mammospheres).45 A recent study shows that NEK2 correlates with migration, invasion, and EMT in hepatocellular cells.113 Some of the genes that were modulated by Nek2 were E-cadherin, N-cadherin, and vimentin, both in cancer cell lines as well as in patient tissues. Gene microarray identified other potential EMT-related genes that might be modulated. Another study correlated NEK2 expression with hepatoma metastasis and showed that NEK2 modulates E-cadherin and MMP9.114 Das et al.115 showed that NEK2 overexpression leads to upregulation of the Wnt orthologue wingless and alteration of E-cadherin, β-catenin, and activation of the Akt pathway; Nek2 cooperates mitogenic pathways (Src or Ras) to cause distant metastasis in Drosophila melanogaster; in contrast, such cooperation was abrogated by PI3K inhibitors. Studies from Mbom et al.116 further investigated how NEK2 regulates β-catenin and found that NEK2 (under the regulation of Plk1) binds and phosphorylates the N-terminus of β-catenin (independently of GSK3β). Phosphorylation of β-catenin prevents ubiquitination and degradation; thus, β-catenin accumulates at centrosomes during mitosis. Moreover, NEK2 has been shown to contribute to drug resistance. One study showed that NEK2 induces drug resistance through the activation of efflux drug pumps in myeloma.117

Surprisingly, there is not any current inhibitor for NEK2 in clinical trials; however, some studies have tested the effects of pharmacological inhibition of NEK2 or downregulation of NEK2 by RNAi in cellular and animal models. Other strategies include blocking of ATP-binding site and interruption of protein interaction. Studies from Kaowinn et al.118 tested the efficacy of CGK062, a small chemical molecule that inhibits CUG2, which is a gene that interacts with NEK2. Findings from this study showed that CGK062 promotes phosphorylation of β-catenin at Ser33/Ser37 and induced its degradation; thus, posing a potential therapeutic use in lung cancer. Another study showed that the treatment of non-small cell lung cancer cells with deguelin, a rotenoid extracted from Mundulea sericea resulted in lower Nek2 levels, reduced migration, and invasion.119 Ectopic expression of Nek2 negated deguelin function by preventing elevation of mRNAs coding for E-cadherin and the downregulation of vimentin mRNA. Another approach currently tested is NEK2 siRNA therapy using a portal venous port-catheter system for liver metastasis in pancreatic cancer.120 In their study, NEK2 siRNA inhibited tumor growth in a subcutaneous xenograft mouse model, prolonged survival time, and prevented liver metastasis. The main limitation of RNAi inhibition is the delivery method. Regarding ATP-binding, site blocking is the poor selectivity. The most promising approach is the interruption of protein inhibition. Notably, INH1, which inhibits the NEK2/Hec1 interaction121 has shown anti-tumoral activity by reducing tumor growth in breast cancer nude mice models. A series of analogs of INH1 have been synthesized to improve its efficacy.

Mps1/TTK

Monopolar spindle 1 kinase (Mps1) or TTK is a serine, threonine, and tyrosine kinase that has multiple roles in mitosis and meiosis.122 TTK is widely distributed in most eukaryotes and has alternative splice forms in humans and mice.122 TTK is found in centrosomes and is required for the assembly of the spindle pole body in yeast and contributes to centrosome duplication in various eukaryotes—reviewed in Pike and Fisk123— albeit its role in centriole duplication in human cells has been challenged.124 TTK localizes at kinetochores and ensures control of proper biorientation of sister chromatids on the mitotic spindle at kinetochores in collaboration with AURORA kinase B and Hec1/NDC80 as its substrate.125,126 Besides the regulation of TTK in the spindle assembly checkpoint, TTK is involved in the exit of mitosis and cytokinesis127 through the interaction with Mob1 and MIP1.128,129 Apart from its roles in mitosis, TTK is involved in the genotoxic stress response (reviewed in Liu and Winey122) and meiosis I and II.130 The C-terminal catalytic domain undergoes auto-phosphorylation at serine, threonine, and tyrosine sites (to a lesser extent) to activate itself.131 Auto-phosphorylation also helps in subcellular localization. Like NEK2, TTK is controlled at the transcriptional level by the E2Fs, specifically, E2F4 and to a lesser degree E2F1 binds to TTK promoter and represses TTK transcription in cells during interphase.92 TTK protein levels also rise upon overexpression of E2F1 or E2F3 in human mammary epithelial cells.35 Meanwhile, IL-2 appears to be an inductor of TTK.132 The major route of TTK inactivation is through degradation in anaphase by the ubiquitin E3 ligase APC/Cdc20.133 Another proposed mechanism of degradation is de-phosphorylation, however; to this date, there has not been any phosphatase identified yet.

TTK is deregulated in several human tumors. Elevated levels of TTK have been reported in thyroid papillary carcinoma, breast cancer, gastric cancer, lung cancer, and many more and its elevation correlates with poor prognosis and tumor aggressiveness—reviewed in Liu and Winey.122 TTK is often overexpressed in hepatocellular carcinomas and promotes proliferation, anchorage-independent growth, and cell migration.134 Previously, we showed that TTK is overexpressed in Her2+ cells that also display elevated levels of centrosome amplification and that its silencing reduces that process in Her2+ cells.135 Recently, we showed the highly novel finding that mitotic kinases may be involved in the early stages of metastasis by showing that pharmacologic inhibition and silencing of TTK reversed EMT, suppressed invasion, and increased the expression of KLF5, an effector of TGF-β signaling; further, TTK inhibition decreased miR-21 but increased miR-200 expression and suppressed TGF-β signaling.136 Thus, TTK may be an excellent target to suppress cancer cell survival and progression to metastasis. Currently, there is an Mps1/TTK inhibitor in clinical trial Phase I (CFI-402257), in which MPS1 inhibitor is administered along with paclitaxel in patients with advanced/metastatic Her2—breast cancer (NCT03568422) to induce tumor death by increasing chromosome mis-segregation—reviewed in Jusino et al.57 Also, this inhibitor (CFI-402257) is used alone to treat advanced solid tumors from patients enrolled in another clinical trial Phase I (NCT02792465).

SGO1

SGO1 (Shugoshin 1) plays a role in mediating the centromeric cohesion of sister chromatids, preventing their premature dissociation during cell division.137 SGO1 is also known as a guardian of the genome due to its role in suppressing chromosomal instability during cell division. SGO1 has several splicing variants and along with SGO2, they conform to the Shugoshin family.138 Here we will focus only on SGO1 due to its novel role in EMT. SGO1 N-terminal homo-dimerizes and interacts with other proteins, while the C-terminal contributes to chromosomal localization.138,139 SGO1 is widely distributed in eukaryotes such as yeast, invertebrates (i.e. Drosophila melanogaster), plants, mice, etc., and induces similar functions to human SGO1 when expressed in these species.139 SGO1 levels are low during G1, gradually increases during S phase, accumulates at G2, and peaks in mitosis (during prometaphase and metaphase), and are followed by a rapid decline upon exiting from mitosis due to proteolytic cleavage dependent on the APC/C complex through removal of Ken box or D box (destruction box) sequences.140143 SGO1 has nuclear localization sequences and phosphorylation sites for CDKs,137 Plk1,144 AURORA B,137 BUB1,145,146 and NEK2A.107 In early mitosis, SGO1 co-localizes at the inner centromeres with Cohesin, and a small fraction localizes at the kinetochores at metaphase.147 SGO1 forms a complex with PP2Ato prevent premature dissociation of Cohesin from sister chromatids preventing phosphorylation of SCC3 by Plk1.148 BUB1 and AURORA B are important for localization of SGO1 in centromeres.145,146 On the other hand, phosphorylation by NEK2A is not required for SGO1 assembly but for chromosome congression.107

Until recently, research on SGO1 was focused on cell embryology; however, some studies have shown the role of SGO1 on tumorigenesis of intestinal cancer,149 liver cancer,150,151 neuroblastoma,152 lung cancer (through Wnt activation),153 and colon cancer.154 An interesting study from Mu et al.155 showed that SGO1 overexpression correlates with poor prognostic factors in prostate cancer. Furthermore, the SGO1 knockdown resulted in the inhibition of prostate cancer cell proliferation, migration, invasion, and changes in EMT (through the rescue of E-cadherin protein levels and decrease in vimentin protein levels). Other studies and ours have shown the role of SGO1 in the suppression of chromosome instability.35,156158

Particularly, our studies show that overexpression of E2F1 and E2F3 leads to overexpression of SGO1 in mammary epithelial cells and that SGO1 guard against centrosome amplification and chromosome instability in these cells35 and that silencing of SGO1 suppresses centrosome amplification in Her2+ cells.135 Since SGO1 is essential for proper spindle assembly checkpoint functioning and chromosome cohesion,137,146,159 and our studies have shown its role in centrosome amplification and chromosome instability,35,135 SGO1 represents a novel drug target for cancer. Furthermore, SGO1 localization to chromosome arms and centromeres is controlled by phosphorylation by the kinases Aurora B160 and BUB1.146 In turn, SGO1 brings the kinase PLK1 to centromeres.161,162 Therefore, Sgo1 is upstream and downstream of proteins currently in clinical trials and its levels and function may affect outcomes of these patients—currently in clinical trials. However, far more research needs to be done to elucidate the mechanism of SGO1 in EMT and how can be targeted, and should be used as a cancer target with care, because it can suppress or induce chromosome instability and centrosome amplification, depending on cell context.

Conclusion

As discussed above, both E2Fs and E2F targets exert a role in EMT. However, since E2Fs are presently undruggable, due to their numerous important cellular functions and the lack of selectivity against individual E2Fs, we propose that E2F targets, such as NEK2, TTK, or SGO1 are further investigated in order to design clinical small molecules that inhibit them and prevent EMT. There is a relatively new classification of EMT. This sub-classification is relatively new and there are no publications that correlate E2Fs with any phase. The article published by Pastushenko et al.16 makes a distinction based on EMT gene expression, cell adhesion, invasiveness, plasticity, stemness, and metastasis. Rather than focusing on pathways that mediate these changes, the author focuses on what type of cancer exemplifies these phases. For example, breast cancer grouped into the Hybrid EMT state category. Since we study E2Fs in the context of breast cancer, our data are consistent with this classification. We have observed that E2Fs associated with the hybrid EMT state, characterized by the expression of certain mesenchymal genes like vimentin and N-cadherin and certain EMT transcription factors while not influencing the expression of other genes such as E-cadherin (Jusino and Saavedra, unpublished). The E2Fs may also help in the late stages of metastasis, where cells can undergo the mesenchymal to epithelial transition (MET) to be able to grow in the metastatic site. However, this is all speculation at this point.

We propose the model that that overexpression of E2F activators (E2F1-3) or downregulation of E2Fs repressors (E2F4-E2F8) leads to the deregulation of mitotic kinases such as NEK2A and Mps1/TTK and/or mitotic regulators such as SGO1 (Figure 1). In turn, these mitotic regulators signal EMT transcription factors and EMT markers (and plenty of studies have been published showing that these mitotic regulators signal EMT). EMT modulation, in turn, leads to invasion and metastasis. As discussed previously, EMT is a complex cellular process that facilitates invasion and migration to distant sites through cytoskeleton remodeling and increased expression and activity of metalloproteases (MMP) that facilitate metastasis. Likewise, we cited several studies that have shown that miRNAs promote EMT through the E2Fs. Therefore, an alternative model is that E2Fs signal EMT independently of these mitotic regulators. Similarly, it is possible that these mitotic regulators signal EMT independently of the E2Fs. In our model, we present two alternatives. The first is that these mitotic regulators signal EMT transcription factors or EMT markers independently of E2Fs or under the regulation of other genes not considered here. The second is that these mitotic regulators lead to chromosome instability and centrosome amplification leading to the formation of pre-invasive structures and eventually metastasis.

Figure 1.

Figure 1.

Model depicting how the E2F transcriptional activators and mitotic kinases under their control may control EMT in cancer. Overexpression of E2F activators (E2F1-3) or downregulation of E2Fs repressors (E2F4-8) leads to mitotic regulator deregulation (NEK2, MPS1/TTK, SGO1), which in turn lead to a cascade of events that ultimately leads to metastasis. Alternative models suggest miRNA may mediate these changes through controlling the E2Fs activators or mitotic regulators, that the E2Fs may regulate EMT independently of these mitotic regulators by regulating EMT transcription factors or markers, or that these mitotic regulators signal EMT and/or lead to metastasis through chromosome instability (CIN), centrosome amplification (CA), or independently of E2Fs. (A color version of this figure is available in the online journal.)

Thus, it remains some questions to be answered in order to elucidate the mechanism by which E2Fs, mitotic regulators, and EMT intertwine to lead to metastasis. EMT also leads to therapy resistance, stemness, and tumor heterogeneity; therefore, understanding the molecular mechanisms that underline this process is critical to improving current cancer therapies.

Authors’ contributions

The general concept and editing were done by Harold I. Saavedra. The literature research, specific sections, and concepts, writing and design of the figure were done by Shirley Jusino.

DECLARATION OF CONFLICTING INTERESTS

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was supported by the NIGMS-RISE Training Program (R25GM082406), by PSM-U54-CA163071 and MCC-U54-CA163068 from the National Institutes of Health. The project was also supported by 2U54MD007587 from the PRCTRC, G12MD007579 from RCMI, The Puerto Rico Science, Technology and Research Trust, and Ponce Medical School Foundation Inc. under the cooperative agreement 2016–00026.

References

  • 1.Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat 1995; 154:8–20 [DOI] [PubMed] [Google Scholar]
  • 2.Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 2006; 7:131–42 [DOI] [PubMed] [Google Scholar]
  • 3.Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009; 139:871–90 [DOI] [PubMed] [Google Scholar]
  • 4.Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2014; 15:178–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Huang RY, Guilford P, Thiery JP. Early events in cell adhesion and polarity during epithelial-mesenchymal transition. J Cell Sci 2012; 125:4417–22 [DOI] [PubMed] [Google Scholar]
  • 6.Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 2007; 7:415–28 [DOI] [PubMed] [Google Scholar]
  • 7.Wheelock MJ, Shintani Y, Maeda M, Fukumoto Y, Johnson KR. Cadherin switching. J Cell Sci 2008; 121:727–35 [DOI] [PubMed] [Google Scholar]
  • 8.Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev 2009; 28:15–33 [DOI] [PubMed] [Google Scholar]
  • 9.Nistico P, Bissell MJ, Radisky DC. Epithelial-mesenchymal transition: general principles and pathological relevance with special emphasis on the role of matrix metalloproteinases. Cold Spring Harb Perspect Biol 2012; 4:pii: a011908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Barrallo-Gimeno A, Nieto MA. The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development 2005; 132:3151–61 [DOI] [PubMed] [Google Scholar]
  • 11.De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nature Rev Cancer 2013; 13:97–110 [DOI] [PubMed] [Google Scholar]
  • 12.Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res 2009; 19:156–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009; 119:1420–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wang Y, Zhou BP. Epithelial-mesenchymal transition – a hallmark of breast cancer metastasis. Cancer Hallm 2013; 1:38–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Creighton CJ, Gibbons DL, Kurie JM. The role of epithelial-mesenchymal transition programming in invasion and metastasis: a clinical perspective. Cancer Manag Res 2013; 5:187–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol 2019; 29:212–26 [DOI] [PubMed] [Google Scholar]
  • 17.Lammens T, Li J, Leone G, De Veylder L. Atypical E2Fs: new players in the E2F transcription factor family. Trends Cell Biol 2009; 19:111–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hiebert SW, Chellappan SP, Horowitz JM, Nevins JR. The interaction of RB with E2F coincides with an inhibition of the transcriptional activity of E2F. Genes Dev 1992; 6:177–85 [DOI] [PubMed] [Google Scholar]
  • 19.Arroyo M, Raychaudhuri P. Retinoblastoma-repression of E2F-dependent transcription depends on the ability of the retinoblastoma protein to interact with E2F and is abrogated by the adenovirus E1A oncoprotein. Nucl Acids Res 1992; 20:5947–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Qin XQ, Chittenden T, Livingston DM, Kaelin WG., Jr. Identification of a growth suppression domain within the retinoblastoma gene product. Genes Dev 1992; 6:953–64 [DOI] [PubMed] [Google Scholar]
  • 21.Weintraub SJ, Prater CA, Dean DC. Retinoblastoma protein switches the E2F site from positive to negative element. Nature 1992; 358:259–61 [DOI] [PubMed] [Google Scholar]
  • 22.Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR. The E2F transcription factor is a cellular target for the RB protein. Cell 1991; 65:1053–61 [DOI] [PubMed] [Google Scholar]
  • 23.Chittenden T, Livingston DM, Kaelin WG., Jr. RB associates with an E2F-like, sequence-specific DNA-binding protein. Cold Spring Harb Symp Quant Biol 1991; 56:187–95 [DOI] [PubMed] [Google Scholar]
  • 24.Bagchi S, Weinmann R, Raychaudhuri P. The retinoblastoma protein copurifies with E2F-I, an E1A-regulated inhibitor of the transcription factor E2F. Cell 1991; 65:1063–72 [DOI] [PubMed] [Google Scholar]
  • 25.Iaquinta PJ, Lees JA. Life and death decisions by the E2F transcription factors. Curr Opin Cell Biol 2007; 19:649–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Attwooll C, Lazzerini Denchi E, Helin K. The E2F family: specific functions and overlapping interests. EMBO J 2004; 23:4709–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Trimarchi JM, Lees JA. Sibling rivalry in the E2F family. Nat Rev Mol Cell Biol 2002; 3:11–20 [DOI] [PubMed] [Google Scholar]
  • 28.Chen HZ, Tsai SY, Leone G. Emerging roles of E2Fs in cancer: an exit from cell cycle control. Nat Rev Cancer 2009; 9:785–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cam H, Dynlacht BD. Emerging roles for E2F: beyond the G1/S transition and DNA replication. Cancer Cell 2003; 3:311–6 [DOI] [PubMed] [Google Scholar]
  • 30.DeGregori J. The genetics of the E2F family of transcription factors: shared functions and unique roles. Biochim Biophys Acta 2002; 1602:131–50 [DOI] [PubMed] [Google Scholar]
  • 31.Stevaux O, Dyson NJ. A revised picture of the E2F transcriptional network and RB function. Curr Opin Cell Biol 2002; 14:684–91 [DOI] [PubMed] [Google Scholar]
  • 32.Saavedra HI, Wu L, de Bruin A, Timmers C, Rosol TJ, Weinstein M, Robinson ML, Leone G. Specificity of E2F1, E2F2, and E2F3 in mediating phenotypes induced by loss of Rb. Cell Growth Differ 2002; 13:215–25 [PubMed] [Google Scholar]
  • 33.Shan B, Lee WH. Deregulated expression of E2F-1 induces S-phase entry and leads to apoptosis. Mol Cell Biol 1994; 14:8166–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Huang YL, Ning G, Chen LB, Lian YF, Gu YR, Wang JL, Chen DM, Wei H, Huang YH. Promising diagnostic and prognostic value of E2Fs in human hepatocellular carcinoma. Cancer Manag Res 2019; 11:1725–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lee M, Rivera-Rivera Y, Moreno CS, Saavedra HI. The E2F activators control multiple mitotic regulators and maintain genomic integrity through Sgo1 and BubR1. Oncotarget 2017; 8:77649–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Liu ZL, Bi XW, Liu PP, Lei DX, Wang Y, Li ZM, Jiang WQ, Xia Y. Expressions and prognostic values of the E2F transcription factors in human breast carcinoma. Cancer Manag Res 2018; 10:3521–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Li Y, Huang J, Yang D, Xiang S, Sun J, Li H, Ren G. Expression patterns of E2F transcription factors and their potential prognostic roles in breast cancer. Oncol Lett 2018; 15:9216–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ziebold U, Lee EY, Bronson RT, Lees JA. E2F3 loss has opposing effects on different pRB-deficient tumors, resulting in suppression of pituitary tumors but metastasis of medullary thyroid carcinomas. Mol Cell Biol 2003; 23:6542–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yamasaki L, Bronson R, Williams BO, Dyson NJ, Harlow E, Jacks T. Loss of E2F-1 reduces tumorigenesis and extends the lifespan of Rb1(+/−) mice. Nat Genet 1998; 18:360–4 [DOI] [PubMed] [Google Scholar]
  • 40.Pierce AM, Schneider-Broussard R, Gimenez-Conti IB, Russell JL, Conti CJ, Johnson DG. E2F1 has both oncogenic and tumor-suppressive properties in a transgenic model. Mol Cell Biol 1999; 19:6408–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wu L, de Bruin A, Wang H, Simmons T, Cleghorn W, Goldenberg LE, Sites E, Sandy A, Trimboli A, Fernandez SA, Eng C, Shapiro C, Leone G. Selective roles of E2Fs for ErbB2- and Myc-mediated mammary tumorigenesis. Oncogene 2015; 34:119–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Andrechek ER. HER2/Neu tumorigenesis and metastasis is regulated by E2F activator transcription factors. Oncogene 2015; 34:217–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yuwanita I, Barnes D, Monterey MD, O'Reilly S, Andrechek ER. Increased metastasis with loss of E2F2 in Myc-driven tumors. Oncotarget 2015; 6:38210–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Saavedra HI, Maiti B, Timmers C, Altura R, Tokuyama Y, Fukasawa K, Leone G. Inactivation of E2F3 results in centrosome amplification. Cancer Cell 2003; 3:333–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lee M, Oprea-Ilies G, Saavedra HI. Silencing of E2F3 suppresses tumor growth of Her2+ breast cancer cells by restricting mitosis. Oncotarget 2015; 6:37316–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lee MY, Moreno CS, Saavedra HI. E2F activators signal and maintain centrosome amplification in breast cancer cells. Mol Cell Biol 2014; 34:2581–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Adon AM, Zeng X, Harrison MK, Sannem S, Kiyokawa H, Kaldis P, Saavedra HI. Cdk2 and Cdk4 regulate the centrosome cycle and are critical mediators of centrosome amplification in p53-null cells. Mol Cell Biol 2010; 30:694–710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Levine MS, Bakker B, Boeckx B, Moyett J, Lu J, Vitre B, Spierings DC, Lansdorp PM, Cleveland DW, Lambrechts D, Foijer F, Holland AJ. Centrosome amplification is sufficient to promote spontaneous tumorigenesis in mammals. Dev Cell 2017; 40:313–22.e5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Godinho SA, Pellman D. Causes and consequences of centrosome abnormalities in cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Okuda M, Horn HF, Tarapore P, Tokuyama Y, Smulian AG, Chan PK, Knudsen ES, Hofmann IA, Snyder JD, Bove KE, Fukasawa K. Nucleophosmin/B23 is a target of CDK2/cyclin E in centrosome duplication. Cell 2000; 103:127–40 [DOI] [PubMed] [Google Scholar]
  • 51.Dzhindzhev NS, Tzolovsky G, Lipinszki Z, Abdelaziz M, Debski J, Dadlez M, Glover DM. Two-step phosphorylation of Ana2 by Plk4 is required for the sequential loading of Ana2 and Sas6 to initiate procentriole formation. Open Biol 2017; 7:170247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vulprecht J, David A, Tibelius A, Castiel A, Konotop G, Liu F, Bestvater F, Raab MS, Zentgraf H, Izraeli S, Kramer A. STIL is required for centriole duplication in human cells. J Cell Sci 2012; 125:1353–62 [DOI] [PubMed] [Google Scholar]
  • 53.Arquint C, Cubizolles F, Morand A, Schmidt A, Nigg EA. The SKP1-Cullin-F-box E3 ligase betaTrCP and CDK2 cooperate to control STIL abundance and centriole number. Open Biol 2018; 8:pii:170-523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tokuyama Y, Horn HF, Kawamura K, Tarapore P, Fukasawa K. Specific phosphorylation of nucleophosmin on Thr(199) by cyclin-dependent kinase 2-cyclin E and its role in centrosome duplication. J Biol Chem 2001; 276:21529–37 [DOI] [PubMed] [Google Scholar]
  • 55.McLamarrah TA, Buster DW, Galletta BJ, Boese CJ, Ryniawec JM, Hollingsworth NA, Byrnes AE, Brownlee CW, Slep KC, Rusan NM, Rogers GC. An ordered pattern of Ana2 phosphorylation by Plk4 is required for centriole assembly. J Cell Biol 2018; 217:1217–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Denu RA, Zasadil LM, Kanugh C, Laffin J, Weaver BA, Burkard ME. Centrosome amplification induces high grade features and is prognostic of worse outcomes in breast cancer. BMC Cancer 2016; 16:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jusino S, Fernandez-Padin FM, Saavedra HI. Centrosome aberrations and chromosome instability contribute to tumorigenesis and intra-tumor heterogeneity. J Cancer Metastasis Treat 2018; 4:pii:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Godinho SA, Picone R, Burute M, Dagher R, Su Y, Leung CT, Polyak K, Brugge JS, Thery M, Pellman D. Oncogene-like induction of cellular invasion from centrosome amplification. Nature 2014; 510:167–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lee M, Oprea-Ilies G, Saavedra HI. Silencing of E2F3 suppresses tumor growth of Her2+ breast cancer cells by restricting mitosis. Oncotarget 2015; 6:37316–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Schaal C, Pillai S, Chellappan SP. The Rb-E2F transcriptional regulatory pathway in tumor angiogenesis and metastasis. Adv Cancer Res 2014; 121:147–82 [DOI] [PubMed] [Google Scholar]
  • 61.Zhang SY, Liu SC, Johnson DG, Klein-Szanto AJ. E2F-1 gene transfer enhances invasiveness of human head and neck carcinoma cell lines. Cancer Res 2000; 60:5972–6 [PubMed] [Google Scholar]
  • 62.Johnson JL, Pillai S, Pernazza D, Sebti SM, Lawrence NJ, Chellappan SP. Regulation of matrix metalloproteinase genes by E2F transcription factors: Rb-Raf-1 interaction as a novel target for metastatic disease. Cancer Res 2012; 72:516–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lu Z, Nian Z, Jingjing Z, Tao L, Quan L. MicroRNA-424/E2F6 feedback loop modulates cell invasion, migration and EMT in endometrial carcinoma. Oncotarget 2017; 8:114281–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ye YY, Mei JW, Xiang SS, Li HF, Ma Q, Song XL, Wang Z, Zhang YC, Liu YC, Jin YP, Hu YP, Jiang L, Liu FT, Zhang YJ, Hao YJ, Liu YB. MicroRNA-30a-5p inhibits gallbladder cancer cell proliferation, migration and metastasis by targeting E2F7. Cell Death Dis 2018; 9:410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pillai S, Trevino J, Rawal B, Singh S, Kovacs M, Li X, Schell M, Haura E, Bepler G, Chellappan S. beta-arrestin-1 mediates nicotine-induced metastasis through E2F1 target genes that modulate epithelial-mesenchymal transition. Cancer Res 2015; 75:1009–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Khan FM, Marquardt S, Gupta SK, Knoll S, Schmitz U, Spitschak A, Engelmann D, Vera J, Wolkenhauer O, Putzer BM. Unraveling a tumor type-specific regulatory core underlying E2F1-mediated epithelial-mesenchymal transition to predict receptor protein signatures. Nat Commun 2017; 8:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wang T, Chen X, Qiao W, Kong L, Sun D, Li Z. Transcription factor E2F1 promotes EMT by regulating ZEB2 in small cell lung cancer. BMC Cancer 2017; 17:719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li Z, Guo Y, Jiang H, Zhang T, Jin C, Young CY, Yuan H. Differential regulation of MMPs by E2F1, Sp1 and NF-kappa B controls the small cell lung cancer invasive phenotype. BMC Cancer 2014; 14:276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chen J, Gong C, Mao H, Li Z, Fang Z, Chen Q, Lin M, Jiang X, Hu Y, Wang W, Zhang X, Chen X, Li H. E2F1/SP3/STAT6 axis is required for IL-4-induced epithelial-mesenchymal transition of colorectal cancer cells. Int J Oncol 2018; 53:567–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang Z, Sun X, Bao Y, Mo J, Du H, Hu J, Zhang X. E2F1 silencing inhibits migration and invasion of osteosarcoma cells via regulating DDR1 expression. Int J Oncol 2017; 51:1639–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liang YX, Lu JM, Mo RJ, He HC, Xie J, Jiang FN, Lin ZY, Chen YR, Wu YD, Luo HW, Luo Z, Zhong WD. E2F1 promotes tumor cell invasion and migration through regulating CD147 in prostate cancer. Int J Oncol 2016; 48:1650–8 [DOI] [PubMed] [Google Scholar]
  • 72.Knoll S, Furst K, Kowtharapu B, Schmitz U, Marquardt S, Wolkenhauer O, Martin H, Putzer BM. E2F1 induces miR-224/452 expression to drive EMT through TXNIP downregulation. EMBO Rep 2014; 15:1315–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Feliciano A, Garcia-Mayea Y, Jubierre L, Mir C, Hummel M, Castellvi J, Hernandez-Losa J, Paciucci R, Sansano I, Sun Y, Ramon YCS, Kondon H, Soriano A, Segura M, Lyakhovich A, ME LL. miR-99a reveals two novel oncogenic proteins E2F2 and EMR2 and represses stemness in lung cancer. Cell Death Dis 2017; 8:e3141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ye J, Zhang W, Liu S, Liu Y, Liu K. miR-363 inhibits the growth, migration and invasion of hepatocellular carcinoma cells by regulating E2F3. Oncol Rep 2017; 38:3677–84 [DOI] [PubMed] [Google Scholar]
  • 75.Liu L, Qiu M, Tan G, Liang Z, Qin Y, Chen L, Chen H, Liu J. miR-200c inhibits invasion, migration and proliferation of bladder cancer cells through down-regulation of BMI-1 and E2F3. J Transl Med 2014; 12:305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ruckert F, Joensson P, Saeger HD, Grutzmann R, Pilarsky C. Functional analysis of LOXL2 in pancreatic carcinoma. Int J Colorectal Dis 2010; 25:303–11 [DOI] [PubMed] [Google Scholar]
  • 77.Hollern DP, Honeysett J, Cardiff RD, Andrechek ER. The E2F transcription factors regulate tumor development and metastasis in a mouse model of metastatic breast cancer. Mol Cell Biol 2014; 34:3229–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Baiz D, Dapas B, Farra R, Scaggiante B, Pozzato G, Zanconati F, Fiotti N, Consoloni L, Chiaretti S, Grassi G. Bortezomib effect on E2F and cyclin family members in human hepatocellular carcinoma cell lines. World J Gastroenterol 2014; 20:795–803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Xie L, Law BK, Aakre ME, Edgerton M, Shyr Y, Bhowmick NA, Moses HL. Transforming growth factor beta-regulated gene expression in a mouse mammary gland epithelial cell line. Breast Cancer Res 2003; 5:R187–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Weijts BG, Bakker WJ, Cornelissen PW, Liang KH, Schaftenaar FH, Westendorp B, de Wolf CA, Paciejewska M, Scheele CL, Kent L, Leone G, Schulte-Merker S, de Bruin A. E2F7 and E2F8 promote angiogenesis through transcriptional activation of VEGFA in cooperation with HIF1. EMBO J 2012; 31:3871–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dang CV, Reddy EP, Shokat KM, Soucek L. Drugging the 'undruggable' cancer targets. Nat Rev Cancer 2017; 17:502–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yan C, Higgins PJ. Drugging the undruggable: transcription therapy for cancer. Biochim Biophys Acta 2013; 1835:76–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kent LN, Leone G. The broken cycle: E2F dysfunction in cancer. Nat Rev Cancer 2019; 19:326–38 [DOI] [PubMed] [Google Scholar]
  • 84.Knudsen ES, Witkiewicz AK. The strange case of CDK4/6 inhibitors: mechanisms, resistance, and combination strategies. Trends Cancer 2017; 3:39–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Whittaker SR, Mallinger A, Workman P, Clarke PA. Inhibitors of cyclin-dependent kinases as cancer therapeutics. Pharmacol Ther 2017; 173:83–105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nokisalmi P, Pesonen S, Escutenaire S, Sarkioja M, Raki M, Cerullo V, Laasonen L, Alemany R, Rojas J, Cascallo M, Guse K, Rajecki M, Kangasniemi L, Haavisto E, Karioja-Kallio A, Hannuksela P, Oksanen M, Kanerva A, Joensuu T, Ahtiainen L, Hemminki A. Oncolytic adenovirus ICOVIR-7 in patients with advanced and refractory solid tumors. Clin Cancer Res 2010; 16:3035–43 [DOI] [PubMed] [Google Scholar]
  • 87.Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene 2010; 29:4018–32 [DOI] [PubMed] [Google Scholar]
  • 88.Wan XB, Long ZJ, Yan M, Xu J, Xia LP, Liu L, Zhao Y, Huang XF, Wang XR, Zhu XF, Hong MH, Liu Q. Inhibition of Aurora-A suppresses epithelial-mesenchymal transition and invasion by downregulating MAPK in nasopharyngeal carcinoma cells. Carcinogenesis 2008; 29:1930–7 [DOI] [PubMed] [Google Scholar]
  • 89.Schultz SJ, Fry AM, Sutterlin C, Ried T, Nigg EA. Cell cycle-dependent expression of Nek2, a novel human protein kinase related to the NIMA mitotic regulator of Aspergillus nidulans. Cell Growth Differ 1994; 5:625–35 [PubMed] [Google Scholar]
  • 90.Hames RS, Fry AM. Alternative splice variants of the human centrosome kinase Nek2 exhibit distinct patterns of expression in mitosis. Biochem J 2002; 361:77–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wu W, Baxter JE, Wattam SL, Hayward DG, Fardilha M, Knebel A, Ford EM, da Cruz e Silva EF, Fry AM. Alternative splicing controls nuclear translocation of the cell cycle-regulated Nek2 kinase. J Biol Chem 2007; 282:26431–40 [DOI] [PubMed] [Google Scholar]
  • 92.Ren B, Cam H, Takahashi Y, Volkert T, Terragni J, Young RA, Dynlacht BD. E2F integrates cell cycle progression with DNA repair, replication, and G(2)/M checkpoints. Genes Dev 2002; 16:245–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lee MY, Moreno CS, Saavedra HI. The E2F activators signal and maintain centrosome amplification in breast cancer cells. Mol Cell Biol 2014; 34:2581–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Harrison Pitner MK, Saavedra HI. Cdk4 and nek2 signal binucleation and centrosome amplification in a her2+ breast cancer model. PLoS One 2013; 8:e65971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zeng X, Shaikh FY, Harrison MK, Adon AM, Trimboli AJ, Carroll KA, Sharma N, Timmers C, Chodosh LA, Leone G, Saavedra HI. The Ras oncogene signals centrosome amplification in mammary epithelial cells through cyclin D1/Cdk4 and Nek2 . Oncogene 2010; 29:5103–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Nabilsi NH, Ryder DJ, Peraza-Penton AC, Poudyal R, Loose DS, Kladde MP. Local depletion of DNA methylation identifies a repressive p53 regulatory region in the NEK2 promoter. J Biol Chem 2013; 288:35940–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Takahashi Y, Iwaya T, Sawada G, Kurashige J, Matsumura T, Uchi R, Ueo H, Takano Y, Eguchi H, Sudo T, Sugimachi K, Yamamoto H, Doki Y, Mori M, Mimori K. Up-regulation of NEK2 by microRNA-128 methylation is associated with poor prognosis in colorectal cancer. Ann Surg Oncol 2014; 21:205–12 [DOI] [PubMed] [Google Scholar]
  • 98.Laoukili J, Kooistra MR, Bras A, Kauw J, Kerkhoven RM, Morrison A, Clevers H, Medema RH. FoxM1 is required for execution of the mitotic programme and chromosome stability. Nat Cell Biol 2005; 7:126–36 [DOI] [PubMed] [Google Scholar]
  • 99.Wonsey DR, Follettie MT. Loss of the forkhead transcription factor FoxM1 causes centrosome amplification and mitotic catastrophe. Cancer Res 2005; 65:5181–9 [DOI] [PubMed] [Google Scholar]
  • 100.Fang Y, Zhang X. Targeting NEK2 as a promising therapeutic approach for cancer treatment. Cell Cycle 2016; 15:895–907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Fry AM, Mayor T, Meraldi P, Stierhof YD, Tanaka K, Nigg EA. C-Nap1, a novel centrosomal coiled-coil protein and candidate substrate of the cell cycle-regulated protein kinase Nek2. J Cell Biol 1998; 141:1563–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bahe S, Stierhof YD, Wilkinson CJ, Leiss F, Nigg EA. Rootletin forms centriole-associated filaments and functions in centrosome cohesion. J Cell Biol 2005; 171:27–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Man X, Megraw TL, Lim YP. Cep68 can be regulated by Nek2 and SCF complex. Eur J Cell Biol 2015; 94:162–72 [DOI] [PubMed] [Google Scholar]
  • 104.Rapley J, Baxter JE, Blot J, Wattam SL, Casenghi M, Meraldi P, Nigg EA, Fry AM. Coordinate regulation of the mother centriole component nlp by nek2 and plk1 protein kinases. Mol Cell Biol 2005; 25:1309–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lou Y, Yao J, Zereshki A, Dou Z, Ahmed K, Wang H, Hu J, Wang Y, Yao X. NEK2A interacts with MAD1 and possibly functions as a novel integrator of the spindle checkpoint signaling. J Biol Chem 2004; 279:20049–57 [DOI] [PubMed] [Google Scholar]
  • 106.Wei R, Ngo B, Wu G, Lee WH. Phosphorylation of the Ndc80 complex protein, HEC1, by Nek2 kinase modulates chromosome alignment and signaling of the spindle assembly checkpoint. Mol Biol Cell 2011; 22:3584–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Fu G, Ding X, Yuan K, Aikhionbare F, Yao J, Cai X, Jiang K, Yao X. Phosphorylation of human Sgo1 by NEK2A is essential for chromosome congression in mitosis. Cell Res 2007; 17:608–18 [DOI] [PubMed] [Google Scholar]
  • 108.Fry AM, Meraldi P, Nigg EA. A centrosomal function for the human Nek2 protein kinase, a member of the NIMA family of cell cycle regulators. EMBO J 1998; 17:470–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gu Z, Zhou W, Huang J, Yang Y, Wendlandt E, Xu H, He X, Tricot G, Zhan F. Nek2 is a novel regulator of B cell development and immunological response. Biomed Res Int 2014; 2014:621082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wai DH, Schaefer KL, Schramm A, Korsching E, Van Valen F, Ozaki T, Boecker W, Schweigerer L, Dockhorn-Dworniczak B, Poremba C. Expression analysis of pediatric solid tumor cell lines using oligonucleotide microarrays. Int J Oncol 2002; 20:441–51 [PubMed] [Google Scholar]
  • 111.Kokuryo T, Senga T, Yokoyama Y, Nagino M, Nimura Y, Hamaguchi M. Nek2 as an effective target for inhibition of tumorigenic growth and peritoneal dissemination of cholangiocarcinoma. Cancer Res 2007; 67:9637–42 [DOI] [PubMed] [Google Scholar]
  • 112.Rajagopalan H, Lengauer C. Aneuploidy and cancer. Nature 2004; 432:338–41 [DOI] [PubMed] [Google Scholar]
  • 113.Zhang Y, Wang W, Wang Y, Huang X, Zhang Z, Chen B, Xie W, Li S, Shen S, Peng B. NEK2 promotes hepatocellular carcinoma migration and invasion through modulation of the epithelial-mesenchymal transition. Oncol Rep 2018; 39:1023–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chang YY, Yen CJ, Chan SH, Chou YW, Lee YP, Bao CY, Huang CJ, Huang W. NEK2 promotes hepatoma metastasis and serves as biomarker for high recurrence risk after hepatic resection. Ann Hepatol 2018; 17:843–56 [DOI] [PubMed] [Google Scholar]
  • 115.Das TK, Dana D, Paroly SS, Perumal SK, Singh S, Jhun H, Pendse J, Cagan RL, Talele TT, Kumar S. Centrosomal kinase Nek2 cooperates with oncogenic pathways to promote metastasis. Oncogenesis 2013; 2:e69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mbom BC, Siemers KA, Ostrowski MA, Nelson WJ, Barth AI. Nek2 phosphorylates and stabilizes beta-catenin at mitotic centrosomes downstream of Plk1. Mol Biol Cell 2014; 25:977–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhou W, Yang Y, Xia J, Wang H, Salama ME, Xiong W, Xu H, Shetty S, Chen T, Zeng Z, Shi L, Zangari M, Miles R, Bearss D, Tricot G, Zhan F. NEK2 induces drug resistance mainly through activation of efflux drug pumps and is associated with poor prognosis in myeloma and other cancers. Cancer Cell 2013; 23:48–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kaowinn S, Oh S, Moon J, Yoo AY, Kang HY, Lee MR, Kim JE, Hwang DY, Youn SE, Koh SS, Chung YH. CGK062, a small chemical molecule, inhibits cancer upregulated gene 2induced oncogenesis through NEK2 and betacatenin. Int J Oncol 2019; 54:1295–305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhao D, Han W, Liu X, Cui D, Chen Y. Deguelin inhibits epithelial-to-mesenchymal transition and metastasis of human non-small cell lung cancer cells by regulating NIMA-related kinase 2. Thorac Cancer 2017; 8:320–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kokuryo T, Hibino S, Suzuki K, Watanabe K, Yokoyama Y, Nagino M, Senga T, Hamaguchi M. Nek2 siRNA therapy using a portal venous port-catheter system for liver metastasis in pancreatic cancer. Cancer Sci 2016; 107:1315–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wu G, Qiu XL, Zhou L, Zhu J, Chamberlin R, Lau J, Chen PL, Lee WH. Small molecule targeting the Hec1/Nek2 mitotic pathway suppresses tumor cell growth in culture and in animal. Cancer Res 2008; 68:8393–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Liu X, Winey M. The MPS1 family of protein kinases. Annu Rev Biochem 2012; 81:561–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pike AN, Fisk HA. Centriole assembly and the role of Mps1: defensible or dispensable? Cell Div 2011; 6:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Stucke VM, Sillje HH, Arnaud L, Nigg EA. Human Mps1 kinase is required for the spindle assembly checkpoint but not for centrosome duplication. EMBO J 2002; 21:1723–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.DeLuca JG, Gall WE, Ciferri C, Cimini D, Musacchio A, Salmon ED. Kinetochore microtubule dynamics and attachment stability are regulated by Hec1. Cell 2006; 127:969–82 [DOI] [PubMed] [Google Scholar]
  • 126.Maciejowski J, George KA, Terret ME, Zhang C, Shokat KM, Jallepalli PV. Mps1 directs the assembly of Cdc20 inhibitory complexes during interphase and mitosis to control M phase timing and spindle checkpoint signaling. J Cell Biol 2010; 190:89–100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Fisk HA, Mattison CP, Winey M. Human Mps1 protein kinase is required for centrosome duplication and normal mitotic progression. Proc Natl Acad Sci USA 2003; 100:14875–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Mohl DA, Huddleston MJ, Collingwood TS, Annan RS, Deshaies RJ. Dbf2-Mob1 drives relocalization of protein phosphatase Cdc14 to the cytoplasm during exit from mitosis. J Cell Biol 2009; 184:527–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mattison CP, Stumpff J, Wordeman L, Winey M. Mip1 associates with both the Mps1 kinase and actin, and is required for cell cortex stability and anaphase spindle positioning. Cell Cycle 2011; 10:783–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Straight PD, Giddings TH, Jr., Winey M. Mps1p regulates meiotic spindle pole body duplication in addition to having novel roles during sporulation. Mol Biol Cell 2000; 11:3525–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Mattison CP, Old WM, Steiner E, Huneycutt BJ, Resing KA, Ahn NG, Winey M. Mps1 activation loop autophosphorylation enhances kinase activity. J Biol Chem 2007; 282:30553–61 [DOI] [PubMed] [Google Scholar]
  • 132.Schmandt R, Hill M, Amendola A, Mills GB, Hogg D. IL-2-induced expression of TTK, a serine, threonine, tyrosine kinase, correlates with cell cycle progression. J Immunol 1994; 152:96–105 [PubMed] [Google Scholar]
  • 133.Palframan WJ, Meehl JB, Jaspersen SL, Winey M, Murray AW. Anaphase inactivation of the spindle checkpoint. Science 2006; 313:680–4 [DOI] [PubMed] [Google Scholar]
  • 134.Liu X, Liao W, Yuan Q, Ou Y, Huang J. TTK activates Akt and promotes proliferation and migration of hepatocellular carcinoma cells. Oncotarget 2015; 6:34309–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Lee MY, Marina M, King JL, Saavedra HI. Differential expression of centrosome regulators in Her2+ breast cancer cells versus non-tumorigenic MCF10A cells. Cell Div 2014; 9:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.King JL, Zhang B, Li Y, Li KP, Ni JJ, Saavedra HI, Dong JT. TTK promotes mesenchymal signaling via multiple mechanisms in triple negative breast cancer. Oncogenesis 2018; 7:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wang X, Dai W. Shugoshin, a guardian for sister chromatid segregation. Exp Cell Res 2005; 310:1–9 [DOI] [PubMed] [Google Scholar]
  • 138.Kitajima TS, Kawashima SA, Watanabe Y. The conserved kinetochore protein shugoshin protects centromeric cohesion during meiosis. Nature 2004; 427:510–7 [DOI] [PubMed] [Google Scholar]
  • 139.Yao Y, Dai W. Shugoshins function as a guardian for chromosomal stability in nuclear division. Cell Cycle 2012; 11:2631–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Karamysheva Z, Diaz-Martinez LA, Crow SE, Li B, Yu H. Multiple anaphase-promoting complex/cyclosome degrons mediate the degradation of human Sgo1. J Biol Chem 2009; 284:1772–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Fu G, Hua S, Ward T, Ding X, Yang Y, Guo Z, Yao X. D-box is required for the degradation of human Shugoshin and chromosome alignment. Biochem Biophys Res Commun 2007; 357:672–8 [DOI] [PubMed] [Google Scholar]
  • 142.McGuinness BE, Hirota T, Kudo NR, Peters JM, Nasmyth K. Shugoshin prevents dissociation of cohesin from centromeres during mitosis in vertebrate cells. PLoS Biol 2005; 3:e86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wang X, Yang Y, Dai W. Differential subcellular localizations of two human Sgo1 isoforms: implications in regulation of sister chromatid cohesion and microtubule dynamics. Cell Cycle 2006; 5:635–40 [PubMed] [Google Scholar]
  • 144.Wang X, Yang Y, Duan Q, Jiang N, Huang Y, Darzynkiewicz Z, Dai W. sSgo1, a major splice variant of Sgo1, functions in centriole cohesion where it is regulated by Plk1. Dev Cell 2008; 14:331–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kitajima TS, Hauf S, Ohsugi M, Yamamoto T, Watanabe Y. Human Bub1 defines the persistent cohesion site along the mitotic chromosome by affecting Shugoshin localization. Curr Biol 2005; 15:353–9 [DOI] [PubMed] [Google Scholar]
  • 146.Tang Z, Sun Y, Harley SE, Zou H, Yu H. Human Bub1 protects centromeric sister-chromatid cohesion through Shugoshin during mitosis. Proc Natl Acad Sci USA 2004; 101:18012–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lee J, Kitajima TS, Tanno Y, Yoshida K, Morita T, Miyano T, Miyake M, Watanabe Y. Unified mode of centromeric protection by shugoshin in mammalian oocytes and somatic cells. Nat Cell Biol 2008; 10:42–52 [DOI] [PubMed] [Google Scholar]
  • 148.Kitajima TS, Sakuno T, Ishiguro K, Iemura S, Natsume T, Kawashima SA, Watanabe Y. Shugoshin collaborates with protein phosphatase 2A to protect cohesin. Nature 2006; 441:46–52 [DOI] [PubMed] [Google Scholar]
  • 149.Rao CV, Sanghera S, Zhang Y, Biddick L, Reddy A, Lightfoot S, Janakiram NB, Mohammed A, Dai W, Yamada HY. Systemic chromosome instability resulted in colonic transcriptomic changes in metabolic, proliferation, and stem cell regulators in Sgo1−/+ mice. Cancer Res 2016; 76:630–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wang LH, Yen CJ, Li TN, Elowe S, Wang WC, Wang LH. Sgo1 is a potential therapeutic target for hepatocellular carcinoma. Oncotarget 2015; 6:2023–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Yamada HY, Zhang Y, Reddy A, Mohammed A, Lightfoot S, Dai W, Rao CV. Tumor-promoting/progressing role of additional chromosome instability in hepatic carcinogenesis in Sgo1 (Shugoshin 1) haploinsufficient mice. Carcinogenesis 2015; 36:429–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Murakami-Tonami Y, Ikeda H, Yamagishi R, Inayoshi M, Inagaki S, Kishida S, Komata Y, Jan K, Takeuchi I, Kondo Y, Maeda T, Sekido Y, Murakami H, Kadomatsu K. SGO1 is involved in the DNA damage response in MYCN-amplified neuroblastoma cells. Sci Rep 2016; 6:31615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Yamada HY, Kumar G, Zhang Y, Rubin E, Lightfoot S, Dai W, Rao CV. Systemic chromosome instability in Shugoshin-1 mice resulted in compromised glutathione pathway, activation of Wnt signaling and defects in immune system in the lung. Oncogenesis 2016; 5:e256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Iwaizumi M, Shinmura K, Mori H, Yamada H, Suzuki M, Kitayama Y, Igarashi H, Nakamura T, Suzuki H, Watanabe Y, Hishida A, Ikuma M, Sugimura H. Human Sgo1 downregulation leads to chromosomal instability in colorectal cancer. Gut 2009; 58:249–60 [DOI] [PubMed] [Google Scholar]
  • 155.Mu J, Fan L, Liu D, Zhu D. Overexpression of shugoshin1 predicts a poor prognosis for prostate cancer and promotes metastasis by affecting epithelial-mesenchymal transition. Onco Targets Ther 2019; 12:1111–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yamada HY, Yao Y, Wang X, Zhang Y, Huang Y, Dai W, Rao CV. Haploinsufficiency of SGO1 results in deregulated centrosome dynamics, enhanced chromosomal instability and colon tumorigenesis. Cell Cycle 2012; 11:479–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Diaz-Martinez LA, Yu H. Sgo1 as a guardian of chromosome stability. Cell Cycle 2012; 11:650–1 [DOI] [PubMed] [Google Scholar]
  • 158.Shi Z, Taylor AW, Hu G, Gill T, Wittert GA. Rice intake, weight change and risk of the metabolic syndrome development among Chinese adults: the Jiangsu Nutrition Study (JIN). Asia Pac J Clin Nutr 2012; 21:35–43 [PubMed] [Google Scholar]
  • 159.Watanabe Y, Kitajima TS. Shugoshin protects cohesin complexes at centromeres. Philos Trans R Soc Lond B Biol Sci 2005; 360:515–21, discussion 21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Lee NR, Kim HS, Kim YS, Kwon MH, Choi KS, Lee CW. Regulation of the subcellular shuttling of Sgo1 between centromeres and chromosome arms by Aurora B-mediated phosphorylation. Biochem Biophys Res Commun 2014; 454:429–35 [DOI] [PubMed] [Google Scholar]
  • 161.Liu H, Rankin S, Yu H. Phosphorylation-enabled binding of SGO1-PP2A to cohesin protects sororin and centromeric cohesion during mitosis. Nat Cell Biol 2013; 15:40–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Pouwels J, Kukkonen AM, Lan W, Daum JR, Gorbsky GJ, Stukenberg T, Kallio MJ. Shugoshin 1 plays a central role in kinetochore assembly and is required for kinetochore targeting of Plk1. Cell Cycle 2007; 6:1579–85 [DOI] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES