IMR Press / CEOG / Volume 50 / Issue 12 / DOI: 10.31083/j.ceog5012272
Open Access Review
The Mechanism and Dynamic Regulation of Epithelial to Mesenchymal Transition in Ovarian Cancer
Show Less
1 Division of Gynecologic Oncology, Department of Obstetrics and Gynecology Prof. Dr. I G.N.G. Ngoerah Hospital, 80113 Denpasar, Bali, Indonesia
2 Faculty of Medicine, Udayana University, 80113 Denpasar, Bali, Indonesia
*Correspondence: sastra@unud.ac.id (I Gde Sastra Winata)
Clin. Exp. Obstet. Gynecol. 2023, 50(12), 272; https://doi.org/10.31083/j.ceog5012272
Submitted: 31 May 2023 | Revised: 17 July 2023 | Accepted: 3 August 2023 | Published: 26 December 2023
(This article belongs to the Special Issue Molecular and Cellular Advances in Gynecologic Cancer Research)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Objective: To understand the basic mechanism and dynamic regulation that underlies the epithelial-to-mesenchymal transition (EMT) in ovarian cancer (OC) cells. Mechanism: A literature review using evidences from several data bases (i.e., PubMed, EMBASE, Web of Science, Medline, Cochrane, Science Direct, and Google Scholar) were conducted to describe the basic mechanism and dynamic regulation of EMT in OC cells. Finding in Brief: EMT is a complex epigenetic reprogramming orchestrated by specific transcription factors (TFs) and multiple upstream activators and regulators, such as transforming growth factor-β (TGF-β), Wnt, Hedgehog, and Hippo signaling pathways. The net result of this cellular reprogramming is the acquisition of mesenchymal phenotypes with increased invasive and metastatic potential, stemness properties and chemoresistance. Recent studies have demonstrated that EMT activation is the result of dynamic and reciprocal interplay between OC cells and their tumor microenvironment (TME). Cellular or non-cellular component of TME, external factors related to TME such as hypoxia, oxidative stress, mechanical forces, as well as exposure to chemotherapy, all play significant role to EMT induction. Current understanding behind the mechanism of EMT induction in cancer cells have proposed the idea that EMT is not merely a binary process involving a complete conversion from epithelial to mesenchymal state, but rather a dynamic process that encompasses a range of hybrid states, a phenotype that has been referred to as “partial EMT”. Cells with partial EMT have been known to be more apoptosis-resistant and have more tumor-initiating potential as compared to those with complete EMT. Conclusions: Understanding the complex regulatory network that underlies EMT in OC cells is crucial in order to gain insight in developing novel and effective treatment strategies for OC.

Keywords
ovarian cancer
epithelial-to-mesenchymal transition
cellular reprogramming
1. Introduction

In 2020, ovarian cancer (OC) became the third most common gynecologic malignancy with a total of 313,959 new cases, and 207,252 new deaths recorded globally [1, 2]. Majority of OC are diagnosed in advance stage due to the ineffective screening, and its silent progression at early stage [3]. OC imposes a significant economic burden with annual average costs being significantly higher in advance stages than early stage OC [4]. Epithelial OC is the commonest histologic type, while only 10% belong to the non-epithelial type. The epithelial subtype has five major histologic types, i.e., serous, mucinous, endometroid, clear cell, and unspecified [5]. Surgical cytoreduction to attain no gross residual disease (R0) followed by adjuvant chemotherapy is the current standard treatment. Recently, maintenance therapies such as poly ADP-ribose polymerase (PARP) inhibitors, bevacizumab, and drugs targeting homologous recombination deficiency (HRD) are incorporated to prolong the survival [6]. However, despite advancement in the treatment of OC, recurrence rate remains high (>45%), and survival for those with advance disease remains dismal. The predictors for recurrence include the extent of carcinomatosis within the peritoneal cavity, the amount of residual disease after cytoreductive surgery, and cellular grade [7].

Epithelial-to-mesenchymal transition (EMT) program has gained popularity among researchers as the responsible cellular mechanism that confers OC cells with increased metastatic potential and drug resistance, thus predisposing to recurrence [8, 9, 10, 11]. EMT is a complex epigenetic reprogramming that results in reversible phenotype transition where cancer cells lose their epithelial phenotype and acquire mesenchymal phenotype [12]. The crucial orchestrator in EMT promotion is a well-known group of transcription factors (TFs), which includes SNAI1 (Snail1), SNAI2 (Snail2/Slug), Twist1/2, and zinc-finger E-box binding homeobox 1/2 (ZEB1/2) [13]. These TFs induce epigenetic silencing of epithelial markers (e.g., E-cadherin, Mucin 1 (MUC1), cytokeratin 18) while upregulating the expression of mesenchymal markers (e.g., N-cadherin, vimentin, fibronectin, matrix metalloproteinases (MMPs)) [14]. The upstream regulators of these EMT-transctipion factors (EMT-TFs) are multiple pathways which are mainly involved in embryonic development, such as transforming growth factor-β (TGF-β), Wnt, Hippo and Hedgehog. These pathways can be activated by numerous signals originating from the OC cells itself or their microenvironment.

This literature review will summarize the recent advances in the mechanisms underlying the complex regulatory network of EMT in OCs, with the focus on its key orchestrators such as EMT-TFs, signaling pathways, upstream activator, as well as the dynamics of its regulation.

2. The Key Players of EMT Regulation in OC

EMT is a complex, reversible cellular process orchestrated by multiple activators and signal transduction pathways (Figs. 1,2). The downstream effector in this process is a series of activated TFs (EMT-TFs) that acts as either activator or repressor of the targets gene, with the end result of phenotypic transition from epithelial cells to more invasive mesenchymal cells. Thus, epigenetic reprogramming is at the heart of EMT regulation [15]. By undergoing EMT, OC cells gain characteristics crucial for distant metastasis, resistance to apoptosis, and thus, recurrence after therapy [16].

Fig. 1.

Schematic illustration of complex regulatory interplays that underlie EMT. Tumor microenvironment (TME) and exposure to chemotherapy are believed to be the activators of EMT. The external factors then drive the expression of specific TFs, and activate signaling transduction pathways (TGF-β, Wnt, Hedgehog, Hippo). Expression of several non-coding RNA also interacts with EMT-TFs and signaling pathways. The net result of this complex regulatory network is the activation of mesenchymal phenotype with increased invasiveness, metastatic potential, stemness, and chemoresistance. EMT, epithelial-to-mesenchymal transition; TGF-β, transforming growth factor-β; EMT-TFs, EMT-transctipion factors; ADSC, adipose-derived stem cells; CAF, cancer associated fibroblasts; EPC, endothelial progenitor cells; ROS, reactive oxygen species; TAM-M2, tumor associated macrophage-M2; MMPs, matrix metalloproteinases; MUC1, Mucin1.

Fig. 2.

Signaling pathways that regulate the induction of EMT. Several signaling pathways play crucial role in EMT induction. All of these important pathways culminate in the activation of genes encoding the key TFs zinc-finger E-box binding homeobox (ZEB), Snail and Twist. Several miRNAs downregulate the expression of these TFs. SMAD 2/3/4, SMAD family member 2/3/4; GSK, glycogen synthase kinase; APC, adenomatous polyposis coli; AKT, AKT serine threonine kinase; JAK, janus kinase; STAT, signal transducer and activator of transcription; TCF, T cell factor; LEF, lymphoid enhancer factor; mTOR, mammalian target of rapamycin; NFκB, Nuclear factor kappa B; RTK, receptor tyrosine kinase; AXIN, Axin; PI3K, phosphatidylinositol 3-kinase.

2.1 Transcription Factors (TFs)
2.1.1 Snail/Slug

Snail and Slug, encoded by the snail family transcriptional repressor 1 (SNAI1) and 2 (SNAI2) gene, respectively, are transcriptional repressors that play important roles in regulating EMT. Snail and Slug activation results from several signaling pathways, such as receptor tyrosine kinases (RTKs), TGF-β, Notch, Wnt, tumor necrosis factor alpha (TNF-α), and bone morphogenetic proteins (BMPs) [17, 18, 19, 20, 21, 22, 23, 24]. Snail transcriptionally represses the expression of CDH1 gene, which encode E-cadherin. Snail also downregulates other epithelial markers, but upregulates the mesenchymal markers and transcriptional repressor ZEB1 [25, 26]. Meanwhile, Slug represses the expression of cell junction components: adherens junction (E-cadherin, β-catenin), tight junction (Occludin, Zona occludens-1 (ZO-1)) and desmosomes (Dsg2) [27]. In OC cell lines, Snail and Slug expressions were mutually exclusive, where Snail downregulates Slug expression [28]. Snail strongly represses epithelial splicing regulatory protein 1 (ESRP1) transcription in OC cells, which resultes in an isoform switching from epithelial spliced variant CD44v to mesenchymal spliced variant CD44s [29, 30]. Slug is involved in the ferroptosis regulation in OC cell line through binding to the promoter of Solute carrier family 7 member 11 (SLC7A11) [31]. Slug is also capable of transforming normal fibroblasts to a cancer associated fibroblast (CAF)-like state [32]. One study demonstrated that among several EMT-TFs (Twist, Slug, ZEB1/2), Slug showed the highest expression. Slug is believed to be the master regulator of EMT [33]. Downregulation of Snail and Slug expression greatly suppressed cell invasiveness and promoted cell apoptosis [31, 34, 35, 36, 37]. Snail inhibition also appears to reduce the expression of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2, chemokines that attract myeloid-derived suppressor cells (MDSCs) [38]. Higher Snail and Slug expression are associated with poorer survival of OC patients [39, 40, 41].

2.1.2 Zinc-Finger E-Box Binding Homeobox (ZEB)

ZEB is the zinc finger E-box binding homeobox family of TFs with its two members, ZEB1 and ZEB2. ZEB contain zinc-finger domain that allows binding at the enhancer boxes within the promoter region of target genes [42]. ZEB can interact with several TFs and cofactors, such as Smads (Suppressor of mothers against decapentaplegic), protein 300 (p300)/P300/CBP-associated factor (pCAF), Brahma-related gene-1 (BRG1), Nucleosome remodeling and deacetylation (NuRD) complex, and C-terminal binding protein (CtBP) [43, 44, 45]. The interaction determines ZEB role either as transcriptional activator or repressor of the target genes. ZEBs are crucial regulators of TGF-β/BMP signaling pathways [46, 47, 48]. ZEB1 synergizes with Smad-mediated transcriptional activation, while ZEB2 represses it [44]. Downregulation of ZEB2 expression in OC decreased the population of cancer stem cells (CSCs) and reduced the expression of Oct4 and Homeobox protein Nanog (Nanog) [49]. Furthermore, ZEB2 knockdown result in downregulation of N-cadherin and vimentin. ZEB2, but not ZEB1, may regulate the expression of membranous E-cadherin during EMT [50]. ZEB1 is associated with worse overall survival (OS) in patients with solid tumors [51].

2.1.3 Twist

Twist is a member of the basic helix-loop-helix (bHLH) family, and acts as either transcription activator or inhibitor [52]. The two Twist genes, i.e., Twist1 (Twist) and Twist2 (Dermo-1), have 90% similarity [53]. Upregulated Twist expression predicted shorter OS in OC patients [54, 55, 56]. Twist1 induce upregulation of Akt upon cisplatin treatment in OC cell lines, which drives resistance [57]. Twist2 also induces chemoresistance in OC cells through induction of Akt serine threonine kinase (Akt)/Glycogen synthase kinase-β (GSK-β) signaling pathways [58]. Twist2 cytoplasmic expression contributes to the maintenance of epithelial characteristics, while nuclear Twist2 induces expression of Vimentin which promotes metastasis. Upregulated expression of Twist2 reduces the expression of E-cadherin while increasing the expression of vimentin [59]. Twist2 also upregulates N-cadherin and β-catenin in human OC cells. Upregulation of β-catenin expression by Twist promotes the activation of Wnt/β-catenin pathway [60]. Under hypoxic conditions, Twist2 is also capable of activating phosphatidylinositol 3-kinase-Akt (PI3K-Akt) pathway, which in turn promotes the survival of cancer cells [61].

2.1.4 Novel TFs

Heat shock transcription factor 1 (HSF1) is a proteotoxic stress-responsive transcription factor that also contribute in EMT. Knockdown of HSF1 expression in OC cell lines impaired TGF-β-induced EMT [62]. Downregulation of HSF1 also results in reduced proliferative activity, and intensified apoptosis [63]. Copy number alteration of HSF1 gene in OC patients is associated with worse outcome [64]. HSF-1 also plays an important role in Akt-induced Slug upregulation [65]. SRY-related HMG-box genes (SOX), a family of pluripotent TFs may also play a role in EMT. Their role in inducing EMT have been demonstrated in several solid cancers [66, 67, 68, 69]. Knockdown of SOX2 induced downregulation of vimentin and upregulation of E-cadherin in OC cell lines [70]. Nanog, another important TFs commonly involved in embryonic development, also contributes to EMT. Nanog regulates EMT via 5 AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway [71]. Silencing of Nanog expression in OC cell lines restores expression of E-cadherin [72]. Inhibition of Nanog attenuates the proliferation, migration, and invasion of OC cell lines. Meanwhile, increased expression of Nanog enhances OC cell migration and invasion [73]. Downregulation of Nanog also results in reduced expression of vimentin, β-catenin, and Snail [74].

2.2 Signaling Pathways
2.2.1 Transforming Growth Factor Beta (TGF-β)

TGF-β is a highly pleiotropic cytokine that is capable of inducing EMT. In ovarian surface epithelium (OSE), TGF-β1 promotes tissue repair after ovulation by induction of EMT [75, 76]. TGF-β is overexpressed in the OC microenvironment. TGF-β1 upregulate the expression of Snail, Slug, Twist, ZEB1, and mesenchymal markers while downregulating E-cadherin [77, 78, 79, 80]. TGF-β inhibition results in reduced expression of Smad2, Smad3, Snail, and vimentin, and increased expression levels of Smad4 and E-cadherin, thus blocking the activation of EMT [81, 82]. TGF-β1 also induces overexpression of SOX2, OCT4a, Nanog, CD44, and CD117 in OC cells [83]. TGF-β also induces EMT in peritoneal mesothelial cells, which is associated with cancer-associated mesothelial cells [84]. In promoting EMT, TGF-β expression is mainly mediated by the activity of Smad protein [81]. TGF-β also capable in inducing the expression of long non-coding RNA (lncRNA) activated by TGF-β (lncRNA ATB), which acts as inhibitor to miR-204-3p [85].

2.2.2 Wingless/Integrated (Wnt)

Wingless/Integrated (Wnt) signaling pathway is one of the main orchestrator of EMT in OC. In promoting EMT, Wnt signaling mainly depends on the activity of β-catenin, which is a transcriptional activator to several EMT-TFs (Snail, ZEB, Twist) [34, 86, 87, 88, 89]. Suppression of the Wnt/β-catenin result in suppression of EMT in OC cells [86, 90, 91, 92]. The Wnt/β-catenin pathway also appears to be activated by Twist, which releases β-catenin from β-catenin/E-cadherin complex and thus, leads to nuclear β-catenin accumulation [60]. C-X-C motif chemokine ligand 14 (CXCL14) can induce EMT in OC cells via activation of the Wnt/β-catenin pathway [93]. The bioactive lipid lysophosphatidic acid (LPA) promotes the nuclear translocation of β-catenin and upregulates the expression of Wnt/β-catenin target genes [94]. The non-canonical Wnt also contributes to EMT via signal transducer and activator of transcription 3 (Stat3) expression [95, 96, 97]. Upregulation of Wnt5a, the non-canonical Wnt ligand, increases the transcriptional activation of Snail and induces EMT via protein kinase Cα (PKCα) [98, 99]. Wnt5a, the activator of non-canonical Wnt pathway, is also capable of inducing EMT in OSE cells [100]. Downregulation of Wnt5a significantly reduces the expression Smad2/3 and Yes-associated protein 1 (YAP1) expression [101].

2.2.3 Hedgehog

Hedgehog (Hh) signaling cascade culminate in a balance between activator or repressor forms of transcription factor Glioma-associated oncogene TFs (Gli) with PTCH1, PTCH2, and GLI1 being the main target genes. Hedgehog (Hh) ligand, Patched (Ptch), and Smoothened (Smo) are proteins involved in the activation of Hh signaling cascade [102]. Hh signaling plays an important role in the regulation of invasiveness, chemoresistance, as well as maintenance of CSCs characteristics [103, 104, 105, 106, 107, 108]. A crosstalk between Sonic Hh-Gli1 signals and PI3K-Akt pathway regulates EMT induction in OC cells [109]. Inhibition of Hh signaling results in inhibition of EMT [110, 111]. Gli1 also regulates the expression of Snail1, Slug, and Twist. Gli1 and Gli2 repress the expression of E-Cadherin [112].

2.2.4 Hippo

The Hippo pathway is a tumor suppressive pathway involved in regulating tissue growth, and their component comprises a pair of related serine/threonine kinases, macrophage stimulating 1 and 2 (MST1 and MST2), large tumor suppressor kinase 1 and 2 (LATS1 and LATS2), and lastly Salvador family WW domain containing protein 1 (SAV1), and Mps one binder 1 (MOB1A and MOB1B) [113]. This pathway downregulates the activity of YAP/TAZ. Following Hippo inactivation, YAP and transcriptional enhanced associate domain (TEAD) form complex within the nucleus to direct transcription of target genes. EMT-TFs are capable of complex formation with YAP/TEAD, which in turn upregulate the expression of YAP target genes in inducing EMT in OC cells [114, 115, 116]. High YAP and TEAD expression is associated with OC progression. Hippo pathway is also involved in regulating chemoresistance in OC cells [117, 118, 119, 120, 121]. Hippo signaling interacts with other EMT-inducing pathways, such as TGF-β and Wnt [101].

2.3 Non-Coding RNA
2.3.1 Micro-RNA

MicroRNA (miRNA) is a single-stranded non-coding RNA that acts as antisense RNA to downregulate expression of the target genes at the post-transcriptional level. Several miRNAs are involved in the regulation of EMT in OC cells. miR-200 family of miRNAs is a well-known EMT suppressor. Low level of miR-200 expression is demonstrated in normal OSE, but the increased expression is present in OC [122]. Overexpression of miR-205 and/or miR-200 family result in downregulation of ZEB1 transcription factor and Wnt5a [123]. miR-200c upregulation induced downregulation of ZEB1 and vimentin, and upregulation of E-cadherin [124]. In OC cells line, miR-200c overexpression decrease Snail, increase E-cadherin, and significantly reduce the invasiveness and tumorigenic potency of OC cell lines [125]. miR-203 expression can attenuate TGF-β pathway in OC cells [126]. Let-7 miRNA family is another EMT suppressor. The overexpression of Let-7g in OC cell lines reduce vimentin, Snail, and Slug expression [127]. miR-16 promotes the inactivation of the Wnt/β-catenin signaling pathway, thus inhibiting EMT. miR-16 upregulates the expression of Cadherin-1 and downregulates the expression of Snail, Slug, Twist 1, vimentin, and Cadherin-2 in OC cell lines [128]. miR-30d represses EMT by targeting Snail [129]. Expression of miR-30d reversed the TGF-β1-induced EMT phenotypes in OC cell lines. Expression of miR-186 in OC cells downregulates the expression of Twist1 and subsequent reversal of EMT [130]. miR‑141 overexpression upregulates E‑cadherin, and decreases cell invasiveness in OC cell line [131]. However, several miRNAs also act as an EMT inducer. miR-1301 upregulates Snail, Slug, and N-cadherin expression, while downregulating E-cadherin [132]. The miR-150-5p also plays important roles in EMT regulation [133]. miR-27a promotes EMT via activation of Wnt/β-catenin signaling pathway [134].

2.3.2 Long Non-Coding RNA

Long non-coding RNA (lncRNA) is non-protein coding RNA with length longer than 200 nucleotides that regulates target gene expression at both transcriptional and post-transcriptional level [135]. lncRNA are capable of direct binding to DNA or RNA to affect the transcription process. lncRNA H19 expression in the OC cells promotes migration and EMT-related activity [136]. Silencing of lncRNA in colon cancer associated transcript 2 (CCAT2) results in EMT inhibition. Knockdown of lncRNA CCAT2 also inhibits the expression of β-catenin and the activity of Wnt signaling pathway [137]. TGF-β treatment in OC cell lines results in the upregulation of lncRNA H19 and downregulation of miR-370-3p. H19 overexpression or miR-370-3p knockdown are capable of promoting TGF-β-induced EMT [138]. lncRNA-ATB downregulation results in EMT suppression [139]. By regulating the expression of LATS2, lncRNA ASAP1 Intronic Transcript 1 (ASAP1-IT1) induces downregulation of YAP1 expression and thus, preventing EMT in OC cells [140].

3. Activation of EMT Program in OC

EMT activation is the result of dynamic and reciprocal interplay between OC cells and their tumor microenvironment (TME). Cellular or non-cellular component of TME can act either as activator or inhibitor to certain signaling transduction pathways associated with EMT. On the other hand, OC cells can induce differentiation of cellular component of TME into certain phenotypes that favor metastatic potential of drug resistance of OC cells. These complex regulations are also affected by external factors related to TME, such as hypoxia, oxidative stress, as well as mechanical forces.

3.1 The Role of Tumor Microenvironment
3.1.1 Cellular Components

TME is the niche or environment in which the cancer cells closely interact with the host stroma, including cellular and non-cellular component of TME. The cellular components include immune cells, endothelial cells and fibroblast, while the non-cellular components include extracellular matrix and cellular metabolites. The cellular components play a more dominant role in promoting EMT. Cancer associated fibroblasts (CAF) are one of the key cellular components of TME that play important role in EMT induction. CAF are more proliferative and have higher metabolic states as compared to normal fibroblasts [141]. CAF-derived exosomes are rich in TGF-β as compared to normal fibroblasts, and are capable of inducing EMT in OC cell lines [142]. CAF highly secretes interleukin 6 (IL‑6) and promotes TGF-β-mediated EMT via the Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (STAT3) pathway [143]. CAFs also secretes periostin, which functions as a ligand for integrin αvβ3. Periostin is also capable of inducing EMT, mediated by TGF-β in OC cells [144]. The increased expression of fibroblast growth factor-1 (FGF-1) in CAFs induces the phosphorylation of fibroblast growth factor receptor-4 (FGFR-4) in OC cell line, which then induces the activation of mitogen-activated protein kinase (MAPK)/Extracellular signal-regulated kinase (ERK) pathway and EMT-associated gene Snail1 and matrix metalloproteinase 3 (MMP3) expression [145]. Stanniocalcin-1 expressed by CAF, is capable of upregulating the expression of fibronectin, vimentin, and Slug [146].

Tumor associated macrophage (TAM) plays a critical role in the interaction between TME and OC cells. TAMs are capable of differentiating into two disctinct phenotypes: M1, which has pro-inflammatory with anti-tumor activity, and M2, which has anti-inflammatory with pro-tumor activity. The path of TAMs is determined by the local TME [147]. In a mouse model of OC, TAM require ZEB1 expression to activate the tumor-promoting functions [148]. OC cells secretes macrophage colony-stimulating factor (M-CSF) to drive the differentiation of M2-TAM [149]. M2-TAMs are capable of inducing EMT of OC cells by releasing chemokine (C-C motif) ligand 18 (CCL18). On the other hand, CCL18 induces M-CSF transcription in OC cells through the activity ZEB1 transcription factor. Thus, a CCL18-ZEB1-M-CSF interacting loop exists between OC cells and TAMs that regulate the tumor progression and metastasis through EMT [149]. OC-derived exosomes containing miR-222-3p and miR‑940 are also capable in inducing TAM polarization into the M2 phenotype [150, 151].

Adipose-derived stem cells (ADSCs) are mesenchymal stem cells obtained from adipose tissues. ADSCs have been shown to affect the proteomic profile of OC cells via paracrine mechanism in favour of OC progression [152]. Secretion of TGF-β from ADSC result in activation of the TGF-β pathway in OC cells and subsequent EMT activation [153]. The relationship between ADSC and OC cells seems to be reciprocal. OC cells are capable of inducing the expression of CAF markers in ADSC, including alpha-smooth muscle actin (α-SMA) and fibroblast activation protein, via the TGF-β1 signaling pathway [154, 155]. The addition of ADSCs into the medium of OC culture significant increase of the paired box 8 (PAX8) level in OC cells [156]. Overexpression of PAX8 lead to upregulation of Snail, Twist and Zeb2 [157].

Endothelial progenitor cells (EPCs), the bone marrow-derived stem cells, play a significant role in tumor angiogenesis and growth. EPCs are recruited into the neovascular bed of the tumor in response to certain signals or cytokines secreted by tumor cells [158, 159, 160]. EPCs are able to invade into the OC cell clusters, whereas normal human microvascular endothelial cells are not capable of invading OC cell clusters [161]. Circulating levels of EPCs are significantly increased in OC patients and correlate with tumor stage and residual tumor size [162]. OC cells cultured in EPC-conditioned media (EPC-CM) demonstrate an increase in TGF-β. EPC-CM also induce loss of cell junctions, reduced expression of E-cadherin, increased expression of N-cadherin, and development of a fibroblastic phenotype in OC cells, which are the consistent feature of EMT [163].

3.1.2 Non-Cellular Components

The non-cellular components of TME also participate in promoting EMT. Collagen I enhance OC cells motility and invasiveness through the increased expression of MMPs and α5β1 integrin. Collagen I matrix upregulate the expression of N-cadherin, vimentin, fibronectin, and transcriptional factors Snail and Slug [164]. Collagen I also upregulates the activity of TGF-β1/Smad4 and Wnt5b/β-catenin signaling cascade [165]. IL-6 treatment downregulates the expression of epithelial markers, and upregulates the expression of mesenchymal markers in OC cell line. Overexpression of IL-6 in OC cells significantly increases the expression of MMP-2 and MMP-9 and thus, enhancing their migration ability [166].

Hypoxia is one of the most important non-cellular factors in inducing EMT. Hypoxia influences cellular processes such as angiogenesis, acquisition of stem cell-like features, chemoresistance, as well as EMT [167, 168, 169, 170]. Hypoxia is the characteristic of the peritoneal environment. Hypoxia induces the stabilization of hypoxia-inducible factor-1α (HIF-1α), which then translocate into the nucleus, to bind to HIF-1β and forming HIF-1 heterodimer. HIF-1 heterodimer acts as transcription factor targeting genes with the hypoxic responsive elements (HRE). Under hypoxic conditions, OC cells presents morphological changes consistent with EMT [171]. HIF-1 expression in OC stem cells results in the induction of Twist1 and E12 expression [172]. Downregulation of HIF-1α expression leads to upregulation of E-cadherin and downregulation of the vimentin [173]. Hypoxia downregulates the expression of miR-210, the EMT repressor [174]. Hypoxia also upregulates the expression of C-X3-C motif chemokine receptor (CX3CR), increasing the chemotactic response to C-X3-C motif chemokine ligand 1 (CX3CL1), and thus leading to tumor progression and metastasis [175]. Hypoxia also upregulates the expression of signal transducer and activator of transcription 4 (STAT4), which contributes to the regulation of EMT [176]. Hypoxic stress downregulated the expression of Sirtuin (silent mating type information regulation 2 homolog) 1 (SIRT1), a negative regulator of HIF-1α [177].

Reactive oxygen species (ROS), such as hydroxyl free radicals, superoxide, and hydrogen peroxide can accumulate within the TME due to active metabolic patterns of OC cells and tumor stromal cells. ROS accumulation can lead to lipid peroxidation, antioxidants deprivation, and ultimately programmed cell death which depend on iron, also known as ferroptosis. However, OC cells develop several mechanisms that confer resistance to ferroptosis [178]. ROS are involved in the regulation of EMT. ROS accumulation induced the increased expression of HIF-1α and subsequent transcriptional induction of lysyl oxidase (LOX), which then decreases the expression of E-cadherin [179]. ROS scavenging negatively affect migration and invasion of OC cells through reversing EMT [180].

OC cells endure several mechanical forces from their TME. The presence of ascites and interstitial fluid confer the shearing force to the OC cells, while tumor expansion against the extracellular matrix and TME exert tension on the tumor periphery. Furthermore, tumor expansion and the hydrostatic pressure of ascites also exert internal compression forces to the OC cells [181]. Those forces give impact into biochemical regulation and signaling pathways of OC cells, including the regulation of EMT. Oscillatory tension significantly decreases the expression of E-cadherin while increasing the expression level of Snail [182]. Tissue stiffness, recapitulated by substrate stiffness in vitro, promotes OC cells proliferation and nuclear translocation of the oncogene YAP. Substrate softening has been demonstrated to promote changes consistent with EMT [183, 184].

3.2 The Role of Chemotherapy

Results from in vitro studies have demonstrated that chemoresistant OC cells express markers of EMT [8, 185, 186, 187, 188, 189, 190]. On the other hand, exposure to chemotherapy has been demonstrated to induce EMT in OC cells. Cisplatin induced EMT-associated morphological changes in OC cells [191, 192, 193]. Receptor cells co-cultured with carboplatin- or etoposide-treated feeder cells in Transwell co-culture system exhibited increased expression of EMT markers vimentin and Snail. The altered microenvironment of either carboplatin- or etoposide-16-treated feeder OC cells also significantly increased the migration of the OC cells [194]. OC cells treated with carboplatin exhibited phenotypic changes consistent with EMT [195]. Clinical studies in OC patients revealed that treatment with platinum-based chemotherapy increased the proportion of EMT-like circulating tumor cells (CTC), accompanied by the “de novo” emergence of PI3Kα+/Twist+ EMT-like CTCs [196]. Continuous exposure to increasing doses of paclitaxel lead to the establishment of OC cell lines that are resistant to paclitaxel and exhibit phenotypic changes consistent with EMT [197]. However, the EMT-inducing effect of chemotherapy, whether it is direct or indirect, remains unclear. The proposed mechanisms are that chemotherapy may directly trigger intracellular signaling, such as orchestrated cellular defense response against platinum toxicity. On the other hand, while killing the tumor cells, chemotherapy indirectly influences the EMT of the remaining cancer cells. Chemoterapy is also believed to induce oxidative stress that is capable of inducing EMT.

4. The Dynamics of EMT Regulation

The traditional concept viewed EMT as a binary process in which the phenotype of cancer cells can change completely into either epithelial or mesenchymal cells. However, a newer concept of “partial EMT” has been proposed, in which cancer cells can undergo phenotypic changes with both epithelial and mesenchymal phenotype [198, 199]. Tumours in a partial EMT state exhibit low expression of EMT-TFs, and co-express both epithelial and mesenchymal genes. A recent study in pancreatic ductal adenocarcinoma cells reported that partial EMT results from different mechanisms underlying complete EMT. Cancer cells lose their epithelial phenotype through an alternative post-translational process of protein relocalizations, which lead to “partial EMT”. In that study, E-cadherin protein was found to be confined to intracellular foci in delaminated cells exhibiting a mesenchymal morphology. Furthermore, cancer cells that exhibit partial EMT, migrate and form circulating tumor cell clusters, rather than disseminate as single cell as in “complete EMT” [200]. However, it remains unclear whether partial EMT represents an intermediate stage where cancer cells are in a paused transitional state within the mesenchymal differentiation continuum, or it is a final fate of the cancer cells. Interestingly, one study support the later hypothesis, in which they found no evidence that complete and partial EMT co-exist within the same tumor [50]. This finding implicate that the tendency of cancer cells to use either a complete or partial EMT program is a specific and stable feature of an individual tumor. Cells with partial EMT have been known to be more resistant to apoptosis and have greater tumor-initiating potential, as compared to those with complete EMT. EMT interconversions are also dynamically regulated during the development and progression of ovarian tumors [50, 201].

Transcription Factor Balance in Partial EMT

A review by Jolly et al. [198] stated that the core regulatory network for EMT or MET (Mesenchymal-to-Epithelial Transition) acts as a “three-way”, which give rise to three distinct phenotypes, i.e., epithelial (E), mesenchymal (M), and hybrid, or partial EMT (pEMT). The core regulatory network depends on two mutually inhibitory loops, i.e., miR-34/SNAIL and miR-200/ZEB. E phenotype is defined as high miR-200/miR-34, low ZEB/SNAIL; M phenotype is defined as low miR-200/miR-34, high ZEB/SNAIL; and partial EMT is defined as low miR-34/ZEB, high SNAIL/miR-200. Another theory proposed that miR-200/ZEB, with input from SNAIL, behaves as a three-way switch allowing for the existence of three phenotypes, i.e., E (high miR-200, low ZEB), M (low miR-200, high ZEB), and E/M or partial EMT (medium miR-200, medium ZEB). Therefore, ZEB activation is a necessary requirement for the acquisition of a complete EMT. However, results from experimental studies observing partial EMT appear to be more consistent with medium miR-200, medium ZEB theory. One study demonstrated that difference in expression and subcellular localization of transcription factor 21 (Tcf21) and Slug (Tcf21/Slug balance) is associated with phenotypic plasticity [202]. Downregulation of Slug by Tcf21 is known to maintain the epithelial properties of high grade serous carcinoma (HGSC). The study identified the association of Tcf21/Slug balance with additional intermediate phenotypic states (i.e., iE or iM), supporting the proposed hypothesis of a multistep EMT program [202]. Tcf21 high-Slug low expression was identified in E phenotype, Tcf21 low-Slug high expression was identified in M phenotype, and Tcf21 moderate-Slug moderate expression (E-M or pEMT) was identified as stable phenotypes. Intermediate E or M represents a metastable state associated with phenotypic switches. Intermediate E (iE) expressed epithelial markers, Snail, Twist1, but lacked Tcf21, while the tumor group with low EMT-TF expression was identified as intermediate M (iM). The dynamic regulation of EMT, which encompasses a range of phenotypic plasticity, is summarized in Fig. 3.

Fig. 3.

Schematic illustration of dynamic regulation underlying phenotypic plasticity in EMT. A newer concept of EMT introduces the concept of partial EMT in which cancer cells exhibit both mesenchymal and epithelial characteristics. This phenotypic plasticity are determined by the balance of expression from certain TFs and their inhibitors (miRNA), such as the balance between miR-34/SNAIL and miR-200/ZEB. The expression and subcellular location of Tcf21 and Slug also influence the phenotype of cancer cells. Cancer cells undergoing partial or hybrid EMT tend to be more invasive, more resistant to apoptosis, and have greater tumor initiating potential.

5. Conclusions

Epithelial-to-mesenchymal transition or EMT is a form of epigenetic cellular reprogramming governed by complex regulatory networks that confers OC cells with increased invasiveness and drug resistance. EMT is orchestrated by multiple TFs, upstream activators, and regulators that result in the acquisition of mesenchymal phenotypes with increased metastatic potential, stemness properties, and chemoresistance. EMT activation is the result of dynamic and reciprocal interplay between OC cells and their tumor microenvironment. EMT is a dynamic process of phenotypic plasticity, which encompasses a range of hybrid states. Understanding this complex regulatory network is crucial in order to gain insight in developing novel and effective treatment strategies for OC.

Abbreviations

ADSC, Adipocyte-derived stem cell; AKT, AKT serine threonine kinase; AMPK, 5 AMP-activated protein kinase; ASAP1-IT1, ASAP1 Intronic Transcript 1; ATB, Activated by TGF-β; bHLH, basic helix-loop-helix; BMP, Bone morphogenetic protein; BRG1, Brahma-related gene-1; CAF, Cancer associated fibroblast; CCAT2, Colon cancer associated transcript 2; CCL18, C-C motif chemokine ligand 18; CD117, Cluster of differentiation 117; CD44s, Cluster of differentiation standard isoform; CD44v, Cluster of differentiation 44 variant isoform; CDH1, Cadherin 1; CSCs, Cancer stem cells; CtBP, C-terminal binding protein; CTC, Circulating tumor cells; CX3CL, C-X3-C motif chemokine ligand ; CX3CR, C-X3-C motif chemokine receptor; CXCL1, C-X-C motif chemokine ligand 1; CXCL14, C-X-C motif chemokine ligand 14; CXCL2, C-X-C motif chemokine ligand 2; EMT, Epithelial-to-mesenchymal transition; EPC-CM, Endothelial progenitor cell-conditioned media; ERK, Extracellular signal-regulated kinase; ESRP1, Epithelial splicing regulatory protein 1; FGF-1, Fibroblast growth factor-1; GLI, Glioma-associated oncogene transcription factors; GSK-β, Glycogen synthase kinase-β; HIF-1α, Hypoxia-inducible factor 1-alpha; HRD, Homologous recombination deficiency; HRE, Hypoxia responsive element; HSF1, Heat shock transcription factor 1; iE, Intermediate epithelial; IL, Interleukin; iM, Intermediate mesenchymal; JAK, Janus kinase; LATS, Large tumor suppressor kinase 1; lncRNA, Long non coding ribonucleic acid; LOX, Lysyl oxidase; LPA, Lysophosphatidic acid; M-CSF, Monocyte-colony stimulating factor; MAPK, Mitogen-activated protein kinase; MDSC, Myeloid-derived suppressor cells; miRNA, Micro ribonucleic acid; MMP, Matrix metalloproteinase; MOB1, Mps one binder 1; MST, Macrophage stimulating; mTOR, Mammalian target of rapamycin; MUC1, Mucin 1; NANOG, Homeobox protein Nanog; NFκB, Nuclear factor kappa B; NuRD, Nucleosome remodeling and deacetylation; OC, Ovarian cancer; OCT4, Octamer-binding transcription factor 4; OSE, Ovarian surface epithelium; p300, Protein 300; PARP, Poly (ADP-ribose) polymerase ; PAX, Paired box; pCAF, P300/CBP-associated factor; pEMT, Partial epithelial-to-mesenchymal transition; PI3K, Phosphatidylinositol 3-kinase; PKCα, Protein kinase C alpha; PTCH, Protein pathced homolog; R0, No gross residual disease; ROS, Reactive oxygen species; RTK, Receptor tyrosine kinase; SAV, Salvador family WW domain containing protein 1; SIRT1, Sirtuin (silent mating type information regulation 2 homolog) 1; SLC7A11, Solute carrier family 7 member 11; SMAD, Suppressor of mothers against decapentaplegic; SMO, Smoothened; SNAI1, Snail family transcriptional repressor 1; SNAI2, Snail family transcriptional repressor 1; SOX, SRY-related HMG-box genes; STAT3, Signal transducer and activator of transcription 3; TAM, Tumour associated macrophage; TAM-M2, Tumour associated macrophage-M2 phenotype; Tcf21, Transcription factor 21; TEAD, Transcriptional enhanced associate domain; TF, Transcription factor; TGF-β, Transforming growth factor-β; TME, Tumour microenvironment; TNF-α, Tumor necrosis factor alpha; TWIST, Twist-related protein; WNT, Wingless/Integrated; YAP, Yes-associated protein; ZEB, Zinc-finger E-box binding homeobox; ZO-1, Zona occludens-1; α-SMA, Alpha-Smooth muscle actin.

Author Contributions

PKAP performed conception and design, acquisition of data, analysis and interpretation of data and drafting the manuscript. INBM and INGB performed conception and design, analysis and interpretation of data, and reviewing the manuscript. IGSW and KYS performed acquisition of data, analysis and interpretation of data and drafting the manuscript. KS performed conception and design, analysis and interpretation of data, reviewing and final approval of published version. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We would like to express our gratitude to all those who helped us during the writing of this manuscript. Thanks to all the peer reviewers for their opinions and suggestions.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: a Cancer Journal for Clinicians. 2021; 71: 209–249.
[2]
Huang J, Chan WC, Ngai CH, Lok V, Zhang L, Lucero-Prisno DE 3rd, et al. Worldwide Burden, Risk Factors, and Temporal Trends of Ovarian Cancer: A Global Study. Cancers. 2022; 14: 2230.
[3]
Narod S. Can advanced-stage ovarian cancer be cured? Nature Reviews. Clinical Oncology. 2016; 13: 255–261.
[4]
Delgado-Ortega L, González-Domínguez A, Borrás JM, Oliva-Moreno J, González-Haba E, Menjón S, et al. The economic burden of disease of epithelial ovarian cancer in Spain: the OvarCost study. The European Journal of Health Economics: HEPAC: Health Economics in Prevention and Care. 2019; 20: 135–147.
[5]
Gaona-Luviano P, Medina-Gaona LA, Magaña-Pérez K. Epidemiology of ovarian cancer. Chinese Clinical Oncology. 2020; 9: 47.
[6]
Kuroki L, Guntupalli SR. Treatment of epithelial ovarian cancer. BMJ (Clinical Research Ed.). 2020; 371: m3773.
[7]
Ntatsis K, Papantoni E, Kyziridis D, Kalakonas A, Hristakis C, Tzavara C, et al. Ovarian cancer: 20-year experience with cytoreductive surgery and perioperative intraperitoneal chemotherapy. Journal of B.U.ON.: Official Journal of the Balkan Union of Oncology. 2021; 26: 1754–1761.
[8]
Wu X, Zhao J, Ruan Y, Sun L, Xu C, Jiang H. Sialyltransferase ST3GAL1 promotes cell migration, invasion, and TGF-β1-induced EMT and confers paclitaxel resistance in ovarian cancer. Cell Death & Disease. 2018; 9: 1102.
[9]
Wang J, Liu L. MiR-149-3p promotes the cisplatin resistance and EMT in ovarian cancer through downregulating TIMP2 and CDKN1A. Journal of Ovarian Research. 2021; 14: 165.
[10]
Liu C, Yu M, Li Y, Wang H, Xu C, Zhang X, et al. Lidocaine inhibits the metastatic potential of ovarian cancer by blocking Na_V 1.5-mediated EMT and FAK/Paxillin signaling pathway. Cancer Medicine. 2021; 10: 337–349.
[11]
Wang J, Liu L. MiR-149-3p promotes the cisplatin resistance and EMT in ovarian cancer through downregulating TIMP2 and CDKN1A. Journal of Ovarian Research. 2021; 14: 165.
[12]
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial-mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm. 2022; 3: e144.
[13]
Goossens S, Vandamme N, Van Vlierberghe P, Berx G. EMT transcription factors in cancer development re-evaluated: Beyond EMT and MET. Biochimica et Biophysica Acta. Reviews on Cancer. 2017; 1868: 584–591.
[14]
Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, et al. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells. 2019; 8: 1118.
[15]
Nowak E, Bednarek I. Aspects of the Epigenetic Regulation of EMT Related to Cancer Metastasis. Cells. 2021; 10: 3435.
[16]
Zhang Y, Huang W, Chen D, Zhao Y, Sun F, Wang Z, et al. Identification of a Recurrence Gene Signature for Ovarian Cancer Prognosis by Integrating Single-Cell RNA Sequencing and Bulk Expression Datasets. Frontiers in Genetics. 2022; 13: 823082.
[17]
Li D, Li L, Yang W, Chen L, Chen X, Wang Q, et al. Prognostic values of SNAI family members in breast cancer patients. Annals of Translational Medicine. 2020; 8: 922.
[18]
Djediai S, Gonzalez Suarez N, El Cheikh-Hussein L, Rodriguez Torres S, Gresseau L, Dhayne S, et al. MT1-MMP Cooperates with TGF-β Receptor-Mediated Signaling to Trigger SNAIL and Induce Epithelial-to-Mesenchymal-like Transition in U87 Glioblastoma Cells. International Journal of Molecular Sciences. 2021; 22: 13006.
[19]
Matsuno Y, Coelho AL, Jarai G, Westwick J, Hogaboam CM. Notch signaling mediates TGF-β1-induced epithelial-mesenchymal transition through the induction of Snai1. The International Journal of Biochemistry & Cell Biology. 2012; 44: 776–789.
[20]
Horvay K, Casagranda F, Gany A, Hime GR, Abud HE. Wnt signaling regulates Snai1 expression and cellular localization in the mouse intestinal epithelial stem cell niche. Stem Cells and Development. 2011; 20: 737–745.
[21]
Wang H, Wang HS, Zhou BH, Li CL, Zhang F, Wang XF, et al. Epithelial-mesenchymal transition (EMT) induced by TNF-α requires AKT/GSK-3β-mediated stabilization of snail in colorectal cancer. PloS One. 2013; 8: e56664.
[22]
Chandran Latha K, Sreekumar A, Beena V, S S BR, Lakkappa RB, Kalyani R, et al. Shear Stress Alterations Activate BMP4/pSMAD5 Signaling and Induce Endothelial Mesenchymal Transition in Varicose Veins. Cells. 2021; 10: 3563.
[23]
Slabáková E, Pernicová Z, Slavíčková E, Staršíchová A, Kozubík A, Souček K. TGF-β1-induced EMT of non-transformed prostate hyperplasia cells is characterized by early induction of SNAI2/Slug. The Prostate. 2011; 71: 1332–1343.
[24]
Sakai D, Tanaka Y, Endo Y, Osumi N, Okamoto H, Wakamatsu Y. Regulation of Slug transcription in embryonic ectoderm by beta-catenin-Lef/Tcf and BMP-Smad signaling. Development, Growth & Differentiation. 2005; 47: 471–482.
[25]
Wang J, He H, Jiang Q, Wang Y, Jia S. CBX6 promotes HCC metastasis via transcription factors Snail/Zeb1-mediated EMT mechanism. OncoTargets and Therapy. 2020; 13: 12489–12500.
[26]
Lu Z, Yuan S, Ruan L, Tu Z, Liu H. Partitioning defective 6 homolog alpha (PARD6A) promotes epithelial-mesenchymal transition via integrin β1-ILK-SNAIL1 pathway in ovarian cancer. Cell Death & Disease. 2022; 13: 304.
[27]
Kurrey NK, K A, Bapat SA. Snail and Slug are major determinants of ovarian cancer invasiveness at the transcription level. Gynecologic Oncology. 2005; 97: 155–165.
[28]
Sundararajan V, Tan M, Tan TZ, Ye J, Thiery JP, Huang RYJ. SNAI1 recruits HDAC1 to suppress SNAI2 transcription during epithelial to mesenchymal transition. Scientific Reports. 2019; 9: 8295.
[29]
Chen L, Yao Y, Sun L, Zhou J, Miao M, Luo S, et al. Snail Driving Alternative Splicing of CD44 by ESRP1 Enhances Invasion and Migration in Epithelial Ovarian Cancer. Cellular Physiology and Biochemistry: International Journal of Experimental Cellular Physiology, Biochemistry, and Pharmacology. 2017; 43: 2489–2504.
[30]
Bhattacharya R, Mitra T, Ray Chaudhuri S, Roy SS. Mesenchymal splice isoform of CD44 (CD44s) promotes EMT/invasion and imparts stem-like properties to ovarian cancer cells. Journal of Cellular Biochemistry. 2018; 119: 3373–3383.
[31]
Jin Y, Chen L, Li L, Huang G, Huang H, Tang C. SNAI2 promotes the development of ovarian cancer through regulating ferroptosis. Bioengineered. 2022; 13: 6451–6463.
[32]
Yoon H, Tang CM, Banerjee S, Yebra M, Noh S, Burgoyne AM, et al. Cancer-associated fibroblast secretion of PDGFC promotes gastrointestinal stromal tumor growth and metastasis. Oncogene. 2021; 40: 1957–1973.
[33]
Ghulam J, Stuerken C, Wicklein D, Pries R, Wollenberg B, Schumacher U. Immunohistochemical Analysis of Transcription Factors and Markers of Epithelial-Mesenchymal Transition (EMT) in Human Tumors. Anticancer Research. 2019; 39: 5437–5448.
[34]
Hojo N, Huisken AL, Wang H, Chirshev E, Kim NS, Nguyen SM, et al. Snail knockdown reverses stemness and inhibits tumour growth in ovarian cancer. Scientific Reports. 2018; 8: 8704.
[35]
Wang Q, López-Ozuna VM, Baloch T, Bithras J, Amin O, Kessous R, et al. Biguanides in combination with olaparib limits tumorigenesis of drug-resistant ovarian cancer cells through inhibition of Snail. Cancer Medicine. 2020; 9: 1307–1320.
[36]
Sun Y, Hu L, Zheng H, Bagnoli M, Guo Y, Rupaimoole R, et al. MiR-506 inhibits multiple targets in the epithelial-to-mesenchymal transition network and is associated with good prognosis in epithelial ovarian cancer. The Journal of Pathology. 2015; 235: 25–36.
[37]
Jiang H, Lin X, Liu Y, Gong W, Ma X, Yu Y, et al. Transformation of epithelial ovarian cancer stemlike cells into mesenchymal lineage via EMT results in cellular heterogeneity and supports tumor engraftment. Molecular Medicine (Cambridge, Mass.). 2012; 18: 1197–1208.
[38]
Taki M, Abiko K, Baba T, Hamanishi J, Yamaguchi K, Murakami R, et al. Snail promotes ovarian cancer progression by recruiting myeloid-derived suppressor cells via CXCR2 ligand upregulation. Nature Communications. 2018; 9: 1685.
[39]
Li J, Li S, Chen R, Lu X. Increased risk of poor survival in ovarian cancer patients with high expression of SNAI2 and lymphovascular space invasion. Oncotarget. 2017; 8: 9672–9685.
[40]
Blechschmidt K, Sassen S, Schmalfeldt B, Schuster T, Höfler H, Becker KF. The E-cadherin repressor Snail is associated with lower overall survival of ovarian cancer patients. British Journal of Cancer. 2008; 98: 489–495.
[41]
Hipp S, Berg D, Ergin B, Schuster T, Hapfelmeier A, Walch A, et al. Interaction of Snail and p38 mitogen-activated protein kinase results in shorter overall survival of ovarian cancer patients. Virchows Archiv: an International Journal of Pathology. 2010; 457: 705–713.
[42]
Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, et al. The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Molecular Cell. 2001; 7: 1267–1278.
[43]
Postigo AA. Opposing functions of ZEB proteins in the regulation of the TGFbeta/BMP signaling pathway. The EMBO Journal. 2003; 22: 2443–2452.
[44]
Xiong M, Jiang L, Zhou Y, Qiu W, Fang L, Tan R, et al. The miR-200 family regulates TGF-β1-induced renal tubular epithelial to mesenchymal transition through Smad pathway by targeting ZEB1 and ZEB2 expression. American Journal of Physiology. Renal Physiology. 2012; 302: F369–F379.
[45]
Furusawa T, Moribe H, Kondoh H, Higashi Y. Identification of CtBP1 and CtBP2 as corepressors of zinc finger-homeodomain factor deltaEF1. Molecular and Cellular Biology. 1999; 19: 8581–8590.
[46]
Gregory PA, Bracken CP, Smith E, Bert AG, Wright JA, Roslan S, et al. An autocrine TGF-beta/ZEB/miR-200 signaling network regulates establishment and maintenance of epithelial-mesenchymal transition. Molecular Biology of the Cell. 2011; 22: 1686–1698.
[47]
Rateitschak K, Kaderali L, Wolkenhauer O, Jaster R. Autocrine TGF-β/ZEB/microRNA-200 signal transduction drives epithelial-mesenchymal transition: Kinetic models predict minimal drug dose to inhibit metastasis. Cellular Signalling. 2016; 28: 861–870.
[48]
Yan Z, Tian X, Wang R, Cheng X, Mi J, Xiong L, et al. Title Prognosis Significance of ZEB2 and TGF-β1 as well as Other Clinical Characteristics in Epithelial Ovarian Cancer. International Journal of Gynecological Cancer: Official Journal of the International Gynecological Cancer Society. 2017; 27: 1343–1349.
[49]
Li Y, Fei H, Lin Q, Liang F, You Y, Li M, et al. ZEB2 facilitates peritoneal metastasis by regulating the invasiveness and tumorigenesis of cancer stem-like cells in high-grade serous ovarian cancers. Oncogene. 2021; 40: 5131–5141.
[50]
Wu DI, Liu L, Ren C, Kong D, Zhang P, Jin X, et al. Epithelial-mesenchymal interconversions and the regulatory function of the ZEB family during the development and progression of ovarian cancer. Oncology Letters. 2016; 11: 1463–1468.
[51]
Chen B, Chen B, Zhu Z, Ye W, Zeng J, Liu G, et al. Prognostic value of ZEB-1 in solid tumors: a meta-analysis. BMC Cancer. 2019; 19: 635.
[52]
Pei H, Li Y, Liu M, Chen Y. Targeting Twist expression with small molecules. MedChemComm. 2016; 8: 268–275.
[53]
Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, et al. A twist code determines the onset of osteoblast differentiation. Developmental Cell. 2004; 6: 423–435.
[54]
Wushou A, Hou J, Zhao YJ, Shao ZM. Twist-1 up-regulation in carcinoma correlates to poor survival. International Journal of Molecular Sciences. 2014; 15: 21621–21630.
[55]
Kim K, Park EY, Yoon MS, Suh DS, Kim KH, Lee JH, et al. The Role of TWIST in Ovarian Epithelial Cancers. Korean Journal of Pathology. 2014; 48: 283–291.
[56]
Hosono S, Kajiyama H, Terauchi M, Shibata K, Ino K, Nawa A, et al. Expression of Twist increases the risk for recurrence and for poor survival in epithelial ovarian carcinoma patients. British Journal of Cancer. 2007; 96: 314–320.
[57]
Roberts CM, Tran MA, Pitruzzello MC, Wen W, Loeza J, Dellinger TH, et al. TWIST1 drives cisplatin resistance and cell survival in an ovarian cancer model, via upregulation of GAS6, L1CAM, and Akt signalling. Scientific Reports. 2016; 6: 37652.
[58]
Wang T, Li Y, Tuerhanjiang A, Wang W, Wu Z, Yuan M, et al. Twist2 contributes to cisplatin-resistance of ovarian cancer through the AKT/GSK-3β signaling pathway. Oncology Letters. 2014; 7: 1102–1108.
[59]
Li X, Yang J, Wang X, Li X, Liang J, Xing H. Role of TWIST2, E-cadherin and Vimentin in epithelial ovarian carcinogenesis and prognosis and their interaction in cancer progression. European Journal of Gynaecological Oncology. 2016; 37: 100–108.
[60]
Mao Y, Xu J, Li Z, Zhang N, Yin H, Liu Z. The role of nuclear β-catenin accumulation in the Twist2-induced ovarian cancer EMT. PloS One. 2013; 8: e78200.
[61]
Mao Y, Xu J, Song G, Zhang N, Yin H. Twist2 promotes ovarian cancer cell survival through activation of Akt. Oncology Letters. 2013; 6: 169–174.
[62]
Powell CD, Paullin TR, Aoisa C, Menzie CJ, Ubaldini A, Westerheide SD. The Heat Shock Transcription Factor HSF1 Induces Ovarian Cancer Epithelial-Mesenchymal Transition in a 3D Spheroid Growth Model. PloS One. 2016; 11: e0168389.
[63]
Chen YF, Wang SY, Yang YH, Zheng J, Liu T, Wang L. Targeting HSF1 leads to an antitumor effect in human epithelial ovarian cancer. International Journal of Molecular Medicine. 2017; 39: 1564–1570.
[64]
Brusselaers N, Ekwall K, Durand-Dubief M. Copy number of 8q24.3 drives HSF1 expression and patient outcome in cancer: an individual patient data meta-analysis. Human Genomics. 2019; 13: 54.
[65]
Carpenter RL, Paw I, Dewhirst MW, Lo HW. Akt phosphorylates and activates HSF-1 independent of heat shock, leading to Slug overexpression and epithelial-mesenchymal transition (EMT) of HER2-overexpressing breast cancer cells. Oncogene. 2015; 34: 546–557.
[66]
Parol-Kulczyk M, Gzil A, Maciejewska J, Bodnar M, Grzanka D. Clinicopathological significance of the EMT-related proteins and their interrelationships in prostate cancer. An immunohistochemical study. PloS One. 2021; 16: e0253112.
[67]
Huang J, Guo L. Knockdown of SOX9 Inhibits the Proliferation, Invasion, and EMT in Thyroid Cancer Cells. Oncology Research. 2017; 25: 167–176.
[68]
Yang LX, Guo HB, Liu SY, Feng HP, Shi J. ETS1 promoted cell growth, metastasis and epithelial-mesenchymal transition process in melanoma by regulating miR-16-mediated SOX4 expression. Melanoma Research. 2021; 31: 298–308.
[69]
Feng M, Fang F, Fang T, Jiao H, You S, Wang X, et al. Sox13 promotes hepatocellular carcinoma metastasis by transcriptionally activating Twist1. Laboratory Investigation; a Journal of Technical Methods and Pathology. 2020; 100: 1400–1410.
[70]
Wen Y, Hou Y, Huang Z, Cai J, Wang Z. SOX2 is required to maintain cancer stem cells in ovarian cancer. Cancer Science. 2017; 108: 719–731.
[71]
Yun H, Han GH, Kim J, Chung JY, Kim JH, Cho H. NANOG regulates epithelial-mesenchymal transition via AMPK/mTOR signalling pathway in ovarian cancer SKOV-3 and A2780 cells. Journal of Cellular and Molecular Medicine. 2022; 26: 5277–5291.
[72]
Qin S, Li Y, Cao X, Du J, Huang X. NANOG regulates epithelial-mesenchymal transition and chemoresistance in ovarian cancer. Bioscience Reports. 2017; 37: BSR20160247.
[73]
Siu MKY, Wong ESY, Kong DSH, Chan HY, Jiang L, Wong OGW, et al. Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of E-cadherin and FoxJ1 and contributes to adverse clinical outcome in ovarian cancers. Oncogene. 2013; 32: 3500–3509.
[74]
Liu S, Sun J, Cai B, Xi X, Yang L, Zhang Z, et al. NANOG regulates epithelial-mesenchymal transition and chemoresistance through activation of the STAT3 pathway in epithelial ovarian cancer. Tumour Biology: the Journal of the International Society for Oncodevelopmental Biology and Medicine. 2016; 37: 9671–9680.
[75]
Carter LE, Cook DP, Collins O, Gamwell LF, Dempster HA, Wong HW, et al. COX2 is induced in the ovarian epithelium during ovulatory wound repair and promotes cell survival†. Biology of Reproduction. 2019; 101: 961–974.
[76]
Zhu Y, Nilsson M, Sundfeldt K. Phenotypic plasticity of the ovarian surface epithelium: TGF-beta 1 induction of epithelial to mesenchymal transition (EMT) in vitro. Endocrinology. 2010; 151: 5497–5505.
[77]
Jiang Y, Zhou T, Shi Y, Feng W, Lyu T. A SMYD3/ITGB6/TGFβ1 Positive Feedback Loop Promotes the Invasion and Adhesion of Ovarian Cancer Spheroids. Frontiers in Oncology. 2021; 11: 690618.
[78]
Cheng JC, Auersperg N, Leung PCK. TGF-beta induces serous borderline ovarian tumor cell invasion by activating EMT but triggers apoptosis in low-grade serous ovarian carcinoma cells. PloS One. 2012; 7: e42436.
[79]
Wang Y, Yang B, Zhao J, Yu X, Liu X, Zhang L, et al. Epithelial mesenchymal transition induced by bone morphogenetic protein 9 hinders cisplatin efficacy in ovarian cancer cells. Molecular Medicine Reports. 2019; 19: 1501–1508.
[80]
Ma Y, Zhang H, Xiong C, Liu Z, Xu Q, Feng J, et al. CD146 mediates an E-cadherin-to-N-cadherin switch during TGF-β signaling-induced epithelial-mesenchymal transition. Cancer Letters. 2018; 430: 201–214.
[81]
Wen H, Qian M, He J, Li M, Yu Q, Leng Z. Inhibiting of self-renewal, migration and invasion of ovarian cancer stem cells by blocking TGF-β pathway. PloS One. 2020; 15: e0230230.
[82]
Rafehi S, Ramos Valdes Y, Bertrand M, McGee J, Préfontaine M, Sugimoto A, et al. TGFβ signaling regulates epithelial-mesenchymal plasticity in ovarian cancer ascites-derived spheroids. Endocrine-related Cancer. 2016; 23: 147–159.
[83]
Mitra T, Prasad P, Mukherjee P, Chaudhuri SR, Chatterji U, Roy SS. Stemness and chemoresistance are imparted to the OC cells through TGFβ1 driven EMT. Journal of Cellular Biochemistry. 2018; 119: 5775–5787.
[84]
Fujikake K, Kajiyama H, Yoshihara M, Nishino K, Yoshikawa N, Utsumi F, et al. A novel mechanism of neovascularization in peritoneal dissemination via cancer-associated mesothelial cells affected by TGF-β derived from ovarian cancer. Oncology Reports. 2018; 39: 193–200.
[85]
Yuan D, Qian H, Guo T, Ye J, Jin C, Liu X, et al. LncRNA-ATB Promotes the Tumorigenesis of Ovarian Cancer via Targeting miR-204-3p. OncoTargets and Therapy. 2020; 13: 573–583.
[86]
Bernaudo S, Salem M, Qi X, Zhou W, Zhang C, Yang W, et al. Cyclin G2 inhibits epithelial-to-mesenchymal transition by disrupting Wnt/β-catenin signaling. Oncogene. 2016; 35: 4816–4827.
[87]
Deng Z, Wang L, Hou H, Zhou J, Li X. Epigenetic regulation of IQGAP2 promotes ovarian cancer progression via activating Wnt/β-catenin signaling. International Journal of Oncology. 2016; 48: 153–160.
[88]
Sun J, Yang X, Zhang R, Liu S, Gan X, Xi X, et al. GOLPH3 induces epithelial-mesenchymal transition via Wnt/β-catenin signaling pathway in epithelial ovarian cancer. Cancer Medicine. 2017; 6: 834–844.
[89]
Dong P, Fu H, Chen L, Zhang S, Zhang X, Li H, et al. PCNP promotes ovarian cancer progression by accelerating β-catenin nuclear accumulation and triggering EMT transition. Journal of Cellular and Molecular Medicine. 2020; 24: 8221–8235.
[90]
Zhang LM, Li M, Tian CC, Wang TT, Mi SF. CCAAT enhancer binding protein α suppresses proliferation, metastasis, and epithelial-mesenchymal transition of ovarian cancer cells via suppressing the Wnt/β-catenin signaling. Neoplasma. 2021; 68: 602–612.
[91]
Duan H, Yan Z, Chen W, Wu Y, Han J, Guo H, et al. TET1 inhibits EMT of ovarian cancer cells through activating Wnt/β-catenin signaling inhibitors DKK1 and SFRP2. Gynecologic Oncology. 2017; 147: 408–417.
[92]
Li R, Dong T, Hu C, Lu J, Dai J, Liu P. Salinomycin repressed the epithelial-mesenchymal transition of epithelial ovarian cancer cells via downregulating Wnt/β-catenin pathway. OncoTargets and Therapy. 2017; 10: 1317–1325.
[93]
Gao LN, Hao M, Liu XH, Zhang L, Dong Y, Zhang YF, et al. CXCL14 facilitates the growth and metastasis of ovarian carcinoma cells via activation of the Wnt/β-catenin signaling pathway. Journal of Ovarian Research. 2021; 14: 159.
[94]
Burkhalter RJ, Westfall SD, Liu Y, Stack MS. Lysophosphatidic Acid Initiates Epithelial to Mesenchymal Transition and Induces β-Catenin-mediated Transcription in Epithelial Ovarian Carcinoma. The Journal of Biological Chemistry. 2015; 290: 22143–22154.
[95]
Chen MW, Yang ST, Chien MH, Hua KT, Wu CJ, Hsiao SM, et al. The STAT3-miRNA-92-Wnt Signaling Pathway Regulates Spheroid Formation and Malignant Progression in Ovarian Cancer. Cancer Research. 2017; 77: 1955–1967.
[96]
Liu L, Wang L, Li X. The roles of HOXB8 through activating Wnt/β-catenin and STAT3 signaling pathways in the growth, migration and invasion of ovarian cancer cells. Cytotechnology. 2022; 74: 77–87.
[97]
Luo M, Zhou L, Zhan SJ, Cheng LJ, Li RN, Wang H, et al. ALPL regulates the aggressive potential of high grade serous ovarian cancer cells via a non-canonical WNT pathway. Biochemical and Biophysical Research Communications. 2019; 513: 528–533.
[98]
Qi H, Sun B, Zhao X, Du J, Gu Q, Liu Y, et al. Wnt5a promotes vasculogenic mimicry and epithelial-mesenchymal transition via protein kinase Cα in epithelial ovarian cancer. Oncology Reports. 2014; 32: 771–779.
[99]
Ford CE, Punnia-Moorthy G, Henry CE, Llamosas E, Nixdorf S, Olivier J, et al. The non-canonical Wnt ligand, Wnt5a, is upregulated and associated with epithelial to mesenchymal transition in epithelial ovarian cancer. Gynecologic Oncology. 2014; 134: 338–345.
[100]
Abedini A, Sayed C, Carter LE, Boerboom D, Vanderhyden BC. Non-canonical WNT5a regulates Epithelial-to-Mesenchymal Transition in the mouse ovarian surface epithelium. Scientific Reports. 2020; 10: 9695.
[101]
Lüönd F, Pirkl M, Hisano M, Prestigiacomo V, Kalathur RK, Beerenwinkel N, et al. Hierarchy of TGFβ/SMAD, Hippo/YAP/TAZ, and Wnt/β-catenin signaling in melanoma phenotype switching. Life Science Alliance. 2022; 5: e202101010.
[102]
Skoda AM, Simovic D, Karin V, Kardum V, Vranic S, Serman L. The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosnian Journal of Basic Medical Sciences. 2018; 18: 8–20.
[103]
Zeng C, Chen T, Zhang Y, Chen Q. Hedgehog signaling pathway regulates ovarian cancer invasion and migration via adhesion molecule CD24. Journal of Cancer. 2017; 8: 786–792.
[104]
Zhang H, Wang Y, Chen T, Zhang Y, Xu R, Wang W, et al. Aberrant Activation Of Hedgehog Signalling Promotes Cell Migration And Invasion Via Matrix Metalloproteinase-7 In Ovarian Cancer Cells. Journal of Cancer. 2019; 10: 990–1003.
[105]
Zhang H, Hu L, Cheng M, Wang Q, Hu X, Chen Q. The Hedgehog signaling pathway promotes chemotherapy resistance via multidrug resistance protein 1 in ovarian cancer. Oncology Reports. 2020; 44: 2610–2620.
[106]
Tai H, Wu Z, Sun S, Zhang Z, Xu C. FGFRL1 Promotes Ovarian Cancer Progression by Crosstalk with Hedgehog Signaling. Journal of Immunology Research. 2018; 2018: 7438608.
[107]
Liu Y, Gao S, Zhu J, Zheng Y, Zhang H, Sun H. Dihydroartemisinin induces apoptosis and inhibits proliferation, migration, and invasion in epithelial ovarian cancer via inhibition of the hedgehog signaling pathway. Cancer Medicine. 2018; 7: 5704–5715.
[108]
Sneha S, Nagare RP, Sidhanth C, Krishnapriya S, Garg M, Ramachandran B, et al. The hedgehog pathway regulates cancer stem cells in serous adenocarcinoma of the ovary. Cellular Oncology (Dordrecht). 2020; 43: 601–616.
[109]
Ke Z, Caiping S, Qing Z, Xiaojing W. Sonic hedgehog-Gli1 signals promote epithelial-mesenchymal transition in ovarian cancer by mediating PI3K/AKT pathway. Medical Oncology (Northwood, London, England). 2015; 32: 368.
[110]
Ma J, Zhou C, Chen X. miR-636 inhibits EMT, cell proliferation and cell cycle of ovarian cancer by directly targeting transcription factor Gli2 involved in Hedgehog pathway. Cancer Cell International. 2021; 21: 64.
[111]
Zhang J, Cui Y, Sun S, Cao J, Fang X. Casticin inhibits the epithelial-mesenchymal transition in ovarian carcinoma via the hedgehog signaling pathway. Oncology Letters. 2018; 15: 4495–4502.
[112]
Tang C, Mei L, Pan L, Xiong W, Zhu H, Ruan H, et al. Hedgehog signaling through GLI1 and GLI2 is required for epithelial-mesenchymal transition in human trophoblasts. Biochimica et Biophysica Acta. 2015; 1850: 1438–1448.
[113]
Harvey KF, Zhang X, Thomas DM. The Hippo pathway and human cancer. Nature Reviews. Cancer. 2013; 13: 246–257.
[114]
Yagi H, Onoyama I, Asanoma K, Hori E, Yasunaga M, Kodama K, et al. Gα13-mediated LATS1 down-regulation contributes to epithelial-mesenchymal transition in ovarian cancer. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 2019; 33: 13683–13694.
[115]
Chen G, Xie J, Huang P, Yang Z. Overexpression of TAZ promotes cell proliferation, migration and epithelial-mesenchymal transition in ovarian cancer. Oncology Letters. 2016; 12: 1821–1825.
[116]
Xia Y, Chang T, Wang Y, Liu Y, Li W, Li M, et al. YAP promotes ovarian cancer cell tumorigenesis and is indicative of a poor prognosis for ovarian cancer patients. PloS One. 2014; 9: e91770.
[117]
Muñoz-Galván S, Felipe-Abrio B, Verdugo-Sivianes EM, Perez M, Jiménez-García MP, Suarez-Martinez E, et al. Downregulation of MYPT1 increases tumor resistance in ovarian cancer by targeting the Hippo pathway and increasing the stemness. Molecular Cancer. 2020; 19: 7.
[118]
Yang WH, Huang Z, Wu J, Ding CKC, Murphy SK, Chi JT. A TAZ-ANGPTL4-NOX2 Axis Regulates Ferroptotic Cell Death and Chemoresistance in Epithelial Ovarian Cancer. Molecular Cancer Research: MCR. 2020; 18: 79–90.
[119]
Xiao L, Shi XY, Li ZL, Li M, Zhang MM, Yan SJ, et al. Downregulation of LINC01508 contributes to cisplatin resistance in ovarian cancer via the regulation of the Hippo-YAP pathway. Journal of Gynecologic Oncology. 2021; 32: e77.
[120]
Mohamed Z, Hassan MK, Okasha S, Mitamura T, Keshk S, Konno Y, et al. miR-363 confers taxane resistance in ovarian cancer by targeting the Hippo pathway member, LATS2. Oncotarget. 2018; 9: 30053–30065.
[121]
Xia Y, Zhang YL, Yu C, Chang T, Fan HY. YAP/TEAD co-activator regulated pluripotency and chemoresistance in ovarian cancer initiated cells. PloS One. 2014; 9: e109575.
[122]
Bendoraite A, Knouf EC, Garg KS, Parkin RK, Kroh EM, O’Briant KC, et al. Regulation of miR-200 family microRNAs and ZEB transcription factors in ovarian cancer: evidence supporting a mesothelial-to-epithelial transition. Gynecologic Oncology. 2010; 116: 117–125.
[123]
Zhang M, Jabbari N, Satpathy M, Matyunina LV, Wang Y, McDonald LD, et al. Sequence diverse miRNAs converge to induce mesenchymal-to-epithelial transition in ovarian cancer cells through direct and indirect regulatory controls. Cancer Letters. 2019; 459: 168–175.
[124]
Chen D, Zhang Y, Wang J, Chen J, Yang C, Cai K, et al. MicroRNA-200c overexpression inhibits tumorigenicity and metastasis of CD117+CD44+ ovarian cancer stem cells by regulating epithelial-mesenchymal transition. Journal of Ovarian Research. 2013; 6: 50.
[125]
Yang C, Li H, Zhang T, Chu Y, Chen D, Zuo J. miR-200c overexpression inhibits the invasion and tumorigenicity of epithelial ovarian cancer cells by suppressing lncRNA HOTAIR in mice. Journal of Cellular Biochemistry. 2020; 121: 1514–1523.
[126]
Wang B, Li X, Zhao G, Yan H, Dong P, Watari H, et al. miR-203 inhibits ovarian tumor metastasis by targeting BIRC5 and attenuating the TGFβ pathway. Journal of Experimental & Clinical Cancer Research: CR. 2018; 37: 235.
[127]
Biamonte F, Santamaria G, Sacco A, Perrone FM, Di Cello A, Battaglia AM, et al. MicroRNA let-7g acts as tumor suppressor and predictive biomarker for chemoresistance in human epithelial ovarian cancer. Scientific Reports. 2019; 9: 5668.
[128]
Li N, Yang L, Sun Y, Wu X. MicroRNA-16 inhibits migration and invasion via regulation of the Wnt/β-catenin signaling pathway in ovarian cancer. Oncology Letters. 2019; 17: 2631–2638.
[129]
Ye Z, Zhao L, Li J, Chen W, Li X. miR-30d Blocked Transforming Growth Factor β1-Induced Epithelial-Mesenchymal Transition by Targeting Snail in Ovarian Cancer Cells. International Journal of Gynecological Cancer: Official Journal of the International Gynecological Cancer Society. 2015; 25: 1574–1581.
[130]
Zhu X, Shen H, Yin X, Long L, Xie C, Liu Y, et al. miR-186 regulation of Twist1 and ovarian cancer sensitivity to cisplatin. Oncogene. 2016; 35: 323–332.
[131]
Ye Q, Lei L, Shao L, Shi J, Jia J, Tong X. MicroRNA 141 inhibits epithelial mesenchymal transition, and ovarian cancer cell migration and invasion. Molecular Medicine Reports. 2017; 16: 6743–6749.
[132]
Yu JL, Gao X. MicroRNA 1301 inhibits cisplatin resistance in human ovarian cancer cells by regulating EMT and autophagy. European Review for Medical and Pharmacological Sciences. 2020; 24: 1688–1696.
[133]
Tung CH, Kuo LW, Huang MF, Wu YY, Tsai YT, Wu JE, et al. MicroRNA-150-5p promotes cell motility by inhibiting c-Myb-mediated Slug suppression and is a prognostic biomarker for recurrent ovarian cancer. Oncogene. 2020; 39: 862–876.
[134]
Zhang LY, Chen Y, Jia J, Zhu X, He Y, Wu LM. MiR-27a promotes EMT in ovarian cancer through active Wnt/β-catenin signalling by targeting FOXO1. Cancer Biomarkers: Section a of Disease Markers. 2019; 24: 31–42.
[135]
Wang JY, Lu AQ, Chen LJ. LncRNAs in ovarian cancer. Clinica Chimica Acta; International Journal of Clinical Chemistry. 2019; 490: 17–27.
[136]
Wu Y, Zhou Y, He J, Sun H, Jin Z. Long non-coding RNA H19 mediates ovarian cancer cell cisplatin-resistance and migration during EMT. International Journal of Clinical and Experimental Pathology. 2019; 12: 2506–2515.
[137]
Wang B, Liu M, Zhuang R, Jiang J, Gao J, Wang H, et al. Long non-coding RNA CCAT2 promotes epithelial-mesenchymal transition involving Wnt/β-catenin pathway in epithelial ovarian carcinoma cells. Oncology Letters. 2018; 15: 3369–3375.
[138]
Li J, Huang Y, Deng X, Luo M, Wang X, Hu H, et al. Long noncoding RNA H19 promotes transforming growth factor-β-induced epithelial-mesenchymal transition by acting as a competing endogenous RNA of miR-370-3p in ovarian cancer cells. OncoTargets and Therapy. 2018; 11: 427–440.
[139]
Yuan D, Zhang X, Zhao Y, Qian H, Wang H, He C, et al. Role of lncRNA-ATB in ovarian cancer and its mechanisms of action. Experimental and Therapeutic Medicine. 2020; 19: 965–971.
[140]
Wang K, Hu YB, Zhao Y, Ye C. Long non coding RNA ASAP1 IT1 suppresses ovarian cancer progression by regulating Hippo/YAP signaling. International Journal of Molecular Medicine. 2021; 47: 44.
[141]
San Francisco IF, DeWolf WC, Peehl DM, Olumi AF. Expression of transforming growth factor-beta 1 and growth in soft agar differentiate prostate carcinoma-associated fibroblasts from normal prostate fibroblasts. International Journal of Cancer. 2004; 112: 213–218.
[142]
Ringuette Goulet C, Bernard G, Tremblay S, Chabaud S, Bolduc S, Pouliot F. Exosomes induce fibroblast differentiation into cancer-associated fibroblasts through TGFβ signaling. Molecular Cancer Research. 2018; 16: 1196–1204.
[143]
Wang L, Zhang F, Cui JY, Chen L, Chen YT, Liu BW. CAFs enhance paclitaxel resistance by inducing EMT through the IL 6/JAK2/STAT3 pathway. Oncology Reports. 2018; 39: 2081–2090.
[144]
Yue H, Li W, Chen R, Wang J, Lu X, Li J. Stromal POSTN induced by TGF-β1 facilitates the migration and invasion of ovarian cancer. Gynecologic Oncology. 2021; 160: 530–538.
[145]
Sun Y, Fan X, Zhang Q, Shi X, Xu G, Zou C. Cancer-associated fibroblasts secrete FGF-1 to promote ovarian proliferation, migration, and invasion through the activation of FGF-1/FGFR4 signaling. Tumour Biology: the Journal of the International Society for Oncodevelopmental Biology and Medicine. 2017; 39: 1010428317712592.
[146]
Yang Y, Yin S, Li S, Chen Y, Yang L. Stanniocalcin 1 in tumor microenvironment promotes metastasis of ovarian cancer. OncoTargets and Therapy. 2019; 12: 2789–2798.
[147]
Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nature Reviews. Immunology. 2011; 11: 750–761.
[148]
Cortés M, Sanchez‐Moral L, de Barrios O, Fernández‐Aceñero MJ, Martínez‐Campanario M, Esteve‐Codina A, et al. Tumor‐associated macrophages (TAMs) depend on ZEB1 for their cancer‐promoting roles. The EMBO Journal. 2017; 36: 3336–3355.
[149]
Long L, Hu Y, Long T, Lu X, Tuo Y, Li Y, et al. Tumor-associated macrophages induced spheroid formation by CCL18-ZEB1-M-CSF feedback loop to promote transcoelomic metastasis of ovarian cancer. Journal for Immunotherapy of Cancer. 2021; 9: e003973.
[150]
Ying X, Wu Q, Wu X, Zhu Q, Wang X, Jiang L, et al. Epithelial ovarian cancer-secreted exosomal miR-222-3p induces polarization of tumor-associated macrophages. Oncotarget. 2016; 7: 43076–43087.
[151]
Chen X, Ying X, Wang X, Wu X, Zhu Q, Wang X. Exosomes derived from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce macrophage M2 polarization. Oncology Reports. 2017; 38: 522–528.
[152]
Zhang Y, Dong W, Wang J, Cai J, Wang Z. Human omental adipose-derived mesenchymal stem cell-conditioned medium alters the proteomic profile of epithelial ovarian cancer cell lines in vitro. OncoTargets and Therapy. 2017; 10: 1655–1663.
[153]
Liu X, Zhao G, Huo X, Wang Y, Tigyi G, Zhu BM, et al. Adipose-Derived Stem Cells Facilitate Ovarian Tumor Growth and Metastasis by Promoting Epithelial to Mesenchymal Transition Through Activating the TGF-β Pathway. Frontiers in Oncology. 2021; 11: 756011.
[154]
Tang H, Chu Y, Huang Z, Cai J, Wang Z. The metastatic phenotype shift toward myofibroblast of adipose-derived mesenchymal stem cells promotes ovarian cancer progression. Carcinogenesis. 2020; 41: 182–193.
[155]
Cho JA, Park H, Lim EH, Kim KH, Choi JS, Lee JH, et al. Exosomes from ovarian cancer cells induce adipose tissue-derived mesenchymal stem cells to acquire the physical and functional characteristics of tumor-supporting myofibroblasts. Gynecologic Oncology. 2011; 123: 379–386.
[156]
Chu Y, Zhu C, Wang Q, Liu M, Wan W, Zhou J, et al. Adipose-derived mesenchymal stem cells induced PAX8 promotes ovarian cancer cell growth by stabilizing TAZ protein. Journal of Cellular and Molecular Medicine. 2021; 25: 4434–4443.
[157]
Di Palma T, Lucci V, de Cristofaro T, Filippone MG, Zannini M. A role for PAX8 in the tumorigenic phenotype of ovarian cancer cells. BMC Cancer. 2014; 14: 292.
[158]
Gao D, Nolan D, McDonnell K, Vahdat L, Benezra R, Altorki N, et al. Bone marrow-derived endothelial progenitor cells contribute to the angiogenic switch in tumor growth and metastatic progression. Biochimica et Biophysica Acta. 2009; 1796: 33–40.
[159]
Su Y, Gao L, Teng L, Wang Y, Cui J, Peng S, et al. Id1 enhances human ovarian cancer endothelial progenitor cell angiogenesis via PI3K/Akt and NF-κB/MMP-2 signaling pathways. Journal of Translational Medicine. 2013; 11: 132.
[160]
Su Y, Zheng L, Wang Q, Bao J, Cai Z, Liu A. The PI3K/Akt pathway upregulates Id1 and integrin α4 to enhance recruitment of human ovarian cancer endothelial progenitor cells. BMC Cancer. 2010; 10: 459.
[161]
Bagley RG, Walter-Yohrling J, Cao X, Weber W, Simons B, Cook BP, et al. Endothelial precursor cells as a model of tumor endothelium: characterization and comparison with mature endothelial cells. Cancer Research. 2003; 63: 5866–5873.
[162]
Su Y, Zheng L, Wang Q, Li W, Cai Z, Xiong S, et al. Quantity and clinical relevance of circulating endothelial progenitor cells in human ovarian cancer. Journal of Experimental & Clinical Cancer Research: CR. 2010; 29: 27.
[163]
Teng L, Peng S, Guo H, Liang H, Xu Z, Su Y, et al. Conditioned media from human ovarian cancer endothelial progenitor cells induces ovarian cancer cell migration by activating epithelial-to-mesenchymal transition. Cancer Gene Therapy. 2015; 22: 518–523.
[164]
Cheng JC, Leung PCK. Type I collagen down-regulates E-cadherin expression by increasing PI3KCA in cancer cells. Cancer Letters. 2011; 304: 107–116.
[165]
Liu M, Zhang X, Long C, Xu H, Cheng X, Chang J, et al. Collagen-based three-dimensional culture microenvironment promotes epithelial to mesenchymal transition and drug resistance of human ovarian cancer in vitro. RSC Advances. 2018; 8: 8910–8919.
[166]
So KA, Min KJ, Hong JH, Lee JK. Interleukin-6 expression by interactions between gynecologic cancer cells and human mesenchymal stem cells promotes epithelial-mesenchymal transition. International Journal of Oncology. 2015; 47: 1451–1459.
[167]
Bao B, Azmi AS, Ali S, Ahmad A, Li Y, Banerjee S, et al. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. Biochimica et Biophysica Acta. 2012; 1826: 272–296.
[168]
Greville G, Llop E, Huang C, Creagh-Flynn J, Pfister S, O’Flaherty R, et al. Hypoxia Alters Epigenetic and N-Glycosylation Profiles of Ovarian and Breast Cancer Cell Lines in-vitro. Frontiers in Oncology. 2020; 10: 1218.
[169]
Sun L, Lin P, Qin Z, Liu Y, Deng LL, Lu C. Hypoxia promotes HO-8910PM ovarian cancer cell invasion via Snail-mediated MT1-MMP upregulation. Experimental Biology and Medicine (Maywood, N.J.). 2015; 240: 1434–1445.
[170]
Seo EJ, Kim DK, Jang IH, Choi EJ, Shin SH, Lee SI, et al. Hypoxia-NOTCH1-SOX2 signaling is important for maintaining cancer stem cells in ovarian cancer. Oncotarget. 2016; 7: 55624–55638.
[171]
Du J, Sun B, Zhao X, Gu Q, Dong X, Mo J, et al. Hypoxia promotes vasculogenic mimicry formation by inducing epithelial-mesenchymal transition in ovarian carcinoma. Gynecologic Oncology. 2014; 133: 575–583.
[172]
Yin G, Alvero AB, Craveiro V, Holmberg JC, Fu HH, Montagna MK, et al. Constitutive proteasomal degradation of TWIST-1 in epithelial-ovarian cancer stem cells impacts differentiation and metastatic potential. Oncogene. 2013; 32: 39–49.
[173]
Liu T, Zhao L, Zhang Y, Chen W, Liu D, Hou H, et al. Ginsenoside 20(S)-Rg3 targets HIF-1α to block hypoxia-induced epithelial-mesenchymal transition in ovarian cancer cells. PloS One. 2014; 9: e103887.
[174]
Ding L, Zhao L, Chen W, Liu T, Li Z, Li X. miR-210, a modulator of hypoxia-induced epithelial-mesenchymal transition in ovarian cancer cell. International Journal of Clinical and Experimental Medicine. 2015; 8: 2299–2307.
[175]
Singh SK, Mishra MK, Singh R. Hypoxia-inducible factor-1α induces CX3CR1 expression and promotes the epithelial to mesenchymal transition (EMT) in ovarian cancer cells. Journal of Ovarian Research. 2019; 12: 42.
[176]
Li Y, Wang J, Chen W, Chen X, Wang J. Overexpression of STAT4 under hypoxia promotes EMT through miR-200a/STAT4 signal pathway. Life Sciences. 2021; 273: 119263.
[177]
Sun L, Li H, Chen J, Iwasaki Y, Kubota T, Matsuoka M, et al. PIASy mediates hypoxia-induced SIRT1 transcriptional repression and epithelial-to-mesenchymal transition in ovarian cancer cells. Journal of Cell Science. 2013; 126: 3939–3947.
[178]
Li L, Qiu C, Hou M, Wang X, Huang C, Zou J, et al. Ferroptosis in Ovarian Cancer: A Novel Therapeutic Strategy. Frontiers in Oncology. 2021; 11: 665945.
[179]
Wang Y, Ma J, Shen H, Wang C, Sun Y, Howell SB, et al. Reactive oxygen species promote ovarian cancer progression via the HIF-1α/LOX/E-cadherin pathway. Oncology Reports. 2014; 32: 2150–2158.
[180]
Zhao Z, Zhao J, Xue J, Zhao X, Liu P. Autophagy inhibition promotes epithelial-mesenchymal transition through ROS/HO-1 pathway in ovarian cancer cells. American Journal of Cancer Research. 2016; 6: 2162–2177.
[181]
Bregenzer ME, Horst EN, Mehta P, Novak CM, Repetto T, Mehta G. The Role of Cancer Stem Cells and Mechanical Forces in Ovarian Cancer Metastasis. Cancers. 2019; 11: 1008.
[182]
Martinez A, Buckley M, Scalise CB, Katre AA, Dholakia JJ, Crossman D, et al. Understanding the effect of mechanical forces on ovarian cancer progression. Gynecologic Oncology. 2021; 162: 154–162.
[183]
Fan Y, Sun Q, Li X, Feng J, Ao Z, Li X, et al. Substrate Stiffness Modulates the Growth, Phenotype, and Chemoresistance of Ovarian Cancer Cells. Frontiers in Cell and Developmental Biology. 2021; 9: 718834.
[184]
McGrail DJ, Kieu QMN, Dawson MR. The malignancy of metastatic ovarian cancer cells is increased on soft matrices through a mechanosensitive Rho-ROCK pathway. Journal of Cell Science. 2014; 127: 2621–2626.
[185]
Deng J, Bai X, Feng X, Ni J, Beretov J, Graham P, et al. Inhibition of PI3K/Akt/mTOR signaling pathway alleviates ovarian cancer chemoresistance through reversing epithelial-mesenchymal transition and decreasing cancer stem cell marker expression. BMC Cancer. 2019; 19: 618.
[186]
Wang Y, Zhao G, Condello S, Huang H, Cardenas H, Tanner EJ, et al. Frizzled-7 Identifies Platinum-Tolerant Ovarian Cancer Cells Susceptible to Ferroptosis. Cancer Research. 2021; 81: 384–399.
[187]
Zhao J, Tan W, Zhang L, Liu J, Shangguan M, Chen J, et al. FGFR3 phosphorylates EGFR to promote cisplatin-resistance in ovarian cancer. Biochemical Pharmacology. 2021; 190: 114536.
[188]
Kielbik M, Szulc-Kielbik I, Klink M. Impact of Selected Signaling Proteins on SNAIL 1 and SNAIL 2 Expression in Ovarian Cancer Cell Lines in Relation to Cells’ Cisplatin Resistance and EMT Markers Level. International Journal of Molecular Sciences. 2021; 22: 980.
[189]
Bahar E, Kim JY, Kim HS, Yoon H. Establishment of Acquired Cisplatin Resistance in Ovarian Cancer Cell Lines Characterized by Enriched Metastatic Properties with Increased Twist Expression. International Journal of Molecular Sciences. 2020; 21: 7613.
[190]
Ricci F, Fratelli M, Guffanti F, Porcu L, Spriano F, Dell’Anna T, et al. Patient-derived ovarian cancer xenografts re-growing after a cisplatinum treatment are less responsive to a second drug re-challenge: a new experimental setting to study response to therapy. Oncotarget. 2017; 8: 7441–7451.
[191]
Baribeau S, Chaudhry P, Parent S, Asselin É. Resveratrol inhibits cisplatin-induced epithelial-to-mesenchymal transition in ovarian cancer cell lines. PloS One. 2014; 9: e86987.
[192]
Latifi A, Abubaker K, Castrechini N, Ward AC, Liongue C, Dobill F, et al. Cisplatin treatment of primary and metastatic epithelial ovarian carcinomas generates residual cells with mesenchymal stem cell-like profile. Journal of Cellular Biochemistry. 2011; 112: 2850–2864.
[193]
Wintzell M, Löfstedt L, Johansson J, Pedersen AB, Fuxe J, Shoshan M. Repeated cisplatin treatment can lead to a multiresistant tumor cell population with stem cell features and sensitivity to 3-bromopyruvate. Cancer Biology & Therapy. 2012; 13: 1454–1462.
[194]
Zhao Y, He M, Cui L, Gao M, Zhang M, Yue F, et al. Chemotherapy exacerbates ovarian cancer cell migration and cancer stem cell-like characteristics through GLI1. British Journal of Cancer. 2020; 122: 1638–1648.
[195]
Pan JX, Qu F, Wang FF, Xu J, Mu LS, Ye LY, et al. Aberrant SERPINE1 DNA methylation is involved in carboplatin induced epithelial-mesenchymal transition in epithelial ovarian cancer. Archives of Gynecology and Obstetrics. 2017; 296: 1145–1152.
[196]
Chebouti I, Kasimir-Bauer S, Buderath P, Wimberger P, Hauch S, Kimmig R, et al. EMT-like circulating tumor cells in ovarian cancer patients are enriched by platinum-based chemotherapy. Oncotarget. 2017; 8: 48820–48831.
[197]
Kajiyama H, Shibata K, Terauchi M, Yamashita M, Ino K, Nawa A, et al. Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. International Journal of Oncology. 2007; 31: 277–283.
[198]
Jolly MK, Boareto M, Huang B, Jia D, Lu M, Ben-Jacob E, et al. Implications of the Hybrid Epithelial/Mesenchymal Phenotype in Metastasis. Frontiers in Oncology. 2015; 5: 155.
[199]
Pastushenko I, Brisebarre A, Sifrim A, Fioramonti M, Revenco T, Boumahdi S, et al. Identification of the tumour transition states occurring during EMT. Nature. 2018; 556: 463–468.
[200]
Aiello NM, Maddipati R, Norgard RJ, Balli D, Li J, Yuan S, et al. EMT Subtype Influences Epithelial Plasticity and Mode of Cell Migration. Developmental Cell. 2018; 45: 681–695.e4.
[201]
Chirshev E, Hojo N, Bertucci A, Sanderman L, Nguyen A, Wang H, et al. Epithelial/mesenchymal heterogeneity of high-grade serous ovarian carcinoma samples correlates with miRNA let-7 levels and predicts tumor growth and metastasis. Molecular Oncology. 2020; 14: 2796–2813.
[202]
Varankar SS, More M, Abraham A, Pansare K, Kumar B, Narayanan NJ, et al. Functional balance between Tcf21-Slug defines cellular plasticity and migratory modalities in high grade serous ovarian cancer cell lines. Carcinogenesis. 2020; 41: 515–526.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top